Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Cell Death Dis ; 15(2): 131, 2024 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-38346958

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is considered one of the most lethal forms of cancer. Although in the last decade, an increase in 5-year patient survival has been observed, the mortality rate remains high. As a first-line treatment for PDAC, gemcitabine alone or in combination (gemcitabine plus paclitaxel) has been used; however, drug resistance to this regimen is a growing issue. In our previous study, we reported MYC/glutamine dependency as a therapeutic target in gemcitabine-resistant PDAC secondary to deoxycytidine kinase (DCK) inactivation. Moreover, enrichment of oxidative phosphorylation (OXPHOS)-associated genes was a common property shared by PDAC cell lines, and patient clinical samples coupled with low DCK expression was also demonstrated, which implicates DCK in cancer metabolism. In this article, we reveal that the expression of most genes encoding mitochondrial complexes is remarkably upregulated in PDAC patients with low DCK expression. The DCK-knockout (DCK KO) CFPAC-1 PDAC cell line model reiterated this observation. Particularly, OXPHOS was functionally enhanced in DCK KO cells as shown by a higher oxygen consumption rate and mitochondrial ATP production. Electron microscopic observations revealed abnormal mitochondrial morphology in DCK KO cells. Furthermore, DCK inactivation exhibited reactive oxygen species (ROS) reduction accompanied with ROS-scavenging gene activation, such as SOD1 and SOD2. SOD2 inhibition in DCK KO cells clearly induced cell growth suppression. In combination with increased anti-apoptotic gene BCL2 expression in DCK KO cells, we finally reveal that venetoclax and a mitochondrial complex I inhibitor are therapeutically efficacious for DCK-inactivated CFPAC-1 cells in in vitro and xenograft models. Hence, our work provides insight into inhibition of mitochondrial metabolism as a novel therapeutic approach to overcome DCK inactivation-mediated gemcitabine resistance in PDAC patient treatment.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Linhagem Celular Tumoral , Desoxicitidina/farmacologia , Desoxicitidina/uso terapêutico , Desoxicitidina Quinase/antagonistas & inibidores , Desoxicitidina Quinase/metabolismo , Resistencia a Medicamentos Antineoplásicos/genética , Gencitabina/farmacologia , Gencitabina/uso terapêutico , Paclitaxel/uso terapêutico , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Espécies Reativas de Oxigênio/metabolismo
2.
Biochem Pharmacol ; 172: 113742, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31812677

RESUMO

BACKGROUND: Deoxycytidine kinase (dCK) is an essential enzyme for production of nucleotides via the salvage pathway; DI-87 is a novel dCK inhibitor in preclinical development for use in anticancer therapy. The current study utilizes PET imaging to evaluate PK-PD relationships and to determine optimal dosing of the drug. METHODS: NSG mice bearing CEM tumors had plasma and tumor PK assessed using mass spectrometry following oral administration of DI-87. dCK inhibition was assessed after a single dose of oral DI-87 followed by a [18F]CFA PET probe and PET imaging. Tumor growth inhibition was assessed by orally administering DI-87 with concurrent intraperitoneal thymidine. RESULTS: DI-87 had an in vitro EC50 of 10.2 nM with low protein binding. Peak DI-87 concentrations were observed between 1-3 h and 3-9 h in plasma and tumor, respectively, with tumor concentrations less than one third of plasma. Full dCK inhibition, as evaluated by PET imaging, was observed as early as 3 h following 25 mg/kg dosing and was maintained for 12 h, with full recovery of enzyme activity after 36 h. When DI-87 was administered as repeated doses in combination with thymidine, full dCK inhibition was maintained at 12 h (25 mg/kg twice daily dose) and led to maximal tumor growth inhibition. CONCLUSIONS: DI-87 is a promising new compound for use in combination therapy against tumors expressing dCK. Utilizing a [18F]CFA PET probe targeting the pathway of interest allowed for efficient and accurate identification of the optimal dose for growth inhibition.


Assuntos
Antineoplásicos/farmacologia , Desoxicitidina Quinase/antagonistas & inibidores , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Quimioterapia Combinada , Humanos , Masculino , Camundongos , Estrutura Molecular , Neoplasias Experimentais
3.
Int J Mol Sci ; 17(11)2016 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-27879648

RESUMO

Deoxycytidine kinase (dCK) is a key enzyme in deoxyribonucleoside salvage and the anti-tumor activity for many nucleoside analogs. dCK is activated in response to ionizing radiation (IR)-induced DNA damage and it is phosphorylated on Serine 74 by the Ataxia-Telangiectasia Mutated (ATM) kinase in order to activate the cell cycle G2/M checkpoint. However, whether dCK plays a role in radiation-induced cell death is less clear. In this study, we genetically modified dCK expression by knocking down or expressing a WT (wild-type), S74A (abrogates phosphorylation) and S74E (mimics phosphorylation) of dCK. We found that dCK could decrease IR-induced total cell death and apoptosis. Moreover, dCK increased IR-induced autophagy and dCK-S74 is required for it. Western blotting showed that the ratio of phospho-Akt/Akt, phospho-mTOR/mTOR, phospho-P70S6K/P70S6K significantly decreased in dCK-WT and dCK-S74E cells than that in dCK-S74A cells following IR treatment. Reciprocal experiment by co-immunoprecipitation showed that mTOR can interact with wild-type dCK. IR increased polyploidy and decreased G2/M arrest in dCK knock-down cells as compared with control cells. Taken together, phosphorylated and activated dCK can inhibit IR-induced cell death including apoptosis and mitotic catastrophe, and promote IR-induced autophagy through PI3K/Akt/mTOR pathway.


Assuntos
Apoptose/efeitos da radiação , Desoxicitidina Quinase/genética , Regulação Neoplásica da Expressão Gênica , Fosfatidilinositol 3-Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética , Serina-Treonina Quinases TOR/genética , Substituição de Aminoácidos , Apoptose/genética , Proteínas Mutadas de Ataxia Telangiectasia/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Autofagia/genética , Autofagia/efeitos da radiação , Desoxicitidina Quinase/antagonistas & inibidores , Desoxicitidina Quinase/metabolismo , Pontos de Checagem da Fase G2 do Ciclo Celular/genética , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos da radiação , Raios gama , Células HeLa , Humanos , Mimetismo Molecular , Mutação , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos da radiação , Ligação Proteica/efeitos da radiação , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/genética , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo
4.
Mol Cancer Ther ; 15(6): 1271-8, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27196770

RESUMO

Inhibition of both the de novo (DNP) and salvage (NSP) pathways of nucleoside synthesis has been demonstrated to impair leukemia cells. We endeavored to determine whether this approach would be efficacious in glioblastoma. To diminish nucleoside biosynthesis, we utilized compound DI-39, which selectively targets NSP, in combination with thymidine (dT), which selectively targets DNP. We employed in vitro and ex vivo models to determine the effects of pretreatment with dT + DI-39 on brain tumor stem cells (BTSC). Here, we demonstrate that this combinatorial therapy elicits a differential response across a spectrum of human patient-derived glioblastoma cultures. As determined by apoptotic markers, most cultures were relatively resistant to treatment, although a subset was highly sensitive. Sensitivity was unrelated to S-phase delay and to DNA damage induced by treatment. Bioinformatics analysis indicated that response across cultures was associated with the transcription factor PAX3 (associated with resistance) and with canonical pathways, including the nucleotide excision repair pathway, PTEN (associated with resistance), PI3K/AKT (associated with sensitivity), and ErbB2-ErbB3. Our in vitro assays demonstrated that, in sensitive cultures, clonal sphere formation was reduced upon removal from pretreatment. In contrast, in a resistant culture, clonal sphere formation was slightly increased upon removal from pretreatment. Moreover, in an intracranial xenograft model, pretreatment of a sensitive culture caused significantly smaller and fewer tumors. In a resistant culture, tumors were equivalent irrespective of pretreatment. These results indicate that, in the subset of sensitive glioblastoma, BTSCs are targeted by inhibition of pyrimidine synthesis. Mol Cancer Ther; 15(6); 1271-8. ©2016 AACR.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Desoxicitidina Quinase/antagonistas & inibidores , Inibidores Enzimáticos/administração & dosagem , Glioblastoma/tratamento farmacológico , Células-Tronco Neoplásicas/efeitos dos fármacos , Pirimidinas/administração & dosagem , Sulfonamidas/administração & dosagem , Timidina/administração & dosagem , Animais , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Inibidores Enzimáticos/farmacologia , Glioblastoma/metabolismo , Humanos , Camundongos , Fator de Transcrição PAX3/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Pirimidinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Sulfonamidas/farmacologia , Timidina/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Proc Natl Acad Sci U S A ; 113(15): 4027-32, 2016 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-27035974

RESUMO

Deoxycytidine kinase (dCK), a rate-limiting enzyme in the cytosolic deoxyribonucleoside (dN) salvage pathway, is an important therapeutic and positron emission tomography (PET) imaging target in cancer. PET probes for dCK have been developed and are effective in mice but have suboptimal specificity and sensitivity in humans. To identify a more suitable probe for clinical dCK PET imaging, we compared the selectivity of two candidate compounds-[(18)F]Clofarabine; 2-chloro-2'-deoxy-2'-[(18)F]fluoro-9-ß-d-arabinofuranosyl-adenine ([(18)F]CFA) and 2'-deoxy-2'-[(18)F]fluoro-9-ß-d-arabinofuranosyl-guanine ([(18)F]F-AraG)-for dCK and deoxyguanosine kinase (dGK), a dCK-related mitochondrial enzyme. We demonstrate that, in the tracer concentration range used for PET imaging, [(18)F]CFA is primarily a substrate for dCK, with minimal cross-reactivity. In contrast, [(18)F]F-AraG is a better substrate for dGK than for dCK. [(18)F]CFA accumulation in leukemia cells correlated with dCK expression and was abrogated by treatment with a dCK inhibitor. Although [(18)F]CFA uptake was reduced by deoxycytidine (dC) competition, this inhibition required high dC concentrations present in murine, but not human, plasma. Expression of cytidine deaminase, a dC-catabolizing enzyme, in leukemia cells both in cell culture and in mice reduced the competition between dC and [(18)F]CFA, leading to increased dCK-dependent probe accumulation. First-in-human, to our knowledge, [(18)F]CFA PET/CT studies showed probe accumulation in tissues with high dCK expression: e.g., hematopoietic bone marrow and secondary lymphoid organs. The selectivity of [(18)F]CFA for dCK and its favorable biodistribution in humans justify further studies to validate [(18)F]CFA PET as a new cancer biomarker for treatment stratification and monitoring.


Assuntos
Nucleotídeos de Adenina/química , Arabinonucleosídeos/química , Biomarcadores Tumorais/química , Desoxicitidina Quinase/análise , Desoxicitidina Quinase/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Animais , Antineoplásicos/química , Linhagem Celular Tumoral , Clofarabina , Meios de Contraste/química , Desoxicitidina Quinase/antagonistas & inibidores , Humanos , Leucemia/enzimologia , Camundongos , Neoplasias/tratamento farmacológico , Pró-Fármacos/química , Ratos
6.
J Med Chem ; 57(22): 9480-94, 2014 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-25341194

RESUMO

Recently, we have shown that small molecule dCK inhibitors in combination with pharmacological perturbations of de novo dNTP biosynthetic pathways could eliminate acute lymphoblastic leukemia cells in animal models. However, our previous lead compound had a short half-life in vivo. Therefore, we set out to develop dCK inhibitors with favorable pharmacokinetic properties. We delineated the sites of the inhibitor for modification, guided by crystal structures of dCK in complex with the lead compound and with derivatives. Crystal structure of the complex between dCK and the racemic mixture of our new lead compound indicated that the R-isomer is responsible for kinase inhibition. This was corroborated by kinetic analysis of the purified enantiomers, which showed that the R-isomer has >60-fold higher affinity than the S-isomer for dCK. This new lead compound has significantly improved metabolic stability, making it a prime candidate for dCK-inhibitor based therapies against hematological malignancies and, potentially, other cancers.


Assuntos
Desoxicitidina Quinase/antagonistas & inibidores , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Inibidores de Proteínas Quinases/química , Animais , Antineoplásicos/química , Sítios de Ligação , Química Farmacêutica/métodos , Simulação por Computador , Cristalografia por Raios X , Desoxicitidina/análogos & derivados , Desenho de Fármacos , Feminino , Humanos , Concentração Inibidora 50 , Camundongos , Camundongos Endogâmicos C57BL , Microssomos/metabolismo , Fosforilação , Tomografia por Emissão de Pósitrons , Estereoisomerismo , Tiazóis/química
8.
J Exp Med ; 211(3): 473-86, 2014 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-24567448

RESUMO

Pharmacological targeting of metabolic processes in cancer must overcome redundancy in biosynthetic pathways. Deoxycytidine (dC) triphosphate (dCTP) can be produced both by the de novo pathway (DNP) and by the nucleoside salvage pathway (NSP). However, the role of the NSP in dCTP production and DNA synthesis in cancer cells is currently not well understood. We show that acute lymphoblastic leukemia (ALL) cells avoid lethal replication stress after thymidine (dT)-induced inhibition of DNP dCTP synthesis by switching to NSP-mediated dCTP production. The metabolic switch in dCTP production triggered by DNP inhibition is accompanied by NSP up-regulation and can be prevented using DI-39, a new high-affinity small-molecule inhibitor of the NSP rate-limiting enzyme dC kinase (dCK). Positron emission tomography (PET) imaging was useful for following both the duration and degree of dCK inhibition by DI-39 treatment in vivo, thus providing a companion pharmacodynamic biomarker. Pharmacological co-targeting of the DNP with dT and the NSP with DI-39 was efficacious against ALL models in mice, without detectable host toxicity. These findings advance our understanding of nucleotide metabolism in leukemic cells, and identify dCTP biosynthesis as a potential new therapeutic target for metabolic interventions in ALL and possibly other hematological malignancies.


Assuntos
Vias Biossintéticas/fisiologia , Desoxicitidina Quinase/antagonistas & inibidores , Nucleotídeos de Desoxicitosina/biossíntese , Erradicação de Doenças/métodos , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Animais , Vias Biossintéticas/efeitos dos fármacos , Nucleotídeos de Desoxicitosina/metabolismo , Camundongos , Tomografia por Emissão de Pósitrons , Timidina/farmacologia
9.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 1): 68-78, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24419380

RESUMO

Deoxycytidine kinase (dCK) is a key enzyme in the nucleoside salvage pathway that is also required for the activation of several anticancer and antiviral nucleoside analog prodrugs. Additionally, dCK has been implicated in immune disorders and has been found to be overexpressed in several cancers. To allow the probing and modulation of dCK activity, a new class of small-molecule inhibitors of the enzyme were developed. Here, the structural characterization of four of these inhibitors in complex with human dCK is presented. The structures reveal that the compounds occupy the nucleoside-binding site and bind to the open form of dCK. Surprisingly, a slight variation in the nature of the substituent at the 5-position of the thiazole ring governs whether the active site of the enzyme is occupied by one or two inhibitor molecules. Moreover, this substituent plays a critical role in determining the affinity, improving it from >700 to 1.5 nM in the best binder. These structures lay the groundwork for future modifications that would result in even tighter binding and the correct placement of moieties that confer favorable pharmacodynamics and pharmacokinetic properties.


Assuntos
Desoxicitidina Quinase/antagonistas & inibidores , Desoxicitidina Quinase/química , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Cristalografia por Raios X , Humanos , Modelos Moleculares , Ligação Proteica , Conformação Proteica/efeitos dos fármacos , Difosfato de Uridina/metabolismo
10.
J Med Chem ; 56(17): 6696-708, 2013 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-23947754

RESUMO

Combined inhibition of ribonucleotide reductase and deoxycytidine kinase (dCK) in multiple cancer cell lines depletes deoxycytidine triphosphate pools leading to DNA replication stress, cell cycle arrest, and apoptosis. Evidence implicating dCK in cancer cell proliferation and survival stimulated our interest in developing small molecule dCK inhibitors. Following a high throughput screen of a diverse chemical library, a structure-activity relationship study was carried out. Positron Emission Tomography (PET) using (18)F-L-1-(2'-deoxy-2'-FluoroArabinofuranosyl) Cytosine ((18)F-L-FAC), a dCK-specific substrate, was used to rapidly rank lead compounds based on their ability to inhibit dCK activity in vivo. Evaluation of a subset of the most potent compounds in cell culture (IC50 = ∼1-12 nM) using the (18)F-L-FAC PET pharmacodynamic assay identified compounds demonstrating superior in vivo efficacy.


Assuntos
Desoxicitidina Quinase/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Tomografia por Emissão de Pósitrons/métodos , Linhagem Celular Tumoral , Cristalografia por Raios X , Humanos , Concentração Inibidora 50 , Cinética , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Método de Monte Carlo , Espectrometria de Massas por Ionização por Electrospray
11.
Biochem Pharmacol ; 84(1): 43-51, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22490700

RESUMO

Deoxycytidine kinase (dCK) (EC 2.7.1.74) is a key enzyme in the activation of several therapeutic nucleoside analogs (NA). Its activity can be increased in vivo by Ser-74 phosphorylation, a property that could be used for enhancing NA activation and clinical efficacy. In line with this, studies with recombinant dCK showed that mimicking Ser-74 phosphorylation by a S74E mutation increases its activity toward pyrimidine analogs. However, purine analogs had not been investigated. Here, we show that the S74E mutation increased the k(cat) for cladribine (CdA) by 8- or 3-fold, depending on whether the phosphoryl donor was ATP or UTP, for clofarabine (CAFdA) by about 2-fold with both ATP and UTP, and for fludarabine (F-Ara-A) by 2-fold, but only with UTP. However, the catalytic efficiencies (k(cat)/Km) were not, or slightly, increased. The S74E mutation also sensitized dCK to feed-back inhibition by dCTP, regardless of the phosphoryl donor. Importantly, we did not observe an increase of endogenous dCK activity toward purine analogs after in vivo-induced increase of Ser-74 phosphorylation. Accordingly, treatment of CLL cells with aphidicolin, which enhances dCK activity through Ser-74 phosphorylation, did not modify the conversion of CdA or F-Ara-A into their active triphosphate form. Nevertheless, the same treatment enhanced activation of gemcitabine (dFdC) into dFdCTP in CLL as well as in HCT-116 cells and produced synergistic cytotoxicity. We conclude that increasing phosphorylation of dCK on Ser-74 might constitute a valuable strategy to enhance the clinical efficacy of some NA, like dFdC, but not of CdA or F-Ara-A.


Assuntos
Antineoplásicos/metabolismo , Desoxicitidina Quinase/metabolismo , Nucleosídeos de Purina/metabolismo , Nucleosídeos de Pirimidina/metabolismo , Serina/metabolismo , Antineoplásicos/química , Antineoplásicos/farmacologia , Afidicolina/farmacologia , Biotransformação , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cladribina/química , Cladribina/metabolismo , Cladribina/farmacologia , Desoxicitidina/análogos & derivados , Desoxicitidina/química , Desoxicitidina/metabolismo , Desoxicitidina/farmacologia , Desoxicitidina Quinase/antagonistas & inibidores , Desoxicitidina Quinase/genética , Ativação Enzimática , Células HCT116 , Células HT29 , Humanos , Cinética , Mutação , Fosforilação , Nucleosídeos de Purina/química , Nucleosídeos de Purina/farmacologia , Nucleosídeos de Pirimidina/química , Nucleosídeos de Pirimidina/farmacologia , Serina/genética , Relação Estrutura-Atividade , Especificidade por Substrato , Vidarabina/análogos & derivados , Vidarabina/química , Vidarabina/metabolismo , Vidarabina/farmacologia , Gencitabina
12.
Curr Med Chem ; 19(7): 1076-87, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22257063

RESUMO

Gemcitabine (dFdC, 2',2'-difluorodeoxycytidine) is a deoxycytidine nucleoside analogue of deoxycytidine in which two fluorine atoms have been inserted into the deoxyribose ring. Like other nucleoside analogues, gemcitabine is a prodrug. It is inactive in its original form, and depends on the intracellular machinery to gain pharmacological activity. What makes gemcitabine different from other nucleoside analogues is that it is actively transported across the cell membrane, it is phosphorylated more efficiently and it is eliminated at a slower rate. These differences, together with self-potentiation mechanisms, masked DNA chain termination and extensive inhibitory efficiency against several enzymes, are the source of gemcitabine's cytotoxic activity against a wide variety of tumors. This unique combination of metabolic properties and mechanistic characteristics is only found in very few other anticancer drugs, and both the FDA and the EMEA have already approved its use for clinical purposes, for the treatment of several types of tumors. In spite of the promising results associated with gemcitabine, the knowledge of its mode of action and of the enzymes it interacts with is still not fully documented. In this article we propose to review all these aspects and summarize the path of gemcitabine inside the cell.


Assuntos
Antimetabólitos Antineoplásicos/uso terapêutico , Desoxicitidina/análogos & derivados , Neoplasias/tratamento farmacológico , Domínio Catalítico , Linhagem Celular Tumoral , Desoxicitidina/uso terapêutico , Desoxicitidina Quinase/antagonistas & inibidores , Desoxicitidina Quinase/química , Humanos , Modelos Moleculares , Nucleosídeos/química , Nucleosídeos/uso terapêutico , Fosforilação , Gencitabina
13.
Oncol Rep ; 23(2): 471-5, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20043109

RESUMO

The prognosis of pancreatic cancer remains poor, and the standard first-line chemotherapy with gemcitabine (GEM) has a response rate of less than 20%. Since expression of deoxycytidine kinase (dCK) seems important for improvement of GEM sensitivity, overexpression of dCK was investigated using pancreatic cancer cell lines (Panc-1, MIAPaCa-2 and BxPC-3). dCK gene was introduced into the cell lines by retrovirus and changes in IC50 were examined. Sensitivity of two pancreatic cancer cell lines to GEM elevated dramatically in comparison with control cells, but change of sensitivity remained at 1.8 times in BxPC-3. Since addition of tetrahydro uridine (THU), an inhibitor of deoxycytidine deaminase (CDA), increased the sensitivity 54-fold, overexpression of CDA seems to be the mechanism for improvement of the sensitivity. In conclusion, dCK is a key enzyme of GEM, but resistance of GEM is not improved in all pancreatic cancer cells by overexpression of dCK. Combination treatment based on expression of GEM metabolism-related gene may become an effective therapy in the future.


Assuntos
Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/genética , Desoxicitidina Quinase/genética , Desoxicitidina/análogos & derivados , Resistencia a Medicamentos Antineoplásicos/genética , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Antimetabólitos Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Citidina Desaminase , Desoxicitidina/uso terapêutico , Desoxicitidina Quinase/antagonistas & inibidores , Relação Dose-Resposta a Droga , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/fisiologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Modelos Biológicos , Nucleosídeo Desaminases/genética , Nucleosídeo Desaminases/fisiologia , Tetra-Hidrouridina/farmacologia , Transfecção , Regulação para Cima/fisiologia , Gencitabina
14.
J Biomol Screen ; 15(1): 72-9, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19959816

RESUMO

Deoxycytidine kinase (dCK) phosphorylates deoxycytidine, deoxyguanosine, and deoxyadenosine and plays an important role in the salvage pathway of nucleoside metabolism. dCK is also required for the phosphorylation of several antiviral and anticancer nucleoside drugs, with resistance to these agents often being associated with a loss or decrease in dCK activity. Data also indicate a role for dCK in immune function, and dCK inhibitors may provide treatment for immune disorders. To identify novel dCK inhibitors, the authors evaluated 2 existing biochemical assays, adapted both to high-throughput screening, and identified several series of hits. They also compared the potency of the hits between purified recombinant and endogenous enzyme. Meanwhile, they also developed a novel cell-based assay that rests on the rescue of cells from dCK-dependent cytotoxic agents such as AraC. A large number of compounds were tested using the 3 assays, and a strong correlation in potency was observed between the biochemical assay using endogenous enzyme and the cell-based assay. The hits identified in these screens have proved to be good starting points for the synthesis of much more potent tool compounds to further investigate the physiological functions of dCK and potentially lead to the development of therapeutic agents.


Assuntos
Desoxicitidina Quinase/antagonistas & inibidores , Ensaios de Triagem em Larga Escala/métodos , Inibidores de Proteínas Quinases/análise , Inibidores de Proteínas Quinases/farmacologia , Morte Celular/efeitos dos fármacos , Humanos , Medições Luminescentes , Proteínas Recombinantes/antagonistas & inibidores , Fatores de Tempo
15.
Bioorg Med Chem Lett ; 19(23): 6780-3, 2009 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-19836229

RESUMO

A series of potent piperidine-linked cytosine derivatives were prepared as inhibitors of deoxycytidine kinase (dCK). Compound 9h was discovered to be a potent inhibitor of dCK and shows a good combination of cellular potency and pharmacokinetic parameters. Compound 9h blocks the incorporation of radiolabeled cytosine into mouse T-cells in vitro, as well as in vivo in mice following a T-cell challenge.


Assuntos
Desoxicitidina Quinase/antagonistas & inibidores , Flucitosina/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Animais , Desenho de Fármacos , Flucitosina/síntese química , Flucitosina/química , Humanos , Camundongos , Estrutura Molecular , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/química , Estereoisomerismo , Relação Estrutura-Atividade
16.
Bioorg Med Chem Lett ; 19(23): 6784-7, 2009 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-19836232

RESUMO

A series of deoxycytidine kinase inhibitors was simultaneously optimized for potency and PK properties. A co-crystal structure then allowed merging this series with a high throughput screening hit to afford a highly potent, selective and orally bioavailable inhibitor, compound 10. This compound showed dose dependent inhibition of deoxycytidine kinase in vivo.


Assuntos
Desoxicitidina Quinase/antagonistas & inibidores , Desoxicitidina/análogos & derivados , Desenho de Fármacos , Inibidores de Proteínas Quinases/farmacologia , Desoxicitidina/síntese química , Desoxicitidina/química , Desoxicitidina/farmacologia , Relação Dose-Resposta a Droga , Modelos Moleculares , Estrutura Molecular , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/química , Estereoisomerismo , Relação Estrutura-Atividade
17.
Anticancer Res ; 28(4B): 2205-12, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18751396

RESUMO

BACKGROUND: The functional roles of deoxycytidine kinase (dCK) in acquired resistance to gemcitabine remain unknown in pancreatic cancer. Here, the functional involvement of dCK in gemcitabine-resistance of pancreatic cancer was investigated. MATERIALS AND METHODS: The levels of the dCK gene as well as other gemcitabine-related genes (hENT1, RRM1 and RRM2) were analyzed in gemcitabine-resistant pancreatic cancer cells (GR cells) using quantitative real-time reverse transcription polymerase chain reaction. The effects of inhibition of these genes on sensitivity to gemcitabine were evaluated. RESULTS: In GR cells, expression of dCK was significantly reduced compared with that of parental cells (p < 0.05). The dCK-targeting siRNA significantly reduced gemcitabine sensitivity (p < 0.01) without affecting cell proliferation. The RRM1- and RRM2-targeting siRNAs increased gemcitabine sensitivity (p < 0.05) and reduced cell proliferation even without gemcitabine treatment. The hENT-targeting siRNA did not affect gemcitabine sensitivity or cell proliferation. CONCLUSION: Down-regulation of dCK specifically enhanced acquired resistance to gemcitabine in pancreatic cancer cells without affecting their proliferation.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Desoxicitidina Quinase/biossíntese , Desoxicitidina/análogos & derivados , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/enzimologia , Antimetabólitos Antineoplásicos/farmacocinética , Linhagem Celular Tumoral , Desoxicitidina/farmacocinética , Desoxicitidina/farmacologia , Desoxicitidina Quinase/antagonistas & inibidores , Desoxicitidina Quinase/genética , Regulação para Baixo , Resistencia a Medicamentos Antineoplásicos , Transportador Equilibrativo 1 de Nucleosídeo/antagonistas & inibidores , Transportador Equilibrativo 1 de Nucleosídeo/biossíntese , Transportador Equilibrativo 1 de Nucleosídeo/genética , Humanos , Neoplasias Pancreáticas/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ribonucleosídeo Difosfato Redutase/antagonistas & inibidores , Ribonucleosídeo Difosfato Redutase/biossíntese , Ribonucleosídeo Difosfato Redutase/genética , Proteínas Supressoras de Tumor/antagonistas & inibidores , Proteínas Supressoras de Tumor/biossíntese , Proteínas Supressoras de Tumor/genética , Gencitabina
18.
Nucleosides Nucleotides Nucleic Acids ; 27(6): 816-20, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18600545

RESUMO

Thymocytes lacking adenosine deaminase (ADA) activity, a purine metabolism enzyme, accumulate intracellular dATP and consequently undergo apoptosis during development. We have analyzed the effect of ADA enzyme inhibition in human thymocyte suspension cultures with regard to accumulation of intracellular dATP and induction of apoptosis. We demonstrate that while inhibition of deoxycytidine kinase will prevent the accumulation of dATP and induction of apoptosis to a large degree, inhibition of both deoxycytidine kinase and adenosine kinase completely abrogates the accumulation of dATP and significantly reduces the induction of apoptosis. Thus, both deoxynucleoside kinases are involved in this model of ADA deficiency.


Assuntos
Adenosina Quinase/antagonistas & inibidores , Apoptose/efeitos dos fármacos , Nucleotídeos de Desoxiadenina/metabolismo , Desoxicitidina Quinase/antagonistas & inibidores , Células Precursoras de Linfócitos T/citologia , Células Precursoras de Linfócitos T/metabolismo , Timo/citologia , Adenosina Desaminase/deficiência , Adenosina Desaminase/metabolismo , Inibidores de Adenosina Desaminase , Antígenos CD34/metabolismo , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Humanos , Lactente , Células Precursoras de Linfócitos T/efeitos dos fármacos , Timo/efeitos dos fármacos , Timo/crescimento & desenvolvimento
19.
Hematol Oncol Clin North Am ; 20(5): 1087-97, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16990109

RESUMO

Cladribine is effective therapy for HCL, and there are several ways to achieve the adequate concentrations of the active metabolites in relevant cells, without the need for long-term continuous infusions. This simplifies therapy, although careful control of patients is required during and after treatment in most instances because of the significant activity of the drug on leukemia cells of various types and also on lymphoid cells and normal stem cells.


Assuntos
Adenosina/análogos & derivados , Adenosina/farmacocinética , Antimetabólitos Antineoplásicos/farmacocinética , Leucemia de Células Pilosas/tratamento farmacológico , Adenosina/história , Adenosina/uso terapêutico , Adenosina Desaminase/metabolismo , Inibidores de Adenosina Desaminase , Antimetabólitos Antineoplásicos/história , Antimetabólitos Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Desoxicitidina Quinase/antagonistas & inibidores , Desoxicitidina Quinase/metabolismo , História do Século XX , História do Século XXI , Humanos , Leucemia de Células Pilosas/enzimologia , Leucemia de Células Pilosas/história , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/metabolismo
20.
Br J Cancer ; 95(3): 289-97, 2006 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-16868547

RESUMO

The present study was performed to investigate the capability of gemcitabine and pemetrexed to synergistically interact with respect to cytotoxicity and apoptosis in T24 and J82 bladder cancer cells, and to establish a correlation between drug activity and gene expression of selected genes in tumour samples. The interaction between gemcitabine and pemetrexed was synergistic; indeed, pemetrexed favoured gemcitabine cytotoxicity by increasing cellular population in S-phase, reducing Akt phosphorylation as well as by inducing the expression of a major gemcitabine uptake system, the human equilibrative nucleoside transporter-1 (hENT1), and the key activating enzyme deoxycytidine kinase (dCK) in both cell lines. Bladder tumour specimens showed an heterogeneous gene expression pattern and patients with higher levels of dCK and hENT1 had better response. Moreover, human nucleoside concentrative transporter-1 was detectable only in 3/12 patients, two of whom presented a complete response to gemcitabine. These data provide evidence that the chemotherapeutic activity of the combination of gemcitabine and pemetrexed is synergistic against bladder cancer cells in vitro and that the assessment of the expression of genes involved in gemcitabine uptake and activation might be a possible determinant of bladder cancer response and may represent a new tool for treatment optimization.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Desoxicitidina/análogos & derivados , Glutamatos/administração & dosagem , Guanina/análogos & derivados , Neoplasias da Bexiga Urinária/tratamento farmacológico , Neoplasias da Bexiga Urinária/metabolismo , Administração Intravesical , Adulto , Idoso , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Desoxicitidina/administração & dosagem , Desoxicitidina/antagonistas & inibidores , Desoxicitidina/farmacologia , Desoxicitidina Quinase/antagonistas & inibidores , Desoxicitidina Quinase/genética , Dipiridamol/farmacologia , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Transportador Equilibrativo 1 de Nucleosídeo/efeitos dos fármacos , Transportador Equilibrativo 1 de Nucleosídeo/genética , Antagonistas de Aminoácidos Excitatórios , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glutamatos/farmacologia , Guanina/administração & dosagem , Guanina/antagonistas & inibidores , Guanina/farmacologia , Humanos , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Pemetrexede , Fosforribosilglicinamido Formiltransferase/antagonistas & inibidores , Fosforribosilglicinamido Formiltransferase/genética , Fosforilação , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fase S/efeitos dos fármacos , Tioinosina/análogos & derivados , Tioinosina/farmacologia , Timidilato Sintase/antagonistas & inibidores , Timidilato Sintase/genética , Resultado do Tratamento , Gencitabina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA