904
Views
47
CrossRef citations to date
0
Altmetric
Review

Kinase inhibitor pharmacokinetics: comprehensive summary and roadmap for addressing inter-individual variability in exposure

, , , , , , , , & show all
Pages 31-49 | Received 23 May 2016, Accepted 23 Aug 2016, Published online: 01 Sep 2016

References

  • Drenberg CD, Baker SD, Sparreboom A. Integrating clinical pharmacology concepts in individualized therapy with tyrosine kinase inhibitors. Clin Pharmacol Ther. 2013;93:215–219.
  • Humphrey R, Brockway-Lunardi L, Bonk D, et al. Opportunities and challenges in the development of experimental drug combinations for cancer. J Nat Cancer Inst. 2011;103:1222–1226.
  • Eckstein N, Röper L, Haas B, et al. Clinical pharmacology of tyrosine kinase inhibitors becoming generic drugs: the regulatory perspective. J Exp Clin Cancer Res. 2014;33:15.
  • Arora A, Scholar EM. Role of tyrosine kinase inhibitors in cancer therapy. J Pharmacol Exp Ther. 2005;315:971–979.
  • Krause DS, Van Etten RA. Tyrosine kinases as targets for cancer therapy. N Engl J Med. 2005;353:172–187.
  • McTigue M, Murray BW, Chen JH, et al. Molecular conformations, interactions, and properties associated with drug efficiency and clinical performance among VEGFR TK inhibitors. Proc Natl Acad Sci. 2012;109:18281–18289.
  • Chen Y, Tortorici MA, Garrett M, et al. Clinical pharmacology of axitinib. Clin Pharmacokinet. 2013;52:713–725.
  • Tortorici MA, Cohen EE, Pithavala YK, et al. Pharmacokinetics of single-agent axitinib across multiple solid tumor types. Cancer Chemother Pharmacol. 2014;74:1279–1289.
  • Nguyen L, Holland J, Miles D, et al. Pharmacokinetic (PK) drug interaction studies of cabozantinib: effect of CYP3A inducer rifampin and inhibitor ketoconazole on cabozantinib plasma PK and effect of cabozantinib on CYP2C8 probe substrate rosiglitazone plasma PK. J Clin Pharmacol. 2015;55:1012–1023.
  • Nair A, Lemery SJ, Yang J, et al. FDA approval summary: lenvatinib for progressive, radio-iodine-refractory differentiated thyroid cancer. Clin Cancer Res. 2015;21:5205–5208.
  • FDA. OFEV (nintedanib) prescribing information 2014. Available from: http://www.accessdata.fda.gov/drugsatfda_docs/label/2014/205832s000lbl.pdf
  • Deng Y, Sychterz C, Suttle AB, et al. Bioavailability, metabolism and disposition of oral pazopanib in patients with advanced cancer. Xenobiotica. 2013;43:443–453.
  • Imbs D-C, Négrier S, Cassier P, et al. Pharmacokinetics of pazopanib administered in combination with bevacizumab. Cancer Chemother Pharmacol. 2014;73:1189–1196.
  • Bruix J, Tak W-Y, Gasbarrini A, et al. Regorafenib as second-line therapy for intermediate or advanced hepatocellular carcinoma: multicentre, open-label, phase II safety study. Eur J Cancer. 2013;49:3412–3419.
  • Strumberg D, Richly H, Hilger RA, et al. Phase I clinical and pharmacokinetic study of the novel Raf kinase and vascular endothelial growth factor receptor inhibitor BAY 43-9006 in patients with advanced refractory solid tumors. J Clin Oncol. 2005;23:965–972.
  • Kane RC, Farrell AT, Saber H, et al. Sorafenib for the treatment of advanced renal cell carcinoma. Clin Cancer Res. 2006;12:7271–7278.
  • Goodman VL, Rock EP, Dagher R, et al. Approval summary: sunitinib for the treatment of imatinib refractory or intolerant gastrointestinal stromal tumors and advanced renal cell carcinoma. Clin Cancer Res. 2007;13:1367–1373.
  • Faivre S, Delbaldo C, Vera K, et al. Safety, pharmacokinetic, and antitumor activity of SU11248, a novel oral multitarget tyrosine kinase inhibitor, in patients with cancer. J Clin Oncol. 2006;24:25–35.
  • Martin P, Oliver S, Kennedy S-J, et al. Pharmacokinetics of vandetanib: three phase I studies in healthy subjects. Clin Ther. 2012;34:221–237.
  • Stopfer P, Marzin K, Narjes H, et al. Afatinib pharmacokinetics and metabolism after oral administration to healthy male volunteers. Cancer Chemother Pharmacol. 2012;69:1051–1061.
  • Wind S, Schmid M, Erhardt J, et al. Pharmacokinetics of afatinib, a selective irreversible ErbB family blocker, in patients with advanced solid tumours. Clin Pharmacokinet. 2013;52:1101–1109.
  • Johnson JR, Cohen M, Sridhara R, et al. Approval summary for erlotinib for treatment of patients with locally advanced or metastatic non-small cell lung cancer after failure of at least one prior chemotherapy regimen. Clin Cancer Res. 2005;11:6414–6421.
  • Frohna P, Lu J, Eppler S, et al. Evaluation of the absolute oral bioavailability and bioequivalence of erlotinib, an inhibitor of the epidermal growth factor receptor tyrosine kinase, in a randomized, crossover study in healthy subjects. J Clin Pharmacol. 2006;46:282–290.
  • Swaisland HC, Smith RP, Laight A, et al. Single-dose clinical pharmacokinetic studies of gefitinib. Clin Pharmacokinet. 2005;44:1165–1177.
  • Ranson M, Wardell S. Gefitinib, a novel, orally administered agent for the treatment of cancer. J Clin Pharm Ther. 2004;29:95–103.
  • Burris HA 3rd, Hurwitz HI, Dees EC, et al. Phase I safety, pharmacokinetics, and clinical activity study of lapatinib (GW572016), a reversible dual inhibitor of epidermal growth factor receptor tyrosine kinases, in heavily pretreated patients with metastatic carcinomas. J Clin Oncol. 2005;23:5305–5313.
  • LoRusso PM, Jones SF, Koch KM, et al. Phase I and pharmacokinetic study of lapatinib and docetaxel in patients with advanced cancer. J Clin Oncol. 2008;26:3051–3056.
  • FDA. TAGRISSO (osimertinib) prescribing information 2015. Available from: http://www.accessdata.fda.gov/drugsatfda_docs/label/2015/208065s000lbl.pdf
  • FDA. ALECENSA (alectinib) prescribing information 2015. Available from: http://www.accessdata.fda.gov/drugsatfda_docs/label/2015/208434s000lbl.pdf
  • Lau YY, Gu W, Lin T, et al. Effects of meal type on the oral bioavailability of the ALK inhibitor, ceritinib, in healthy adult subjects. J Clin Pharmacol. 2016;56:559–566.
  • Ou SH. Crizotinib: a novel and first-in-class multitargeted tyrosine kinase inhibitor for the treatment of anaplastic lymphoma kinase rearranged non-small cell lung cancer and beyond. Drug Des Dev Ther. 2011;5:471–485.
  • Yamazaki S, Johnson TR, Smith BJ. Prediction of drug-drug interactions with crizotinib as the CYP3A substrate using a physiologically based pharmacokinetic model. Drug Metab Dispos. 2015;43:1417–1429.
  • Takahashi RH, Choo EF, Ma S, et al. Absorption, metabolism, excretion, and the contribution of intestinal metabolism to the oral disposition of [14C]cobimetinib, a MEK inhibitor, in humans. Drug Metab Dispos. 2016;44:28–39.
  • Falchook GS, Long GV, Kurzrock R, et al. Dose selection, pharmacokinetics, and pharmacodynamics of BRAF inhibitor dabrafenib (GSK2118436). Clin Cancer Res. 2014;20:4449–4458.
  • Infante JR, Fecher LA, Falchook GS, et al. Safety, pharmacokinetic, pharmacodynamic, and efficacy data for the oral MEK inhibitor trametinib: a phase 1 dose-escalation trial. Lancet Oncol. 2012;13:773–781.
  • Kim G, McKee AE, Ning YM, et al. FDA approval summary: vemurafenib for treatment of unresectable or metastatic melanoma with the BRAFV600E mutation. Clin Cancer Res. 2014;20:4994–5000.
  • Solimando DA, Waddell JA. Drug monographs: bosutinib and regorafenib. Hosp Pharm. 2013;48:190–194.
  • Brave M, Goodman V, Kaminskas E, et al. Sprycel for chronic myeloid leukemia and Philadelphia chromosome-positive acute lymphoblastic leukemia resistant to or intolerant of imatinib mesylate. Clin Cancer Res. 2008;14:352–359.
  • Peng B, Hayes M, Resta D, et al. Pharmacokinetics and pharmacodynamics of imatinib in a phase I trial with chronic myeloid leukemia patients. J Clin Oncol. 2004;22:935–942.
  • Hazarika M, Jiang X, Liu Q, et al. Tasigna for chronic and accelerated phase Philadelphia chromosome–positive chronic myelogenous leukemia resistant to or intolerant of imatinib. Clin Cancer Res. 2008;14:5325–5331.
  • Narasimhan NI, Dorer DJ, Davis J, et al. Evaluation of pharmacokinetics and safety of ponatinib in subjects with chronic hepatic impairment and matched healthy subjects. Cancer Chemother Pharmacol. 2014;74:341–348.
  • de Jong J, Sukbuntherng J, Skee D, et al. The effect of food on the pharmacokinetics of oral ibrutinib in healthy participants and patients with chronic lymphocytic leukemia. Cancer Chemother Pharmacol. 2015;75:907–916.
  • Jin F, Robeson M, Zhou H, et al. The pharmacokinetics and safety of idelalisib in subjects with moderate or severe hepatic impairment. J Clin Pharmacol. 2015;55:944–952.
  • FDA. JAKAFI (ruxolitinib) prescribing information 2014. Available from: http://www.accessdata.fda.gov/drugsatfda_docs/label/2014/202192s009lbl.pdf
  • Dowty ME, Lin J, Ryder TF, et al. The pharmacokinetics, metabolism, and clearance mechanisms of tofacitinib, a Janus kinase inhibitor, in humans. Drug Metab Dispos. 2014;42:759–773.
  • FDA. IBRANCE (palbociclib) prescribing information 2015. Available from: http://www.accessdata.fda.gov/drugsatfda_docs/label/2015/207103s000lbl.pdf
  • Burns K, Nair PC, Rowland A, et al. The nonspecific binding of tyrosine kinase inhibitors to human liver microsomes. Drug Metab Dispos. 2015;43:1934–1937.
  • Di Gion P, Kanefendt F, Lindauer A, et al. Clinical pharmacokinetics of tyrosine kinase inhibitors: focus on pyrimidines, pyridines and pyrroles. Clin Pharmacokinet. 2011;50:551–603.
  • Scheffler M, Di Gion P, Doroshyenko O, et al. Clinical pharmacokinetics of tyrosine kinase inhibitors: focus on 4-anilinoquinazolines. Clin Pharmacokinet. 2011;50:371–403.
  • Budha NR, Frymoyer A, Smelick GS, et al. Drug absorption interactions between oral targeted anticancer agents and PPIs: is pH-dependent solubility the Achilles heel of targeted therapy? Clin Pharmacol Ther. 2012;92:203–213.
  • Scripture CD, Figg WD. Drug interactions in cancer therapy. Nat Rev Cancer. 2006;6:546–558.
  • van Erp NP, Gelderblom H, Guchelaar H-J. Clinical pharmacokinetics of tyrosine kinase inhibitors. Cancer Treat Rev. 2009;35:692–706.
  • de Graan A-JM, Lancaster CS, Obaidat A, et al. Influence of polymorphic OATP1B-type carriers on the disposition of docetaxel. Clin Cancer Res. 2012;18:4433–4440.
  • Filppula AM, Neuvonen M, Laitila J, et al. Autoinhibition of CYP3A4 leads to important role of CYP2C8 in imatinib metabolism: variability in CYP2C8 activity may alter plasma concentrations and response. Drug Metab Dispos. 2013;41:50–59.
  • Filppula AM, Neuvonen PJ, Backman JT. In vitro assessment of time-dependent inhibitory effects on CYP2C8 and CYP3A activity by fourteen protein kinase inhibitors. Drug Metab Dispos. 2014;42:1202–1209.
  • van Erp NP, Gelderblom H, Karlsson MO, et al. Influence of CYP3A4 inhibition on the steady-state pharmacokinetics of imatinib. Clin Cancer Res. 2007;13:7394–7400.
  • Khan MS, Barratt DT, Somogyi AA. Impact of CYP2C8*3 polymorphism on in vitro metabolism of imatinib to N-desmethyl imatinib. Xenobiotica. 2016;46:278–287.
  • van Leeuwen RWF, Van Gelder T, Mathijssen RHJ, et al. Drug-drug interactions with tyrosine-kinase inhibitors: a clinical perspective. Lancet Oncol. 2013;15:e315–e326.
  • Reyner EL, Sevidal S, West MA, et al. In vitro characterization of axitinib interactions with human efflux and hepatic uptake transporters: implications for disposition and drug interactions. Drug Metab Dispos. 2013;41:1575–1583.
  • van Leeuwen RWF, Peric R, Hussaarts KGAM, et al. Influence of the acidic beverage cola on the absorption of erlotinib in patients with non–small-cell lung cancer. J Clin Oncol. 2016;34:1309–1314.
  • Singh BN. Effects of food on clinical pharmacokinetics. Clin Pharmacokinet. 1999;37:213–255.
  • Jain RK, Brar SS, Lesko LJ. Food and oral antineoplastics: more than meets the eye. Clin Cancer Res. 2010;16:4305–4307.
  • Ling J, Fettner S, Lum BL, et al. Effect of food on the pharmacokinetics of erlotinib, an orally active epidermal growth factor receptor tyrosine-kinase inhibitor, in healthy individuals. Anti-Cancer Drugs. 2008;19:209–216.
  • Bello CL, Sherman L, Zhou J, et al. Effect of food on the pharmacokinetics of sunitinib malate (SU11248), a multi-targeted receptor tyrosine kinase inhibitor: results from a phase I study in healthy subjects. Anti-Cancer Drugs. 2006;17:353–358.
  • Koch KM, Reddy NJ, Cohen RB, et al. Effects of food on the relative bioavailability of lapatinib in cancer patients. J Clin Oncol. 2009;27:1191–1196.
  • Polasek TM, Lin FPY, Miners JO, et al. Perpetrators of pharmacokinetic drug–drug interactions arising from altered cytochrome P450 activity: a criteria-based assessment. Br J Clin Pharmacol. 2011;71:727–736.
  • Ieiri I, Tsunemitsu S, Maeda K, et al. Mechanisms of pharmacokinetic enhancement between ritonavir and saquinavir; micro/small dosing tests using midazolam (CYP3A4), fexofenadine (p-glycoprotein), and pravastatin (OATP1B1) as probe drugs. J Clin Pharmacol. 2013;53:654–661.
  • Ulvestad M, Skottheim IB, Jakobsen GS, et al. Impact of OATP1B1, MDR1, and CYP3A4 expression in liver and intestine on interpatient pharmacokinetic variability of atorvastatin in obese subjects. Clin Pharmacol Ther. 2013;93:275–282.
  • Kim R, Wandel C, Leake B, et al. Interrelationship between substrates and inhibitors of human CYP3A and P-glycoprotein. Pharm Res. 1999;16:408–414.
  • Shao J, Markowitz JS, Bei D, et al. Enzyme- and transporter-mediated drug interactions with small molecule tyrosine kinase inhibitors. J Pharm Sci. 2014;103:3810–3833.
  • Lees J, Chan A. Polypharmacy in elderly patients with cancer: clinical implications and management. Lancet Oncol. 2011;12:1249–1257.
  • Eskens F, Verweij J. The clinical toxicity profile of vascular endothelial growth factor (VEGF) and vascular endothelial growth factor receptor (VEGFR) targeting angiogenesis inhibitors; A review. Eur J Cancer. 2006;42:3127–3139.
  • Yu H, Steeghs N, Nijenhuis C, et al. Practical guidelines for therapeutic drug monitoring of anticancer tyrosine kinase inhibitors: focus on the pharmacokinetic targets. Clin Pharmacokinet. 2014;53:305–325.
  • Gao B, Yeap S, Clements A, et al. Evidence for therapeutic drug monitoring of targeted anticancer therapies. J Clin Oncol. 2012;30:4017–4025.
  • Klümpen H-J, Samer CF, Mathijssen RHJ, et al. Moving towards dose individualization of tyrosine kinase inhibitors. Cancer Treat Rev. 2011;37:251–260.
  • Kloth JS, Klumpen HJ, Yu H, et al. Predictive value of CYP3A and ABCB1 phenotyping probes for the pharmacokinetics of sunitinib: the ClearSun study. Clin Pharmacokinet. 2014;53:261–269.
  • de Wit D, Guchelaar H-J, den Hartigh J, et al. Individualized dosing of tyrosine kinase inhibitors: are we there yet? Drug Discov Today. 2015;20:18–36.
  • Demetri GD, Wang Y, Wehrle E, et al. Imatinib plasma levels are correlated with clinical benefit in patients with unresectable/metastatic gastrointestinal stromal tumors. J Clin Oncol. 2009;27:3141–3147.
  • Houk BE, Bello CL, Poland B, et al. Relationship between exposure to sunitinib and efficacy and tolerability endpoints in patients with cancer: results of a pharmacokinetic/pharmacodynamic meta-analysis. Cancer Chemother Pharmacol. 2010;66:357–371.
  • Widmer N, Decosterd LA, Leyvraz S, et al. Relationship of imatinib-free plasma levels and target genotype with efficacy and tolerability. Br J Cancer. 2008;98:1633–1640.
  • Rini BI, Garrett M, Poland B, et al. Axitinib in metastatic renal cell carcinoma: results of a pharmacokinetic and pharmacodynamic analysis. J Clin Pharmacol. 2013;53:491–504.
  • Calvo E, Malik SN, Siu LL, et al. Assessment of erlotinib pharmacodynamics in tumors and skin of patients with head and neck cancer. Ann Oncol. 2007;18:761–767.
  • Rixe O, Dutcher J, Motzer R, et al. Diastolic blood pressure (dBP) and pharmacokinetics (PK) as predictors of axitinib efficacy in metastatic renal cell cancer (mRCC). J Clin Oncol. 2009;27:abstr 5045.
  • Tiseo M, Andreoli R, Gelsomino F, et al. Correlation between erlotinib pharmacokinetics, cutaneous toxicity and clinical outcomes in patients with advanced non-small cell lung cancer (NSCLC). Lung Cancer. 2014;83:265–271.
  • Zhao Y-Y, Li S, Zhang Y, et al. The relationship between drug exposure and clinical outcomes of non-small cell lung cancer patients treated with gefitinib. Med Oncol. 2011;28:697–702.
  • Picard S, Titier K, Etienne G, et al. Trough imatinib plasma levels are associated with both cytogenetic and molecular responses to standard-dose imatinib in chronic myeloid leukemia. Blood. 2007;109:3496–3499.
  • Larson RA, Druker BJ, Guilhot F, et al. Imatinib pharmacokinetics and its correlation with response and safety in chronic-phase chronic myeloid leukemia: a subanalysis of the IRIS study. Blood. 2008;111:4022–4028.
  • Takahashi N, Miura M, Kuroki J, et al. Multicenter phase II clinical trial of nilotinib for patients with imatinib-resistant or -intolerant chronic myeloid leukemia from the East Japan CML study group evaluation of molecular response and the efficacy and safety of nilotinib. Biomarker Res. 2014;2:6.
  • Larson RA, Yin OQ, Hochhaus A, et al. Population pharmacokinetic and exposure-response analysis of nilotinib in patients with newly diagnosed Ph+ chronic myeloid leukemia in chronic phase. Eur J Clin Pharmacol. 2012;68:723–733.
  • Hurwitz HI, Dowlati A, Saini S, et al. Phase I trial of pazopanib in patients with advanced cancer. Clin Cancer Res. 2009;15:4220–4227.
  • Suttle AB, Ball HA, Molimard M, et al. Relationships between pazopanib exposure and clinical safety and efficacy in patients with advanced renal cell carcinoma. Br J Cancer. 2014;111:1909–1916.
  • Yau T, Chen P-J, Chan P, et al. Phase I dose-finding study of pazopanib in hepatocellular carcinoma: evaluation of early efficacy, pharmacokinetics, and pharmacodynamics. Clin Cancer Res. 2011;17:6914–6923.
  • Fukudo M, Ito T, Mizuno T, et al. Exposure-toxicity relationship of sorafenib in Japanese patients with renal cell carcinoma and hepatocellular carcinoma. Clin Pharmacokinet. 2014;53:185–196.
  • Goulooze S, Martin J, Smith A. Therapeutic drug monitoring trials for gastrointestinal stromal tumors treated with sunitinib cannot be expected to detect clinical outcome improvement. ASCEPT-APSA Joint Scientific Meeting; 2015; Hobart. p. 524.
  • Lankheet NA, Kloth JS, Gadellaa-van Hooijdonk CG, et al. Pharmacokinetically guided sunitinib dosing: a feasibility study in patients with advanced solid tumours. Br J Cancer. 2014;110:2441–2449.
  • Zhang A, Fox P, Coulter S, et al. Effect of toxicity-adjusted dose (TAD) of sunitinib on intra-patient variation of trough levels: a longitudinal study in metastatic renal cell cancer (mRCC). J Clin Oncol. 2014;32(suppl; abstr 2597).
  • Mangoni A, Woodman R, Kichenadasse G, et al. Anti-VEGF-induced hypertension and cancer outcomes: translating research into clinical practice. Exp Rev Prec Med Drug Dev. 2016;1:125–127.
  • Shah DR, Shah RR, Morganroth J. Tyrosine kinase inhibitors: their on-target toxicities as potential indicators of efficacy. Drug Saf. 2013;36:413–426.
  • Steeghs N, Gelderblom H, Roodt JO, et al. Hypertension and rarefaction during treatment with telatinib, a small molecule angiogenesis inhibitor. Clin Cancer Res. 2008;14:3470–3476.
  • Sorich M, Rowland A, Kichenadasse G, et al. Risk factors of proteinuria in renal cell carcinoma patients treated with VEGF inhibitors: a secondary analysis of pooled clinical trial data. Br J Cancer. 2016;114:1313–1317.
  • Riesenbeck LM, Bierer S, Hoffmeister I, et al. Hypothyroidism correlates with a better prognosis in metastatic renal cancer patients treated with sorafenib or sunitinib. World J Urol. 2011;29:807–813.
  • Daimon M, Kato T, Kaino W, et al. Thyroid dysfunction in patients treated with tyrosine kinase inhibitors, sunitinib, sorafenib and axitinib, for metastatic renal cell carcinoma. Jpn J Clin Oncol. 2012;42:742–747.
  • Clemons J, Gao D, Naam M, et al. Thyroid dysfunction in patients treated with sunitinib or sorafenib. Clin Genitourin Cancer. 2012;10:225–231.
  • Pérez-Soler R. Can rash associated with HER1/EGFR inhibition be used as a marker of treatment outcome? Oncology. 2003;17:23–28.
  • Perez-Soler R. Rash as a surrogate marker for efficacy of epidermal growth factor receptor inhibitors in lung cancer. Clin Lung Cancer. 2006;8(Suppl 1):S7–S14.
  • Wacker B, Nagrani T, Weinberg J, et al. Correlation between development of rash and efficacy in patients treated with the epidermal growth factor receptor tyrosine kinase inhibitor erlotinib in two large phase III studies. Clin Cancer Res. 2007;13:3913–3921.
  • Vincenzi B, Santini D, Russo A, et al. Early skin toxicity as a predictive factor for tumor control in hepatocellular carcinoma patients treated with sorafenib. Oncologist. 2010;15:85–92.
  • Petrelli F, Borgonovo K, Cabiddu M, et al. Relationship between skin rash and outcome in non-small-cell lung cancer patients treated with anti-EGFR tyrosine kinase inhibitors: a literature-based meta-analysis of 24 trials. Lung Cancer. 2012;78:8–15.
  • Mita AC, Papadopoulos K, De Jonge MJ, et al. Erlotinib ‘dosing-to-rash’: a phase II intrapatient dose escalation and pharmacologic study of erlotinib in previously treated advanced non-small cell lung cancer. Br J Cancer. 2011;105:938–944.
  • Lyman GH. Impact of chemotherapy dose intensity on cancer patient outcomes. J Natl Compr Canc Netw. 2009;7:99–108.
  • Mantyh PW. Cancer pain and its impact on diagnosis, survival and quality of life. Nat Rev Neurosci. 2006;7:797–809.
  • Santos FP, Kantarjian H, Fava C, et al. Clinical impact of dose reductions and interruptions of second-generation tyrosine kinase inhibitors in patients with chronic myeloid leukaemia. Br J Haematol. 2010;150:303–312.
  • Khosravan R, Huang X, Wiltshire R, et al. A retrospective analysis of data from two trials of sunitinib in patients with advanced renal cell carcinoma (RCC): pitfalls of efficacy subgroup analyses based on dose-reduction status. J Clin Oncol. 2012;suppl 5:abstr 363.
  • Bjarnason GA, Khalil B, Hudson JM, et al. Outcomes in patients with metastatic renal cell cancer treated with individualized sunitinib therapy: correlation with dynamic microbubble ultrasound data and review of the literature. Urol Oncol. 2014;32:480–487.
  • Najjar YG, Mittal K, Elson P, et al. A 2 weeks on and 1 week off schedule of sunitinib is associated with decreased toxicity in metastatic renal cell carcinoma. Eur J Cancer. 2014;50:1084–1089.
  • Foulon V, Schöffski P, Wolter P. Patient adherence to oral anticancer drugs: an emerging issue in modern oncology. Acta Clin Belg. 2011;66:85–96.
  • Gandhi S, Day L, Paramsothy T, et al. Oral anticancer medication adherence, toxicity reporting, and counseling: a study comparing health care providers and patients. J Oncol Prac. 2015;11:498–504.
  • Sorich MJ, Wiese MD, Rowland A, et al. Extended RAS mutations and anti-EGFR monoclonal antibody survival benefit in metastatic colorectal cancer: a meta-analysis of randomized controlled trials. Ann Oncol. 2015;26:13–21.
  • Rowland A, Dias MM, Wiese MD, et al. Meta-analysis of BRAF mutation as a predictive biomarker of benefit from anti-EGFR monoclonal antibody therapy for RAS wild-type metastatic colorectal cancer. Br J Cancer. 2015;112:1888–1894.
  • Rowland A, Dias MM, Wiese MD, et al. Meta-analysis comparing the efficacy of anti-EGFR monoclonal antibody therapy between KRAS G13D and other KRAS mutant metastatic colorectal cancer tumours. Eur J Cancer. 2016;55:122–130.
  • Elens L, Van Gelder T, Hesselink DA, et al. CYP3A4*22: promising newly identified CYP3A4 variant allele for personalizing pharmacotherapy. Pharmacogenomics. 2013;14:47–62.
  • Shin K-H, Choi MH, Lim KS, et al. Evaluation of endogenous metabolic markers of hepatic CYP3A activity using metabolic profiling and midazolam clearance. Clin Pharmacol Ther. 2013;94:601–609.
  • Ravegnini G, Sammarini G, Angelini S, et al. Pharmacogenetics of tyrosine kinase inhibitors in gastrointestinal stromal tumor and chronic myeloid leukemia. Expert Opin Drug Metabol Toxicol. 2016;12:733–742.
  • Wang D, Sadee W. The making of a CYP3A biomarker panel for guiding drug therapy. J Personal Med. 2012;2:175–191.
  • Niemi M, Pasanen MK, Neuvonen PJ. Organic anion transporting polypeptide 1B1: a genetically polymorphic transporter of major importance for hepatic drug uptake. Pharmacol Rev. 2011;63:157–181.
  • Lancaster CS, Sprowl JA, Walker AL, et al. Modulation of OATP1B-type transporter function alters cellular uptake and disposition of platinum chemotherapeutics. Mol Cancer Ther. 2013;12:1537–1544.
  • Snyder BD, Rowland A, Polasek TM, et al. Evaluation of felodipine as a potential perpetrator of pharmacokinetic drug-drug interactions. Eur J Clin Pharmacol. 2014;70:1115–1122.
  • Mårde Arrhén Y, Nylén H, Lövgren-Sandblom A, et al. A comparison of 4β-hydroxycholesterol : cholesterol and 6β-hydroxycortisol : cortisol as markers of CYP3A4 induction. Br J Clin Pharmacol. 2013;75:1536–1540.
  • Shen H, Dai J, Liu T, et al. Coproporphyrins I and III as functional markers of OATP1B activity: in vitro and in vivo evaluation in preclinical species. J Pharmacol Exp Ther. 2016;357:382–393.
  • Karapetis CS, Khambata-Ford S, Jonker DJ, et al. K-ras mutations and benefit from cetuximab in advanced colorectal cancer. N Engl J Med. 2008;359:1757–1765.
  • Rowland A, Dias MM, Wiese MD, et al. Reply: comment ‘meta-analysis of BRAF mutation as a predictive biomarker of benefit from anti-EGFR monoclonal antibody therapy for RAS wild-type metastatic colorectal cancer’. Br J Cancer. 2015;113:1635.
  • Rowland A, Karapetis C, Mangoni A, et al. Clinical translation of predictive markers for anti-EGFR monoclonal antibody therapy in metastatic colorectal cancer. Transl Cancer Res. 2016;5:S31–S34.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.