Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Br J Cancer ; 129(9): 1373-1382, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37524968

RESUMO

BACKGROUND: Successful immunotherapy is restricted to some cancers only, and combinatorial strategies with other drugs could help to improve their efficacy. Here, we monitor T cells in NSCLC model after treatment with cytotoxics (CT) and anti-VEGF drugs, to understand when immune checkpoint inhibitors should be best associated next. METHODS: In vivo study was performed on BALB/c mice grafted with KLN205 cells. Eight treatments were tested including control, cisplatin and pemetrexed as low (LD CT) and full (MTD CT) dose as single agents, flat dose anti-VEGF and the association anti-VEGF + CT. Full immunomonitoring was performed by flow cytometry on tumor, spleen and blood over 3 weeks. RESULTS: Immunomodulatory effect was dependent upon both treatments and time. In tumors, combination groups shown numerical lower Treg cells on Day 21. In spleen, anti-VEGF and LD CT group shown higher CD8/Treg ratio on Day 7; on Day 14, higher T CD4 were observed in both combination groups. Finally, in blood, Tregs were lower and CD8/Treg ratio higher, on Day 14 in both combination groups. On Day 21, CD4 and CD8 T cells were higher in the anti-VEGF + MTD CT group. CONCLUSIONS: Anti-VEGF associated to CT triggers notable increase in CD8/Tregs ratio. Regarding the scheduling, a two-week delay after using anti-VEGF and CT could be the best sequence to optimize antitumor efficacy.


Assuntos
Antineoplásicos , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Camundongos , Animais , Pemetrexede , Cisplatino , Neoplasias Pulmonares/patologia , Linfócitos T Reguladores , Linfócitos T CD8-Positivos
6.
ESMO Open ; 8(3): 101577, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37267808

RESUMO

BACKGROUND: The chemotherapeutic agent 5-fluorouracil (5-FU) is catabolized by dihydropyrimidine dehydrogenase (DPD), the deficiency of which may lead to severe toxicity or death. Since 2019, DPD deficiency testing, based on uracilemia, is mandatory in France and recommended in Europe before initiating fluoropyrimidine-based regimens. However, it has been recently shown that renal impairment may impact uracil concentration and thus DPD phenotyping. PATIENTS AND METHODS: The impact of renal function on uracilemia and DPD phenotype was studied on 3039 samples obtained from three French centers. We also explored the influence of dialysis and measured glomerular filtration rate (mGFR) on both parameters. Finally, using patients as their own controls, we assessed as to what extent modifications in renal function impacted uracilemia and DPD phenotyping. RESULTS: We observed that uracilemia and DPD-deficient phenotypes increased concomitantly to the severity of renal impairment based on the estimated GFR, independently and more critically than hepatic function. This observation was confirmed with the mGFR. The risk of being classified 'DPD deficient' based on uracilemia was statistically higher in patients with renal impairment or dialyzed if uracilemia was measured before dialysis but not after. Indeed, the rate of DPD deficiency decreased from 86.4% before dialysis to 13.7% after. Moreover, for patients with transient renal impairment, the rate of DPD deficiency dropped dramatically from 83.3% to 16.7% when patients restored their renal function, especially in patients with an uracilemia close to 16 ng/ml. CONCLUSIONS: DPD deficiency testing using uracilemia could be misleading in patients with renal impairment. When possible, uracilemia should be reassessed in case of transient renal impairment. For patients under dialysis, testing of DPD deficiency should be carried out on samples taken after dialysis. Hence, 5-FU therapeutic drug monitoring would be particularly helpful to guide dose adjustments in patients with elevated uracil and renal impairment.


Assuntos
Deficiência da Di-Hidropirimidina Desidrogenase , Di-Hidrouracila Desidrogenase (NADP) , Humanos , Di-Hidrouracila Desidrogenase (NADP)/genética , Deficiência da Di-Hidropirimidina Desidrogenase/complicações , Deficiência da Di-Hidropirimidina Desidrogenase/induzido quimicamente , Deficiência da Di-Hidropirimidina Desidrogenase/tratamento farmacológico , Antimetabólitos Antineoplásicos/efeitos adversos , Fluoruracila/uso terapêutico , Uracila/uso terapêutico
8.
ESMO Open ; 7(6): 100646, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36521418

RESUMO

BACKGROUND: Anti-programmed cell death protein 1 (PD1)/programmed death-ligand 1 (PD-L1) agents have only moderate antitumor activity in some advanced solid tumors (AST), including breast cancer (BC), prostate cancer (PC), cervical cancer (CC), and head and neck cancer (HNC). Combining anti-PD-L1 with anti-cytotoxic T-lymphocyte-associated protein (CTLA) and chemotherapy may significantly improve efficacy. PATIENTS AND METHODS: MOVIE is a multicohort phase I/II study examining the combination of anti-PD-L1 durvalumab (Durv; 1500 mg IV Q4W) plus anti-CTLA tremelimumab (Trem; 75 mg IV Q4W) with metronomic vinorelbine (MVino; 20-40 mg orally daily) in various AST resistant to conventional therapies. The primary objective of the phase I part was to determine the maximum tolerated dose (MTD) and recommended dose for phase II (RP2D). RESULTS: Among the 14 patients enrolled during phase I, including 13 women and 1 man, 9 had BC, 1 PC, 2 CC, and 2 miscellaneous cancers with high mutational loads. Median age was 53 years. A total of 12 patients were assessable for the dose-escalation part in which only one dose-limiting toxicity (DLT) was observed [one neutropenia without fever, grade (G) 4]. Two (14.3%), four (28.6%), and four (28.6%) patients had G ≥3 adverse events (AEs) related to MVino, Durv, and Trem, respectively. Treatment-related events included mostly clinical AEs with asthenia (eight G2; three G3), colitis (one G2, one G3), diarrhea (one G3), nausea (two G2), dry skin (two G2), maculopapular rash (one G3), and hyperthyroidism (three G2). No toxic death was reported. Preliminary data showed one patient (CC) who presented a complete response and four patients with stable disease (SD). CONCLUSIONS: MTD was not reached and dose level 2 (MVino 40 mg, Durv 1500 mg, Trem 75 mg) was selected as RP2D. The safety profile of the combination was manageable and consistent with previous reports of Trem + Durv or MVino. Phase II is currently ongoing in BC, PC, CC, HNC, and miscellaneous cohorts.


Assuntos
Antineoplásicos , Neoplasias de Cabeça e Pescoço , Neoplasias do Colo do Útero , Masculino , Humanos , Feminino , Pessoa de Meia-Idade , Vinorelbina/farmacologia , Filmes Cinematográficos , Antineoplásicos/efeitos adversos , Neoplasias de Cabeça e Pescoço/induzido quimicamente
9.
Sci Rep ; 10(1): 4147, 2020 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-32139753

RESUMO

Developing targeted nanoparticles is a rising strategy to improve drug delivery in oncology. Antibodies are the most commonly used targeting agents. However, determination of their optimal number at the surface remains a challenging issue, mainly due to the difficulties in measuring precisely surface coating levels when prototyping nanoparticles. We developed an original quantitative assay to measure the exact number of coated antibodies per nanoparticle. Using flow cytometry optimized for submicron particle analysis and beads covered with known amounts of human IgG-kappa mimicking various amounts of antibodies, this new method was tested as part of the prototyping of docetaxel liposomes coated with trastuzumab against Her2+ breast cancer. This quantification method allowed to discriminate various batches of immunoliposomes depending on their trastuzumab density on nanoparticle surface (i.e., 330 (Immunoliposome-1), 480 (Immunoliposome-2) and 690 (Immunoliposome-3), p = 0.004, One-way ANOVA). Here we showed that optimal number of grafted antibodies on nanoparticles should be finely tuned and highest density of targeting agent is not necessarily associated with highest efficacy. Overall, this new method should help to better prototype third generation nanoparticles.


Assuntos
Docetaxel/química , Lipossomos/química , Trastuzumab/química , Análise de Variância , Citometria de Fluxo , Nanopartículas/química
10.
Crit Rev Oncol Hematol ; 127: 1-5, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29891106

RESUMO

There is a rising evidence that the proverbial statement "No pain, No gain" first coined at the light of pioneering clinical experiences with canonical chemotherapy still holds true in the era of modern treatments of cancer. This close relationship between the occurrence of specific drug-related toxicity and treatment outcome has been confirmed since then with a large variety of treatments, ranging from cytotoxics, hormonotherapy, targeted therapy and much interestingly even with the latest immune checkpoint inhibitors. In the current context of precision medicine, and along with the constant quest for identifying predictive biomarkers, close monitoring of treatment-related toxicities could therefore be convenient to help predicting therapeutic response, but presents several caveats. The purpose of this review is to briefly describe these relationships across the different treatments, to comment on possible underlying mechanisms and to comment on possible strategies aiming at exploiting this relationship while keeping the maximal safety ensured in patients with cancer. In particular, this review will investigate on how drug exposure along with germinal and somatic genetic issues does impact on the "No Pain, No Gain" aphorism, and why the temptation to use treatment-related toxicities as a cheap and convenient way to predict clinical outcome or to adapt dosing should be resisted. We do advocate instead for developing comprehensive genomic support along with extensive biomathematical modeling to better customize dosing and shift towards a new "No Pain, Maximal Gain" paradigm.


Assuntos
Biomarcadores Tumorais , Imunoterapia/efeitos adversos , Neoplasias/diagnóstico , Neoplasias/terapia , Dor/etiologia , Medicina de Precisão , Biomarcadores Farmacológicos/análise , Biomarcadores Tumorais/análise , Biomarcadores Tumorais/fisiologia , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/diagnóstico , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/fisiopatologia , Genômica/métodos , Humanos , Dor/diagnóstico , Medicina de Precisão/efeitos adversos , Medicina de Precisão/métodos , Medicina de Precisão/tendências , Prognóstico , Resultado do Tratamento
11.
Clin Cancer Res ; 6(4): 1529-35, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10778986

RESUMO

We investigated the effects of 2'-deoxyinosine (d-Ino), a modulator yielding thymidine phosphorylase activity, on cellular pharmacology of 5-fluorouracil (FUra) in various human colorectal cell lines and its antitumoral activity when combined with FUra in human xenografts. Associating d-Ino with FUra increased by 38 up to 700 times the sensitivity of HT29 and FUra-resistant SW620 lines, respectively, but not of CaCO2 cells, although high levels of intracellular FdUMP and subsequent higher thymidylate synthase inhibition were observed. Cell death studies confirmed the ability of d-Ino to enhance both early and late apoptosis induced by FUra in HT29 and SW620 but not in CaCo2. Similarly, we showed that associating d-Ino increased by 68 up to 101% the Fas overexpression induced by FUra in HT29 and SW620 but not in CaCo2 cells. Anti-Fas and anti-FasL antibodies both partly reversed this increase of cell sensitivity, thus confirming the role Fas plays in the modulation of FUra toxicity by d-Ino. This Fas component could explain the discrepancy between the lines because CaCO2 has been described as insensitive to Fas-mediated apoptosis. Antitumor activity of the combination was next investigated in nude mice transplanted with SW620. Results showed that although FUra alone has little effect on SW620 xenografts (P > 0.05), associating d-Ino significantly reduced the tumor growth by 57% (P < 0.05). This study suggests that it is possible to reduce both in vitro and in vivo resistance to FUra by modulating the way the drug is converted after cellular uptake.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias do Colo/tratamento farmacológico , Neoplasias Colorretais/tratamento farmacológico , Análise de Variância , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Neoplasias do Colo/patologia , Neoplasias Colorretais/patologia , Sinergismo Farmacológico , Fluoruracila/administração & dosagem , Fluoruracila/metabolismo , Humanos , Concentração Inibidora 50 , Inosina/administração & dosagem , Inosina/análogos & derivados , Camundongos , Camundongos Nus , Transplante de Neoplasias , Timidilato Sintase/antagonistas & inibidores , Timidilato Sintase/metabolismo , Nucleotídeos de Timina/metabolismo , Transplante Heterólogo , Trítio , Células Tumorais Cultivadas , Receptor fas/efeitos dos fármacos , Receptor fas/metabolismo
12.
Mol Cancer Ther ; 1(2): 133-9, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12467230

RESUMO

We reported previously that 5-fluorouracil (FUra) efficacy could be enhanced by increasing tumoral thymidine phosphorylase (TP) activity. Potentiated TP yield was achieved by either transfecting cells with human TP gene (A. Evrard et al., Br. J. Cancer, 80: 1726-1733, 1999) or associating FUra with 2'-deoxyinosine (d-Ino), a modulator providing the tumors with TP cofactor deoxyribose 1-phosphate (J. Ciccolini et al., Clin. Cancer Res., 6: 1529-1535, 2000). The purpose of the present work was to study the effects of a combined modulation (TP gene transfer + use of d-Ino) on the sensitivity to FUra of the LS174T human colorectal cell line. Results showed a near 4000 times increase of cell sensitivity in vitro after double (genetic + biochemical) modulation. This potentiation of tumor response was accompanied by a total change in the FUra anabolic pathway with a 5000% increase of cytosolic fluorodeoxyuridine monophosphate, a stronger and longer inhibition of thymidylate synthase, and 300% augmentation of DNA damage. Besides, whereas thymidine failed to inhibit FUra cytotoxicity in LS174T wild-type cells, the potentiation of the antitumor activity observed in the modulating regimen was partly reversed by thymidine, indicative of thymidylate synthase as the main drug target. The impact of this double modulation was next investigated in xenograft-bearing nude mice. Results showed that whereas FUra alone was completely ineffective on wild-type tumor growth, the size of TP-transfected tumors in animals treated with the FUra/d-Ino combination was reduced by 80% (P < 0.05). Our results suggest that FUra exhibits stronger antiproliferative activity when activated via TP through the DNA pathway and that high tumoral TP activity therefore leads to enhanced sensitivity to fluoropyrimidines.


Assuntos
Neoplasias Colorretais/terapia , Fluoruracila/uso terapêutico , Terapia Genética , Inosina/análogos & derivados , Inosina/uso terapêutico , Timidina Fosforilase/genética , Animais , Apoptose/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Terapia Combinada , Sinergismo Farmacológico , Técnicas de Transferência de Genes , Humanos , Técnicas In Vitro , Camundongos , Camundongos Nus , Timidina Fosforilase/metabolismo , Timidilato Sintase/antagonistas & inibidores , Timidilato Sintase/metabolismo , Nucleotídeos de Timina/metabolismo , Trítio , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo
13.
Fundam Clin Pharmacol ; 14(2): 147-54, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10796062

RESUMO

An HPLC method was developed for in vitro detection and monitoring of intracellular metabolites of [3H]-5-fluorouracil (FUra). Results showed a preferential activation of FUra to ribonucleoside and ribonucleotide derivatives (FURd, FUMP, FUDP and FUTP) in the human colorectal HT29 cell line. We screened various agents so as to determine if they could act as modulators of metabolism and/or toxicity of FUra by reversing the activation pathway of FUra from ribo- to deoxyribonucleotides, thus enhancing FdUMP formation. Different drugs (efflux inhibitors, catabolism inhibitors and enzymatic cofactors) were tested for enhancement of cytotoxicity when associated with FUra. The most promising agents were further studied by assessment of their ability to modulate intracellular activation of FUra to enhance thymidylate synthase (TS) inhibition by FUra and to increase the subsequent induction of apoptosis. 2'-Deoxyinosine (d-Ino), a deoxyribose 1-phosphate donor increasing thymidine phosphorylase activity, stood out as the best modulating agent we screened. Results showed an up to 30-fold increase of cytotoxicity along with a stronger inhibition of TS when FUra was associated with d-Ino, while FUra alone exhibited a lesser effect on TS activity. Besides, HPLC analysis revealed a complete reversal of the activation pathway of FUra, thus leading to an intracellular accumulation of deoxyribonucleotides. Assessment of cell cycle distribution showed a marked increase (+480%) of apoptosis in cells exposed to FUra/d-Ino compared to FUra alone. The HPLC method we developed is a convenient tool for assessing to what extent modulators will actually act on the intracellular activation of FUra. This study confirms the potentiality of d-Ino to modulate FUra metabolism in vitro. It proved to be an agent able to orientate the mechanism of action of FUra towards the inhibition of TS in cells where the normal activation pathway of the drug does not result in the intracellular accumulation of the active metabolite FdUMP.


Assuntos
Desoxirribonucleotídeos/metabolismo , Fluoruracila/metabolismo , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Sistema Livre de Células/enzimologia , Cromatografia Líquida de Alta Pressão , Fluoruracila/farmacologia , Células HT29 , Humanos , Inosina/análogos & derivados , Inosina/metabolismo , Inosina/farmacologia , Timidina Fosforilase/efeitos dos fármacos , Timidina Fosforilase/metabolismo , Timidilato Sintase/antagonistas & inibidores , Timidilato Sintase/metabolismo , Trítio
14.
J Chemother ; 15(3): 266-74, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12868554

RESUMO

The human liver metabolism of paclitaxel (Taxol), an anticancer drug, leads to three metabolites: 6alpha-hydroxypaclitaxel, 3'-p-hydroxypaclitaxel and 6alpha,3'-p-dihydroxypaclitaxel. The inter-individual variability of paclitaxel metabolism was investigated first in vitro using 22 human liver microsomes. Three metabolites have been detected by HPLC. This preliminary work revealed marked inter-individual differences in paclitaxel metabolism. The amount of major metabolite 6alpha-hydroxypaclitaxel formed varied 16-fold (0.7 to 11.5 nmol/mg/h). We next studied the effect of 29 compounds (antineoplastics, antiemetics, histamine-2 receptor antagonist, antalgics, antifungals, antivirals, psychotropics, antibiotic, corticoid, antiarrhythmic, calcium channel blocker) on paclitaxel metabolism in human liver microsomes. Among the compounds studied, quercetin, antifungal drugs such as ketoconazole and miconazole, and the antineoplastic drug doxorubicin inhibited formation of 6alpha-hydroxypaclitaxel. Dixon plots indicated that quercetin and doxorubicin inhibited 6alpha-hydroxypaclitaxel formation through a competitive mechanism with a Ki of 10.1 microM and 64.8 microM, respectively. The inhibition of this metabolite by ketoconazole was through a noncompetitive mechanism with a Ki of 11.8 microM. Our data thus suggest that special attention should be paid when these drugs are combined in clinical practice.


Assuntos
Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Microssomos Hepáticos/efeitos dos fármacos , Paclitaxel/metabolismo , Paclitaxel/farmacologia , Adulto , Disponibilidade Biológica , Células Cultivadas , Cromatografia Líquida de Alta Pressão , Relação Dose-Resposta a Droga , Interações Medicamentosas , Feminino , Humanos , Masculino , Variações Dependentes do Observador , Sensibilidade e Especificidade
15.
J Chemother ; 22(2): 129-33, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20435574

RESUMO

The combination of capecitabine and the tyrosine kinase inhibitor erlotinib has recently been tested in patients with gemcitabine-refractory pancreatic tumors, with limited success. To understand this lack of efficacy, we studied the molecular effects of these agents in Capan-1 and Capan-2 human pancreatic resistant cancer cells. Erlotinib up-regulated thymidine phosphorylase (+50%) and downregulated dihydropyrimidine dehydrogenase (+55%) in a cell-dependent manner, thus suggesting that the combination should result in synergism. However, only mild additivity was achieved at best when combining both drugs, and several sequences tested even led to strong antagonism. Further experiments were performed to understand this lack of efficacy. We found that the fluoropyrimidine down-regulated EGFR expression by 30%, an unexpected finding resulting in a possible reduction in efficacy when cells were subsequently exposed to erlotinib. We also observed marked drug-induced over-expression of both cytosolic and extracellular vascular endothelial growth factor (VEGF) secretion, thus possibly triggering proliferation. These preliminary findings strongly suggest that these observations could be new mechanisms in the development of acquired drug resistance in pancreatic cancer cells.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Desoxicitidina/análogos & derivados , Resistencia a Medicamentos Antineoplásicos , Fluoruracila/análogos & derivados , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/farmacologia , Capecitabina , Linhagem Celular Tumoral , Proliferação de Células , Desoxicitidina/farmacologia , Di-Hidrouracila Desidrogenase (NADP)/biossíntese , Interações Medicamentosas , Receptores ErbB/biossíntese , Cloridrato de Erlotinib , Fluoruracila/farmacologia , Humanos , Neoplasias Pancreáticas , Timidina Fosforilase/biossíntese , Fator A de Crescimento do Endotélio Vascular/biossíntese
16.
Curr Med Chem ; 16(33): 4361-71, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19835568

RESUMO

Anticancer drugs are essential agents in the global strategy developed to fight cancer. Still, narrow therapeutic indices, erratic pharmacokinetics profiles and lack of selectivity towards malignant tissues often hamper their efficacy at the bedside, when they not cause severe toxicities. In this respect, developing innovative drug delivery strategies that would selectively target malignant tissues is still an ongoing story, both in experimental and in clinical oncology. Delivery systems such as liposomes are usually required when an existing formulation is not satisfactory, because encapsulation is expected to provide higher therapeutic efficacy and safety. Such significant improvement in therapeutic efficacy and/or therapeutic indices has already been achieved in patients with some liposome-encapsulated drugs such as anthracyclines. It is now possible to develop a wide range of vectors varying in size, composition, and surface morphology suitable for a variety of therapeutic applications, including for targeting tumor tissues. Reformulation of anticancer drugs in liposomes remains a challenging opportunity to stretch the therapeutic indices of many cytotoxic drugs, through the optimization of their distribution in the body. Despite these promising and exciting perspectives in oncology, to date only few drugs (e.g., anthracyclines) have actually made their way as liposomes from the bench to the bedside. However, as target therapies have brought a new hope in the cancer war in the 2000's, developing now targeted delivery systems is more and more seen as the next step to further improve clinical outcome in cancer patients. This review covers the achievements, limits, and new expectancies of anticancer drugs as candidates for liposomal encapsulation.


Assuntos
Antineoplásicos/administração & dosagem , Lipossomos/química , Anticorpos Imobilizados/química , Antineoplásicos/química , Antineoplásicos/farmacocinética , Portadores de Fármacos , Nanotecnologia , Fosfolipídeos/química
17.
Br J Cancer ; 97(7): 919-26, 2007 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-17848948

RESUMO

Drug resistance is a major cause of treatment failure in cancer chemotherapy, including that with the extensively prescribed antimetabolite, 5-fluorouracil (5-FU). In this study, we tried to reverse 5-FU resistance by using a double-punch strategy: combining 5-FU with a biochemical modulator to improve its tumoural activation and encapsulating both these agents in one same stealth liposome. Experiments carried out in the highly resistant, canonical SW620 human colorectal model showed a up to 80% sensitisation to 5-FU when these cells were treated with our liposomal formulation. Results with this formulation demonstrated 30% higher tumoural drug uptake, better activation with increased active metabolites including critical-5-fluoro-2-deoxyuridine-5-monophosphate, superior inhibition (98%) of tumour thymidylate synthase, and subsequently, higher induction of both early and late apoptosis. Drug monitoring showed that higher and sustained exposure was achieved in rats treated with liposomal formulation. When examined in a xenograft animal model, our dual-agent liposomal formulation caused a 74% reduction in tumour size with a mean doubling in survival time, whereas standard 5-FU failed to exhibit significant antiproliferative activity as well as to increase the lifespan of tumour-bearing mice. Taken collectively, our data suggest that resistance to 5-FU can be overcome through a better control of its intratumoural activation and the use of an encapsulated formulation.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias Colorretais/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos , Fluoruracila/farmacologia , Lipossomos , Animais , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Feminino , Humanos , Técnicas In Vitro , Camundongos , Camundongos Nus , Timidilato Sintase/antagonistas & inibidores , Células Tumorais Cultivadas/efeitos dos fármacos
18.
J Chromatogr B Biomed Sci Appl ; 759(2): 299-306, 2001 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-11499483

RESUMO

A new rapid and sensitive high-performance liquid chromatographic method for analysis of docetaxel (Taxotere) in human plasma was developed and validated. After adding an internal standard (paclitaxel, Taxol), plasma was extracted following a simple liquid-liquid extraction with diethyl ether. Extraction efficiency averaged 95% for docetaxel. Separation was performed using a Nucleosil (C18) 5 microm column, monitored at 227 nm. The isocratic mobile phase consisted of acetonitrile-acetate buffer, pH 5-tetrahydrofuran (45:50:5, v/v) pumped at a flow-rate of 1.8 ml/min. The limit of quantification for docetaxel in plasma was 12.5 ng/ml. Retention times for docetaxel and paclitaxel were 7.7 and 9 min, respectively. Standard curves were linear over a range of 25-1,000 ng/ml. This new method is rapid since it does not require time-consuming extraction procedures, or complex chromatographic conditions. This rapidity, along with the lack of chromatographic interferences with various other drugs likely to be administered to the cancer patients (pain killers, corticoids, antiemetics drugs) make this method suitable for daily routine analysis of Taxotere, a major anticancer drug extensively used in clinical oncology.


Assuntos
Antineoplásicos Fitogênicos/sangue , Cromatografia Líquida de Alta Pressão/métodos , Paclitaxel/análogos & derivados , Paclitaxel/sangue , Taxoides , Antineoplásicos Fitogênicos/farmacocinética , Docetaxel , Humanos , Paclitaxel/farmacocinética , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Espectrofotometria Ultravioleta
19.
J Clin Pharm Ther ; 29(4): 307-15, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15271097

RESUMO

BACKGROUND: Dihydropyrimidine dehydrogenase (DPD) gene polymorphism may lead to severe toxicity with 5-fluorouracil (5-FU), a major anticancer drug extensively used in clinical oncology. Drug monitoring combined with early detection of patients at risk would enable timely dose adaptation so as to maintain drug concentrations within a therapeutic window. However, the best method to identify such patients remains to be determined. OBJECTIVE: The aim of this study was to develop a rapid and simple high-performance liquid chromatographic (HPLC) method for estimating uracil/dihydrouracil (U/UH2) ratio in plasma, as an index of DPD status, and for assaying 5-FU as part of drug level monitoring. METHOD: Assay of 5-FU, and U/UH2 detection were performed on a HPLC system equipped with UV detector. Analytes were separated at room temperature using a 5 microm particles, 25 cm RP-18 X-Terra column. The mobile-phase consisted of a KH(2)PO(4) salt solution (0.05 m) + 0.1% triethylamine (TEA) pumped at 0.4 mL/min. Detection of 5-FU and 5-bromouracil were performed at 254 nm; U and UH2 elution was monitored at 210 nm. RESULTS: The method was sensitive and specific for assaying 5-FU within the 5-500 ng/mL concentration range, which covers exposure levels currently met in clinical practice. The method was simple, and relatively cheap, and rapid, with an analytical run time of about 30 min. Data from a patient with 5-FU toxicity suggest that the method was capable of identifying DPD metabolic phenotype in cancer patients, based on measurement of plasma U/UH2 ratio. CONCLUSION: The method described should be suitable both for detecting patients at high risk of 5-FU toxicity, and for drug level monitoring during chemotherapy.


Assuntos
Antimetabólitos Antineoplásicos/sangue , Antineoplásicos/metabolismo , Cromatografia Líquida de Alta Pressão/métodos , Di-Hidrouracila Desidrogenase (NADP)/metabolismo , Fluoruracila/sangue , Uracila/análogos & derivados , Uracila/sangue , Antimetabólitos Antineoplásicos/efeitos adversos , Antimetabólitos Antineoplásicos/farmacocinética , Di-Hidrouracila Desidrogenase (NADP)/genética , Feminino , Fluoruracila/efeitos adversos , Fluoruracila/farmacocinética , Humanos , Pessoa de Meia-Idade , Polimorfismo Genético
20.
Br J Cancer ; 80(11): 1726-33, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10468288

RESUMO

5-Fluorouracil (5-FU) and 5'-deoxy-5-fluorouridine (5'-DFUR), a prodrug of 5-FU, are anticancer agents activated by thymidine phosphorylase (TP). Transfecting the human TP cDNA into cancer cells in order to sensitize them to these pyrimidine antimetabolites may be an important approach in human cancer gene therapy research. In this study, an expression vector containing the human TP cDNA (pcTP5) was transfected into LS174T human colon carcinoma cells. Eight stable transfectants were randomly selected and analysed. The cytotoxic effects of 5-FU and 5'-DFUR were higher in TP-transfected cells as compared to wild-type cells. The maximal decreases in the IC50 were 80-fold for 5-FU and 40-fold for 5'-DFUR. The increase in sensitivity to these pyrimidines of TP-transfected cells significantly correlated with the increase in both TP activity and TP expression. Transfected clone LS174T-c2 but not wild-type cells exhibited formation of [3H]FdUMP from [3H]5-FU. In addition the LS174T-c2 clone enhanced the cytotoxic effect of 5'-DFUR, but also that of 5-FU, towards co-cultured parental cells. For both anti-cancer agents, this bystander effect did not require cell-cell contact. These results show that both 5-FU or 5'-DFUR could be used together with a TP-suicide vector in cancer gene therapy.


Assuntos
Antineoplásicos/toxicidade , Sobrevivência Celular/efeitos dos fármacos , Floxuridina/toxicidade , Fluoruracila/toxicidade , Timidina Fosforilase/metabolismo , Adenocarcinoma , Sobrevivência Celular/fisiologia , Clonagem Molecular , Neoplasias Colorretais , DNA Complementar , Fluoruracila/farmacocinética , Humanos , Proteínas Recombinantes/metabolismo , Timidina Fosforilase/genética , Transfecção , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA