Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
Toxicol Appl Pharmacol ; 403: 115163, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32730777

RESUMEN

During its clinical development fialuridine caused liver toxicity and the death of five patients. This case remains relevant due to the continued development of mechanistically-related compounds against a back-drop of simple in vitro models which remain limited for the preclinical detection of such delayed toxicity. Here, proteomic investigation of a differentiated, HepaRG, and proliferating, HepG2 cell model was utilised to confirm the presence of the hENT1 transporter, thymidine kinase-1 and -2 (TK1, TK2) and thymidylate kinase, all essential in order to reproduce the cellular activation and disposition of fialuridine in the clinic. Acute metabolic modification assays could only identify mitochondrial toxicity in HepaRG cells following extended dosing, 2 weeks. Toxic effects were observed around 10 µM, which is within a range of 10-15 X approximate Cmax. HepaRG cell death was accompanied by a significant decrease in mitochondrial DNA content, indicative of inhibition of mitochondrial replication, and a subsequent reduction in mitochondrial respiration and the activity of mitochondrial respiratory complexes, not replicated in HepG2 cells. The structural epimer of fialuridine, included as a pharmacological negative control, was shown to have no cytotoxic effects in HepaRG cells up to 4 weeks. Overall, these comparative studies demonstrate the HepaRG model has translational relevance for fialuridine toxicity and therefore may have potential in investigating the inhibition of mitochondrial replication over prolonged exposure for other toxicants.


Asunto(s)
Antivirales/farmacología , Arabinofuranosil Uracilo/análogos & derivados , Hepatocitos/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Arabinofuranosil Uracilo/farmacología , Línea Celular Tumoral , Replicación del ADN/efectos de los fármacos , ADN Mitocondrial/fisiología , Relación Dosis-Respuesta a Droga , Humanos , Mitocondrias/fisiología
2.
Antimicrob Agents Chemother ; 58(12): 7312-23, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25267682

RESUMEN

The susceptibilities of gammaherpesviruses, including Epstein-Barr virus (EBV), Kaposi's sarcoma-associated herpesvirus (KSHV), and animal rhadinoviruses, to various nucleoside analogs was investigated in this work. Besides examining the antiviral activities and modes of action of antivirals currently marketed for the treatment of alpha- and/or betaherpesvirus infections (including acyclovir, ganciclovir, penciclovir, foscarnet, and brivudin), we also investigated the structure-activity relationship of various 5-substituted uridine and cytidine molecules. The antiviral efficacy of nucleoside derivatives bearing substitutions at the 5 position was decreased if the bromovinyl was replaced by chlorovinyl. 1-ß-D-Arabinofuranosyl-(E)-5-(2-bromovinyl)uracil (BVaraU), a nucleoside with an arabinose configuration of the sugar ring, exhibited no inhibitory effect against rhadinoviruses but was active against EBV. On the other hand, the fluoroarabinose cytidine analog 2'-fluoro-5-iodo-aracytosine (FIAC) showed high selectivity indices against gammaherpesviruses that were comparable to those of brivudin. Additionally, we selected brivudin- and acyclovir-resistant rhadinoviruses in vitro and characterized them by phenotypic and genotypic (i.e., sequencing of the viral thymidine kinase, protein kinase, and DNA polymerase) analysis. Here, we reveal key amino acids in these enzymes that play an important role in substrate recognition. Our data on drug susceptibility profiles of the different animal gammaherpesvirus mutants highlighted cross-resistance patterns and indicated that pyrimidine nucleoside derivatives are phosphorylated by the viral thymidine kinase and purine nucleosides are preferentially activated by the gammaherpesvirus protein kinase.


Asunto(s)
Antivirales/farmacología , Farmacorresistencia Viral/genética , Herpesvirus Humano 4/efectos de los fármacos , Herpesvirus Humano 8/efectos de los fármacos , Rhadinovirus/efectos de los fármacos , Proteínas Virales/química , Aciclovir/análogos & derivados , Aciclovir/química , Aciclovir/farmacología , Secuencia de Aminoácidos , Animales , Antivirales/química , Arabinofuranosil Uracilo/análogos & derivados , Arabinofuranosil Uracilo/química , Arabinofuranosil Uracilo/farmacología , Bromodesoxiuridina/análogos & derivados , Bromodesoxiuridina/química , Bromodesoxiuridina/farmacología , Citarabina/análogos & derivados , Citarabina/química , Citarabina/farmacología , ADN Polimerasa Dirigida por ADN/química , ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Foscarnet/química , Foscarnet/farmacología , Ganciclovir/química , Ganciclovir/farmacología , Guanina , Herpesvirus Humano 4/enzimología , Herpesvirus Humano 4/genética , Herpesvirus Humano 8/enzimología , Herpesvirus Humano 8/genética , Humanos , Datos de Secuencia Molecular , Proteínas Quinasas/química , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Rhadinovirus/enzimología , Rhadinovirus/genética , Alineación de Secuencia , Relación Estructura-Actividad , Timidina Quinasa/química , Timidina Quinasa/genética , Timidina Quinasa/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo
3.
Biomacromolecules ; 14(3): 801-10, 2013 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-23373724

RESUMEN

1ß-d-Arabinofuranosylcytosine (Cytarabine, Ara-C) is a key drug in the treatment of acute myeloid leukemia. Ara-C has a number of limitations such as a rapid deactivation by cytidine deaminase leading to the formation of a biologically inactive metabolite, Ara-U (1ß-d-arabinofuranosyluracil), a low lipophilicity, and fast clearance from the body. To address these problems, we developed a conjugate in which hydroxyl-terminated PAMAM dendrimer, G4-OH ["D"] and PEG were used as carriers for the drug (Ara-C). The conjugates were synthesized using an efficient multistep protection/deprotection method resulting in the formation of a covalent bond between the primary hydroxyl group of Ara-C and dendrimer/PEG. The structure, physicochemical properties, and drug release kinetics were characterized extensively. (1)H NMR and MALDI-TOF mass spectrometry suggested covalent attachment of 10 Ara-C molecules to the dendrimer. The release profile of Ara-C in human plasma and in PBS buffer (pH 7.4) showed that the conjugates released the drug over 14 days in PBS, with the release sped up in plasma. In PBS, while most of the drug is released from PEG-Ara-C, the dendrimer continues to release the drug in a sustained fashion. The results also suggested that the formation of the inactive form of Ara-C (Ara-U) was delayed upon conjugation of Ara-C to the polymers. The inhibition of cancer growth by the dendrimer-Ara-C and PEG-Ara-C conjugates was evaluated in A549 human adenocarcinoma epithelial cells. Both dendrimer- and PEG-Ara-C conjugates were 4-fold more effective in inhibition of A549 cells compared to free Ara-C after 72 h of treatment.


Asunto(s)
Citarabina/farmacología , Dendrímeros/química , Polietilenglicoles/química , Arabinofuranosil Uracilo/sangre , Arabinofuranosil Uracilo/química , Arabinofuranosil Uracilo/farmacología , Línea Celular Tumoral , Fenómenos Químicos , Cromatografía Líquida de Alta Presión , Citarabina/sangre , Citarabina/química , Citidina Desaminasa/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Leucemia Mieloide Aguda/tratamiento farmacológico , Espectroscopía de Resonancia Magnética , Tamaño de la Partícula , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
4.
BMC Gastroenterol ; 12: 4, 2012 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-22230186

RESUMEN

BACKGROUND: Clevudine is a nucleoside analog reverse transcriptase inhibitor that exhibits potent antiviral activity against hepatitis B virus (HBV) without serious side effects. However, mitochondrial myopathy has been observed in patients with chronic HBV infection taking clevudine. Moreover, the development of diabetes was recently reported in patients receiving long-term treatment with clevudine. In this study, we investigated the effects of clevudine on mitochondrial function and insulin release in a rat clonal ß-cell line, INS-1E. METHODS: The mitochondrial DNA (mtDNA) copy number and the mRNA levels were measured by using quantitative PCR. MTT analysis, ATP/lactate measurements, and insulin assay were performed. RESULTS: Both INS-1E cells and HepG2 cells, which originated from human hepatoma, showed dose-dependent decreases in mtDNA copy number and cytochrome c oxidase-1 (Cox-1) mRNA level following culture with clevudine (10 µM-1 mM) for 4 weeks. INS-1E cells treated with clevudine had reduced total mitochondrial activities, lower cytosolic ATP contents, enhanced lactate production, and more lipid accumulation. Insulin release in response to glucose application was markedly decreased in clevudine-treated INS-1E cells, which might be a consequence of mitochondrial dysfunction. CONCLUSIONS: Our data suggest that high-dose treatment with clevudine induces mitochondrial defects associated with mtDNA depletion and impairs glucose-stimulated insulin secretion in insulin-releasing cells. These findings partly explain the development of diabetes in patients receiving clevudine who might have a high susceptibility to mitochondrial toxicity.


Asunto(s)
Antivirales/farmacología , Arabinofuranosil Uracilo/análogos & derivados , Glucosa/farmacología , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/fisiología , Adenosina Trifosfato/metabolismo , Animales , Antivirales/efectos adversos , Arabinofuranosil Uracilo/efectos adversos , Arabinofuranosil Uracilo/farmacología , Línea Celular , Variaciones en el Número de Copia de ADN/efectos de los fármacos , ADN Mitocondrial/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Complejo IV de Transporte de Electrones/metabolismo , Células Hep G2 , Humanos , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Lactatos/metabolismo , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas
5.
Antivir Chem Chemother ; 20(4): 153-60, 2010 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-20231780

RESUMEN

BACKGROUND: Nucleoside analogues always require phosphorylation to be active. This appears to be a particular limitation for uridine-based nucleosides. Our ProTide method allows the direct use of masked membrane-soluble preformed nucleoside phosphates, bypassing the need for the initial phosphorylation step. We herein applied it to some novel 5-trimethylsilyl arabinosyl uridines. METHODS: 5-Trimethylsilyl-1-beta-D-(arabinofuranosyl)uracil was prepared in six steps starting from uridine, and five phosphoramidate ProTide derivatives were synthesized. These compounds were investigated for activity against a range of DNA and RNA viruses, including herpes simplex virus type-1 and type-2, vaccinia virus and HIV. RESULTS: Overall, these compounds did not show significant antiviral activity against any of the viruses tested. CONCLUSIONS: The inactivity of the ProTides of this nucleoside could correspond with poor ProTide activation in vitro, poor onward metabolism or low activity of the putative monophosphate metabolite.


Asunto(s)
Antivirales/química , Antivirales/farmacología , Arabinofuranosil Uracilo/análogos & derivados , Arabinofuranosil Uracilo/farmacología , Amidas/síntesis química , Amidas/química , Amidas/farmacología , Animales , Antivirales/síntesis química , Arabinofuranosil Uracilo/síntesis química , Línea Celular Tumoral , Chlorocebus aethiops , Efecto Citopatogénico Viral/efectos de los fármacos , Perros , Fibroblastos/virología , Humanos , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Ácidos Fosfóricos/síntesis química , Ácidos Fosfóricos/química , Ácidos Fosfóricos/farmacología , Compuestos de Trimetilsililo/síntesis química , Compuestos de Trimetilsililo/química , Compuestos de Trimetilsililo/farmacología , Replicación Viral/efectos de los fármacos
6.
Bioorg Med Chem ; 18(7): 2439-46, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20299228

RESUMEN

2'-Beta-D-arabinouridine (AraU), the uridine analogue of the anticancer agent AraC, was synthesized and evaluated for antiviral activity and cytotoxicity. In addition, a series of AraU monophosphate prodrugs in the form of triester phosphoramidates (ProTides) were also synthesized and tested against a range of viruses, leukaemia and solid tumour cell lines. Unfortunately, neither the parent compound (AraU) nor any of its ProTides showed antiviral activity, nor potent inhibitory activity against any of the cancer cell lines. Therefore, the metabolism of AraU phosphoramidates to release AraU monophosphate was investigated. The results showed carboxypeptidase Y, hog liver esterase and crude CEM tumor cell extracts to hydrolyse the ester motif of phosphoramidates with subsequent loss of the aryl group, while molecular modelling studies suggested that the AraU l-alanine aminoacyl phosphate derivative might not be a good substrate for the phosphoramidase enzyme Hint-1. These findings are in agreement with the observed disappearance of intact prodrug and concomitant appearance of the corresponding phosphoramidate intermediate derivative in CEM cell extracts without measurable formation of araU monophosphate. These findings may explain the poor antiviral/cytostatic potential of the prodrugs.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Antivirales/síntesis química , Antivirales/farmacología , Arabinofuranosil Uracilo/síntesis química , Arabinofuranosil Uracilo/farmacología , Profármacos/síntesis química , Profármacos/farmacología , Animales , Antineoplásicos/farmacocinética , Antivirales/farmacocinética , Arabinofuranosil Uracilo/farmacocinética , Catepsina A/antagonistas & inhibidores , Catepsina A/metabolismo , Línea Celular Tumoral , Cromatografía en Capa Delgada , Diseño de Fármacos , Esterasas/antagonistas & inhibidores , Humanos , Indicadores y Reactivos , Hígado/efectos de los fármacos , Hígado/enzimología , Linfocitos/efectos de los fármacos , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Profármacos/farmacocinética , Relación Estructura-Actividad , Porcinos , Virus/efectos de los fármacos
7.
Antivir Ther ; 12(5): 761-8, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17713159

RESUMEN

AIMS: This study was conducted to clarify the resistance profile of a novel mutation pattern emerging during lamivudine (3TC) therapy and showing cross-resistance to adefovir dipivoxil (ADV) in a patient with chronic hepatitis B. METHODS AND RESULTS: Successful suppression of hepatitis B virus (HBV) replication by sequential therapy of 9 MU thrice weekly interferon (IFN) and 3TC was followed by genotypical resistance detected at month 28 of therapy (month 19 of lamivudine treatment). ADV was added to 3TC therapy on month 44 of antiviral treatment. Neither alanine aminotransferase normalization nor a stable decrease in HBV viral load was observed, although ADV was used for more than 40 months. The HBV pol region was amplified from serum samples obtained before and after ADV treatment. The complete genome was cloned into a TA vector. PCR products and 7-10 clones from each cloned vector were sequenced. A novel mutation, A181S, in the reverse transcriptase gene leading to a conversion of W172C in the overlapping surface antigen gene was detected along with a M2041 mutation. The complete genome comprising the A181S+M2041 pattern was cloned into an expression vector and its in vitro susceptibility to 3TC, ADV, tenofovir (PMPA), clevudine (L-FMAU) and emtricitabine (FTC) were determined in transiently transfected Huh7 cells. This mutation pattern displayed more than 1000-fold resistance to the nucleoside analogues 3TC and FTC and approximately sixfold resistance to L-FMAU, while it confers 28.23- and 5.57-fold resistance for the nucleotide analogues ADV and PMPA, respectively. CONCLUSION: A new mutation pattern, A181S+M2041, arising under lamivudine treatment confers cross-resistance to ADV both in vivo and in vitro.


Asunto(s)
Adenina/análogos & derivados , Farmacorresistencia Viral Múltiple/genética , Virus de la Hepatitis B/efectos de los fármacos , Hepatitis B Crónica/tratamiento farmacológico , Lamivudine/uso terapéutico , Mutación , Organofosfonatos/uso terapéutico , ADN Polimerasa Dirigida por ARN/genética , Inhibidores de la Transcriptasa Inversa/uso terapéutico , Adenina/farmacología , Adenina/uso terapéutico , Adulto , Arabinofuranosil Uracilo/análogos & derivados , Arabinofuranosil Uracilo/farmacología , Secuencia de Bases , Línea Celular Tumoral , Clonación Molecular , Análisis Mutacional de ADN , ADN Viral/sangre , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Relación Dosis-Respuesta a Droga , Emtricitabina , Virus de la Hepatitis B/enzimología , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/crecimiento & desarrollo , Hepatitis B Crónica/sangre , Hepatitis B Crónica/genética , Humanos , Lamivudine/farmacología , Masculino , Datos de Secuencia Molecular , Organofosfonatos/farmacología , ADN Polimerasa Dirigida por ARN/metabolismo , Inhibidores de la Transcriptasa Inversa/farmacología , Tenofovir , Factores de Tiempo , Transfección , Resultado del Tratamiento , Carga Viral , Replicación Viral/efectos de los fármacos
8.
Eur J Nucl Med Mol Imaging ; 34(6): 822-9, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17206416

RESUMEN

PURPOSE: FIAU and FEAU were evaluated in vitro and in vivo as markers for HSV1-tk gene expression. METHODS: In vitro and biodistribution studies were performed in wild type and transduced HT-29 cells using [14C]FIAU and [3H]FEAU. PET imaging was performed using [18F]FIAU and [18F]FEAU. RESULTS: In vitro uptake of [14C]FIAU in tk-positive cells was 39-fold, 49-fold, and 43-fold higher (p<0.001) than in wild type cells at 30, 60, and 120 min, respectively. Uptake of [3H]FEAU in transduced cells was 46-fold, 62-fold, and 121-fold higher (p<0.001) than in wild type cells at the same time points. In vivo uptake of [14C]FIAU at 2 h in HSV1-tk positive tumors was 15.48+/-3.94, 6.7-fold higher (p<0.001) than in wild type tumors. Uptake of [3H]FEAU in transduced tumors was 9.98+/-1.99, 5.0-fold higher (p<0.001) than in wild type tumors. Micro-PET images using [18F]FIAU and [18F]FEAU also showed very high uptake in HSV-tk tumors. CONCLUSION: [18F]FIAU and [18F]FEAU appear to be potential PET imaging agents for gene expression.


Asunto(s)
Arabinofuranosil Uracilo/farmacología , Terapia Genética/métodos , Animales , Radioisótopos de Carbono , ADN/metabolismo , Radioisótopos de Flúor , Técnicas de Transferencia de Gen , Genes Reporteros , Técnicas Genéticas , Terapia Genética/instrumentación , Humanos , Ratones , Ratones Desnudos , Tomografía de Emisión de Positrones/instrumentación , Tomografía de Emisión de Positrones/métodos , Radiofármacos/farmacología , Timidina Quinasa/metabolismo
9.
J Nucl Med ; 47(7): 1161-71, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16818951

RESUMEN

UNLABELLED: The ability to monitor tumor responses during prodrug activation gene therapy and other anticancer gene therapies is critical for their translation into clinical practice. Previously, we demonstrated the feasibility of noninvasive in vivo imaging with 131I-5-iodo-2'-fluoro-1-beta-D-arabinofuranosyluracil (131I-FIAU) for monitoring herpes simplex virus type 1 thymidine kinase (HSV1-tk) cancer gene expression in an experimental animal model. Here we tested the efficacy of SPECT with 123I-FIAU and PET with 5-18F-fluoro-2'-deoxyuridine (18F-FUdR), 2-18F-fluoroethyl-L-tyrosine (18F-FET), and 18F-FDG for monitoring tumor responses during prodrug activation gene therapy with HSV1-tk and ganciclovir (GCV). METHODS: In the flanks of FVB/N female mice, 4 tumors per animal were established by subcutaneous injection of 1 x 10(5) cells of NG4TL4 sarcoma cells, HSV1-tk-transduced NG4TL4-STK cells, or a mixture of these cells in different proportions to model different efficacies of transfection and HSV1-tk gene expression levels in tumors. Ten days later, the animals were treated with GCV (10 mg/kg/d intraperitoneally) for 7 d. Gamma-Imaging with 123I-FIAU and PET with 18F-FUdR, 18F-FET, and 18F-FDG were performed before and after initiation of therapy with GCV in the same animal. RESULTS: Before GCV treatment, no significant difference in weight and size was found in tumors that expressed different HSV1-tk levels, suggesting similar in vivo proliferation rates for NG4TL4 and NG4TL4-STK sarcomas. The accumulation of 123I-FIAU at 24 h after injection was directly proportional to the percentage of NG4TL4-STK cells in the tumors. The 123I-FIAU accumulation at 4 and 7 d of GCV therapy decreased significantly compared with pretreatment levels and was proportional to the percentage of HSV1-tk-positive tumor cells. Tumor uptake of 18F-FUdR in all HSV1-tk-expressing tumors also decreased significantly compared with pretreatment levels and was proportional to the percentage of HSV1-tk-positive tumor cells. The accumulation of 18F-FET decreased minimally (about 1.5-fold) and 18F-FDG decreased only 2-fold after 7 d of GCV therapy, and the degree of reduction was proportional to the percentage of HSV1-tk-positive tumor cells. CONCLUSION: We have shown that gamma-camera imaging with 123I-FIAU was the most reliable method for prediction of tumor response to GCV therapy, which was proportional to the magnitude of HSV1-tk expression in tumor tissue. 123I-FIAU imaging can be used to verify the efficacy of elimination of HSV1-tk-expressing cells by therapy with GCV. PET with 18F-FUdR reliably visualizes proliferating tumor tissue and is most suitable for the assessment of responses in tumors undergoing HSV1-tk plus GCV prodrug activation gene therapy. PET with 18F-FDG or 18F-FET can be used as additional "surrogate" biomarkers of the treatment response, although these radiotracers are less sensitive than 18F-FUdR for monitoring tumor responses to prodrug activation gene therapy with HSV1-tk and GCV in this sarcoma model.


Asunto(s)
Arabinofuranosil Uracilo/análogos & derivados , Floxuridina/farmacología , Fluorodesoxiglucosa F18/farmacología , Ganciclovir/farmacología , Terapia Genética/métodos , Herpesvirus Humano 1/enzimología , Radioisótopos de Yodo/farmacología , Neoplasias/genética , Timidina Quinasa/metabolismo , Tirosina/análogos & derivados , Animales , Antivirales/farmacología , Arabinofuranosil Uracilo/farmacología , Línea Celular Tumoral , Femenino , Humanos , Ratones , Neoplasias/patología , Profármacos , Tirosina/farmacología
10.
Expert Opin Investig Drugs ; 14(10): 1277-84, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16185170

RESUMEN

Chronic hepatitis B virus (HBV) infection is a major health problem that is responsible for < or = 1 million deaths and 500,000 cases of hepatocellular carcinoma worldwide each year. Drugs that are currently approved by the FDA for the treatment of chronic HBV consist of two groups: the immunomodulators, such as conventional IFN-alpha and pegylated IFN-alpha2a; and nucleoside/nucleotide analogues, such as lamivudine, adefovir dipivoxil and entecavir. However, due to the limitations of these agents, newer agents with improved efficacy are currently being developed. One nucleoside/nucleotide analogue that is drawing a wide range of interest is clevudine, which is an analogue of the unnatural beta-L configuration. In the woodchuck hepatitis virus (WHV), clevudine 10 mg/kg has proven to be effective in suppressing viral replication with < or = 9 log10 decreases in WHV. At this dose, a significant reduction of intrahepatic WHV RNA and covalently closed circular WHV DNA levels can also be observed. Treatment with clevudine 10 mg/kg can confer additional antiviral benefit in the form of a more sustained reduction in WHV replication, serum woodchuck hepatitis surface antigen and intrahepatic woodchuck hepatitis core antigen expression following the withdrawal of clevudine. In humans, clevudine 10, 50, 100 or 200 mg/day for 28 days can reduce the median HBV DNA by -2.5, -2.7, -3 and -2.6 log10, respectively. More importantly, this suppression of antiviral activity is maintained at 12 and 24 weeks post treatment. Based on the early results of clevudine, more large-scale human studies with clevudine monotherapy or combination therapy is eagerly awaited.


Asunto(s)
Arabinofuranosil Uracilo/análogos & derivados , Virus de la Hepatitis B/efectos de los fármacos , Hepatitis B Crónica/tratamiento farmacológico , Animales , Antivirales/química , Antivirales/farmacología , Antivirales/uso terapéutico , Arabinofuranosil Uracilo/química , Arabinofuranosil Uracilo/farmacología , Arabinofuranosil Uracilo/uso terapéutico , Evaluación Preclínica de Medicamentos/tendencias , Virus de la Hepatitis B/patogenicidad , Hepatitis B Crónica/virología , Humanos
11.
Stud Health Technol Inform ; 107(Pt 1): 454-8, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15360854

RESUMEN

Drug Interaction Ontology (DIO) was developed for formal representation of pharmacological knowledge. It provides a fundamental framework for accumulation of reusable knowledge components in molecular pharmacology. Ontology was employed and implemented as a relational model. Some features include: 1) Drug-biomolecule interaction was assumed as a primitive knowledge element. 2) Symbolic representation was developed for drug-biomolecule interaction. Consequences of two conjugated units of interaction were defined by using symbols. These are applied for query development for identification of possible drug-drug interaction. 3) The triadic relationship model was developed as a ground model for bio-logical interactions and/or function, including semantic ones. One application of DIO is to support hypothesis generation of drug interaction by providing new hypotheses from a structured database storing literature information on known drug-biomolecule interactions. A knowledge base using DIO that contains information beginning with anti-cancer drugs is now under development. Detection of possible drug interaction was tested and its capacity to lead clinically known ones was confirmed. The system generated theoretically possible drug-drug interactions, which implies potential usefulness of new drugs to be tested before actual clinical application. In this paper, sorivudine and 5-fluorouracil mediated by dihydropyrimidine dehydrogenase are presented.


Asunto(s)
Arabinofuranosil Uracilo/análogos & derivados , Interacciones Farmacológicas , Vocabulario Controlado , Arabinofuranosil Uracilo/farmacología , Inteligencia Artificial , Biología Computacional , Dihidrouracilo Deshidrogenasa (NADP)/farmacología , Fluorouracilo/farmacología , Humanos
12.
Hepatology ; 36(3): 710-22, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12198665

RESUMEN

To design combination strategies for chronic hepatitis B therapy, we evaluated in vitro the inhibitory activity of 4 nucleoside analogs, (-)FTC, L-FMAU, DXG, and DAPD, in comparison with lamivudine (3TC) and PMEA. In a cell-free assay for the expression of wild-type duck hepatitis B virus (DHBV) reverse transcriptase, DAPD-TP was found to be the most active on viral minus strand DNA synthesis, including the priming reaction, followed by 3TC-TP, (-)FTC-TP, and DXG-TP, whereas L-FMAU-TP was a weak inhibitor. In cell culture experiments, important differences in drug concentration allowing a 50% inhibition of viral replication or polymerase activity (IC50s) were observed depending on the cell type used, showing that antiviral effect of nucleoside analogs may depend on their intracellular metabolism. IC50s obtained for wild-type DHBV replication in primary duck hepatocytes were much lower than with DHBV transfected LMH cells. IC50s were also significantly lower in the 2.2.1.5 and HepG2 cells compared with HBV transfected HuH7 cells. Moreover, L-FMAU inhibited preferentially HBV plus strand DNA synthesis in these cell lines. The antiviral effect of these inhibitors was also evaluated against 3TC-resistant mutants of the DHBV and HBV polymerases. These mutants were found to be cross resistant to (-)FTC. By contrast, the double DHBV polymerase mutant was sensitive to DXG-TP and DAPD-TP. Moreover, both purine analogs remained active against DHBV and HBV 3TC-resistant mutants in transfected LMH and HepG2 cells, respectively. In conclusion, the unique mechanism of action of these new inhibitors warrants further evaluation in experimental models to determine their capacity to delay or prevent the selection of drug resistant mutants.


Asunto(s)
Trifosfato de Arabinofuranosil Citosina/análogos & derivados , Citidina Trifosfato/análogos & derivados , Dioxolanos/farmacología , Guanosina/análogos & derivados , Infecciones por Hepadnaviridae/tratamiento farmacológico , Virus de la Hepatitis B del Pato/genética , Hepatitis Viral Animal/tratamiento farmacológico , Nucleósidos de Purina/farmacología , Tiofenos , Animales , Trifosfato de Arabinofuranosil Citosina/química , Trifosfato de Arabinofuranosil Citosina/farmacología , Arabinofuranosil Uracilo/análogos & derivados , Arabinofuranosil Uracilo/química , Arabinofuranosil Uracilo/farmacología , Carcinoma Hepatocelular , Citidina Trifosfato/química , Citidina Trifosfato/farmacología , Replicación del ADN/efectos de los fármacos , ADN Viral/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Dioxolanos/química , Farmacorresistencia Viral , Regulación Viral de la Expresión Génica/efectos de los fármacos , Guanosina/química , Guanosina/farmacología , Virus de la Hepatitis B , Hepatocitos/citología , Hepatocitos/virología , Humanos , Lamivudine/farmacología , Neoplasias Hepáticas , Mutación , Nucleósidos de Purina/química , Inhibidores de la Transcriptasa Inversa/farmacología , Células Tumorales Cultivadas
13.
J Virol ; 76(12): 6356-63, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12021368

RESUMEN

Covalently closed circular DNA (cccDNA) is a crucial intermediate in the replication of hepadnaviruses. We inhibited the replication of duck hepatitis B virus in congenitally infected ducks with a combination of lamivudine and a dideoxyguanosine prodrug. Inhibition of viral replication should prevent renewal of the cccDNA pool, and its decay was measured in liver biopsy samples collected over a 5-month period. In three ducks, the cccDNA pools declined exponentially, with half-lives ranging from 35 to 57 days. In two others, the pools declined exponentially for about 70 days but then stabilized at about 6 copies/diploid genome. The selection of drug-resistant virus mutants is an unlikely explanation for this unexpected stabilization of cccDNA levels. Liver sections stained for the cell division marker PCNA showed that animals in which cccDNA loss was continuous had significantly greater numbers of PCNA-positive nuclei than did those animals in which cccDNA levels had plateaued.


Asunto(s)
ADN Circular/fisiología , ADN Viral/fisiología , Virus de la Hepatitis B del Pato/fisiología , Hepatitis Viral Animal/virología , Replicación Viral/efectos de los fármacos , Animales , Antivirales/farmacología , Arabinofuranosil Uracilo/análogos & derivados , Arabinofuranosil Uracilo/farmacología , Patos , Femenino , Semivida , Infecciones por Hepadnaviridae/virología , Virus de la Hepatitis B del Pato/efectos de los fármacos , Virus de la Hepatitis B del Pato/genética , Hígado/virología , Masculino , Datos de Secuencia Molecular
14.
J Virol ; 76(3): 1213-23, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11773397

RESUMEN

Administration of either lamivudine (2'-deoxy-3'-thiacytidine) or L-FMAU (2'-fluoro-5-methyl-beta-L-arabinofuranosyluracil) to woodchucks chronically infected with woodchuck hepatitis virus (WHV) induces a transient decline in virus titers. However, within 6 to 12 months, virus titers begin to increase towards pretreatment levels. This is associated with the emergence of virus strains with mutations of the B and C regions of the viral DNA polymerase (T. Zhou et al., Antimicrob. Agents Chemother. 43:1947-1954, 1999; Y. Zhu et al., J. Virol. 75:311-322, 2001). The present study was carried out to determine which of the mutants that we have identified conferred resistance to lamivudine and/or to L-FMAU. When inserted into a laboratory strain of WHV, each of the mutations, or combinations of mutations, of regions B and C produced a DNA replication-competent virus and typically conferred resistance to both nucleoside analogs in cell culture. Sequencing of the polymerase active site also occasionally revealed other mutations, but these did not appear to contribute to drug resistance. Moreover, in transfected cells, most of the mutants synthesized viral DNA nearly as efficiently as wild-type WHV. Computational models suggested that persistence of several of the WHV mutants as prevalent species in the serum and, by inference, liver for up to 6 months following drug withdrawal required a replication efficiency of at least 10 to 30% of that of the wild type. However, their delayed emergence during therapy suggested replication efficiency in the presence of the drug that was still well below that of wild-type WHV in the absence of the drug.


Asunto(s)
Antivirales/farmacología , Arabinofuranosil Uracilo/farmacología , ADN Polimerasa Dirigida por ADN/fisiología , Farmacorresistencia Viral Múltiple , Farmacorresistencia Viral , Virus de la Hepatitis B de la Marmota/enzimología , Lamivudine/farmacología , Secuencia de Aminoácidos , Animales , Arabinofuranosil Uracilo/análogos & derivados , Sitios de Unión , ADN Polimerasa Dirigida por ADN/genética , Genes Virales/fisiología , Virus de la Hepatitis B de la Marmota/efectos de los fármacos , Virus de la Hepatitis B de la Marmota/genética , Virus de la Hepatitis B de la Marmota/fisiología , Humanos , Marmota , Datos de Secuencia Molecular , Mutagénesis , Mutagénesis Insercional , Homología de Secuencia de Aminoácido , Factores de Tiempo , Células Tumorales Cultivadas , Replicación Viral
15.
Antimicrob Agents Chemother ; 45(9): 2495-501, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11502520

RESUMEN

Prolonged treatment of chronic hepatitis B virus (HBV) infection with lamivudine ([-]-beta-L-2',3'-dideoxy-3' thiacytidine) or famciclovir may select for viral mutants that are drug resistant due to point mutations in the polymerase gene. Determining whether such HBV mutants are sensitive to new antiviral agents is therefore important. We used a transient transfection system to compare the sensitivities of wild-type HBV and four lamivudine- and/or famciclovir-resistant HBV mutants to adefovir [9-(2-phosphonyl-methoxyethyl)-adenine; PMEA] and the nucleoside analogues (-)-beta-D-2, 6-diaminopurine dioxolane (DAPD) and 2'-fluoro-5-methyl-beta-L-arabinofuranosyluracil (L-FMAU). The drug-resistant mutants contained amino acid substitutions in the polymerase protein. We found that the M550I and M550V plus L526M substitutions, which confer lamivudine resistance, did not confer cross-resistance to adefovir or DAPD, but conferred cross-resistance to L-FMAU. The M550V substitution in isolation conferred a similar phenotype to M550I, except that it did not confer significant resistance to L-FMAU. The L526M substitution, which is associated with famciclovir resistance, conferred cross-resistance to L-FMAU but not to adefovir or DAPD. Inhibition of HBV secretion by DAPD, L-FMAU, and adefovir did not always correlate with inhibition of the generation of intracellular HBV replicative intermediates, suggesting that these analogs may preferentially inhibit specific stages of the viral replication cycle.


Asunto(s)
Antivirales/farmacología , Arabinofuranosil Uracilo/farmacología , Dioxolanos/farmacología , Virus de la Hepatitis B/efectos de los fármacos , Organofosfonatos , Nucleósidos de Purina/farmacología , Adenina/análogos & derivados , Adenina/farmacología , Arabinofuranosil Uracilo/análogos & derivados , Farmacorresistencia Microbiana , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/fisiología , Humanos , Lamivudine/farmacología , Pruebas de Sensibilidad Microbiana , Mutagénesis Sitio-Dirigida , Transfección , Células Tumorales Cultivadas , Replicación Viral
16.
Biochem Pharmacol ; 61(6): 727-32, 2001 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-11266658

RESUMEN

Introduction of a bulky lipophilic acyl entity at the 2'-OH position of both 1-beta-D-arabinofuranosylthymine (araT) and (E)-5-(2-bromovinyl)-1-beta-D-arabinofuranosyluracil (BVaraU), consistently resulted in a marked ( approximately 10-fold) increase in the inhibitory activity of these new arabinosyl nucleoside analogues for the mitochondrial thymidine kinase (TK-2)-catalysed conversion of 2 microM [methyl-(3)H]dThd to [methyl-(3)H]dTMP. The most potent derivatives were inhibitory to [methyl-(3)H]dThd phosphorylation by TK-2 within the lower micromolar concentration range. Substitution of the arabinosyl nucleoside derivatives with the acyl groups also dramatically increased the selectivity of these compounds. The inhibitory activity of araT and BVaraU to dThd phosphorylation by other related nucleoside kinases, including herpes simplex virus type 1 TK, varicella-zoster virus TK, and cytosolic TK-1, was completely annihilated upon 2'-O-acyl substitution (IC(50) > or = 1000 microM). Kinetic analysis revealed purely competitive inhibition of 2'-O-acyl-BVaraU against TK-2-catalysed thymidine phosphorylation (K(i)/K(m): 2.3). However, 2'-O-acyl-BVaraU was extremely poorly converted to the corresponding arabinosyl nucleoside 5'-monophosphate by TK-2 as revealed by [gamma-(32)P]phosphate transfer studies from [gamma-(32)P]ATP. Thus, the 2'-O-acyl derivatives of BVaraU did not behave as substrates, but rather as potent and highly selective inhibitors of TK-2. This is the first report on such a highly selective arabinosyl nucleoside inhibitor of mitochondrial TK-2, and opens perspectives for the rational design of selective mitochondrial TK-2 inhibitors.


Asunto(s)
Arabinofuranosil Uracilo/análogos & derivados , Arabinofuranosil Uracilo/farmacología , Arabinonucleósidos/farmacología , Mitocondrias/efectos de los fármacos , Timidina Quinasa/antagonistas & inhibidores , Timidina/análogos & derivados , Timidina/farmacología , Adenosina Trifosfato/metabolismo , Antineoplásicos/farmacología , Antivirales/farmacología , Arabinofuranosil Uracilo/química , Arabinonucleósidos/química , Unión Competitiva , Humanos , Cinética , Mitocondrias/enzimología , Mitocondrias/metabolismo , Radioisótopos de Fósforo , Fosforilación/efectos de los fármacos , Timidina/química , Timidina/metabolismo , Timidina Quinasa/metabolismo , Tritio
17.
Cancer Lett ; 162 Suppl: S33-S37, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11164188

RESUMEN

Several types of virus were found to have a strong association with different types of cancers. Thus, a selective antiviral compound without toxicity upon long-term usage will be useful not only for the treatment of viral diseases but also for the prevention or the delayed onset of those cancers, which have a strong association with viruses. L(-)Nucleoside analogs were discovered recently in my laboratory as an important class of antiviral and anticancer chemical entities. L(-)SddC (3TC, Lamivudine), FTC, Fd4C, and L(-)FMAU are potent anti-HBV compounds with different pharmacological profiles. These compounds may be useful in the prevention or delayed onset of hepatocellular carcinoma associated with HBV. L(-)I-OddU is the most potent anti-Epstein-Barr Virus (EBV) compound without cytotoxicity and animal toxicity upon long-term dosing which gives the pharmacological levels of the drug in plasma. This compound may have the potential to prevent B-cell lymphoma associated with patients undergoing organ transplants in addition to its potential use for the treatment of EBV infection.


Asunto(s)
Antivirales/farmacología , Neoplasias/tratamiento farmacológico , Nucleósidos/farmacología , Virosis/prevención & control , Zalcitabina/análogos & derivados , Animales , Antivirales/química , Arabinofuranosil Uracilo/análogos & derivados , Arabinofuranosil Uracilo/farmacología , Dioxolanos/farmacología , Virus de la Hepatitis B/efectos de los fármacos , Herpesvirus Humano 4/efectos de los fármacos , Humanos , Neoplasias/etiología , Nucleósidos/química , Estereoisomerismo , Relación Estructura-Actividad , Uracilo/análogos & derivados , Uracilo/farmacología , Virosis/complicaciones , Virosis/virología , Zalcitabina/farmacología
18.
Circulation ; 102(9): 948-50, 2000 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-10961956

RESUMEN

BACKGROUND: Gene therapy is a promising approach for the treatment of cardiac diseases. Coexpression of therapeutic genes with a suitable marker gene would allow for the noninvasive imaging of successful gene transfer and expression via radiolabeled marker substrates. In the present study, such an approach was first applied to cardiac tissue. METHODS AND RESULTS: The combination of the herpesvirus thymidine kinase reporter gene (HSV1-tk) and radiolabeled 2'-fluoro-2'-deoxy-5-iodo-1-beta-D-arabinofuranosyluracil (FIAU) was evaluated. H9c2 rat cardiomyoblasts were infected in vitro with a replication-defective HSV1-tk-containing adenovirus and a negative control virus. The intracellular uptake of [(14)C]FIAU increased with increasing multiplicity of infection and with time after infection. Uptake in negative controls remained <15% of positive controls. Additionally, vectors were applied intramyocardially in Wistar rats. The marker substrate [(125)I]FIAU was injected intravenously 3 days later, and animals were killed after 24 hours. Autoradiographically, regional transgene expression was clearly identified in animals receiving the adenovirus containing HSV1-tk (3. 4+/-2.2-fold increase of radioactivity at vector administration site compared with remote myocardium), whereas nonspecific uptake in negative controls was low (<10% of positive controls). CONCLUSIONS: Using an adenoviral vector, HSV1-tk can be successfully expressed in cardiac cells in vitro and in vivo, yielding high uptake of radiolabeled FIAU. The results suggest that imaging transgene expression in the heart is feasible and may be used to monitor gene therapy noninvasively.


Asunto(s)
Adenoviridae/genética , Arabinofuranosil Uracilo/análogos & derivados , Arabinofuranosil Uracilo/farmacología , Genes Reporteros , Miocardio/metabolismo , Radiofármacos/farmacología , Animales , Autorradiografía , Radioisótopos de Carbono , Línea Celular , Expresión Génica , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos , Inyecciones Intravenosas , Radioisótopos de Yodo , Miocardio/citología , Miocardio/enzimología , Ratas , Ratas Wistar , Simplexvirus/enzimología , Simplexvirus/genética , Timidina Quinasa/genética , Factores de Tiempo
19.
Clin Cancer Res ; 5(8): 1976-81, 1999 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10473074

RESUMEN

Most tumors are resistant to therapy by thymidylate synthase (TS) inhibitors due to their high levels of TS. Instead of inhibiting TS, we hypothesized that it was possible to use this enzyme to activate suicide prodrugs (deoxyuridine analogues) to more toxic species (thymidine analogues). Tumors with high levels of TS could be particularly sensitive to deoxyuridine analogues because they would be more efficient in producing the toxic methylated species. Furthermore, the accumulation of methylated species within tumors could be visualized externally if a tracer dose of the deoxyuridine analogue was tagged with an isotope, preferably a positron emitter, such as 18F. Higher accumulation of isotope indicates higher activity of TS and lower sensitivity of the tumor to TS inhibitors, but perhaps more sensitivity to therapy with deoxyuridine analogues as suicide prodrugs. 2'-F-ara-deoxyuridine (FAU) was used as a prototype to demonstrate these concepts experimentally. FAU readily entered cells and was phosphorylated, methylated, and subsequently incorporated into cellular DNA. Among different cell lines, FAU produced varying degrees of growth inhibition. Greater DNA incorporation (e.g., for CEM and U-937 cells) was reflected as increased toxicity. FAU produced less DNA incorporation in Raji or L1210 cells, and growth rate was minimally decreased. As the first demonstration that cells with high levels of TS activity can be more vulnerable to therapy than cells with low TS activity, this preliminary work suggests a new therapeutic approach for common human tumors that were previously resistant. Furthermore, it appears that the TS activity of tumors could be noninvasively imaged in situ by tracer doses of [18F]FAU and that this phenotypic information could guide patient therapy.


Asunto(s)
Floxuridina/metabolismo , Neoplasias/metabolismo , Profármacos/metabolismo , Timidilato Sintasa/metabolismo , Animales , Arabinofuranosil Uracilo/análogos & derivados , Arabinofuranosil Uracilo/metabolismo , Arabinofuranosil Uracilo/farmacología , División Celular/efectos de los fármacos , ADN/efectos de los fármacos , ADN/metabolismo , Radioisótopos de Flúor , Humanos , Metilación , Ratones , Neoplasias/patología , Fosforilación , Profármacos/farmacocinética , Células Tumorales Cultivadas
20.
J Med Chem ; 42(17): 3243-50, 1999 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-10464011

RESUMEN

Continuing our studies on ribonucleotide reductase (RNR) mechanism-based inhibitors, we have now prepared the diphosphates (DP) of 2'-O-allyl-1-beta-D-arabinofuranosyl-uracil and -cytosine and 2'-O-allyl-9-beta-D-arabinofuranosyl-adenine and evaluated their inhibitory activity against recombinant murine RNR. 2'-O-Allyl-araUDP proved to be inhibitory to RNR at an IC(50) of 100 microM, whereas 2'-O-allyl-araCDP was only marginally active (IC(50) 1 mM) and 2'-O-allyl-araADP was completely inactive. The susceptibility of the parent nucleosides to phosphorylation by thymidine kinase and 2'-deoxycytidine kinase was also investigated, and all nucleosides proved to be poor substrates for the above-cited kinases. Moreover, prodrugs of 2'-O-allyl-araU and -araC monophosphates, namely 2'-O-allyl-5'-(phenylethoxy-L-alanyl phosphate)-araU and -araC, were prepared and tested against tumor cell proliferation but proved to be inactive. A molecular modeling study has been conducted in order to explain our results. The data confirm that for both the natural and analogue nucleoside diphosphates, the principal determinant interaction with the active site of RNR is with the diphosphate group, which forms strong hydrogen bonds with Glu623, Thr624, Ser625, and Thr209. Our findings indicate that the poor phosphorylation may represent an explanation for the lack of marked in vitro cytostatic activity of the test compounds.


Asunto(s)
Antineoplásicos/síntesis química , Arabinofuranosil Uracilo/síntesis química , Citarabina/síntesis química , Ribonucleótido Reductasas/antagonistas & inhibidores , Vidarabina/síntesis química , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Arabinofuranosil Uracilo/química , Arabinofuranosil Uracilo/farmacología , Citarabina/química , Citarabina/farmacología , Desoxicitidina Quinasa/química , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Ratones , Modelos Moleculares , Fosforilación , Profármacos/síntesis química , Profármacos/química , Profármacos/farmacología , Proteínas Recombinantes/antagonistas & inhibidores , Relación Estructura-Actividad , Timidina Quinasa/química , Células Tumorales Cultivadas , Vidarabina/química , Vidarabina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA