Skip to main content
Top
Published in: Cancer Chemotherapy and Pharmacology 5/2010

01-04-2010 | Original Article

Limited inter-occasion variability in relation to inter-individual variability in chemotherapy-induced myelosuppression

Authors: Emma K. Hansson, Johan E. Wallin, Henrik Lindman, Marie Sandström, Mats O. Karlsson, Lena E. Friberg

Published in: Cancer Chemotherapy and Pharmacology | Issue 5/2010

Login to get access

Abstract

Purpose

A previously developed semi-physiological model of chemotherapy-induced myelosuppression has shown consistent system-related parameter and inter-individual variability (IIV) estimates across drugs. A requirement for dose individualization to be useful is relatively low variability between treatment courses (inter-occasion variability [IOV]) in relation to IIV. The objective of this study was to evaluate and compare magnitudes of IOV and IIV in myelosuppression model parameters across six different anti-cancer drug treatments.

Methods

Neutrophil counts from several treatment courses following therapy with docetaxel, paclitaxel, epirubicin-docetaxel, 5-fluorouracil–epirubicin–cyclophosphamide, topotecan, and etoposide were included in the analysis. The myelosuppression model was fitted to the data using NONMEM VI. IOV in the model parameters baseline neutrophil counts (ANC0), mean transit time through the non-mitotic maturation chain (mean transit time [MTT]), and the parameter describing the concentration–effect relationship (slope), were evaluated for statistical significance (P < 0.001).

Results

Inter-occasion variability in MTT was significant for all the investigated datasets, except for topotecan, and was of similar magnitude (8–16 CV%). IOV in slope was significant for docetaxel, topotecan, and etoposide (19–39 CV%). For all six investigated datasets, the IOV in myelosuppression parameters was lower than the IIV. There was no indication of systematic shifts in the system- or drug sensitivity-related parameters over time across datasets.

Conclusion

This study indicates that the semi-physiological model of chemotherapy-induced myelosuppression has potential to be used for prediction of the time-course of myelosuppression in future courses and is, thereby, a valuable step towards individually tailored anticancer drug therapy.
Literature
1.
go back to reference Sawyer M, Ratain MJ (2001) Body surface area as a determinant of pharmacokinetics and drug dosing. Invest New Drugs 19:171–177CrossRefPubMed Sawyer M, Ratain MJ (2001) Body surface area as a determinant of pharmacokinetics and drug dosing. Invest New Drugs 19:171–177CrossRefPubMed
2.
go back to reference Crawford J, Dale DC, Lyman GH (2004) Chemotherapy-induced neutropenia—risks, consequences, and new directions for its management. Cancer 100:228–237CrossRefPubMed Crawford J, Dale DC, Lyman GH (2004) Chemotherapy-induced neutropenia—risks, consequences, and new directions for its management. Cancer 100:228–237CrossRefPubMed
3.
go back to reference Mayers C, Panzarella T, Tannock IF (2001) Analysis of the prognostic effects of inclusion in a clinical trial and of myelosuppression on survival after adjuvant chemotherapy for breast carcinoma. Cancer 91:2246–2257CrossRefPubMed Mayers C, Panzarella T, Tannock IF (2001) Analysis of the prognostic effects of inclusion in a clinical trial and of myelosuppression on survival after adjuvant chemotherapy for breast carcinoma. Cancer 91:2246–2257CrossRefPubMed
4.
go back to reference Saarto T, Blomqvist C, Rissanen P, Auvinen A, Elomaa I (1997) Haematological toxicity: A marker of adjuvant chemotherapy efficacy in stage II and III breast cancer. Br J Cancer 75:301–305PubMed Saarto T, Blomqvist C, Rissanen P, Auvinen A, Elomaa I (1997) Haematological toxicity: A marker of adjuvant chemotherapy efficacy in stage II and III breast cancer. Br J Cancer 75:301–305PubMed
5.
go back to reference Cameron DA, Massie C, Kerr G, Leonard RCF (2003) Moderate neutropenia with adjuvant CMF confers improved survival in early breast cancer. Br J Cancer 89:1837–1842CrossRefPubMed Cameron DA, Massie C, Kerr G, Leonard RCF (2003) Moderate neutropenia with adjuvant CMF confers improved survival in early breast cancer. Br J Cancer 89:1837–1842CrossRefPubMed
6.
go back to reference Poikonen P, Saarto T, Lundin J, Joensuu H, Blomqvist C (1999) Leucocyte nadir as a marker for chemotherapy efficacy in node-positive breast cancer treated with adjuvant CMF. Br J Cancer 80:1763–1766CrossRefPubMed Poikonen P, Saarto T, Lundin J, Joensuu H, Blomqvist C (1999) Leucocyte nadir as a marker for chemotherapy efficacy in node-positive breast cancer treated with adjuvant CMF. Br J Cancer 80:1763–1766CrossRefPubMed
7.
go back to reference Wallin J, Friberg LE, Karlsson MO (2009) A tool for neutrophil guided dose adaptation in chemotherapy. Comp Meth Prog Biomed 93:283–291CrossRef Wallin J, Friberg LE, Karlsson MO (2009) A tool for neutrophil guided dose adaptation in chemotherapy. Comp Meth Prog Biomed 93:283–291CrossRef
8.
go back to reference Friberg LE, Henningsson A, Maas H, Nguyen L, Karlsson MO (2002) Model of chemotherapy-induced myelosuppression with parameter consistency across drugs. J Clin Oncol 20:4713–4721CrossRefPubMed Friberg LE, Henningsson A, Maas H, Nguyen L, Karlsson MO (2002) Model of chemotherapy-induced myelosuppression with parameter consistency across drugs. J Clin Oncol 20:4713–4721CrossRefPubMed
9.
go back to reference Troconiz IF, Garrido MJ, Segura C et al (2006) Phase I dose-finding study and a pharmacokinetic/pharmacodynamic analysis of the neutropenic response of intravenous diflomotecan in patients with advanced malignant tumours. Cancer Chemother Pharmacol 57:727–735CrossRefPubMed Troconiz IF, Garrido MJ, Segura C et al (2006) Phase I dose-finding study and a pharmacokinetic/pharmacodynamic analysis of the neutropenic response of intravenous diflomotecan in patients with advanced malignant tumours. Cancer Chemother Pharmacol 57:727–735CrossRefPubMed
10.
go back to reference Kathman SJ, Williams DH, Hodge JP, Dar M (2007) A Bayesian population PK-PD model of ispinesib-induced myelosuppression. Clin Pharmacol Ther 81:88–94CrossRefPubMed Kathman SJ, Williams DH, Hodge JP, Dar M (2007) A Bayesian population PK-PD model of ispinesib-induced myelosuppression. Clin Pharmacol Ther 81:88–94CrossRefPubMed
11.
go back to reference Leger F, Loos WJ, Bugat R et al (2004) Mechanism-based models for topotecan-induced neutropenia. Clin Pharmacol Ther 76:567–578CrossRefPubMed Leger F, Loos WJ, Bugat R et al (2004) Mechanism-based models for topotecan-induced neutropenia. Clin Pharmacol Ther 76:567–578CrossRefPubMed
12.
go back to reference Brain EGC, Rezai K, Lokiec F, Gutierrez M, Urien S (2008) Population pharmacokinetics and exploratory pharmacodynamics of ifosfamide according to continuous or short infusion schedules: an n = 1 randomized study. Br J Clin Pharmacol 65:607–610CrossRefPubMed Brain EGC, Rezai K, Lokiec F, Gutierrez M, Urien S (2008) Population pharmacokinetics and exploratory pharmacodynamics of ifosfamide according to continuous or short infusion schedules: an n = 1 randomized study. Br J Clin Pharmacol 65:607–610CrossRefPubMed
13.
go back to reference Latz JE, Rusthoven JJ, Karlsson MO, Ghosh A, Johnson RD (2006) Clinical application of a semimechanistic-physiologic population PK/PD model for neutropenia following pemetrexed therapy. Cancer Chemother Pharmacol 57:427–435CrossRefPubMed Latz JE, Rusthoven JJ, Karlsson MO, Ghosh A, Johnson RD (2006) Clinical application of a semimechanistic-physiologic population PK/PD model for neutropenia following pemetrexed therapy. Cancer Chemother Pharmacol 57:427–435CrossRefPubMed
14.
go back to reference Sandstrom M, Lindman H, Nygren P, Johansson M, Bergh J, Karlsson MO (2006) Population analysis of the pharmacokinetics and the haematological toxicity of the fluorouracil-epirubicin-cyclophosphamide regimen in breast cancer patients. Cancer Chemother Pharmacol 58:143–156CrossRefPubMed Sandstrom M, Lindman H, Nygren P, Johansson M, Bergh J, Karlsson MO (2006) Population analysis of the pharmacokinetics and the haematological toxicity of the fluorouracil-epirubicin-cyclophosphamide regimen in breast cancer patients. Cancer Chemother Pharmacol 58:143–156CrossRefPubMed
15.
go back to reference Sandstrom M, Lindman H, Nygren P, Lidbrink E, Bergh J, Karlsson MO (2005) Model describing the relationship between pharmacokinetics and hematologic toxicity of the epirubicin-docetaxel regimen in breast cancer patients. J Clin Oncol 23:413–421 Jan 20CrossRefPubMed Sandstrom M, Lindman H, Nygren P, Lidbrink E, Bergh J, Karlsson MO (2005) Model describing the relationship between pharmacokinetics and hematologic toxicity of the epirubicin-docetaxel regimen in breast cancer patients. J Clin Oncol 23:413–421 Jan 20CrossRefPubMed
16.
go back to reference Joerger M, Huitema ADR, Richel DJ et al (2007) Population pharmacokinetics and pharmacodynamics of paclitaxel and carboplatin in ovarian cancer patients: A study by the European Organization for Research and Treatment of Cancer-Pharmacology and Molecular Mechanisms Group and New Drug Development Group. Clin Cancer Res 13:6410–6418CrossRefPubMed Joerger M, Huitema ADR, Richel DJ et al (2007) Population pharmacokinetics and pharmacodynamics of paclitaxel and carboplatin in ovarian cancer patients: A study by the European Organization for Research and Treatment of Cancer-Pharmacology and Molecular Mechanisms Group and New Drug Development Group. Clin Cancer Res 13:6410–6418CrossRefPubMed
17.
go back to reference O’Shaughnessy J, Miles D, Vukelja S et al (2002) Superior survival with capecitabine plus docetaxel combination therapy in anthracycline-pretreated patients with advanced breast cancer: phase III trial results. J Clin Oncol 20:2812–2823CrossRefPubMed O’Shaughnessy J, Miles D, Vukelja S et al (2002) Superior survival with capecitabine plus docetaxel combination therapy in anthracycline-pretreated patients with advanced breast cancer: phase III trial results. J Clin Oncol 20:2812–2823CrossRefPubMed
18.
go back to reference Henningsson A, Sparreboom A, Sandstrom M et al (2003) Population pharmacokinetic modelling of unbound and total plasma concentrations of paclitaxel in cancer patients. Eur J Cancer 39:1105–1114CrossRefPubMed Henningsson A, Sparreboom A, Sandstrom M et al (2003) Population pharmacokinetic modelling of unbound and total plasma concentrations of paclitaxel in cancer patients. Eur J Cancer 39:1105–1114CrossRefPubMed
19.
go back to reference Vanwarmerdam LJC, Huinink WWB, Rodenhuis S et al (1995) Phase-I clinical and pharmacokinetic study of topotecan administered by a 24-hour continuous-infusion. J Clin Oncol 13:1768–1776 Jul Vanwarmerdam LJC, Huinink WWB, Rodenhuis S et al (1995) Phase-I clinical and pharmacokinetic study of topotecan administered by a 24-hour continuous-infusion. J Clin Oncol 13:1768–1776 Jul
20.
go back to reference Ratain MJ, Mick R, Schilsky RL, Vogelzang NJ, Berezin F (1991) Pharmacologically based dosing of etoposide—a means of safely increasing dose intensity. J Clin Oncol 9:1480–1486PubMed Ratain MJ, Mick R, Schilsky RL, Vogelzang NJ, Berezin F (1991) Pharmacologically based dosing of etoposide—a means of safely increasing dose intensity. J Clin Oncol 9:1480–1486PubMed
21.
go back to reference Ratain MJ, Schilsky RL, Choi KE et al (1989) Adaptive-control of etoposide administration—impact of interpatient pharmacodynamic variability. Clin Pharmacol Ther 45:226–233PubMed Ratain MJ, Schilsky RL, Choi KE et al (1989) Adaptive-control of etoposide administration—impact of interpatient pharmacodynamic variability. Clin Pharmacol Ther 45:226–233PubMed
22.
go back to reference Beal S, Sheiner L (2006) NONMEM users guides. NONMEM Project Group, University of California at San Francisco ed, San Francisco Beal S, Sheiner L (2006) NONMEM users guides. NONMEM Project Group, University of California at San Francisco ed, San Francisco
23.
go back to reference Jonsson EN, Karlsson MO (1999) Xpose: an Splus based population pharmacokinetic/pharmacodynamic model building aid for NONMEM. Comput Methods Programs Biomed 58:51–64CrossRefPubMed Jonsson EN, Karlsson MO (1999) Xpose: an Splus based population pharmacokinetic/pharmacodynamic model building aid for NONMEM. Comput Methods Programs Biomed 58:51–64CrossRefPubMed
24.
go back to reference Bruno R, Vivier N, Vergniol JC, DePhillips SL, Montay G, Sheiner LB (1996) A population pharmacokinetic model for docetaxel (Taxotere(R)): model building and validation. J Pharmacokinet Biopharm 24:153–172CrossRefPubMed Bruno R, Vivier N, Vergniol JC, DePhillips SL, Montay G, Sheiner LB (1996) A population pharmacokinetic model for docetaxel (Taxotere(R)): model building and validation. J Pharmacokinet Biopharm 24:153–172CrossRefPubMed
25.
go back to reference Leger F, Loos WJ, Fourcade J et al (2004) Factors affecting pharmacokinetic variability of oral topotecan: a population analysis. Br J Cancer 90:343–347CrossRefPubMed Leger F, Loos WJ, Fourcade J et al (2004) Factors affecting pharmacokinetic variability of oral topotecan: a population analysis. Br J Cancer 90:343–347CrossRefPubMed
26.
go back to reference Toffoli G, Corona G, Sorio R et al (2001) Population pharmacokinetics and pharmacodynamics of oral etoposide. Br J Clin Pharmacol 52:511–519CrossRefPubMed Toffoli G, Corona G, Sorio R et al (2001) Population pharmacokinetics and pharmacodynamics of oral etoposide. Br J Clin Pharmacol 52:511–519CrossRefPubMed
27.
go back to reference Cartwright GE, Athens JW, Wintrobe MM (1964) The kinetics of granulopoiesis in normal man. Blood 24:780–803PubMed Cartwright GE, Athens JW, Wintrobe MM (1964) The kinetics of granulopoiesis in normal man. Blood 24:780–803PubMed
28.
go back to reference Karlsson MO, Port RE, Ratain MJ, Sheiner LB (1994) A population-model for the leukopenic effect of etoposide. Clin Pharmacol Ther 55:152 152 Karlsson MO, Port RE, Ratain MJ, Sheiner LB (1994) A population-model for the leukopenic effect of etoposide. Clin Pharmacol Ther 55:152 152
29.
go back to reference Friberg LE, Brindley CJ, Karlsson MO, Devlin AJ (2000) Models of schedule dependent haematological toxicity of 2′-deoxy-2′-methylidenecytidine (DMDC). Eur J Clin Pharmacol 56:567–574CrossRefPubMed Friberg LE, Brindley CJ, Karlsson MO, Devlin AJ (2000) Models of schedule dependent haematological toxicity of 2′-deoxy-2′-methylidenecytidine (DMDC). Eur J Clin Pharmacol 56:567–574CrossRefPubMed
30.
go back to reference Gisleskog PO, Karlsson MO, Beal SL (2002) Use of prior information to stabilize a population data analysis. J Pharmacokinet Pharmacodyn 29:473–505CrossRefPubMed Gisleskog PO, Karlsson MO, Beal SL (2002) Use of prior information to stabilize a population data analysis. J Pharmacokinet Pharmacodyn 29:473–505CrossRefPubMed
31.
go back to reference Karlsson MO, Sheiner LB (1993) The importance of modeling interoccasion variability in population pharmacokinetic analyses. J Pharmacokinet Biopharm 21:735–750CrossRefPubMed Karlsson MO, Sheiner LB (1993) The importance of modeling interoccasion variability in population pharmacokinetic analyses. J Pharmacokinet Biopharm 21:735–750CrossRefPubMed
32.
go back to reference Gibiansky L (2007) Precision of parameter estimates: covariance step ($COV) versus bootstrap procedure (Abstr 1106) [wwwpage-meetingorg/?abstract=1106], p 16 Gibiansky L (2007) Precision of parameter estimates: covariance step ($COV) versus bootstrap procedure (Abstr 1106) [wwwpage-meetingorg/?abstract=1106], p 16
33.
go back to reference Karlsson MO, Holford NHG (2008) A tutorial on visual predictive checks (Abstr 1434) [wwwpage-meetingorg/?abstract=1434], p 17 Karlsson MO, Holford NHG (2008) A tutorial on visual predictive checks (Abstr 1434) [wwwpage-meetingorg/?abstract=1434], p 17
34.
go back to reference Kloft C, Wallin J, Henningsson A, Chatelut E, Karlsson MO (2006) Population pharmacokinetic-pharmacodynamic model for neutropenia with patient subgroup identification: Comparison across anticancer drugs. Clin Cancer Res 12:5481–5490CrossRefPubMed Kloft C, Wallin J, Henningsson A, Chatelut E, Karlsson MO (2006) Population pharmacokinetic-pharmacodynamic model for neutropenia with patient subgroup identification: Comparison across anticancer drugs. Clin Cancer Res 12:5481–5490CrossRefPubMed
35.
go back to reference Jelliffe RW, Schumitzky A, Bayard D et al (1998) Model-based, goal-oriented, individualised drug therapy—linkage of population modelling new ‘multiple model’ dosage design, Bayesian feedback and individualised target goals. Clin Pharmacokinet 34:57–77CrossRefPubMed Jelliffe RW, Schumitzky A, Bayard D et al (1998) Model-based, goal-oriented, individualised drug therapy—linkage of population modelling new ‘multiple model’ dosage design, Bayesian feedback and individualised target goals. Clin Pharmacokinet 34:57–77CrossRefPubMed
36.
go back to reference de Jonge ME, Huitema ADR, Schellens JHM, Rodenhuis S, Beijnen JH (2005) Individualised cancer chemotherapy: Strategies and performance of prospective studies on therapeutic drug monitoring with dose adaptation—a review. Clin Pharmacokinet 44:147–173CrossRefPubMed de Jonge ME, Huitema ADR, Schellens JHM, Rodenhuis S, Beijnen JH (2005) Individualised cancer chemotherapy: Strategies and performance of prospective studies on therapeutic drug monitoring with dose adaptation—a review. Clin Pharmacokinet 44:147–173CrossRefPubMed
37.
go back to reference Hon YY, Evans WE (1998) Making TDM work to optimize cancer chemotherapy: a multidisciplinary team approach. Clin Chem 44:388–400PubMed Hon YY, Evans WE (1998) Making TDM work to optimize cancer chemotherapy: a multidisciplinary team approach. Clin Chem 44:388–400PubMed
Metadata
Title
Limited inter-occasion variability in relation to inter-individual variability in chemotherapy-induced myelosuppression
Authors
Emma K. Hansson
Johan E. Wallin
Henrik Lindman
Marie Sandström
Mats O. Karlsson
Lena E. Friberg
Publication date
01-04-2010
Publisher
Springer-Verlag
Published in
Cancer Chemotherapy and Pharmacology / Issue 5/2010
Print ISSN: 0344-5704
Electronic ISSN: 1432-0843
DOI
https://doi.org/10.1007/s00280-009-1089-3

Other articles of this Issue 5/2010

Cancer Chemotherapy and Pharmacology 5/2010 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine