Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
Cancer Res ; 47(15): 3911-9, 1987 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-3300957

RESUMO

The role of thymidine in the treatment of cancer has been under clinical investigation for nearly a decade. Clinical trials have demonstrated that it lacks antitumor activity in its own right. In this review, the mechanism of action and rationale for the use of thymidine as a biochemical modulator of standard agents such as 5-fluorouracil, 1-beta-D-arabinofuranosylcytosine, and methotrexate are summarized. With this background, the clinical trials which have been conducted with thymidine, either alone or in combination, are described. We suggest a number of further studies of the role of thymidine in the biochemical modulation of antimetabolites.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Neoplasias/tratamento farmacológico , Timidina/farmacologia , Animais , Antimetabólitos Antineoplásicos/administração & dosagem , Antimetabólitos Antineoplásicos/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Ácido Aspártico/administração & dosagem , Ácido Aspártico/análogos & derivados , Ácido Aspártico/farmacologia , Citarabina/administração & dosagem , Citarabina/metabolismo , Replicação do DNA/efeitos dos fármacos , DNA de Neoplasias/biossíntese , Desoxicitidina Quinase/antagonistas & inibidores , Avaliação de Medicamentos , Sinergismo Farmacológico , Fluoruracila/administração & dosagem , Fluoruracila/metabolismo , Antagonistas do Ácido Fólico , Humanos , Interfase/efeitos dos fármacos , Cinética , Leucemia/tratamento farmacológico , Metotrexato/administração & dosagem , Metotrexato/farmacologia , Camundongos , Neoplasias/metabolismo , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/metabolismo , Inibidores da Síntese de Ácido Nucleico , Nucleotídeos/metabolismo , Ácido Fosfonoacéticos/administração & dosagem , Ácido Fosfonoacéticos/análogos & derivados , Ácido Fosfonoacéticos/farmacologia , RNA Neoplásico/biossíntese , Especificidade da Espécie , Timidina/administração & dosagem , Timidina/efeitos adversos , Timidina/metabolismo
11.
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
12.
Biochem Pharmacol ; 70(3): 426-32, 2005 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-15950950

RESUMO

Continuous in vitro cultivation of human lymphoid H9 cells in the presence of 0.5microM arabinosyl-cytosine (araC) resulted in cell variant, H9-araC cells, that was >600-fold resistant to the drug and cross resistant to its analogs and other unrelated nucleosides, e.g. dideoxycytidine (5-fold), thiacytidine (2-fold), 2-fluoro-adenine arabinoside (8.3-fold), and 2-chloro-deoxyadenosine (2.1-fold). Compared to the parental cell line, the resistant cells accumulated <1% araCTP, and had reduced deoxycytidine kinase (dCK) activity (31.4%) and equilibrative nucleoside transporter 1 (ENT1) protein. The expression of the dCK gene in araC resistant cells was reduced to 60% of H9 cells, which correlated with lower dCK protein and activity. Whereas, there was no difference in the expression of ENT1 mRNA between the cell lines, ENT1 protein content was much lower in the resistant cells than in H9 cells. The concentrative nucleoside transporter (CNT3) was slightly increased in H9-araC cells, but CNT2, and MDR1 remained unaffected. Although a definitive correlation remains to be established, the amount of Sp1 protein, a transcription factor, that regulates the expressions of dCK, nucleoside transporters and other cellular proteins, was found reduced in H9-araC cells. Like ENT1, the Sp1 mRNA levels remained unaffected in H9-araC whereas protein contents were reduced. These observations are indicative of differences in the production and/or turnover of ENT1 and Sp1 proteins in H9-araC cells. Since nucleoside transporters and dCK play an important role in the activity of potential antiviral and anticancer deoxynucleoside analogs, understanding of their regulation is important. These studies show that the exposure of cells to araC, in vitro, is capable of simultaneously affecting more than one target site to confer resistance. The importance of this observation in the clinical use of araC remains to be determined.


Assuntos
Citarabina/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Linfócitos/efeitos dos fármacos , Linfócitos/metabolismo , Linhagem Celular , Desoxicitidina Quinase/antagonistas & inibidores , Desoxicitidina Quinase/genética , Desoxicitidina Quinase/metabolismo , Resistencia a Medicamentos Antineoplásicos/fisiologia , Transportador Equilibrativo 1 de Nucleosídeo/antagonistas & inibidores , Transportador Equilibrativo 1 de Nucleosídeo/biossíntese , Transportador Equilibrativo 1 de Nucleosídeo/genética , Transportador Equilibrativo 2 de Nucleosídeo/antagonistas & inibidores , Transportador Equilibrativo 2 de Nucleosídeo/genética , Humanos , Ligação Proteica/efeitos dos fármacos , RNA Mensageiro/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição Sp1/antagonistas & inibidores , Fator de Transcrição Sp1/metabolismo
13.
Gene ; 150(2): 351-4, 1994 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-7821805

RESUMO

In order to study the mutational inactivation of deoxycytidine kinase (Dck) in a rat model for acute myeloid leukemia we have cloned the complete coding region of the rat Dck gene. Using primers chosen from the human Dck cDNA sequence, we obtained a rat-specific probe via PCR and used it to isolate two clones from a rat lymphocyte cDNA library. The ORF showed 89.7 and 92.2% nucleotide identity with the human and mouse Dck, respectively, and encodes a 260-amino-acid protein, that is 91.9 and 94.6% homologous to human and mouse Dck, respectively. Northern blot analysis of rat tissues revealed high expression of a 4.1-kb Dck transcript in the thymus, whereas spleen, liver and lung samples showed weak expression of the gene. This tissue-specific expression pattern was confirmed by cDNA-PCR analysis.


Assuntos
Desoxicitidina Quinase/genética , Ratos/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , Sequência Conservada , Primers do DNA , DNA Complementar , Desoxicitidina Quinase/antagonistas & inibidores , Desoxicitidina Quinase/biossíntese , Humanos , Camundongos , Dados de Sequência Molecular , Fases de Leitura Aberta , Especificidade de Órgãos , Reação em Cadeia da Polimerase , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/biossíntese , Homologia de Sequência de Aminoácidos
14.
J Med Chem ; 26(12): 1691-6, 1983 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-6644738

RESUMO

Various new 5-substituted 3'-azido- and 3'-amino derivatives of 2'-deoxyuridine and 2'-deoxycytidine have been synthesized and biologically evaluated. Among these compounds, 3'-amino-2',3'-dideoxy-5-fluorouridine (3), 3'-amino-2',3'-dideoxycytidine (7a), and 3'-amino-2',3'-dideoxy-5-fluorocytidine (7c) were found to be the most active against murine L1210 and sarcoma 180 neoplastic cells in vitro, with an ED50 of 15 and 1 microM, 0.7 and 4 microM, and 10 and 1 microM, respectively. The 3'-azido derivatives, 2 and 6c, were less active in comparison with their 3'-amino counterparts. In addition, the 5-fluoro-3'-amino nucleosides, 3 and 7c, were tested against L1210 leukemia bearing CDF1 mice. Our preliminary findings indicate that compound 7c (6 X 200 mg/kg) was as active as the positive control, 5-fluorouracil (6 X 20 mg/kg), yielding a T/C X 100 of 146 and 129, respectively. However, 3 was found to be inactive in this experiment.


Assuntos
Antineoplásicos/síntese química , Desoxicitidina/análogos & derivados , Desoxiuridina/análogos & derivados , Animais , Desoxicitidina/síntese química , Desoxicitidina/uso terapêutico , Desoxicitidina Quinase/antagonistas & inibidores , Desoxiuridina/síntese química , Desoxiuridina/uso terapêutico , Leucemia L1210/tratamento farmacológico , Camundongos , Sarcoma 180/tratamento farmacológico , Timidina Quinase/antagonistas & inibidores
15.
Biochem Pharmacol ; 38(16): 2601-7, 1989 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-2548513

RESUMO

The kinetics and regulation of nucleoside phosphorylation by highly purified human deoxycytidine kinase from leukemic lymphoblasts were studied. The phosphorylation of purine nucleosides by this enzyme showed sensitivity to the endogenous inhibitors dCTP and UDP three times greater than the phosphorylation of dCyd. Examination of nucleotide pools in human T and B lymphoblasts disclosed that the levels of dCTP and UDP in these cells were sufficient to regulate kinase activity. The enhanced sensitivity of the kinase to dCTP and UDP was related to its reduced ability to interact with purine nucleosides. Comparison of the phosphorylation kinetics for pyrimidine and purine dideoxynucleosides used in antiviral therapy showed that the purine nucleosides were at least 50-fold less efficient as enzyme substrates. These results suggest that the phosphorylation of pharmacologically active purine nucleosides by deoxycytidine kinase is regulated by cellular nucleotide pools.


Assuntos
Desoxicitidina Quinase/antagonistas & inibidores , Nucleosídeos/fisiologia , Nucleotídeos/fisiologia , Fosfotransferases/antagonistas & inibidores , Nucleosídeos de Purina/metabolismo , Linfócitos T/metabolismo , Linfócitos B/metabolismo , Linhagem Celular , Desoxicitidina Quinase/fisiologia , Humanos , Fosforilação
16.
Biochem Pharmacol ; 50(2): 163-8, 1995 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-7632159

RESUMO

Human and mouse deoxycytidine kinase (dCK) (EC 2.7.1.74) were cloned and expressed in Escherichia coli. Michaelis-Menten kinetics were determined for the purified enzymes with 2'-deoxycytidine (dCyd), 2'-deoxyadenosine (dAdo), 2-chloro-2'-deoxyadenosine (CdA), 2',3'-dideoxycytidine (ddCyd) and 9-beta-D-arabinofuranosylguanine (araG) as substrates and ATP and UTP as phosphate donors. Both human and mouse dCK showed highest affinity to dCyd with Km values of 0.05-0.2 microM. The anti-leukaemic compound CdA was the superior substrate of the nucleoside analogues tested. Both enzymes were able to efficiently utilize ATP and UTP as phosphate donors. However, the use of UTP instead of ATP as phosphate donor decreased Km values for all substrates investigated. The kinetic properties of mouse and human dCK differed in that the human enzyme showed higher affinity for the substrates dAdo, CdA, ddCyd and araG. The human enzyme also showed higher affinity for ATP and UTP. The ability to phosphorylate dCyd was, however, similar for both human and mouse dCK. At physiological concentration of the feedback inhibitor dCTP, mouse dCK showed lower activity than human dCK for all substrates investigated.


Assuntos
Desoxicitidina Quinase/metabolismo , Trifosfato de Adenosina/metabolismo , Aminoácidos/análise , Animais , Clonagem Molecular , Desoxicitidina Quinase/antagonistas & inibidores , Desoxicitidina Quinase/química , Nucleotídeos de Desoxicitosina/farmacologia , Escherichia coli/enzimologia , Humanos , Cinética , Camundongos , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Uridina Trifosfato/metabolismo
17.
Biochem Pharmacol ; 43(2): 197-206, 1992 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-1739408

RESUMO

Deoxycytidine (dCyd) kinase has been purified to homogeneity from human leukemic spleen, and the capacity of the enzyme to phosphorylate 2',3'-dideoxynucleoside (ddN) analogs that are clinically effective inhibitors of human immunodeficiency virus (HIV) replication was evaluated. Cytosine-containing ddN analogs, such as 2',3'-dideoxycytidine, 2',3'-dideoxy-2',3'-dehydrocytidine, and cytallene, were efficiently phosphorylated by dCyd kinase, while no phosphorylation of purine-containing ddN analogs was detected. dCyd kinase was completely inactive toward 2',3'-dideoxyadenosine (ddAdo), 2',3'-dideoxyinosine, 2',3'-dideoxyguanosine, and adenallene, although it was capable of phosphorylating both 2'-deoxyadenosine (dAdo) and 2'-deoxyguanosine (dGuo). The abilities of wild type and mutant human T lymphoblastoid CEM cells to accumulate ddAdo in situ and in vitro were also ascertained. Comparison of the abilities of intact wild type CEM cells and derivatives deficient in nucleoside transport, dCyd kinase, and/or adenosine (Ado) kinase to accumulate [3H]ddAdo-derived radioactivity revealed no significant differences among the wild type and mutant strains. However, ddAdo phosphorylating activity was decreased in extracts from Ado kinase-deficient cells but not in lysates prepared from cells genetically deficient in dCyd kinase activity. In comparative growth rate experiments, wild type, nucleoside transport-deficient, and dCyd kinase-deficient CEM cells were equally sensitive to ddAdo toxicity, while, interestingly, a deficiency in Ado kinase correlated with a 5-fold decreased growth sensitivity to the purine ddN. Insertion of an adenine phosphoribosyltransferase deficiency into the CEM cell lines did not influence ddAdo toxicity or incorporation rate. These results imply that Ado kinase may be an important factor in ddAdo phosphorylation by CEM cells. Furthermore, these studies demonstrate that cytosine- and purine-containing ddNs are transported and activated by independent pathways and, therefore, have important implications for anti-HIV therapy in that pyrimidine and purine ddNs might be used in combination for the treatment of acquired immunodeficiency syndrome.


Assuntos
Antivirais/metabolismo , Desoxicitidina Quinase/antagonistas & inibidores , Didesoxinucleosídeos/metabolismo , Adenina Fosforribosiltransferase/deficiência , Linhagem Celular/efeitos dos fármacos , Linhagem Celular/metabolismo , Cromatografia Líquida de Alta Pressão , Desoxiadenosinas/metabolismo , Desoxicitidina/metabolismo , Desoxicitidina Quinase/isolamento & purificação , Didesoxiadenosina/metabolismo , Didesoxiadenosina/farmacologia , Didesoxinucleosídeos/isolamento & purificação , Humanos , Fosforilação , Especificidade por Substrato
18.
Leuk Res ; 27(5): 445-53, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12620296

RESUMO

AraC resistance in vitro is explained by inactivation of dCK, while resistance to DNR is described by overexpression of multidrug efflux pumps like Pgp or MRP. Thus far, no correlation between resistance mechanisms in vitro and in patients with AML has been documented. We generated AraC and DNR double resistant cell lines to investigate resistance mechanisms of both agents. In these cell lines involvement of dCK was extensively investigated and Pgp expression and activity was determined. Our data implicate that similar resistance mechanisms like inactivation of dCK coincided by alternatively spliced dCK forms and overexpression of Pgp are induced in single-as well as in double resistant leukemic cell lines.


Assuntos
2-Cloroadenosina/análogos & derivados , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/biossíntese , Azacitidina/análogos & derivados , Citarabina/farmacologia , Daunorrubicina/farmacologia , Desoxicitidina Quinase/antagonistas & inibidores , Resistência a Múltiplos Medicamentos/genética , Resistencia a Medicamentos Antineoplásicos/genética , Leucemia/patologia , Proteínas de Neoplasias/antagonistas & inibidores , 2-Cloroadenosina/farmacologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Processamento Alternativo , Animais , Azacitidina/farmacologia , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/genética , Butionina Sulfoximina/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Análise Mutacional de DNA , DNA de Neoplasias/genética , Decitabina , Desoxiadenosinas/farmacologia , Desoxicitidina Quinase/biossíntese , Desoxicitidina Quinase/genética , Glutationa/antagonistas & inibidores , Humanos , Idarubicina/farmacologia , Leucemia/genética , Metotrexato/farmacologia , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas/efeitos dos fármacos , Verapamil/farmacologia
19.
Adv Enzyme Regul ; 33: 39-59, 1993.
Artigo em Inglês | MEDLINE | ID: mdl-8356916

RESUMO

(1) Deoxycytidine kinase activity increased in a transformation- and progression-linked fashion in rat hepatomas of different proliferation rates. The activity also increased and was growth rate-linked in a series of tissue culture cell lines of human and animal tumors. (2) Deoxycytidine kinase activity was stringently linked with expression of the neoplastic proliferative program as it sharply increased in log phase in tissue culture cells of hepatoma 3924A and several human carcinoma strains. (3) Deoxycytidine kinase is subject to nutritional and hormonal regulation. On starvation the activity in liver decreased and on refeeding it returned to normal. Steroid hormone increased liver enzymic activity. Deoxycytidine kinase is substrate-inducible, since deoxycytidine injections in rat led to a 2- to 3-fold increase in hepatic enzyme activity. (4) Actinomycin or cycloheximide treatment blocked the increase in liver deoxycytidine kinase activity induced by steroid or deoxycytidine treatment. Therefore, it is assumed that the rise in deoxycytidine kinase activity requires new RNA and protein synthesis. (5) Cycloheximide treatment of rats carrying hepatomas yielded a t1/2 = 3.4 hr in the tumor for deoxycytidine kinase activity which was the shortest among the examined enzymes of purine and pyrimidine biosynthesis. (6) Actinomycin treatment of rats carrying hepatomas yielded a t1/2 of 5.8 hr for deoxycytidine kinase activity in the tumor which was one of the shortest in the examined enzymes of purine and pyrimidine biosynthesis. (7) Difluorodeoxycytidine (DFDC) is a competitive inhibitor (Ki = 7-28 microM) of deoxycytidine kinase from rat hepatoma and from human pancreatic carcinoma and ovarian carcinoma cells in culture.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Desoxicitidina Quinase/antagonistas & inibidores , Desoxicitidina Quinase/metabolismo , Desoxicitidina/análogos & derivados , Animais , Divisão Celular , Cicloeximida/farmacologia , Dactinomicina/farmacologia , Desoxicitidina/farmacologia , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/patologia , Nucleotídeos de Pirimidina/biossíntese , Ratos , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo , Células Tumorais Cultivadas/patologia , Gencitabina
20.
Oncol Res ; 4(11-12): 517-22, 1992.
Artigo em Inglês | MEDLINE | ID: mdl-1299379

RESUMO

The activity of deoxycytidine kinase (EC 2.7.1.74), an important pyrimidine salvage enzyme, was elevated 5- to 30-fold in human ovarian carcinoma and OVCAR-5 cells, in human colon carcinoma and HT-29 cells, in rat hepatoma 3924A solid tumors and cells, and in rat sarcoma as compared with the respective control normal cells. There was an inverse relationship between cell doubling time and deoxycytidine kinase activity in 8 cancer cell lines, with rapidly growing HL-60 cells (20 hr) showing the highest, and slower-growing lung H69 cells (60 hr) the smallest, increase in enzyme activity. In time-sequence studies in human HL-60, OVCAR-5, PANC-1, and rat hepatoma 3924A cells, there was a significant rise in deoxycytidine kinase activity after 3-6 hr of seeding, with peak increases (3.5- to 4-fold) at 48-72 hr in the log phase in comparison with values of the respective plateau phase cells (96-144 hr). In extracts of various cancer cells, the high deoxycytidine kinase activity was competitively inhibited by difluorodeoxycytidine (DFDC), with Ki = 7 to 30 microM. The Km for deoxycytidine in various carcinoma cell lines ranged from 0.3 to 0.7 mM and addition of DFDC increased the apparent Km from 0.7 to 4 mM. Deoxycytidine kinase activity in human HL-60 cells was inhibited by the end product, dCTP, with IC50 = 3 microM; dCTP elevated the Km for deoxycytidine from 0.35 to 0.9 mM. dTTP reversed the inhibition by dCTP.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Desoxicitidina Quinase/metabolismo , Desoxicitidina/análogos & derivados , Neoplasias/enzimologia , Animais , Divisão Celular , Desoxicitidina/farmacologia , Desoxicitidina Quinase/antagonistas & inibidores , Humanos , Técnicas In Vitro , Cinética , Neoplasias/patologia , Ratos , Células Tumorais Cultivadas , Gencitabina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA