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








Base de dados
Intervalo de ano de publicação
1.
Langmuir ; 21(15): 6950-6, 2005 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-16008408

RESUMO

We report the controlled heterocoagulation of platelets and spheres, leading to the formation of colloidally stable, anisotropic hybrid particles. Anionically charged, nanosized polymer latex spherical particles were heterocoagulated on the surface of cationically charged hexagonal gibbsite platelets via the adsorption of a single layer of spheres onto both sides of the hexagonal platelets. The latex particles were annealed at a temperature above the Tg of the latex polymer, resulting in a thin polymer layer covering the gibbsite platelets. This heterocoagulation approach enabled the encapsulation of hydrophilic inorganic particles with polymer latexes and the formation of anisotropic hybrid particles.

2.
Nucleosides Nucleotides Nucleic Acids ; 23(8-9): 1329-33, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15571253

RESUMO

UNLABELLED: Gemcitabine is a deoxycytidine analog, which can be inactivated by deamination catalyzed by deoxycytidine deaminase (dCDA). Altered transport over the cell membrane is a mechanism of resistance to gemcitabine. To facilitate accumulation, the fatty acid derivative CP-4125 was synthesized. Since, the fatty acid is acylated at the site of action of dCDA, a decreased deamination was expected. CP-4125 was equally active as gemcitabine in a panel of rodent and human cell lines and in human melanoma xenografts bearing mice. In contrast to gemcitabine, CP-4125 was not deaminated but inhibited deamination of deoxycytidine and gemcitabine. Pools of the active triphosphate of gemcitabine increased for over 20 hr after CP-4125 exposure, while these pools decreased directly after removal of gemcitabine. IN CONCLUSION: CP-4125 is an interesting new gemcitabine derivative.


Assuntos
Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Ácidos Graxos/metabolismo , Leucemia/tratamento farmacológico , Animais , Antimetabólitos Antineoplásicos/farmacologia , Carbono/química , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Proliferação de Células , Citidina Desaminase , DNA/química , Dano ao DNA , Humanos , Concentração Inibidora 50 , Melanoma/patologia , Camundongos , Camundongos Nus , Modelos Químicos , Transplante de Neoplasias , Nucleosídeo Desaminases/metabolismo , Fosforilação , Ratos , Fatores de Tempo , Gencitabina
3.
Biochem Pharmacol ; 67(3): 503-11, 2004 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-15037202

RESUMO

1-beta-D-arabinofuranosylcytosine (ara-C) is a deoxycytidine analog with activity in leukemia, which requires phosphorylation by deoxycytidine kinase (dCK) to allow formation of its active phosphate 1-beta-D-arabinofuranosylcytosine triphosphate, but can be deaminated by deoxycytidine deaminase. Altered membrane transport is also a mechanism of drug resistance. In order to facilitate ara-C uptake and prolong retention in the cell, lipophilic prodrugs were synthesized. Fatty acid groups with a varying acyl chain length and number of double bonds were esterified at the 5' position on the sugar moiety of ara-C. The compounds were tested in two pairs of ara-C resistant leukemic cell lines (murine L1210 and rat BCLO and their resistant variants L4A6 and Bara-C, respectively) and two pairs of cell lines with a resistance to gemcitabine, another deoxycytidine analog (human ovarian cancer A2780 and murine colon cancer C26-A and their resistant variants AG6000 and C26-G, respectively). L4A6, Bara-C and AG6000 have varying degrees of decreased dCK activity, while the mechanism for C26-G is not yet clear. In the parent cell lines, ara-C was more active, but in the resistant variants several of the analogs were more active, while the degree of cross-resistance varied. In AG6000 with a total dCK deficiency, all compounds were inactive. Structure-activity relation analysis showed that ara-C derivatives with shorter acyl chains and more double bonds were more active in the parental and drug resistant cells. Further mechanistic studies were performed with the elaidic acid derivative of ara-C (CP-4055). CP-4055 inhibited deamination of dCyd partly and induced DNA synthesis inhibition effectively in C26-A and C26-G cells, but the retention of inhibition was much longer for CP-4055 than for ara-C. In contrast to ara-C, CP-4055 inhibited RNA synthesis for 60% after drug exposure. In conclusion, CP-4055 seems to be a promising prodrug, whose effects were different and longer lasting than for the parent drug.


Assuntos
Antineoplásicos/farmacologia , Citarabina/farmacologia , Ácidos Graxos/química , Animais , Linhagem Celular Tumoral , Citarabina/análogos & derivados , Citidina Desaminase , DNA/biossíntese , DNA/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Leucemia/patologia , Nucleosídeo Desaminases/metabolismo , RNA/biossíntese , RNA/efeitos dos fármacos , Ratos
4.
Cancer Chemother Pharmacol ; 52(1): 1-12, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12739060

RESUMO

S-1 is an oral formulation of ftorafur (FT), oxonic acid and 5-chloro-2,4-dihydroxypyridine (CDHP) at a molar ratio of 1:0.4:1. FT is a 5-fluorouracil (5-FU) prodrug, CDHP is a dihydropyrimidine dehydrogenase (DPD) inhibitor and oxonic acid is an inhibitor of 5-FU phosphoribosylation in the gastrointestinal mucosa and was included to prevent gastrointestinal toxicity. We determined the pharmacokinetics of S-1 in 28 patients at doses of 25, 35, 40 and 45 mg/m(2). The plasma C(max) values of FT, 5-FU, oxonic acid and CDHP increased dose-dependently and after 1-2 h were in the ranges 5.8-13 microM, 0.4-2.4 microM, 0.026-1.337 microM, and 1.1-3.6 microM, respectively. Uracil levels, indicative of DPD inhibition, also increased dose-dependently from basal levels of 0.03-0.25 microM to 3.6-9.4 microM after 2-4 h, and 0.09-0.9 microM was still present after 24 h. The pharmacokinetics of CDHP and uracil were linear over the dose range. The areas under the plasma concentration curves (AUC) for CDHP and uracil were in the ranges 418-1735 and 2281-8627 micromol x min/l, respectively. The t(1/2) values were in the ranges 213-692 and 216-354 min, respectively. Cumulative urinary excretion of FT was predominantly as 5-FU and was 2.2-11.9%; the urinary excretion of both fluoro-beta-alanine and uracil was generally maximal between 6 and 18 h. During 28-day courses with twice-daily S-1 administration, 5-FU and uracil generally increased. Before each intake of S-1, 5-FU varied between 0.5 and 1 microM and uracil was in the micromolar range (up to 7 microM), indicating that effective DPD inhibition was maintained during the course. In a biopsy of an esophageal adenocarcinoma metastasis that had regressed, thymidylate synthase, the target of 5-FU, was inhibited 50%, but increased four- to tenfold after relapse in subsequent biopsies. In conclusion, oral S-1 administration resulted in prolonged exposure to micromolar 5-FU concentrations due to DPD inhibition, and the decrease in uracil levels after 6 h followed the pattern of CDHP and indicates reversible DPD inhibition.


Assuntos
Antimetabólitos Antineoplásicos/farmacocinética , Neoplasias/metabolismo , Ácido Oxônico/farmacocinética , Piridinas/farmacocinética , Tegafur/farmacocinética , Adulto , Idoso , Antimetabólitos Antineoplásicos/sangue , Antimetabólitos Antineoplásicos/uso terapêutico , Área Sob a Curva , Disponibilidade Biológica , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Feminino , Fluoruracila/sangue , Fluoruracila/farmacocinética , Fluoruracila/urina , Meia-Vida , Humanos , Masculino , Taxa de Depuração Metabólica , Pessoa de Meia-Idade , Neoplasias/tratamento farmacológico , Ácido Oxônico/sangue , Ácido Oxônico/uso terapêutico , Piridinas/análise , Piridinas/sangue , Piridinas/uso terapêutico , Tegafur/análise , Tegafur/sangue , Tegafur/uso terapêutico , Distribuição Tecidual , Uracila/farmacocinética
5.
Int J Oncol ; 22(1): 201-7, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12469205

RESUMO

Gemcitabine (dFdC) and cisplatin (CDDP) act synergistically by an increase in platinum-DNA adduct formation. Since ribonucleotide (NTP) and deoxyribonucleotide (dNTP) levels are essential for DNA-synthesis and repair of DNA damage, we investigated whether disturbances might account for differences in effects between sensitive and resistant cell lines. The human ovarian cancer cell line A2780, its CDDP-resistant variant ADDP and its dFdC-resistant variant AG6000 were exposed for 24 h to dFdC or CDDP alone, or a combination causing moderate to strong growth inhibition. In AG6000 cells UTP levels were 2-fold lower and in ADDP cells almost 2-fold higher than in A2780 cells. Levels of dTTP, dATP and dCTP were 2-5-fold lower in the resistant cell lines. Drug treatment affected NTP and dNTP levels most pronounced in A2780 cells. dFdC alone, at 1.5 nM to 1 micro M increased ATP, GTP and CTP pools 1.2 to 2.0-fold, while 10 micro M dFdC increased UTP 2.5-fold. Combination of dFdC and CDDP increased all NTP levels at low dFdC and CDDP concentrations more than 1.2-fold, but at 20 micro M CDDP only CTP increased 2.4-fold. Only 1.5 nM dFdC increased all dNTP pools more than 1.6-fold, but at 0.1 and 1 micro M dFdC, dATP and dGTP decreased down to 10-fold, while dTTP increased 3-5-fold. CDDP and the combination increased all dNTP pools over 1.4 and 1.9-fold, respectively. In AG6000 cells dFdC and CDDP hardly affected the NTP and dNTP status, except at the high concentrations, which decreased ATP, GTP and UTP levels 1.2-1.8-fold. Both CDDP alone and the combination increased dTTP, dCTP and dATP pools up to 1.6-fold. In ADDP cells NTP and most dNTP levels were hardly affected, except dGTP levels which decreased to non-detectable levels. In conclusion, both dFdC and CDDP induce concentration and combination dependent changes in NTP and dNTP pools.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Cisplatino/administração & dosagem , Desoxicitidina/análogos & derivados , Desoxicitidina/administração & dosagem , Desoxirribonucleotídeos/análise , Neoplasias Ovarianas/tratamento farmacológico , Ribonucleotídeos/análise , Relação Dose-Resposta a Droga , Feminino , Humanos , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Células Tumorais Cultivadas , Gencitabina
6.
Int J Oncol ; 19(1): 157-62, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11408937

RESUMO

The main mechanism of action of the anticancer drug gemcitabine is assumed to be incorporation of its triphosphate (dFdCTP) into DNA, resulting in inhibition of DNA polymerization, inhibition of DNA synthesis and repair. Another mechanism is inhibition of ribonucleotide reductase leading to imbalance in the deoxyribonucleotide (dNTP) pools. One assay to measure dNTP pools is based on oligonucleotide elongation mediated by DNA polymerase. Since the latter may be affected by dFdCTP, we studied the effect of 0.1-600 pmol dFdCTP on this assay; 10 pmol and more dFdCTP significantly increased the average dpm of the blank (absence of other dNTP) and that of the calibration line of dATP (1.4-1.6-fold); 0.1 pmol and more increased that of the standard dGTP curve significantly (1.1-1.8-fold); 10-75 pmol decreased that of dCTP while 75 and 100 pmol significantly increased that of dCTP (1.3-fold); 50 pmol significantly increased that of dTTP (1.3-1.5-fold). For dATP, dGTP and dTTP, a saturation was reached at 100 pmol dFdCTP, but not yet for dCTP. To minimize these effects, we added an excess of 200 pmol dFdCTP to all samples and calibration lines when measuring dNTP levels of gemcitabine treated samples. In this way the effects of gemcitabine on dNTP levels were studied in human A2780 ovarian, HT29 colon, K562 myelogenous leukemia, H322 non-small cell lung cancer cell lines and the murine lung cancer cell line Lewis Lung. In all cell lines, intrinsic dTTP pools (3-77 pmol/106 cells) were the highest, followed by dATP (1.5-31), dCTP (0.7-27) and (nd-14) dGTP. Exposure to 1 and 10 microM gemcitabine for 4-h concentration dependently decreased dATP 3-10-fold and dGTP to undetectable levels, but dCTP at most 3-fold, while dTTP increased. In conclusion, dFdCTP affects dNTP measurements with the DNA polymerase elongation assay, but its effect could be controlled by addition of similar amounts of dFdCTP to each assay.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , DNA Polimerase Dirigida por DNA/metabolismo , Desoxicitidina/farmacologia , Desoxirribonucleotídeos/metabolismo , Células Tumorais Cultivadas/efeitos dos fármacos , DNA de Neoplasias/biossíntese , DNA de Neoplasias/metabolismo , Desoxicitidina/análogos & derivados , Humanos , Inibidores da Síntese de Ácido Nucleico , Gencitabina
7.
Biochem Pharmacol ; 61(11): 1401-8, 2001 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-11331076

RESUMO

Multidrug resistance (MDR), characterized by a cross-resistance to many natural toxin-related compounds, may be caused either by overexpression of a drug efflux pump such as P-glycoprotein, (P-gP), multidrug resistance proteins MRP1-3, or BCRP/MXR or, in the case of DNA topoisomerase II active drugs, by a decrease in the enzymatic activity of the target molecule termed altered topoisomerase MDR (at-MDR). However, human small cell lung carcinoma (SCLC) cell lines showed a collateral sensitivity to 2',2'-difluorodeoxycytidine (gemcitabine, dFdC) and 1-beta-D-arabinofuranosylcytosine (ara-C). H69/DAU, a daunorubicin (DAU)-resistant variant of H69 with a P-gP overexpression, and NYH/VM, a VM-26 (teniposide)-resistant variant of NYH with an at-MDR, were both 2-fold more sensitive to gemcitabine and 7- and 2-fold more sensitive to ara-C, respectively. MDR variants had a 4.3- and 2.0-fold increased activity of deoxycytidine kinase (dCK), respectively. dCK catalyzes the first rate-limiting activation step of both gemcitabine and ara-C. In addition, deoxycytidine deaminase, responsible for inactivation of dFdC and ara-C, was 9.0-fold lower in H69/DAU cells. The level of thymidine kinase 2, a mitochondrial enzyme that can also phosphorylate deoxycytidine and gemcitabine, was not significantly different between the variants. These differences most likely caused an increased accumulation of the active metabolites (dFdCTP, 2.1- and 1.6-fold in NYH/VM and H69/DAU cells, respectively) and of ara-CTP (1.3-fold in NYH/VM cells). Ara-CTP accumulation was not detectable in either H69 variant. The pools of all ribonucleoside and deoxyribonucleoside triphosphates were at least 3- to 4-fold higher in the NYH variants compared to the H69 variants; for dCTP and dGTP this difference was even larger. The higher ribonucleotide pools might explain the >10-fold higher accumulation of dFdCTP in NYH compared to H69 variants. Since dCTP is low, H69 cells might not need a high ara-CTP accumulation to inhibit DNA polymerase. This might be related to the lack of ara-CTP in H69 variants. In addition, the increased CTP, ATP, and UTP pools in the MDR variants might explain the increased ara-CTP and dFdCTP accumulation. In conclusion, the MDR variants of the human SCLC cell lines were collaterally sensitive due to an increased dCK activity, and consequently an increased ara-CTP and dFdCTP accumulation.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Citarabina/farmacologia , Desoxicitidina/farmacologia , Trifosfato de Adenosina/metabolismo , Antimetabólitos Antineoplásicos/farmacologia , Antineoplásicos/farmacologia , Arabinofuranosilcitosina Trifosfato/metabolismo , Carcinoma de Células Pequenas , Sobrevivência Celular/efeitos dos fármacos , Citidina Desaminase , Daunorrubicina/farmacologia , Desoxicitidina/análogos & derivados , Desoxicitidina Quinase/metabolismo , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Ensaios de Seleção de Medicamentos Antitumorais , Sinergismo Farmacológico , Guanosina Trifosfato/metabolismo , Humanos , Neoplasias Pulmonares , Nucleosídeo Desaminases/metabolismo , Teniposídeo/farmacologia , Timidina Quinase/metabolismo , Células Tumorais Cultivadas , Uridina Trifosfato/metabolismo , Gencitabina
8.
Cancer Res ; 61(9): 3675-81, 2001 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-11325838

RESUMO

Plasma levels of folates and thymidine in mice are about 10-fold higher than in humans and may influence the therapeutic efficacy of thymidylate synthase (TS) inhibitors, such as 5-fluorouracil (5FU) and the antifolates pemetrexed (MTA) and raltitrexed (RTX). Therefore, we tested their therapeutic efficacy in various murine tumor models, grown in mice on a normal and a folate-depleted diet, with high and low thymidine kinase (TK) levels. MTA and RTX were inactive against Colon-26-10 [doubling times gained by treatment; growth delay factor (GDF), 0.5 and 0.3, respectively], whereas 5FU was very active (GDF, >10; complete cures). Colon-26-10/F, grown in mice on a folate-depleted diet, was more sensitive to RTX and MTA (GDF, 2.1 and 1.3, respectively) but not to 5FU (GDF, 1.2); however, leucovorin reversed the effect leading to cures. Folate depletion did not reverse resistance of Colon-26A and Colon-26G (low TK) to MTA and RTX, whereas leucovorin only enhanced the 5FU effect in Colon-26A and Colon-26A/F. Folic acid at 15 mg/kg did not improve the therapeutic efficacy of MTA in folate-deficient mice. The folate-depleted diet decreased the reduced folates in Colon-26A/F and Colon-26-G/F tumors less (4-5-fold; P < 0.01) than in Colon-26-10/F tumors (8-fold; P < 0.001). Folate depletion increased TS levels 2-3-fold in all of the models and TK levels 6-fold (P < 0.01) in Colon-26G/F, explaining the lack of activity of MTA and RTX in Colon-26G/F. In contrast, TK-deficient FM3A/TK tumors were much more sensitive to RTX, MTA, and 5FU than parent FM3A tumors, which have comparable TS levels. The rate of thymidine phosphorylysis varied considerably in all of the tumors without a clear relation to antitumor activity. In conclusion, tumor folates may potentiate (5FU) or protect (antifolates). Murine tumor models should combine low folates and low thymidine rescue to optimize preclinical testing of antifolates.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Antagonistas do Ácido Fólico/farmacologia , Ácido Fólico/metabolismo , Guanina/análogos & derivados , Timidina/metabolismo , Timidilato Sintase/antagonistas & inibidores , Animais , Antimetabólitos Antineoplásicos/administração & dosagem , Divisão Celular/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Dieta , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Feminino , Fluoruracila/administração & dosagem , Fluoruracila/farmacologia , Ácido Fólico/administração & dosagem , Ácido Fólico/sangue , Glutamatos/farmacologia , Guanina/farmacologia , Leucovorina/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Pemetrexede , Quinazolinas/farmacologia , Tiofenos/farmacologia , Timidina/sangue , Timidina Quinase/metabolismo , Timidilato Sintase/metabolismo
9.
Eur J Cancer ; 37(4): 492-8, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11267859

RESUMO

Acute myeloid leukaemia (AML) is thought to be methotrexate (MTX)-resistant. However, a small study suggested that acute monocytic leukemia (AML-M5) is sensitive to MTX. We measured MTX accumulation/polyglutamylation in 20 AML-nonM5, 37 AML-M5 and 83 common/preB-acute lymphoblastic leukaemia (c/preB-ALL) samples. Membrane transport was determined in 11 childhood AMLs (including 3 AML-M5) and in 25 c/preB-ALL samples. MTX sensitivity was determined in 23 AML-nonM5, 15 AML-M5 and 63 common/preB-ALL samples using the thymidylate synthase (TS) inhibition assay. MTX transport was higher in AML samples compared with c/preB-ALL precluding a transport defect in AML. Accumulation of long-chain polyglutamates MTX-Glu(4-6) was 3-fold lower for AML-nonM5 compared with c/preB-ALL cells (median 268 versus 889 pmol MTX-Glu(4-6)/10(9) cells; P < or = 0.001); for AML-M5 samples, median accumulation of MTX-Glu(4-6) was 0 pmol/10(9) cells (P < or = 0.001). After short-term MTX exposure, AML-nonM5 was 6-fold more resistant to MTX compared with c/preB-ALL cells (2.16 versus 0.39 microM; P < 0.001), while AML-M5 was 2-fold more resistant (P = 0.02). In both AML-nonM5 and AML-M5 cells, MTX resistance was circumvented by continuous MTX exposure (median TSI(50) values: 0.052 and 0.041 microM, respectively) compared with a c/preB-ALL value of 0.066 microM. In conclusion, AML-M5 is relatively sensitive to MTX compared with other AML-subtypes even though polyglutamylation of MTX is poor. Using continuous exposure, AML-nonM5 and AML-M5 cells were at least as sensitive to MTX as c/preB-ALL cells. This report suggests that MTX might be an overlooked drug in the treatment of childhood AML.


Assuntos
Antimetabólitos Antineoplásicos/uso terapêutico , Leucemia Monocítica Aguda/tratamento farmacológico , Metotrexato/uso terapêutico , Antimetabólitos Antineoplásicos/farmacocinética , Transporte Biológico , Criança , Resistencia a Medicamentos Antineoplásicos , Humanos , Leucemia Monocítica Aguda/enzimologia , Metotrexato/farmacocinética , Timidilato Sintase/antagonistas & inibidores , Células Tumorais Cultivadas
10.
Eur J Cancer ; 36(18): 2420-9, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11094319

RESUMO

The combination of 2',2'-difluorodeoxycytidine (gemcitabine, dFdC) and cis-diammine-dichloroplatinum(II) (cisplatin, CDDP) is increasingly applied in clinical oncology. We studied the underlying mechanisms of the in vivo schedule dependency and supraadditive interaction between dFdC and CDDP in C57/B16 mice bearing Lewis lung (LL) tumours. Mice were treated with CDDP (6 mg/kg) and dFdC (60 mg/kg) either simultaneously or in a 4 or 24 h interval with dFdC preceding CDDP or vice versa. Four, 8 (in some cases 12) and 24 h after treatment mice were sacrificed and tumours, kidneys, blood and bone marrow (BM) were collected. Since CDDP acts by formation of Platinum (Pt)-DNA adducts and dFdC by incorporation of its triphosphate (dFdCTP) into DNA, we measured total Pt levels, dFdCTP accumulation and Pt-DNA adducts by atomic absorption spectrometry (AAS), high performance liquid chromatography (HPLC) and 2P-postlabelling, respectively. These levels were related to the previously determined antitumour efficacy and toxicity of the dFdC/CDDP combination. Peak dFdCTP accumulation in tumours (11 pmol/mg) was found 4 h after dFdC treatment, while CDDP tended to reduce this in a time-dependent way. Peak levels of total Pt in tumours were found 4 h after CDDP treatment (581 fmol/mg) and dropped 1.8-fold after simultaneous treatment with dFdC (P = 0.04). Treatment with dFdC 4 h after or simultaneously with CDDP increased Pt retention (level 24 h after CDDP treatment) 1.4- and 1.6-fold (P = 0.04 and P = 0.03, respectively). Peak Pt-DNA adduct levels in tumours were also found 4 h after CDDP treatment (7 fmol/microg DNA) and were decreased 3-fold by dFdC treatment 24 h prior to CDDP (P = 0.04). Pt-DNA adduct retention was only decreased when dFdC was given 4 h before CDDP (8-fold (P < 0.01)). The retention and the area-under the concentration time curve of Pt-DNA adducts were related to decreased tumour doubling time (linear regression coefficient (R) = 0.95; P < 0.05, 0.96 P = 0.04 and 0.90; P = 0.04. Pt-DNA adduct levels in the BM cells reached a plateau level 4-24 h after CDDP treatment (approximately 10 fmol/microg DNA), which was increased by dFdC when given either simultaneously with, 4 h before or 4 h after CDDP (6-, 3- and 5-fold at 28 h, 8 h and 28 h, respectively (P < or = 0.04)). Peak Pt-DNA adduct formation (24 h: 8 fmol/microg DNA) in kidneys was enhanced by dFdC when given simultaneously with or 4 h before CDDP (4 h timepoint) (P < 0.01). However, retention was 4- and 6-fold decreased when dFdC was given 4 or 24 h after CDDP, respectively (P < or = 0.01). dFdC given 24 h before CDDP decreased all Pt-DNA adduct levels in kidneys 3-fold or more (P < or = 0.03). Pt-DNA adduct levels were inversely related to kidney toxicity when the most toxic schedule was excluded from the analysis. Peak levels of total Pt in kidneys were reached 24 h after CDDP treatment (4.3 fmol/mg) and the 8 h levels were increased 2-fold by dFdC when given 4 h after CDDP (P = 0.07).


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma Pulmonar de Lewis/tratamento farmacológico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Células da Medula Óssea/metabolismo , Cisplatino/administração & dosagem , Cisplatino/farmacocinética , Desoxicitidina/administração & dosagem , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacocinética , Feminino , Rim/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Gencitabina
11.
Br J Cancer ; 83(8): 1069-76, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10993656

RESUMO

Gemcitabine and paclitaxel are active agents in the treatment of non-small-cell lung cancer (NSCLC). To optimize treatment drug combinations, simultaneously and 4 and 24 h intervals, were studied using DNA flow cytometry and multiple drug effect analysis in the NSCLC cell lines H460, H322 and Lewis Lung. All combinations resulted in comparable cytotoxicity, varying from additivity to antagonism (combination index: 1.0-2.6). Gemcitabine caused a S (48%) and G1 (64%) arrest at IC-50 and 10 x IC-50 concentrations, respectively. Paclitaxel induced G2/M arrest (70%) which was maximal within 24 h at 10 x IC-50. Simultaneous treatment increased S-phase arrest, while at the 24 h interval after 72 h the first drug seemed to dominate the effect. Apoptosis was more pronounced when paclitaxel preceded gemcitabine (20% for both intervals) as compared to the reverse sequence (8%, P = 0.173 for the 4 h and 12%, P = 0.051 for the 24 h time interval). In H460 cells, paclitaxel increased 2-fold the accumulation of dFdCTP, the active metabolite of gemcitabine, in contrast to H322 cells. Paclitaxel did not affect deoxycytidine kinase levels, but ribonucleotide levels increased possibly explaining the increase in dFdCTP. Paclitaxel did not affect gemcitabine incorporation into DNA, but seemed to increase incorporation into RNA. Gemcitabine almost completely inhibited DNA synthesis in both cell lines (70-89%), while paclitaxel had a minor effect and did not increase that of gemcitabine. In conclusion, various gemcitabine-paclitaxel combinations did not show sequence dependent cytotoxic effects; all combinations were not more than additive. However, since paclitaxel increased dFdCTP accumulation, gemcitabine incorporation into RNA and the apoptotic index, the administration of paclitaxel prior to gemcitabine might be favourable as compared to reversed sequences.


Assuntos
Antimetabólitos Antineoplásicos/toxicidade , Ciclo Celular/fisiologia , Desoxicitidina/análogos & derivados , Paclitaxel/toxicidade , Animais , Antimetabólitos Antineoplásicos/farmacocinética , Biotransformação , Carcinoma Pulmonar de Células não Pequenas , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , DNA de Neoplasias/biossíntese , Desoxicitidina/farmacocinética , Desoxicitidina/toxicidade , Humanos , Cinética , Neoplasias Pulmonares , Camundongos , RNA Neoplásico/biossíntese , Células Tumorais Cultivadas , Gencitabina
12.
J Lab Clin Med ; 135(4): 303-8, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10779045

RESUMO

There is considerable evidence that the extracellular fluid volume (ECV) may change in disease states or during longitudinal Intervention studies. Therefore, the measurement of ECV is Important for studying body composition in patients and laboratory animals. We present a modified plasma bromide (Br-, non-radioactive) assay using anion-exchange chromatography, in which a small blood sample of 200 microL (100 microL of plasma) appeared to be enough to reproducibly measure ECV. The inter- and intra-assay accuracy of the Br- analysis ranged from -1.6% to 0.9% and from -0.5% to 0%, respectively. The inter- and intra-assay precision ranged from 1.3% to 1.7% and from 0.6% to 1.2%, respectively. This modified precise and accurate Br- analysis in a small blood sample was applied to investigate whether the ECV changed in rats after ovariectomy. Ovariectomy significantly (P < .05) reduced the ECV as compared with results in SHAM rats. This observation indicates that a change in clinical condition may change ECV, which has consequences for the determination of, for example, the fractional absorption and the relative bioavailability of compounds principally distributed through the ECV.


Assuntos
Água Corporal , Brometos/sangue , Espaço Extracelular/metabolismo , Ovariectomia , Volume Plasmático , Animais , Água Corporal/metabolismo , Cromatografia por Troca Iônica/métodos , Feminino , Ratos , Ratos Wistar , Reprodutibilidade dos Testes
13.
Clin Cancer Res ; 6(3): 1063-72, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10741735

RESUMO

5-Fluorouracil (5FU)-based therapy is given to patients with advanced colorectal cancer and as adjuvant treatment. Thymidylate synthase (TS) is the target for 5FU, and may have a prognostic role for the outcome of 5FU-based therapy together with proliferation markers such as p53 and Ki67. Thymidine phosphorylase (TP, also known as platelet-derived endothelial cell growth factor) may be of importance both in the 5FU drug activation pathway and in tumor angiogenesis, similar to vascular endothelial growth factor (VEGF). TS and TP levels were determined biochemically in fresh-frozen tumor specimens of 32 untreated patients with colorectal cancer, whereas in paraffin-embedded tissue samples, immunohistochemistry was performed for TS, TP, and additional prognostic markers such as p53, Ki67, and VEGF as well as microvessel density. All factors were correlated with patient characteristics such as age, gender, Dukes' stage, angio-invasion, and differentiation grade. TS and TP as measured by various assays were correlated with overall and disease-free survival in this patient group. TP enzyme activity and protein expression correlated with each other. A significant correlation was found between TP enzyme activity and 5-fluoro-2'-deoxyuridine-5'-monophosphate binding activity. VEGF expression correlated significantly with TP immunostaining and Ki67 index. Survival analysis revealed a significant relation of TS levels to the overall survival in this small patient group and a significant correlation between TP activity and disease-free survival. TS and TP both were of prognostic significance in these patients with colorectal cancer. The interesting relationship of TS and TP with angiogenesis and proliferation needs further investigation.


Assuntos
Neoplasias Colorretais/metabolismo , Timidina Fosforilase/análise , Timidilato Sintase/análise , Idoso , Biomarcadores/análise , Neoplasias Colorretais/patologia , Fatores de Crescimento Endotelial/análise , Feminino , Humanos , Imuno-Histoquímica , Antígeno Ki-67/análise , Linfocinas/análise , Masculino , Pessoa de Meia-Idade , Neovascularização Patológica/patologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/análise , Prognóstico , Análise de Sobrevida , Timidina Fosforilase/metabolismo , Timidilato Sintase/metabolismo , Proteína Supressora de Tumor p53/análise , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
14.
Biochim Biophys Acta ; 1474(1): 5-12, 2000 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-10699484

RESUMO

To gain a more detailed insight into the metabolism of 2', 2'-difluoro-2'-deoxycytidine (dFdC, gemcitabine, Gemzar) and its effect on normal ribonucleotide (NTP) metabolism in relation to sensitivity, we studied the accumulation of dFdCTP and the changes in NTP pools after dFdC exposure in a panel of 21 solid tumour and leukaemia cell lines. Both sensitivity to dFdC and accumulation of dFdCTP were clearly cell line-dependent: in this panel of cell lines, the head and neck cancer (HNSCC) cell line 22B appeared to be the most sensitive, whereas the small cell lung cancer (SCLC) cell lines were the least sensitive to dFdC. The human leukaemia cell line CCRF-CEM accumulated the highest concentration of dFdCTP, whereas the non-SCLC cell lines accumulated the least. Not only the amount of dFdCTP accumulation was clearly related to the sensitivity for dFdC (R=-0.61), but also the intrinsic CTP/UTP ratio (R=0.97). NTP pools were affected considerably by dFdC treatment: in seven cell lines dFdC resulted in a 1.7-fold depletion of CTP pools, in two cell lines CTP pools were unaffected, but in 12 cell lines CTP pools increased about 2-fold. Furthermore, a 1.6-1.9-fold rise in ATP, UTP and GTP pools was shown in 20, 19 and 20 out of 21 cell lines, respectively. Only the UTP levels after treatment with dFdC were clearly related to the amount of dFdCTP accumulating in the cell (R=0.64 (P<0.01)), but not to the sensitivity to dFdC treatment. In conclusion, we demonstrate that besides the accumulation of dFdCTP, the CTP/UTP ratio was clearly related to the sensitivity to dFdC. Furthermore, the UTP levels and the CTP/UTP ratio after treatment were related to dFdCTP accumulation. Therefore, both the CTP and UTP pools appear to play an important role in the sensitivity to dFdC.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Desoxicitidina/análogos & derivados , Ribonucleotídeos/metabolismo , Animais , Desoxicitidina/farmacologia , Feminino , Neoplasias de Cabeça e Pescoço , Humanos , Leucemia , Neoplasias Pulmonares , Camundongos , Neoplasias Ovarianas , Ratos , Ribonucleotídeo Redutases/antagonistas & inibidores , Células Tumorais Cultivadas , Gencitabina
15.
J Clin Oncol ; 17(7): 2190-7, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10561275

RESUMO

PURPOSE: To assess possible pharmacokinetic and pharmacodynamic interactions between gemcitabine and paclitaxel in a phase I/II study in non-small-cell lung cancer (NSCLC) patients. PATIENTS AND METHODS: Eighteen patients with advanced NSCLC received the following in a 3-week schedule: gemcitabine 1,000 mg/m(2) (30 minutes, days 1 and 8) and paclitaxel 150 (n = 9) or 200 mg/m(2) (n = 9) before gemcitabine (3 hours, day 1). Plasma pharmacokinetics and pharmacodynamics in mononuclear cells were studied. RESULTS: Gemcitabine did not influence paclitaxel pharmacokinetics at 150 and 200 mg/m(2) (area under the concentration-time curve [AUC], 7.7 and 8.8 micromol/ L. h, respectively; maximum plasma concentration [C(max)], 3.2 and 4.0 micromol/L, respectively), and paclitaxel did not influence that of gemcitabine (C(max), 30 +/- 3 micromol/L) and 2',2'-difluorodeoxyuridine. Paclitaxel, however, dose-dependently increased the C(max) of gemcitabine triphosphate (dFdCTP), the active metabolite of gemcitabine, from 55 +/- 10 to 106 +/- 16 pmol/10(6) cells.( )No significant difference in the AUC of dFdCTP was observed. Moreover, the gemcitabine-paclitaxel combination significantly increased ribonucleotide levels, most pronounced for adenosine triphosphate (six- to seven-fold). Postinfusion paclitaxel AUC was related to pretreatment hepatic function (bilirubin: r = 0. 79; P <.001) and to the percentage decrease in platelets (r = 0.61; P =.009). The latter was also related to the duration of paclitaxel concentration above 0.1 micromol/L (r = 0.62; P =.007). Gemcitabine C(max )was related to the percentage decrease in platelets (r = 0. 58; P =.01), pretreatment hepatic function (bilirubin: r = 0.77; P <. 001), and to plasma creatinine (r = 0.5; P =.03). The pharmacokinetics and pharmacodynamics were not related to response or survival. CONCLUSION: Gemcitabine and paclitaxel pharmacokinetics were related to the percentage decrease in platelets. Paclitaxel did not affect the pharmacokinetics of gemcitabine, nor did gemcitabine affect the pharmacokinetics of paclitaxel, but paclitaxel increased dFdCTP accumulation. This might enhance the antitumor activity of gemcitabine.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Desoxicitidina/análogos & derivados , Neoplasias Pulmonares/tratamento farmacológico , Paclitaxel/farmacocinética , Idoso , Carcinoma Pulmonar de Células não Pequenas/sangue , Células Cultivadas , Desoxicitidina/administração & dosagem , Desoxicitidina/farmacocinética , Relação Dose-Resposta a Droga , Interações Medicamentosas , Feminino , Humanos , Técnicas In Vitro , Infusões Intravenosas , Leucócitos Mononucleares/efeitos dos fármacos , Modelos Lineares , Neoplasias Pulmonares/sangue , Masculino , Taxa de Depuração Metabólica , Pessoa de Meia-Idade , Paclitaxel/administração & dosagem , Estatísticas não Paramétricas , Gencitabina
16.
Br J Haematol ; 106(1): 78-85, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10444166

RESUMO

Resistant variants of the human leukaemic line K562 were developed using selection with the deoxynucleoside analogues cytosine arabinoside, 2-chlorodeoxyadenosine, fludarabine and gemcitabine. The resistant lines displayed a high degree of cross resistance to all deoxynucleoside analogues, with little or no cross resistance to other agents. There was a profound accumulation defect of all nucleoside analogues in the resistant variants but no significant defect in nucleoside transport in any of the variants. 5' nucleotidase activity was strongly increased and deoxycytidine kinase activity was moderately reduced in all of the resistant variants, resulting in reduced accumulation of triphosphate analogues. In addition a deletion in one of the alleles of the deoxycytidine kinase was detected in the fludarabine-resistant line. Ribonucleotide reductase activity was found to be strongly increased in the gemcitabine-selected line and purine nucleoside phosphorylase was increased in the 2-chlorodeoxyadenosine-selected line. Free nucleotide pools were increased in the 2-chlorodeoxyadenosine-selected line. There was no expression of the mdr1 gene by the resistant lines. Karyotypic analysis and FISH experiments using a 6q21 specific probe showed alterations in the 6(q16-q22) region which contains the 5'-nucleotidase gene. Early events in the activation and degradation of deoxynucleoside analogues appear to constitute common mechanisms of resistance to these compounds.


Assuntos
Antineoplásicos/uso terapêutico , Cladribina/uso terapêutico , Citarabina/uso terapêutico , Desoxicitidina/análogos & derivados , Leucemia Eritroblástica Aguda/tratamento farmacológico , Vidarabina/análogos & derivados , Desoxicitidina/uso terapêutico , Desoxicitidina Quinase/metabolismo , Resistencia a Medicamentos Antineoplásicos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Células K562 , Leucemia Eritroblástica Aguda/enzimologia , Fenótipo , Vidarabina/uso terapêutico , Gencitabina
17.
Nucleosides Nucleotides ; 18(4-5): 877-8, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10432699

RESUMO

Lipophilic derivatives of ara-C were developed with the aim to improve drug penetration and retention in solid tumors. Ara-C was esterified at the 5'-position with fatty acids (16-22 C-atoms, 0-3 double bonds). The derivatives were inactive in cell lines with various forms of ara-C and 2',2'-difluorodeoxycytidine (dFdC, gemcitabine) resistance, including deoxycytidine kinase (dCK) deficiency. The activity in the parent cell lines correlated negatively with chain length and positively with double bonds.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Citarabina/análogos & derivados , Animais , Antimetabólitos Antineoplásicos/química , Citarabina/química , Citarabina/farmacologia , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Ensaios de Seleção de Medicamentos Antitumorais , Camundongos , Ratos , Relação Estrutura-Atividade , Células Tumorais Cultivadas , Gencitabina
18.
Ann Oncol ; 10(4): 441-8, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10370787

RESUMO

PURPOSE: To determine possible schedule dependent pharmacokinetic and pharmacodynamic interactions between gemcitabine (2',2'-difluorodeoxycytidine, dFdC) and cisplatin (cis-diammine-dichloroplatinum, CDDP) in patients with advanced stage solid tumors in a phase I trial. PATIENTS AND METHODS: A total of 33 patients with advanced stage solid tumors were treated with gemcitabine (30-min infusion, 800 mg/m2) and cisplatin (one-hour infusion, 50 mg/m2). Sixteen patients had a four-hour interval between gemcitabine (days 1, 8, 15) and cisplatin (days 1 and 8), followed by the reverse schedule and seventeen patients had a 24-hour interval between gemcitabine (days 1, 8, 15) and cisplatin (days 2 and 9), followed by the reverse schedule. Gemcitabine and cisplatin pharmacokinetics were measured in plasma and white blood cells (WBC), isolated from blood samples taken at several time points after the start of treatment. RESULTS: A four-hour time interval between both agents did not reveal major differences in plasma pharmacokinetics of gemcitabine, dFdU (deaminated gemcitabine) and platinum (Pt), and of gemcitabine-triphosphate (dFdCTP) accumulation and Pt-DNA adduct formation in WBC between the two different sequences of gemcitabine and cisplatin. In the patients treated with the 24-hour interval, cisplatin before gemcitabine did not significantly change peak gemcitabine levels and the AUC of plasma dFdU, but tended to increase dFdCTP AUC in WBC 1.5-fold (P < 0.06). Gemcitabine before cisplatin decreased the plasma AUC of Pt 2.1-fold (P = 0.03). No significant differences in Pt-DNA adduct levels in WBC were found, although gemcitabine before cisplatin tended to increase the 24-hour retention of Pt-DNA adducts. Creatinine clearance on day 28 was related to the peak plasma levels of total Pt (linear regression coefficient (r) = 0.47, P = 0.02, n = 26). Furthermore, the increase in the Pt-GG to Pt-AG ratio 24 hours after cisplatin treatment was related to the overall response of patients (r = 0.89, P < 0.01, n = 8). CONCLUSIONS: Of all schedules the treatment of patients with cisplatin 24 hours before gemcitabine led to the highest dFdCTP accumulation in WBC and total Pt levels in plasma. These characteristics formed the basis for further investigation of this schedule in a phase II clinical study.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Neoplasias/sangue , Neoplasias/tratamento farmacológico , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Área Sob a Curva , Cisplatino/administração & dosagem , Cisplatino/sangue , Adutos de DNA/análise , Desoxicitidina/administração & dosagem , Desoxicitidina/análogos & derivados , Desoxicitidina/sangue , Relação Dose-Resposta a Droga , Esquema de Medicação , Feminino , Humanos , Infusões Intravenosas , Leucócitos/química , Modelos Lineares , Masculino , Pessoa de Meia-Idade , Estatísticas não Paramétricas , Resultado do Tratamento , Gencitabina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA