Safety and tolerability of Bruton's tyrosine kinase inhibitors in the treatment of Waldenström macroglobulinemia

Authors

  • Anthea Peters, MD, FRCPC

DOI:

https://doi.org/10.58931/cht.2022.1S0628

Abstract

Waldenström macroglobulinemia (WM) is an indolent B-cell non-Hodgkin lymphoma (NHL) characterized by malignant B cells that produce IgM monoclonal protein. Like other indolent B-cell NHLs, treatment is indicated when patients are symptomatic with lymphadenopathy, splenomegaly or have detrimental cytopenias, but uniquely hyperviscosity and other complications related to gammopathy may present a need for treatment. Currently in Canada chemoimmunotherapy using bendamustine and rituximab (BR) is the favored therapeutic combination for treatment-naïve patients with WM due to a superior efficacy and toxicity profile compared to rituximab plus CHOP and a fixed duration schedule. The availability of Bruton’s tyrosine kinase inhibitors (BTKi) have transformed the treatment landscape for patients with WM, particularly in the relapsed setting. Ibrutinib, a once-daily BTKi, was approved by Health Canada (HC) for WM in 2016 based on two non-randomized studies showing high response rates in heavily pretreated rituximab-refractory patients with sustained efficacy (86% progression-free survival (PFS) at 18 months) and acceptable tolerability. Ibrutinib forms an irreversible covalent bond to the cysteine residue (C481) at the active binding site of BTK. Patients with mutated MYD88 (MYD88MUT), who represent over 90% of patients with WM, have a higher rate of response with ibrutinib than those without (MYD88WT). Aside from its impressive efficacy, its oral administration offers a major advantage in terms of convenience for patients and lower administrative costs for publicly funded health care systems such as in Canada.

The toxicity of ibrutinib has become of greater concern with the ongoing emergence of trial data and real-world clinical experience with the drug. The indefinite duration of treatment results in extended exposure of a primarily elderly patient population to toxicities such as increased bleeding risk, atrial fibrillation, hypertension, and infection. Discontinuation of ibrutinib due to adverse events occurs at a higher rate in the real-world compared to clinical trials and has been associated with an inferior overall survival in WM.

Fortunately, several BTKis have recently emerged. Both acalabrutinib and zanubrutinib also bind irreversibly to BTK at C481, but both have more selectivity for targeting BTK than ibrutinib. Data has demonstrated that tighter and more selective binding of BTK does translate into fewer adverse effects for patients. The most informative study to date in this regard is the randomized phase 3 clinical trial comparing ibrutinib to zanubrutinib, the ASPEN study. ASPEN is the largest multicentre phase 3 trial to date randomizing 201 patients with MYD88MUT WM to receive ibrutinib at 420 mg daily (n=99) vs. zanubrutinib 160 mg twice daily (n=102), with 37 patients being treatment-naïve and 164 being relapsed or refractory (R/R) with a median of 1 prior line of therapy (range 1-8). The median age at enrolment was 70 however more patients randomized to zanubrutinib than to ibrutinib were >75 years old (33% vs 22%, respectively). Although not statistically significant, a higher rate of complete response (CR)/very good partial response (VGPR) was observed for zanubrutinib vs ibrutinib (28% vs 19%, respectively). With a median follow up of 19.4 months the major response rates (77% vs. 78%) and PFS (85% vs. 84% at 18 months) were also not statistically different in patients receiving zanubrutinib vs. ibrutinib, respectively. However, zanubrutinib was associated with a trend towards less toxicity. In particular, the incidences of all grade atrial fibrillation, diarrhea, bruising, muscle spasms, peripheral edema, and pneumonia were over 10% higher among ibrutinib-treated patients. The incidence of neutropenia was higher for zanubrutinib-treated patients (> 10% difference), although grade ≥3 infection rates were similar in both arms (1.2 and 1.1 events per 100 person-months). Further, more ibrutinib-treated patients required dose reductions (23% vs. 14%) and discontinued treatment (9% vs. 4%) due to adverse events. An analysis of quality of life (QoL) instruments demonstrated a trend towards greater improvement in the zanubrutinib arm, particularly in the subgroup of patients who achieved VGPR. These QoL improvements were most notable in validated instrument subscales encompassing appetite, dyspnea, fatigue, physical functioning and role functioning. The ASPEN trial formed the basis for recent approval of zanubrutinib for WM by HC. Although provincial funding is not yet in place, clinicians in Quebec can access zanubrutinib through the Patient d’exception mechanism.

As Canadian hematologists will be able to choose a BTKi for their patient with R/R WM, it is important to review the toxicities reported in trials. Toxicities of BTKis and their management will be discussed in this article.

Author Biography

Anthea Peters, MD, FRCPC

Dr. Anthea Peters is a hematologist based at the Cross Cancer Institute in Edmonton, Alberta and an Associate Professor in the Department of Oncology at the University of Alberta. She completed her MD at the University of Saskatchewan, internal medicine residency training at the University of Alberta and hematology training as well as a lymphoma fellowship at the University of Calgary. Her clinical and research interests are centered around lymphoma and chronic lymphocytic leukemia (CLL), with a special interest in post-transplant lymphoproliferative disorders. She is currently the clinical lead for lymphoma and CLL for Northern Alberta.

References

Castillo JJ, Advani RH, Branagan AR, Buske C, Dimopoulos MA, D’Sa S, et al. Consensus treatment recommendations from the tenth International Workshop for Waldenstrom Macroglobulinaemia. Lancet Haematol. 2020;7(11):e827-e37.

Rummel MJ, Niederle N, Maschmeyer G, Banat GA, von Grunhagen U, Losem C, et al. Bendamustine plus rituximab versus CHOP plus rituximab as first-line treatment for patients with indolent and mantle-cell lymphomas: an open-label, multicentre, randomised, phase 3 non-inferiority trial. Lancet. 2013;381(9873):1203-10.

Larose F, Chen, C.I. Bruton’s Tyrosine Kinase Inhibitors for the Treatment of Waldenstrom’s Macroglobulinemia: A Canadian Perspective. touchREVIEWS in Oncology & Haematology. 2021;17(2).

Treon SP, Tripsas CK, Meid K, Warren D, Varma G, Green R, et al. Ibrutinib in previously treated Waldenstrom’s macroglobulinemia. N Engl J Med. 2015;372(15):1430-40.

Dimopoulos MA, Trotman J, Tedeschi A, Matous JV, Macdonald D, Tam C, et al. Ibrutinib for patients with rituximab-refractory Waldenstrom’s macroglobulinaemia (iNNOVATE): an open-label substudy of an international, multicentre, phase 3 trial. Lancet Oncol. 2017;18(2):241-50.

Lipsky A, Lamanna N. Managing toxicities of Bruton tyrosine kinase inhibitors. Hematology Am Soc Hematol Educ Program. 2020;2020(1):336-45.

Dimopoulos MA, Tedeschi A, Trotman J, Garcia-Sanz R, Macdonald D, Leblond V, et al. Phase 3 Trial of Ibrutinib plus Rituximab in Waldenstrom’s Macroglobulinemia. N Engl J Med. 2018;378(25):2399-410.

Treon SP, Xu L, Yang G, Zhou Y, Liu X, Cao Y, et al. MYD88 L265P somatic mutation in Waldenstrom’s macroglobulinemia. N Engl J Med. 2012;367(9):826-33.

Gustine JN, Meid K, Dubeau T, Severns P, Hunter ZR, Guang Y, et al. Ibrutinib discontinuation in Waldenstrom macroglobulinemia: Etiologies, outcomes, and IgM rebound. Am J Hematol. 2018;93(4):511-7.

Tam CS, Opat S, D’Sa S, Jurczak W, Lee HP, Cull G, et al. A randomized phase 3 trial of zanubrutinib vs ibrutinib in symptomatic Waldenstrom macroglobulinemia: the ASPEN study. Blood. 2020;136(18):2038-50.

Trotman J, Opat S, Gottlieb D, Simpson D, Marlton P, Cull G, et al. Zanubrutinib for the treatment of patients with Waldenstrom macroglobulinemia: 3 years of follow-up. Blood. 2020;136(18):2027-37.

Owen RG, McCarthy H, Rule S, D’Sa S, Thomas SK, Tournilhac O, et al. Acalabrutinib monotherapy in patients with Waldenstrom macroglobulinemia: a single-arm, multicentre, phase 2 study. Lancet Haematol. 2020;7(2):e112-e21.

Treon SP, Meid K, Gustine J, Yang G, Xu L, Liu X, et al. Long-Term Follow-Up of Ibrutinib Monotherapy in Symptomatic, Previously Treated Patients With Waldenstrom Macroglobulinemia. J Clin Oncol. 2021;39(6):565-75.

Gustine JN, Meid K, Dubeau TE, Treon SP, Castillo JJ. Atrial fibrillation associated with ibrutinib in Waldenstrom macroglobulinemia. Am J Hematol. 2016;91(6):E312-3.

Abeykoon JP, Zanwar S, Ansell SM, Gertz MA, Kumar S, Manske M, et al. Ibrutinib monotherapy outside of clinical trial setting in Waldenstrom macroglobulinaemia: practice patterns, toxicities and outcomes. Br J Haematol. 2020;188(3):394-403.

Published

2022-06-15

How to Cite

1.
Peters A. Safety and tolerability of Bruton’s tyrosine kinase inhibitors in the treatment of Waldenström macroglobulinemia. Can Hematol Today [Internet]. 2022 Jun. 15 [cited 2024 Apr. 24];1(S06). Available from: https://canadianhematologytoday.com/article/view/28

Issue

Section

Articles