#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Resistance-associated mutations in chronic lymphocytic leukaemia patients treated with ibrutinib, idelalisib and venetoclax


Authors: L. Sedlaříková 1;  T. Papajík 2;  A. Petráčková 1;  P. Turcsányi 2;  E. Kriegová 1
Authors‘ workplace: Ústav imunologie, Lékařská fakulta Univerzity Palackého a Fakultní nemocnice, Olomouc 1;  Hemato-onkologická klinika, Lékařská fakulta Univerzity Palackého a Fakultní nemocnice, Olomouc 2
Published in: Transfuze Hematol. dnes,26, 2020, No. 1, p. 46-54.
Category: Review/Educational Papers

Overview

Targeted BCR signalling pathway inhibitors (ibrutinib, idelalisib) and BCL2 inhibitors (venetoclax) appear promising in patients with chronic lymphocytic leukaemia (CLL), as they have shown significant clinical efficacy in relapsed/refractory high-risk patients with deletion 17p and/or TP53 mutation, as well as in first-line treatment. Nevertheless, there is a growing body of evidence regarding the emergence of mutations leading to clinical resistance during treatment. So far, most information has been reported in patients treated with ibrutinib (an irreversible inhibitor of Bruton’s tyrosine kinase, BTK), where resistance-associated mutations have been found in the BTK and PLCG2 genes. For idelalisib (an inhibitor of the δ isoform of phosphatidylinositol 3-kinase, PI3K), no resistance-associated mutation and/or aberrant signalling pathway have been reported, but a number of various genetic aberrations associated with CLL have been found in relapsing patients. Recently, mutations in the BCL2 gene have been reported in resistant patients treated with venetoclax (BCL2 inhibitor). In most patients who progressed on novel agents, the mutations associated with resistance occurred at low allelic frequencies between the second and fourth years of therapy. Frequently, several subclones are present which can be detected several months prior to clinical relapse. In this review, we report state-of-the-art data about known gene variants associated with resistance to ibrutinib, idelalisib and venetoclax, and the latest findings of their clinical impact. Nevertheless, recent findings suggest that there are also alternative mechanisms of acquired resistance to treatment with these inhibitors, which will certainly become the subject of further studies. We also discuss recent findings in the development of next-generation pathway inhibitors for the treatment of CLL patients who have acquired resistant mutations to current inhibitors.

Keywords:

chronic lymphocytic leukaemia – targeted therapy – signalling pathway inhibitors – mutations associated with resistance – resistance to treatment


Sources
  1. Byrd JC, Furman RR, Coutre SE, et al. Targeting BTK with ibrutinib in relapsed chronic lymphocytic leukemia. N Engl J Med. 2013;369:32–42.
  2. Byrd JC, Hillmen P, O‘Brien S, et al. Long-term follow-up of the RESONATE phase 3 trial of ibrutinib vs ofatumumab. Blood. 2019;133(19):2031–2042.
  3. Burger JA, Tedeschi A, Barr PM, et al. Ibrutinib as initial therapy for patients with chronic lymphocytic leukemia. N Engl J Med. 2015;373(25):2425–2437.
  4. Barr PM, Robak T, Owen C, et al. Sustained efficacy and detailed clinical follow-up of first-line ibrutinib treatment in older patients with chronic lymphocytic leukemia: extended phase 3 results from RESONATE-2. Haematologica. 2018;103(9):1502–1510.
  5. Woyach JA, Furman RR, Liu TM, et al. Resistance mechanisms for the Bruton‘s tyrosine kinase inhibitor ibrutinib. N Engl J Med. 2014;370(24):2286–2294.
  6. Cheng S, Guo A, Lu P, Ma J, Coleman M, Wang YL. Functional characterization of BTK(C481S) mutation that confers ibrutinib resistance: exploration of alternative kinase inhibitors. Leukemia. 2015;29(4):895–900.
  7. Hamasy A, Wang Q, Blomberg KE, et al. Substitution scanning identifies a novel, catalytically active ibrutinib-resistant BTK cysteine 481 to threonine (C481T) variant. Leukemia. 2017;31(1):177–185.
  8. Ahn IE, Underbayev C, Albitar A et al. Clonal evolution leading to ibrutinib resistance in chronic lymphocytic leukemia. Blood. 2017;129(11):1469–1479.
  9. Woyach JA, Ruppert AS, Guinn D, et al. BTKC481S-mediated resistance to ibrutinib in chronic lymphocytic leukemia. J Clin Oncol. 2017;35(13):1437–1443.
  10. Kadri S, Lee J, Fitzpatrick C, et al. Clonal evolution underlying leukemia progression and Richter transformation in patients with ibrutinib-relapsed CLL. Blood Adv. 2017;1(12):715–727.
  11. Jones D, Woyach JA, Zhao W, et al. PLCG2 C2 domain mutations co-occur with BTK and PLCG2 resistance mutations in chronic lymphocytic leukemia undergoing ibrutinib treatment. Leukemia. 2017;31(7):1645–1647.
  12. Maddocks KJ, Ruppert AS, Lozanski G, et al. Etiology of ibrutinib therapy discontinuation and outcomes in patients with chronic lymphocytic leukemia. JAMA Oncol. 2015;1(1):80–87.
  13. Sharma S, Galanina N, Guo A, et al. Identification of a structurally novel BTK mutation that drives ibrutinib resistance in CLL. Oncotarget. 2016;7(42):68833–68841.
  14. Quinquenel A, Fornecker LM, Letestu R, et al. Prevalence of BTK and PLCG2 mutations in a real-life CLL cohort still on ibrutinib after 3 years: a FILO group study. Blood. 2019;134(7):641–644.
  15. Liu TM, Woyach JA, Zhong Y, et al. Hypermorphic mutation of phospholipase Cγ2 acquired in ibrutinib-resistant CLL confers BTK independency upon B-cell receptor activation. Blood. 2015;126(1):61–68.
  16. Burger JA, Landau DA, Taylor-Weiner A, et al. Clonal evolution in patients with chronic lymphocytic leukaemia developing resistance to BTK inhibition. Nat Commun. 2016;7:11589.
  17. Maffei R, Fiorcari S, Martinelli S, et al. Targeting neoplastic B cells and harnessing microenvironment: the „double face“ of ibrutinib and idelalisib. J Hematol Oncol. 2015;8:60.
  18. Lampson BL, Brown JR. Are BTK and PLCG2 mutations necessary and sufficient for ibrutinib resistance in chronic lymphocytic leukemia? Expert Rev Hematol. 2018;11(3):185–194.
  19. Hoellenriegel J, Meadows SA, Sivina M, et al. The phosphoinositide 3‘-kinase delta inhibitor, CAL-101, inhibits B-cell receptor signaling and chemokine networks in chronic lymphocytic leukemia. Blood. 2011;118(13):3603–3612.
  20.  Brown JR, Byrd JC, Coutre SE, et al. Idelalisib, an inhibitor of phosphatidylinositol 3-kinase p110δ, for relapsed/refractory chronic lymphocytic leukemia. Blood. 2014;123(22):3390–3397.
  21. Burger JA, Wiestner A. Targeting B cell receptor signalling in cancer: preclinical and clinical advances. Nat Rev Cancer. 2018;18(3):148–167.
  22. Arnason JE, Brown JR. Targeting B cell signaling in chronic lymphocytic leukemia. Curr Oncol Rep. 2017;19(9):61.
  23. Scheffold A, Jebaraj BMC, Tausch E, et al. IGF1R as druggable target mediating PI3K-δ inhibitor resistance in a murine model of chronic lymphocytic leukemia. Blood. 2019; 134(6):534–547.
  24. Ghia P, Ljungström V, Tausch E, et al. Whole-exome sequencing revealed no recurrent mutations within the PI3K pathway in relapsed chronic lymphocytic leukemia patients progressing under idelalisib treatment. Blood. 2016;128:2770.
  25. Stilgenbauer S, Eichhorst B, Schetelig J, et al. Venetoclax in relapsed or refractory chronic lymphocytic leukaemia with 17p deletion: a multicentre, open-label, phase 2 study. Lancet Oncol. 2016;17:768–778.
  26. Jones JA, Mato AR, Wierda WG, et al. Venetoclax for chronic lymphocytic leukaemia progressing after ibrutinib: an interim analysis of a multicentre, open-label, phase 2 trial. Lancet Oncol. 2018; 19(1):65–75.
  27. Roberts AW, Davids MS, Pagel JM, et al. Targeting BCL2 with venetoclax in relapsed chronic lymphocytic leukemia. N Engl J Med. 2016;374:311–322.
  28. Blombery P, Anderson MA, Gong JN, et al. Acquisition of the recurrent Gly101Val mutation in BCL2 confers resistance to venetoclax in patients with progressive chronic lymphocytic leukemia. Cancer Discov. 2019;9(3):342–353.
  29. Tausch E, Close W, Dolnik A, et al. Venetoclax resistance and acquired BCL2 mutations in chronic lymphocytic leukemia. Haematologica. 2019;104(9):e434–e437.
  30. Herling CD, Abedpour N, Weiss J, et al. Clonal dynamics towards the development of venetoclax resistance in chronic lymphocytic leukemia. Nat Commun. 2018;9(1):727.
  31. Jennings LJ, Arcila ME, Corless C, et al. Guidelines for validation of next-generation sequencing-based oncology panels: a joint consensus recommendation of the Association for Molecular Pathology and College of American Pathologists. J Mol Diagn. 2017;19(3):341–365.
  32. Bacher U, Shumilov E, Flach J, et al. Challenges in the introduction of next-generation sequencing (NGS) for diagnostics of myeloid malignancies into clinical routine use. Blood Cancer J. 2018;8:113.
  33. Merker JD, Devereaux K, Iafrate AJ, et al. Proficiency testing of standardized samples shows very high interlaboratory agreement for clinical nextgeneration sequencing-based oncology assays Arch Pathol Lab Med. 2019;143:463–471.
  34. Petrackova A, Vasinek M, Sedlarikova L, et al. Standardisation of sequencing coverage depth in NGS: recommendation for detection of clonal and subclonal mutations in cancer diagnostics. Front Oncol. 2019;9:851.
  35. Sutton LA. Mechanisms of resistance to targeted therapies in chronic lymphocytic leukemia. HemaSphere. 2019;3(S2):p40–p43.
  36. Fürstenau M, Hallek M, Eichhorst B. Sequential and combination treatments with novel agents in chronic lymphocytic leukemia. Haematologica. 2019;104(11):2144–2154.
  37. Deng J, Isik E, Fernandes SM, et al. Bruton‘s tyrosine kinase inhibition increases BCL-2 dependence and enhances sensitivity to venetoclax in chronic lymphocytic leukemia. Leukemia. 2017;31(10):2075–2084.
  38. Bose P, Gandhi V. Recent therapeutic advances in chronic lymphocytic leukemia. F1000Res. 2017;6:1924.
  39. 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.
  40. Walter HS, Rule SA, Dyer MJ, et al. A phase 1 clinical trial of the selective BTK inhibitor ONO/GS-4059 in relapsed and refractory mature B-cell malignancies. Blood. 2016;127(4):411–419.
  41. Thompson PA, Burger JA. Bruton‘s tyrosine kinase inhibitors: first and second generation agents for patients with Chronic Lymphocytic Leukemia (CLL). Expert Opin Investig Drugs. 2018;27(1):31–42.
  42. Crawford JJ, Johnson AR, Misner DL, et al. Discovery of GDC-0853: a potent, selective, and noncovalent bruton‘s tyrosine kinase inhibitor in early clinical development. J Med Chem. 2018;61(6):2227–2245.
  43. Reiff SD, Muhowski EM, Guinn D, et al. Noncovalent inhibition of C481S Bruton tyrosine kinase by GDC-0853: a new treatment strategy for ibrutinib-resistant CLL. Blood. 2018;132(10):1039–1049.
  44. Wang S, Mondal S, Zhao C, et al. Noncovalent inhibitors reveal BTK gatekeeper and auto-inhibitory residues that control its transforming activity. JCI Insight. 2019;4(12):pii:127566.
  45. Woyach JA. Patterns of resistance to B cell-receptor pathway antagonists in chronic lymphocytic leukemia and strategies for management. Hematology Am Soc Hematol Educ Program. 2015;2015:355–360.
Labels
Haematology Internal medicine Clinical oncology

Article was published in

Transfusion and Haematology Today

Issue 1

2020 Issue 1

Most read in this issue
Login
Forgotten password

Enter the email address that you registered with. We will send you instructions on how to set a new password.

Login

Don‘t have an account?  Create new account

#ADS_BOTTOM_SCRIPTS#