Skip to main content
Top
Published in: Cancer Chemotherapy and Pharmacology 6/2014

01-12-2014 | Original Article

Pharmacokinetics of single-agent axitinib across multiple solid tumor types

Authors: Michael A. Tortorici, Ezra E. W. Cohen, Yazdi K. Pithavala, May Garrett, Ana Ruiz-Garcia, Sinil Kim, John P. Fruehauf

Published in: Cancer Chemotherapy and Pharmacology | Issue 6/2014

Login to get access

Abstract

Purpose

Axitinib, a potent and selective inhibitor of vascular endothelial growth factor receptors, showed antitumor activity as a single agent against several solid tumor types in Phase II and III trials. This study was conducted to evaluate axitinib pharmacokinetics across a variety of solid tumors.

Methods

The current study analyzed the pharmacokinetics of axitinib in 110 patients with non-small cell lung cancer (NSCLC), thyroid cancer, or melanoma from three Phase II trials plus 127 healthy volunteers, using nonlinear mixed-effects modeling. Boxplots of maximum observed plasma concentration (C max) and area under the plasma concentration–time curve (AUC) of data from these tumor populations was compared to C max and AUC from the final population pharmacokinetic model developed for metastatic renal cell carcinoma (mRCC) to compare axitinib pharmacokinetics across different tumor types.

Results

Axitinib disposition based on data from 237 subjects was best described using a two-compartment model with first-order absorption and lag time. Population estimates for systemic clearance, central volume of distribution, absorption rate constant, absolute bioavailability, and lag time were 20.1 L/h, 56.2 L, 1.26/h−1, 0.663, and 0.448 h, respectively. Statistically significant covariates included gender on clearance, and body weight on central volume of distribution. However, predicted changes due to gender and body weight were found not clinically meaningful. The final analysis indicated that the pharmacokinetic model for mRCC was able to successfully describe axitinib pharmacokinetics in patients with NSCLC, thyroid cancer, and melanoma.

Conclusion

The pharmacokinetics of axitinib appears to be similar across a variety of tumor types.
Appendix
Available only for authorised users
Literature
1.
go back to reference Hu-Lowe DD, Zou HY, Grazzini ML, Hallin ME, Wickman GR, Amundson K, Chen JH, Rewolinski DA, Yamazaki S, Wu EY, McTigue MA, Murray BW, Kania RS, O’Connor P, Shalinsky DR, Bender SL (2008) Nonclinical antiangiogenesis and antitumor activities of axitinib (AG-013736), an oral, potent, and selective inhibitor of vascular endothelial growth factor receptor tyrosine kinases 1, 2, 3. Clin Cancer Res 14:7272–7283PubMedCrossRef Hu-Lowe DD, Zou HY, Grazzini ML, Hallin ME, Wickman GR, Amundson K, Chen JH, Rewolinski DA, Yamazaki S, Wu EY, McTigue MA, Murray BW, Kania RS, O’Connor P, Shalinsky DR, Bender SL (2008) Nonclinical antiangiogenesis and antitumor activities of axitinib (AG-013736), an oral, potent, and selective inhibitor of vascular endothelial growth factor receptor tyrosine kinases 1, 2, 3. Clin Cancer Res 14:7272–7283PubMedCrossRef
2.
go back to reference Choueiri TK (2008) Axitinib, a novel anti-angiogenic drug with promising activity in various solid tumors. Curr Opin Investig Drugs 9:658–671PubMed Choueiri TK (2008) Axitinib, a novel anti-angiogenic drug with promising activity in various solid tumors. Curr Opin Investig Drugs 9:658–671PubMed
3.
go back to reference Kelly RJ, Rixe O (2009) Axitinib–a selective inhibitor of the vascular endothelial growth factor (VEGF) receptor. Target Oncol 4:297–305PubMedCrossRef Kelly RJ, Rixe O (2009) Axitinib–a selective inhibitor of the vascular endothelial growth factor (VEGF) receptor. Target Oncol 4:297–305PubMedCrossRef
4.
go back to reference Rixe O, Bukowski RM, Michaelson MD, Wilding G, Hudes GR, Bolte O, Motzer RJ, Bycott P, Liau KF, Freddo J, Trask PC, Kim S, Rini BI (2007) Axitinib treatment in patients with cytokine-refractory metastatic renal-cell cancer: a phase II study. Lancet Oncol 8:975–984PubMedCrossRef Rixe O, Bukowski RM, Michaelson MD, Wilding G, Hudes GR, Bolte O, Motzer RJ, Bycott P, Liau KF, Freddo J, Trask PC, Kim S, Rini BI (2007) Axitinib treatment in patients with cytokine-refractory metastatic renal-cell cancer: a phase II study. Lancet Oncol 8:975–984PubMedCrossRef
5.
go back to reference Rini BI, Wilding G, Hudes G, Stadler WM, Kim S, Tarazi J, Rosbrook B, Trask PC, Wood L, Dutcher JP (2009) Phase II study of axitinib in sorafenib-refractory metastatic renal cell carcinoma. J Clin Oncol 27:4462–4468PubMedCrossRef Rini BI, Wilding G, Hudes G, Stadler WM, Kim S, Tarazi J, Rosbrook B, Trask PC, Wood L, Dutcher JP (2009) Phase II study of axitinib in sorafenib-refractory metastatic renal cell carcinoma. J Clin Oncol 27:4462–4468PubMedCrossRef
6.
go back to reference Cohen EE, Rosen LS, Vokes EE, Kies MS, Forastiere AA, Worden FP, Kane MA, Sherman E, Kim S, Bycott P, Tortorici M, Shalinsky DR, Liau KF, Cohen RB (2008) Axitinib is an active treatment for all histologic subtypes of advanced thyroid cancer: results from a phase II study. J Clin Oncol 26:4708–4713PubMedCrossRef Cohen EE, Rosen LS, Vokes EE, Kies MS, Forastiere AA, Worden FP, Kane MA, Sherman E, Kim S, Bycott P, Tortorici M, Shalinsky DR, Liau KF, Cohen RB (2008) Axitinib is an active treatment for all histologic subtypes of advanced thyroid cancer: results from a phase II study. J Clin Oncol 26:4708–4713PubMedCrossRef
7.
go back to reference Schiller JH, Larson T, Ou SH, Limentani S, Sandler A, Vokes E, Kim S, Liau K, Bycott P, Olszanski AJ, von Pawel J (2009) Efficacy and safety of axitinib in patients with advanced non-small-cell lung cancer: results from a phase II study. J Clin Oncol 27:3836–3841PubMedCrossRef Schiller JH, Larson T, Ou SH, Limentani S, Sandler A, Vokes E, Kim S, Liau K, Bycott P, Olszanski AJ, von Pawel J (2009) Efficacy and safety of axitinib in patients with advanced non-small-cell lung cancer: results from a phase II study. J Clin Oncol 27:3836–3841PubMedCrossRef
8.
go back to reference Fruehauf JP, Lutzky J, McDermott DF, Brown CK, Meric JB, Rosbrook B, Shalinsky DR, Liau KF, Niethammer AG, Kim S, Rixe O (2011) Multicenter, phase II study of axitinib, a selective second-generation inhibitor of vascular endothelial growth factor receptors 1, 2, 3, in patients with metastatic melanoma. Clin Cancer Res 17:7462–7469PubMedCrossRef Fruehauf JP, Lutzky J, McDermott DF, Brown CK, Meric JB, Rosbrook B, Shalinsky DR, Liau KF, Niethammer AG, Kim S, Rixe O (2011) Multicenter, phase II study of axitinib, a selective second-generation inhibitor of vascular endothelial growth factor receptors 1, 2, 3, in patients with metastatic melanoma. Clin Cancer Res 17:7462–7469PubMedCrossRef
9.
go back to reference Rini BI, Escudier B, Tomczak P, Kaprin A, Szczylik C, Hutson TE, Michaelson MD, Gorbunova VA, Gore ME, Rusakov IG, Negrier S, Ou Y-C, Castellano D, Lim HY, Uemura H, Tarazi J, Cella D, Chen C, Rosbrook B, Kim S, Motzer RJ (2011) Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma (AXIS): a randomised phase 3 trial. Lancet 378:1931–1939PubMedCrossRef Rini BI, Escudier B, Tomczak P, Kaprin A, Szczylik C, Hutson TE, Michaelson MD, Gorbunova VA, Gore ME, Rusakov IG, Negrier S, Ou Y-C, Castellano D, Lim HY, Uemura H, Tarazi J, Cella D, Chen C, Rosbrook B, Kim S, Motzer RJ (2011) Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma (AXIS): a randomised phase 3 trial. Lancet 378:1931–1939PubMedCrossRef
10.
go back to reference Boxenbaum H, Battle M (1995) Effective half-life in clinical pharmacology. J Clin Pharmacol 35:763–766PubMedCrossRef Boxenbaum H, Battle M (1995) Effective half-life in clinical pharmacology. J Clin Pharmacol 35:763–766PubMedCrossRef
11.
go back to reference Rugo HS, Herbst RS, Liu G, Park JW, Kies MS, Steinfeldt HM, Pithavala YK, Reich SD, Freddo JL, Wilding G (2005) Phase I trial of the oral antiangiogenesis agent AG-013736 in patients with advanced solid tumors: pharmacokinetic and clinical results. J Clin Oncol 23:5474–5483PubMedCrossRef Rugo HS, Herbst RS, Liu G, Park JW, Kies MS, Steinfeldt HM, Pithavala YK, Reich SD, Freddo JL, Wilding G (2005) Phase I trial of the oral antiangiogenesis agent AG-013736 in patients with advanced solid tumors: pharmacokinetic and clinical results. J Clin Oncol 23:5474–5483PubMedCrossRef
13.
go back to reference Pithavala YK, Tortorici M, Toh M, Garrett M, Hee B, Kuruganti U, Ni G, Klamerus KJ (2010) Effect of rifampin on the pharmacokinetics of Axitinib (AG-013736) in Japanese and Caucasian healthy volunteers. Cancer Chemother Pharmacol 65:563–570PubMedCentralPubMedCrossRef Pithavala YK, Tortorici M, Toh M, Garrett M, Hee B, Kuruganti U, Ni G, Klamerus KJ (2010) Effect of rifampin on the pharmacokinetics of Axitinib (AG-013736) in Japanese and Caucasian healthy volunteers. Cancer Chemother Pharmacol 65:563–570PubMedCentralPubMedCrossRef
14.
go back to reference Chen Y, Jiang J, Zhang J, Tortorici MA, Pithavala YK, Lu L, Ni G, Hu P (2011) A Phase I study to evaluate the pharmacokinetics of axitinib (AG-13736) in healthy Chinese volunteers. Int J Clin Pharmacol Ther 49:679–687PubMedCrossRef Chen Y, Jiang J, Zhang J, Tortorici MA, Pithavala YK, Lu L, Ni G, Hu P (2011) A Phase I study to evaluate the pharmacokinetics of axitinib (AG-13736) in healthy Chinese volunteers. Int J Clin Pharmacol Ther 49:679–687PubMedCrossRef
15.
go back to reference Pithavala YK, Tong W, Mount J, Rahavendran SV, Garrett M, Hee B, Selaru P, Sarapa N, Klamerus KJ (2012) Effect of ketoconazole on the pharmacokinetics of axitinib in healthy volunteers. Invest New Drugs 30:273–281PubMedCentralPubMedCrossRef Pithavala YK, Tong W, Mount J, Rahavendran SV, Garrett M, Hee B, Selaru P, Sarapa N, Klamerus KJ (2012) Effect of ketoconazole on the pharmacokinetics of axitinib in healthy volunteers. Invest New Drugs 30:273–281PubMedCentralPubMedCrossRef
16.
go back to reference Pithavala YK, Chen Y, Toh M, Selaru P, LaBadie RR, Garrett M, Hee B, Mount J, Ni G, Klamerus KJ, Tortorici MA (2012) Evaluation of the effect of food on the pharmacokinetics of axitinib in healthy volunteers. Cancer Chemother Pharmacol 70:103–112PubMedCrossRef Pithavala YK, Chen Y, Toh M, Selaru P, LaBadie RR, Garrett M, Hee B, Mount J, Ni G, Klamerus KJ, Tortorici MA (2012) Evaluation of the effect of food on the pharmacokinetics of axitinib in healthy volunteers. Cancer Chemother Pharmacol 70:103–112PubMedCrossRef
17.
go back to reference Larson RA, Druker BJ, Guilhot F, O’Brien SG, Riviere GJ, Krahnke T, Gathmann I, Wang Y (2008) Imatinib pharmacokinetics and its correlation with response and safety in chronic-phase chronic myeloid leukemia: a subanalysis of the IRIS study. Blood 111:4022–4028PubMedCrossRef Larson RA, Druker BJ, Guilhot F, O’Brien SG, Riviere GJ, Krahnke T, Gathmann I, Wang Y (2008) Imatinib pharmacokinetics and its correlation with response and safety in chronic-phase chronic myeloid leukemia: a subanalysis of the IRIS study. Blood 111:4022–4028PubMedCrossRef
18.
go back to reference Houk BE, Bello CL, Poland B, Rosen LS, Demetri GD, Motzer RJ (2010) Relationship between exposure to sunitinib and efficacy and tolerability endpoints in patients with cancer: results of a pharmacokinetic/pharmacodynamic meta-analysis. Cancer Chemother Pharmacol 66:357–371PubMedCrossRef Houk BE, Bello CL, Poland B, Rosen LS, Demetri GD, Motzer RJ (2010) Relationship between exposure to sunitinib and efficacy and tolerability endpoints in patients with cancer: results of a pharmacokinetic/pharmacodynamic meta-analysis. Cancer Chemother Pharmacol 66:357–371PubMedCrossRef
19.
go back to reference Garrett M, Poland B, Brennan M, Hee B, Pithavala YK, Amantea MA (2014) Population pharmacokinetic analysis of axitinib in healthy volunteers. Br J Clin Pharmacol 77:480–492PubMedCrossRef Garrett M, Poland B, Brennan M, Hee B, Pithavala YK, Amantea MA (2014) Population pharmacokinetic analysis of axitinib in healthy volunteers. Br J Clin Pharmacol 77:480–492PubMedCrossRef
20.
go back to reference Rini BI, Garrett M, Poland B, Dutcher JP, Rixe O, Wilding G, Stadler WM, Pithavala YK, Kim S, Tarazi J, Motzer RJ (2013) Axitinib in metastatic renal cell carcinoma: results of a pharmacokinetic and pharmacodynamic analysis. J Clin Pharmacol 53:491–504PubMedCentralPubMedCrossRef Rini BI, Garrett M, Poland B, Dutcher JP, Rixe O, Wilding G, Stadler WM, Pithavala YK, Kim S, Tarazi J, Motzer RJ (2013) Axitinib in metastatic renal cell carcinoma: results of a pharmacokinetic and pharmacodynamic analysis. J Clin Pharmacol 53:491–504PubMedCentralPubMedCrossRef
21.
go back to reference Lindbom L, Pihlgren P, Jonsson EN (2005) PsN-Toolkit–a collection of computer intensive statistical methods for non-linear mixed effect modeling using NONMEM. Comput Methods Programs Biomed 79:241–257PubMedCrossRef Lindbom L, Pihlgren P, Jonsson EN (2005) PsN-Toolkit–a collection of computer intensive statistical methods for non-linear mixed effect modeling using NONMEM. Comput Methods Programs Biomed 79:241–257PubMedCrossRef
22.
go back to reference Hooker AC, Staatz CE, Karlsson MO (2007) Conditional weighted residuals (CWRES): a model diagnostic for the FOCE method. Pharm Res 24:2187–2197PubMedCrossRef Hooker AC, Staatz CE, Karlsson MO (2007) Conditional weighted residuals (CWRES): a model diagnostic for the FOCE method. Pharm Res 24:2187–2197PubMedCrossRef
23.
24.
go back to reference Garrett M, Amantea MA, Pithavala YK, Ruiz A (2010) Evaluation of the impact of omitting drug concentration data below the lower limit of quantification (LLOQ) on the pharmacokinetics of axitinib (AG-013736), an anti-angiogenic agent. Clin Pharmacol Thera 87(suppl 1; abstr PIII-33):S78–S79 Garrett M, Amantea MA, Pithavala YK, Ruiz A (2010) Evaluation of the impact of omitting drug concentration data below the lower limit of quantification (LLOQ) on the pharmacokinetics of axitinib (AG-013736), an anti-angiogenic agent. Clin Pharmacol Thera 87(suppl 1; abstr PIII-33):S78–S79
25.
26.
go back to reference Houk BE, Bello CL, Kang D, Amantea M (2009) A population pharmacokinetic meta-analysis of sunitinib malate (SU11248) and its primary metabolite (SU12662) in healthy volunteers and oncology patients. Clin Cancer Res 15:2497–2506PubMedCrossRef Houk BE, Bello CL, Kang D, Amantea M (2009) A population pharmacokinetic meta-analysis of sunitinib malate (SU11248) and its primary metabolite (SU12662) in healthy volunteers and oncology patients. Clin Cancer Res 15:2497–2506PubMedCrossRef
27.
go back to reference Jain L, Woo S, Gardner ER, Dahut WL, Kohn EC, Kummar S, Mould DR, Giaccone G, Yarchoan R, Venitz J, Figg WD (2011) Population pharmacokinetic analysis of sorafenib in patients with solid tumours. Br J Clin Pharmacol 72:294–305PubMedCentralPubMedCrossRef Jain L, Woo S, Gardner ER, Dahut WL, Kohn EC, Kummar S, Mould DR, Giaccone G, Yarchoan R, Venitz J, Figg WD (2011) Population pharmacokinetic analysis of sorafenib in patients with solid tumours. Br J Clin Pharmacol 72:294–305PubMedCentralPubMedCrossRef
28.
go back to reference Tortorici MA, Toh M, Rahavendran SV, LaBadie RR, Alvey CW, Marbury T, Fuentes E, Green M, Ni G, Hee B, Pithavala YK (2011) Influence of mild and moderate hepatic impairment on axitinib pharmacokinetics. Invest New Drugs 29:1370–1380PubMedCentralPubMedCrossRef Tortorici MA, Toh M, Rahavendran SV, LaBadie RR, Alvey CW, Marbury T, Fuentes E, Green M, Ni G, Hee B, Pithavala YK (2011) Influence of mild and moderate hepatic impairment on axitinib pharmacokinetics. Invest New Drugs 29:1370–1380PubMedCentralPubMedCrossRef
29.
go back to reference Smith BJ, Pithavala Y, Bu H, Kang P, Hee B, Deese AJ, Pool WF, Klamerus KJ, Wu EY, Dalvie DK (2014) Pharmacokinetics, metabolism, and excretion of [14C]axitinib, a vascular endothelial growth factor receptor tyrosine kinase inhibitor, in humans. Drug Metab Dispos 42:918–931PubMedCrossRef Smith BJ, Pithavala Y, Bu H, Kang P, Hee B, Deese AJ, Pool WF, Klamerus KJ, Wu EY, Dalvie DK (2014) Pharmacokinetics, metabolism, and excretion of [14C]axitinib, a vascular endothelial growth factor receptor tyrosine kinase inhibitor, in humans. Drug Metab Dispos 42:918–931PubMedCrossRef
30.
go back to reference Lamba JK, Lin YS, Schuetz EG, Thummel KE (2002) Genetic contribution to variable human CYP3A-mediated metabolism. Adv Drug Deliv Rev 54:1271–1294PubMedCrossRef Lamba JK, Lin YS, Schuetz EG, Thummel KE (2002) Genetic contribution to variable human CYP3A-mediated metabolism. Adv Drug Deliv Rev 54:1271–1294PubMedCrossRef
31.
go back to reference Zhao W, Elie V, Roussey G, Brochard K, Niaudet P, Leroy V, Loirat C, Cochat P, Cloarec S, Andre JL, Garaix F, Bensman A, Fakhoury M, Jacqz-Aigrain E (2009) Population pharmacokinetics and pharmacogenetics of tacrolimus in de novo pediatric kidney transplant recipients. Clin Pharmacol Ther 86:609–618PubMedCrossRef Zhao W, Elie V, Roussey G, Brochard K, Niaudet P, Leroy V, Loirat C, Cochat P, Cloarec S, Andre JL, Garaix F, Bensman A, Fakhoury M, Jacqz-Aigrain E (2009) Population pharmacokinetics and pharmacogenetics of tacrolimus in de novo pediatric kidney transplant recipients. Clin Pharmacol Ther 86:609–618PubMedCrossRef
32.
go back to reference Han JY, Lim HS, Shin ES, Yoo YK, Park YH, Lee JE, Jang IJ, Lee DH, Lee JS (2006) Comprehensive analysis of UGT1A polymorphisms predictive for pharmacokinetics and treatment outcome in patients with non-small-cell lung cancer treated with irinotecan and cisplatin. J Clin Oncol 24:2237–2244PubMedCrossRef Han JY, Lim HS, Shin ES, Yoo YK, Park YH, Lee JE, Jang IJ, Lee DH, Lee JS (2006) Comprehensive analysis of UGT1A polymorphisms predictive for pharmacokinetics and treatment outcome in patients with non-small-cell lung cancer treated with irinotecan and cisplatin. J Clin Oncol 24:2237–2244PubMedCrossRef
33.
go back to reference Brennan M, Williams JA, Chen Y, Tortorici M, Pithavala Y, Liu YC (2012) Meta-analysis of contribution of genetic polymorphisms in drug-metabolizing enzymes or transporters to axitinib pharmacokinetics. Eur J Clin Pharmacol 68:645–655PubMedCrossRef Brennan M, Williams JA, Chen Y, Tortorici M, Pithavala Y, Liu YC (2012) Meta-analysis of contribution of genetic polymorphisms in drug-metabolizing enzymes or transporters to axitinib pharmacokinetics. Eur J Clin Pharmacol 68:645–655PubMedCrossRef
Metadata
Title
Pharmacokinetics of single-agent axitinib across multiple solid tumor types
Authors
Michael A. Tortorici
Ezra E. W. Cohen
Yazdi K. Pithavala
May Garrett
Ana Ruiz-Garcia
Sinil Kim
John P. Fruehauf
Publication date
01-12-2014
Publisher
Springer Berlin Heidelberg
Published in
Cancer Chemotherapy and Pharmacology / Issue 6/2014
Print ISSN: 0344-5704
Electronic ISSN: 1432-0843
DOI
https://doi.org/10.1007/s00280-014-2606-6

Other articles of this Issue 6/2014

Cancer Chemotherapy and Pharmacology 6/2014 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