Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Bone Marrow Transplant ; 29(4): 313-9, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11896428

RESUMEN

Taurolidine has been shown to have remarkable cytotoxic activity against selected human tumor cells at concentrations that spare normal cells. In this study we have extended this observation and assessed the ability of Taurolidine to purge tumor cells from chimeric mixtures of bone marrow (BM) and neoplastic cells. Normal murine BM and human leukemic (HL-60) or ovarian (PA-1) tumor cell lines were used as models. Exposure of tumor cells to 2.5 mM Taurolidine for 1 h resulted in the complete elimination of viable cells. In contrast, exposure of BM to 5 mMTaurolidine for 1 h reduced CFU-GM, BFU-E and CFU-GEEM colony formation by only 23.0%, 19.6% and 25.2%, respectively. Inhibition of long-term BM culture (LTBMC) growth following a 1 h exposure to 5 mM Taurolidine also was approximately 20% compared to untreated LTBMC. Finally, chimeric cultures were generated from BM and HL-60GR or PA-1GR cells (tumor cells transfected with the geneticin resistance gene). Exposure of these chimeric cultures to 5 mM Taurolidine for 1 h totally eliminated viable cancer cells while minimally reducing viable BM cells. This finding was confirmed by subsequent positive selection for surviving tumor cells with geneticin. These findings reveal that Taurolidine holds promise for use in BM purging.


Asunto(s)
Antineoplásicos/farmacología , Purgación de la Médula Ósea/métodos , Taurina/farmacología , Tiadiazinas/farmacología , Animales , Trasplante de Médula Ósea , Quimera , Evaluación Preclínica de Medicamentos , Resistencia a Medicamentos/genética , Femenino , Gentamicinas/farmacología , Células HL-60 , Humanos , Ratones , Ratones Endogámicos C57BL , Taurina/análogos & derivados , Trasplante Autólogo , Células Tumorales Cultivadas , Ensayo de Tumor de Célula Madre
2.
Cancer Res ; 61(18): 6816-21, 2001 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-11559556

RESUMEN

Bis-(1,1-dioxoperhydro-1,2,4-thiadiazinyl-4)methane (taurolidine) is a synthetic broad-spectrum antibiotic that reacts with bacterial cell membrane components to prevent adhesion to epithelial cell surfaces. Reflecting the key role of adhesion in the growth and development of human solid tumors, studies were initiated to assess the antiproliferative activity of this agent in selected human and murine tumor cell lines. A 3-day exposure to Taurolidine inhibited the growth of all of the cell lines evaluated with IC(50)s ranging from 9.6-34.2 microM. Studies to identify the mechanism responsible for this effect were conducted in NIH-3T3 murine fibroblasts and the PA-1 and SKOV-3 human ovarian tumor cells. These studies revealed that a 48-h exposure to taurolidine had little effect on cell cycle distribution in PA-1 and SKOV-3 cells but significantly increased the appearance of DNA debris in the sub-G(0)/G(1) region, an effect consistent with an induction of apoptosis. In contrast, in NIH-3T3 cells, taurolidine exposure did not increase DNA debris in the sub-G(0)/G(1) region. Additional studies assessed phosphotidylserine externalization after a 24-h exposure to taurolidine using annexin-V binding as a cell surface marker. These studies revealed that taurolidine increased the percentage of annexin-V-positive cells by 4-fold and 3-fold in PA-1 and SKOV-3 cells, respectively. In NIH-3T3 cells, taurolidine exposure slightly increased ( approximately 5%) annexin-V binding. Parallel studies revealed that exposure to taurolidine also resulted in poly(ADP-ribose) polymerase cleavage in both ovarian tumor cell lines but not in NIH-3T3 cells. Finally, murine-based studies were conducted to assess the antineoplastic activity of three consecutive daily i.p. bolus injections of taurolidine at doses ranging from 5-mg injection/mouse to 30-mg injection/mouse. The 20-mg injection dose produced approximately 10% mortality and was identified as the maximally tolerated dose in this model. Administration of this regimen to nude mice bearing i.p. human ovarian tumor xenografts significantly inhibited both tumor formation and growth. These findings are discussed in light of their clinical implications.


Asunto(s)
Antineoplásicos/farmacología , Taurina/farmacología , Tiadiazinas/farmacología , Células 3T3/efectos de los fármacos , Animales , Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , División Celular/efectos de los fármacos , Fragmentación del ADN , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Ratones , Ratones Desnudos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Taurina/análogos & derivados , Taurina/toxicidad , Tiadiazinas/toxicidad , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Leuk Res ; 24(6): 519-25, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10781687

RESUMEN

Amifostine (AMF), a phosphorylated aminothiol, has been used to treat myelodysplastic syndrome (MDS), where it produces a stimulatory effect on hematopoiesis in bone marrow. To determine if AMF also produced a direct effect on human MDS cells, we planned a study to evaluate the effect of a continuous exposure to AMF on a human MDS cell line. AMF was shown to have a growth-inhibitory effect on MDS cells, with an IC(50) of 14 microM after a 5 day exposure. Cell cycle analysis revealed that a 5 day exposure to 20 microM AMF increased the percentage of cells in G0/G1 and this was accompanied by a decrease in the percentage of cells in S phase. Cytoflorometric and agarose-gel electrophoretic analysis revealed that this effect correlated with cell membrane alterations and DNA fragmentation consistent with an induction of apoptosis without affecting the expression of p53 protein or inducing any lymphoid or myeloid differentiation in the MDS cell line. We conclude that the continuous exposure of a human MDS cell line to AMF is cytotoxic and associated with an induction of apoptosis independent of alterations in p53 expression.


Asunto(s)
Amifostina/toxicidad , Apoptosis/efectos de los fármacos , Síndromes Mielodisplásicos/patología , Protectores contra Radiación/toxicidad , División Celular/efectos de los fármacos , Separación Celular , Citometría de Flujo , Humanos , Inmunofenotipificación , Células Tumorales Cultivadas
4.
Biochem Pharmacol ; 53(4): 571-80, 1997 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-9105409

RESUMEN

We reported that 3'-azidothymidine-3'-deoxythymidine (AZT) plus 5-fluorouracil or methotrexate produces additive cytotoxicity in HCT-8 cells: a reflection of increased AZT metabolism when de novo thymidylate (dTMP) synthesis was inhibited. We now report that AZT plus human recombinant interferon alpha-2a (rIFN-alpha 2a) produces synergistic growth inhibition in these cells. Evaluation of the effect of rIFN-alpha 2a on dTMP metabolism revealed that exposure to rIFN-alpha 2a (+/-AZT) did not affect dTMP synthase activity significantly but increased thymidine (dThd) kinase activity significantly. Consequently, AZT nucleotide production and incorporation into DNA were increased by coexposure to rIFN-alpha 2a. This alone, however, cannot explain the observed synergism. Therefore, the effect of these agents on DNA excision/repair processes was assessed. Isotope clearance studies demonstrated that rIFN-alpha 2a did not alter the rate of [3H]AZT excision from DNA. In contrast, filter-elution studies revealed that rIFN-alpha 2a (+/-AZT) produced more DNA damage and delayed repair compared with the effects produced by AZT alone. Since DNA polymerases alpha and beta are directly involved in gap-filling repair synthesis, experiments next assessed the effect of rIFN-alpha 2a and/or 3'- azido-3'-deoxythymidine-5'-triphosphate (AZTTP) on their activities. Polymerase alpha was inhibited slightly by AZTTP but not by rIFN-alpha 2a. Polymerase beta activity, however, was inhibited dramatically by rIFN-alpha 2a + AZTTP. Finally, western analysis revealed that a 24-hr exposure to 5000 IU/mL rIFN-alpha 2a (+/-20 microM AZT) significantly reduced wild-type p53 expression compared with AZT-exposed cells. We conclude that rIFN-alpha 2a enhances AZT-induced tumor cell growth inhibition by (i) increasing AZT metabolism, and (ii) inhibiting DNA repair and p53-mediated cell cycle control processes.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Interferón-alfa/administración & dosificación , Zidovudina/administración & dosificación , División Celular/efectos de los fármacos , Fragmentación del ADN , Reparación del ADN/efectos de los fármacos , Humanos , Interferón alfa-2 , Inhibidores de la Síntesis del Ácido Nucleico , Proteínas Recombinantes , Timidina Quinasa/efectos de los fármacos , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/análisis
5.
Cancer Res ; 56(11): 2522-6, 1996 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-8653690

RESUMEN

G3361/CP cells, a cisplatin (CDDP)-resistant subclone of the human melanoma cell line G3361, overexpress wild-type p53 protein and demonstrate an increase in the percentage of cells in G0--G1 arrest compared to parental cells. Exposing G3361/CP cells to human recombinant IFN-alpha2a reduces the high basal levels of p53, releases G3361/CP cells from G0-G1 into S phase, and abrogates CDDP resistance. These findings suggest that recombinant IFN-alpha2a disrupts p53-mediated cell cycle regulation to restore CDDP sensitivity in G3361/CP cells.


Asunto(s)
Cisplatino/administración & dosificación , Genes p53 , Interferón-alfa/farmacología , Melanoma/genética , Proteína p53 Supresora de Tumor/metabolismo , Supervivencia Celular/efectos de los fármacos , Daño del ADN , Resistencia a Medicamentos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Interferón alfa-2 , ARN Mensajero/genética , Proteínas Recombinantes , Células Tumorales Cultivadas
6.
Biochem Pharmacol ; 48(9): 1797-805, 1994 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-7980649

RESUMEN

We have reported that 3'-azido-3'-deoxythymidine (AZT) possesses significant cytotoxicity in human tumor models when combined with agents that inhibit de novo thymidylate (dTMP) synthesis, such as 5-fluorouracil (FUra) and methotrexate (MTX). To aid in the further development of these and related cancer chemotherapeutic regimens, this study was undertaken to identify the biochemical processes relevant to the induction of AZT cytotoxicity in the model human colon tumor cell line HCT-8. The IC50 of AZT in this cell line after a 5-day exposure was 55 microM. In cells incubated for 5 days with various concentrations of [3H]AZT alone, both [3H]AZT nucleotide pools and [3H]AZT incorporation into DNA increased as the concentration of AZT in the medium increased. In addition, a 5-day exposure to AZT, at medium concentrations < or = 100 microM, resulted in a reduction in dTMP synthase (EC 2.1.1.45; methylene tetrahydrofolate:deoxyuridine-5'-monophosphate C methyltransferase) and dTHd kinase (EC 2.7.1.27; ATP: thymidine phosphotransferase) activities, compared with cells incubated without drug. The IC50 of AZT was unchanged when the medium concentration of dThd was increased from 0.1 to 50 microM. Increasing the concentration of dThd to 50 microM also did not affect intracellular pools of [3H]AZTDP and [3H]AZTTP or the degree to which [3H]AZT was incorporated into cellular DNA, but did reduce intracellular [3H]AZTMP by approximately 75%. The degree to which 3'-amino-3'-deoxythymidine (AMT) was generated from AZT and incorporated into DNA also was not affected by varying the medium concentration of dThd. However, the amount of [3H]-AMT detected in DNA, < or = 3 pmol/10(6) cells at medium concentrations of [3H]AZT < or = 100 microM, was below that associated with significant cytotoxicity in these cells. These data support the notion that, in this model, AZT cytotoxicity is determined by the relative size of AZTTP pools and its utilization in DNA synthesis. Studies to verify this relationship assessed the effect of alterations in the concentration of dTTP and [3H]AZTTP on [3H]AZT incorporation into newly synthesized DNA in vitro, using DNA polymerases isolated from HCT-8 cells. The results of these studies confirmed that alterations in the concentration of either dTTP or AZTTP to reduce the dTTP/AZTTP ratio resulted in an increase in AZT incorporation into DNA. These findings are discussed in light of their biochemical implications and relevance to ongoing clinical trials.


Asunto(s)
Neoplasias del Colon/metabolismo , Zidovudina/metabolismo , Muerte Celular , Neoplasias del Colon/tratamiento farmacológico , Didesoxinucleótidos , Humanos , Timidina Quinasa/metabolismo , Timidilato Sintasa/metabolismo , Nucleótidos de Timina/análisis , Células Tumorales Cultivadas/efectos de los fármacos , Zidovudina/análogos & derivados , Zidovudina/análisis , Zidovudina/farmacología
7.
Cancer Res ; 52(15): 4069-73, 1992 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-1638518

RESUMEN

We have reported that 5-fluorouracil can increase the cytotoxic and antineoplastic activity of 3'-azido-3'-deoxythymidine (AZT). To further evaluate the antineoplastic utility of AZT we now have assessed its effect in combination with methotrexate (MTX) in the human colon tumor model HCT-8. Incubation of these cells for 5 days in AZT and MTX caused a reduction in the 50% inhibitory concentration of AZT and isobologram analysis revealed additive effects which were reversed by the addition of 50 microM thymidine to the incubation media. This enhanced cytotoxicity appeared not to be related to an effect of AZT on MTX activity; in whole-cell assays the ability of MTX to inhibit de novo dTMP synthesis and deplete intracellular pools of dTTP was not affected by AZT. In contrast, although MTX did not alter AZT triphosphate production, it did affect AZT triphosphate utilization in DNA synthesis. Incubation of cells for 24 h in [3H]AZT alone (5 microM, 3 microCi/ml) resulted in 6.6 pmol AZT incorporated into cellular DNA/10(6) cells. Coincubation of these cells in [3H]AZT (5 microM) plus 5 or 15 nM MTX increased AZT incorporation into DNA to 8.0 and 20.5 pmol/10(6) cells, respectively. Biochemically, this effect appeared to correlate with the concentration-dependent ability of 5 or 15 nM MTX to deplete intracellular dTTP pools, which were reduced by 25 and 49%, respectively. Further evidence of the relationship between intracellular dTTP pools and AZT cytotoxicity was that, in the presence of both MTX and 50 microM thymidine, intracellular dTTP pools remained near normal levels and the incorporation of 5 microM AZT into DNA was not enhanced. Therapeutically, studies conducted in athymic (nude) mice bearing HCT-8 xenografts that received six weekly cycles of MTX (87.5 mg/kg) and AZT (300 mg/kg) revealed that the two-drug regimen exerted superior antineoplastic effects compared to either drug alone (treated versus control approximately 0.9 for AZT or MTX and approximately 0.3 for MTX plus AZT). In addition, the combination did not increase toxicity compared to therapy with MTX alone. These findings are discussed in light of their biochemical and clinical implications.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias del Colon/tratamiento farmacológico , ADN de Neoplasias/metabolismo , Metotrexato/farmacología , Zidovudina/metabolismo , Zidovudina/farmacología , Animales , Biotransformación , Línea Celular , Supervivencia Celular/efectos de los fármacos , Neoplasias del Colon/metabolismo , Humanos , Metotrexato/uso terapéutico , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Timidilato Sintasa/metabolismo , Trasplante Heterólogo , Zidovudina/uso terapéutico
8.
Biochem Pharmacol ; 41(12): 2031-6, 1991 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-2039551

RESUMEN

The concentration of uridine (Urd) in murine tissues appears to be controlled by Urd catabolism, concentrative Urd transport, and the non-concentrative, facilitated diffusion of Urd. Previous reports document the tissue-specific disruption of these processes, and subsequently intracellular pools of free Urd in mice, by the administration of exogenous Urd (250 mg/kg) or the Urd phosphorylase (EC 2.4.2.3; uracil:ribose-1-phosphate phosphotransferase) inhibitor 5-benzylacyclouridine (BAU) (240 mg/kg). We now report the effect of combinations of BAU (120 mg/kg, p.o.), the nucleoside transport inhibitor dipyridamole (DP) (25 mg/kg, i.p.), and exogenous Urd (250 mg/kg, i.v.) on Urd pools in mice. This dose of BAU increased Urd pools 2- to 6-fold, in a tissue-specific manner, for up to 5 hr. DP increased Urd pools 3-fold in spleen, over a 4-hr period, but did not affect other tissues. Administration of BAU 1 hr prior to exogenous Urd resulted in a 50- to 100-fold expansion of tissue normal after 6 hr. Administration of DP 1 hr prior to exogenous Urd caused a tissue-specific 40- to 100-fold increase in Urd pools which, except in spleen, returned to normal within 2 hr. The marked additive effects of these combinations were in contrast to those obtained following the administration of BAU 1 hr prior to DP. This regimen increased Urd pools from 4- to 9-fold, in a tissue-specific manner. In addition, Urd pools remained elevated for up to 9 hr, except in spleen where the Urd concentration was elevated for up to 15 hr. Analysis of enzyme activities indicated that DP does not enhance the inhibitory effect of BAU against murine liver Urd phosphorylase. However, DP did inhibit plasma clearance of BAU, and this effect may partially explain the apparent synergistic effect of this combination. In spite of the prolonged and dramatic expansion of tissue Urd pools produced by BAU + DP, the total Ura nucleotide content in spleen, gut and colon tumor 38 (CT38) increased by less than 70% over a 12-hr period following administration of this combination. These findings are discussed in light of their biochemical and therapeutic implications.


Asunto(s)
Dipiridamol/farmacología , Uracilo/análogos & derivados , Uridina Fosforilasa/antagonistas & inhibidores , Uridina/metabolismo , Administración Oral , Animales , Dipiridamol/administración & dosificación , Sinergismo Farmacológico , Femenino , Inyecciones Intravenosas , Ratones , Ratones Endogámicos , Factores de Tiempo , Distribución Tisular , Uracilo/administración & dosificación , Uracilo/farmacocinética , Uracilo/farmacología , Uridina/administración & dosificación , Uridina/sangre , Uridina/farmacología
9.
Cancer Res ; 50(13): 4026-31, 1990 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-2354452

RESUMEN

Preliminary in vitro studies suggest that the combination of 5-fluorouracil (FUra) and 3'-azido-3'-deoxythymidine (AZT) is more cytotoxic than either agent alone. Therefore, a biochemical and therapeutic evaluation of this combination was initiated. Quantitation of the cytotoxicity of FUra plus AZT against the growth of HCT-8 cells in vitro revealed that 1 microM FUra (approximately 10% inhibitory concentration) increased the cytotoxicity of AZT and decreased its 50% inhibitory concentration by 60%. Similarly, incubating HCT-8 cells in 5 microM AZT (approximately 10% inhibitory concentration) decreased the 50% inhibitory concentration of FUra by over 50%. Biochemical analysis indicated that AZT did not affect FUra-induced inhibition of thymidylate synthase or [3H]-FUra incorporation into nucleic acids. In contrast, incubation in 5 microM FUra increased the incorporation of [3H]-AZT (5 microM) into the nucleic acid fraction of these cells by 52% (P less than 0.05). Therapeutic evaluation of this combination in athymic (nude) mice bearing HCT-8 xenographs revealed that, while weekly FUra (85 mg/kg) or AZT (600 mg/kg) exerts minimal antineoplastic activity (after 4 courses, treatment/control = 0.81 and 0.70, respectively), their combination, at the same doses, inhibited tumor growth by nearly 70% (P less than 0.01 versus FUra alone). FUra-related host toxicity was not increased by the addition of AZT. Higher doses of FUra alone were not more effective than FUra plus AZT. In vivo, AZT did not affect the incorporation of [3H]-FUra into the nucleic acid fraction of various murine tissues, including HCT-8 xenografts. FUra, however, increased [3H]-AZT incorporation into nucleic acids in a tissue-specific manner. In the presence of FUra, the incorporation of [3H]-AZT in spleen, liver, and gut increased 40, 21, and 4%, respectively, while in HCT-8 xenografts [3H]-AZT incorporation increased more than 2-fold. Analysis of the activities of selected enzymes involved in pyrimidine metabolism suggests that this tissue-specific effect may be related to the pyrimidine salvage capacity of these tissues. These findings are described in light of their potential impact on human colon cancer chemotherapy.


Asunto(s)
Neoplasias del Colon/metabolismo , Fluorouracilo/farmacología , Timidilato Sintasa/antagonistas & inhibidores , Zidovudina/farmacología , Animales , División Celular/efectos de los fármacos , Neoplasias del Colon/patología , ADN de Neoplasias/metabolismo , Esquema de Medicación , Sinergismo Farmacológico , Femenino , Fluorouracilo/metabolismo , Fluorouracilo/toxicidad , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Nucleósidos de Pirimidina/metabolismo , Células Tumorales Cultivadas , Zidovudina/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...