A RANDOMIZED PHASE 3 TRIAL OF ZANUBRUTINIB VERSUS IBRUTINIB IN SYMPTOMATIC WALDENSTRÖM MACROGLOBULINEMIA:THE ASPEN STUDY.

Bruton tyrosine kinase (BTK) inhibition is an effective treatment approach for patients with Waldenström macroglobulinemia (WM). The phase 3 ASPEN study compared the efficacy and safety of ibrutinib, a first-generation BTK inhibitor, with zanubrutinib, a novel, highly selective BTK inhibitor, in patients with WM. Patients with MYD88L265P disease were randomly assigned 1:1 to treatment with either ibrutinib or zanubrutinib. The primary endpoint was the proportion of patients achieving a complete or very good partial response (CR or VGPR) by independent review. Key secondary endpoints included major response rate (MRR), progression-free survival (PFS), duration of response (DOR), disease burden, and safety. A total of 201 patients were randomized, and 199 received ≥1 dose of study treatment. No patient achieved a CR. Twenty-nine (28%) zanubrutinib and 19 (19%) ibrutinib patients achieved a VGPR, a non-statistically significant difference (P = .09). MRRs were 77% and 78% , respectively. Median DOR and PFS were not reached; 84% and 85% of ibrutinib and zanubrutinib patients were progression-free at 18 months. Incidence of atrial fibrillation, contusion, diarrhea, peripheral edema, hemorrhage, muscle spasms, and pneumonia, as well as adverse events leading to treatment discontinuation, were all lower among zanubrutinib recipients. Incidence of neutropenia was higher with zanubrutinib, although grade ≥3 infection rates were similar in both arms (1.2 and 1.1 events/100 person-months). These results demonstrate that zanubrutinib and ibrutinib are highly effective in the treatment of WM, but zanubrutinib treatment was associated with a trend toward better response quality and less toxicity, particularly cardiovascular toxicity.

[1]  W. Novotny,et al.  Three-year follow-up of treatment-naïve and previously treated patients with Waldenström macroglobulinemia (WM) receiving single-agent zanubrutinib. , 2020 .

[2]  Sun Ku Lee,et al.  Acalabrutinib monotherapy in patients with Waldenström macroglobulinemia: a single-arm, multicentre, phase 2 study. , 2019, The Lancet. Haematology.

[3]  J. Byrd,et al.  Hypertension and Incident Cardiovascular Events Following Ibrutinib Initiation. , 2019, Blood.

[4]  R. Elstrom,et al.  Phase 1 study of selective BTK inhibitor zanubrutinib in B-cell malignancies and safety and efficacy evaluation in CLL. , 2019, Blood.

[5]  G. Keller,et al.  Ibrutinib Displays Atrial-Specific Toxicity in Human Stem Cell-Derived Cardiomyocytes , 2019, Stem cell reports.

[6]  M. Gertz Waldenström macroglobulinemia: 2019 update on diagnosis, risk stratification, and management , 2018, American journal of hematology.

[7]  S. Treon,et al.  Ibrutinib Monotherapy in Symptomatic, Treatment-Naïve Patients With Waldenström Macroglobulinemia. , 2018, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[8]  C. Tam,et al.  Cardiac side effects of bruton tyrosine kinase (BTK) inhibitors , 2018, Leukemia and Lymphoma.

[9]  M. Dimopoulos,et al.  Phase 3 Trial of Ibrutinib plus Rituximab in Waldenström's Macroglobulinemia , 2018, The New England journal of medicine.

[10]  R. Hendriks,et al.  Role of Bruton’s tyrosine kinase in B cells and malignancies , 2018, Molecular Cancer.

[11]  R. Advani,et al.  Long-Term Follow-up of Previously Treated Patients Who Received Ibrutinib for Symptomatic Waldenstrom's Macroglobulinemia: Update of Pivotal Clinical Trial , 2017 .

[12]  A. Kater,et al.  Incidence and management of toxicity associated with ibrutinib and idelalisib: a practical approach , 2017, Haematologica.

[13]  J. Byrd,et al.  Characterization of atrial fibrillation adverse events reported in ibrutinib randomized controlled registration trials , 2017, Haematologica.

[14]  O. McCarty,et al.  Ibrutinib‐associated bleeding: pathogenesis, management and risk reduction strategies , 2017, Journal of thrombosis and haemostasis : JTH.

[15]  M. Dimopoulos,et al.  Ibrutinib for patients with rituximab-refractory Waldenström's macroglobulinaemia (iNNOVATE): an open-label substudy of an international, multicentre, phase 3 trial. , 2017, The Lancet. Oncology.

[16]  R. García-Sanz WM, MYD88, and CXCR4: following the thread. , 2016, Blood.

[17]  Wei Zhang,et al.  HCK is a survival determinant transactivated by mutated MYD88, and a direct target of ibrutinib. , 2016, Blood.

[18]  S. Treon,et al.  Atrial fibrillation associated with ibrutinib in Waldenström macroglobulinemia , 2016, American journal of hematology.

[19]  R. Advani,et al.  Clonal architecture of CXCR4 WHIM‐like mutations in Waldenström Macroglobulinaemia , 2016, British journal of haematology.

[20]  Lei Lei,et al.  A Simple Method for Estimating Confidence Intervals for Exposur e Adjusted Incidence Rate and Its Applications to Clinical Trials , 2015 .

[21]  R. Advani,et al.  Ibrutinib in previously treated Waldenström's macroglobulinemia. , 2015, The New England journal of medicine.

[22]  S. Treon,et al.  CXCR4 WHIM‐like frameshift and nonsense mutations promote ibrutinib resistance but do not supplant MYD88L265P‐directed survival signalling in Waldenström macroglobulinaemia cells , 2015, British journal of haematology.

[23]  C. Tam,et al.  Ibrutinib treatment affects collagen and von Willebrand factor-dependent platelet functions. , 2014, Blood.

[24]  M. Keating,et al.  Ibrutinib inhibits collagen-mediated but not ADP-mediated platelet aggregation , 2014, Leukemia.

[25]  A. Roccaro,et al.  C1013G/CXCR4 acts as a driver mutation of tumor progression and modulator of drug resistance in lymphoplasmacytic lymphoma. , 2014, Blood.

[26]  S. Rodig,et al.  The WHIM-like CXCR4S338X somatic mutation activates AKT and ERK, and promotes resistance to ibrutinib and other agents used in the treatment of Waldenstrom’s Macroglobulinemia , 2014, Leukemia.

[27]  S. Treon,et al.  The genomic landscape of Waldenstrom macroglobulinemia is characterized by highly recurring MYD88 and WHIM-like CXCR4 mutations, and small somatic deletions associated with B-cell lymphomagenesis. , 2014, Blood.

[28]  R. Advani,et al.  Treatment recommendations for patients with Waldenström's Macroglobulinemia (WM) and related disorders: consensus from the Seventh International Workshop on WM , 2014 .

[29]  E. Kimby,et al.  Response assessment in Waldenström macroglobulinaemia: update from the VIth International Workshop , 2013, British journal of haematology.

[30]  N. Harris,et al.  MYD88 L265P somatic mutation in Waldenström's macroglobulinemia. , 2012, The New England journal of medicine.

[31]  Hsiuyi Tseng,et al.  Attainment of complete/very good partial response following rituximab‐based therapy is an important determinant to progression‐free survival, and is impacted by polymorphisms in FCGR3A in Waldenstrom macroglobulinaemia , 2011, British journal of haematology.

[32]  J. Crowley,et al.  International prognostic scoring system for Waldenstrom macroglobulinemia. , 2009, Blood.

[33]  Tosiya Sato,et al.  On the variance estimator for the Mantel-Haenszel risk difference , 1989 .

[34]  Ron Brookmeyer,et al.  A Confidence Interval for the Median Survival Time , 1982 .

[35]  W. Haenszel,et al.  Statistical aspects of the analysis of data from retrospective studies of disease. , 1959, Journal of the National Cancer Institute.

[36]  E. Kaplan,et al.  Nonparametric Estimation from Incomplete Observations , 1958 .

[37]  M. Greenwood A Report on the Natural Duration of Cancer. , 1926 .