Pharmacodynamics and pharmacokinetics of ticagrelor vs. clopidogrel in patients with acute coronary syndromes and chronic kidney disease

Heyang Wang, Jing Qi, Yi Li, Yunbiao Tang, Chao Li, Jing Li, Yaling Han, Heyang Wang, Jing Qi, Yi Li, Yunbiao Tang, Chao Li, Jing Li, Yaling Han

Abstract

Background: Pivotal clinical trials found that ticagrelor reduced ischaemic complications to a greater extent than clopidogrel, and also that the benefit gradually increased with the reduction in creatinine clearance. However, the underlying mechanisms remains poorly explored.

Methods: This was a single-centre, prospective, randomized clinical trial involving 60 hospitalized Adenosine Diphosphate (ADP) P2Y12 receptor inhibitor-naïve patients with chronic kidney disease (CKD) (estimated glomerular filtration rate <60 ml min-1 1.73 m-2 ) and non-ST-elevation acute coronary syndromes (NSTE-ACS). Eligible patients were randomly assigned in a 1:1 ratio to receive ticagrelor (180 mg loading dose, then followed by 90 mg twice daily) or clopidogrel (600 mg loading dose, then followed by 75 mg once daily). The primary endpoint was the P2Y12 reactive unit (PRU) value assessed by VerifyNow at 30 days. The plasma concentrations of ticagrelor and clopidogrel and their active metabolites were measured in the first 10 patients in each group at baseline, and at 1 h, 2 h, 4 h, 8 h, 12 h and 24 h after the loading dose.

Results: Baseline characteristics were well matched between the two groups. Our results indicated a markedly lower PRU in patients treated with ticagrelor vs. clopidogrel at 30 days (32.6 ± 11.29 vs. 203.7 ± 17.92; P < 0.001) as well as at 2 h, 8 h and 24 h after the loading dose (P < 0.001). Ticagrelor and its active metabolite AR-C124910XX showed a similar time to reach maximum concentration (Cmax ) of 8 h, with the maximum concentration (Cmax ) of 355 (242.50-522.00) ng ml-1 and 63.20 (50.80-85.15) ng ml-1 , respectively. Both clopidogrel and its active metabolite approached the Cmax at 2 h, with a similar Cmax of 8.67 (6.64-27.75) ng ml-1 vs. 8.53 (6.94-15.93) ng ml-1 .

Conclusion: Ticagrelor showed much more potent platelet inhibition in comparison with clopidogrel in patients with CKD and NSTE-ACS.

Keywords: chronic kidney disease; non-ST-elevation acute coronary syndromes; pharmacodynamics; ticagrelor.

© 2017 The British Pharmacological Society.

Figures

Figure 1
Figure 1
Flow chart of the COmparison of The Pharmacodynamics and pharmacokinetics of Ticagrelor vs. clopidogrel in patients with non–ST‐elevation acute coronary syndromes and Chronic Kidney Disease (OPT‐CKD) trial. Eligible patients were randomly assigned in a 1:1 ratio to receive ticagrelor or clopidogrel (R 1:1). ASA, aspirin; CKD, chronic kidney disease; CLO, clopidogrel; CYP, cytochrome P450; eGFR, estimated glomerular filtration rate; HPR, high on‐treatment platelet reactivity; IPA, inhibition of platelet aggregation; LD, loading dose; NSTE‐ACS, non‐ST‐elevation acute coronary syndromes; PRU, platelet reactive unit; qd, once daily; TIC, ticagrelor *Plasma concentrations of clopidogrel, clopidogrel active metabolite‐derivatized (CAMD), ticagrelor and its active metabolite AR‐C124910XX were assessed predose and 1, 2, 4, 8, 12 and 24 h after the loading dose
Figure 2
Figure 2
Platelet reaction units (A) and inhibition of platelet aggregation (B) by protocol time and treatment. Data are expressed as mean ± standard error *P < 0.001, ticagrelor vs. clopidogrel
Figure 3
Figure 3
Correlation between baseline estimated glomerular filtration rate (eGFR) (A) and cytochrome B450 2C19 genotype (B) and 30‐day inhibition of platelet aggregation in non‐ST‐elevation acute coronary syndromes patients with chronic kidney disease. EM, extensive metabolizer; IM, intermediate metabolizer; PM, poor metabolizer
Figure 4
Figure 4
Overview of clopidogrel and ticagrelor pharmacokinetics on day 1. Plasma concentrations of clopidogrel (A), clopidogrel active metabolite‐derivatized (CAMD) (B), ticagrelor (C) and its active metabolite AR‐C124910XX (D) were measured before and 1–24 h after the administration of a loading dose of 600 mg clopidogrel or 180 mg ticagrelor. Maximum concentration (Cmax) and time to reach Cmax (Tmax) were established directly from the measured plasma concentration of each patient and presented as medians with interquartile range. Individual data are also shown, and the pharmacokinetic trend was demonstrated via the connecting curves of mean values

References

    1. Best PJ, Steinhub SR, Berger PB, Dasgupta A, Brennan DM, Szczech LA, et al The efficacy and safety of short‐ and long‐term dual antiplatelet therapy in patients with mild or moderate chronic kidney disease: results from the Clopidogrel for the Reduction of Events During Observation (CREDO) trial. Am Heart J 2008; 155: 687–693.
    1. Dasgupta A, Steinhubl SR, Bhatt DL, Berger PB, Shao M, Mak KH, et al Clinical outcomes of patients with diabetic nephropathy randomized to clopidogrel plus aspirin versus aspirin alone (a post hoc analysis of the Clopidogrel for High Atherothrombotic Risk and Ischemic Stabilization, Management, and Avoidance [CHARISMA] trial). Am J Cardiol 2009; 103: 1359–1363.
    1. Park SH, Kim W, Park CS, Kang WY, Hwang SH, Kim W. A comparison of clopidogrel responsiveness in patients with versus without chronic renal failure. Am J Cardiol 2009; 104: 1292–1295.
    1. Angiolillo DJ, Bernardo E, Capodanno D, Vivas D, Sabaté M, Ferreiro JL, et al Impact of chronic kidney disease on platelet function profiles in diabetes mellitus patients with coronary artery disease taking dual antiplatelet therapy. J Am Coll Cardiol 2010; 55: 1139–1146.
    1. Guo LZ, Kim MH, Shim CH, Choi SY, Serebruany VL. Impact of renal impairment on platelet reactivity and clinical outcomes during chronic dual antiplatelet therapy following coronary stenting. Eur Heart J Cardiovasc Pharmacother 2016; 2: 145–151.
    1. Geisler T, Grass D, Bigalke B, Stellos K, Drosch T, Dietz K, et al The residual platelet aggregation after deployment of intracoronary stent (PREDICT) score. J Thromb Haemost 2008; 6: 54–61.
    1. Dreisbach AW. The influence of chronic renal failure on drug metabolism and transport. Clin Pharmacol Ther 2009; 86: 553–556.
    1. Husted S, van Giezen JJ. Ticagrelor: the first reversibly binding oral P2Y receptor antagonist. Cardiovasc Ther 2009; 27: 259–274.
    1. James S, Budaj A, Aylward P, Buck KK, Cannon CP, Cornel JH, et al Ticagrelor versus clopidogrel in acute coronary syndromes in relation to renal function: results from the platelet inhibition and patient outcomes (PLATO) trial. Circulation 2010; 122: 1056–1067.
    1. Ma YC, Zuo L, Chen JH, Luo Q, Yu XQ, Li Y, et al Modified glomerular filtration rate estimating equation for Chinese patients with chronic kidney disease. J Am Soc Nephrol 2006; 17: 2937–2944.
    1. Price MJ, Coleman JL, Steinhubl SR, Wong GB, Cannon CP, Teirstein PS. Onset and offset of platelet inhibition after high‐dose clopidogrel loading and standard daily therapy measured by a point‐of‐care assay in healthy volunteers. Am J Cardiol 2006; 98: 681–684.
    1. Angiolillo DJ, Curzen N, Gurbel P, Vaitkus P, Lipkin F, Li W, et al Pharmacodynamic evaluation of switching from ticagrelor to prasugrel in patients with stable coronary artery disease: results of the SWAP‐2 study (switching anti platelet‐2). J Am Coll Cardiol 2014; 63: 1500–1509.
    1. Sillén H, Cook M, Davis P. Determination of ticagrelor and two metabolites in plasma samples by liquid chromatography and mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2010; 878: 2299–2306.
    1. Peer CJ, Spencer SD, Van Den Berg DA, Pacanowski MA, Horenstein RB, Figg WD. A sensitive and rapid ultra HPLCMS/MS method for the simultaneous detection of clopidogrel and its derivatized active thiol metabolite in human plasma. J Chromatogr B Analyt Technol Biomed Life Sci 2012; 880: 132–139.
    1. Gugliucci A, Kinugasa E, Kotani K, Caccavello R, Kimura S. Serum paraoxonase 1 (PON1) lactonase activity is lower in end‐stage renal disease patients than in healthy control subjects and increases after hemodialysis. Clin Chem Lab Med 2011; 49: 61–67.
    1. Dohi T, Kasai T, Miyauchi K, Takasu K, Kajimoto K, Kubota N, et al Prognostic impact of chronic kidney disease on 10‐year clinical outcomes among patients with acute coronary syndrome. J Cardiol 2012; 60: 438–442.
    1. Chen Y, Dong W, Wan Z, Li Z, Cong H, Hong T, et al Ticagrelor versus clopidogrel in Chinese patients with acute coronary syndrome: a pharmacodynamic analysis. Int J Cardiol 2015; 201: 545–546.
    1. Franchi F, Rollini F, Cho JR, Bhatti M, DeGroat C, Ferrante E, et al Impact of escalating loading dose regimens of ticagrelor in patients with ST‐segment elevation myocardial infarction undergoing primary percutaneous coronary intervention: results of a prospective randomized pharmacokinetic and pharmacodynamic investigation. JACC Cardiovasc Interv 2015; 8: 1457–1467.
    1. Tang XF, Wang J, Zhang JH, Meng XM, Xu B, Qiao SB, et al Effect of the CYP2C19 2 and 3 genotypes, ABCB1 C3435T and PON1 Q192R alleles on the pharmacodynamics and adverse clinical events of clopidogrel in Chinese people after percutaneous coronary intervention. Eur J Clin Pharmacol 2013; 69: 1103–1112.
    1. Price MJ, Clavijo L, Angiolillo DJ, Carlson G, Caplan R, Teng R, et al A randomised trial of the pharmacodynamic and pharmacokinetic effects of ticagrelor compared with clopidogrel in Hispanic patients with stable coronary artery disease. J Thromb Thrombolysis 2015; 39: 8–14.
    1. Karaźniewicz‐Łada M, Danielak D, Burchardt P, Kruszyna L, Komosa A, Lesiak M, et al Clinical pharmacokinetics of clopidogrel and its metabolites in patients with cardiovascular diseases. Clin Pharmacokinet 2014; 53: 155–164.
    1. Danielak D, Karaźniewicz‐Łada M, Wiśniewska K, Bergus P, Burchardt P, Komosa A, et al Impact of CYP3A4*1G allele on clinical pharmacokinetics and pharmacodynamics of clopidogrel. Eur J Drug Metab Pharmacokinet 2017; 42: 99–107.

Source: PubMed

3
구독하다