Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
DNA Repair (Amst) ; 57: 116-124, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28732309

RESUMEN

Zebularine is a second-generation, highly stable hydrophilic inhibitor of DNA methylation with oral bioavailability that preferentially target cancer cells. It acts primarily as a trap for DNA methyl transferases (DNMTs) protein by forming covalent complexes between DNMT protein and zebularine-substrate DNA. It's well documented that replication-blocking DNA lesions can cause replication fork collapse and thereby to the formation of DNA double-strand breaks (DSB). DSB are dangerous lesions that can lead to potentially oncogenic genomic rearrangements or cell death. The two major pathways for repair of DSB are non-homologous end joining (NHEJ) and homologous recombination (HR). Recently, multiple functions for the HR machinery have been identified at arrested forks. Here we investigate in more detail the importance of the lesions induced by zebularine in terms of DNA damage and cytotoxicity as well as the role of HR in the repair of these lesions. When we examined the contribution of NHEJ and HR in the repair of DSB induced by zebularine we found that these breaks were preferentially repaired by HR. Also we show that the production of DSB is dependent on active replication. To test this, we determined chromosome damage by zebularine while transiently inhibiting DNA synthesis. Here we report that cells deficient in single-strand break (SSB) repair are hypersensitive to zebularine. We have observed more DSB induced by zebularine in XRCC1 deficient cells, likely to be the result of conversion of SSB into toxic DSB when encountered by a replication fork. Furthermore we demonstrate that HR is required for the repair of these breaks. Overall, our data suggest that zebularine induces replication-dependent DSB which are preferentially repaired by HR.


Asunto(s)
Citidina/análogos & derivados , Roturas del ADN de Doble Cadena , Replicación del ADN/efectos de los fármacos , Reparación del ADN por Recombinación , Animales , Línea Celular , Cricetulus/genética , Cricetulus/metabolismo , Citidina/farmacología , Citidina/toxicidad , ADN/efectos de los fármacos , ADN/metabolismo , Roturas del ADN de Cadena Simple , Metilasas de Modificación del ADN/antagonistas & inhibidores , Reparación del ADN , Femenino
2.
Antiviral Res ; 143: 151-161, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28412183

RESUMEN

Recent cases of severe toxicity during clinical trials have been associated with antiviral ribonucleoside analogs (e.g. INX-08189 and balapiravir). Some have hypothesized that the active metabolites of toxic ribonucleoside analogs, the triphosphate forms, inadvertently target human mitochondrial RNA polymerase (POLRMT), thus inhibiting mitochondrial RNA transcription and protein synthesis. Others have proposed that the prodrug moiety released from the ribonucleoside analogs might instead cause toxicity. Here, we report the mitochondrial effects of several clinically relevant and structurally diverse ribonucleoside analogs including NITD-008, T-705 (favipiravir), R1479 (parent nucleoside of balapiravir), PSI-7851 (sofosbuvir), and INX-08189 (BMS-986094). We found that efficient substrates and chain terminators of POLRMT, such as the nucleoside triphosphate forms of R1479, NITD-008, and INX-08189, are likely to cause mitochondrial toxicity in cells, while weaker chain terminators and inhibitors of POLRMT such as T-705 ribonucleoside triphosphate do not elicit strong in vitro mitochondrial effects. Within a fixed 3'-deoxy or 2'-C-methyl ribose scaffold, changing the base moiety of nucleotides did not strongly affect their inhibition constant (Ki) against POLRMT. By swapping the nucleoside and prodrug moieties of PSI-7851 and INX-08189, we demonstrated that the cell-based toxicity of INX-08189 is mainly caused by the nucleoside component of the molecule. Taken together, these results show that diverse 2' or 4' mono-substituted ribonucleoside scaffolds cause mitochondrial toxicity. Given the unpredictable structure-activity relationship of this ribonucleoside liability, we propose a rapid and systematic in vitro screen combining cell-based and biochemical assays to identify the early potential for mitochondrial toxicity.


Asunto(s)
Antivirales/toxicidad , Mitocondrias/efectos de los fármacos , Ribonucleósidos/química , Ribonucleósidos/toxicidad , Adenosina/análogos & derivados , Amidas/toxicidad , Línea Celular/efectos de los fármacos , Citidina/análogos & derivados , Citidina/toxicidad , ARN Polimerasas Dirigidas por ADN/efectos de los fármacos , Guanosina Monofosfato/análogos & derivados , Guanosina Monofosfato/toxicidad , Humanos , Concentración 50 Inhibidora , Proteínas Mitocondriales/metabolismo , Nucleósidos/toxicidad , Profármacos/farmacología , Biosíntesis de Proteínas/efectos de los fármacos , Pirazinas/toxicidad , ARN/metabolismo , ARN Mitocondrial , Sofosbuvir/toxicidad , Relación Estructura-Actividad , Sitio de Iniciación de la Transcripción/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
3.
Proc Natl Acad Sci U S A ; 113(48): 13774-13779, 2016 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-27849610

RESUMEN

Routine dietary consumption of foods that contain aflatoxins is the second leading cause of environmental carcinogenesis worldwide. Aflatoxin-driven mutagenesis is initiated through metabolic activation of aflatoxin B1 (AFB1) to its epoxide form that reacts with N7 guanine in DNA. The resulting AFB1-N7-dG adduct undergoes either spontaneous depurination or imidazole-ring opening yielding formamidopyrimidine AFB1 (AFB1-Fapy-dG). Because this latter adduct is known to persist in human tissues and contributes to the high frequency G-to-T mutation signature associated with many hepatocellular carcinomas, we sought to establish the identity of the polymerase(s) involved in processing this lesion. Although our previous biochemical analyses demonstrated the ability of polymerase ζ (pol ζ) to incorporate an A opposite AFB1-Fapy-dG and extend from this mismatch, biological evidence supporting a unique role for this polymerase in cellular tolerance following aflatoxin exposure has not been established. Following challenge with AFB1, survival of mouse cells deficient in pol ζ (Rev3L-/-) was significantly reduced relative to Rev3L+/- cells or Rev3L-/- cells complemented through expression of the wild-type human REV3L. Furthermore, cell-cycle progression of Rev3L-/- mouse embryo fibroblasts was arrested in late S/G2 following AFB1 exposure. These Rev3L-/- cells showed an increase in replication-dependent formation of γ-H2AX foci, micronuclei, and chromosomal aberrations (chromatid breaks and radials) relative to Rev3L+/- cells. These data suggest that pol ζ is essential for processing AFB1-induced DNA adducts and that, in its absence, cells do not have an efficient backup polymerase or a repair/tolerance mechanism facilitating survival.


Asunto(s)
Carcinoma Hepatocelular/genética , Proteínas de Unión al ADN/genética , ADN Polimerasa Dirigida por ADN/genética , Neoplasias Hepáticas/genética , Aflatoxina B1/análogos & derivados , Aflatoxina B1/genética , Aflatoxina B1/toxicidad , Aflatoxinas/toxicidad , Animales , Carcinoma Hepatocelular/inducido químicamente , Carcinoma Hepatocelular/patología , Supervivencia Celular/efectos de los fármacos , Aberraciones Cromosómicas/efectos de los fármacos , Citidina/análogos & derivados , Citidina/genética , Citidina/toxicidad , Aductos de ADN/efectos de los fármacos , Aductos de ADN/genética , Daño del ADN/efectos de los fármacos , Reparación del ADN/genética , ADN Polimerasa Dirigida por ADN/química , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Neoplasias Hepáticas/inducido químicamente , Neoplasias Hepáticas/patología , Ratones , Mutagénesis/efectos de los fármacos , Mutagénesis/genética , Mutación
4.
DNA Repair (Amst) ; 9(2): 153-60, 2010 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-20005183

RESUMEN

Most bacteria, including Escherichia coli, lack an enzyme that can phosphorylate deoxycytidine and its analogs. Consequently, most studies of toxicity and mutagenicity of cytosine analogs use ribonucleosides such as 5-azacytidine (AzaC) and zebularine (Zeb) instead of their deoxynucleoside forms, 5-aza-2'-deoxycytidine (AzadC) and 2'-deoxy-zebularine (dZeb). The former analogs are incorporated into both RNA and DNA creating complex physiological responses in cells. To circumvent this problem, we introduced into E. coli the Drosophila deoxynucleoside kinase (Dm-dNK), which has a relaxed substrate specificity, and tested these cells for sensitivity to AzadC and dZeb. We find that Dm-dNK expression increases substantially sensitivity of cells to these analogs and dZeb is very mutagenic in cells expressing the kinase. Furthermore, toxicity of dZeb in these cells requires DNA mismatch correction system suggesting a mechanism for its toxicity and mutagenicity. The fluorescence properties of dZeb were used to quantify the amount of this analog incorporated into cellular DNA of mismatch repair-deficient cells expressing Dm-dNK and the results showed that in a mismatch correction-defective strain a high percentage of DNA bases may be replaced with the analog without long term toxic effects. This study demonstrates that the mechanism by which Zeb and dZeb cause cell death is fundamentally different than the mechanism of toxicity of AzaC and AzadC. It also opens up a new way to study the mechanism of action of deoxycytidine analogs that are used in anticancer chemotherapy.


Asunto(s)
Desoxicitidina/análogos & derivados , Desoxicitidina/toxicidad , Drosophila melanogaster/enzimología , Escherichia coli/efectos de los fármacos , Mutagénesis/efectos de los fármacos , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Animales , Antineoplásicos/farmacología , Azacitidina/química , Azacitidina/toxicidad , Citidina/análogos & derivados , Citidina/química , Citidina/toxicidad , ADN/metabolismo , Metilación de ADN/efectos de los fármacos , Reparación de la Incompatibilidad de ADN/efectos de los fármacos , Drosophila melanogaster/efectos de los fármacos , Fluorescencia , Pruebas de Sensibilidad Microbiana , Fosforilación/efectos de los fármacos
5.
J Med Chem ; 52(17): 5394-407, 2009 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-19725579

RESUMEN

The application of a phosphoramidate prodrug approach to 2'-C-methylcytidine (NM107), the first nucleoside inhibitor of the hepatitis C virus (HCV) NS5B polymerase, is reported. 2'-C-Methylcytidine, as its valyl ester prodrug (NM283), was efficacious in reducing the viral load in patients infected with HCV. Several of the phosphoramidates prepared demonstrated a 10- to 200-fold superior potency with respect to the parent nucleoside in the cell-based replicon assay. This is due to higher levels of 2'-C-methylcytidine triphosphate in the cells. These prodrugs are efficiently activated and converted to the triphosphate in hepatocytes of several species. Our SAR studies ultimately led to compounds that gave high levels of NTP in hamster and rat liver after subcutaneous dosing and that were devoid of the toxic phenol moiety usually found in ProTides.


Asunto(s)
Amidas/metabolismo , Amidas/uso terapéutico , Antivirales/metabolismo , Citidina/análogos & derivados , Hepatitis C/tratamiento farmacológico , Ácidos Fosfóricos/metabolismo , Ácidos Fosfóricos/uso terapéutico , Profármacos/metabolismo , Profármacos/uso terapéutico , Amidas/farmacología , Amidas/toxicidad , Animales , Antivirales/farmacología , Antivirales/uso terapéutico , Antivirales/toxicidad , Línea Celular , Citidina/metabolismo , Citidina/farmacología , Citidina/uso terapéutico , Citidina/toxicidad , Hepacivirus/efectos de los fármacos , Hepacivirus/fisiología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hepatocitos/virología , Humanos , Ácidos Fosfóricos/farmacología , Ácidos Fosfóricos/toxicidad , Polifosfatos/metabolismo , Profármacos/farmacología , Profármacos/toxicidad , Relación Estructura-Actividad , Replicación Viral/efectos de los fármacos
6.
Leuk Res ; 31(11): 1545-51, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17459472

RESUMEN

The experimental cytotoxic drug cyclopentenyl cytosine (CPEC) is a non-competitive inhibitor of the enzyme cytidine triphosphate (CTP) synthethase. We evaluated the in vitro and in vivo antitumor activity of CPEC on human acute lymphoblastic leukemia (ALL) cell lines. CPEC displayed anti-leukemic activity with IC50 (after 3 days of incubation) ranging from 6 to 15 nM. Subsequently the in vivo activity of CPEC against primary human ALL was evaluated in a xenogeneic model of human ALL using NOD/scid mice inoculated with primary human ALL cells. In the model, only a marginal anti-leukemic activity was observed at 1.5 mg kg(-1) (5 days per week) and 5 mg kg(-1) (2 days per week), however, this activity was associated with severe systemic toxicity. The observed toxicity was not specific for the NOD/scid model, as toxicity at comparable treatment intensity was also observed in Balb/c mice. In conclusion, although CPEC showed antitumor activity against human ALL cells in vitro, its activity in the in vivo human leukemia model was only marginal and accompanied by severe toxicity.


Asunto(s)
Antineoplásicos/toxicidad , Citidina/análogos & derivados , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Animales , Citidina/toxicidad , Humanos , Cariotipificación , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones SCID , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética
7.
Antimicrob Agents Chemother ; 49(7): 2589-97, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15980324

RESUMEN

Beta-D-2',3'-dideoxy-3'-oxa-5-fluorocytidine (D-FDOC) is an effective inhibitor of human immunodeficiency virus 1 (HIV-1) and HIV-2, simian immunodeficiency virus, and hepatitis B virus (HBV) in vitro. The purpose of this study was to evaluate the intracellular metabolism of d-FDOC in human hepatoma (HepG2), human T-cell lymphoma (CEM), and primary human peripheral blood mononuclear (PBM) cells by using tritiated compound. By 24 h, the levels of D-FDOC-triphosphate (D-FDOC-TP) were 2.8 +/- 0.4, 6.7 +/- 2.3, and 2.0 +/- 0.1 pmol/10(6) cells in HepG2, CEM, and primary human PBM cells, respectively. Intracellular D-FDOC-TP concentrations remained greater than the 50% inhibitory concentration for HIV-1 reverse transcriptase for up to 24 h after removal of the drug from cell cultures. In addition to d-FDOC-monophosphate (D-FDOC-MP), -diphosphate (D-FDOC-DP), and -TP, D-FDOC-DP-ethanolamine and d-FDOC-DP-choline were detected in all cell extracts as major intracellular metabolites. D-FDOC was not a substrate for Escherichia coli thymidine phosphorylase. No toxicity was observed in mice given D-FDOC intraperitoneally for 6 days up to a dose of 100 mg/kg per day. Pharmacokinetic studies in rhesus monkeys indicated that D-FDOC has a t(1/2) of 2.1 h in plasma and an oral bioavailability of 38%. The nucleoside was excreted unchanged primary in the urine, and no metabolites were detected in plasma or urine. These results suggest that further safety and pharmacological studies are warranted to assess the potential of this nucleoside for the treatment of HIV- and HBV-infected individuals.


Asunto(s)
Antivirales , Citidina , Citidina/análogos & derivados , Animales , Antivirales/administración & dosificación , Antivirales/metabolismo , Antivirales/farmacocinética , Antivirales/toxicidad , Línea Celular Tumoral , Citidina/administración & dosificación , Citidina/metabolismo , Citidina/farmacocinética , Citidina/toxicidad , Escherichia coli/enzimología , Femenino , Humanos , Leucocitos Mononucleares/metabolismo , Macaca mulatta , Masculino , Ratones , Timidina Fosforilasa/metabolismo
8.
Arch Toxicol ; 79(5): 268-76, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15902424

RESUMEN

The experimental anticancer drug cyclopentenyl cytosine (CPEC) was associated with cardiotoxicity in a phase I study. The aim of the present study was twofold; first we investigated whether the observed effects could be reproduced in in-vitro and in-vivo rat models. Second, we intended to investigate the underlying mechanism of the possible cardiotoxicity of CPEC. Effects on frequency and contractility were studied on the isolated atria of 18 male Wistar rats. Atria were incubated with 0.1 mmol L(-1) (n = 6) or 1 mmol L(-1) (n = 6) CPEC for 1.5 h and compared with control atria (incubation with buffer solution, n = 6). The cardiac apoptosis-inducing potential was studied in-vivo on 66 rats by 99mTc-Annexin V scintigraphy, followed by postmortem determination of radioactivity in tissues, histological confirmation with the TUNEL assay (late-phase apoptosis), and immunohistochemical staining for cleaved caspase 3 and cytochrome C (early-phase apoptosis). Serum levels of the necrotic cardiomyopathy marker troponin T were also determined. No effect on heart frequency was found in the isolated atria after CPEC treatment. A trend towards a decrease of contraction force was observed. However, the differences were not statistically significant. 99mTc-Annexin V scintigraphy showed no increase in cardiac uptake ratio upon CPEC treatment in the in-vivo rat model, which was confirmed by determination of radioactivity in heart versus blood ratios. At each section a few individual isolated late apoptotic cells (< 5) could be identified by the TUNEL assay in the highest CPEC dose group (90 mg kg(-1)) but not in controls or in rats treated with 60 mg kg(-1) CPEC. Staining for the early apoptosis markers cleaved caspase 3 and cytochrome C did not reveal any significant differences between treated and control rats. Cardiac troponin T levels were not increased after CPEC treatment. CPEC does not affect heart frequency or contraction force in our cardiotoxicity models. Moreover, we did not find an indication of CPEC-induced apoptosis in heart tissue.


Asunto(s)
Antineoplásicos/toxicidad , Cardiomiopatías/inducido químicamente , Citidina/análogos & derivados , Corazón/efectos de los fármacos , Animales , Anexina A5/metabolismo , Apoptosis/efectos de los fármacos , Cardiomiopatías/sangre , Cardiomiopatías/patología , Citidina/toxicidad , Modelos Animales de Enfermedad , Corazón/diagnóstico por imagen , Atrios Cardíacos/diagnóstico por imagen , Atrios Cardíacos/efectos de los fármacos , Atrios Cardíacos/patología , Atrios Cardíacos/fisiopatología , Etiquetado Corte-Fin in Situ , Masculino , Contracción Miocárdica/efectos de los fármacos , Miocardio/patología , Compuestos de Organotecnecio , Ratas , Ratas Wistar , Tomografía Computarizada de Emisión de Fotón Único , Troponina T/sangre
9.
Jpn J Cancer Res ; 92(3): 343-51, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11267946

RESUMEN

We examined the effects of dosage schedule on antitumor activity in vitro and in vivo to determine the optimal administration schedule for a new nucleoside antimetabolite 1-(3-C-ethynyl-beta-D-ribo-pentofuranosyl)cytosine (ECyd, TAS-106). The cytotoxicity of TAS-106 in vitro against human tumors was evaluated at three drug exposure periods. TAS-106 exhibited fairly potent cytotoxicity even with 4 h exposure, and nearly equivalent and sufficiently potent cytotoxicity with 24 and 72 h exposures. These results suggest that long-term exposure to TAS-106 will not be required to achieve maximal cytotoxicity. The antitumor activity of TAS-106 in vivo was compared in nude rat models bearing human tumors on three administration schedules, once weekly, 3 times weekly, and 5 times weekly for 2 or 4 consecutive weeks. TAS-106 showed strong antitumor activity without serious toxicity on all three schedules, but the antitumor activity showed no obvious schedule-dependency in these models. When tumor-bearing nude rats were given a single i.v. dose of [(3)H]TAS-106, tumor tissue radioactivity tended to remain high for longer periods of time as compared to the radioactivity in various normal tissues. Furthermore, when the metabolism of TAS-106 in the tumor was examined, it was found that TAS-106 nucleotides (including the active metabolite, the triphosphate of TAS-106) were retained at high concentrations for prolonged periods. These pharmacodynamic features of TAS-106 may explain the strong antitumor activity without serious toxicity, observed on intermittent administration schedules, in nude rat models with human tumors. We therefore consider TAS-106 to be a promising compound which merits further investigation in patients with solid tumors.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacocinética , Antimetabolitos Antineoplásicos/uso terapéutico , Supervivencia Celular/efectos de los fármacos , Citidina/farmacocinética , Citidina/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Gástricas/tratamiento farmacológico , Animales , Antimetabolitos Antineoplásicos/toxicidad , Neoplasias de la Mama , Neoplasias del Colon , Citidina/análogos & derivados , Citidina/toxicidad , Femenino , Humanos , Masculino , Neoplasias Pancreáticas , Ratas , Ratas Endogámicas F344 , Ratas Desnudas , Distribución Tisular , Trasplante Heterólogo , Células Tumorales Cultivadas
10.
Nucleic Acids Symp Ser ; (42): 133-4, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10780415

RESUMEN

The molecular mechanism of cell death induced by 1-(3-C-ethynyl-beta-D-ribo-pentofuranosyl)cytosine (ECyd: Figure 1), a potent inhibitor of RNA synthesis, was performed using mouse mammary tumor FM3A cells and human fibrosarcoma HT1080 cells. ECyd induced the characteristics of apoptosis on these cells, such as morphological changes, DNA fragmentations (Figure 2), and caspase-3-like protease activation. General caspases inhibitor (Z-Asp-CH2-DCB) inhibited these changes and cell death. We also found that ECyd induced DNA and 28S ribosomal RNA (rRNA) fragmentations. Though the mechanisms of rRNA fragmentations haven't revealed, it suggests that translational function of the treated cells should be disturbed. These results indicate that antitumor mechanism of ECyd are characteristics of apoptosis on the cells and rRNA fragmentations is one of the death events resulted inhibition of RNA synthesis.


Asunto(s)
Antimetabolitos Antineoplásicos/toxicidad , Citidina/análogos & derivados , Inhibidores de la Síntesis del Ácido Nucleico/toxicidad , Transcripción Genética/efectos de los fármacos , Animales , Caspasa 3 , Caspasas/metabolismo , Citidina/toxicidad , Fragmentación del ADN , ADN de Neoplasias/efectos de los fármacos , Activación Enzimática , Femenino , Fibrosarcoma , Humanos , Neoplasias Mamarias Experimentales , Ratones , ARN Ribosómico 28S/genética , ARN Ribosómico 28S/efectos de la radiación , Células Tumorales Cultivadas
11.
Nucleic Acids Symp Ser ; (37): 137-8, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-9586037

RESUMEN

The cytotoxic mechanisms of 1-(3-C-ethynyl-beta-D-ribo-pentofuranosyl)cytosine (ECyd) and 1-(3-C-ethynyl-beta-D-ribo-pentofuranosyl)uracil (EUrd) were studied with mouse mammary tumor FM3A cells and human fibrosarcoma HT1080 cells. ECyd and EUrd are converted to ECyd 5'-triphosphate (ECTP) in the cells. ECTP has also outstanding stability in the cells; the half life of ECTP in FM3A cells was more than 3 days. The metabolisms and mechanisms of these analogues may play a key role in a potent antitumor activities against slow-growing solid tumors.


Asunto(s)
Antimetabolitos Antineoplásicos/toxicidad , Citidina/análogos & derivados , Uridina/análogos & derivados , Animales , Antimetabolitos Antineoplásicos/química , Antimetabolitos Antineoplásicos/farmacocinética , Biotransformación , Muerte Celular/efectos de los fármacos , Línea Celular , Citidina/química , Citidina/farmacocinética , Citidina/toxicidad , Femenino , Fibrosarcoma , Humanos , Neoplasias Mamarias Experimentales , Ratones , Modelos Químicos , Células Tumorales Cultivadas , Uridina/química , Uridina/farmacocinética , Uridina/toxicidad
12.
Biochem Pharmacol ; 49(2): 173-80, 1995 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-7840794

RESUMEN

Among nine compounds surveyed, cytidine was found to be the most effective in reversing the antiproliferative effects of cyclopentenyl cytosine (CPEC) on human T-lymphoblasts (MOLT-4) in culture. Cytidine, at concentrations of 1-25 microM, enabled cells to maintain normal logarithmic growth when added up to 12 hr after exposure to a 200 nM concentration of the oncolytic nucleoside, CPEC. The most abundant CPEC metabolite, CPEC-5'-triphosphate, is a potent [K1 approximately 6 microM] inhibitor of CTP synthetase (EC 6.3.4.2). Accumulation of this inhibitor resulted in a depletion of CTP levels to 17% of their original cellular concentration. Exogenous cytidine reversed CPEC-induced cellular cytotoxicity by suppressing the formation of CPEC-5'-triphosphate by 70%, and by partially replenishing intracellular CTP to at least 60-70% of its original concentration. In vivo, cytidine (500 mg/kg) administered intraperitoneally 4 hr after each daily dose of CPEC (LD10-LD100) for 9 days reduced the toxicity and abolished the lethality of CPEC to non-tumored mice. Of greater practical importance is the finding that, under these experimental conditions, cytidine did not curtail the antineoplastic properties of CPEC in L1210 tumor-bearing mice. Moreover, the concentration range over which CPEC exhibited antineoplastic activity was extended with cytidine administration.


Asunto(s)
Antineoplásicos/uso terapéutico , Citidina/análogos & derivados , Citidina/farmacología , Leucemia L1210/tratamiento farmacológico , Animales , Citidina/antagonistas & inhibidores , Citidina/sangre , Citidina/uso terapéutico , Citidina/toxicidad , Citidina Trifosfato/análisis , Interacciones Farmacológicas , Leucemia L1210/sangre , Masculino , Ratones , Análisis de Supervivencia , Células Tumorales Cultivadas
13.
Carcinogenesis ; 15(10): 2319-24, 1994 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-7525096

RESUMEN

We have investigated the genotoxicity of two 3'-derivatives of cytidine, 2,3'-O-cyclocytidine (3'-cycloC) and beta-xylocytidine (xyloC), in human leukemia and solid tumor cell lines. Both derivatives were found to be cytotoxic at micromolar concentrations. For example, in the alveolar tumor cell line A549 which was included in all experiments as a reference, drug concentrations required to induce 50% inhibition of cell growth (D50 values) equalled 55 microM for 3'-cycloC and 80 microM for xyloC. Compared with the response of this reference cell line, none of the solid tumor cell lines tested--representing five different malignancies--displayed significant hypersensitivity to these drugs, while the acute lymphoblastic leukemia cell lines proved to be hypersensitive (range of D50 values, 5-13 microM). To gain insight into the modes of cytotoxic action of xyloC and 3'-cycloC, we compared the effect on DNA metabolism of these compounds with that of 1-beta-D-arabinofuranosylcytosine (araC), a potent inhibitor of semi-conservative DNA replication and long-patch excision repair. As seen with araC, the xylo compound strongly inhibited both DNA replicative synthesis and the repair of DNA damage induced by UV light and 60Co gamma-radiation. In gamma-irradiated A549 cells, the extent of repair inhibition by 1 mM xyloC was approximately 40% of that inhibited by araC, and concomitant exposure of the irradiated cultures to xyloC plus araC gave rise to a synergistic response. Since araC was employed at a concentration (0.1 mM) which produced a maximal effect on DNA repair when applied alone, the observed synergistic response implies that the mode of action of xyloC on DNA repair is different from that of araC. In contrast to that observed with xyloC, 3'-cycloC proved to be a very weak inhibitor of DNA replication and repair, strongly suggesting that the genotoxic action of the latter analog may be through a mechanism other than inhibition of DNA synthesis.


Asunto(s)
Ancitabina/toxicidad , Citarabina/toxicidad , Citidina/análogos & derivados , Leucemia/tratamiento farmacológico , Leucemia/genética , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Protocolos de Quimioterapia Combinada Antineoplásica/toxicidad , División Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Niño , Citarabina/administración & dosificación , Citidina/toxicidad , Daño del ADN , Reparación del ADN/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , ADN de Neoplasias/biosíntesis , ADN de Neoplasias/efectos de los fármacos , ADN de Neoplasias/efectos de la radiación , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Humanos , Leucemia/patología , Neoplasias/patología , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/efectos de la radiación
14.
Cancer Chemother Pharmacol ; 34(2): 103-8, 1994.
Artículo en Inglés | MEDLINE | ID: mdl-8194161

RESUMEN

We studied the toxicity of a new experimental anticancer drug, cyclopentenyl cytosine (CPE-C), to human and murine hematopoietic progenitor cells in vitro. Due to CPE-C's in vivo myelotoxicity, it was important to characterize its potential adverse effects on human marrow cells during preclinical development of the drug. Marrow cells were exposed to CPE-C for either 1 h prior to addition in clonal assays or continuously during their culture period. The inhibitory effects of CPE-C on myeloid (CFU-gm) and erythroid (CFU-e, BFU-e) colony formation were concentration- and time-dependent, with continuous CPE-C exposure being significantly more inhibitory than 1-h exposure. The results of both exposure experiments were combined to investigate colony inhibition as a function of overall drug exposure (concentration x time, AUC) and data analyzed by the nonlinear Emax equation. Human and murine CFU-gm had similar AUC-response curves and IAUC70 values (i.e., AUC at 70% colony inhibition) of 40.8 and 41.9 microM h, respectively. In contrast, murine CFU-e and BFU-e were more sensitive to CPE-C, having lower IAUC70 values (both, 21.1 microM h) than human CFU-e and BFU-e (107.8 and 33.0 microM h, respectively). This difference was most prominent with the late erythroid progenitor, CFU-e, in that the human cells were 5 times more resistant to inhibition by CPE-C. CPE-C was myelotoxic in vitro to human and murine marrow cells and toxicity correlated with overall drug exposure.


Asunto(s)
Antineoplásicos/toxicidad , Médula Ósea/efectos de los fármacos , Citidina/análogos & derivados , Animales , Células de la Médula Ósea , Carmustina/toxicidad , Recuento de Células/efectos de los fármacos , Células Clonales/citología , Células Clonales/efectos de los fármacos , Ensayo de Unidades Formadoras de Colonias , Citidina/toxicidad , Relación Dosis-Respuesta a Droga , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Ratones , Factores de Tiempo
15.
Cancer Res ; 52(12): 3503-5, 1992 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-1596909

RESUMEN

Cyclopentenylcytosine (CPE-C), a synthetic cytidine analogue with significant preclinical antitumor activity against both solid tumor xenografts and 1-beta-D-arabinofuranosylcytosine resistant murine leukemia cell lines, will soon enter phase I clinical trials. Unlike 1-beta-D-arabinofuranosylcytosine which is activated by deoxycytidine kinase, the enzyme responsible for the phosphorylation of CPE-C is uridine/cytidine kinase. Preclinical pharmacokinetic studies of CPE-C in nonhuman primates revealed that the primary route of elimination in this species was deamination to cyclopentenyluridine (CPE-U), an inhibitor of uridine/cytidine kinase. Since CPE-C is likely to be deaminated in humans, we investigated the modulating effect of CPE-U on the in vitro cytotoxicity of CPE-C in Molt-4 lymphoblasts. Concurrent exposure of cells to cytotoxic concentrations of CPE-C and 50 microM CPE-U resulted in the rescue of 50% of cells and exposure to CPE-U concentrations in excess of 100 microM resulted in the rescue of greater than 90% of cells. Progressive attenuation of the rescue effect was observed with delayed administration of CPE-U and no cells were rescued when addition of CPE-C was delayed for more than 2 h. At the intracellular level it was observed that the formation of the cytotoxic metabolite, cyclopentenylcytosine triphosphate, was blocked by increasing concentrations of CPE-U presumably secondary to inhibition of uridine/cytidine kinase by CPE-U. Although CPE-U can modulate the cytotoxic effects of CPE-C in vitro, the minimum CPE-U levels that are required for modulation coupled with the available preclinical pharmacokinetic data from nonhuman primates suggests that this modulation is not likely to impact on the antitumor effects of CPE-C in humans.


Asunto(s)
Citidina/análogos & derivados , Uracilo/análogos & derivados , Supervivencia Celular/efectos de los fármacos , Citidina/administración & dosificación , Citidina/toxicidad , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Humanos , Leucemia/patología , Células Tumorales Cultivadas , Uracilo/farmacología
16.
Biochem Pharmacol ; 41(12): 1821-9, 1991 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-1710119

RESUMEN

Cyclopentenylcytosine (Ce-Cyd) is a broad-spectrum antiviral agent active against DNA viruses [herpes (cytomegalo), pox (vaccinia)], (+)RNA viruses [picorna (polio, Coxsackie, rhino), toga (Sindbis, Semliki forest), corona], (-)RNA viruses [orthomyxo (influenza), paramyxo (parainfluenza, measles), arena (Junin, Tacaribe), rhabdo (vesicular stomatitis)] and (+/-)RNA viruses (reo). Ce-Cyd is a more potent antiviral agent than its saturated counterpart, cyclopentylcytosine (carbodine, C-Cyd). Ce-Cyd also has potent cytocidal activity against a number of tumor cell lines. The putative target enzyme for both the antiviral and antitumor action of Ce-Cyd is assumed to be the CTP synthetase that converts UTP to CTP. In keeping with this hypothesis was the finding that the antiviral and cytocidal effects of Ce-Cyd are readily reversed by Cyd and, to a lesser extent, Urd, but not by other nucleosides such as dThd or dCyd. In contrast, pyrazofurin and 6-azauridine, two nucleoside analogues that are assumed to interfere with OMP decarboxylase, another enzyme involved in the biosynthesis of pyrimidine ribonucleotides, potentiate the cytocidal activity of Ce-Cyd. Ce-Cyd should be further pursued, as such and in combination with OMP decarboxylase inhibitors, for its therapeutic potential in the treatment of both viral and neoplastic diseases.


Asunto(s)
Antivirales/farmacología , Ligasas de Carbono-Nitrógeno , Citidina/análogos & derivados , Ligasas/antagonistas & inhibidores , Uracilo/análogos & derivados , Adenosina/análogos & derivados , Adenosina/farmacología , Amidas , Antivirales/toxicidad , Azauridina/farmacología , División Celular/efectos de los fármacos , Citarabina/farmacología , Citidina/farmacología , Citidina/toxicidad , Desoxicitidina/farmacología , Sinergismo Farmacológico , Fluorouracilo/farmacología , Células HeLa/efectos de los fármacos , Humanos , Nucleósidos/farmacología , Pirazoles , Ribavirina/farmacología , Ribonucleósidos/farmacología , Ribosa , Timidina/farmacología , Células Tumorales Cultivadas , Uracilo/farmacología , Replicación Viral/efectos de los fármacos , Virus/efectos de los fármacos
17.
Cancer Res ; 50(22): 7285-92, 1990 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-2224859

RESUMEN

We have established an estrogen receptor- and progesterone receptor-negative, hormone-nonresponsive breast cancer cell line from a receptor-positive, hormone-responsive line grown under estrogen-free conditions. T47D breast cancer cells were cultured under estrogenized conditions (in phenol red-containing medium supplemented with whole fetal bovine serum) and cloned to produce line T47D:A18. The parental T47D line was also estrogen deprived (in phenol red-free medium supplemented with dextran-coated charcoal-treated fetal bovine serum) for more than 1 year and subsequently clone T47D:C4 was established. T47D:A18 was estrogen receptor and progesterone receptor positive as determined by both ligand binding assay analysis and enzyme immunoassay analysis. T47D:C4 cells were estrogen receptor and progesterone receptor negative and mRNA for these receptors was not detected. Incubation of hormone-responsive T47D:A18 cells with 17 beta-estradiol caused a 3-fold increase in cell growth over 8 days when compared to control. This stimulation of growth was completely inhibited by the anti-estrogens 4-hydroxytamoxifen (0.1 microM) and ICI 164,384 (1.0 microM). Receptor-negative T47D:C4 cells were refractory to the effects of both 17 beta-estradiol and the antiestrogens. T47D:A18 cells grown under both estrogen-containing and estrogen-free conditions expressed low levels of transforming growth factor (TGF)-alpha and epidermal growth factor receptor mRNA. In the presence of estrogen, high levels of TGF-beta 1 mRNA were detected in T47D:A18 cells. These levels decreased when T47D:A18 cells were grown in estrogen-free media. Conversely, TGF-beta 2 mRNA was not detected in T47D:A18 cells cultured under estrogenic conditions; however, message was detected after the cells were cultured under estrogen-free conditions. T47D:C4 cells expressed low levels of TGF-alpha, epidermal growth factor receptor, TGF-beta 1, and TGF-beta 2 mRNA. These studies characterize a novel hormone-nonresponsive cell line which has been established from a hormone-responsive cell line grown under estrogen-free and drug-free conditions. Further analysis of these lines should provide valuable information concerning the development of antiestrogen-resistant breast cancer.


Asunto(s)
Neoplasias de la Mama/patología , Citidina/análogos & derivados , Estradiol/farmacología , Receptores de Estrógenos/fisiología , Células Tumorales Cultivadas , Northern Blotting , División Celular/efectos de los fármacos , Células Clonales , Citidina/administración & dosificación , Citidina/toxicidad , Dermatoglifia del ADN , ADN de Neoplasias/análisis , Receptores ErbB/genética , Estradiol/análogos & derivados , Expresión Génica/efectos de los fármacos , Humanos , Técnicas In Vitro , Alcamidas Poliinsaturadas , ARN Mensajero/genética , Receptores de Estrógenos/genética , Receptores de Progesterona/genética , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacología , Factor de Crecimiento Transformador alfa/genética , Factor de Crecimiento Transformador beta/genética
18.
Cancer Chemother Pharmacol ; 20(2): 101-8, 1987.
Artículo en Inglés | MEDLINE | ID: mdl-3664929

RESUMEN

Administration of high-dose uridine or cytidine (3500 mg/kg) resulted in severe hypothermia of 6-10 degrees C in mice. This effect of uridine was observed in three different mouse strains, C57B1/6, Balb/c, and Swiss. A high-dose of uridine also caused hypothermia in Wistar rats. Co-infusion of uridine with benzylacyclouridine, an inhibitor of uridine phosphorylase, partially prevented uridine-mediated hypothermia in mice. A low dose of uridine (100 mg/kg) resulted in a slight increase in temperature. Plasma pharmacokinetics of uridine (at 3500 mg/kg) were studied in two mouse strains, C57B1/6 and Balb/c, and those of cytidine only in C57B1/6 mice. Peak plasma concentrations of uridine in both strains after uridine administration were about 20 mM (at 30-60 min). The peak plasma concentration of cytidine in C57B1/6 mice after cytidine administration was about 12 mM and that of uridine, 1.3 mM. The mean residence time for uridine was about 105 min. The area under the plasma concentration-time curve for uridine was about 50 mmol h/l, and that for cytidine, about 25 mmol h/l. In various tissues of C57B1/6 mice the levels of uridine, uracil and total uracil and cytosine nucleotide pools were determined before and 2 h after uridine administration. Uridine levels increased about 53-fold in liver, about 70-fold in a colon tumor, and only about 7-fold in brain, while the corresponding uracil levels increased about 9-fold, 4-fold and 11-fold, respectively. Total uracil nucleotide pools increased about 8-fold, 3.2-fold and 1.6-fold, respectively. Cytosine nucleotide pools did not increase in the brain. In conclusion, high-dose uridine administration caused severe hypothermia. Plasma levels of uridine and uracil were enhanced to a considerably higher extent than the levels in the tissues. The hypothermia might be related to breakdown products of uridine, since inhibition of uridine breakdown partially prevented hypothermia and since in brain uracil nucleotide levels were only slightly increased after uridine administration, while those of uracil were more markedly increased than in other tissues.


Asunto(s)
Hipotermia/inducido químicamente , Uridina/farmacocinética , Animales , Temperatura Corporal/efectos de los fármacos , Encéfalo/metabolismo , Neoplasias del Colon/metabolismo , Citidina/sangre , Citidina/toxicidad , Citidina Desaminasa/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Femenino , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratas , Ratas Endogámicas , Tetrahidrouridina/toxicidad , Timidina/análogos & derivados , Timidina/farmacología , Uracilo/análogos & derivados , Uracilo/sangre , Uracilo/farmacología , Uridina/sangre , Uridina/toxicidad , Uridina Fosforilasa/antagonistas & inhibidores
19.
Int J Immunopharmacol ; 8(4): 427-31, 1986.
Artículo en Inglés | MEDLINE | ID: mdl-3744641

RESUMEN

The intermediary metabolism of pyrimidine nucleosides was studied in a line of human B lymphoblasts (Raji) in which pyrimidine de novo synthesis deficiency was pharmacologically induced by pyrazofurin. It was found that Raji cells are cytidine deaminase deficient that cytidine has a synergistic effect on the toxicity of pyrazofurin towards these cytidine deaminase deficient cells, affecting both the proliferation and the viability of the cells. Indirect evidences suggest that this synergistic toxicity is not mediated by an effect on nucleoside diphosphate reductase nor on the first steps of pyrimidine de novo synthesis.


Asunto(s)
Antibióticos Antineoplásicos/toxicidad , Citidina Desaminasa/deficiencia , Citidina/toxicidad , Nucleósido Desaminasas/deficiencia , Ribonucleósidos/toxicidad , Amidas , Linfoma de Burkitt/enzimología , Linfoma de Burkitt/patología , División Celular/efectos de los fármacos , Línea Celular , Replicación del ADN/efectos de los fármacos , Sinergismo Farmacológico , Humanos , Cinética , Purinas/farmacología , Pirazoles , Pirimidinas/farmacología , Ribosa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA