Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
2.
Cancers (Basel) ; 15(4)2023 Feb 10.
Article in English | MEDLINE | ID: mdl-36831478

ABSTRACT

PURPOSE: To perform a scoping review on the use of Patient-Reported Outcome Measures (PROMs) in randomized trials on systemic therapy in patients with metastatic colorectal cancer (mCRC) between 2010 and 2021. METHODS: First, a search on clinicaltrials.gov was performed, looking for randomized trials in mCRC. The use of PROMs was analyzed quantitatively. Subsequently, we assessed the completeness of PROM reporting based on the CONSORT PRO extension in publications related to the selected trials acquired using Embase and PubMed. RESULTS: A total of 46/176 trials were registered on clinicaltrials.gov used PROMs. All these trials used validated PROM instruments. The EORTC QLQ-C30 was most frequently used (37 times), followed by the EQ-5D (21 times) and the EORTC QLQ-CR29 (six times). A total of 56/176 registered trials were published. In 35% (n = 20), the results of the PROMs were available. Overall, 7/20 (35%) trials documented all items of the CONSORT PRO extension and quality of reporting according to the CONSORT PRO extension was higher than in the period 2004-2012. In 3/20 (15%) of the published trials, the results of PROMs were not discussed nor included in the positioning of the new treatment compared to the reference treatment. CONCLUSION: When PROMs are used, the quality of reporting on patient-reported outcomes is improving, but this must continue in order to optimize the translation of trial results to individual patient values.

3.
Clin Pharmacokinet ; 56(4): 317-337, 2017 04.
Article in English | MEDLINE | ID: mdl-27641154

ABSTRACT

Cancer treatment is becoming more and more individually based as a result of the large inter-individual differences that exist in treatment outcome and toxicity when patients are treated using population-based drug doses. Polymorphisms in genes encoding drug-metabolizing enzymes and transporters can significantly influence uptake, metabolism, and elimination of anticancer drugs. As a result, the altered pharmacokinetics can greatly influence drug efficacy and toxicity. Pharmacogenetic screening and/or drug-specific phenotyping of cancer patients eligible for treatment with chemotherapeutic drugs, prior to the start of anticancer treatment, can identify patients with tumors that are likely to be responsive or resistant to the proposed drugs. Similarly, the identification of patients with an increased risk of developing toxicity would allow either dose adaptation or the application of other targeted therapies. This review focuses on the role of genetic polymorphisms significantly altering the pharmacokinetics of anticancer drugs. Polymorphisms in DPYD, TPMT, and UGT1A1 have been described that have a major impact on the pharmacokinetics of 5-fluorouracil, mercaptopurine, and irinotecan, respectively. For other drugs, however, the association of polymorphisms with pharmacokinetics is less clear. To date, the influence of genetic variations on the pharmacokinetics of the increasingly used monoclonal antibodies has hardly been investigated. Some studies indicate that genes encoding the Fcγ-receptor family are of interest, but more research is needed to establish if screening before the start of therapy is beneficial. Considering the profound impact of polymorphisms in drug transporters and drug-metabolizing enzymes on the pharmacokinetics of chemotherapeutic drugs and hence, their toxicity and efficacy, pharmacogenetic and pharmacokinetic profiling should become the standard of care.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Genotype , Neoplasms/genetics , Neoplasms/metabolism , Polymorphism, Single Nucleotide/genetics , Animals , Antineoplastic Agents/therapeutic use , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Genetic Variation/drug effects , Genetic Variation/physiology , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Humans , Neoplasms/drug therapy , Pharmacogenetics/methods , Pharmacogenetics/trends , Polymorphism, Single Nucleotide/drug effects
4.
Br J Clin Pharmacol ; 81(3): 553-61, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26551538

ABSTRACT

AIM: Dihydropyrimidine dehydrogenase (DPD) deficiency can lead to severe toxicity following 5-fluorouracil (5FU) or capecitabine (CAP) treatment. Uracil (U) can be used as a probe to determine systemic DPD activity. The present study was performed to assess the sensitivity and specificity of a U loading dose for detecting DPD deficiency. METHODS: Cancer patients with Common Toxicity Score (CTC) grade III or IV toxicity after the first or second cycle of 5-FU or CAP treatment were asked to participate. Based on DPD activity in PBMCs, patients were divided into two groups: DPD activity in peripheral blood mononuclear cells (PBMCs) <5 nmol mg(-1) *h(-1) (deficient group) and ≥ 5 nmol mg(-1) *h(-1) . U 500 mg m(-2) was administered orally and plasma concentrations of U and dihydrouracil (DHU) were determined. In the deficient group, polymerase chain reaction amplification of all 23 coding exons and flanking intronic regions of DPYD was performed. A U pharmacokinetic model was developed and used to determine the maximum enzymatic conversion capacity (Vmax ) of the DPD enzyme for each patient. The sensitivity and specificity of Vmax, U concentration and the U/DHU concentration ratio were determined. RESULTS: A total of 47 patients were included (19 DPD deficient, 28 DPD normal). Of the pharmacokinetic parameters investigated, a sensitivity and specificity of 80% and 98%, respectively, was obtained for the U/DHU ratio at t = 120 min. CONCLUSIONS: The high sensitivity of the U/DHU ratio at t = 120 min for detecting DPD deficiency, as defined by DPD activity in PBMCs, showed that the oral U loading dose can effectively identify patients with reduced DPD activity.


Subject(s)
Dihydropyrimidine Dehydrogenase Deficiency/diagnosis , Dihydrouracil Dehydrogenase (NADP)/metabolism , Uracil/administration & dosage , Uracil/pharmacokinetics , Administration, Oral , Case-Control Studies , Dihydropyrimidine Dehydrogenase Deficiency/blood , Female , Humans , Leukocytes, Mononuclear , Male , Middle Aged , Predictive Value of Tests , Sensitivity and Specificity , Uracil/analogs & derivatives , Uracil/blood
5.
Cancer Chemother Pharmacol ; 76(1): 47-52, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25957957

ABSTRACT

PURPOSE: Dihydropyrimidine dehydrogenase (DPD) deficiency can lead to severe toxicity in patients treated with a standard dose of a fluoropyrimidine such as 5-fluorouracil or capecitabine (CAP). Administration of oral uracil and subsequent measurement of uracil and dihydrouracil (DHU) plasma concentrations has been used to identify patients with DPD deficiency. Liver metastasis might influence systemic DPD activity. The aim of the study was to investigate the effect of metastatic disease on the pharmacokinetics of uracil and DHU after oral administration of uracil. METHODS: 500 mg/m(2) uracil was administered orally to 12 subjects with stages II-III colorectal cancer (CRC) who were treated in the adjuvant setting and to 12 subjects with stage IV metastasized CRC, all treated with CAP containing therapy. All subjects had a normal DPD activity defined as >6 nmol/mg/h determined in peripheral blood mononuclear cells. RESULTS: The mean uracil clearance [CL 51.7 (SD 6.4) vs. 46.7 (SD 13.0) l/h], area under the curve [AUC0-220min 20.6 (SD 6.4) vs. 21.0 (SD 5.7) h mg/l], elimination half-life [t 1/2 21 (SD 7) vs. 21 (SD 8) min], maximum concentration time [T max 27 (SD 9) vs. 25 (SD 9) min], volume of distribution [V 26.58 (SD 10.11) vs. 21.10 (SD 8.48) l] and the elimination constant [k el 2.01 (SD 0.56) vs. 2.41 (SD 0.72) h(-1)] did not differ significantly (p > 0.05) non-metastatic CRD versus metastatic CRC. CONCLUSIONS: Metastasis does not alter uracil pharmacokinetics and is similar in CRC patients with and without metastasis. Therefore, the uracil test dose could be used as a DPD phenotype test in both adjuvantly treated and metastatic CRC patients using similar cutoff criteria to identify patients with DPD deficiency.


Subject(s)
Colorectal Neoplasms/enzymology , Uracil/pharmacokinetics , Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bevacizumab , Capecitabine , Colorectal Neoplasms/blood , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Dihydropyrimidine Dehydrogenase Deficiency/blood , Dihydropyrimidine Dehydrogenase Deficiency/enzymology , Dihydrouracil Dehydrogenase (NADP)/blood , Fluorouracil/administration & dosage , Fluorouracil/analogs & derivatives , Humans , Neoplasm Metastasis , Neoplasm Staging , Organoplatinum Compounds/administration & dosage , Oxaliplatin , Uracil/administration & dosage , Uracil/analogs & derivatives , Uracil/blood
6.
BMJ Open ; 4(10): e006082, 2014 Oct 08.
Article in English | MEDLINE | ID: mdl-25296655

ABSTRACT

INTRODUCTION: Nursing home residents often have a high number of comorbidities resulting in polypharmacy. Inappropriate prescribing is therefore likely to occur, which in turn is expected to worsen cognitive impairment, to increase the fall risk and to decrease residents' quality of life. The objective of the 'Discontinuing Inappropriate Medication in Nursing Home Residents' (DIM-NHR) study is to examine the efficacy and cost-effectiveness of the Multidisciplinary Multistep Medication Review (3MR) that is aimed at optimising prescribing and discontinuing inappropriate medication. METHODS: A cluster randomised controlled trial will be conducted. Elderly care physicians and their wards (clusters) will be randomised. Data will be collected at baseline and 4 months after the 3MR has taken place. Six hundred nursing home residents will be recruited of whom more than half are expected to suffer from dementia. The 3MR will be based on consensus criteria and the relevant literature and will be performed by the patient's elderly care physician in collaboration with a pharmacist. ANALYSIS: Primary outcomes-the difference in proportion of residents who successfully discontinued inappropriate medication between the intervention and control group at follow-up. Secondary outcomes-undertreatment, exposure to anticholinergic and sedative medicines, neuropsychiatric symptoms, cognitive function, falls, hospital admission, quality of life and cost-effectiveness. ETHICS AND DISSEMINATION: Participant burden will be kept at a minimum. The elderly care physician will remain free to adjust medication when symptoms relapse or adverse events occur, rendering serious adverse events highly unlikely. Study findings will be published in peer-reviewed journals and a 3MR toolkit will be developed. TRIAL REGISTRATION NUMBER: This study has been registered at http://www.ClinicalTrials.gov (trial registration number: NCT01876095).


Subject(s)
Dementia , Drug Utilization Review/methods , Inappropriate Prescribing/prevention & control , Nursing Homes , Polypharmacy , Accidental Falls/prevention & control , Aged , Aged, 80 and over , Cholinergic Antagonists/therapeutic use , Disease Progression , Hospitalization/economics , Humans , Hypnotics and Sedatives/therapeutic use , Inappropriate Prescribing/economics , Quality Improvement , Quality of Life
7.
Pharmacogenomics ; 14(7): 799-811, 2013 May.
Article in English | MEDLINE | ID: mdl-23651027

ABSTRACT

5-fluorouracil (5-FU) remains the cornerstone of all currently applied regimens for the treatment of patients with cancers of the gastrointestinal tract, breast, and head and neck. Unfortunately, a large variation in the clearance of 5-FU has been observed between patients, suggesting that some patients might receive nonoptimal 5-FU doses. However, therapeutic drug monitoring of 5-FU has been shown to result in reduced intra- and inter-individual variability in 5-FU plasma levels and pharmacokinetically guided dose adjustments of 5-FU-containing therapy results in a significantly improved efficacy and tolerability. To date, compartmental Michaelis-Menten elimination-based modeling has proven to be a sensitive and accurate tool for analyzing the pharmacokinetics of 5-FU and to identify patients with a dihydropyrimidine dehydrogenase deficiency. These Michaelis-Menten models also allow the use of a limited sampling strategy and offer the opportunity to predict a priori the 5-FU plasma concentrations in patients receiving adapted doses of 5-FU.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacokinetics , Fluorouracil/administration & dosage , Fluorouracil/pharmacokinetics , Neoplasms/drug therapy , Neoplasms/metabolism , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Dihydropyrimidine Dehydrogenase Deficiency/metabolism , Dihydrouracil Dehydrogenase (NADP)/metabolism , Drug Monitoring/methods , Humans
8.
Clin Pharmacokinet ; 51(3): 163-74, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22339448

ABSTRACT

BACKGROUND AND OBJECTIVE: Dihydropyrimidine dehydrogenase (DPD) is the initial enzyme in the catabolism of 5-fluorouracil (5FU) and DPD deficiency is an important pharmacogenetic syndrome. So far, only very limited information is available regarding the pharmacokinetics of 5FU in patients with a (partial) DPD deficiency and no limited sampling models have been developed taking into account the non-linear pharmacokinetic behaviour of 5FU. The aim of this study was to evaluate the pharmacokinetics of 5FU and to develop a limited sampling strategy to detect decreased 5FU elimination in patients with a c.1905+1G>A-related DPD deficiency. METHODS: Thirty patients, heterozygous for the c.1905+1G>A mutation in DPYD, and 18 control patients received a dose of 5FU 300 mg/m2 and/or 5FU 450 mg/m2, followed by pharmacokinetic analysis of the 5FU plasma levels. A population pharmacokinetic analysis was performed in order to develop a compartmental pharmacokinetic model suitable for a limited sampling strategy. Clinical aspects of treating DPD-deficient patients with 5FU-based chemotherapy were assessed from the retrospectively collected clinical data. RESULTS: In a two-compartment model with Michaelis-Menten elimination, the mean maximum enzymatic conversion capacity (V(max)) value was 40% lower in DPD-deficient patients compared with controls (p < 0.001). Using a limited sampling strategy, with V(max) values calculated from 5FU concentrations at 30 or 60 minutes, significant differences were observed between DPD-deficient patients and controls at both dose levels (p < 0.001). The positive predictive value and negative predictive value for V(max), calculated from 5FU levels at 60 minutes, were 96% and 88%, respectively, in patients treated with a single dose of 5FU 300 mg/m2. All seven DPD-deficient patients (two males and five females) who had been genotyped prior to initiation of standard 5FU-containing chemotherapy developed grade 3-4 toxicity, with one case of lethal toxicity in a female patient. No grade 4 toxicity or lethal outcome was observed in 13 DPD-deficient patients treated with reduced doses of 5FU. The average dose of 5FU in DPD-deficient patients with mild toxicity (grade ≤2) was 61 ± 16% of the normal 5FU dose (n = 10). CONCLUSIONS: Profound differences in the elimination of 5FU could be detected between DPD-deficient patients and control patients. Pharmacokinetic 5FU profiling, using a single 5FU concentration at 60 minutes, may be useful for identification of DPD-deficient patients in order to reduce severe toxicity. Furthermore, treatment of DPD-deficient patients with standard 5FU-containing chemotherapy was associated with severe (lethal) toxicity.


Subject(s)
Antimetabolites, Antineoplastic/pharmacokinetics , Bayes Theorem , Dihydropyrimidine Dehydrogenase Deficiency/genetics , Dihydrouracil Dehydrogenase (NADP)/genetics , Drug Monitoring/methods , Fluorouracil/pharmacokinetics , Models, Biological , Mutation , Neoplasms/drug therapy , Adult , Aged , Antimetabolites, Antineoplastic/adverse effects , Antimetabolites, Antineoplastic/blood , Biotransformation , Dihydropyrimidine Dehydrogenase Deficiency/blood , Dihydropyrimidine Dehydrogenase Deficiency/enzymology , Dihydrouracil Dehydrogenase (NADP)/metabolism , Female , Fluorouracil/adverse effects , Fluorouracil/blood , Heterozygote , Humans , Linear Models , Logistic Models , Male , Metabolic Clearance Rate , Middle Aged , Neoplasms/blood , Netherlands , Pharmacogenetics , Phenotype , ROC Curve , Retrospective Studies
9.
Cancer Chemother Pharmacol ; 68(6): 1611-7, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21590448

ABSTRACT

PURPOSE: Dihydropyrimidine dehydrogenase (DPD) deficiency can lead to severe toxicity in patients treated with standard doses of 5-fluorouracil (5-FU). Oral uracil administration and subsequent measurement of uracil and dihydrouracil (DHU) plasma concentrations might detect patients with DPD deficiency. This study compares the pharmacokinetics of uracil and DHU after oral uracil administration in subjects with normal and deficient DPD status. METHODS: Five hundred milligrams of uracil per metre square was administered orally to 11 subjects with normal DPD status and to 10 subjects with reduced DPD activity. Repeated administration (n = 3) of this dose was performed in 4 subjects, and 1,000 mg uracil/m(2) was administered to 4 subjects to assess intra-individual variation and linearity of pharmacokinetics. RESULTS: In subjects with normal DPD status, 500 mg/m(2) uracil resulted in uracil C (max) levels of 14.4 ± 4.7 mg/L at T (max) = 30.0 ± 11.6 min, and in DPD-deficient subjects, 20.0 ± 4.5 mg/L at 31.5 ± 1.1 min. The uracil AUC(0>180) was 31.2 ± 5.1 mg L/h in DPD-deficient subjects, which was significantly higher (P < 0.05) than in the subjects with normal DPD status (13.8 ± 3.9 mg L/h). Repeated uracil dosing showed reproducible uracil PK in subjects with normal DPD status, and dose elevation of uracil suggested linear pharmacokinetics. CONCLUSION: The pharmacokinetics of uracil differs significantly between subjects with a normal DPD activity and those with a deficient DPD status. The AUC and C (max) of uracil can be useful as a diagnostic tool to differentiate patients with regard to DPD status.


Subject(s)
Dihydropyrimidine Dehydrogenase Deficiency/diagnosis , Uracil/pharmacokinetics , Administration, Oral , Adult , Aged , Dihydropyrimidine Dehydrogenase Deficiency/metabolism , Female , Humans , Male , Middle Aged , Uracil/administration & dosage
10.
Hum Genet ; 128(5): 529-38, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20803296

ABSTRACT

Dihydropyrimidine dehydrogenase (DPD) is the initial enzyme acting in the catabolism of the widely used antineoplastic agent 5-fluorouracil (5FU). DPD deficiency is known to cause a potentially lethal toxicity following administration of 5FU. Here, we report novel genetic mechanisms underlying DPD deficiency in patients presenting with grade III/IV 5FU-associated toxicity. In one patient a genomic DPYD deletion of exons 21-23 was observed. In five patients a deep intronic mutation c.1129-5923C>G was identified creating a cryptic splice donor site. As a consequence, a 44 bp fragment corresponding to nucleotides c.1129-5967 to c.1129-5924 of intron 10 was inserted in the mature DPD mRNA. The deleterious c.1129-5923C>G mutation proved to be in cis with three intronic polymorphisms (c.483 + 18G>A, c.959-51T>G, c.680 + 139G>A) and the synonymous mutation c.1236G>A of a previously identified haplotype. Retrospective analysis of 203 cancer patients showed that the c.1129-5923C>G mutation was significantly enriched in patients with severe 5FU-associated toxicity (9.1%) compared to patients without toxicity (2.2%). In addition, a high prevalence was observed for the c.1129-5923C>G mutation in the normal Dutch (2.6%) and German (3.3%) population. Our study demonstrates that a genomic deletion affecting DPYD and a deep intronic mutation affecting pre-mRNA splicing can cause severe 5FU-associated toxicity. We conclude that screening for DPD deficiency should include a search for genomic rearrangements and aberrant splicing.


Subject(s)
Antimetabolites, Antineoplastic/toxicity , Dihydrouracil Dehydrogenase (NADP)/genetics , Fluorouracil/toxicity , Introns , Mutation, Missense , RNA Splicing , Sequence Deletion , Adult , Aged , Comparative Genomic Hybridization , Dihydrouracil Dehydrogenase (NADP)/metabolism , Female , Gene Rearrangement , Humans , Male , Middle Aged , Polymerase Chain Reaction , Polymorphism, Genetic , RNA, Messenger , Sequence Analysis, DNA
11.
Eur J Clin Pharmacol ; 66(6): 611-7, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20213492

ABSTRACT

OBJECTIVE: To study the impact of the 79A>C polymorphism in the cytidine deaminase (CDA) gene on the pharmacokinetics of gemcitabine and its metabolite 2',2'-difluorodeoxyuridine (dFdU) in non-small-cell lung cancer (NSCLC) patients. PATIENTS AND METHODS: Patients (n = 20) received gemcitabine 1,125 mg/m(2) as a 30 min i.v. infusion as part of treatment for NSCLC. Plasma samples were collected during 0-6 h after gemcitabine administration. Gemcitabine and dFdU were quantified by high performance liquid chromatography with ultraviolet detection. The CDA 79A>C genotype was determined with PCR and DNA sequencing. RESULTS: Gemcitabine was rapidly cleared from plasma and undetectable after 3 h. The allele frequency of the 79A>C polymorphism was 0.40. Diplotypes were distributed as A/A n = 8, A/C n = 8 ,and C/C n = 4. No significant differences were found between the different CDA genotypes and gemcitabine or dFdU AUC, clearance, or half-life. CONCLUSION: The 79A>C polymorphism in the CDA gene does not have a major consistent and signficant impact on gemcitabine pharmacokinetics.


Subject(s)
Antimetabolites, Antineoplastic/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/blood , Cytidine Deaminase/genetics , Deoxycytidine/analogs & derivatives , Lung Neoplasms/blood , Polymorphism, Single Nucleotide , Adult , Aged , Antimetabolites, Antineoplastic/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacokinetics , Female , Floxuridine/analogs & derivatives , Floxuridine/pharmacokinetics , Gene Frequency , Genotype , Humans , Lung Neoplasms/drug therapy , Male , Metabolic Clearance Rate , Middle Aged , Gemcitabine
12.
Ther Drug Monit ; 32(3): 364-8, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20335827

ABSTRACT

AIM: The excretion in saliva of gemcitabine and its metabolite 2',2'-difluorodeoxyuridine (dFdU) as well as epirubicin (Epi) and its metabolite epirubicinol (Epi-ol) was studied in patients with non-small cell lung cancer, treated with gemcitabine plus epirubicin. METHODS: Patients (n = 12) were treated with gemcitabine 1125 mg/m, followed by Epi 100 mg/m. Blood, saliva, and oral mucosa cells were collected during 22 hours for analysis of gemcitabine, Epi, and their metabolites. Gemcitabine, dFdU, Epi, and Epi-ol were quantified by high-performance liquid chromatography. RESULTS: Gemcitabine was cleared rapidly from plasma and undetectable after 3 hours in all patients. Gemcitabine was detectable in saliva during only the first hour after infusion. The Cmax in saliva was 0.66 +/- 0.61 mg/L, and the saliva to plasma ratio (S/P ratio) was 0.038 +/- 0.037. The Cmax of dFdU was reached 1.5-2 hours after gemcitabine infusion and was 1.03 +/- 0.63 mg/L. The dFdU S/P ratios gradually increased from 0.021 +/- 0.013 at t = 1 hour to 0.050 +/- 0.027 at t = 6 hours after infusion. Epi displayed a triexponential plasma concentration-time profile. The Epi and Epi-ol concentrations in saliva at t = 6 hours after administration were 55 +/- 27 and 9 +/- 9 microg/L, respectively, and decreased to 28 +/- 14 and 7 +/- 4 microg/L, respectively, at t = 22 hours. The corresponding S/P ratios were 1.28 +/- 0.73 and 0.36 +/- 0.31 at t = 6 hours and 1.72 +/- 1.00 and 0.62 +/- 0.34 at t = 22 hours, respectively. The amount of Epi in mucosal cells ranged from 135-598 ng per 10 cells at t = 3 hours and decreased to 33-196 ng per 10 cells at t = 22 hours. CONCLUSION: Gemcitabine and Epi, as well as their main metabolites dFdU and Epi-ol, are excreted in detectable amounts in saliva, although their absolute concentrations remain relatively low.


Subject(s)
Antimetabolites, Antineoplastic/blood , Carcinoma, Non-Small-Cell Lung/pathology , Deoxycytidine/analogs & derivatives , Epirubicin/blood , Saliva/chemistry , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/metabolism , Chromatography, High Pressure Liquid/methods , Deoxycytidine/blood , Deoxycytidine/metabolism , Epirubicin/metabolism , Humans , Infusions, Intravenous , Lung Neoplasms/blood , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Gemcitabine
13.
Ther Drug Monit ; 27(1): 25-30, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15665742

ABSTRACT

The authors developed a simple and sensitive, fully validated HPLC-UV method for the determination of both 5-FU and its metabolite DHFU in small-volume plasma samples. The analytes were separated on a 4.6 x 250 mm ID Atlantis dC18 5-microm column with isocratic elution at room temperature. Chlorouracil was used as internal standard. The analytes were detected with an UV diode array detector. DHFU was detected at 205 nm, 5-FU at 266 nm, and chlorouracil at both wavelengths. The limits of quantification in plasma were 0.040 mug /mL for 5-FU and 0.075 microg/mL for DHFU. Linearity, accuracy, precision, recovery, dilution, freeze-thaw stability, and stability in the sample compartment were evaluated. The method appeared linear over a range from 0.04 to 15.90 microg/mL for 5-FU and from 0.075 to 3.84 microg/mL for DHFU. The method appeared very suitable for therapeutic drug monitoring and pharmacokinetic studies of 5-FU because of its simple extraction and small sample volume. Problems in earlier published methods with interfering peaks and variable retention times were overcome. The method appeared also to be suitable for detection of uracil and its metabolite dihydrouracil in plasma.


Subject(s)
Chromatography, High Pressure Liquid/methods , Fluorouracil/analogs & derivatives , Fluorouracil/blood , Spectrophotometry, Ultraviolet/methods , Dihydropyrimidine Dehydrogenase Deficiency , Dihydrouracil Dehydrogenase (NADP)/genetics , Drug Monitoring/methods , Drug Monitoring/trends , Fluorouracil/chemistry , Fluorouracil/therapeutic use , Humans
14.
Ther Drug Monit ; 25(4): 433-40, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12883225

ABSTRACT

We present a high-performance liquid chromatography (HPLC) method suitable for the analysis of epirubicin and its metabolite epirubicinol in saliva and plasma. Preparation of saliva and plasma samples was performed by extraction of analytes with a chloroform:2-propanol mixture (6:1, vol/vol) and evaporation of the organic phase to dryness under vacuum at a temperature of approximately 45 degrees C. The chromatographic analysis was carried out by reversed-phase isocratic elution of the anthracyclines with a Chromsep stainless steel HPLC column (150 x 4.6 mm I.D.) filled with Nucleosil 100 S C(18) material, particle size 5 micro m. The detection was accomplished by spectrofluorimetry at excitation and emission wavelengths of 474 and 551 nm, respectively. The anthracyclines eluted within 10 min of injection, and the method appeared to be specific. The method is linear over a concentration range of 5 to 1000 micro g/L for epirubicin and 2 to 400 micro g/L for epirubicinol (r > 0.99) in both saliva and plasma. The recoveries from saliva and plasma of epirubicin, epirubicinol, and the internal standard doxorubicin were 88.9 and 69.0%, 87.6 and 77.3%, and 80 and 67.9%, respectively. The lower limit of quantification was 5 micro g/L for epirubicin and 2 micro g/L for epirubicinol. The method proved to be precise and accurate, as the within-day and between-day coefficients of variation were less than 10%. Overall results indicate that our method is suitable for the bioanalysis of epirubicin and epirubicinol in saliva as well as plasma.


Subject(s)
Antibiotics, Antineoplastic/analysis , Doxorubicin/analogs & derivatives , Doxorubicin/analysis , Epirubicin/analysis , Epirubicin/blood , Saliva/chemistry , Antibiotics, Antineoplastic/blood , Calibration , Chromatography, High Pressure Liquid , Doxorubicin/blood , Humans , Linear Models , Sensitivity and Specificity , Spectrometry, Fluorescence
15.
Cancer Chemother Pharmacol ; 51(2): 167-73, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12647019

ABSTRACT

PURPOSE: The influence of liver metastases on the pharmacokinetics of 5-fluorouracil (5-FU) and its metabolite 5,6-dihydrofluorouracil (DHFU) was studied in patients with liver metastases from gastrointestinal cancer ( n=16) and compared with a control group of patients with nonmetastatic gastrointestinal cancer ( n=18). METHODS: Patients were assigned to two different groups based on the presence of liver metastases. The percentage of hepatic replacement was determined with CT and ultrasonography and classified as <25%, 25-50% or >50% of the total liver volume. Chemotherapy consisted of leucovorin 20 mg/m(2) per day plus 5-FU 425 mg/m(2) per day, both for 5 days. Blood sampling was carried out on the first day of the first chemotherapy cycle. 5-FU and DHFU were quantified in plasma by HPLC. A four-compartment parent drug-metabolite model with nonlinear Michaelis-Menten elimination from the central compartment of the parent drug (5-FU) was applied to describe 5-FU and DHFU pharmacokinetics. RESULTS: No effect of liver metastases on 5-FU clearance was observed. The effects of 18 covariables on pharmacokinetic parameters were also studied in a univariate correlation analysis. Body surface area was positively correlated with the distribution volume of 5-FU in the central compartment and with V(max) ( r=0.65 and r=0.54, respectively). CONCLUSIONS: There is no need for dose adjustment of 5-FU as a standard procedure in patients with liver metastases and mild to moderate elevations in liver function tests.


Subject(s)
Fluorouracil/analogs & derivatives , Fluorouracil/pharmacokinetics , Liver Neoplasms/secondary , Aged , Female , Fluorouracil/adverse effects , Gastrointestinal Neoplasms/pathology , Humans , Liver/physiopathology , Liver Neoplasms/metabolism , Liver Neoplasms/physiopathology , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...