A dose-ranging study of ticagrelor in children aged 3-17 years with sickle cell disease: A 2-part phase 2 study

Lewis L Hsu, Sharada Sarnaik, Suzan Williams, Carl Amilon, Jenny Wissmar, Anders Berggren, HESTIA1 Investigators, Lewis L Hsu, Sharada Sarnaik, Suzan Williams, Carl Amilon, Jenny Wissmar, Anders Berggren, HESTIA1 Investigators

Abstract

Antiplatelet treatment is a potential therapeutic approach for sickle cell disease (SCD). Ticagrelor inhibits platelet aggregation and is approved for adults with acute coronary syndrome and following myocardial infarction. HESTIA1 (NCT02214121) was a 2-part, phase 2 dose-finding study generating ticagrelor exposure, platelet inhibition, and safety data in children with SCD (3-17 years). In part A (n = 45), patients received 2 ticagrelor single doses, 0.125-2.25 mg/kg (washout ≥7 days), then 7 days of twice-daily (bid) dosing with 0.125, 0.563, or 0.75 mg/kg. In the 4-week blinded Part B extension (optional), patients received ticagrelor (0.125, 0.563, or 0.75 mg/kg bid; n = 16) or placebo (n = 7). Platelet reactivity decreased from baseline to 2 hours postdosing, and returned to near baseline after 6 hours postdosing. Dose-dependent platelet inhibition was seen with ticagrelor; mean relative P2Y12 reaction unit inhibition 2 hours after a single dose ranged from 6% (0.125 mg/kg) to 73% (2.25 mg/kg). Ticagrelor plasma exposure increased approximately dose proportionally. No patients experienced a hemorrhagic event during treatment. No differences were seen between groups in pain ratings and analgesic use during Part B. Ticagrelor was well tolerated with no safety concerns, no discontinuations due to adverse events (AEs), and reported AEs were mainly due to SCD. In conclusion, a dose-exposure-response relationship for ticagrelor was demonstrated in children with SCD for the first time. These data are important for future pediatric studies of the efficacy and safety of ticagrelor in SCD.

© 2018 The Authors. American Journal of Hematology published by Wiley Periodicals, Inc.

Figures

Figure 1
Figure 1
Boxplots of absolute (A) and relative (B) PRU at 2 hours postdose after single ticagrelor doses in part A. A, the observed PRU values at 2 hours post a single dose at both visits 2 and 3. PRU % inhibition was calculated by the dividing the observed PRU value at 2 hours by the individual baseline PRU value (1−[observed PRU/baseline PRU]) × 100, values above 100% and below 0% were imputed to 100% and 0%, respectively (Figure B only). For patients missing a visit 2 baseline PRU, the visit 3 predose value (following a 1‐week wash‐out) was used. The boxplot whiskers display the 5th and 95th percentile of the data, values beyond this (outliers) are excluded from the graphs. Abbreviation: PRU, P2Y12 reactivity unit
Figure 2
Figure 2
Boxplots of observed plasma concentrations at 2 hours postdose of (A) ticagrelor and (B) AR‐C124910XX following a single ticagrelor dose. The boxplot whiskers display the 5th and 95th percentile of the data, values beyond this (outliers) are excluded from the graphs. Values below the limit of quantification of the bioanalysis method are not included

References

    1. Ataga KI, Brittain JE, Desai P, et al. Association of coagulation activation with clinical complications in sickle cell disease. PLoS One. 2012;7:e29786.
    1. Zhang D, Xu C, Manwani D, Frenette PS. Neutrophils, platelets, and inflammatory pathways at the nexus of sickle cell disease pathophysiology. Blood. 2016;127:801‐809.
    1. Owusu‐Ansah A, Ihunnah CA, Walker AL, Ofori‐Acquah SF. Inflammatory targets of therapy in sickle cell disease. Transl Res. 2016;167:281‐297.
    1. Sparkenbaugh E, Pawlinski R. Prothrombotic aspects of sickle cell disease. J Thromb Haemost. 2017;15:1307‐1316.
    1. Ansari J, Moufarrej YE, Pawlinski R, Gavins FNE. Sickle cell disease: a malady beyond a hemoglobin defect in cerebrovascular disease. Expert Rev Hematol. 2018;11:45‐55.
    1. Noubouossie D, Key NS, Ataga KI. Coagulation abnormalities of sickle cell disease: relationship with clinical outcomes and the effect of disease modifying therapies. Blood Rev. 2016;30:245‐256.
    1. Bennewitz MF, Jimenez MA, Vats R, et al. Lung vaso‐occlusion in sickle cell disease mediated by arteriolar neutrophil‐platelet microemboli. JCI Insight. 2017;2:e89761.
    1. Heeney MM, Hoppe CC, Abboud MR, et al. A multinational trial of prasugrel for sickle cell vaso‐occlusive events. N Engl J Med. 2016;374:625‐635.
    1. Jakubowski JA, Hoppe CC, Zhou C, et al. Real‐time dose adjustment using point‐of‐care platelet reactivity testing in a double‐blind study of prasugrel in children with sickle cell anaemia. Thromb Haemost. 2017;117:580‐588.
    1. Husted S, van Giezen JJ. Ticagrelor: the first reversibly binding oral P2Y12 receptor antagonist. Cardiovasc Ther. 2009;27:259‐274.
    1. van Giezen JJ, Nilsson L, Berntsson P, et al. Ticagrelor binds to human P2Y(12) independently from ADP but antagonizes ADP‐induced receptor signaling and platelet aggregation. J Thromb Haemost. 2009;7:1556‐1565.
    1. Nylander S, Schulz R. Effects of P2Y12 receptor antagonists beyond platelet inhibition – comparison of ticagrelor with thienopyridines. Br J Pharmacol. 2016;173:1163‐1178.
    1. Armstrong D, Summers C, Ewart L, Nylander S, Sidaway JE, van Giezen JJ. Characterization of the adenosine pharmacology of ticagrelor reveals therapeutically relevant inhibition of equilibrative nucleoside transporter 1. J Cardiovasc Pharmacol Ther. 2014;19:209‐219.
    1. Nylander S, Femia EA, Scavone M, et al. Ticagrelor inhibits human platelet aggregation via adenosine in addition to P2Y12 antagonism. J Thromb Haemost. 2013;11:1867‐1176.
    1. Wallentin L, Becker RC, Budaj A, et al. Ticagrelor versus clopidogrel in patients with acute coronary syndromes. N Engl J Med. 2009;361:1045‐1057.
    1. Bonaca MP, Bhatt DL, Cohen M, et al. Long‐term use of ticagrelor in patients with prior myocardial infarction. N Engl J Med. 2015;372:1791‐1800.
    1. Brilinta (ticagrelor) tablets, for oral use. US prescribing Information. Revised March 2018. Available at: . Last accessed April 19, 2018.
    1. Butler K, Teng R. Pharmacokinetics, pharmacodynamics, safety and tolerability of multiple ascending doses of ticagrelor in healthy volunteers. Br J Clin Pharmacol. 2010;70:65‐77.
    1. Teng R, Butler K. Pharmacokinetics, pharmacodynamics, tolerability and safety of single ascending doses of ticagrelor, a reversibly binding oral P2Y(12) receptor antagonist, in healthy subjects. Eur J Clin Pharmacol. 2010;66:487‐496.
    1. Husted S, Emanuelsson H, Heptinstall S, Sandset PM, Wickens M, Peters G. Pharmacodynamics, pharmacokinetics, and safety of the oral reversible P2Y12 antagonist AZD6140 with aspirin in patients with atherosclerosis: a double‐blind comparison to clopidogrel with aspirin. Eur Heart J. 2006;27:1038‐1047.
    1. Teng R. Ticagrelor: pharmacokinetic, pharmacodynamic and pharmacogenetic profile: an update. Clin Pharmacokinet. 2015;54:1125‐1138.
    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. Teng R, Oliver S, Hayes MA, Butler K. Absorption, distribution, metabolism, and excretion of ticagrelor in healthy subjects. Drug Metab Dispos. 2010;38:1514‐1521.
    1. Söderlund F, Asztély A‐K, Jeppsson A, et al. In vitro anti‐platelet potency of ticagrelor in blood samples from infants and children. Thromb Res. 2015;136:620‐624.
    1. Cabannes R, Lonsdorfer J, Castaigne JP, Ondo A, Plassard A, Zohoun I. Clinical and biological double‐blind‐study of ticlopidine in preventive treatment of sickle‐cell disease crises. Agents Actions Suppl. 1984;15:199‐212.
    1. Ataga KI, Kutlar A, Kanter J, et al. Crizanlizumab for the prevention of pain crises in sickle cell disease. N Engl J Med. 2017;376:429‐439.
    1. Matsui NM, Borsig L, Rosen SD, Yaghmai M, Varki A, Embury SH. P‐selectin mediates the adhesion of sickle erythrocytes to the endothelium. Blood. 2001;98:1955‐1962.
    1. Ware RE, de Montalembert M, Tshilol L, et al. Sickle cell disease. Lancet. 2017;390:311‐323.
    1. Novelli EM, Gladwin MT. Crises in sickle cell disease. Chest. 2016;149:1082‐1093.
    1. Subramaniam S, Chao JH. Managing acute complication of sickle cell disease in pediatric patients. Pediatr Emerg Med Pract. 2013;13:1‐28.
    1. Chakravorty S, Williams TN. Sickle cell disease: a neglected chronic disease of increasing global health importance. Arch Dis Child. 2015;100:48‐53.
    1. Heeney MM, Abboud MR, Amilon C, et al. Ticagrelor versus placebo for the reduction of vaso‐occlusive crises in pediatric sickle cell disease: design of a randomized, double‐blind, parallel‐group, multicenter, phase 3 study (HESTIA3). HemaSphere. 2018;2(S1):669 abstract PS1460.

Source: PubMed

3
Abonnieren