Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Pediatr Hematol Oncol ; 31(8): 731-42, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25007236

ABSTRACT

A physiologically based pharmacokinetic (PBPK) model of the DNA-alkylating agent busulfan was slightly modified and scaled from adults to children in order to predict the systemic busulfan drug exposure in children. Capitalizing on the recent major software release of PK-Sim®, we refined our PBPK model by implementing glutathione S transferase (GST) in 11 organs using the software integrated enzyme expression database. In addition, two irreversible binding processes (i.e., DNA and plasma protein binding) were applied by using Koff and KD values. The model was scaled from adults to children. Simulations were computed and compared to concentration-time data after intravenous (i.v.) busulfan administration to 36 children. Based on the results, an age-dependent enzyme activity and maturation ratio was tailored and evaluated with an external dataset consisting of 23 children. Initial adult to children scaling indicated lower clearance values for children in comparison to adults. Subsequent age-dependent maturation ratio resulted in three different age groups: Activity of busulfan-glutathione conjugate formation was 80%, 61%, and 89% in comparison to adults for children with an age of up to 2 years, > 2-6 years, and > 6-18 years, respectively. Patients of the evaluation dataset were simulated with a mean percentage error (MPE) for all patients of 3.9% with 3/23 children demonstrating a MPE of > ±30%. The PBPK model parameterization sufficiently described the observed concentration-time data of the validation dataset while showing an adequate predictive performance. This PBPK model could be helpful to determine the first dose of busulfan in children.


Subject(s)
Alkylating Agents/pharmacokinetics , Busulfan/pharmacokinetics , Adolescent , Adult , Alkylating Agents/blood , Busulfan/blood , Canada , Child , Child, Preschool , Hematopoietic Stem Cell Transplantation , Humans , Infant , Infant, Newborn , Infusions, Intravenous , Models, Biological , Software , Transplantation Conditioning
2.
Cancer Chemother Pharmacol ; 73(3): 475-84, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24414550

ABSTRACT

PURPOSE: Tamoxifen is a key therapeutic option for breast cancer treatment. Understanding its complex metabolism and pharmacokinetics is important for dose optimization. We examined the possibility of utilizing archival formalin-fixed paraffin-embedded (FFPE) tissue as an alternative sample source for quantification since well-annotated retrospective samples were always limited. METHODS: Six 15 µm sections of FFPE tissues were deparaffinized with xylene and purified using solid-phase extraction. Tamoxifen and its metabolites were separated and detected by liquid chromatography-tandem mass spectrometry using multiple-reaction monitoring. RESULTS: This method was linear between 0.4 and 200 ng/g for 4-hydroxy-tamoxifen and endoxifen, and 4-2,000 ng/g for tamoxifen and N-desmethyl-tamoxifen. Inter- and intra-assay precisions were <9 %, and mean accuracies ranged from 81 to 106 %. Extraction recoveries were between 83 and 88 %. The validated method was applied to FFPE tissues from two groups of patients, who received 20 mg/day of tamoxifen for >6 months, and were classified into breast tumor recurrence and non-recurrence. Our preliminary data show that levels of tamoxifen metabolites were significantly lower in patients with recurrent cancer, suggesting that inter-individual variability in tamoxifen metabolism might partly account for the development of cancer recurrence. Nevertheless, other causes such as non-compliance or stopping therapy of tamoxifen could possibly lead to the concentration differences. CONCLUSIONS: The ability to successfully study tamoxifen metabolism in such tissue samples will rapidly increase our knowledge of how tamoxifen's action, metabolism and tissue distribution contribute to breast cancer control. However, larger population studies are required to understand the underlying mechanism of tamoxifen metabolism for optimization of its treatment.


Subject(s)
Breast Neoplasms/chemistry , Breast Neoplasms/metabolism , Chromatography, Liquid/methods , Tamoxifen/isolation & purification , Tamoxifen/metabolism , Tandem Mass Spectrometry/methods , Breast Neoplasms/drug therapy , Female , Formaldehyde , Humans , Neoplasm Recurrence, Local/chemistry , Neoplasm Recurrence, Local/metabolism , Paraffin , Pilot Projects , Retrospective Studies , Tamoxifen/analogs & derivatives , Tamoxifen/chemistry
3.
Cancer Chemother Pharmacol ; 72(5): 991-1000, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24061863

ABSTRACT

PURPOSE: A physiologically based pharmacokinetic (PBPK) model was established and evaluated describing the pharmacokinetics (PK) of the DNA-alkylating agent Busulfan in adults in order to predict the systemic Busulfan drug exposure in both plasma and toxicity-related organs. METHODS: A generic PBPK model was tailored to describe Busulfan PK by implementing compound-specific physicochemical and metabolism data. With regard to possible influences of glutathione S transferase (GST) variations on Busulfan PK, two different PBPK model parameterizations were investigated: a first parameterization with individual GST activity (expressed as different estimated V(max) values) for each patient, and a resulting second model parameterization with a mean GST activity for all patients. Simulations were computed and compared to concentration-time data after intravenous Busulfan administration to 108 adults serving as development dataset. Subsequently, appropriateness of the PBPK model was evaluated with an external dataset not used for model development, consisting of 95 adults. RESULTS: Both PBPK model parameterizations of Busulfan successfully described the observed plasma concentrations. For the validation dataset, calculated PK parameters were as follows: clearance 0.16 ± 0.03 L/h/kg and volume of distribution 0.65 ± 0.06 L/kg (mean ± standard deviation). Mean absolute percentage error was less than 30 % for each PK parameter. Mass balances for distribution and excretion were in good agreement with the literature data. CONCLUSIONS: Both PBPK model parameterizations sufficiently described the observed concentration-time data while showing an adequate predictive performance. The model should be further evaluated for its ability to explain the between-subject variability in intravenous Busulfan PK parameters.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacokinetics , Busulfan/pharmacokinetics , Intestine, Small/metabolism , Liver/metabolism , Models, Biological , Adolescent , Adult , Aged , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/chemistry , Antineoplastic Agents, Alkylating/metabolism , Artificial Intelligence , Busulfan/administration & dosage , Busulfan/chemistry , Busulfan/metabolism , Chemical Phenomena , Computer Simulation , Dose-Response Relationship, Drug , Drug Monitoring/methods , Female , Glutathione Transferase/metabolism , Humans , Infusions, Intravenous , Intestine, Small/drug effects , Intestine, Small/enzymology , Liver/drug effects , Liver/enzymology , Male , Metabolic Detoxication, Phase II , Middle Aged , Young Adult
4.
Article in English | MEDLINE | ID: mdl-23777614

ABSTRACT

An improved quantitative assay was developed and validated for fludarabine in human plasma. Fludarabine and its internal standard, cladribine, were separated on a C18 analytical column after sample purification by strong anion-exchange solid-phase extraction. Quantitation was performed by electrospray triple-quadrupole mass spectrometry in positive ionization mode using multiple-reaction monitoring. This assay had excellent inter- and intra-assay precisions within 8%, and accuracies ranging from 100 to 116%. The method was linear within the concentration range of 0.2-250ng/mL using 100µL of plasma with mean R(2)=0.9999. The extraction recoveries were 85% for fludarabine and 95% for the internal standard, which represent a significant improvement over the previously published methods. We utilized this method for pharmacokinetic (PK) investigations in 215 patients. Interference peaks constantly observed in each blank plasma sample were well resolved from fludarabine using our optimized LC-MS/MS conditions, demonstrating the reliability of this improved assay. The validated method will be further applied to PK studies within our bone marrow transplant program, which will allow for optimal dose and scheduling of fludarabine in these patients.


Subject(s)
Antineoplastic Agents/blood , Chromatography, Liquid/methods , Tandem Mass Spectrometry/methods , Vidarabine/analogs & derivatives , Antineoplastic Agents/chemistry , Cladribine , Drug Stability , Humans , Linear Models , Reproducibility of Results , Sensitivity and Specificity , Vidarabine/blood , Vidarabine/chemistry
5.
Biol Blood Marrow Transplant ; 18(2): 295-301, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21801704

ABSTRACT

Intravenous (i.v.) busulfan (Bu) administered once daily in myeloablative transplant regimens is convenient, effective, and relatively well tolerated. Therapeutic drug monitoring is recommended as nonrelapse mortality increases when daily exposure, as determined by the area under the plasma concentration versus time curve (AUC), exceeds 6000 µM·min. We describe sequential studies to achieve accurate prediction of treatment doses of Bu based on the kinetics of a smaller test dose. A total of 335 patients with hematologic malignancies were given daily i.v. Bu 3.2 mg/kg × 4 and fludarabine 50 mg/m(2) × 5. Pharmacokinetic monitoring was conducted for both the test dose and first treatment dose of Bu (day -5). Three different test dose schedules were evaluated: 12 mg Bu administered over 20 minutes, 0.8 mg/kg over 3 hours, and 0.8 mg/kg infused at 80 mg/h. The 3.2 mg/kg treatment doses were infused over a fixed time of 3 hours for the first 2 test dose trials and at a fixed rate of 80 mg/h for the final protocol. All test dose infusions were on day -7. In the first 2 schedules, Bu administered over a fixed time had significantly higher clearance for the test dose compared with the treatment dose. However, when both the test and the treatment doses were administered at the same infusion rate, clearance of the drug between the 2 dosing days was equivalent. Predicted day -5 AUC (AUC(-5)) showed a high linear correlation (r(2) = 0.74) to the actual AUC(-5). The error of these predictions was <20% in 98% of patients and <10% in 80%. In 24 individuals, the test dose predicted an AUC >5500 µM·min; therefore, the first Bu treatment dose was reduced to a desired target AUC. All adjusted doses fell within 20% of the targeted exposure. We conclude that a test dose strategy for therapeutic drug monitoring of daily i.v. Bu is accurate if the test and treatment doses are infused at the same rate. This approach allows targeting of therapeutic doses of Bu to desired levels and the potential for improved safety and efficacy.


Subject(s)
Busulfan/administration & dosage , Busulfan/pharmacokinetics , Monitoring, Physiologic , Myeloablative Agonists/administration & dosage , Myeloablative Agonists/pharmacokinetics , Transplantation Conditioning/methods , Adolescent , Adult , Aged , Busulfan/adverse effects , Female , Hematologic Neoplasms/therapy , Hematopoietic Stem Cell Transplantation/methods , Humans , Male , Middle Aged , Myeloablative Agonists/adverse effects , Time Factors , Transplantation, Homologous
6.
Biol Blood Marrow Transplant ; 14(2): 220-8, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18215782

ABSTRACT

Low plasma busulfan (Bu) area under the concentration-time curve (AUC) is associated with graft failure and relapsed leukemias, and high AUC with toxicities when Bu is used orally or i.v. 4 times daily combined with cyclophosphamide in myeloablative hematopoietic stem cell transplantation (SCT) conditioning regimens. We report Bu AUC and its association with clinical outcomes in 130 patients with hematologic malignancies given a once-daily i.v. Bu (3.2 mg/kg days -5 to -2) and fludarabine (Flu, 50 mg/m(2) days -6 to -2) regimen. Total-body irradiation (TBI) 200 cGy x 2 was added for 51 patients with acute leukemias. Plasma AUC varied 3.6-fold (2184-7794 microM.min, median 4699 microM.min). Patients with an AUC >6000 microM.min had lower overall survival (OS) than those with AUC < or =6000 microM.min at 12 months (38% versus 74%) and 36 months (23% versus 68%, P < .001). This effect was apparent in patients with standard-risk and high-risk disease, and persisted when potential confounders were considered (hazard ratio 3.2, 95% confidence interval 1.7-6.3). Nonrelapse mortality (NRM) at 100 days (6% versus 19%) and progression free survival (PFS; 58% versus 16%) at 3 years were better with AUC < or =6000 microM.min. These data support a role for therapeutic dose monitoring and dose adjustment with daily i.v. busulfan.


Subject(s)
Busulfan/administration & dosage , Busulfan/pharmacokinetics , Hematopoietic Stem Cell Transplantation/methods , Transplantation Conditioning/methods , Vidarabine/analogs & derivatives , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols , Area Under Curve , Busulfan/toxicity , Drug Monitoring/methods , Graft Survival , Hematologic Neoplasms/therapy , Hematopoietic Stem Cell Transplantation/mortality , Humans , Middle Aged , Transplantation Conditioning/mortality , Transplantation, Homologous , Treatment Outcome , Vidarabine/administration & dosage , Whole-Body Irradiation
SELECTION OF CITATIONS
SEARCH DETAIL
...