45 results on '"Huitema, Alwin D. R."'
Search Results
2. Development of a population pharmacokinetic/pharmacodynamic model for various oral paclitaxel formulations co-administered with ritonavir and thrombospondin-1 based on data from early phase clinical studies.
- Author
-
van Eijk, Maarten, Yu, Huixin, Sawicki, Emilia, de Weger, Vincent A, Nuijen, Bastiaan, Dorlo, Thomas P. C., Beijnen, Jos H, Huitema, Alwin D R, van Eijk, Maarten, Yu, Huixin, Sawicki, Emilia, de Weger, Vincent A, Nuijen, Bastiaan, Dorlo, Thomas P. C., Beijnen, Jos H, and Huitema, Alwin D R
- Abstract
PURPOSE: Orally administered paclitaxel offers increased patient convenience while providing a method to prolong exposure without long continuous, or repeated, intravenous infusions. The oral bioavailability of paclitaxel is improved through co-administration with ritonavir and application of a suitable pharmaceutical formulation, which addresses the dissolution-limited absorption of paclitaxel. We aimed to characterize the pharmacokinetics of different paclitaxel formulations, co-administered with ritonavir, and to investigate a pharmacodynamic relationship between low-dose metronomic (LDM) treatment with oral paclitaxel and the anti-angiogenic marker thrombospondin-1 (TSP-1). METHODS: Fifty-eight patients treated with different oral paclitaxel formulations were included for pharmacokinetic analysis. Pharmacodynamic data was available for 36 patients. All population pharmacokinetic/pharmacodynamic modelling was performed using non-linear mixed-effects modelling. RESULTS: A pharmacokinetic model consisting of gut, liver, central, and peripheral compartments was developed for paclitaxel. The gastrointestinal absorption rate was modelled with a Weibull function. Relative gut bioavailabilities of the tablet and capsule formulations, as fractions of the gut bioavailability of the drinking solution, were estimated to be 0.97 (95%CI: 0.67-1.33) and 0.46 (95%CI: 0.34-0.61), respectively. The pharmacokinetic/pharmacodynamic relationship between paclitaxel and TSP-1 was modelled using a turnover model with paclitaxel plasma concentrations driving an increase in TSP-1 formation rate following an Emax relationship with an EC50 of 284 ng/mL (95%CI: 122-724). CONCLUSION: The developed pharmacokinetic model adequately described the paclitaxel plasma concentrations for the different oral formulations co-administered with ritonavir. This model, and the established pharmacokinetic/pharmacodynamic relationship with TSP-1, may facilitate future development of oral paclitaxel.
- Published
- 2022
- Full Text
- View/download PDF
3. Pharmacokinetic/Pharmacodynamic Modelling of Allopurinol, its Active Metabolite Oxypurinol, and Biomarkers Hypoxanthine, Xanthine and Uric Acid in Hypoxic-Ischemic Encephalopathy Neonates
- Author
-
Chu, Wan-Yu, Annink, Kim V, Nijstad, A Laura, Maiwald, Christian A, Schroth, Michael, Bakkali, Loubna El, van Bel, Frank, Benders, Manon J N L, van Weissenbruch, Mirjam M, Hagen, Anja, Franz, Axel R, Dorlo, Thomas P C, Allegaert, Karel; https://orcid.org/0000-0001-9921-5105, Huitema, Alwin D R, ALBINO Study Group, Chu, Wan-Yu, Annink, Kim V, Nijstad, A Laura, Maiwald, Christian A, Schroth, Michael, Bakkali, Loubna El, van Bel, Frank, Benders, Manon J N L, van Weissenbruch, Mirjam M, Hagen, Anja, Franz, Axel R, Dorlo, Thomas P C, Allegaert, Karel; https://orcid.org/0000-0001-9921-5105, Huitema, Alwin D R, and ALBINO Study Group
- Abstract
BACKGROUND Allopurinol, an xanthine oxidase (XO) inhibitor, is a promising intervention that may provide neuroprotection for neonates with hypoxic-ischemic encephalopathy (HIE). Currently, a double-blind, placebo-controlled study (ALBINO, NCT03162653) is investigating the neuroprotective effect of allopurinol in HIE neonates. OBJECTIVE The aim of the current study was to establish the pharmacokinetics (PK) of allopurinol and oxypurinol, and the pharmacodynamics (PD) of both compounds on hypoxanthine, xanthine, and uric acid in HIE neonates. The dosage used and the effect of allopurinol in this population, either or not undergoing therapeutic hypothermia (TH), were evaluated. METHODS Forty-six neonates from the ALBINO study and two historical clinical studies were included. All doses were administered on the first day of life. In the ALBINO study (n = 20), neonates received a first dose of allopurinol 20 mg/kg, and, in the case of TH (n = 13), a second dose of allopurinol 10 mg/kg. In the historical cohorts (n = 26), neonates (all without TH) received two doses of allopurinol 20 mg/kg in total. Allopurinol and oxypurinol population PK, and their effects on inhibiting conversions of hypoxanthine and xanthine to uric acid, were assessed using nonlinear mixed-effects modelling. RESULTS Allopurinol and oxypurinol PK were described by two sequential one-compartment models with an autoinhibition effect on allopurinol metabolism by oxypurinol. For allopurinol, clearance (CL) was 0.83 L/h (95% confidence interval [CI] 0.62-1.09) and volume of distribution (V$_{d}$) was 2.43 L (95% CI 2.25-2.63). For metabolite oxypurinol, CL and V$_{d}$ relative to a formation fraction (f$_{m}$) were 0.26 L/h (95% CI 0.23-0.3) and 11 L (95% CI 9.9-12.2), respectively. No difference in allopurinol and oxypurinol CL was found between TH and non-TH patients. The effect of allopurinol and oxypurinol on XO inhibition was described by a turnover model of hypoxanthine with sequential metabolites xa
- Published
- 2022
4. Semi-mechanistic Modeling of Hypoxanthine, Xanthine, and Uric Acid Metabolism in Asphyxiated Neonates
- Author
-
Chu, Wan-Yu, Allegaert, Karel, Dorlo, Thomas P C, Huitema, Alwin D R, ALBINO Study Group, Chu, Wan-Yu, Allegaert, Karel, Dorlo, Thomas P C, Huitema, Alwin D R, and ALBINO Study Group
- Abstract
BACKGROUND AND OBJECTIVE Previously, we developed a pharmacokinetic-pharmacodynamic model of allopurinol, oxypurinol, and biomarkers, hypoxanthine, xanthine, and uric acid, in neonates with hypoxic-ischemic encephalopathy, in which high initial biomarker levels were observed suggesting an impact of hypoxia. However, the full pharmacodynamics could not be elucidated in our previous study. The current study included additional data from the ALBINO study (NCT03162653) placebo group, aiming to characterize the dynamics of hypoxanthine, xanthine, and uric acid in neonates with hypoxic-ischemic encephalopathy. METHODS Neonates from the ALBINO study who received allopurinol or placebo mannitol were included. An extended population pharmacokinetic-pharmacodynamic model was developed based on the mechanism of purine metabolism, where synthesis, salvage, and degradation via xanthine oxidoreductase pathways were described. The initial level of the biomarkers was a combination of endogenous turnover and high disease-related amounts. Model development was accomplished by nonlinear mixed-effects modeling (NONMEM®, version 7.5). RESULTS In total, 20 neonates treated with allopurinol and 17 neonates treated with mannitol were included in this analysis. Endogenous synthesis of the biomarkers reduced with 0.43% per hour because of precursor exhaustion. Hypoxanthine was readily salvaged or degraded to xanthine with rate constants of 0.5 1/h (95% confidence interval 0.33-0.77) and 0.2 1/h (95% confidence interval 0.09-0.31), respectively. A greater salvage was found in the allopurinol treatment group consistent with its mechanism of action. High hypoxia-induced initial levels of biomarkers were quantified, and were 1.2-fold to 2.9-fold higher in neonates with moderate-to-severe hypoxic-ischemic encephalopathy compared with those with mild hypoxic-ischemic encephalopathy. Half-maximal xanthine oxidoreductase inhibition was achieved with a combined allopurinol and oxypurinol c
- Published
- 2022
5. Population Pharmacokinetics of Intracellular 5-Fluorouridine 5'-Triphosphate and its Relationship with Hand-and-Foot Syndrome in Patients Treated with Capecitabine
- Author
-
Apotheek Opleiding, Apotheek O&O&O, Cancer, Janssen, Julie M, Jacobs, Bart A W, Roosendaal, Jeroen, Derissen, Ellen J B, Marchetti, Serena, Beijnen, Jos H, Huitema, Alwin D R, Dorlo, Thomas P C, Apotheek Opleiding, Apotheek O&O&O, Cancer, Janssen, Julie M, Jacobs, Bart A W, Roosendaal, Jeroen, Derissen, Ellen J B, Marchetti, Serena, Beijnen, Jos H, Huitema, Alwin D R, and Dorlo, Thomas P C
- Published
- 2021
6. Determination of the absolute bioavailability of oral imatinib using a stable isotopically labeled intravenous imatinib-d8 microdose
- Author
-
Afd Pharmacoepi & Clinical Pharmacology, Pharmacoepidemiology and Clinical Pharmacology, Roosendaal, Jeroen, Groenland, Stefanie L, Rosing, Hilde, Lucas, Luc, Venekamp, Nikkie, Nuijen, Bastiaan, Huitema, Alwin D R, Beijnen, Jos H, Steeghs, Neeltje, Afd Pharmacoepi & Clinical Pharmacology, Pharmacoepidemiology and Clinical Pharmacology, Roosendaal, Jeroen, Groenland, Stefanie L, Rosing, Hilde, Lucas, Luc, Venekamp, Nikkie, Nuijen, Bastiaan, Huitema, Alwin D R, Beijnen, Jos H, and Steeghs, Neeltje
- Published
- 2020
7. Quantification of T Cell Binding Polyclonal Rabbit Anti-thymocyte Globulin in Human Plasma with Liquid Chromatography Tandem-Mass Spectrometry
- Author
-
Apotheek KF TDM, Arts-assistenten Kinderen, CTI, Apotheek Onderzoek, Cancer, CTI Research, CDL Patiëntenzorg MI, Infection & Immunity, Apotheek Klinische Farmacie, Amrani, Mohsin El, Admiraal, Rick, Willaert, Lobke, Ebskamp-van Raaij, Lysette J C, Lacna, Amelia M, Hack, C Erik, Huitema, Alwin D R, Nierkens, Stefan, van Maarseveen, Erik M, Apotheek KF TDM, Arts-assistenten Kinderen, CTI, Apotheek Onderzoek, Cancer, CTI Research, CDL Patiëntenzorg MI, Infection & Immunity, Apotheek Klinische Farmacie, Amrani, Mohsin El, Admiraal, Rick, Willaert, Lobke, Ebskamp-van Raaij, Lysette J C, Lacna, Amelia M, Hack, C Erik, Huitema, Alwin D R, Nierkens, Stefan, and van Maarseveen, Erik M
- Published
- 2020
8. Phase I pharmacological study of continuous chronomodulated capecitabine treatment
- Author
-
Afd Pharmacoepi & Clinical Pharmacology, Pharmacoepidemiology and Clinical Pharmacology, Roosendaal, Jeroen, Jacobs, Bart A W, Pluim, Dick, Rosing, Hilde, de Vries, Niels, van Werkhoven, Erik, Nuijen, Bastiaan, Beijnen, Jos H, Huitema, Alwin D R, Schellens, Jan H M, Marchetti, Serena, Afd Pharmacoepi & Clinical Pharmacology, Pharmacoepidemiology and Clinical Pharmacology, Roosendaal, Jeroen, Jacobs, Bart A W, Pluim, Dick, Rosing, Hilde, de Vries, Niels, van Werkhoven, Erik, Nuijen, Bastiaan, Beijnen, Jos H, Huitema, Alwin D R, Schellens, Jan H M, and Marchetti, Serena
- Published
- 2020
9. Individualized dosing of oral targeted therapies in oncology is crucial in the era of precision medicine
- Author
-
Afd Pharmacoepi & Clinical Pharmacology, Pharmacoepidemiology and Clinical Pharmacology, Groenland, Stefanie L, Mathijssen, Ron H J, Beijnen, Jos H, Huitema, Alwin D R, Steeghs, Neeltje, Afd Pharmacoepi & Clinical Pharmacology, Pharmacoepidemiology and Clinical Pharmacology, Groenland, Stefanie L, Mathijssen, Ron H J, Beijnen, Jos H, Huitema, Alwin D R, and Steeghs, Neeltje
- Published
- 2019
10. Does Older Age Lead to Higher Risk for Neutropenia in Patients Treated with Paclitaxel?
- Author
-
Afd Pharmacoepi & Clinical Pharmacology, Pharmacoepidemiology and Clinical Pharmacology, Crombag, Marie-Rose B S, Koolen, Stijn L W, Wijngaard, Sophie, Joerger, Markus, Dorlo, Thomas P C, van Erp, Nielka P, Mathijssen, Ron H J, Beijnen, Jos H, Huitema, Alwin D R, Afd Pharmacoepi & Clinical Pharmacology, Pharmacoepidemiology and Clinical Pharmacology, Crombag, Marie-Rose B S, Koolen, Stijn L W, Wijngaard, Sophie, Joerger, Markus, Dorlo, Thomas P C, van Erp, Nielka P, Mathijssen, Ron H J, Beijnen, Jos H, and Huitema, Alwin D R
- Published
- 2019
11. Impact of Older Age on the Exposure of Paclitaxel: a Population Pharmacokinetic Study
- Author
-
Afd Pharmacoepi & Clinical Pharmacology, Pharmacoepidemiology and Clinical Pharmacology, Crombag, Marie-Rose B S, de Vries Schultink, Aurelia H M, Koolen, Stijn L W, Wijngaard, Sophie, Joerger, Markus, Schellens, Jan H M, Dorlo, Thomas P C, van Erp, Nielka P, Mathijssen, Ron H J, Beijnen, Jos H, Huitema, Alwin D R, Afd Pharmacoepi & Clinical Pharmacology, Pharmacoepidemiology and Clinical Pharmacology, Crombag, Marie-Rose B S, de Vries Schultink, Aurelia H M, Koolen, Stijn L W, Wijngaard, Sophie, Joerger, Markus, Schellens, Jan H M, Dorlo, Thomas P C, van Erp, Nielka P, Mathijssen, Ron H J, Beijnen, Jos H, and Huitema, Alwin D R
- Published
- 2019
12. Cytochrome P450 3A4, 3A5, and 2C8 expression in breast, prostate, lung, endometrial, and ovarian tumors: relevance for resistance to taxanes
- Author
-
Afd Pharmacoepi & Clinical Pharmacology, Pharmacoepidemiology and Clinical Pharmacology, van Eijk, Maarten, Boosman, René J, Schinkel, Alfred H, Huitema, Alwin D R, Beijnen, Jos H, Afd Pharmacoepi & Clinical Pharmacology, Pharmacoepidemiology and Clinical Pharmacology, van Eijk, Maarten, Boosman, René J, Schinkel, Alfred H, Huitema, Alwin D R, and Beijnen, Jos H
- Published
- 2019
13. An Antiestrogenic Activity Score for tamoxifen and its metabolites is associated with breast cancer outcome
- Author
-
Afd Pharmacoepi & Clinical Pharmacology, Pharmacoepidemiology and Clinical Pharmacology, de Vries Schultink, Aurelia H M, Alexi, X, van Werkhoven, E., Madlensky, L, Natarajan, L, Flatt, S W, Zwart, Wilbert, Linn, Sabine C., Parker, B A, Wu, A H B, Pierce, J P, Huitema, Alwin D R, Beijnen, J H, Afd Pharmacoepi & Clinical Pharmacology, Pharmacoepidemiology and Clinical Pharmacology, de Vries Schultink, Aurelia H M, Alexi, X, van Werkhoven, E., Madlensky, L, Natarajan, L, Flatt, S W, Zwart, Wilbert, Linn, Sabine C., Parker, B A, Wu, A H B, Pierce, J P, Huitema, Alwin D R, and Beijnen, J H
- Published
- 2017
14. Exposure-survival analyses of pazopanib in renal cell carcinoma and soft tissue sarcoma patients: opportunities for dose optimization
- Author
-
Afd Pharmacoepi & Clinical Pharmacology, Pharmacoepidemiology and Clinical Pharmacology, Verheijen, Remy B., Swart, L E, Beijnen, J H, Schellens, J H M, Huitema, Alwin D R, Steeghs, N., Afd Pharmacoepi & Clinical Pharmacology, Pharmacoepidemiology and Clinical Pharmacology, Verheijen, Remy B., Swart, L E, Beijnen, J H, Schellens, J H M, Huitema, Alwin D R, and Steeghs, N.
- Published
- 2017
15. Clinical pharmacokinetics of an amorphous solid dispersion tablet of elacridar
- Author
-
Pharmacoepidemiology and Clinical Pharmacology, Afd Pharmacoepi & Clinical Pharmacology, Sawicki, Emilia, Verheijen, Remy B., Huitema, Alwin D R, van Tellingen, Olaf, Schellens, Jan H M, Nuijen, Bastiaan, Beijnen, Jos H., Steeghs, Neeltje, Pharmacoepidemiology and Clinical Pharmacology, Afd Pharmacoepi & Clinical Pharmacology, Sawicki, Emilia, Verheijen, Remy B., Huitema, Alwin D R, van Tellingen, Olaf, Schellens, Jan H M, Nuijen, Bastiaan, Beijnen, Jos H., and Steeghs, Neeltje
- Published
- 2017
16. Development of a Tumour Growth Inhibition Model to Elucidate the Effects of Ritonavir on Intratumoural Metabolism and Anti-tumour Effect of Docetaxel in a Mouse Model for Hereditary Breast Cancer
- Author
-
Pharmacoepidemiology and Clinical Pharmacology, Sub Clinical Pharmacology, Yu, Huixin, Hendrikx, Jeroen J M A, Rottenberg, Sven, Schellens, Jan H M, Beijnen, Jos H., Huitema, Alwin D R, Pharmacoepidemiology and Clinical Pharmacology, Sub Clinical Pharmacology, Yu, Huixin, Hendrikx, Jeroen J M A, Rottenberg, Sven, Schellens, Jan H M, Beijnen, Jos H., and Huitema, Alwin D R
- Published
- 2016
17. A phase 0 clinical trial of novel candidate extended-release formulations of capecitabine
- Author
-
Sub Clinical Pharmacology, Pharmacoepidemiology and Clinical Pharmacology, Jacobs, Bart A W, Meulenaar, Jelte, Rosing, Hilde, Pluim, Dick, Tibben, Matthijs M., de Vries, Niels, Nuijen, Bastiaan, Huitema, Alwin D R, Beijnen, Jos H., Schellens, Jan H M, Marchetti, Serena, Sub Clinical Pharmacology, Pharmacoepidemiology and Clinical Pharmacology, Jacobs, Bart A W, Meulenaar, Jelte, Rosing, Hilde, Pluim, Dick, Tibben, Matthijs M., de Vries, Niels, Nuijen, Bastiaan, Huitema, Alwin D R, Beijnen, Jos H., Schellens, Jan H M, and Marchetti, Serena
- Published
- 2016
18. Baclofen overdose treated with continuous venovenous hemofiltration
- Author
-
Pharmacoepidemiology and Clinical Pharmacology, Sub Clinical Pharmacology, Meulendijks, Didier, Khan, Saheed, Koks, Cornelis H W, Huitema, Alwin D R, Schellens, Jan H M|info:eu-repo/dai/nl/073926272, Beijnen, Jos H.|info:eu-repo/dai/nl/071919570, Pharmacoepidemiology and Clinical Pharmacology, Sub Clinical Pharmacology, Meulendijks, Didier, Khan, Saheed, Koks, Cornelis H W, Huitema, Alwin D R, Schellens, Jan H M|info:eu-repo/dai/nl/073926272, and Beijnen, Jos H.|info:eu-repo/dai/nl/071919570
- Published
- 2015
19. Therapeutic drug monitoring of oral targeted antineoplastic drugs.
- Author
-
Mueller-Schoell A, Groenland SL, Scherf-Clavel O, van Dyk M, Huisinga W, Michelet R, Jaehde U, Steeghs N, Huitema ADR, and Kloft C
- Subjects
- Administration, Oral, Antineoplastic Agents pharmacokinetics, Humans, Antineoplastic Agents therapeutic use, Drug Monitoring
- Abstract
Purpose: This review provides an overview of the current challenges in oral targeted antineoplastic drug (OAD) dosing and outlines the unexploited value of therapeutic drug monitoring (TDM). Factors influencing the pharmacokinetic exposure in OAD therapy are depicted together with an overview of different TDM approaches. Finally, current evidence for TDM for all approved OADs is reviewed., Methods: A comprehensive literature search (covering literature published until April 2020), including primary and secondary scientific literature on pharmacokinetics and dose individualisation strategies for OADs, together with US FDA Clinical Pharmacology and Biopharmaceutics Reviews and the Committee for Medicinal Products for Human Use European Public Assessment Reports was conducted., Results: OADs are highly potent drugs, which have substantially changed treatment options for cancer patients. Nevertheless, high pharmacokinetic variability and low treatment adherence are risk factors for treatment failure. TDM is a powerful tool to individualise drug dosing, ensure drug concentrations within the therapeutic window and increase treatment success rates. After reviewing the literature for 71 approved OADs, we show that exposure-response and/or exposure-toxicity relationships have been established for the majority. Moreover, TDM has been proven to be feasible for individualised dosing of abiraterone, everolimus, imatinib, pazopanib, sunitinib and tamoxifen in prospective studies. There is a lack of experience in how to best implement TDM as part of clinical routine in OAD cancer therapy., Conclusion: Sub-therapeutic concentrations and severe adverse events are current challenges in OAD treatment, which can both be addressed by the application of TDM-guided dosing, ensuring concentrations within the therapeutic window.
- Published
- 2021
- Full Text
- View/download PDF
20. Correction to: Therapeutic drug monitoring of oral targeted antineoplastic drugs.
- Author
-
Mueller-Schoell A, Groenland SL, Scherf-Clavel O, van Dyk M, Huisinga W, Michelet R, Jaehde U, Steeghs N, Huitema ADR, and Kloft C
- Published
- 2021
- Full Text
- View/download PDF
21. Quantification of the pharmacokinetic-toxicodynamic relationship of oral docetaxel co-administered with ritonavir.
- Author
-
Yu H, Janssen JM, de Weger VA, Nuijen B, Stuurman RE, Marchetti S, Schellens JHM, Beijnen JH, Dorlo TPC, and Huitema ADR
- Subjects
- Administration, Oral, Computer Simulation, Humans, Antineoplastic Agents administration & dosage, Antineoplastic Agents adverse effects, Antineoplastic Agents pharmacokinetics, Antineoplastic Agents toxicity, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols pharmacokinetics, Antineoplastic Combined Chemotherapy Protocols toxicity, Docetaxel administration & dosage, Docetaxel adverse effects, Docetaxel pharmacokinetics, Docetaxel toxicity, Models, Biological, Ritonavir administration & dosage, Ritonavir adverse effects, Ritonavir pharmacokinetics, Ritonavir toxicity
- Abstract
Introduction Oral formulations of docetaxel have successfully been developed as an alternative for intravenous administration. Co-administration with the enzyme inhibitor ritonavir boosts the docetaxel plasma exposure. In dose-escalation trials, the maximum tolerated doses for two different dosing regimens were established and dose-limiting toxicities (DLTs) were recorded. The aim of current analysis was to develop a pharmacokinetic (PK)-toxicodynamic (TOX) model to quantify the relationship between docetaxel plasma exposure and DLTs. Methods A total of 85 patients was included in the current analysis, 18 patients showed a DLT in the four-week observation period. A PK-TOX model was developed and simulations based on the PK-TOX model were performed. Results The final PK-TOX model was characterized by an effect compartment representing the toxic effect of docetaxel, which was linked to the probability of developing a DLT. Simulations of once-weekly, once-daily 60 mg and once-weekly, twice-daily 30 mg followed by 20 mg of oral docetaxel suggested that 14% and 34% of patients, respectively, would have a probability >25% to develop a DLT in a four-week period. Conclusions A PK-TOX model was successfully developed. This model can be used to evaluate the probability of developing a DLT following treatment with oral docetaxel and ritonavir in different dosing regimens.
- Published
- 2020
- Full Text
- View/download PDF
22. Evaluation of dose-tapering strategies for intravenous tocilizumab in rheumatoid arthritis patients using model-based pharmacokinetic/pharmacodynamic simulations.
- Author
-
Bastida C, Huitema ADR, l'Ami MJ, Ruiz-Esquide V, Wolbink GJ, Sanmartí R, and Soy D
- Subjects
- Administration, Intravenous, Algorithms, Antibodies, Monoclonal, Humanized pharmacokinetics, Antirheumatic Agents pharmacokinetics, Computer Simulation, Drug Tapering, Humans, Remission Induction, Antibodies, Monoclonal, Humanized administration & dosage, Antirheumatic Agents administration & dosage, Arthritis, Rheumatoid drug therapy, Models, Biological
- Abstract
Purpose: Tocilizumab is a humanized monoclonal antibody approved for rheumatoid arthritis treatment. In clinical practice, empirical dose-tapering strategies are implemented in patients showing sustained remission or low disease activity (LDA) to avoid overtreatment and reduce costs. Since rational adaptive-dosing algorithms taking the full pharmacokinetic (PK)/pharmacodynamic (PD) characteristics into account are currently lacking, we aimed to develop novel tapering strategies and compare them with currently used empirical ones., Methods: Four strategies were simulated on a virtual population. In all of them, the same initial dose was administered every 28 days for six consecutive months. Then, different strategies were considered: (1) label-dosing; (2) mild empirical dose-tapering; (3) intense empirical dose-tapering; (4) therapeutic drug monitoring (TDM)-guided dose-tapering. The different strategies were evaluated on the proportion of patients who maintain remission/LDA 1 year after the intervention. Cost-savings of direct drug costs were also estimated as relative dose intensity., Results: The overall proportion of simulated patients in remission/LDA after 1 year of the intervention was comparable between the mild empirical and the TDM-guided dose-tapering strategies, and much lower for the intense empirical dose-tapering strategy (80.3%, 78.2%, and 69.0%, respectively). Likewise, 1-year flare rates were lower for the mild empirical and TDM-guided tapering strategies. The relative dose intensity was lowest for the intense empirical dose-tapering, followed by the TDM-guided and the mild empirical dose-tapering approaches (61.2%, 71.0%, and 80.4%, respectively)., Conclusion: We demonstrated that the TDM-guided strategy using model-based algorithms performed similarly to mild empirical dose-tapering strategies in overall remission/LDA rates but is superior in cost-savings.
- Published
- 2020
- Full Text
- View/download PDF
23. Determination of the absolute bioavailability of oral imatinib using a stable isotopically labeled intravenous imatinib-d8 microdose.
- Author
-
Roosendaal J, Groenland SL, Rosing H, Lucas L, Venekamp N, Nuijen B, Huitema ADR, Beijnen JH, and Steeghs N
- Subjects
- Administration, Intravenous, Administration, Oral, Aged, Antineoplastic Agents blood, Antineoplastic Agents pharmacokinetics, Biological Availability, Chromatography, Liquid, Deuterium, Female, Gastrointestinal Stromal Tumors blood, Gastrointestinal Stromal Tumors drug therapy, Gastrointestinal Stromal Tumors metabolism, Humans, Imatinib Mesylate blood, Imatinib Mesylate pharmacokinetics, Isotope Labeling, Male, Middle Aged, Protein Kinase Inhibitors blood, Protein Kinase Inhibitors pharmacokinetics, Tandem Mass Spectrometry, Antineoplastic Agents administration & dosage, Imatinib Mesylate administration & dosage, Protein Kinase Inhibitors administration & dosage
- Abstract
Purpose: The aim of this study was to ascertain whether the absolute bioavailability of oral imatinib (Glivec®) during steady state plasma pharmacokinetics in cancer patients could be determined through a concomitant intravenous administration of a single 100 μg microdose of deuterium labeled imatinib (imatinib-d8). Secondly, the usefulness of liquid chromatography-tandem mass spectrometry (LC-MS/MS) was investigated for simultaneous analysis of orally and intravenously administered imatinib., Methods: Included patients were on a stable daily dose of 400 mg oral imatinib prior to study participation. On day 1, patients received a 100 μg intravenous imatinib-d8 microdose 2.5 h after intake of the oral dose. Plasma samples were collected for 48 h. Imatinib and imatinib-d8 concentrations were simultaneously quantified using a validated LC-MS/MS assay. The absolute bioavailability was calculated by comparing the dose-normalized exposure with unlabeled and stable isotopically labeled imatinib in plasma., Results: A total of six patients were enrolled. All patients had a history of gastrointestinal stromal tumors (GIST). The median absolute bioavailability of oral imatinib at steady state was 76% (range 44-106%). Imatinib and imatinib-d8 plasma concentrations were quantified in all collected plasma samples, with no samples below the limit of quantification for imatinib-d8., Conclusion: The absolute bioavailability of imatinib was successfully estimated at steady state plasma pharmacokinetics using the stable isotopically labeled microdose trial design. This study exhibits the use of a stable isotopically labeled intravenous microdose to determine the absolute bioavailability of an oral anticancer agent in patients with LC-MS/MS as the analytical tool.
- Published
- 2020
- Full Text
- View/download PDF
24. Mass balance and metabolite profiling of 14 C-guadecitabine in patients with advanced cancer.
- Author
-
Roosendaal J, Rosing H, Lucas L, Gebretensae A, Huitema ADR, van Dongen MG, Beijnen JH, and Oganesian A
- Subjects
- Aged, Antineoplastic Agents blood, Antineoplastic Agents urine, Azacitidine blood, Azacitidine pharmacokinetics, Azacitidine urine, Carbon Radioisotopes, Female, Humans, Male, Middle Aged, Neoplasms blood, Neoplasms urine, Antineoplastic Agents pharmacokinetics, Azacitidine analogs & derivatives, Neoplasms metabolism
- Abstract
Purpose The objective of this mass balance trial was to determine the excretory pathways and metabolic profile of the novel anticancer agent guadecitabine in humans after administration of a
14 C-radiolabeled dose of guadecitabine. Experimental design Included patients received at least one cycle of 45 mg/m2 guadecitabine subcutaneously as once-daily doses on Days 1 to 5 of a 28-day cycle, of which the 5th (last) dose in the first cycle was spiked with14 C-radiolabeled guadecitabine. Using different mass spectrometric techniques in combination with off-line liquid scintillation counting, the exposure and excretion of14 C-guadecitabine and metabolites in the systemic circulation, excreta, and intracellular target site were established. Results Five patients were enrolled in the mass balance trial.14 C-guadecitabine radioactivity was rapidly and almost exclusively excreted in urine, with an average amount of radioactivity recovered of 90.2%. After uptake in the systemic circulation, guadecitabine was converted into ß-decitabine (active anomer), and from ß-decitabine into the presumably inactive metabolites M1-M5. All identified metabolites in plasma and urine were ß-decitabine related products, suggesting almost complete conversion via cleavage of the phosphodiester bond between ß-decitabine and deoxyguanosine prior to further elimination. ß-decitabine enters the intracellular activation pathway, leading to detectable ß-decitabine-triphosphate and DNA incorporated ß-decitabine levels in peripheral blood mononuclear cells, providing confirmation that the drug reaches its DNA target site. Conclusion The metabolic and excretory pathways of guadecitabine and its metabolites were successfully characterized after subcutaneous guadecitabine administration in cancer patients. These data support the clinical evaluation of safety and efficacy of the subcutaneous guadecitabine drug product.- Published
- 2020
- Full Text
- View/download PDF
25. Individualized dosing of oral targeted therapies in oncology is crucial in the era of precision medicine.
- Author
-
Groenland SL, Mathijssen RHJ, Beijnen JH, Huitema ADR, and Steeghs N
- Subjects
- Administration, Oral, Humans, Antineoplastic Agents administration & dosage, Drug Monitoring, Molecular Targeted Therapy, Neoplasms drug therapy, Precision Medicine
- Abstract
Purpose: While in the era of precision medicine, the right drug for each patient is selected based on molecular tumor characteristics, most novel oral targeted anticancer agents are still being administered using a one-size-fits-all fixed dosing approach. In this review, we discuss the scientific evidence for dose individualization of oral targeted therapies in oncology, based on therapeutic drug monitoring (TDM)., Methods: Based on literature search and our own experiences, seven criteria for drugs to be suitable candidates for TDM will be addressed: (1) absence of an easily measurable biomarker for drug effect; (2) long-term therapy; (3) availability of a validated sensitive bioanalytical method; (4) significant variability in pharmacokinetic exposure; (5) narrow therapeutic range; (6) defined and consistent exposure-response relationships; (7) feasible dose-adaptation strategies., Results: All of these requirements are met for most oral targeted therapies in oncology. Also, prospective studies have already shown TDM to be feasible for imatinib, pazopanib, sunitinib, everolimus, and endoxifen., Conclusions: In order to realize the full potential of personalized medicine in oncology, patients should not only be treated with the right drug, but also at the right dose. TDM could be a suitable tool to achieve this.
- Published
- 2019
- Full Text
- View/download PDF
26. Translational PK-PD modeling analysis of MCLA-128, a HER2/HER3 bispecific monoclonal antibody, to predict clinical efficacious exposure and dose.
- Author
-
de Vries Schultink AHM, Doornbos RP, Bakker ABH, Bol K, Throsby M, Geuijen C, Maussang D, Schellens JHM, Beijnen JH, and Huitema ADR
- Subjects
- Animals, Area Under Curve, Cell Line, Tumor, Cell Proliferation drug effects, Computer Simulation, Dose-Response Relationship, Immunologic, Female, Humans, Macaca fascicularis, Mice, Mice, SCID, Treatment Outcome, Tumor Burden, Xenograft Model Antitumor Assays, Antibodies, Bispecific pharmacokinetics, Antibodies, Bispecific pharmacology, Antibodies, Monoclonal pharmacokinetics, Antibodies, Monoclonal pharmacology, Immunoglobulin G pharmacology, Models, Biological, Translational Research, Biomedical
- Abstract
Introduction MCLA-128 is a bispecific monoclonal antibody targeting the HER2 and HER3 receptors. Pharmacokinetics (PK) and pharmacodynamics (PD) of MCLA-128 have been evaluated in preclinical studies in cynomolgus monkeys and mice. The aim of this study was to characterize the PK and PD of MCLA-128 and to predict a safe starting dose and efficacious clinical dose for the First-In-Human study. Methods A PK-PD model was developed based on PK data from cynomolgus monkeys and tumor growth data from a mouse JIMT-1 xenograft model. Allometric scaling was used to scale PK parameters between species. Simulations were performed to predict the safe and efficacious clinical dose, based on AUCs, receptor occupancies and PK-PD model simulations. Results MCLA-128 PK in cynomolgus monkeys was described by a two-compartment model with parallel linear and nonlinear clearance. The xenograft tumor growth model consisted of a tumor compartment with a zero-order growth rate and a first-order dying rate, both affected by MCLA-128. Human doses of 10 to 480 mg q3wk were predicted to show a safety margin of >10-fold compared to the cynomolgus monkey AUC at the no-observed-adverse-effect-level (NOAEL). Doses of ≥360 mg resulted in predicted receptor occupancies above 99% (C
max and Cave) . These doses showed anti-tumor efficacy in the PK-PD model. Conclusions This analysis predicts that a flat dose of 10 to 480 mg q3wk is suitable as starting dose for a First-in-Human study with MCLA-128. Flat doses ≥360 mg q3wk are expected to be efficacious in human, based on receptor occupancies and PK-PD model simulations.- Published
- 2018
- Full Text
- View/download PDF
27. Pharmacodynamic modeling of cardiac biomarkers in breast cancer patients treated with anthracycline and trastuzumab regimens.
- Author
-
de Vries Schultink AHM, Boekhout AH, Gietema JA, Burylo AM, Dorlo TPC, van Hasselt JGC, Schellens JHM, and Huitema ADR
- Subjects
- Adult, Aged, Cardiotoxicity metabolism, Female, Humans, Middle Aged, Muscle Cells drug effects, Muscle Cells metabolism, Natriuretic Peptide, Brain metabolism, Peptide Fragments metabolism, Troponin T metabolism, Anthracyclines therapeutic use, Antineoplastic Agents, Immunological therapeutic use, Biomarkers, Tumor metabolism, Breast Neoplasms drug therapy, Breast Neoplasms metabolism, Heart Ventricles metabolism, Trastuzumab therapeutic use
- Abstract
Trastuzumab is associated with cardiotoxicity, manifesting as a decrease of the left-ventricular ejection fraction (LVEF). Administration of anthracyclines prior to trastuzumab increases risk of cardiotoxicity. High-sensitive troponin T and N-terminal-pro-brain natriuretic peptide (NT-proBNP) are molecular markers that may allow earlier detection of drug-induced cardiotoxicity. In this analysis we aimed to quantify the kinetics and exposure-response relationships of LVEF, troponin T and NT-proBNP measurements, in patients receiving anthracycline and trastuzumab. Repeated measurements of LVEF, troponin T and NT-proBNP and dosing records of anthracyclines and trastuzumab were available from a previously published clinical trial. This trial included 206 evaluable patients with early breast cancer. Exposure to anthracycline and trastuzumab was simulated based on available dosing records and by using a kinetic-pharmacodynamic (K-PD) and a fixed pharmacokinetic (PK) model from literature, respectively. The change from baseline troponin T was described with a direct effect model, affected by simulated anthracycline concentrations, representing myocyte damage. The relationship between trastuzumab and LVEF was described by an indirect effect compartment model. The EC
50 for LVEF decline was significantly affected by the maximum troponin T concentration after anthracycline treatment, explaining 15.1% of inter-individual variability. In this cohort, NT-proBNP changes could not be demonstrated to be related to anthracycline or trastuzumab treatment. Pharmacodynamic models for troponin T and LVEF were successfully developed, identifying maximum troponin T concentration after anthracycline treatment as a significant determinant for trastuzumab-induced LVEF decline. These models can help identify patients at risk of drug-induced cardiotoxicity and optimize cardiac monitoring strategies.- Published
- 2018
- Full Text
- View/download PDF
28. Standard Clinical Protocol for Bidirectional Hyperthermic Intraperitoneal Chemotherapy (HIPEC): Systemic Leucovorin, 5-Fluorouracil, and Heated Intraperitoneal Oxaliplatin in a Chloride-Containing Carrier Solution.
- Author
-
Mehta AM, Huitema AD, Burger JW, Brandt-Kerkhof AR, van den Heuvel SF, and Verwaal VJ
- Subjects
- Administration, Intravenous, Blood Glucose metabolism, Chlorides administration & dosage, Chlorides blood, Cytoreduction Surgical Procedures, Dialysis Solutions administration & dosage, Dialysis Solutions chemistry, Evidence-Based Medicine, Female, Fluorouracil administration & dosage, Humans, Infusions, Parenteral, Leucovorin administration & dosage, Male, Middle Aged, Organoplatinum Compounds administration & dosage, Oxaliplatin, Peritoneal Neoplasms secondary, Potassium blood, Sodium blood, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Colorectal Neoplasms pathology, Hyperthermia, Induced, Peritoneal Neoplasms therapy
- Abstract
Background: Intraperitoneal chemotherapy has an established role in the treatment of selected patients with colorectal peritoneal metastases. Oxaliplatin is highly suitable as a chemotherapeutic agent for hyperthermic intraperitoneal chemotherapy (HIPEC), but its use to date has been limited because of the morbidity caused by severe electrolyte and glycemic imbalances associated with 5% glucose as its carrier solution. This report provides an overview of the development, rationale, and application of intraperitoneal chemotherapy and the use of various drugs and carrier solutions. A novel, evidence-based protocol for bidirectional oxaliplatin-based HIPEC in a physiologic carrier solution (Dianeal PD4 dextrose 1.36%) is presented, and its impact on electrolyte and glucose levels is demonstrated., Methods: After implementation of the new protocol, the serum electrolyte (sodium, potassium, and chloride) levels, glucose levels, and intravenous insulin requirements were intensively measured in eight consecutive cases immediately before HIPEC (T = 0), immediately after HIPEC (T = 30), 1 h after HIPEC (T = 60), and 3 h after HIPEC (T = 180)., Results: The median sodium levels were 140 mmol/L at T = 0, 138 mmol/L at T = 30, 140 mmol/L at T = 60, and 140 mmol/L at T = 180. The respective median potassium levels were 4.6, 4.2, 3.7, and 3.9 mmol/L, and the respective median chloride levels were 112, 111, 111, and 112 mmol/L. The respective median glucose levels were 9, 11.5, 10.7, and 8.6 mmol/L. The median insulin requirements were respectively 0.5, 1.5, 1.2, and 0 U/h. None of the patients were diabetic., Conclusion: Using a novel protocol for bidirectional oxaliplatin-based HIPEC in Dianeal instead of 5% glucose, the observed fluctuations in this study were minimal and not clinically relevant compared with historical values for electrolyte and glycemic changes using 5% glucose as a HIPEC carrier solution. This novel protocol leads to only minimal and clinically irrelevant electrolyte and glycemic disturbances, and its adoption as the standard protocol for oxaliplatin-based HIPEC should be considered.
- Published
- 2017
- Full Text
- View/download PDF
29. Clinical pharmacokinetics of an amorphous solid dispersion tablet of elacridar.
- Author
-
Sawicki E, Verheijen RB, Huitema AD, van Tellingen O, Schellens JH, Nuijen B, Beijnen JH, and Steeghs N
- Subjects
- Acridines adverse effects, Acridines chemistry, Adult, Biological Availability, Drug Liberation, Female, Humans, Male, Middle Aged, Powders, Tablets, Tetrahydroisoquinolines adverse effects, Tetrahydroisoquinolines chemistry, Acridines administration & dosage, Acridines pharmacokinetics, Tetrahydroisoquinolines administration & dosage, Tetrahydroisoquinolines pharmacokinetics
- Abstract
Elacridar is an inhibitor of the permeability glycoprotein (P-gp) and the breast cancer resistance protein (BCRP) and is a promising absorption enhancer of drugs that are substrates of these drug-efflux transporters. However, elacridar is practically insoluble in water, resulting in low bioavailability which currently limits its clinical application. We evaluated the in vitro dissolution and clinical pharmacokinetics of a novel amorphous solid dispersion (ASD) tablet containing elacridar. The dissolution from ASD tablets was compared to that from a crystalline powder mixture in a USP type II dissolution apparatus. The pharmacokinetics of the ASD tablet were evaluated in an exploratory clinical study at oral doses of 25, 250, or 1000 mg in 12 healthy volunteers. A target C
max was set at ≥ 200 ng/mL based on previous clinical data. The in vitro dissolution from the ASD tablet was 16.9 ± 3.7 times higher compared to that from a crystalline powder mixture. Cmax and AUC0-∞ increased linearly with dose over the explored range. The target Cmax of ≥ 200 ng/mL was achieved at the 1000-mg dose level. At this dose, the Cmax and AUC0-∞ were 326 ± 67 ng/mL and 13.4 ± 8.6 · 103 ng · h/mL, respectively. In summary, the ASD tablet was well tolerated, resulted in relevant pharmacokinetic exposure, and can be used for proof-of-concept clinical studies.- Published
- 2017
- Full Text
- View/download PDF
30. Pharmacokinetics and excretion of (14)C-omacetaxine in patients with advanced solid tumors.
- Author
-
Nijenhuis CM, Hellriegel E, Beijnen JH, Hershock D, Huitema AD, Lucas L, Mergui-Roelvink M, Munteanu M, Rabinovich-Guilatt L, Robertson P Jr, Rosing H, Spiegelstein O, and Schellens JH
- Subjects
- Adult, Aged, Antineoplastic Agents, Phytogenic adverse effects, Antineoplastic Agents, Phytogenic blood, Antineoplastic Agents, Phytogenic urine, Carbon Radioisotopes, Feces chemistry, Female, Harringtonines adverse effects, Harringtonines blood, Harringtonines urine, Homoharringtonine, Humans, Male, Middle Aged, Neoplasms blood, Neoplasms urine, Antineoplastic Agents, Phytogenic pharmacokinetics, Harringtonines pharmacokinetics, Neoplasms metabolism
- Abstract
Background Omacetaxine mepesuccinate is indicated in adults with chronic myeloid leukemia resistant and/or intolerant to ≥ 2 tyrosine kinase inhibitor treatments. This phase I study assessed the disposition, elimination, and safety of (14)C-omacetaxine in patients with solid tumors. Methods The study comprised a 7-days pharmacokinetic assessment followed by a treatment period of ≤ six 28-days cycles. A single subcutaneous dose of 1.25 mg/m(2) (14)C-omacetaxine was administered to six patients. Blood, urine, and feces were collected through 168 h or until radioactivity excreted within 24 h was <1 % of the dose. Total radioactivity (TRA) was measured in all matrices and concentrations of omacetaxine, 4'-desmethylhomoharringtonine (4'-DMHHT), and cephalotaxine were measured in plasma and urine. For each treatment cycle, patients received 1.25 mg/m(2) omacetaxine twice daily for 7 days. Results Mean TRA recovered was approximately 81 % of the dose, with approximately half of the radioactivity recovered in feces and half in urine. Approximately 20 % of the dose was excreted unchanged in urine; cephalotaxine (0.4 % of dose) and 4' DMHHT (9 %) were also present. Plasma concentrations of TRA were higher than the sum of omacetaxine and known metabolites, suggesting the presence of other (14)C-omacetaxine-derived compounds. Fatigue and anemia were common, consistent with the known toxicity profile of omacetaxine. Conclusion Renal and hepatic processes contribute to the elimination of (14)C-omacetaxine-derived radioactivity in cancer patients. In addition to omacetaxine and its known metabolites, other (14)C-omacetaxine-derived materials appear to be present in plasma and urine. Omacetaxine was adequately tolerated, with no new safety signals., Competing Interests: Compliance with ethical standards This study was conducted in full accordance with the International Conference on Harmonisation Good Clinical Practice Consolidated Guideline (E6) and any applicable national and local laws and regulations. Written informed consent was obtained from each patient before any study procedures or assessments were initiated. Potential conflicts of interest are as follows: CMN, JHB, ADRH, LL, MM-R, HR, and JHMS report employment from Netherlands Cancer Institute and research support from Teva Branded Pharmaceutical Products R&D during the conduction of the study; EH, LR-G, and PR Jr. report employment and stock options from Teva Branded Pharmaceutical Products R&D during the conduction of the study; OS and DH report employment from Teva Branded Pharmaceutical Products R&D during the conduction of the study; MM has nothing to disclose.
- Published
- 2016
- Full Text
- View/download PDF
31. Baclofen overdose treated with continuous venovenous hemofiltration.
- Author
-
Meulendijks D, Khan S, Koks CH, Huitema AD, Schellens JH, and Beijnen JH
- Subjects
- Humans, Metabolic Clearance Rate, Middle Aged, Muscle Relaxants, Central blood, Muscle Relaxants, Central pharmacokinetics, Muscle Relaxants, Central poisoning, Baclofen blood, Baclofen pharmacokinetics, Baclofen poisoning, Drug Overdose surgery, Hemofiltration
- Abstract
Purpose: Overdose with baclofen, a derivative of the inhibitory neurotransmitter γ-aminobutyric acid, may lead to severe respiratory and central nervous system depression and can be life-threatening. Prolonged half-lives of baclofen, of up to 34 h, have been reported in patients after overdose. Hemodialysis has proven to be a successful approach to improve clearance of baclofen, but the value of continuous venovenous hemofiltration (CVVH) is unclear. We applied CVVH in a patient with acute baclofen overdose., Methods: Pharmacokinetic measurements of baclofen in serum and hemofiltrate were made at six time points after hospital admission. Baclofen concentration-time data were analyzed using non-compartmental methods, and the relative contribution of clearance by hemofiltration to total baclofen clearance was calculated., Results: Baclofen concentrations in serum varied between 1.81 and 0.05 mg/L. Concentrations of baclofen in hemofiltrate were within the same range (between 0.74 and 0.05 mg/L), and the elimination half-life during hemofiltration was estimated at 4.8 h. Total clearance and clearance via hemofiltration were estimated at 6.6 and 2.4 L/h, indicating that clearance could be increased by approximately 57 % by applying hemofiltration., Conclusions: The presented case demonstrates the usefulness of CVVH in the treatment of baclofen overdose and indicates that CVVH can be used as an alternative to hemodialysis in patients with overdose of baclofen.
- Published
- 2015
- Full Text
- View/download PDF
32. Pharmacokinetics and excretion of (14)C-lenvatinib in patients with advanced solid tumors or lymphomas.
- Author
-
Dubbelman AC, Rosing H, Nijenhuis C, Huitema AD, Mergui-Roelvink M, Gupta A, Verbel D, Thompson G, Shumaker R, Schellens JH, and Beijnen JH
- Subjects
- Adult, Antineoplastic Agents adverse effects, Antineoplastic Agents therapeutic use, Carbon Radioisotopes blood, Carbon Radioisotopes pharmacokinetics, Carbon Radioisotopes urine, Feces chemistry, Female, Humans, Male, Middle Aged, Neoplasms drug therapy, Phenylurea Compounds adverse effects, Phenylurea Compounds therapeutic use, Protein Kinase Inhibitors adverse effects, Protein Kinase Inhibitors therapeutic use, Quinolines adverse effects, Quinolines therapeutic use, Treatment Outcome, Antineoplastic Agents pharmacokinetics, Neoplasms metabolism, Phenylurea Compounds pharmacokinetics, Protein Kinase Inhibitors pharmacokinetics, Quinolines pharmacokinetics
- Abstract
Lenvatinib is an orally available multi-targeted tyrosine kinase inhibitor with anti-angiogenic and antitumor activity. To get more insight into the disposition of lenvatinib, a mass balance study was performed in patients with advanced solid tumors. A single oral 24 mg (100 μCi) dose of (14)C-lenvatinib was administered to six patients, followed by collection of blood, plasma, urine and feces for 7 to 10 days. The collected material was analyzed for total radioactivity, unchanged lenvatinib and selected metabolites. The safety and antitumor effect of a daily oral dose of 24 mg non-labeled lenvatinib were assessed in the extension phase of the study. Peak plasma concentrations of lenvatinib and total radioactivity were reached 1.6 and 1.4 h after administration, respectively, and their terminal phase half-lifes were 34.5 and 17.8 h, respectively. Unchanged lenvatinib systemic exposure accounted for 60 % of the total radioactivity in plasma. Peak concentrations of the analyzed metabolite were over 700-fold lower than the peak plasma concentration of lenvatinib. Ten days after the initial dose, the geometric mean (± CV) recovery of administered dose was 89 % ±10 %, with 64 % ±11 % recovered in feces and 25 % ±18 % in urine. Unchanged lenvatinib in urine and feces accounted for 2.5 % ±68 % of the administered dose, indicating a major role of metabolism in the elimination of lenvatinib. In conclusion, lenvatinib is rapidly absorbed and extensively metabolized, with subsequent excretion in urine and, more predominantly, in feces. Additionally, lenvatinib showed acceptable safety and preliminary antitumor activity.
- Published
- 2015
- Full Text
- View/download PDF
33. Design of informative renal impairment studies: evaluation of the impact of design stratification on bias, precision and dose adjustment error.
- Author
-
van Hasselt JG, Schellens JH, Beijnen JH, and Huitema AD
- Subjects
- Dose-Response Relationship, Drug, Humans, Models, Biological, Pharmacokinetics, Renal Insufficiency metabolism, Research Design
- Abstract
Purpose: Renal impairment (RI) studies are conducted to estimate the impact of RI on pharmacokinetics (PK). In some disease areas, these studies can be difficult to conduct, for instance due to the limited number of eligible patients. The objective of this analysis was to evaluate bias and precision of population PK parameters, and the dose adjustment error (DAE) for RI studies i) with different levels of study design imbalance in the stratification of subjects across RI categories, and ii) that include additional patients in the control arm of RI studies, that may be available from previously conducted PK studies., Methods: Study designs were simulated and re-estimated using a hypothetical 2-compartmental PK model with varying magnitude of the fraction of renal elimination (FR) and magnitude of between-subject variability (BSV). The DAE was computed based on the difference between the theoretical necessary dose adjustment versus the empirical estimated dose adjustment to reach a similar exposure as controls., Results: Although some design imbalance may still lead to DAEs of acceptable magnitude (DAE < -11.05-14.44 inter-quartile range, IQR), at least some patients are necessary in the more severe RI groups. When 100 additional patients with normal renal function were included in a sub-informative design, the DAE changed from < -7.63-16.64 IQR to < -8.89-8.69 IQR., Conclusions: We quantified the impact of study design imbalance on bias and precision of PK parameters and DAE, as may occur for RI studies in some indications. Adding additional data from earlier studies to the analysis dataset improves the bias and precision of PK parameters.
- Published
- 2014
- Full Text
- View/download PDF
34. The effect of seasonal variation and secretion of sunitinib in sweat on the development of hand-foot syndrome.
- Author
-
Lankheet NA, Huitema AD, Mallo H, Adriaansz S, Haanen JB, Schellens JH, Beijnen JH, and Blank CU
- Subjects
- Aged, Antineoplastic Agents adverse effects, Antineoplastic Agents blood, Female, Hand-Foot Syndrome etiology, Humans, Indoles adverse effects, Indoles blood, Male, Middle Aged, Pyrroles adverse effects, Pyrroles blood, Seasons, Sunitinib, Antineoplastic Agents pharmacokinetics, Hand-Foot Syndrome metabolism, Indoles pharmacokinetics, Pyrroles pharmacokinetics, Sweat metabolism
- Abstract
Background: Hand-foot syndrome (HFS) is a side effect of sunitinib with considerable impact on quality of life. Seasonal variation and hyperhydrosis are possibly correlated to occurrence of HFS. Therefore, we proposed to study the prevalence of HFS in different seasons retrospectively and to study the relationship between sunitinib sweat secretion and HFS prospectively., Patients and Methods: A retrospective cohort of 19 patients treated with sunitinib was used to determine seasonal prevalence of HFS. In a prospective study, sunitinib and N-desethyl sunitinib levels in sweat patches of 25 patients treated with sunitinib were quantified and correlated to severity of HFS., Results: In the retrospective cohort, the patients suffered from more severe HFS during summertime compared with the rest of the year. In the prospective study, the cumulative amounts of sunitinib plus metabolite measured in the patches of the on-treatment phase (median 129.4 ng/patch) were higher than the off-treatment phase (median 39.5 ng/patch). A tendency was observed towards increasing amounts of drug per patch with increasing severity of HFS., Conclusion: Patients experienced more HFS in summer time compared to other seasons. However, no statistically significant correlation between sunitinib sweat secretion and severity of HFS could be demonstrated within our patient cohort.
- Published
- 2013
- Full Text
- View/download PDF
35. Pharmacokinetic evaluation of three oral formulations of docetaxel boosted with ritonavir: two single-drug formulations vs. a fixed-dose combination tablet.
- Author
-
Moes JJ, Stuurman FE, Hendrikx JJ, Marchetti S, Huitema AD, Beijnen JH, Schellens JH, and Nuijen B
- Abstract
The ability to deliver the potent anti-cancer agent docetaxel via the oral route may enable the development of promising new treatment regimens with reduced toxicity, increased efficacy, and increased patient convenience. Recently, we were able to overcome the low oral bioavailability of docetaxel by concomitant administration of the pharmacokinetic booster ritonavir and the design of an oral solid dispersion formulation of docetaxel (ModraDoc001 10-mg capsule). Further research lead to the development of a docetaxel tablet (ModraDoc003 10-mg tablet) and a fixed-dose combination (FDC) tablet of docetaxel and ritonavir (ModraDoc004 10/50-mg tablet). In this clinical proof-of-concept study the exposure to docetaxel and ritonavir was compared between the single agent formulations and the FDC tablet. Six evaluable patients received 40 mg docetaxel and 200 mg of ritonavir once a week according to a cross-over design. No significant differences were found in the exposure to docetaxel and ritonavir between the single agent formulations and the FDC tablet. There was, however, a tendency towards a higher exposure to docetaxel after the administration of the FDC tablet, which could be an effect of the simultaneous release of docetaxel and ritonavir in the gastrointestinal tract. The FDC tablet of docetaxel and ritonavir is a pharmaceutically and clinically feasibly option in the development of patient convenient oral anti-cancer therapy with docetaxel.
- Published
- 2013
- Full Text
- View/download PDF
36. Two-stage model-based design of cancer phase I dose escalation trials: evaluation using the phase I program of barasertib (AZD1152).
- Author
-
Keizer RJ, Zandvliet AS, Beijnen JH, Schellens JH, and Huitema AD
- Subjects
- Antineoplastic Agents administration & dosage, Antineoplastic Agents pharmacokinetics, Antineoplastic Agents therapeutic use, Dose-Response Relationship, Drug, Humans, Leukocyte Count, Maximum Tolerated Dose, Organophosphates pharmacokinetics, Quinazolines pharmacokinetics, Clinical Trials, Phase I as Topic methods, Models, Biological, Organophosphates administration & dosage, Organophosphates therapeutic use, Quinazolines administration & dosage, Quinazolines therapeutic use
- Abstract
Introduction: Modeling and simulation of pharmacokinetics and pharmacodynamics has previously been shown to be potentially useful in designing Phase I programs of novel anti-cancer agents that show hematological toxicity. In this analysis, a two-stage model-based trial design was evaluated retrospectively using data from the Phase I program with the aurora kinase inhibitor barasertib., Methods: Data from two Phase I trials and four regimens were used (n = 79). Using barasertib-hydroxy QPA plasma concentrations and neutrophil count data from only study 1A, a PKPD model was developed and subsequently used to predict the MTD and a safe starting dose for the other trials., Results: The PKPD model based on data from the first study adequately described the time course of neutrophil count fluctuation. The two-stage model-based design provided safe starting doses for subsequent phase I trials for barasertib. Predicted safe starting dose levels were higher than those used in two subsequent trials, but lower than used in the other trial., Discussion: The two-stage approach could have been applied safely to define starting doses for alternative dosing strategies with barasertib. The limited improvement in efficiency for the phase I program of barasertib may have been due to the fact that starting doses for the studied phase I trials were already nearly optimal., Conclusion: Application of the two-stage model-based trial design in Phase I programs with novel anti-cancer drugs that cause haematological toxicity is feasible, safe, and may lead to a reduction in the number of patient treated at sub-therapeutic dose-levels.
- Published
- 2012
- Full Text
- View/download PDF
37. Predictive ability of a semi-mechanistic model for neutropenia in the development of novel anti-cancer agents: two case studies.
- Author
-
Soto E, Keizer RJ, Trocóniz IF, Huitema AD, Beijnen JH, Schellens JH, Wanders J, Cendrós JM, Obach R, Peraire C, Friberg LE, and Karlsson MO
- Subjects
- Adult, Aged, Antineoplastic Agents administration & dosage, Camptothecin administration & dosage, Camptothecin adverse effects, Female, Humans, Leukocyte Count, Male, Middle Aged, Neutropenia blood, Predictive Value of Tests, Sulfonamides administration & dosage, Treatment Outcome, Antineoplastic Agents adverse effects, Camptothecin analogs & derivatives, Drug Discovery, Models, Biological, Neutropenia chemically induced, Sulfonamides adverse effects
- Abstract
In cancer chemotherapy neutropenia is a common dose-limiting toxicity. An ability to predict the neutropenic effects of cytotoxic agents based on proposed trial designs and models conditioned on previous studies would be valuable. The aim of this study was to evaluate the ability of a semi-mechanistic pharmacokinetic/pharmacodynamic (PK/PD) model for myelosuppression to predict the neutropenia observed in Phase I clinical studies, based on parameter estimates obtained from prior trials. Pharmacokinetic and neutropenia data from 5 clinical trials for diflomotecan and from 4 clinical trials for indisulam were used. Data were analyzed and simulations were performed using the population approach with NONMEM VI. Parameter sets were estimated under the following scenarios: (a) data from each trial independently, (b) pooled data from all clinical trials and (c) pooled data from trials performed before the tested trial. Model performance in each of the scenarios was evaluated by means of predictive (visual and numerical) checks. The semi-mechanistic PK/PD model for neutropenia showed adequate predictive ability for both anti-cancer agents. For diflomotecan, similar predictions were obtained for the three scenarios. For indisulam predictions were better when based on data from the specific study, however when the model parameters were conditioned on data from trials performed prior to a specific study, similar predictions of the drug related-neutropenia profiles and descriptors were obtained as when all data were used. This work provides further indication that modeling and simulation tools can be applied in the early stages of drug development to optimize future trials.
- Published
- 2011
- Full Text
- View/download PDF
38. A model of hypertension and proteinuria in cancer patients treated with the anti-angiogenic drug E7080.
- Author
-
Keizer RJ, Gupta A, Mac Gillavry MR, Jansen M, Wanders J, Beijnen JH, Schellens JH, Karlsson MO, and Huitema AD
- Subjects
- Antihypertensive Agents administration & dosage, Antihypertensive Agents therapeutic use, Blood Pressure drug effects, Dose-Response Relationship, Drug, Drug Administration Schedule, Humans, Hypertension drug therapy, Neoplasms metabolism, Neoplasms urine, Proteinuria prevention & control, Urinalysis, Angiogenesis Inhibitors adverse effects, Angiogenesis Inhibitors pharmacokinetics, Angiogenesis Inhibitors therapeutic use, Hypertension chemically induced, Models, Biological, Neoplasms drug therapy, Phenylurea Compounds adverse effects, Phenylurea Compounds pharmacokinetics, Phenylurea Compounds therapeutic use, Proteinuria chemically induced, Quinolines adverse effects, Quinolines pharmacokinetics, Quinolines therapeutic use
- Abstract
Hypertension and proteinuria are commonly observed side-effects for anti-angiogenic drugs targeting the VEGF pathway. In most cases, hypertension can be controlled by prescription of anti-hypertensive (AH) therapy, while proteinuria often requires dose reductions or dose delays. We aimed to construct a pharmacokinetic-pharmacodynamic (PK-PD) model for hypertension and proteinuria following treatment with the experimental VEGF-inhibitor E7080, which would allow optimization of treatment, by assessing the influence of anti-hypertensive medication and dose reduction or dose delays in treating and avoiding toxicity. Data was collected from a phase I study of E7080 (n = 67), an inhibitor of multiple tyrosine kinases, among which VEGF. Blood pressure and urinalysis data were recorded weekly. Modeling was performed in NONMEM, and direct and indirect response PK-PD models were evaluated. A previously developed PK model was used. An indirect response PK-PD model described the increase in BP best, while the probability of developing proteinuria toxicity in response to exposure to E7080, was best described by a Markov transition model. This model may guide clinical interventions and provide treatment recommendations for E7080, and may serve as a template model for other drugs in this class.
- Published
- 2010
- Full Text
- View/download PDF
39. Two-stage model-based clinical trial design to optimize phase I development of novel anticancer agents.
- Author
-
Zandvliet AS, Karlsson MO, Schellens JH, Copalu W, Beijnen JH, and Huitema AD
- Subjects
- Antineoplastic Agents pharmacokinetics, Computer Simulation, Dose-Response Relationship, Drug, Humans, Sulfonamides administration & dosage, Sulfonamides pharmacology, Antineoplastic Agents pharmacology, Clinical Trials, Phase I as Topic, Drug Design, Models, Statistical
- Abstract
Background: The phase I program of anticancer agents usually consists of multiple dose escalation studies to select a safe dose for various administration schedules. We hypothesized that pharmacokinetic and pharmacodynamic (PK-PD) modeling of an initial phase I study (stage 1) can be used for selection of an optimal starting dose for subsequent studies (stage 2) and that a post-hoc PK-PD analysis enhances the selection of a recommended dose for phase II evaluation. The aim of this analysis was to demonstrate that this two-stage model-based design, which does not interfere in the conduct of trials, is safe, efficient and effective., Methods: PK and PD data of dose escalation studies were simulated for nine compounds and for five administration regimens (stage 1) for drugs with neutropenia as dose-limiting toxicity. PK-PD models were developed for each simulated study and were used to determine a starting dose for additional phase I studies (stage 2). The model-based design was compared to a conventional study design regarding safety (number of dose-limiting toxicities (DLTs)), efficiency (number of patients treated with a dose below the recommended dose) and effectiveness (precision of dose selection). Retrospective data of the investigational anticancer drug indisulam were used to show the applicability of the model-based design., Results: The model-based design was as safe as the conventional design (median number of DLTs = 3) and resulted in a reduction of the number of patients who were treated with a dose below the recommended dose (-27%, power 89%). A post-hoc model-based determination of the recommended dose for future phase II studies was more precise than the conventional selection of the recommended dose (root mean squared error 8.3% versus 30%)., Conclusions: A two-stage model-based phase I design is safe for anticancer agents with dose-limiting myelosuppression and may enhance the efficiency of dose escalation studies by reducing the number of patients treated with a dose below the recommended dose and by increasing the precision of dose selection for phase II evaluation.
- Published
- 2010
- Full Text
- View/download PDF
40. Application of population pharmacokinetic modeling in early clinical development of the anticancer agent E7820.
- Author
-
Keizer RJ, Zamacona MK, Jansen M, Critchley D, Wanders J, Beijnen JH, Schellens JH, and Huitema AD
- Subjects
- Administration, Oral, Adult, Aged, Antineoplastic Agents administration & dosage, Female, Food, Humans, Indoles administration & dosage, Male, Middle Aged, Sulfonamides administration & dosage, Antineoplastic Agents pharmacokinetics, Indoles pharmacokinetics, Models, Biological, Sulfonamides pharmacokinetics
- Abstract
The aim of this study was to assess the population pharmacokinetics (PopPK) of the novel oral anti-cancer agent E7820. Both a non-linear mixed effects modeling analysis and a non-compartmental analysis (NCA) were performed and results were compared. Data were obtained from a phase I dose escalation study in patients with malignant solid tumors or lymphomas. E7820 was administered daily for 28 days, followed by a washout period of 7 days prior to the start of subsequent cycles. A one compartment model with linear elimination from the central compartment was shown to give adequate fit, while absorption was described using a turnover model. Final population parameter estimates of basic PK parameters obtained with the PopPK method were (RSE): clearance, 6.24 L/h (7.1%), volume of distribution, 66.0 L (8.5%), mean transit time to the absorption compartment, 0.638 h (6.5%). The intake of food prior to dose administration slowed absorption (2.8-fold, RSE 13%) and increased relative bioavailability of E7820 by 36% (RSE 14%), although the effect on C (max) and AUC was not significant. Comparison with the NCA approach showed approximately equal PK parameter estimates and food effect measures, although specific advantages of PopPK included efficiency in use of data and more appropriate assessment of variability.
- Published
- 2009
- Full Text
- View/download PDF
41. Covariate-based dose individualization of the cytotoxic drug indisulam to reduce the risk of severe myelosuppression.
- Author
-
Zandvliet AS, Schellens JH, Copalu W, Beijnen JH, and Huitema AD
- Subjects
- Adult, Aged, Aged, 80 and over, Antineoplastic Agents administration & dosage, Antineoplastic Agents adverse effects, Antineoplastic Agents pharmacokinetics, Body Surface Area, Clinical Trials as Topic, Computer Simulation, Cytochrome P-450 Enzyme System genetics, Cytotoxins pharmacokinetics, Female, Hematologic Diseases diagnosis, Humans, Male, Middle Aged, Models, Biological, Models, Statistical, Neutropenia chemically induced, Neutropenia diagnosis, Neutropenia prevention & control, Racial Groups, Risk, Risk Factors, Sex Factors, Sulfonamides pharmacokinetics, Thrombocytopenia chemically induced, Thrombocytopenia diagnosis, Thrombocytopenia prevention & control, Young Adult, Algorithms, Cytotoxins administration & dosage, Cytotoxins adverse effects, Hematologic Diseases chemically induced, Hematologic Diseases prevention & control, Sulfonamides administration & dosage, Sulfonamides adverse effects
- Abstract
Aim: Chemotherapy with indisulam causes myelosuppression. This study aimed to evaluate the influence of patient-related covariates on pharmacokinetics and pharmacodynamics, to identify patients at risk for severe myelosuppression and to develop a dosing algorithm for treatment optimization., Methods: Pharmacokinetic and pharmacodynamic data of 412 patients were available. Non-linear mixed effects modeling was used to determine the relative risk of dose-limiting myelosuppression for various covariates (demographics, physical condition, prior treatment, comedication, CYP2C genotype and biochemistry)., Results: Body surface area (BSA), race and CYP2C genotype had a significant impact on indisulam elimination (P < 0.001). Low BSA, Japanese race, variant CYP2C genotype, low baseline neutrophil and thrombocyte counts and female sex were clinically relevant risk factors of dose-limiting myelosuppression (RR > 1.1). A dosing strategy was developed to optimize treatment for patient subgroups., Conclusions: This study has identified covariates related to an increased risk of myelosuppression after indisulam therapy. Dose individualization may contribute to treatment optimization.
- Published
- 2009
- Full Text
- View/download PDF
42. A semi-physiological population pharmacokinetic model describing the non-linear disposition of indisulam.
- Author
-
Zandvliet AS, Schellens JH, Copalu W, Beijnen JH, and Huitema AD
- Subjects
- Algorithms, Antineoplastic Agents administration & dosage, Antineoplastic Agents pharmacokinetics, Area Under Curve, Asian People, Blood Proteins metabolism, Carbonic Anhydrases metabolism, Clinical Trials, Phase I as Topic, Computer Simulation, Dose-Response Relationship, Drug, Erythrocytes metabolism, Extracellular Fluid metabolism, Humans, Metabolic Clearance Rate, Multivariate Analysis, Protein Binding, Regression Analysis, Sulfonamides administration & dosage, Tissue Distribution, White People, Models, Biological, Sulfonamides pharmacokinetics
- Abstract
The pharmacokinetic profile of the anti-cancer agent indisulam is non-linear. In addition to non-linear clearance, this may partially be explained by saturable distribution to red blood cells and saturable plasma protein binding. The aims of this study were to develop a semi-physiological population pharmacokinetic model of indisulam and to examine the impact of protein binding and distribution to red blood cells. Indisulam concentrations in plasma, plasma ultrafiltrate and in red blood cells from multiple phase I studies in Caucasian and Japanese patients were used to develop a pharmacokinetic model using NONMEM. This model comprised four physiological compartments: plasma, red blood cells, interstitial fluid and tissue. A simulation study was performed to examine the contribution of physiological processes in indisulam pharmacokinetics. Plasma albumin concentrations were predictive for the maximal protein binding capacity and considerably influenced total plasma concentrations of indisulam, whereas free plasma concentrations remained relatively unaffected. The maximal specific red blood cell binding capacity was 64.0 ( +/-5.9) mg/L and corresponded to the typical red blood cell carbonic anhydrase concentration. Simulation studies demonstrated that the hematocrit does not have a clinically relevant impact on indisulam disposition. This semi-physiological model allowed adequate prediction of the time profiles of indisulam concentrations in all monitored compartments for a large range of dose levels and several treatment regimens. The model has elucidated the mechanism and the role of saturable plasma protein binding and saturable distribution to red blood cells in indisulam pharmacokinetics and provides a basis for rationale PK-PD relationships.
- Published
- 2006
- Full Text
- View/download PDF
43. Semi-physiological model describing the hematological toxicity of the anti-cancer agent indisulam.
- Author
-
van Kesteren C, Zandvliet AS, Karlsson MO, Mathôt RA, Punt CJ, Armand JP, Raymond E, Huitema AD, Dittrich C, Dumez H, Roché HH, Droz JP, Ravic M, Yule SM, Wanders J, Beijnen JH, Fumoleau P, and Schellens JH
- Subjects
- Adult, Aged, Antineoplastic Agents administration & dosage, Clinical Trials, Phase I as Topic, Drug Administration Schedule, Female, Humans, Male, Middle Aged, Sulfonamides administration & dosage, Antineoplastic Agents adverse effects, Models, Biological, Neutropenia chemically induced, Sulfonamides adverse effects, Thrombocytopenia chemically induced
- Abstract
Indisulam (N-(3-chloro-7-indolyl)-1,4-benzenedisulfonamide, GOAL, E7070) is a novel anti-cancer drug currently in phase II clinical development for the treatment of solid tumors. Phase I dose-escalation studies were conducted comparing four treatment schedules. Neutropenia and thrombocytopenia were dose limiting in all schedules. The aim of this study was to describe the extent and the time course of the hematological toxicity and its possible schedule dependency using a semi-physiological model. Data from 142 patients were analyzed using NONMEM. The semi-physiological model comprised a progenitor blood cell compartment, linked to the central circulation compartment, through 3 transition compartments representing the maturation chain in the bone marrow. Plasma concentrations of the drug were assumed to reduce the proliferation rate in the progenitor compartment according to a linear function. A feedback mechanism was included in the model representing the rebound effect of endogenous growth factors. The model was validated using a posterior predictive check. The model adequately described the extent and time course of neutropenia and thrombocytopenia. The mean transition time (MTT, i.e. maturation time in bone marrow) of neutrophils was increased by 47% in patients who received indisulam as a weekly dose administered for four out of every six weeks. For platelets, MTT was increased by 33% in patients who received this schedule and also in patients who received a continuous 120-h infusion. The validation procedure indicated that the model adequately predicts the nadir value of neutrophils and platelets and the time to reach this nadir. A semi-physiological model was successfully applied to describe the time course and extent of the neutropenia and thrombocytopenia after indisulam administration for four treatment schedules.
- Published
- 2005
- Full Text
- View/download PDF
44. Integrated Population Pharmacokinetic Model of both cyclophosphamide and thiotepa suggesting a mutual drug-drug interaction.
- Author
-
de Jonge ME, Huitema AD, Rodenhuis S, and Beijnen JH
- Subjects
- Adolescent, Adult, Computer Simulation, Drug Interactions, Female, Humans, Male, Middle Aged, Models, Biological, Cyclophosphamide pharmacokinetics, Thiotepa pharmacokinetics
- Abstract
Purpose/aims: Cyclophosphamide (CP) and thiotepa (TT) are frequently administered simultaneously in high-dose chemotherapy regimens. The prodrug CP shows strong autoinduction resulting in increased formation of its activated metabolite 4-hydroxycyclophosphamide (4OHCP). TT inhibits this bioactivation of CP. Previously, we successfully modelled CP bioactivation and the effect of TT on the autoinduction. Recently we suggested that CP may also induce the conversion of TT in to its metabolite tepa (T). The aim of the current study was to investigate whether the influence of CP on TT metabolism can be described with a population pharmacokinetic model and whether this interaction can be incorporated in an integrated model describing both CP and TT pharmacokinetics., Methods: Plasma samples were collected from 49 patients receiving 86 courses of a combination of high-dose CP (4000 or 6000 mg/m2), TT (320 or 480 mg/m2) and carboplatin (1067 or 1600 mg/m2) given in short infusions during four consecutive days. For each patient, approximately 20 plasma samples were available per course. Concentrations of CP, 4OHCP, TT and T were determined using GC and HPLC. Kinetic data were processed using NONMEM., Results: The pharmacokinetics of TT and T were described with a two-compartment model. TT was eliminated through a non-inducible and an inducible pathway, the latter resulting information of T (ClindTT = 12.4 l/hr, ClnonindTT = 17.0 l/hr). Induction of TT metabolism was mediated by a hypothetical amount of enzyme, different from that involved in CP induction, whose amount increased with time in the presence of CP. The amount of enzyme followed a zero-order formation and a decrease with a first-order elimination rate constant of 0.0343 hr(-1) (t1/2 = 20 hr). This model was significantly better than a model lacking the induction by CP. The model was successfully incorporated into the previously published pharmacokinetic model for CP, and resulted in comparable parameter estimates for this compound and its metabolite 4OHCP., Conclusion: The pharmacokinetics of TT, when administered in combination with CP, were successfully described. The model confirms induction of TT metabolism with time and it appears likely that CP is responsible for this phenomenon. The existence of a mutual pharmacokinetic interaction between CP and TT, as described in our integrated model, may be relevant in clinical practice.
- Published
- 2004
- Full Text
- View/download PDF
45. Incidence and risk factors for nevirapine-associated rash.
- Author
-
de Maat MM, ter Heine R, Mulder JW, Meenhorst PL, Mairuhu AT, van Gorp EC, Huitema AD, and Beijnen JH
- Subjects
- Adult, Ambulatory Care, Anti-HIV Agents blood, Anti-HIV Agents therapeutic use, Dose-Response Relationship, Drug, Drug Therapy, Combination, Female, HIV Infections drug therapy, Humans, Logistic Models, Male, Nevirapine blood, Nevirapine therapeutic use, Protease Inhibitors therapeutic use, Reverse Transcriptase Inhibitors therapeutic use, Risk Factors, Time Factors, Anti-HIV Agents adverse effects, Exanthema chemically induced, Nevirapine adverse effects
- Abstract
Objective: To determine the incidence of rash in HIV-1 infected individuals starting a nevirapine-containing regimen in an unselected outpatient clinic population. Possible risk factors including plasma concentrations of nevirapine were evaluated for their relationship with the occurrence of a rash., Methods: The occurrence of rash was extracted from the outpatient medical records or based on a prescription of the antihistaminic cetirizine as documented by the community pharmacy within the first 90 days of nevirapine use. During regular visits to the clinic blood samples were collected for the determination of nevirapine plasma concentrations. Possible risk factors such as demographics, immunology, virology, clinical chemistry and antiretroviral pretreatment were collected at baseline for each patient. In addition, concomitantly used drugs during the nevirapine-based regimen were recorded. The association between these factors and the occurrence of rash was studied. Primary outcome was the onset of rash within the first 90 days after initiation of a nevirapine-containing regimen., Results: Data from 216 HIV-1-infected patients were used in this study. Thirty-eight patients (17.6%) developed a rash of some grade that led to discontinuation of nevirapine in seven patients (3.2% of the included patients). The median time to occurrence of rash was 26 days (interquartile range 17-46 days). The multivariate analysis showed that patients pretreated with antiretroviral drugs less than 12 months before the initiation of a nevirapine-containing regimen had a more than 2.5-fold increased risk of developing rash. Furthermore, nevirapine plasma concentrations were also significantly related to the occurrence of rash. A more than twofold increased risk for developing rash was observed for patients with nevirapine plasma concentrations above 5.3 mg/l., Conclusions: This is the first study demonstrating that patients with antiretroviral pretreatment less than 12 months and with nevirapine plasma concentrations above 5.3 mg/l during the first 90 days of treatment are at a higher risk for the development of rash. It is therefore advised to monitor this group of patients carefully when initiating nevirapine-containing therapy.
- Published
- 2003
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.