Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Br J Cancer ; 129(9): 1373-1382, 2023 10.
Article in English | MEDLINE | ID: mdl-37524968

ABSTRACT

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.


Subject(s)
Antineoplastic Agents , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Mice , Animals , Pemetrexed , Cisplatin , Lung Neoplasms/pathology , T-Lymphocytes, Regulatory , CD8-Positive T-Lymphocytes
2.
ESMO Open ; 8(3): 101577, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37267808

ABSTRACT

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.


Subject(s)
Dihydropyrimidine Dehydrogenase Deficiency , Dihydrouracil Dehydrogenase (NADP) , Humans , Dihydrouracil Dehydrogenase (NADP)/genetics , Dihydropyrimidine Dehydrogenase Deficiency/complications , Dihydropyrimidine Dehydrogenase Deficiency/chemically induced , Dihydropyrimidine Dehydrogenase Deficiency/drug therapy , Antimetabolites, Antineoplastic/adverse effects , Fluorouracil/therapeutic use , Uracil/therapeutic use
3.
ESMO Open ; 7(6): 100646, 2022 12.
Article in English | MEDLINE | ID: mdl-36521418

ABSTRACT

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.


Subject(s)
Antineoplastic Agents , Head and Neck Neoplasms , Uterine Cervical Neoplasms , Male , Humans , Female , Middle Aged , Vinorelbine/pharmacology , Motion Pictures , Antineoplastic Agents/adverse effects , Head and Neck Neoplasms/chemically induced
6.
Sci Rep ; 10(1): 4147, 2020 03 05.
Article in English | MEDLINE | ID: mdl-32139753

ABSTRACT

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.


Subject(s)
Docetaxel/chemistry , Liposomes/chemistry , Trastuzumab/chemistry , Analysis of Variance , Flow Cytometry , Nanoparticles/chemistry
9.
Crit Rev Oncol Hematol ; 127: 1-5, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29891106

ABSTRACT

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.


Subject(s)
Biomarkers, Tumor , Immunotherapy/adverse effects , Neoplasms/diagnosis , Neoplasms/therapy , Pain/etiology , Precision Medicine , Biomarkers, Pharmacological/analysis , Biomarkers, Tumor/analysis , Biomarkers, Tumor/physiology , Drug-Related Side Effects and Adverse Reactions/diagnosis , Drug-Related Side Effects and Adverse Reactions/physiopathology , Genomics/methods , Humans , Pain/diagnosis , Precision Medicine/adverse effects , Precision Medicine/methods , Precision Medicine/trends , Prognosis , Treatment Outcome
11.
J Chemother ; 22(2): 129-33, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20435574

ABSTRACT

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.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm , Fluorouracil/analogs & derivatives , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Capecitabine , Cell Line, Tumor , Cell Proliferation , Deoxycytidine/pharmacology , Dihydrouracil Dehydrogenase (NADP)/biosynthesis , Drug Interactions , ErbB Receptors/biosynthesis , Erlotinib Hydrochloride , Fluorouracil/pharmacology , Humans , Pancreatic Neoplasms , Thymidine Phosphorylase/biosynthesis , Vascular Endothelial Growth Factor A/biosynthesis
12.
Curr Med Chem ; 16(33): 4361-71, 2009.
Article in English | MEDLINE | ID: mdl-19835568

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/administration & dosage , Liposomes/chemistry , Antibodies, Immobilized/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Drug Carriers , Nanotechnology , Phospholipids/chemistry
13.
Br J Cancer ; 97(7): 919-26, 2007 Oct 08.
Article in English | MEDLINE | ID: mdl-17848948

ABSTRACT

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.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm , Fluorouracil/pharmacology , Liposomes , Animals , Cell Cycle/drug effects , Cell Proliferation/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Female , Humans , In Vitro Techniques , Mice , Mice, Nude , Thymidylate Synthase/antagonists & inhibitors , Tumor Cells, Cultured/drug effects
14.
J Clin Pharm Ther ; 29(4): 307-15, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15271097

ABSTRACT

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.


Subject(s)
Antimetabolites, Antineoplastic/blood , Antineoplastic Agents/metabolism , Chromatography, High Pressure Liquid/methods , Dihydrouracil Dehydrogenase (NADP)/metabolism , Fluorouracil/blood , Uracil/analogs & derivatives , Uracil/blood , Antimetabolites, Antineoplastic/adverse effects , Antimetabolites, Antineoplastic/pharmacokinetics , Dihydrouracil Dehydrogenase (NADP)/genetics , Female , Fluorouracil/adverse effects , Fluorouracil/pharmacokinetics , Humans , Middle Aged , Polymorphism, Genetic
15.
J Clin Pharm Ther ; 28(5): 403-8, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14632965

ABSTRACT

Various studies suggested that cytotoxicity induced by 5-fluorouracil (5-FU) is an apoptotic mechanism possibly mediated by the Fas/FasL system. In this preliminary work, we studied retrospectively the role the Fas/FasL expression as a predictive response factor with fluoropyrimidine-based chemotherapies. We developed a real-time PCR method for measuring Fas and FasL expression in various biopsies from patients treated with a FUFOL-like protocol. No correlation was found between Fas or FasL expression and overall survival or partial response. However, the PCR assay was simple and convenient to use for quantitation of Fas/FasL in tumor biopsies.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Apoptosis/drug effects , Colonic Neoplasms/drug therapy , Fluorouracil/therapeutic use , Membrane Glycoproteins/metabolism , Adult , Aged , Antimetabolites, Antineoplastic/adverse effects , Colonic Neoplasms/mortality , Colonic Neoplasms/pathology , DNA, Complementary/genetics , Fas Ligand Protein , Female , Fluorouracil/adverse effects , Humans , Liver Neoplasms/secondary , Male , Middle Aged , Polymerase Chain Reaction , Prognosis , Retrospective Studies
16.
J Chemother ; 15(3): 266-74, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12868554

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Microsomes, Liver/drug effects , Paclitaxel/metabolism , Paclitaxel/pharmacology , Adult , Biological Availability , Cells, Cultured , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Drug Interactions , Female , Humans , Male , Observer Variation , Sensitivity and Specificity
17.
Br J Cancer ; 89(3): 585-92, 2003 Aug 04.
Article in English | MEDLINE | ID: mdl-12888834

ABSTRACT

ZD1839 ('Iressa'), an orally active, selective epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor, is currently being investigated in clinical trials as a treatment for cancer. 'Iressa' is a trademark of the AstraZeneca group of companies. We have previously demonstrated a synergistic interaction between ZD1839 and cisplatin/5-fluorouracil (5FU) in CAL33, a human head and neck cancer cell line that markedly expresses EGFR. This study examined the effects of this drug combination on the cell cycle, cell cycle regulators, apoptosis-related factors, EGFR-related signalling and DNA repair in CAL33 cells. The cells were incubated with ZD1839 alone for 48 h, then cisplatin and 5FU were added. Exposure to the drug combination continued for a further 48 h. ZD1839 alone induced accumulation of cells in the G0/G1 phase of the cell cycle at 24 h accompanied by a concomitant increase in p21, p27 and Bax, a significant decrease in Bcl2 and a decrease in Akt phosphorylation. A decrease in DNA-PK was observed at 48 h. ZD1839 alone had no effect on caspase-3 activity, but addition of ZD1839 to cisplatin-5FU led to a significant increase in caspase-3 activity at 96 h. Thus, ZD1839 affects key cellular pathways controlling cell proliferation, apoptosis and DNA repair. These data provide a rationale to support clinical trials combining ZD1839 and cisplatin-5FU and other protocols that combine EGFR-targeting agents with chemotherapy or radiotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Apoptosis , Cell Cycle/drug effects , Cisplatin/pharmacology , Cisplatin/pharmacokinetics , DNA Repair/drug effects , Fluorouracil/pharmacology , Fluorouracil/pharmacokinetics , Head and Neck Neoplasms/pathology , Quinazolines/pharmacology , Quinazolines/pharmacokinetics , Drug Interactions , Epidermal Growth Factor/antagonists & inhibitors , ErbB Receptors/biosynthesis , Gefitinib , Humans , Protein-Tyrosine Kinases/antagonists & inhibitors , Signal Transduction , Tumor Cells, Cultured
18.
J Clin Pharm Ther ; 28(2): 109-16, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12713607

ABSTRACT

OBJECTIVE: A Bayesian population pharmacokinetics study of data from routine therapeutic drug monitoring cisplatin during a 5-day infusion of cisplatin. METHODS: A total of 95 kinetics data sets (58 patients) were available to perform this study. Individual pharmacokinetic parameters, estimated from 20 courses of treatment in 18 patients, were used to calculate the population parameters (cl: 0.175 +/- 0.034 L/h; t(1/2): 327 +/- 91 h). The accuracy of Bayesian forecasting was tested by comparing in 40 other patients the clearance values calculated either from a complete kinetics profile (eight sampling times) or from three early samples and the new population parameters. Finally, drug monitoring efficacy was assessed by comparing the target Cmax values with the Cmax obtained after dose adjustment based upon early Bayesian estimation of the individual pharmacokinetic parameters. RESULTS: : No significant difference was found between Bayesian and experimental clearances. Besides, dose-individualization proved to successfully adjust Cmax values around their respective target. CONCLUSION: The new reference pharmacokinetic population parameters lead to accurate estimation of individual pharmacokinetic parameters from a limited number of samples, thus allowing efficient therapeutic drug monitoring during 5-day infusion regimens of cisplatin.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Cisplatin/pharmacokinetics , Neoplasms/metabolism , Adolescent , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Area Under Curve , Bayes Theorem , Cisplatin/administration & dosage , Cisplatin/therapeutic use , Female , Half-Life , Humans , Infusions, Intravenous , Male , Metabolic Clearance Rate , Middle Aged , Neoplasms/drug therapy
19.
J Clin Pharm Ther ; 27(3): 189-95, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12081632

ABSTRACT

BACKGROUND: Methotrexate (MTX) infusions may induce severe side-effects, and alkaline hydration along with folinic acid rescue is a common way to reduce such toxic risks. The purpose of this study was to develop an adaptive rescue strategy based upon the early detection of patients with impaired MTX elimination. METHODS AND RESULTS: In this study, we propose a simple population-based Bayesian approach for predicting MTX plasma concentration from a limited number of samples, so as to adapt both duration and dosage of the rescue agent to be used next. Ten kinetic profiles obtained after 10 courses of MTX (1.5 g/m2) in seven patients with inflammatory breast cancer were used to establish the population pharmacokinetic parameters (Cl, 8.16 L/h; t1/2, 12.7 h). This population was next involved in the Bayesian estimation of MTX individual pharmacokinetic parameters from only two blood samples (T24 and T36 h), thus allowing one to forecast the elimination of this drug by predicting MTX levels at 48 h. According to the MTX concentrations predicted during the elimination phase, folinic acid rescue was then prolonged in patients likely to be overexposed. CONCLUSION: The Bayesian estimation presented in this study was an easy and convenient method to efficiently detect patients with impaired MTX elimination in routine clinical practice. This information enabled the introduction of strategies for minimizing the risk of severe drug toxicity.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Breast Neoplasms/drug therapy , Leucovorin/administration & dosage , Methotrexate/administration & dosage , Adult , Aged , Antimetabolites, Antineoplastic/adverse effects , Antimetabolites, Antineoplastic/pharmacokinetics , Bayes Theorem , Breast Neoplasms/blood , Dose-Response Relationship, Drug , Female , Humans , Leucovorin/therapeutic use , Methotrexate/adverse effects , Methotrexate/pharmacokinetics , Middle Aged , Models, Biological , Time Factors
20.
Br J Cancer ; 86(7): 1162-8, 2002 Apr 08.
Article in English | MEDLINE | ID: mdl-11953866

ABSTRACT

The combination of 5-fluorouracil-folinic acid and oxaliplatin has led to a significant improvement of chemotherapy efficacy in advanced pretreated colorectal cancer. The objective of the present study was, considering the oxaplatin-5-fluorouracil-folinic acid combination, to examine the impact of one given drug on the cellular determinants of cytotoxic activity of the other drug. These cellular factors were analysed on the human colon cancer cell line WiDr in clinically relevant conditions of drug exposure ('De Gramont' schedule) with oxaliplatin-folinic acid during 2 h followed by 5-fluorouracil 48 h. The DNA binding of oxaliplatin was significantly reduced by the presence of 5-fluorouracil but this effect was time-dependent and after 50 h the platinum incorporated into DNA was identical in controls and in the drug combination. In the presence of oxaliplatin, there was less formation of FUH(2) which is the first catabolite produced in the cascade of 5-fluorouracil metabolic degradation. The effects of drugs on cell cycle were quite different from one drug to the other with oxaliplatin inducing a shift towards G(2) accumulation and 5-fluorouracil-folinic acid to a greater proportion of cells in G(1)-S. When oxaliplatin and 5-fluorouracil-folinic acid were combined the cell cycle effects were very similar to that of the 5-fluorouracil-folinic acid sequence alone. Oxaliplatin was able to reduce thymidylate synthase activity with a marked impact 28 h after the beginning of cell exposure to the drug. The 5-fluorouracil-folinic acid drug sequence led to a profound reduction in thymidylate synthase activity and this decrease was not markedly enhanced by the presence of oxaliplatin. Regarding apoptosis, changes in mitochondrial membrane permeability were observed in the presence of the tested drugs and the impact of 5-fluorouracil-folinic acid was greater than that of oxaliplatin. The addition of oxaliplatin did not amplify the action of 5-fluorouracil-folinic acid upon mitochondrial membrane permeability change. The presence of oxaliplatin itself did not modify the intracellular concentration of total reduced folates. The fact that oxaliplatin may reduce 5-fluorouracil catabolism could be central in explaining the supra-additive interaction between these drugs.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Colonic Neoplasms/pathology , Fluorouracil/pharmacology , Organoplatinum Compounds/pharmacology , Cell Cycle , Cell Membrane , DNA Damage , Drug Administration Schedule , Drug Interactions , Fluorouracil/administration & dosage , Fluorouracil/pharmacokinetics , Humans , Leucovorin/administration & dosage , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/pharmacokinetics , Oxaliplatin , Permeability , Thymidylate Synthase/pharmacology , Toxicity Tests , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...