Dose modifications in Asian cancer patients with hepatic dysfunction receiving weekly docetaxel: A prospective pharmacokinetic and safety study

Nicholas Li-Xun Syn, Lingzhi Wang, Andrea Li-Ann Wong, Mu-Yar Soe, Benjamin Chuah, Daniel Chan, Sing-Huang Tan, Ross Andrew Soo, Soo-Chin Lee, Boon-Cher Goh, Wei-Peng Yong, Nicholas Li-Xun Syn, Lingzhi Wang, Andrea Li-Ann Wong, Mu-Yar Soe, Benjamin Chuah, Daniel Chan, Sing-Huang Tan, Ross Andrew Soo, Soo-Chin Lee, Boon-Cher Goh, Wei-Peng Yong

Abstract

Hepatic dysfunction may modify the safety profile and pharmacokinetics of docetaxel in cancer patients, but no validated guideline exists to guide dose modification necessitated by this uncommon comorbidity. We carried out the first prospective study of a personalized dosage regimen for cancer patients with liver dysfunction treated with docetaxel. Weekly dosages were stratified by hepatic dysfunction classification as such: Category 1, normal; Category 2, mild--alkaline phosphatase, aspartate aminotransferase, and/or alanine aminotransferase ≤5× upper limit of normal (ULN), and total bilirubin within normal range; and Category 3, moderate--any alkaline phosphatase, and aspartate aminotransferase or alanine aminotransferase ≤5-10× ULN, and/or total bilirubin ≤1-1.5× ULN. Category 1, 2 and 3 patients received starting dosages of 40, 30, and 20 mg/m(2) docetaxel, respectively. Pharmacokinetics were evaluated on day 1 and 8 of the first treatment cycle, and entered into a multilevel model to delineate interindividual and interoccasion variability. Adverse event evaluation was carried out weekly for two treatment cycles. We found that docetaxel clearance was significantly different between patient categories (P < 0.001). Median clearance was 22.8, 16.4, and 11.3 L/h/m(2) in Categories 1, 2, and 3, respectively, representing 28% and 50% reduced clearance in mild and moderate liver dysfunction patients, respectively. However, docetaxel exposure (area under the concentration-time curve) and docetaxel-induced neutropenia (nadir and the maximum percentage decrease in neutrophil count) were not significantly different between categories. Median area under the concentration-time curve was 1.74, 1.83, and 1.77 mg·h/L in Categories 1, 2, and 3, respectively. The most common Grade 3/4 toxicity was neutropenia (30.0%). An unplanned comparison with the Child-Pugh and National Cancer Institute Organ Dysfunction Working Group grouping systems suggests that the proposed classification system appears to more effectively discriminate patients by docetaxel clearance and dose requirements. (ClinicalTrials.gov registration no. NCT00703378).

Keywords: Antineoplastic combined chemotherapy protocols; biomarkers; neutropenia; pharmacokinetics; taxoids.

© 2015 The Authors. Cancer Science published by John Wiley & Sons Australia, Ltd on behalf of Japanese Cancer Association.

Figures

Figure 1
Figure 1
Non‐compartmental pharmacokinetics of docetaxel in cancer patients categorized according to hepatic dysfunction: Category 1, normal; Category 2, mild – alkaline phosphatase, aspartate aminotransferase, and/or alanine aminotransferase ≤5× upper limit of normal (ULN), and total bilirubin within normal range; and Category 3, moderate – any alkaline phosphatase, and aspartate aminotransferase or alanine aminotransferase ≤5–10× ULN, and/or total bilirubin ≤1–1.5× ULN. (a) Mean docetaxel concentrations. (b) Docetaxel clearance. (c) Docetaxel exposure. (d) Changes in docetaxel clearance between baseline and repeat pharmacokinetic measurement. AUC, area under the concentration–time curve.
Figure 2
Figure 2
Pharmacodynamics of docetaxel‐induced neutropenia. (a) Mean ± SD of nadir absolute neutrophil count (ANC). (b) Mean ± SD of the maximum decrease between ANC at baseline compared to nadir. Three patients from Category 1 were switched to the 3‐weekly cycle when the repeat pharmacokinetic assessment was carried out, and were thus excluded from pharmacodynamic analysis. Hepatic dysfunction categories were defined as: Category 1, normal; Category 2, mild – alkaline phosphatase, aspartate aminotransferase, and/or alanine aminotransferase ≤5× upper limit of normal (ULN), and total bilirubin within normal range; and Category 3, moderate – any alkaline phosphatase, and aspartate aminotransferase or alanine aminotransferase ≤5–10× ULN, and/or total bilirubin ≤1–1.5× ULN. Std, standard.

References

    1. Edwards BK, Noone AM, Mariotto AB et al Annual Report to the Nation on the status of cancer, 1975–2010, featuring prevalence of comorbidity and impact on survival among persons with lung, colorectal, breast, or prostate cancer. Cancer 2014; 120: 1290–314.
    1. Field KM, Michael M. Part II: Liver function in oncology: towards safer chemotherapy use. Lancet Oncol 2008. Dec; 9: 1181–90.
    1. Tran A, Jullien V, Alexandre J et al Pharmacokinetics and toxicity of docetaxel: role of CYP3A, MDR1, and GST polymorphisms. Clin Pharmacol Ther 2006. Jun; 79: 570–80.
    1. Field KM, Dow C, Michael M. Part I: Liver function in oncology: biochemistry and beyond. Lancet Oncol 2008. Nov; 9: 1092–101.
    1. Clarke SJ, Rivory LP. Clinical pharmacokinetics of docetaxel. Clin Pharmacokinet 1999. Feb; 36 (2) : 99–114.
    1. Minami H, Kawada K, Sasaki Y et al Population pharmacokinetics of docetaxel in patients with hepatic dysfunction treated in an oncology practice. Cancer Sci 2009; 100 (1) : 144–9.
    1. Bruno R, Hille D, Riva A et al Population pharmacokinetics/pharmacodynamics of docetaxel in phase II studies in patients with cancer. J Clin Oncol 1998. Jan; 16 (1) : 187–96.
    1. Pugh RN, Murray‐Lyon IM, Dawson JL, Pietroni MC, Williams R. Transection of the oesophagus for bleeding oesophageal varices. Br J Surg 1973. Aug; 60: 646–9.
    1. Patel H, Egorin MJ, Remick SC et al Comparison of Child‐Pugh (CP) criteria and NCI organ dysfunction working group (NCI‐ODWG) criteria for hepatic dysfunction (HD): Implications for chemotherapy dosing. ASCO Meet Abstr 2004. Jul; 22 (Suppl): 6051.
    1. Millward MJ, Boyer MJ, Lehnert M et al Docetaxel and carboplatin is an active regimen in advanced non‐small‐cell lung cancer: a phase II study in Caucasian and Asian patients. Ann Oncol 2003. Mar; 14: 449–54.
    1. Kenmotsu H, Tanigawara Y. Pharmacokinetics, dynamics and toxicity of docetaxel: why the Japanese dose differs from the Western dose. Cancer Sci 2015. May; 106: 497–504.
    1. Goh BC, Lee SC, Wang LZ et al Explaining interindividual variability of docetaxel pharmacokinetics and pharmacodynamics in Asians through phenotyping and genotyping strategies. J Clin Oncol 2002; 20: 3683–90.
    1. D'Argenio D, Schumitzky A. ADAPT II User's Guide: Pharmacokinetic/pharmacodynamic systems analysis software. Biomed Simulations Resour Los Angeles; 1997.
    1. Wang LZ, Goh BC, Grigg ME, Lee SC, Khoo YM, Lee HS. A rapid and sensitive liquid chromatography/tandem mass spectrometry method for determination of docetaxel in human plasma. Rapid Commun Mass Spectrom 2003. Jan; 17: 1548–52.
    1. Minami H, Kawada K, Sasaki Y et al Pharmacokinetics and pharmacodynamics of protein‐unbound docetaxel in cancer patients. Cancer Sci 2006; 97: 235–41.
    1. Loos WJ, Baker SD, Verweij J, Boonstra JG, Sparreboom A. Clinical pharmacokinetics of unbound docetaxel: role of polysorbate 80 and serum proteins. Clin Pharmacol Ther 2003. Oct; 74: 364–71.
    1. Mauri D, Kamposioras K, Tsali L et al Overall survival benefit for weekly vs. three‐weekly taxanes regimens in advanced breast cancer: a meta‐analysis. Cancer Treat Rev 2010. Feb; 36: 69–74.
    1. Di Maio M, Perrone F, Chiodini P et al Individual patient data meta‐analysis of docetaxel administered once every 3 weeks compared with once every week second‐line treatment of advanced non‐small‐cell lung cancer. J Clin Oncol 2007. Apr; 25: 1377–82.

Source: PubMed

3
Suscribir