Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Br J Cancer ; 111(7): 1319-26, 2014 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-25003660

RESUMO

BACKGROUND: Topoisomerase I (Topo I) poisons (e.g., camptothecin (CPT)), used to treat cancer, cause DNA breaks that are most cytotoxic during S phase. PARP-1 promotes DNA repair and PARP inhibitors (PARPi) sensitise cells to Topo I poisons. We aimed to determine whether chemosensitisation is also S phase specific using rucaparib, a potent PARPi in advanced clinical evaluation. METHODS: The impact of rucaparib, on CPT-induced cytotoxicity was measured in human colon cancer (LoVo) and leukaemic (K562) cells in asynchronous and cell cycle phase-separated cultures. Topoisomerase I and PARP levels and activity and the effect of rucaparib on DNA single-strand breaks (SSBs), double-strand breaks (DSBs) and collapsed replication fork induction and repair were determined in cell cycle phase-separated cells. RESULTS: The cytotoxicity of CPT was greatest during S phase, partially attributable to high Topo I activity, and rucaparib preferentially sensitised S-phase cells. Rucaparib increased CPT-induced DNA SSBs in all phases of the cell cycle, and increased DSB and γH2AX foci in S and G2, with γH2AX foci being highest in S-phase cells. Repair of SSBs and DSBs was most rapid during S then G2 phases and was substantially hindered by rucaparib. CONCLUSIONS: Rucaparib preferentially sensitises S-phase cells by increasing the frequency of collapsed replication forks.


Assuntos
Camptotecina/farmacologia , Indóis/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases , Pontos de Checagem da Fase S do Ciclo Celular/efeitos dos fármacos , Inibidores da Topoisomerase I/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Quebras de DNA de Cadeia Dupla , Reparo do DNA/efeitos dos fármacos , DNA Topoisomerases Tipo I/metabolismo , Ensaios de Seleção de Medicamentos Antitumorais , Sinergismo Farmacológico , Humanos , Células K562 , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/metabolismo
2.
Br J Cancer ; 111(1): 94-100, 2014 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-24867690

RESUMO

BACKGROUND: Patients with malignant pleural effusions (MPEs) generally have advanced disease with poor survival and few therapeutic options. Cells within MPEs may be used to stratify patients for targeted therapy. Targeted therapy with poly(ADP ribose) polymerase inhibitors (PARPi) depends on identifying homologous recombination DNA repair (HRR)-defective cancer cells. We aimed to determine the feasibility of assaying HRR status in MPE cells. METHODS: A total of 15 MPE samples were collected from consenting patients with non-small-cell lung cancer (NSCLC), mesothelioma and ovarian and breast cancer. Primary cultures were confirmed as epithelial by pancytokeratin, and HRR status was determined by the detection of γH2AX and RAD51 foci following a 24-h exposure to rucaparib, by immunofluorescence microscopy. Massively parallel next-generation sequencing of DNA repair genes was performed on cultured MPE cells. RESULTS: From 15 MPE samples, 13 cultures were successfully established, with HRR function successfully determined in 12 cultures. Four samples - three NSCLC and one mesothelioma - were HRR defective and eight samples - one NSCLC, one mesothelioma, one sarcomatoid, one breast and four ovarian cancers - were HRR functional. No mutations in DNA repair genes were associated with HRR status, but there was probable loss of heterozygosity of FANCG, RPA1 and PARP1. CONCLUSIONS: HRR function can be successfully detected in MPE cells demonstrating the potential to stratify patients for targeted therapy with PARPi.


Assuntos
Derrame Pleural Maligno/genética , Reparo de DNA por Recombinação , Idoso , Idoso de 80 Anos ou mais , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Estudos de Viabilidade , Feminino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Derrame Pleural Maligno/patologia
3.
Br J Cancer ; 110(8): 1977-84, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24556618

RESUMO

BACKGROUND: Poly(ADP-ribose) polymerase-1 (PARP) inhibitors (PARPi) exploit tumour-specific defects in homologous recombination DNA repair and continuous dosing is most efficacious. Early clinical trial data with rucaparib suggested that it caused sustained PARP inhibition. Here we investigate the mechanism of this durable inhibition and potential exploitation. METHODS: Uptake and retention of rucaparib and persistence of PARP inhibition were determined by radiochemical and immunological assays in human cancer cell lines. The pharmacokinetics and pharmacodynamics of rucaparib were determined in tumour-bearing mice and the efficacy of different schedules of rucaparib was determined in mice bearing homologous recombination DNA repair-defective tumours. RESULTS: Rucaparib accumulation is carrier mediated (Km=8.4±1.2 µM, Vmax=469±22 pmol per 10(6) cells per 10 min), reaching steady-state levels >10 times higher than the extracellular concentration within 30 min. Rucaparib is retained in cells and inhibits PARP ≥50% for ≥72 h days after a 30-min pulse of 400 nM. In Capan-1 tumour-bearing mice rucaparib accumulated and was retained in the tumours, and PARP was inhibited for 7 days following a single dose of 10 mg kg(-1) i.p or 150 mg kg(-1) p.o. by 70% and 90%, respectively. Weekly dosing of 150 mg kg(-1) p.o once a week was as effective as 10 mg kg(-1) i.p daily for five days every week for 6 weeks in delaying Capan-1 tumour growth. CONCLUSIONS: Rucaparib accumulates and is retained in tumour cells and inhibits PARP for long periods such that weekly schedules have equivalent anticancer activity to daily dosing in a pre-clinical model, suggesting that clinical evaluation of alternative schedules of rucaparib should be considered.


Assuntos
Inibidores Enzimáticos/administração & dosagem , Indóis/administração & dosagem , Poli(ADP-Ribose) Polimerases/genética , Animais , Linhagem Celular Tumoral , Reparo do DNA/efeitos dos fármacos , Esquema de Medicação , Inibidores Enzimáticos/sangue , Inibidores Enzimáticos/farmacocinética , Recombinação Homóloga/efeitos dos fármacos , Humanos , Indóis/sangue , Indóis/farmacocinética , Camundongos , Inibidores de Poli(ADP-Ribose) Polimerases , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Br J Pharmacol ; 169(8): 1745-65, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23682925

RESUMO

UNLABELLED: The DNA damage response (DDR), consisting of an orchestrated network of proteins effecting repair and signalling to cell cycle arrest, to allow time to repair, is essential for cell viability and to prevent DNA damage being passed on to daughter cells. The DDR is dysregulated in cancer with some pathways up-regulated and others down-regulated or lost. Up-regulated pathways can confer resistance to anti-cancer DNA damaging agents. Therefore, inhibitors of key components of these pathways have the potential to prevent this therapeutic resistance. Conversely, defects in a particular DDR pathway may lead to dependence on a complementary pathway. Inhibition of this complementary pathway may result in tumour-specific cell killing. Thus, inhibitors of the DDR have the potential to increase the efficacy of DNA damaging chemotherapy and radiotherapy and have single-agent activity against tumours with a specific DDR defect. This review describes the compounds that have been designed to inhibit specific DDR targets and summarizes the pre-clinical and clinical evaluation of these inhibitors of DNA damage signalling and repair. LINKED ARTICLES: This article is part of a themed section on Emerging Therapeutic Aspects in Oncology. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2013.169.issue-8.


Assuntos
Antineoplásicos/farmacologia , Dano ao DNA/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Animais , Pontos de Checagem do Ciclo Celular , Reparo do DNA/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Humanos , Regulação para Cima
5.
Cancer Treat Rev ; 38(2): 89-100, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21715099

RESUMO

Homologous recombination (HR) is a process by which DNA double strand breaks are repaired through the alignment of homologous sequences of DNA. Interest continues to increase in HR pathway function due to the development of new therapeutic agents which selectively exploit DNA damage repair pathways. Currently the most promising of these new agents are inhibitors of poly(ADP ribose) polymerase (PARP). The response of cancers known to be deficient in HR, due to BRCA1 or 2 mutations has been demonstrated, and a wider use of PARP inhibitors in cancers with mutations of other HR pathway genes has been suggested. With ongoing clinical studies into the use of PARP inhibitors, further understanding of the HR pathway, to allow patient selection by cancer biology, is now essential. Numerous studies have investigated individual aberrations of genes involved in the HR pathway. Here we collate this evidence to give an overview of the role of the HR pathway in human cancer.


Assuntos
Recombinação Homóloga , Neoplasias/genética , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Feminino , Humanos , Masculino
6.
Breast ; 21(1): 20-6, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21983489

RESUMO

In trials in triple negative breast cancer (TNBC), oestrogen and progesterone receptor negativity should be defined as < 1% positive cells. Negativity is a ratio of <2 between Her2 gene copy number and centromere of chromosome 17 or a copy number of 4 or less. In routine practice, immunohistochemistry is acceptable given stringent quality assurance. Triple negativity emerging after neoadjuvant treatment differs from primary TN and such patients should not enter TNBC trials. Patients relapsing with TN metastases should be eligible even if their primary was positive. Rare TN subtypes such as apocrine, adenoid-cystic and low-grade metaplastic tumours should be excluded. TN and basal-like (BL) signatures overlap but are not equivalent. Since the significance of basal cytokeratin or EGFR overexpression is not known and we lack validated assays, these features should not be used to subclassify TN tumours. Tissue collection in trials is mandatory so the effect on outcome of different tumour phenotypes and BRCA mutation can be explored. No prospective studies have established that TN tumours have particular sensitivity or resistance to any specific chemotherapy agent or radiation. TNBC patients should be treated according to tumour and clinical characteristics.


Assuntos
Neoplasias da Mama/terapia , Metástase Neoplásica , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Ensaios Clínicos como Assunto , Feminino , Genes erbB-2/fisiologia , Humanos , Estadiamento de Neoplasias , Projetos de Pesquisa
7.
Ther Adv Med Oncol ; 3(6): 257-67, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22084640

RESUMO

The modulation of DNA repair pathways for therapeutic benefit in cancer has now become a reality with the development of poly (ADP-ribose) polymerase inhibitors (PARPi). PARP is involved in single-strand DNA breaks, which in the presence of defective homologous recombination repair lead to double-strand DNA breaks, the most lethal form of DNA damage. These agents therefore may be the drugs of choice for BRCA mutant breast and ovarian cancers. PARPi result in synergistic antitumor effects when combined with cisplatin, temozolomide, topoisomerase inhibitors and ionizing radiation. The indications for PARPi lie beyond BRCA mutations and may include genomic and functional defects in DNA repair and damage response pathways. Several PARPi are in the clinical development phase at this time and, given the recent failure of a phase III clinical trial of iniparib in triple-negative breast cancer, the identification of structural and functional differences between these inhibitors becomes critical. Acquired resistance to PARPi is being noted and represents an important limitation in this field. A concise review of the literature in this field is presented.

8.
Br J Cancer ; 105(8): 1114-22, 2011 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-21989215

RESUMO

Historically, PARP inhibitors (PARPi) were developed to potentiate the cytotoxic effect of certain chemotherapeutic agents and are currently being investigated in combination with chemotherapy in diverse cancer types. These agents are also radiosensitisers and clinical trials of PARPi with concurrent radiation are required. It has long been recognised that defective DNA repair pathways lead to tumour susceptibility. Recent studies indicate that tumour cells with defective homologous recombination (HR) repair pathways, the classic example being BRCA mutations, are exquisitely sensitive to PARPi. Defects in HR are not restricted to BRCA-associated tumours and other cancer types may be enriched for HR defects and hence susceptible to PARP inhibition. The identification of predictive markers for sensitivity to PARP inhibition is a priority area for research.


Assuntos
Reparo do DNA , Poli(ADP-Ribose) Polimerases/metabolismo , Antineoplásicos/uso terapêutico , Ensaios Clínicos como Assunto , Inibidores Enzimáticos/uso terapêutico , Humanos , Inibidores de Poli(ADP-Ribose) Polimerases , Radiossensibilizantes/uso terapêutico
9.
Br J Cancer ; 105(3): 372-81, 2011 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-21730979

RESUMO

BACKGROUND: The ataxia telangiectasia mutated and Rad3-related kinase (ATR) has a key role in the signalling of stalled replication forks and DNA damage to cell cycle checkpoints and DNA repair. It has long been recognised as an important target for cancer therapy but inhibitors have proved elusive. As NU6027, originally developed as a CDK2 inhibitor, potentiated cisplatin in a CDK2-independent manner we postulated that it may inhibit ATR. METHODS: Cellular ATR kinase activity was determined by CHK1 phosphorylation in human fibroblasts with inducible dominant-negative ATR-kinase dead expression and human breast cancer MCF7 cells. Cell cycle effects and chemo- and radiopotentiation by NU6027 were determined in MCF7 cells and the role of mismatch repair and p53 was determined in isogenically matched ovarian cancer A2780 cells. RESULTS: NU6027 is a potent inhibitor of cellular ATR activity (IC(50)=6.7 µM) and enhanced hydroxyurea and cisplatin cytotoxicity in an ATR-dependent manner. NU6027 attenuated G2/M arrest following DNA damage, inhibited RAD51 focus formation and increased the cytotoxicity of the major classes of DNA-damaging anticancer cytotoxic therapy but not the antimitotic, paclitaxel. In A2780 cells sensitisation to cisplatin was greatest in cells with functional p53 and mismatch repair (MMR) and sensitisation to temozolomide was greatest in p53 mutant cells with functional MMR. Importantly, NU6027 was synthetically lethal when DNA single-strand break repair is impaired either through poly(ADP-ribose) polymerase (PARP) inhibition or defects in XRCC1. CONCLUSION: NU6027 inhibits ATR, impairing G2/M arrest and homologous recombination thus increasing sensitivity to DNA-damaging agents and PARP inhibitors. It provides proof of concept data for clinical development of ATR inhibitors.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Proteínas de Ciclo Celular/antagonistas & inibidores , Compostos Nitrosos/uso terapêutico , Neoplasias Ovarianas/tratamento farmacológico , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Pirimidinas/uso terapêutico , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Neoplasias da Mama/genética , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Dano ao DNA/efeitos dos fármacos , Reparo de Erro de Pareamento de DNA/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Feminino , Genes p53 , Humanos , Leucemia L1210 , Camundongos , Neoplasias Ovarianas/genética , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/metabolismo
10.
Br J Cancer ; 103(10): 1588-96, 2010 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-20978505

RESUMO

BACKGROUND: Temozolomide shows activity against medulloblastoma, the most common malignant paediatric brain tumour. Poly(ADP-ribose) polymerase (PARP) inhibitors enhance temozolomide activity in extracranial adult and paediatric human malignancies. METHODS: We assessed the effect of AG-014699, a clinically active PARP inhibitor, on temozolomide-induced growth inhibition in human medulloblastoma models. Pharmacokinetic, pharmacodynamic and toxicity assays were performed in tumour-bearing mice. RESULTS: Sensitivity to temozolomide in vitro was consistent with methylguanine methyltransferase (MGMT) and DNA mismatch repair (MMR) status; MGMT(+) MMR(+) D384Med cells (temozolomide GI(50)=220 µM), representative of most primary medulloblastomas, were sensitised fourfold by AG-014699; MGMT⁻ MMR(+) D425Med cells were hypersensitive (GI(50)=9 µM) and not sensitised by AG-014699, whereas MGMT(+) MMR⁻ temozolomide-resistant D283Med cells (GI50=807 µM) were sensitised 20-fold. In xenograft models, co-administration of AG-014699 produced an increase in temozolomide-induced tumour growth delay in D384Med xenografts. Consistent with the in vitro data, temozolomide caused complete tumour regressions of D425Med xenografts, whereas D283Med xenografts were relatively resistant. AG-014699 was not toxic, accumulated and reduced PARP activity ≥75% in xenograft and brain tissues. CONCLUSION: We show for the first time central nervous system penetration and inhibition of brain PARP activity by AG-014699. Taken together with our in vitro chemosensitisation and toxicity data, these findings support further evaluation of the clinical potential of AG-014699-temozolomide combinations in intra-cranial malignancies.


Assuntos
Antineoplásicos Alquilantes/uso terapêutico , Neoplasias do Sistema Nervoso Central/patologia , Dacarbazina/análogos & derivados , Inibidores de Poli(ADP-Ribose) Polimerases , Animais , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Neoplasias do Sistema Nervoso Central/tratamento farmacológico , Neoplasias do Sistema Nervoso Central/enzimologia , Criança , Reparo de Erro de Pareamento de DNA/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Dacarbazina/uso terapêutico , Humanos , Indóis/uso terapêutico , Meduloblastoma/tratamento farmacológico , Meduloblastoma/enzimologia , Meduloblastoma/patologia , Camundongos , Camundongos Nus , Poli(ADP-Ribose) Polimerase-1 , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Temozolomida , Transplante Heterólogo
11.
Br J Cancer ; 102(2): 342-50, 2010 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-20010939

RESUMO

BACKGROUND: Cellular proliferation, driven by cyclin-dependent kinases (CDKs) and their cyclin partners, is deregulated in cancer. Anti-estrogens, such as tamoxifen, antagonise estrogen-induced ERalpha transactivation of cyclin D1, resulting in reduced CDK4/6 activity, p27(Kip1)-mediated inhibition of CDK2 and growth arrest. We hypothesised that direct inhibition of CDK2 and CDK1 may overcome the major clinical problem of anti-estrogen resistance. METHODS: The cellular effects of CDK2/1 siRNA knockdown and purine-based CDK2/1 inhibitors, NU2058 and NU6102, were measured in anti-estrogen-sensitive and resistant breast cancer cell lines. RESULTS: CDK2 knockdown caused G1 accumulation, whereas CDK1 depletion caused G2/M slowing, and dual CDK1/2 depletion resulted in further G2/M accumulation and cell death in both anti-estrogen-sensitive and resistant cells, confirming CDK2 and CDK1 as targets for breast cancer therapy. In contrast to tamoxifen, which only affected hormone-sensitive cells, NU2058 and NU6102 reduced CDK2-mediated phosphorylation of pRb, E2F transcriptional activity and proliferation, ultimately resulting in cell death, in both anti-estrogen-sensitive and resistant cells. Both drugs caused G2/M arrest, reflective of combined CDK2/1 knockdown, with a variable degree of G1 accumulation. CONCLUSION: These studies confirm the therapeutic potential of CDK2 and CDK1 inhibitors for cancer therapy, and support their use as an alternative treatment for endocrine-resistant breast cancer.


Assuntos
Antineoplásicos/farmacologia , Proteína Quinase CDC2/metabolismo , Proliferação de Células/efeitos dos fármacos , Quinase 2 Dependente de Ciclina/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Neoplasias da Mama , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Moduladores de Receptor Estrogênico/farmacologia , Feminino , Humanos
12.
Br J Cancer ; 101(2): 256-62, 2009 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-19568233

RESUMO

BACKGROUND: Poly(ADP-ribose) polymerase-1 (PARP-1) is a DNA-binding enzyme activated by DNA breaks and involved in DNA repair and other cellular processes. Poly(ADP-ribose) polymerase activity can be higher in cancer than in adjacent normal tissue, but cancer predisposition is reported to be greater in individuals with a single-nucleotide polymorphism (SNP) V762A (T2444C) in the catalytic domain that reduces PARP-1 activity. METHODS: To resolve these divergent observations, we determined PARP-1 polymorphisms, PARP-1 protein expression and activity in a panel of 19 solid and haematological, adult and paediatric human cancer cell lines. RESULTS: There was a wide variation in PARP activity in the cell line panel (coefficient of variation, CV=103%), with the lowest and the highest activity being 2460 pmol PAR/10(6) (HS-5 cells) and 85 750 pmol PAR/10(6) (NGP cells). Lower variation (CV=32%) was observed in PARP-1 protein expression with the lowest expression being 2.0 ng microg(-1) (HS-5 cells) and the highest being 7.1 ng microg(-1) (ML-1 cells). The mean activity in the cancer cells was 45-fold higher than the mean activity in normal human lymphocytes and the PARP-1 protein levels were 23-fold higher. CONCLUSIONS: Surprisingly, there was no significant correlation between PARP activity and PARP-1 protein level or the investigated polymorphisms, T2444C and CA.


Assuntos
Neoplasias/enzimologia , Neoplasias/genética , Poli(ADP-Ribose) Polimerases/genética , Linhagem Celular Tumoral , Humanos , Linfócitos/enzimologia , Repetições de Microssatélites , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/biossíntese , Poli(ADP-Ribose) Polimerases/metabolismo , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas
13.
Br J Radiol ; 81 Spec No 1: S6-11, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18820000

RESUMO

Poly(ADP-ribose) polymerase-1 (PARP-1) facilitates DNA single-strand break-base excision repair to maintain genomic stability. Inhibition or loss of PARP activity leads to a recombinogenic phenotype characterized by increased sister chromatid exchange. Deficiency in homologous recombination (HR) owing to loss of BRCA1 or BRCA2 is associated with hereditary cancers of the breast, ovary, pancreas and prostate. We investigated the therapeutic potential of PARP inhibitors in HR and BRCA2-defective cells. We exposed cells defective in the HR component XRCC3 (irs1SF) and BRCA2 (V-C8) and their parental (AA8, V79) or deficiency corrected (CXR3, V-C8+B2) cells to the PARP inhibitors NU1025 and AG14361. Mice bearing BRCA2-deficient and BRCA2-proficient tumours were treated with AG14361. All HR-defective cells were hypersensitive to normally non-cytotoxic concentrations of PARP inhibitors. Cells lacking BRCA2 were 20 times more sensitive to PARP inhibitor-induced cytotoxicity. Three out of five BRCA2-defective xenografts responded to the potent PARP inhibitor, AG14361, and one tumour regressed completely, compared with non-responses in the BRCA2-proficient tumours treated with AG14361 or any mice treated with vehicle control. Untreated PARP-1(-/-) mouse embryo fibroblasts (MEFs) accumulated more DNA double-strand breaks than did PARP-1(+/+) MEFs. We believe the underlying cytotoxic mechanism is due to PARP inhibitor-mediated suppression of repair of DNA single-strand breaks, which are converted to DNA double-strand breaks at replication. These replication-associated double-strand breaks, which are normally repaired by HR, become cytotoxic in cells defective in HR. Using a DNA repair inhibitor alone to selectively kill a tumour represents an exciting new concept in cancer therapy.


Assuntos
Proteína BRCA2/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases , Recombinação Genética/efeitos dos fármacos , Animais , Azulenos/uso terapêutico , Proteína BRCA2/deficiência , Benzodiazepinas/uso terapêutico , Quebras de DNA de Cadeia Dupla , Quebras de DNA de Cadeia Simples , Inibidores Enzimáticos/uso terapêutico , Feminino , Camundongos , Camundongos Nus , Neoplasias Experimentais/tratamento farmacológico , Quinazolinas/uso terapêutico
14.
Oncogene ; 26(55): 7611-9, 2007 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-17599054

RESUMO

Antiandrogens are initially effective in controlling prostate cancer (CaP), the second most common cancer in men, but resistance, associated with the loss of androgen-regulated cell cycle control, is a major problem. At present there is no effective treatment for androgen-independent prostate cancer (AIPC). Cellular proliferation is driven by cyclin-dependent kinases (CDKs) with kinase inhibitors (for example, p27) applying the breaks. We present the first investigation of the therapeutic potential of CDK inhibitors, using the guanine-based CDK inhibitor NU2058 (CDK2 IC(50)=17 microM, CDK1 IC(50)=26 microM), in comparison with the antiandrogen bicalutamide (Casodex) in AIPC cells. A panel of AIPC cells was found to be resistant to Casodex-induced growth inhibition, but with the exception of PC3 (GI(50)=38 microM) and CWR22Rv1 (GI(50)=46 microM) showed similar sensitivity to NU2058 (GI(50)=10-17 microM) compared to androgen-sensitive LNCaP cells (GI(50)=15 microM). In LNCaP cells and their Casodex-resistant derivative, LNCaP-cdxR, growth inhibition by NU2058 was accompanied by a concentration-dependent increase in p27 levels, reduced CDK2 activity and pRb phosphorylation, a decrease in early gene expression and G1 cell cycle phase arrest in both cell lines. In response to Casodex, there were similar observations in LNCaP cells (GI(50)=6+/-3 microM Casodex) but not in LNCaP-cdxR cells (GI(50)=24+/-5 microM Casodex).


Assuntos
Anilidas/uso terapêutico , Quinases Ciclina-Dependentes/antagonistas & inibidores , Guanina/análogos & derivados , Nitrilas/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Compostos de Tosil/uso terapêutico , Androgênios/metabolismo , Proteína Quinase CDC2/antagonistas & inibidores , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Quinase 2 Dependente de Ciclina/antagonistas & inibidores , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Guanina/uso terapêutico , Humanos , Masculino , Fosforilação/efeitos dos fármacos , Proteína do Retinoblastoma/metabolismo
15.
Transfus Med ; 17(3): 197-9, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17561862

RESUMO

Delayed haemolytic transfusion reactions (DHTRs) are relatively common following blood transfusions. In the UK, DHTRs were responsible for 10.2% of all serious transfusion-related hazards between 1996 and 2003. In most cases, there is minor or no morbidity, and the reaction may go unnoticed. However, in some cases, a large fall in haemoglobin level and major morbidity may occur. Even in such serious cases, the true cause of the patient's clinical deterioration may be unrecognized if serological investigation is not carried out immediately because of the often transient nature of the antibodies involved. Failure to make the correct diagnosis will lead to inappropriate treatment and expose the patient to risk of further serious transfusion reactions. We describe a case of DHTR caused by anti-Jk(b) antibodies, which illustrates the difficulty in diagnosing this common but under-recognized reaction and shows the transient nature of the antibody involved, which became undetectable within 4 weeks.


Assuntos
Incompatibilidade de Grupos Sanguíneos/imunologia , Hipersensibilidade Tardia/etiologia , Linfoma não Hodgkin/terapia , Reação Transfusional , Idoso , Antígenos de Grupos Sanguíneos/imunologia , Tipagem e Reações Cruzadas Sanguíneas/efeitos adversos , Tipagem e Reações Cruzadas Sanguíneas/métodos , Hemólise/fisiologia , Humanos , Masculino , Resultado do Tratamento
16.
Clin Lab Haematol ; 27(3): 206-8, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15938729

RESUMO

We present a 54-year-old man who underwent human leucocyte antigen-identical sibling nonmyeloablative peripheral blood stem cell transplant for primary refractory T-cell prolymphocytic leukaemia (T-PLL). His clinical course was complicated by fulminant haemolysis and acute renal failure at the time of engraftment because of minor ABO incompatibility between the donor and the recipient. This case highlights the curative potential of nonmyeloablative transplantation for T-PLL as well as the potential severity of immune haemolysis secondary to minor ABO incompatibility.


Assuntos
Sistema ABO de Grupos Sanguíneos/efeitos adversos , Injúria Renal Aguda/etiologia , Incompatibilidade de Grupos Sanguíneos/complicações , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Hemólise , Leucemia Prolinfocítica/complicações , Leucemia de Células T/complicações , Incompatibilidade de Grupos Sanguíneos/sangue , Transplante de Células-Tronco Hematopoéticas/métodos , Humanos , Leucemia Prolinfocítica/sangue , Leucemia Prolinfocítica/terapia , Leucemia de Células T/sangue , Leucemia de Células T/terapia , Masculino , Pessoa de Meia-Idade
18.
Br J Cancer ; 85(5): 764-71, 2001 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-11531265

RESUMO

ZD2767P is a phenol mustard glutamate prodrug which is currently being developed for Antibody Directed Enzyme Prodrug Therapy (ADEPT). In ZD2767 ADEPT an active bi-functional alkylating drug, ZD2767D (4-[N,N-bis(2-iodoethyl)amino]phenol), is generated following cleavage of ZD2767P by the bacterial enzyme carboxypeptidase G2 (CPG2) which is targeted to the tumour by conjugation to the F(ab')(2)fragment of the anti-CEA antibody A5B7. The aim of the studies described here was to identify the mode of cell death induced by ZD2767P + CPG2 in comparison to the established nitrogen mustard chlorambucil. The contribution of bifunctional and monofunctional ZD2767 DNA lesions to cell death induction was investigated using a monofunctional ZD2767D analogue. Apoptosis in LoVo tumour cells was studied by three different methods (nuclear morphology, annexin V staining and TUNEL). Levels of apoptosis detected using the three assays were similar, and each drug treatment produced apoptosis at levels above those in control cells at concentrations which resulted in tumour cell growth inhibition. The bi-functional compounds, ZD2767P + CPG2 and chlorambucil, induced apoptosis in a concentration and time dependent manner, with equitoxic concentrations producing equivalent levels of apoptosis. In contrast, the mono-functional ZD2767D analogue at 100 microM resulted in the maximal level of apoptosis at 25 h with no further increase over the following 72 h. These studies have demonstrated that apoptosis is the mechanism of cell death induced by the ZD2767 ADEPT system, and that levels of apoptosis produced by ZD2767 are similar to those observed at equitoxic concentrations of the classical nitrogen mustard chlorambucil. The mono-functional ZD2767 analogue also induced apoptosis, but with a different time course and characteristics. In conjunction with previous data, these studies have shown that the potent activity of ZD2767 can be attributed to the ability of the compound to induce bifunctional DNA lesions and engage apoptosis.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Apoptose , Clorambucila/farmacologia , DNA de Neoplasias/efeitos dos fármacos , Compostos de Mostarda Nitrogenada/farmacologia , Pró-Fármacos/farmacologia , gama-Glutamil Hidrolase/farmacologia , Dano ao DNA , Relação Dose-Resposta a Droga , Fatores de Tempo , Células Tumorais Cultivadas/efeitos dos fármacos
19.
Clin Cancer Res ; 7(7): 2105-13, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11448930

RESUMO

The activity of antimetabolite inhibitors of de novo deoxyribonucleotide biosynthesis can be compromised by the salvage of extracellular preformed nucleosides and nucleobases. Dipyridamole (DP) is a nucleoside transport inhibitor that has been used clinically in an attempt to increase antimetabolite activity; however, DP binds tightly to the serum protein alpha1-acid glycoprotein (AGP) thereby rendering this therapeutic strategy largely ineffective. Four novel DP analogues (NU3076, NU3084, NU3108, and NU3121) have been developed with substitutions at the 2,6- and 4,8-positions of the pyrimidopyrimidine ring. The novel DP analogues inhibit thymidine (dThd) uptake into L1210 cells in vitro (NU3076 IC(50), 0.25 microM; NU3084 IC(50), 0.27 microM; NU3108 IC(50), 0.31 microM; NU3121 IC(50), 0.26 microM; and DP IC(50), 0.37 microM), but, unlike DP, their activity remains largely unaffected in the presence of 5 mg/ml AGP. The four DP analogues inhibit dThd and hypoxanthine rescue from Alimta (multitargeted antifolate)-induced growth inhibition in A549 and COR L23 human lung carcinoma cell lines in the presence of 2.5 mg/ml AGP, whereas the activity of DP is completely abolished. i.p. administration of 10 mg/kg NU3108, NU3121, and DP produced peak plasma concentrations of 4.4, 2.1, and 6.7 microM, respectively, and levels were sustained above 1 microM for approximately 45 min (DP) and 120 min (NU3108 and NU3121). [3H]thymidine incorporation into COR L23 xenografts grown in CD1 nude mice was reduced by 64% (NU3108), 44% (NU3121), and 65% (DP) 2 h after administration of the nucleoside transport inhibitors. In conclusion, two novel DP analogues (NU3108 and NU3121) have been identified that do not bind to AGP and that display superior pharmacokinetic profiles in comparison to DP and inhibit [3H]thymidine incorporation into human tumor xenografts in vivo.


Assuntos
Proteínas de Transporte/antagonistas & inibidores , Dipiridamol/farmacologia , Antagonistas do Ácido Fólico/farmacologia , Guanina/análogos & derivados , Proteínas de Membrana/antagonistas & inibidores , Animais , Proteínas de Transporte/metabolismo , Divisão Celular/efeitos dos fármacos , Dipiridamol/química , Dipiridamol/farmacocinética , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Feminino , Glutamatos/farmacologia , Guanina/farmacologia , Humanos , Hipoxantina/farmacologia , Fígado/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Proteínas de Transporte de Nucleosídeos , Orosomucoide/farmacologia , Pemetrexede , Tetra-Hidrofolatos/farmacologia , Timidina/metabolismo , Fatores de Tempo , Trítio , Células Tumorais Cultivadas
20.
Clin Cancer Res ; 7(7): 2114-23, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11448931

RESUMO

The impact of p53 status on cellular sensitivity to antifolate drugs has been examined in seven human cell lines (A549, MCF7, T-47D, CCRF-CEM, COR-L23, A2780, and HCT-116) and p53 nonfunctional counterparts of two of the cell lines (HCT-116/N7 and A2780/CP70). p53 status was determined by sequencing and functional assays. The sensitivities of the cell lines to growth inhibition (sulphorhodamine B assay) produced by four antifolate drugs (Alimta, methotrexate, raltitrexed, and lometrexol) were studied. There was no clear relationship between functional p53 status and sensitivity to methotrexate or lometrexol, whereas a functional p53 status was possibly associated with resistance to Alimta- and raltitrexed-induced growth inhibition. In contrast, in the two pairs of related human tumor cell lines (HCT-116 and HCT-116/N7 and A2780 and A2780/CP70) cells with functional p53 were more sensitive to Alimta- and raltitrexed-induced growth inhibition (P = 0.002). Detailed studies were performed with the A2780 cell lines, and in the parental cells sensitivity to Alimta- and raltitrexed-induced cytotoxicity (clonogenic assay) was similar to the sensitivity determined in the sulphorhodamine B assay. However, in A2780/CP70 cells, 1 microM of drug resulted in only 40-60% growth inhibition yet > or = 85% cytotoxicity. After Alimta and raltitrexed exposure for < or = 72 h, there were no differences between the A2780 and A278/CP70 cell lines in cell cycle phase distribution, absolute cell number, or the induction of apoptosis. However, the cellular protein content of the A2780/CP70 cells was 3-6-fold higher than in A2780 cells after Alimta and raltitrexed treatment, suggesting that cells without functional p53 can maintain protein synthesis in the absence of cell division (unbalanced cell growth). In conclusion, the apparent impact of functional p53 status on sensitivity to antifolate drugs may depend upon the phenotypic/genotypic background as well as the assay used to measure cellular sensitivity.


Assuntos
Antagonistas do Ácido Fólico/farmacologia , Guanina/análogos & derivados , Proteína Supressora de Tumor p53/genética , Apoptose/efeitos dos fármacos , Western Blotting , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/genética , Ciclinas/metabolismo , Análise Mutacional de DNA , DNA de Neoplasias/química , DNA de Neoplasias/genética , Relação Dose-Resposta a Droga , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Genótipo , Glutamatos/farmacologia , Guanina/farmacologia , Humanos , Metotrexato/farmacologia , Mutação , Pemetrexede , Quinazolinas/farmacologia , Sensibilidade e Especificidade , Tetra-Hidrofolatos/farmacologia , Tiofenos/farmacologia , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...