Page 193 - 2019_11 Resto del Mondo-web
P. 193

Ibrutinib and acalabrutinib: effects on platelet functions
form of Src-kinases) were investigated in washed platelets from the two groups of healthy donors. Ibrutinib, at the clinically relevant dose of 0.5 mM, inhibited phosphoryla- tion at both sites, with a significantly more intense effect in the ibrutinib HS group. The strong inhibition of phospho- rylation observed in the ibrutinib HS group correlated with the inhibition of platelet aggregation. The weaker inhibi- tion of phosphorylation in the ibrutinib LS group was accompanied by a weaker decrease of aggregation. Acalabrutinib efficiently inhibited Btk phosphorylation, sig- nificantly decreased PLCγ2 Tyr-753 but had no effect on Src phosphorylation in either group, even at a dose of 2 mM which is above the 1.3 mM mean peak of free plasma drug concentrations measured in patients.9 These data are consis- tent with the better selectivity of acalabrutinib on Btk, also shown by the whole tyrosine phosphorylation profile of collagen-stimulated platelets. However, it is worth noting that while a dose of 2 mM acalabrutinib had no effect on Src activation it did decrease platelet aggregation significantly in the ibrutinib HS group.
A relevant clinical scenario is the association of Btk inhibitors with dual antiplatelet therapy in patients with
coronary intervention with stent placement. The current dual antiplatelet therapy is based on aspirin and a P2Y12 ADP receptor antagonist such as clopidogrel, prasugrel, ticagrelor or cangrelor. Our data are consistent with those of a previous study showing that ibrutinib amplifies the effect of cangrelor on platelets12 and also demonstrate that acalabrutinib strongly potentiated the effect of indomethacin or cangrelor on platelet aggregation induced by collagen both in the ibrutinib HS and LS groups of healthy donors. This is important information for guiding therapeutic strategies in patients under antiplatelet therapy at high risk of bleeding.
In conclusion, this study provides new insights into the impact of the first- and second-generation Btk inhibitors, ibrutinib and acalabrutinib, on platelets and contributes to the improvement of evidence-based recommendations for a safer use of these targeted therapies.
Acknowledgments
This work was supported by grants from Inserm, Fondation pour la Recherche Médicale (DEQ20170336737) and Janssen. We thank the Genotoul imaging facility of I2MC (Inserm U1048). BP is a scholar of the Institut Universitaire de France.
cardiovascular diseases, particularly after
percutaneous
References
1. Thompson PA, Burger JA. Bruton's tyrosine kinase inhibitors: first and second genera- tion agents for patients with chronic lym- phocytic leukemia (CLL). Expert Opin Investig Drugs. 2018;27(1):31-42.
2. Seiler T, Dreyling M. Bruton's tyrosine kinase inhibitors in B-cell lymphoma: cur- rent experience and future perspectives. Expert Opin Investig Drugs. 2017;26(8):909- 915.
3. Chakraborty R, Kapoor P, Ansell SM, Gertz MA. Ibrutinib for the treatment of Waldenström macroglobulinemia. Expert Rev Hematol. 2015;8(5):569-579.
4. ShatzelJJ,OlsonSR,TaoDL,McCartyOJT, Danilov AV, Deloughery TG. Ibrutinib-asso- ciated bleeding: pathogenesis, management and risk reduction strategies. J Thromb Haemost. 2017;15(5):835-847.
5. Caron F, Leong DP, Hillis C, Fraser G, Siegal D. Current understanding of bleeding with ibrutinib use : a systematic review and meta- analysis. Blood Adv. 2017;1(12):772-778.
6. Rushworth SA, MacEwan DJ, Bowles KM. Ibrutininb in relapsed chronic lymphocytic leukemia. N Engl J Med. 2013;369(13):1277- 1278.
7. Byrd JC, Fruman RR, Coutre SE, et al. Targeting BTK with ibrutininb in relapsed chronic lymphocytic leukemia. N Engl J Med. 2013;369(1):32-42.
8. Wang ML, Rule S, Martin P, Goy A, et al. Targeting BTK with ibrutinib in relapsed or refractory mantle-cell lymphoma. N Engl J Med. 2013;369(6):507-516.
9. Byrd JC, Harrington B, O’Brien S, et al. Acalabrutinib (ACP-196) in relapsed chronic lymphocytic leukemia. N Engl J Med. 2016;374(4):323-332.
10. Levade M, David E, Garcia C, et al. Ibrutinib treatment affects collagen and von Willebrand factor-dependent platelet func- tions. Blood. 2014;124(26):3991-3995.
11.
12.
13.
14.
15.
16.
17.
18.
19. 20.
Kamel S, Horton L, Ysebaert L, et al. Ibrutinib inhibits collagen-mediated but not ADP-mediated platelet aggregation. Leukemia. 2015;29(4):783-787.
Bye AP, Unsworth AJ, Vaiyapuri S, Stainer AR, Fry MJ, Gibbins JM. Ibrutinib inhibits platelet integrin αIIbβ3 outside-in signaling and thrombus stability but not adhesion to collagen. Arterioscler Thromb Vasc Biol. 2015;35(11):2326-2335.
Rigg RA, Aslan JE, Healy LD, et al. Oral administration of Bruton's tyrosine kinase inhibitors impairs GPVI-mediated platelet function. Am J Physiol Cell Physiol. 2016;310(5):C373-380.
Quek LS, Bolen J, Watson SP. A role for Bruton’s tyrosine kinase (Btk) in platelet acti- vation by collagen. Cur Biol. 1998;8(20): 1137-1140.
Atkinson BT, Ellmeier W, Watson SP. Tec regulates platelet activation by GPVI in the absence of Btk. Blood. 2003;102(10):3592- 3599.
Liu J, Fitzgerald ME, Berndt MC, Jackson CW, Gartner TK. Bruton tyrosine kinase is essential for botrocetin/VWF-induced sig- naling and GPIb-dependent thrombus for- mation in vivo. Blood. 2006;108(8):2596- 2603.
Futatani T, Watanabe C, Baba Y, Tsukada S, Ochs HD. Bruton’s tyrosine kinase is present in normal platelets and its absence identifies patients X-linked agammaglobulinaemia and carrier females. Br J Haematol. 2001;114(1):141-149.
Winkelstein JA, Marino MC, Lederman HM, et al. X-linked agammaglobulinemia: report on a United States registry of 201 patients. Medicine. 2006;85(4):193-202.
Wu J, Zhang M, Liu D. Acalabrutinib (ACP- 196): a selective second-generation BTK inhibitor. J Hematol Oncol. 2016;9:21.
Barf T, Covey T, Izumi R, et al. Acalabrutinib (ACP-196): a covalent bruton tyrosine kinase inhibitor with a differentiat-
ed selectivity and in vivo potency profile. J
Pharmacol Exp Ther. 2017;363(2):240-252. 21. Chen J, Kinoshita T, Gururaja T, et al. The effect of Bruton’s tyrosine kinase (BTK) inhibitors on collagen-induced platelet aggregation, BTK, and tyrosine kinase expressed in hepatocellular carcinoma (TEC). Eur J Haematol. 2018 Jun 20. [Epub
ahead of print]
22. Senis YA, Mazharian A, Mori J. Src family
kinases: at the forefront of platelet activa-
tion. Blood. 2014;124(13):2013-2024.
23. Gratacap MP, Martin V, Valéra MC, et al. The new tyrosine-kinase inhibitor and anti- cancer drug dasatinib reversibly affects platelet activation in vitro and in vivo.
Blood. 2009;114(9):1884-1892.
24. Bye AP, Unsworth AJ, Desborough MJ, et al.
Severe platelet dysfunction in NHL patients receiving ibrutinib is absent in patients receiving acalabrutinib. Blood Adv. 2017;1 (26):2610-2623.
25. Busygina K, Jamasbi J, Seiler T, et al. Oral Bruton tyrosine kinase inhibitors selectively block atherosclerotic plaque-triggered thrombus formation. Blood. 2018;131(24): 2605-2616.
26. Laurent PA, Séverin S, Hechler B, et al. Platelet PI3kbeta and GSK3 regulate throm- bus stability at a high shear rate. Blood. 2015;125(5):881-888.
27. Nicolson PLR, Hughes CE, Watson S, et al. Inhibition of Btk-specific concentrations of ibrutinib and acalabrutinib delays but does not block platelet aggregation to GPVI. Haematologica. 2018;103(12):2097-2108.
28. Aguilar C. Ibrutinib-related bleeding: patho- genesis, clinical implications and manage- ment. Blood Coagul Fibrinolysis. 2018;29(6):481-487.
29. Mato AR, Nabhan C, Thompson MC, et al. Toxicities and outcomes of 616 ibrutinib- treated patients in the united states: a real- world analysis. Haematologica. 2018;103(5): 874-879.
haematologica | 2019; 104(11)
2299


































































































   191   192   193   194   195