aI
(μM) represents the Cmax of patients with solid tumours,
which were cited from (Di Gion et al., 2011; Gschwind et al., 2005;
Sparano et al., 2009) for imatinib, (Di Gion et al., 2011) for sunitinib
and (Scheffler, Di Gion, Doroshyenko, Wolf & Fuhr, 2011) for gefitinib.
b The AUC ratio was calculated based on Equations
(2)–(4).
Discussion
There is extensive clinical application of rivaroxaban in combination
with TKIs, and cancer patients are one of the main groups that receive
rivaroxaban treatment. Due to the hypercoagulability of cancer patients
and the thrombogenicity of anti-cancer agents, anticoagulant therapy has
become an essential treatment for cancer patients. Increasing evidence
has indicated that DOACs are safer and more convenient than classical
anticoagulation medications, which is reflected in increasing clinical
guidelines recommending that DOACs replace classical anticoagulation
medications for cancer patient VTE therapy. In our previous study,
CYP2J2, but not CYP3A4, was found to be dominant in the P450 metabolism
of rivaroxaban with a contribution of 41.1%. However, few published
reports have targeted CYP2J2 to explore the safety of rivaroxaban
combinations, and even fewer of these have looked at rivaroxaban in
combination with TKIs. Therefore, the estimation of DDI risk between
TKIs and rivaroxaban based on CYP2J2 and CYP3A4 is necessary and
meaningful for clinical practice. This was the focus of the present
study.
All three TKIs were found to have a remarkable inhibitory effect on
CYP3A4-mediated rivaroxaban metabolism. Imatinib showed the strongest
reversible inhibitory effect towards CYP3A4 with a
Ki value of 1.92 μM. The inhibition of CYP3A4 by
sunitinib or gefitinib was also potent, with Kivalues of 13.24 and 4.91 μM, respectively. In addition to reversible
inhibition, the three TKIs also effected time-dependent inactivation of
CYP3A4, especially sunitinib, which greatly increased the DDI risk in
combination with rivaroxaban. Compared with reversible inhibition,
mechanism-dependent inactivation is always more frequently related to
unfavourable DDIs in clinical practice (Kalgutkar, Obach & Maurer,
2007). More importantly, the expression of CYP3A4—which is abundantly
expressed P450 isoform in the liver in most individuals—showed more
than 100-fold population variability (Zanger & Schwab, 2013). And
meanwhile, it has been reported that sunitinib has a 10-fold higher
concentration in the liver than in blood (Lau et al., 2015). Thus, being
cautious about the CYP3A4 enzyme expression in patients who take both
rivaroxaban and TKIs is necessary.
The present study found that CYP2J2, which dominates the metabolism of
rivaroxaban, was inhibited by the TKIs. Notably, the three TKIs showed
different inhibitory activity: imatinib and gefitinib were potent
inhibitors of CYP2J2 with Ki values of 3.53 and
2.99
μM,
respectively, but sunitinib had almost no inhibitory effect on CYP2J2.
Additionally, there was no irreversible inhibition of CYP2J2 by the
three TKIs, with the results showing IC50 shifts of less
than 1.5-fold. The basic principle of mechanism-based inactivation is
the bioactivation of inhibitors, which is achieved by their metabolism
by the inhibited enzymes (Kalgutkar, Obach & Maurer, 2007). Therefore,
the results might suggest that the three TKIs were not or were rarely
metabolised by CYP2J2.
Although the three TKIs did not irreversibly inactivate CYP2J2-mediated
rivaroxaban metabolism, the DDI risk produced by inhibiting CYP2J2
cannot be ignored. The distribution of CYP2J2 in vivo is a potential
factor that may increase the DDI risk of rivaroxaban in combination with
TKIs. CYP2J2 was first detected in the liver but was since identified as
an enzyme that is mainly distributed in the heart (Das, Weigle, Arnold,
Kim, Carnevale & Huff, 2020); indeed, the mRNA levels of CYP2J2 in the
cardiovascular system exceed those of other detected isozymes by 3
million to 62 times (Michaud, Frappier, Dumas & Turgeon, 2010; Wu,
Moomaw, Tomer, Falck & Zeldin, 1996). Although CYP2J2 is not usually
considered to be a DDI target due to its lower content in the liver, if
the heart is set as the organ for potential DDIs, there may be an
extremely high risk of DDI between rivaroxaban and TKIs.
In addition to its physiological distribution in the cardiovascular
system, CYP2J2 has also recently been found to be highly expressed in
various tumour tissues (Das, Weigle, Arnold, Kim, Carnevale & Huff,
2020; Jiang et al., 2005; Karkhanis, Hong & Chan, 2017). Due to the
overexpression of receptors in tumours (such
as
PDGFR and EGFR), TKIs would accumulate in the tumour tissue. Therefore,
the DDI risks of rivaroxaban in combination with TKIs for cancer
patients may be higher than in our predictions, and CYP2J2 may need to
become a target for DDI assessments involving rivaroxaban.
Imatinib was predicted to have moderate DDI risk when combined with
rivaroxaban. Based on the inhibitory constants of imatinib on CYP2J2 and
CYP3A4, and the metabolic contributions of two isoforms, imatinib was
predicted to yield at most a 2.44-fold rivaroxaban AUC increase.
Therefore, according to the FDA’s guidelines for the relative risk of
DDIs, a moderate DDI risk may exist for the combination of rivaroxaban
and imatinib (Table 6). This result was predicted from the
pharmacokinetic data of patients with solid tumours who were received
TKIs. Notably, cancer patients have poorer metabolic function than
normal patients, which may directly lead to higher plasma concentrations
of TKIs for cancer patients. Therefore, using pharmacokinetic data from
cancer patients may make our prediction more accurate. However,
metabolic enzyme activity in the normal population is subject to
individual variability, so factors such as hepatic blood flow and
genetic polymorphism cannot be neglected (Zanger & Schwab, 2013).
Factors that influence the enzyme activity of cancer patients may be
more difficult to determine due to the more complicated in vivo
environment. All these factors would affect the accuracy of our
prediction in clinical practice. Additionally, the plasma concentrations
of anti-cancer drugs in patients with solid tumours that are refractory
to standard therapy are much higher than those of normal patients, which
may result from large doses of anti-cancer drugs and metabolic function
disorders (Scheffler, Di Gion, Doroshyenko, Wolf & Fuhr, 2011).
Furthermore, it has been reported that lower plasma concentrations of
TKIs (< 1100 ng·ml–1) are closely related
to the much faster development of progressive disease and lower
objective response rates for GIST patients (von Mehren & Widmer, 2011).
All these factors would produce great diversity in the pharmacokinetic
data of TKIs in cancer patients. Thus, a rough in vitro prediction may
neglect to indicate the risk of severe DDI in some individuals.
Therefore, an individual physiologically-based pharmacokinetic model for
cancer patients would be encouraged in the prediction of the
pharmacokinetic behaviour of rivaroxaban in combination with TKIs.
In summary, all three TKIs (imatinib, sunitinib and gefitinib) showed
inhibitory effects on CYP2J2- and CYP3A4-mediated rivaroxaban
metabolism. Imatinib and gefitinib exerted significant reversible
inhibition of CYP2J2 and CYP3A4, while sunitinib only showed reversible
inhibition of CYP3A4. The three TKIs also demonstrated time-dependent
inactivation of CYP3A4, with this effect being slight on CYP2J2.
Furthermore, the combination of rivaroxaban with imatinib was predicted
to constitute a moderate DDI risk. Our results provide data for the
clinical safety assessment of the combination of rivaroxaban with
imatinib, sunitinib and gefitinib in cancer patients, and also give new
insights for DDI assessment involving rivaroxaban.
Allison SE, Chen Y, Petrovic N, Zhang J, Bourget K, Mackenzie PI,
et al. (2017). Activation of ALDH1A1 in MDA-MB-468 breast cancer cells
that over-express CYP2J2 protects against paclitaxel-dependent cell
death mediated by reactive oxygen species. Biochem Pharmacol
143: 79-89.
Blom JW, Doggen CJ, Osanto S, & Rosendaal FR (2005). Malignancies,
prothrombotic mutations, and the risk of venous thrombosis. Jama
293: 715-722.
Burotto M, Manasanch EE, Wilkerson J, & Fojo T (2015). Gefitinib and
erlotinib in metastatic non-small cell lung cancer: a meta-analysis of
toxicity and efficacy of randomized clinical trials. Oncologist
20: 400-410.
Cheng AL, Kang YK, Lin DY, Park JW, Kudo M, Qin S, et al. (2013).
Sunitinib versus sorafenib in advanced hepatocellular cancer: results of
a randomized phase III trial. Journal of clinical oncology : official
journal of the American Society of Clinical Oncology 31:4067-4075.
Cheong EJ, Goh JJ, Hong Y, Venkatesan G, Liu Y, Chiu GN, et al.(2017). Application of Static Modeling –in the Prediction of In Vivo
Drug-Drug Interactions between Rivaroxaban and Antiarrhythmic Agents
Based on In Vitro Inhibition Studies. Drug Metab Dispos 45:260-268.
Crusz SM, & Balkwill FR (2015). Inflammation and cancer: advances and
new agents. Nat Rev Clin Oncol 12: 584-596.
Das A, Weigle AT, Arnold WR, Kim JS, Carnevale LN, & Huff HC (2020).
CYP2J2 Molecular Recognition: A New Axis for Therapeutic Design.
Pharmacol Ther 215: 107601.
Dhillon S (2015). Gefitinib: a review of its use in adults with advanced
non-small cell lung cancer. Target Oncol 10: 153-170.
Di Gion P, Kanefendt F, Lindauer A, Scheffler M, Doroshyenko O, Fuhr
U, et al. (2011). Clinical pharmacokinetics of tyrosine kinase
inhibitors: focus on pyrimidines, pyridines and pyrroles. Clin
Pharmacokinet 50: 551-603.
Diakos CI, Charles KA, McMillan DC, & Clarke SJ (2014). Cancer-related
inflammation and treatment effectiveness. Lancet Oncol 15:e493-503.
Douketis JD, Crowther MA, Foster GA, & Ginsberg JS (2001). Does the
location of thrombosis determine the risk of disease recurrence in
patients with proximal deep vein thrombosis? Am J Med 110:515-519.
Fahmi OA, Maurer TS, Kish M, Cardenas E, Boldt S, & Nettleton D (2008).
A combined model for predicting CYP3A4 clinical net drug-drug
interaction based on CYP3A4 inhibition, inactivation, and induction
determined in vitro. Drug Metab Dispos 36: 1698-1708.
Farmakis D, Parissis J, & Filippatos G (2014). Insights into
onco-cardiology: atrial fibrillation in cancer. J Am Coll Cardiol
63: 945-953.
Gschwind HP, Pfaar U, Waldmeier F, Zollinger M, Sayer C, Zbinden
P, et al. (2005). Metabolism and disposition of imatinib mesylate
in healthy volunteers. Drug Metab Dispos 33: 1503-1512.
Guilhot F (2004). Indications for imatinib mesylate therapy and clinical
management. Oncologist 9: 271-281.
Isoherranen N, Lutz JD, Chung SP, Hachad H, Levy RH, &
Ragueneau-Majlessi I (2012). Importance of multi-p450 inhibition in
drug-drug interactions: evaluation of incidence, inhibition magnitude,
and prediction from in vitro data. Chem Res Toxicol 25:2285-2300.
January CT, Wann LS, Calkins H, Chen LY, Cigarroa JE, Cleveland JC,
Jr., et al. (2019). 2019 AHA/ACC/HRS Focused Update of the 2014
AHA/ACC/HRS Guideline for the Management of Patients With Atrial
Fibrillation: A Report of the American College of Cardiology/American
Heart Association Task Force on Clinical Practice Guidelines and the
Heart Rhythm Society in Collaboration With the Society of Thoracic
Surgeons. Circulation 140: e125-e151.
Jiang JG, Chen CL, Card JW, Yang S, Chen JX, Fu XN, et al.(2005). Cytochrome P450 2J2 promotes the neoplastic phenotype of
carcinoma cells and is up-regulated in human tumors. Cancer Res
65: 4707-4715.
Jiao Q, Bi L, Ren Y, Song S, Wang Q, & Wang YS (2018). Advances in
studies of tyrosine kinase inhibitors and their acquired resistance. Mol
Cancer 17: 36.
Kalgutkar AS, Obach RS, & Maurer TS (2007). Mechanism-based
inactivation of cytochrome P450 enzymes: chemical mechanisms,
structure-activity relationships and relationship to clinical drug-drug
interactions and idiosyncratic adverse drug reactions. Curr Drug Metab
8: 407-447.
Kalra S, Rini BI, & Jonasch E (2015). Alternate sunitinib schedules in
patients with metastatic renal cell carcinoma. Ann Oncol 26:1300-1304.
Karkhanis A, Hong Y, & Chan ECY (2017). Inhibition and inactivation of
human CYP2J2: Implications in cardiac pathophysiology and opportunities
in cancer therapy. Biochem Pharmacol 135: 12-21.
Kuczynski EA, Lee CR, Man S, Chen E, & Kerbel RS (2015). Effects of
Sorafenib Dose on Acquired Reversible Resistance and Toxicity in
Hepatocellular Carcinoma. Cancer Res 75: 2510-2519.
Lau CL, Chan ST, Selvaratanam M, Khoo HW, Lim AY, Modamio P, et
al. (2015). Sunitinib-ibuprofen drug interaction affects the
pharmacokinetics and tissue distribution of sunitinib to brain, liver,
and kidney in male and female mice differently. Fundamental & clinical
pharmacology 29: 404-416.
Levitan N, Dowlati A, Remick SC, Tahsildar HI, Sivinski LD, Beyth
R, et al. (1999). Rates of initial and recurrent thromboembolic
disease among patients with malignancy versus those without malignancy.
Risk analysis using Medicare claims data. Medicine (Baltimore)
78: 285-291.
Li JN, Cao YF, He RR, Ge GB, Guo B, & Wu JJ (2018). Evidence for
Shikonin acting as an active inhibitor of human carboxylesterases 2:
Implications for herb-drug combination. Phytother Res 32:1311-1319.
Michaud V, Frappier M, Dumas MC, & Turgeon J (2010). Metabolic activity
and mRNA levels of human cardiac CYP450s involved in drug metabolism.
PLoS One 5: e15666.
Monreal M, Falgá C, Valdés M, Suárez C, Gabriel F, Tolosa C, et
al. (2006). Fatal pulmonary embolism and fatal bleeding in cancer
patients with venous thromboembolism: findings from the RIETE registry.
J Thromb Haemost 4: 1950-1956.
Nattel S, Burstein B, & Dobrev D (2008). Atrial remodeling and atrial
fibrillation: mechanisms and implications. Circulation Arrhythmia and
electrophysiology 1: 62-73.
Nattel S, & Harada M (2014). Atrial remodeling and atrial fibrillation:
recent advances and translational perspectives. J Am Coll Cardiol
63: 2335-2345.
Nebot N, Crettol S, d’Esposito F, Tattam B, Hibbs DE, & Murray M
(2010). Participation of CYP2C8 and CYP3A4 in the N-demethylation of
imatinib in human hepatic microsomes. Br J Pharmacol 161:1059-1069.
Ning J, Liu T, Dong P, Wang W, Ge G, Wang B, et al. (2019).
Molecular Design Strategy to Construct the Near-Infrared Fluorescent
Probe for Selectively Sensing Human Cytochrome P450 2J2. J Am Chem Soc
141: 1126-1134.
O’Brien SG, Guilhot F, Larson RA, Gathmann I, Baccarani M, Cervantes
F, et al. (2003). Imatinib compared with interferon and low-dose
cytarabine for newly diagnosed chronic-phase chronic myeloid leukemia. N
Engl J Med 348: 994-1004.
Onaitis M, D’Amico T, Zhao Y, O’Brien S, & Harpole D (2010). Risk
factors for atrial fibrillation after lung cancer surgery: analysis of
the Society of Thoracic Surgeons general thoracic surgery database. Ann
Thorac Surg 90: 368-374.
Prandoni P, Lensing AW, Piccioli A, Bernardi E, Simioni P, Girolami
B, et al. (2002). Recurrent venous thromboembolism and bleeding
complications during anticoagulant treatment in patients with cancer and
venous thrombosis. Blood 100: 3484-3488.
Prins MH, Lensing AW, Bauersachs R, van Bellen B, Bounameaux H, Brighton
TA, et al. (2013). Oral rivaroxaban versus standard therapy for
the treatment of symptomatic venous thromboembolism: a pooled analysis
of the EINSTEIN-DVT and PE randomized studies. Thrombosis journal
11: 21.
Saliba W, Rennert HS, Gronich N, Gruber SB, & Rennert G (2018).
Association of atrial fibrillation and cancer: Analysis from two large
population-based case-control studies. PLoS One 13: e0190324.
Sanfilippo KM, & Wang TF (2019). Prevention and Treatment of
Cancer-Associated Venous Thromboembolism: a Review. Curr Treat Options
Cardiovasc Med 21: 70.
Scheffler M, Di Gion P, Doroshyenko O, Wolf J, & Fuhr U (2011).
Clinical pharmacokinetics of tyrosine kinase inhibitors: focus on
4-anilinoquinazolines. Clin Pharmacokinet 50: 371-403.
Shalhoub J, Norrie J, Baker C, Bradbury AW, Dhillon K, Everington
T, et al. (2017). Graduated Compression Stockings as an Adjunct
to Low Dose Low Molecular Weight Heparin in Venous Thromboembolism
Prevention in Surgery: A Multicentre Randomised Controlled Trial. Eur J
Vasc Endovasc Surg 53: 880-885.
Song AB, Rosovsky RP, Connors JM, & Al-Samkari H (2019). Direct oral
anticoagulants for treatment and prevention of venous thromboembolism in
cancer patients. Vascular health and risk management 15:175-186.
Sparano BA, Egorin MJ, Parise RA, Walters J, Komazec KA, Redner
RL, et al. (2009). Effect of antacid on imatinib absorption.
Cancer Chemother Pharmacol 63: 525-528.
Streiff MB (2016). Thrombosis in the setting of cancer. Hematology Am
Soc Hematol Educ Program 2016: 196-205.
Streiff MB, Holmstrom B, Angelini D, Ashrani A, Bockenstedt PL, Chesney
C, et al. (2020). NCCN Guidelines Insights: Cancer-Associated
Venous Thromboembolic Disease, Version 1.2020. J Natl Compr Canc Netw
16: 1289-1303.
Timp JF, Braekkan SK, Versteeg HH, & Cannegieter SC (2013).
Epidemiology of cancer-associated venous thrombosis. Blood 122:1712-1723.
Tirumani SH, Jagannathan JP, Krajewski KM, Shinagare AB, Jacene H, &
Ramaiya NH (2013). Imatinib and beyond in gastrointestinal stromal
tumors: A radiologist’s perspective. AJR Am J Roentgenol 201:801-810.
von Mehren M, & Widmer N (2011). Correlations between imatinib
pharmacokinetics, pharmacodynamics, adherence, and clinical response in
advanced metastatic gastrointestinal stromal tumor (GIST): an emerging
role for drug blood level testing? Cancer Treat Rev 37:291-299.
Wertheimer C, Siedlecki J, Kook D, Mayer WJ, Wolf A, Klingenstein
A, et al. (2015). EGFR inhibitor Gefitinib attenuates posterior
capsule opacification in vitro and in the ex vivo human capsular bag
model. Graefes Arch Clin Exp Ophthalmol 253: 409-417.
Wu S, Moomaw CR, Tomer KB, Falck JR, & Zeldin DC (1996). Molecular
cloning and expression of CYP2J2, a human cytochrome P450 arachidonic
acid epoxygenase highly expressed in heart. J Biol Chem 271:3460-3468.
Zamorano JL, Lancellotti P, Rodriguez Muñoz D, Aboyans V, Asteggiano R,
Galderisi M, et al. (2016). 2016 ESC Position Paper on cancer
treatments and cardiovascular toxicity developed under the auspices of
the ESC Committee for Practice Guidelines: The Task Force for cancer
treatments and cardiovascular toxicity of the European Society of
Cardiology (ESC). Eur Heart J 37: 2768-2801.
Zanger UM, & Schwab M (2013). Cytochrome P450 enzymes in drug
metabolism: regulation of gene expression, enzyme activities, and impact
of genetic variation. Pharmacol Ther 138: 103-141.
Zhao T, Chen Y, Wang D, Wang L, Dong P, Zhao S, et al. (2021).
Identifying the Dominant Contribution of Human Cytochrome P450 2J2 to
the Metabolism of Rivaroxaban, an Oral Anticoagulant. Cardiovasc Drugs
Ther.