Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Carcinogenesis ; 42(7): 940-950, 2021 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-33640964

RESUMEN

The 5-year survival rate of pancreatic ductal carcinoma (PDAC) patients is <10% despite progress in clinical medicine. Strategies to prevent the development of PDAC are urgently required. The flavonoids Luteolin (Lut) and hesperetin (Hes) may be cancer-chemopreventive, but effects on pancreatic carcinogenesis in vivo have not been studied. Here, the chemopreventive effects of Lut and Hes on pancreatic carcinogenesis are assessed in the BOP-induced hamster PDAC model. Lut but not Hes suppressed proliferation of pancreatic intraepithelial neoplasia (PanIN) and reduced the incidence and multiplicity of PDAC in this model. Lut also inhibited the proliferation of hamster and human pancreatic cancer cells in vitro. Multi-blot and microarray assays revealed decreased phosphorylated STAT3 (pSTAT3) and dihydropyrimidine dehydrogenase (DPYD) on Lut exposure. To explore the relationship between DPYD and STAT3 activity, the former was silenced by RNAi or overexpressed using expression vectors, and the latter was inactivated by small molecule inhibitors or stimulated by IL6 in human PDAC cells. DPYD knock-down decreased, and overexpression increased, pSTAT3 and cell proliferation. DPYD expression was decreased by inactivation of STAT3 and increased by its activation. The frequency of pSTAT3-positive cells and DPYD expression was significantly correlated and was decreased in parallel by Lut in the hamster PDAC model. Finally, immunohistochemical analysis in 73 cases of human PDAC demonstrated that DPYD expression was positively correlated with the Ki-67 labeling index, and high expression was associated with poor prognosis. These results indicate that Lut is a promising chemopreventive agent for PDAC, targeting a novel STAT3-DPYD pathway.


Asunto(s)
Carcinoma Ductal Pancreático/tratamiento farmacológico , Dihidrouracilo Deshidrogenasa (NADP)/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Luteolina/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Factor de Transcripción STAT3/metabolismo , Anciano , Animales , Apoptosis , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Proliferación Celular , Cricetinae , Femenino , Humanos , Masculino , Ratones , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Pronóstico , Factor de Transcripción STAT3/genética , Tasa de Supervivencia , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Cancer Chemother Pharmacol ; 84(6): 1157-1166, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31482228

RESUMEN

Cancer caused by fundamental defects in cell cycle regulation leads to uncontrolled growth of cells. In spite of the treatment with chemotherapeutic agents of varying nature, multiple resistance mechanisms are identified in cancer cells. Similarly, numerous variations, which decrease the metabolism of chemotherapeutics agents and thereby increasing the toxicity of anticancer drugs have been identified. 5-Fluorouracil (5-FU) is an anticancer drug widely used to treat many cancers in the human body. Its broad targeting range is based upon its capacity to act as a uracil analogue, thereby disrupting RNA and DNA synthesis. Dihydropyrimidine dehydrogenase (DPD) is an enzyme majorly involved in the metabolism of pyrimidines in the human body and has the same metabolising effect on 5-FU, a pyrimidine analogue. Multiple mutations in the DPD gene have been linked to 5-FU toxicity and inadequate dosages. DPD inhibitors have also been used to inhibit excessive degradation of 5-FU for meeting appropriate dosage requirements. This article focusses on the role of dihydropyrimidine dehydrogenase in the metabolism of the anticancer drug 5-FU and other associated drugs.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Dihidrouracilo Deshidrogenasa (NADP)/metabolismo , Inhibidores Enzimáticos/farmacología , Fluorouracilo/farmacología , Neoplasias/tratamiento farmacológico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Dihidrouracilo Deshidrogenasa (NADP)/antagonistas & inhibidores , Dihidrouracilo Deshidrogenasa (NADP)/genética , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Inhibidores Enzimáticos/uso terapéutico , Fluorouracilo/uso terapéutico , Humanos , Mutación , Neoplasias/genética , Resultado del Tratamiento
4.
Biomed Pharmacother ; 105: 53-65, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29843045

RESUMEN

Despite continuous research on chemotherapeutic agents, different mechanisms of resistance have become a major pitfall in cancer chemotherapy. Although, exhaustive efforts are being made by several researchers to target resistance against chemotherapeutic agents, there is another class of resistance mechanism which is almost carrying on unattended. This class of resistance includes pharmacokinetics resistance such as efflux by ABC transporters and drug metabolizing enzymes. ABC transporters are the membrane bound proteins which are responsible for the movement of substrates through the cell membrane. Drug metabolizing enzymes are an integral part of phase-II metabolism that helps in the detoxification of exogenous, endogenous and xenobiotics substrates. These include uridine diphospho-glucuronosyltransferases (UGTs), glutathione-S-transferases (GSTs), dihydropyrimidine dehydrogenases (DPDs) and thiopurine methyltransferases (TPMTs). These enzymes may affect the role of drugs in both positive as well negative manner, depending upon the type of tissue and cells present and when present in tumors, can result in drug resistance. However, the underlying mechanism of resistance by drug metabolizing enzymes is still not clear. Here, we have tried to cover various aspects of these enzymes in relation to anticancer drugs.


Asunto(s)
Antineoplásicos/farmacocinética , Dihidrouracilo Deshidrogenasa (NADP)/antagonistas & inhibidores , Resistencia a Antineoplásicos/efectos de los fármacos , Glucuronosiltransferasa/antagonistas & inhibidores , Glutatión Transferasa/antagonistas & inhibidores , Metiltransferasas/antagonistas & inhibidores , Antineoplásicos/metabolismo , Dihidrouracilo Deshidrogenasa (NADP)/genética , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Glucuronosiltransferasa/genética , Glutatión Transferasa/genética , Humanos , Metiltransferasas/genética , Modelos Moleculares , Estructura Molecular , Conformación Proteica
5.
Drug Des Devel Ther ; 11: 1693-1705, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28652707

RESUMEN

To reduce 5-fluorouracil (5-FU)-induced serious toxicities without loss of antitumor activity, we have developed DFP-11207, a novel fluoropyrimidine, which consists of 1-ethoxymethyl-5-fluorouracil (EM-FU; a precursor form of 5-FU), 5-chloro-2,4-dihydroxypyridine (CDHP; an inhibitor of 5-FU degradation), and citrazinic acid (CTA; an inhibitor of 5-FU phosphorylation). In vitro studies of DFP-11207 indicated that it strongly inhibited the degradation of 5-FU by dihydropyrimidine dehydrogenase (DPD) in homogenates of the rat liver, and also inhibited the phosphorylation of 5-FU by orotate phosphoribosyltransferase (OPRT) in tumor tissues in a similar magnitude of potency by CDHP and CTA, respectively. Especially, DFP-11207 inhibited the intracellular phosphorylation of 5-FU in tumor cells in a dose-dependent manner whereas CTA alone did not protect intracellular 5-FU phosphorylation. These results postulate that DFP-11207 rapidly entered into the cell and the free CTA produced from DFP-11207 inhibited the phosphorylation of 5-FU in the cell. Furthermore, following oral administration of DFP-11207, CTA was found to be highly retained in the gastrointestinal (GI) tract compared to other tissues in rats. Interestingly, EM-FU, the prodrug of 5-FU was found to specifically produce 5-FU by various species of liver microsomes. When DFP-11207 was administered to rats, the plasma level of 5-FU was persisted for a long-time with lower Cmax and longer half-life than that from other 5-FU prodrugs. The antitumor activity of DFP-11207 was evaluated in human tumor xenografts in nude rats and found that DFP-11207 showed an antitumor activity in a dose-dependent fashion and its efficacy is equivalent to reference 5-FU drugs. In striking contrast, DFP-11207 manifested no or less 5-FU-related toxicities, such as a decrease in body weights, GI injury, and myelosuppression, especially thrombocytopenia. Taken together, the preclinical evaluation of DFP-11207 strongly indicates that DFP-11207 be a potential new version of the oral fluoropyrimidine prodrug for further clinical development.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Diseño de Fármacos , Fluorouracilo/farmacología , Hidrocarburos Fluorados/farmacología , Profármacos/farmacología , Pirimidinas/farmacología , Activación Metabólica , Administración Oral , Animales , Antimetabolitos Antineoplásicos/administración & dosificación , Antimetabolitos Antineoplásicos/farmacocinética , Antimetabolitos Antineoplásicos/toxicidad , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Dihidrouracilo Deshidrogenasa (NADP)/antagonistas & inhibidores , Dihidrouracilo Deshidrogenasa (NADP)/metabolismo , Relación Dosis-Respuesta a Droga , Estabilidad de Medicamentos , Fluorouracilo/administración & dosificación , Fluorouracilo/farmacocinética , Fluorouracilo/toxicidad , Células HT29 , Humanos , Hidrocarburos Fluorados/administración & dosificación , Hidrocarburos Fluorados/farmacocinética , Hidrocarburos Fluorados/toxicidad , Hidrólisis , Hígado/efectos de los fármacos , Hígado/metabolismo , Orotato Fosforribosiltransferasa/antagonistas & inhibidores , Orotato Fosforribosiltransferasa/metabolismo , Fosforilación , Profármacos/administración & dosificación , Profármacos/farmacocinética , Profármacos/toxicidad , Pirimidinas/administración & dosificación , Pirimidinas/farmacocinética , Pirimidinas/toxicidad , Ratas Endogámicas F344 , Ratas Desnudas , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Expert Opin Drug Deliv ; 14(12): 1355-1366, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28379040

RESUMEN

INTRODUCTION: Sixty years since its introduction, 5-FU still forms the core of chemotherapy regimens for many types of malignancies. 5-FU is a time-dependent drug but is rapidly degraded in plasma by dihydropyrimidine dehydrogenase (DPD). Although originally developed in an intravenous form, 5-FU oral prodrugs were developed with the goal of improving efficacy and minimizing toxicity as well as to capitalize on the advantages of oral drug administration. The inactive 5-FU prodrug is gradually converted into the active form in the systemic circulation. UFT, S-1, and capecitabine are oral 5-FU prodrugs currently in clinical use. However, the efficacy of 5-FU can be further improved by its combination with DPD inhibitors and biochemical modulators, such as uracil and leucovorin, in addition to modifying administration schedules. Areas covered: We focused on the drug delivery of oral 5-FU prodrugs, their pharmacokinetics, and the development of DPD inhibitors. Since oral 5-FU prodrugs have been formulated into combination drugs, we also discussed the regulatory approval of combination drugs. Expert opinion: Many regimens that include intravenously administered 5-FU can be replaced by oral 5-FU prodrugs. Patients would benefit from development of combination 5-FU oral prodrug formulations and its associated path through the combination drug regulatory approval process.


Asunto(s)
Antimetabolitos Antineoplásicos/administración & dosificación , Antineoplásicos/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Fluorouracilo/administración & dosificación , Neoplasias/tratamiento farmacológico , Administración Intravenosa , Administración Oral , Antimetabolitos Antineoplásicos/uso terapéutico , Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Dihidrouracilo Deshidrogenasa (NADP)/antagonistas & inhibidores , Inhibidores Enzimáticos/administración & dosificación , Fluorouracilo/uso terapéutico , Humanos , Profármacos/administración & dosificación , Pirimidinas
7.
Curr Pharm Des ; 23(14): 2028-2034, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28128059

RESUMEN

BACKGROUND: Fluoropyrimidines are widely used in the treatment of solid tumors and remain the backbone of many combination chemotherapy regimens. Despite their clinical benefit, they are associated with frequent gastrointestinal and hematological toxicities, which often lead to treatment discontinuation. Fluoropyrimidines undergo complex anabolic and catabolic biotransformation. Enzymes involved in this pathway include dihydropyrimidine dehydrogenase (DPD), which breaks down 5-FU and its prodrugs. Candidate gene approaches have demonstrated associations between 5-FU treatment outcomes and germline polymorphisms in DPD. The aim of this review is to report and discuss the latest results on fluoropyrimidine pharmacogenetics. METHODS: Literature from PubMed databases and bibliography from retrieved publications have been analyzed according to terms such DPD, DPYD, fluoropyrimdines, polymorphisms, toxicity, pharmacogenetics. RESULTS: To date, many sequence variations have been identified within DPYD gene, although the majority of these have no functional consequences on enzymatic activity. Nowadays, there is a general agreement on the clinical significance of the importance of DPD deficiency in patients who suffer from severe, life-threatening drug toxicity although preemptive testing is not applied to all patients. CONCLUSION: Considering the published literature, clinicians are strongly encouraged to consider testing for DPD poor metabolizer variants as a rational pre-treatment screening for patients candidate to a fluoropyrimidine-based regimens, in order to prevent toxicities and personalise treatments.


Asunto(s)
Dihidrouracilo Deshidrogenasa (NADP)/genética , Neoplasias/tratamiento farmacológico , Farmacogenética , Antimetabolitos Antineoplásicos/efectos adversos , Antimetabolitos Antineoplásicos/metabolismo , Antimetabolitos Antineoplásicos/farmacología , Dihidrouracilo Deshidrogenasa (NADP)/antagonistas & inhibidores , Dihidrouracilo Deshidrogenasa (NADP)/metabolismo , Fluorouracilo/efectos adversos , Fluorouracilo/metabolismo , Fluorouracilo/farmacología , Humanos , Neoplasias/metabolismo
8.
J Enzyme Inhib Med Chem ; 31(6): 1335-41, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26758717

RESUMEN

Dihydropyrimidine dehydrogenase (DPD, E.C. 1.3.1.2) was purified from sheep liver with a yield of 16.7%, purification fold of 407.5 and specific activity of 0.705 EU/mg proteins. The purification procedure consisted of ammonium sulphate fractionation, DEAE ion exchange chromatography and 2',5'-ADP Sepharose-4B affinity chromatography. The molecular weight determined by SDS-PAGE and was found 111 kDa. Optimum pH, ionic strength temperature and stable pH were determined as 8.0, 0.9 mM, 50 °C and 6.0, respectively. The kinetic parameters (Km and Vmax) of the enzyme were determined with NADPH as 22.97 µM and 0.17 EU/mL, respectively. The same parameters were determined with uracil as 17.46 µM and 0.14 EU/mL, respectively. Additionally, in vitro inhibitory effects of some antidepressant drugs including escitalopram, fluoxetine, mirtazapine, haloperidol and some anaesthetic drugs including propofol and lidocaine were investigated against DPD. In addition, IC50 values for each active drug obtained for escitalopram, fluoxetine, mirtazapine, haloperidol, propofol and lidocaine were determined as 1736.11, 13.24, 86.65, 99.03, 0.21 and 15.07 µM, respectively.


Asunto(s)
Anestésicos/farmacología , Antidepresivos/farmacología , Dihidrouracilo Deshidrogenasa (NADP)/aislamiento & purificación , Hígado/enzimología , Animales , Dihidrouracilo Deshidrogenasa (NADP)/antagonistas & inhibidores , Dihidrouracilo Deshidrogenasa (NADP)/metabolismo , Electroforesis en Gel de Poliacrilamida , Cinética , Ovinos
9.
Life Sci ; 144: 138-47, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26656468

RESUMEN

AIMS: Some compounds derived from Chinese medicine have demonstrated great prospective roles in sensitization to chemotherapy. This study aimed to investigate the combination of luteolin and 5-fluorouracil on proliferations of hepatocellular carcinoma cells and the potential mechanisms. MAIN METHODS: The antitumor effects of luteolin, 5-fluorouracil, and their combinations were detected by 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium/phenazine methosulfate assay, and isobole method was used to evaluate drug combinations. CellTiter-Blue and Caspase-Glo 3/7 assay were used for assessment of cell viability and apoptosis after treatment with luteolin, 5-fluorouracil and their combinations. Cell cycle distributions and apoptosis were detected by PI staining, Hoechst 33342 staining and FITC-Annexin V/PI staining. Bcl-2, bax, p53 and PARP expressions were determined by Western blot. Furthermore, mRNA levels of 5-fluorouracil metabolism related enzymes were detected by RT-PCR. KEY FINDINGS: Drug combination study showed that luteolin could synergize the antitumor effects of 5-fluorouracil at different dose ratios (luteolin: 5-fluorouracil=10:1, 20:1, 40:1) against HepG2 and Bel7402 cells. Cell viability and cell apoptosis analysis showed that the synergistic growth inhibition caused by combined luteolin and 5-fluorouracil was closely related to apoptosis. Further mechanism studies showed that the synergistic effects of drug combinations were related with enhanced bax/bcl-2 ratios and p53 expressions, and induced PARP cleavage. Also, combined luteolin and 5-fluorouracil could significantly decrease the dihydropyrimidine dehydrogenase. SIGNIFICANCE: These results showed that luteolin could synergize the antitumor effects of 5-fluorouracil on HepG2 and Bel7402 cells, which might be related with apoptosis and regulation of 5-fluorouracil metabolism.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Fluorouracilo/farmacología , Neoplasias Hepáticas Experimentales/tratamiento farmacológico , Neoplasias Hepáticas Experimentales/metabolismo , Luteolina/farmacología , Animales , Proteínas Reguladoras de la Apoptosis/biosíntesis , Proteínas Reguladoras de la Apoptosis/genética , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Dihidrouracilo Deshidrogenasa (NADP)/antagonistas & inhibidores , Sinergismo Farmacológico , Humanos , Neoplasias Hepáticas Experimentales/patología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Estudios Prospectivos , ARN Mensajero/biosíntesis , ARN Mensajero/genética
10.
Int J Mol Sci ; 16(10): 23668-82, 2015 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-26457704

RESUMEN

MiR-302b is a member of miR-302-367 cluster. The miR-302-367 cluster played important roles in maintaining pluripotency in human embryonic stem cells (hESCs) and has been proved to be capable of suppressing cell growth in several types of cancer cell lines including Hepatocellular Carcinoma (HCC) Cell lines. However, the role that miR-302b plays in the 5-Fluorouracil (5-FU) sensitivity of HCC has not been known. This study showed that miR-302b could enhance the sensitivity to 5-FU in HCC cell lines and verified its two putative targeted genes responsible for its 5-FU sensitivity.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma Hepatocelular/tratamiento farmacológico , Dihidrouracilo Deshidrogenasa (NADP)/antagonistas & inhibidores , Fluorouracilo/uso terapéutico , Neoplasias Hepáticas/tratamiento farmacológico , MicroARNs/metabolismo , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/antagonistas & inhibidores , Línea Celular Tumoral , Terapia Combinada , Humanos
11.
Int J Oncol ; 46(3): 989-98, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25573239

RESUMEN

Although cytotoxic chemotherapy is essential in epidermal growth factor receptor (EGFR)­mutated non­small cell lung cancer (NSCLC), it is unclear which regimen is most effective. We retrospectively compared the efficacy of standard platinum­based chemotherapy with that of combination chemotherapy using vinorelbine (VNR) plus dihydropyrimidine dehydrogenase­inhibitory fluoropyrimidine (DIF) in EGFR­mutated lung adenocarcinomas, and we investigated a potential mechanism by which the combination chemotherapy of VNR + DIF was favorable in the treatment of EGFR­mutated lung adenocarcinoma in vitro. In our retrospective analysis, the response rate and disease control rate afforded by the VNR + DIF treatment tended to be better than those by platinum­based chemotherapy, and the progression­free survival of the 24 VNR + DIF­treated patients was significantly longer than that of the 15 platinum­based chemotherapy patients. In EGFR­mutated PC9 cells, VNR induced EGFR dephosphorylation at a clinically achievable concentration. 1BR3­LR cells, a line of fibroblast cells transfected with a mutant EGFR construct, were completely resistant to gefitinib in the medium containing 10% fetal bovine serum (FBS), whereas the sensitivity of these cells to gefitinib was increased in 0.5% FBS­containing medium. Similarly, the sensitivity of 1BR3­LR cells to VNR was increased when they were cultured in low­serum condition. In addition, sodium orthovanadate (Na3VO4) inhibited the EGFR dephosphorylation induced by VNR or gefitinib and suppressed the cell growth inhibition by these agents in PC9 cells. VNR and gefitinib showed synergistic cell growth inhibition in combination with 5­fluorouracil (5­FU) in PC9 cells. We propose that the EGFR dephosphorylation induced by VNR is related to cell growth inhibitory activity of VNR, and that this is one of the mechanisms of the synergistic effect of VNR + 5­FU in EGFR­mutated lung cancer cells. In conclusion, the combination chemotherapy of VNR + DIF may be a promising treatment for NSCLC patients with EGFR mutations.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Receptores ErbB/genética , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Adenocarcinoma/mortalidad , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral/efectos de los fármacos , Dihidrouracilo Deshidrogenasa (NADP)/antagonistas & inhibidores , Combinación de Medicamentos , Receptores ErbB/metabolismo , Femenino , Fluorouracilo/administración & dosificación , Gefitinib , Humanos , Estimación de Kaplan-Meier , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Mutación , Ácido Oxónico/administración & dosificación , Quinazolinas/farmacología , Estudios Retrospectivos , Tegafur/administración & dosificación , Resultado del Tratamiento , Vinblastina/administración & dosificación , Vinblastina/análogos & derivados , Vinblastina/farmacología , Vinorelbina
12.
J Oral Pathol Med ; 44(3): 201-7, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25169655

RESUMEN

BACKGROUND: Cancer stem cells (CSCs) are involved in both tumourigenesis and in tumour recurrence after therapy. In head and neck squamous cell carcinoma (HNSCC), there are two biologically different CSC phenotypes both of which express high levels of CD44 but differ in their expression levels of epithelial-specific antigen (ESA). One phenotype is CD44(high)/ESA(high) and has epithelial features (Epi-CSCs), while the other is CD44(high) /ESA(low), has undergone epithelial to mesenchymal transition (EMT-CSCs), has mesenchymal features and is migratory (Biddle et al., 2011). CSCs are resistant to therapeutically induced apoptosis but the molecular mechanisms by which they develop apoptotic resistance remains unclear. However, glycogen synthase kinase 3ß (GSK3ß) contributes to regulation of both the self-renewal and switching of these two CSC phenotypes (Shigeishi et al., 2013). METHODS: CD44(high) /ESA(low), CD44(high) /ESA(high) and CD44(low) cells were FACS sorted from the HNSCC cell line LUC4, and 5-FU-induced apoptosis was analysed by Annexin V staining followed by flow cytometry analysis. RESULTS: CD44(high) /ESA(low) cells exhibited marked resistance to 5-FU-induced apoptosis and had high expression of dihydropyrimidine dehydrogenase (DPD). The DPD inhibitor, 5-chloro-2, 4-dihydroxypyridine (CDHP) significantly enhanced 5-FU-induced apoptosis of CD44(high)/ESA(low) cells. Inhibition of GSK3ß induced CD44(high) /ESA(low) cells to undergo mesenchymal-to-epithelial transition (MET) to CD44(high)/ESA(high) cells and pre-existing CD44(high) /ESA(high) cells to differentiate. Apoptosis induced by 5-FU was thus facilitated. Combination of both CDHP and GSK3ß inhibitors markedly enhanced 5-FU-induced apoptosis of CD44(high) /ESA(low) cells. CONCLUSIONS: Our results suggest potentially new approaches for the elimination of the therapy resistant HNSCC CSC population.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Fluorouracilo/farmacología , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Neoplasias de Cabeza y Cuello/patología , Células Madre Neoplásicas/efectos de los fármacos , Piridinas/farmacología , Antígenos de Neoplasias/análisis , Biomarcadores de Tumor/análisis , Complejo CD3/análisis , Moléculas de Adhesión Celular/análisis , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Separación Celular/métodos , Dihidrouracilo Deshidrogenasa (NADP)/análisis , Dihidrouracilo Deshidrogenasa (NADP)/antagonistas & inhibidores , Resistencia a Antineoplásicos/efectos de los fármacos , Molécula de Adhesión Celular Epitelial , Transición Epitelial-Mesenquimal/efectos de los fármacos , Epitelio/patología , Citometría de Flujo/métodos , Glucógeno Sintasa Quinasa 3 beta , Humanos , Receptores de Hialuranos/análisis , Células Madre Neoplásicas/patología
13.
Cent Nerv Syst Agents Med Chem ; 13(2): 108-13, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23597150

RESUMEN

Xanthine Oxidase (XO; EC. 1.1.3.22) and Dihydropyrimidine Dehydrogenase (DPD; EC. 1.3.1.2) are two enzymes responsible for the last steps of purine and pyrimidine catabolism, and hydroxylation of a wide variety of pyrimidine, pterin, and aldehyde substrates. Elion showed that purine isomers can be converted to various nucleotides, which influence pyrimidine metabolism (Elion, 1978). The current study is devoted to delineating the correlation between survival of human brain derived cells in culture and the activities of XO and DPD. Cultivation of (E90) brain cells was performed by the modified method of Mattson (1990). XO activity was measured by the formation of uric acid in the tissue. DPD activity was evaluated by the reduction of NADPH and the associated absorbance decrease at 320 nm. Cell death was detected by Trypan Blue dye leakage. During our investigation, we noticed a reversed correlation between the activities of XO and DPD over 12 days under normal conditions as well as in the presence of the XO and DPD inhibitors, allopurinol and dipyridamole. During the treatment period of 12 days, as well as from days 7-12 with the inhibitors, we observed cell protection, whereas treatment from days 1-7 elevated the percentage of dead cells in culture. A low dosage of allopurinol over 12 days also stimulated cell growth and increased their number in culture. We concluded that timely inhibition of XO as well as DPD activities might initiate cell growth and prevent their death. However, the main influence as the final enzyme of purine metabolism in the processes of cell proliferation belongs to XO in contrast to DPD.


Asunto(s)
Encéfalo/enzimología , Dihidrouracilo Deshidrogenasa (NADP)/fisiología , Proteínas del Tejido Nervioso/fisiología , Xantina Oxidasa/fisiología , Alopurinol/farmacología , Encéfalo/citología , Encéfalo/embriología , Supervivencia Celular , Células Cultivadas , Dihidrouracilo Deshidrogenasa (NADP)/antagonistas & inhibidores , Dipiridamol/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Cultivo Primario de Células , Purinas/metabolismo , Xantina Oxidasa/antagonistas & inhibidores
15.
Gan To Kagaku Ryoho ; 39(4): 667-70, 2012 Apr.
Artículo en Japonés | MEDLINE | ID: mdl-22504699

RESUMEN

We report two cases of advanced gastric cancer. The first was a 77-year-old man who had experienced distal gastrectomy about 35 years ago. He complained of abdominal bloating, and a gastrointestinal scope showed that he had advanced gastric cancer. CT scan revealed massive ascites. Dissemination of the peritoneum was suspected, and chemotherapy using S-1 (80mg/m², biweekly)plus paclitaxel (50mg/m², on days 1 and 8) was selected, He had no major side effects and the ascites disappeared. He was able to receive 18 courses on an outpatient basis. The second case was a 79-year-old man who had total gastrectomy performed 1 year ago. Invasion to the diaphragm and lymph node metastasis were detected. We selected S-1 (80 mg/m²)as adjuvant chemotherapy but that caused severe fatigue. Eventually he refused the drug. Six month later, he had abdominal bloating and CT scan revealed that he had massive ascites. UFT-E (1. 5 g/body) was administered and paclitaxe (l 50 mg/m²) was added. The ascites disappeared and he has had a stable life. DIF (S-1, UFT) plus paclitaxel is considered to be a useful chemotherapy combination against advanced gastric cancer that has peritoneal dissemination or ascites, even for older patients.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Ascitis/etiología , Neoplasias Peritoneales/tratamiento farmacológico , Peritonitis/complicaciones , Calidad de Vida , Neoplasias Gástricas/tratamiento farmacológico , Anciano , Dihidrouracilo Deshidrogenasa (NADP)/antagonistas & inhibidores , Combinación de Medicamentos , Humanos , Masculino , Ácido Oxónico/administración & dosificación , Paclitaxel/administración & dosificación , Neoplasias Peritoneales/secundario , Neoplasias Gástricas/patología , Tegafur/administración & dosificación , Tomografía Computarizada por Rayos X
16.
J Radiat Res ; 53(2): 250-6, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22510597

RESUMEN

Gimeracil, an inhibitor of dihydropyrimidine dehydrogenase (DPYD), partially inhibits homologous recombination (HR) repair and has a radiosensitizing effect as well as enhanced sensitivity to Camptothecin (CPT). DPYD is the target protein for radiosensitization by Gimeracil. We investigated the mechanisms of sensitization of radiation and CPT by DPYD inhibition using DLD-1 cells treated with siRNA for DPYD. We investigated the focus formation of various kinds of proteins involved in HR and examined the phosphorylation of RPA by irradiation using Western blot analysis. DPYD depletion by siRNA significantly restrained the formation of radiation-induced foci of Rad51 and RPA, whereas it increased the number of foci of NBS1. The numbers of colocalization of NBS1 and RPA foci in DPYD-depleted cells after radiation were significantly smaller than in the control cells. These results suggest that DPYD depletion is attributable to decreased single-stranded DNA generated by the Mre11/Rad50/NBS1 complex-dependent resection of DNA double-strand break ends. The phosphorylation of RPA by irradiation was partially suppressed in DPYD-depleted cells, suggesting that DPYD depletion may partially inhibit DNA repair with HR by suppressing phosphorylation of RPA. DPYD depletion showed a radiosensitizing effect as well as enhanced sensitivity to CPT. The radiosensitizing effect of DPYD depletion plus CPT was the additive effect of DPYD depletion and CPT. DPYD depletion did not have a cell-killing effect, suggesting that DPYD depletion may not be so toxic. Considering these results, the combination of CPT and drugs that inhibit DPYD may prove useful for radiotherapy as a method of radiosensitization.


Asunto(s)
Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/patología , Daño del ADN/efectos de los fármacos , Dihidrouracilo Deshidrogenasa (NADP)/antagonistas & inhibidores , Dihidrouracilo Deshidrogenasa (NADP)/metabolismo , Tolerancia a Radiación/efectos de los fármacos , Proteína de Replicación A/metabolismo , Antineoplásicos Fitogénicos/administración & dosificación , Camptotecina , Línea Celular Tumoral , Humanos , Fosforilación/efectos de los fármacos , Fosforilación/efectos de la radiación , Dosis de Radiación
17.
Cancer Sci ; 102(9): 1712-6, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21668582

RESUMEN

Gimeracil (5-chloro-2, 4-dihydroxypyridine) is an inhibitor of dihydropyrimidine dehydrogenase (DPYD), which degrades pyrimidine including 5-fluorouracil in the blood. Gimeracil was originally added to an oral fluoropyrimidine derivative S-1 to yield prolonged 5-fluorouracil concentrations in serum and tumor tissues. We have already reported that gimeracil had radiosensitizing effects by partially inhibiting homologous recombination (HR) in the repair of DNA double strand breaks. We investigated the mechanisms of gimeracil radiosensitization. Comet assay and radiation-induced focus formation of various kinds of proteins involved in HR was carried out. siRNA for DPYD were transfected to HeLa cells to investigate the target protein for radiosensitization with gimeracil. SCneo assay was carried out to examine whether DPYD depletion by siRNA inhibited HR repair of DNA double strand breaks. Tail moments in neutral comet assay increased in gimeracil-treated cells. Gimeracil restrained the formation of foci of Rad51 and replication protein A (RPA), whereas it increased the number of foci of Nbs1, Mre11, Rad50, and FancD2. When HeLa cells were transfected with the DPYD siRNA before irradiation, the cells became more radiosensitive. The degree of radiosensitization by transfection of DPYD siRNA was similar to that of gimeracil. Gimeracil did not sensitize DPYD-depleted cells. Depletion of DPYD by siRNA significantly reduced the frequency of neopositive clones in SCneo assay. Gimeracil partially inhibits the early step in HR. It was found that DPYD is the target protein for radiosensitization by gimeracil. The inhibitors of DPYD, such as gimeracil, could enhance the efficacy of radiotherapy through partial suppression of HR-mediated DNA repair.


Asunto(s)
Reparación del ADN , Dihidrouracilo Deshidrogenasa (NADP)/antagonistas & inhibidores , Piridinas/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Recombinación Genética , Línea Celular Tumoral , Inhibidores Enzimáticos , Células HeLa , Humanos , ARN Interferente Pequeño/farmacología , Transfección
18.
Anticancer Res ; 31(2): 625-32, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21378348

RESUMEN

UNLABELLED: The purpose of this study was to investigate the effect of gimeracil (CDHP), a reversible dihydropyrimidine dehydrogenase (DPD) inhibitor, on the pharmacokinetics of 5-fluorouracil (5-FU) and other related metabolites by comparing the pharmacokinetic (PK) profile of S-1 (tegafur [FT] + CDHP + oteracil potassium [Oxo]) to that of FT alone. PATIENTS AND METHODS: Patients with advanced solid tumors received single oral doses of S-1 (50 mg) and FT (800 mg) on days 1 and 8 in a randomized crossover fashion. Plasma samples were collected on days 1, 2, 3, 8, 9 and 10. Single-dose PK parameters were determined for FT, 5-FU and α-fluoro-ß-alanine (FBAL). Following the single-dose crossover period, patients entered an extension phase and received treatment with S-1 b.i.d. for 14 days followed by a 7-day rest, repeated every 3 weeks. RESULTS: A total of 12 patients were enrolled; median age was 59 years and mean body surface area was 1.94 m(2). Following S-1 administration, 5-FU exposure was significantly greater (approximately 3-fold) compared to FT alone (p ≤ 0.0007 for AUC0-inf, AUC0-last, and C(max) of 5-FU) despite the 16-fold higher dose of FT administered alone compared to S-1, while plasma concentrations of FT and FBAL were significantly lower with S-1 (p < 0.0001 for all comparisons). Following both single- and multiple-dose administration of S-1, the average maximum DPD inhibition was observed at 4 h post-dose. The extent of inhibition was similar following single and multiple dosing. Following single- and multiple-dose administration of S-1, plasma concentrations of uracil returned to baseline levels within approximately 48 h of dosing, indicating reversibility of DPD inhibition by CDHP. CONCLUSION: Despite the differences in the FT dose administered, exposure to 5-FU was significantly greater following S-1 administration compared to FT administration. Conversely, exposure to FT and FBAL were significantly less following S-1 administration compared to FT administration. Thus, the DPD inhibitory action of CDHP contributes to a decrease in 5-FU catabolism and to significantly higher blood levels of 5-FU compared to FT alone.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Fluorouracilo/farmacocinética , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Piridinas/farmacología , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Estudios Cruzados , Dihidrouracilo Deshidrogenasa (NADP)/antagonistas & inhibidores , Dihidrouracilo Deshidrogenasa (NADP)/metabolismo , Combinación de Medicamentos , Interacciones Farmacológicas , Femenino , Fluorouracilo/antagonistas & inhibidores , Humanos , Masculino , Persona de Mediana Edad , Ácido Oxónico/administración & dosificación , Ácido Oxónico/farmacocinética , Piridinas/administración & dosificación , Piridinas/farmacocinética , Tegafur/administración & dosificación , Tegafur/farmacocinética
19.
Cancer Chemother Pharmacol ; 68(2): 497-504, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21079960

RESUMEN

PURPOSE: Malignant pleural mesothelioma (MPM) is a highly lethal neoplasm. S-1 has been developed as a novel oral antineoplastic agent based on the modulation of 5-fluorouracil (5-FU) bioactivity. This study was conducted to investigate the preclinical therapeutic effect of S-1 on MPM. METHODS: We used three human MPM cell lines, Y-MESO-14, NCI-H290 and MSTO-211H. In vitro proliferation of human MPM cells was determined by MTT assay. Human MPM cells were orthotopically implanted into thoracic cavity of SCID mice. Tumor-bearing mice were treated with S-1 or vehicle. RESULTS: The combination of 5-FU and 5-chloro-2,4-dihydroxypyridine (CDHP) was more effective than 5-FU alone in inhibiting MPM cell proliferation in vitro. This combination was most effective in Y-MESO-14 cells, which co-expressed high protein level of dihydropyrimidine dehydrogenase (DPD) and thymidine phosphorylase (TP). In vivo data showed that treatment with S-1 significantly reduced thoracic tumors and pleural effusion produced by Y-MESO-14 cells. Moreover, treatment with S-1 prolonged the survival of Y-MESO-14 cell-bearing SCID mice. CONCLUSIONS: We demonstrated that S-1 was effective for inhibiting the proliferation of MPM cells, particularly with both DPD and TP expressions, suggesting that S-1 might be therapeutically effective for control of MPM.


Asunto(s)
Antimetabolitos Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Mesotelioma/tratamiento farmacológico , Ácido Oxónico/uso terapéutico , Neoplasias Pleurales/tratamiento farmacológico , Piridinas/uso terapéutico , Tegafur/uso terapéutico , Animales , Antimetabolitos Antineoplásicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Línea Celular Tumoral , Dihidrouracilo Deshidrogenasa (NADP)/antagonistas & inhibidores , Dihidrouracilo Deshidrogenasa (NADP)/metabolismo , Combinación de Medicamentos , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/uso terapéutico , Humanos , Masculino , Mesotelioma/metabolismo , Mesotelioma/patología , Ratones , Ratones SCID , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Ácido Oxónico/administración & dosificación , Pentosiltransferasa/antagonistas & inhibidores , Neoplasias Pleurales/metabolismo , Neoplasias Pleurales/patología , Piridinas/administración & dosificación , Distribución Aleatoria , Análisis de Supervivencia , Tegafur/administración & dosificación , Timidina Fosforilasa/metabolismo , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...