Abstract
The current therapeutic approach in Waldenström’s macroglobulinemia (WM) is being driven by insights in disease biology and genomic landscape. Bruton’s tyrosine kinase (BTK) plays a key role in signaling pathways for the survival of WM clone. BTK inhibition has changed the treatment landscape of the disease. Ibrutinib has resulted in deep and durable responses both as an upfront and salvage treatment with a manageable toxicity profile. However, the need for fewer off-target effects and deeper responses has resulted in the clinical development of second-generation BTK inhibitors. Zanubrutinib has resulted in clinically meaningful antitumor activity, including deep and durable responses, with a low discontinuation rate due to treatment-related toxicities. Cardiovascular adverse events seem to be milder compared with ibrutinib. Interestingly, the efficacy of zanubrutinib in WM is significant both for MYD88L265P and MYD88WT patients. Although the randomized, phase III ASPEN clinical trial did not meet its primary endpoint in terms of showing a superiority of zanubrutinib in deep responses compared with ibrutinib, secondary efficacy and safety endpoints underscore the potential clinical role of zanubrutinib in the treatment algorithm of WM independent of the MYD88 mutational status. Combination regimens and non-covalent BTK inhibitors are emerging as promising treatment strategies. Long-term data will determine whether next-generation BTK inhibitors are more potent and safer compared with ibrutinib, and whether they are able to overcome resistance to ibrutinib, either alone or in combination with inhibitors of other interrelated molecular pathways.
Keywords: Bruton’s tyrosine kinase, ibrutinib, MYD88, Waldenstrom’s macroglobulinemia, zanubrutinib
Introduction
Waldenström’s macroglobulinemia (WM) is an indolent B-cell lymphoma characterized by lymphoplasmacytic cell infiltration of the bone marrow and/or lymphatic tissue, along with secretion of monoclonal immunoglobulin M (IgM) in the serum. WM is a rare malignancy accounting for 1–2% of all hematological cancers.1–5 The disease incidence is estimated approximately at five cases per 1 million person-years.6 It increases with age,7,8 and there is a higher prevalence among White males. Interestingly, a significant familial predisposition has been also reported.9,10
Any lymphoplasmacytic infiltration is sufficient for the diagnosis of WM,1,11 whereas a lower BM clonal cell infiltration is associated with a more indolent disease course.12,13 Risk stratification in symptomatic WM patients is based on the International Prognostic Scoring System for WM. A three-level risk category is defined based on age, b2-microglobulin, hemoglobin, platelet, and IgM levels.14,15
Compared with healthy individuals, patients with WM present with a distinct molecular profile including but not limited to enhanced expression of the VDJ recombination genes DNTT, RAG1, and RAG2, as well as upregulation of genes involved in MYD88 and CXCR4 signaling.16,17 Patterns of aberrant methylation and unique epigenomics signatures have been also described in patients with WM.18
The diverse clinical presentation of patients with WM is attributed to the diverse bone marrow and/or organ infiltration by clonal cells and the immunological and physiochemical properties of monoclonal IgM that are primarily present in each patient. It has to be noted that treatment will not be required at the time diagnosis in up to one third of patients. The median time of asymptomatic patients to acquire features of symptomatic disease has been estimated at 5–10 years.13,19,20 Validated scoring systems are useful in stratifying asymptomatic patients with WM based on risk of progression to symptomatic disease.12 The consensus criteria for treatment initiation should be evaluated in combination with individualized clinical judgment, taking into consideration the disease complexity.21–23
Currently, WM remains incurable with an estimated median overall survival of 10–12 years.24 Taking into consideration the indolent course of the disease and the advanced age of patients, almost half of the patients with WM die of causes unrelated to the disease. Furthermore, estimates of median survival may not reflect the survival benefit of novel therapies that have been introduced in the therapeutic armamentarium during the recent years. Although WM is an orphan disease, there are several treatment options for both newly diagnosed and relapsed WM patients. Chemoimmunotherapy, including the anti-CD20 antibody rituximab in combination with cyclophosphamide, bendamustine, or bortezomib and dexamethasone, has been considered the standard of care for many years. These treatment regimens have high response rates, induce durable responses, and present a manageable toxicity profile.22,25 Novel treatment options are imperative due to either treatment intolerance or disease refractoriness.26,27 In this context, inhibition of Bruton’s tyrosine kinase (BTK) has come to the foreground of research efforts. Ibrutinib, a first-in-class BTK inhibitor, has been the first (and only) agent to receive USA Food and Drug Administration (FDA) and European Medicines Agency (EMA) approval for the treatment of patients with WM. However, acquired resistance and intolerance to ibrutinib have led to the clinical development of next-generation BTK inhibitors.
BTK inhibition in WM
BTK was discovered in 199328 and received its name after Ogden Bruton, a pediatrician who described Bruton’s agammaglobulinemia (X-linked agammaglobulinemia), a primary immunodeficiency syndrome which involves a mutation in BTK.29 BTK is a non-receptor tyrosine kinase, which plays a central role in B-cell signaling and is necessary for normal B-cell development in the bone marrow involved in adaptive and innate receptor-mediated signals. It has an upstream role in the molecular cascade that follows the activation of the B-cell receptor (BCR) and leads to downstream pathway activation of phosphoinositide-3-kinase (P13K)–protein kinase B (AKT) pathway, phospholipase C (PLC), protein kinase C (PKC), and nuclear factor κB (NF-κB). This signaling cascade promotes B-cell differentiation, proliferation, and survival.30–32 BTK also has a role in the signaling of G-coupled chemokine receptors (like CXCR4), cytokine receptors (CD19, CD38 CD40), tumor necrosis family receptors (TNFRs), integrins and toll-like receptors (TLRs), such as TLR/MYD88.33,34
In WM, there is constitutive activation of BTK secondary to multiple mutations, detected by multiple methods in whole bone marrow, CD19+ selected cells, peripheral blood and cell-free deoxyribonucleic acid (DNA).35–39 The first mutation described using whole-genome sequencing of CD19+ bone marrow cells, which is found in up to 90% of WM patients, is the somatic activating mutation of myeloid differentiation factor, MYD88L265P Leu265Pro.40–42 MYD88-activating mutations promote Myddosome self-assembly and trigger TLR activation via BTK interaction and signaling of interleukin 1 (IL-1), IRAK4/IRAK1 and NF-κB.43–45 Around 5–10% patients will have other MYD88 mutations or wild-type MYD88.46 MYD88WT often has mutations in the NF-κB pathway, which are downstream to BTK and therefore show different response patterns to BTK inhibition.47,48 In addition to BTK, MYD88 mutations transactivate another tyrosine kinase, hematopoietic cell kinase (HCK) which is also involved in pro-survival signaling.49 Interestingly, HCK is also found to be a highly relevant target molecule of ibrutinib.49
In 20–40% of patients with WM, the somatic, subclonal, activating mutation in the CXCR4 gene (C-terminal of the C-X-X chemokine receptor type 4) is identified. It is analogous to the germline mutation observed in patients with WHIM (warts, hypogammaglobulinemia, infections, and myelokathexis) syndrome (CXCR4WHIM).50,51 The same patient may harbor different CXCR4 mutations, and this is most likely linked to genomic instability.52,53 The mutations in the C-terminal domain of the CXCR4 receptor lead to a permanently activated state by blocking the internalization of the receptor that normally occurs after SDF-1α stimulation.54 CXCR4 activation promotes AKT kinase and extracellular-regulated kinase (ERK) function, which may be associated with resistance to ibrutinib therapy.55 CXCR4WHIM status is associated with lower responses to BTK inhibition,55,56 which has provided the rationale for the clinical development of anti-CXCR4 monoclonal antibodies, such as ulocuplumab, and small molecules, such as mavorixafor.57,58 Combining BTK and CXCR4 inhibition has resulted in disease responses independent of mutational status in preclinical studies.59
Overall, patients with different MYD88 and CXCR4 mutational status have distinct clinical presentations and sensitivity to BTK inhibition. MYD88L265P/CXCR4MUT patients have higher levels of bone marrow infiltration, and serum IgM and MYD88WT/CXCR4WT have the lowest levels of IgM, bone marrow infiltration and respond less well to BTK inhibition.53,60
Ibrutinib
Ibrutinib is a first-in-class, orally administered BTK inhibitor. It binds irreversibly and covalently with a cysteine residue on site 481 within the binding site of BTK. In several B-cell lymphomas ibrutinib has shown potent and sustained single-agent activity.61 Ibrutinib, like all BTK inhibitors, activates apoptosis, inhibits DNA replication, and blocks pro-survival signaling pathways. It also exerts immunomodulatory effects on macrophages and the tumor microenvironment. It inhibits HCK and causes inactivation of downstream transcription factors including NF-κB, STAT3, and AL-1 and downregulation of cytokines and chemokines.
Ibrutinib is indicated for the treatment of chronic lymphocytic leukemia/small lymphocytic leukemia, marginal zone, and mantle-cell lymphoma.62 It is indicated for the treatment of patients with relapsed/refractory WM, but also for treatment-naïve, newly diagnosed patients with WM. In Europe, ibrutinib is indicated in the first line only for patients who are considered unsuitable for chemoimmunotherapy. It should be administered continuously until disease progression or unacceptable toxicity. MYD88 and CXCR4 testing is recommended before treatment initiation. Drug interruption or dose modifications are required when potent CYP3A inhibitors or inducers are co-administered or in the case of hepatic impairment, due to the fact that ibrutinib is primarily metabolized in the liver by CYP3A.63 Ibrutinib is the only FDA- and EMA-approved drug for WM, which has changed the treatment and outcome landscape for the disease.
Table 1 summarizes the most important clinical data of ibrutinib in patients with WM. Following encouraging initial preliminary results, a phase II trial demonstrated the efficacy of ibrutinib in the relapsed/refractory disease setting.64,65 The median time to first response was 4 weeks and the response rates increased with increased follow up, whereas no IgM flares were reported. The mutational status was a predictor of response. The overall response rates were higher among patients with MYD88L265PCXCR4WT (100%) and lowest for those with MYD88WTCXCR4WT (71%). The progression-free survival (PFS) at 47 months was inferior for MYD88L265PCXCR4WHIM (45 months) compared with patients with MYD88L265PCXCR4WT (not reached) and lowest (21 months) for those with MYD88WTCXCR4WT.66 In addition to the above, a sub-analysis of the phase III iNNOVATE trial confirmed the efficacy of ibrutinib in rituximab refractory patients (n = 31).67 The response rates were similar for MYD88L265PCXCR4WHIM and MYD88L265PCXCR4WT patients, but the time to response was prolonged in the latter group.68 Furthermore, ibrutinib monotherapy showed high efficacy and safety among newly diagnosed patients (n = 30), who were all MYD88L265P, in a prospective phase II study. Patients with CXCR4WHIM had inferior responses compared with those with CXCR4WT.69
Table 1.
Patients (n) | Design/disease setting | Treatment | Outcomes | Comments | |
---|---|---|---|---|---|
Advani et al.64 | 4 RR | Phase I prospective | PO ibrutinib | ORR: 3/4 patients MRR: not reported PFS: not reported OS: not reported |
|
Treon et al.65,66 | 63 RR | Phase II prospective | PO ibrutinib | At 47.1 months: ORR: 90.5% MRR: 73% PFS: 2 years 69.1% OS: 2 years 95.2% Median time to response: 4 weeks |
Response and PFS lower in CXCR4WHIM and MYD88wt
Median PFS at 47.1 months: not reached for MYD88L265P, 45 months for MYD88L265P/CXCR4WHIM, and 21 months for MYD88WT/CXCR4WT |
Dimopoulos et al.68 | 31 RR (rituximab refractory) | Phase III open label | PO ibrutinib | ORR: 90% MRR: 71% PFS: 18 months 86% OS: 18 months 97% |
Response and PFS similar, in MYD88L265P CXCR4WHIM, but slower ORR and MRR 88% for CXCR4WT/MYD88L265P versus 71% for CXCR4WHIM/MYD88L265P |
Dimopoulos et al.70 | 150 (TN and RR) | Phase III double blind, randomized 1:1 | Ibrutinib + rituximab (Ibr/R) versus placebo + rituximab (Pcb/R) | At 30-month FU: ORR: 92% in Ibr/R arm versus 72% in Pcb/R arm MRR: 47% in Ibr/R arm versus 32% in Pcb/R OS: 94% in Ibr/R arm versus 92% in Pcb/R 30-month PFS: 82% in Ibr/R versus 25% in Pcb/R Median PFS: not reached in Ibr/R versus 20.3 months in Pcb/R |
PFS and RR not affected by MYD88/CXCR4 mutation status but major response lower in CXCR4WHIM |
Treon et al.69 | 30 TN MYD88L265P |
Phase II prospective | PO ibrutinib | ORR:100% MRR: 83% PFS: 18 months 92% OS: 18 months 100% |
Median time to response 4 weeks MRR was 94% in WT CXCR4 MRR was 71% in CXCR4 mutated |
MRR, major response rate; ORR, overall response rate; OS, overall survival; PFS, progression-free survival; PO, per os (orally); RR, relapsed/refractory; TN, treatment naïve; WM, Waldenström’s macroglobulinemia; WT, wild type.
iNNOVATE is a double-blind, randomized, placebo-controlled trial that included 150 treatment-naïve and relapsed/refractory patients with WM who were randomized (1:1) to receive either rituximab with placebo or rituximab with ibrutinib. All previously treated patients were rituximab sensitive, whereas 45% had not received any prior treatment. Among the patients receiving ibrutinib–rituximab, ibrutinib prevented the rituximab-induced IgM flare, whereas IgM levels showed a rapid reduction, and hemoglobin level increases were sustainable.70 The addition of ibrutinib to rituximab resulted in a significant PFS benefit both in treatment-naïve and relapsed/refractory patients with WM, independently of the MYD88/CXCR4 mutational status. However, major response rates were lower in patients with CXCR4WHIM. No novel toxicities emerged, whereas the treatment discontinuation rate due to toxicity was similar between the two treatment groups. Furthermore, there are still unanswered questions regarding the role of ibrutinib in the therapeutic algorithm of WM.71 Despite the superiority of ibrutinib–rituximab compared with rituximab, there are no data available regarding the comparison of ibrutinib–rituximab with ibrutinib. Further studies evaluating the efficacy of ibrutinib–rituximab compared to other established regimens for WM both in the upfront and in the relapsed/refractory setting are needed.
Ibrutinib is also an effective treatment option for Bing–Neel syndrome as it crosses the blood–brain barrier into the central nervous system.72 A recent multicenter study enrolled 28 patients with Bing–Neel who received ibrutinib. A total of 85% of patients showed improvement or resolution of their symptoms, and 83% of them showed improvement in the associated abnormalities in the imaging studies, as well.73
Ibrutinib has a favorable toxicity profile; however, it has multiple off-target effects because it is a non-selective agent and inhibits other molecules beyond BTK including epidermal growth-factor receptor (EGFR), ErbB2, proto-oncogene tyrosine-protein kinase (SRC), interleukin-2-inducible T-cell kinase (ITK), tyrosine-protein kinase (TEC), and HCK. Common adverse events are gastrointestinal disturbances, upper respiratory-tract infections, rash, fatigue, and dyspnea.74 Severe neutropenia and thrombocytopenia have been reported in up to 15% of the patients receiving ibrutinib in the abovementioned clinical trials, whereas less than 10% of the patients experienced severe infections, bleeding and arrhythmias, especially atrial fibrillation.75 The arrhythmogenic potential of ibrutinib seems to be mediated by the inhibition of the PI3K–AKT cardioprotective pathway.76–78 Systematic reviews of ibrutinib studies in patients with B-cell malignancies suggest a relative risk for atrial fibrillation and hypertension of 4.69 [95% confidence interval (CI): 2.17–7.64] and 2.82 (95% CI: 1.52–5.23), respectively, whereas cases of life-threatening arrhythmias have been also reported.79,80 Treatment discontinuation due to new onset of atrial fibrillation is not recommended, but patients should receive cardiologic consultation and proper anticoagulation prophylaxis.81 Another pooled analysis of clinical trial data has suggested an increased relative risk of bleeding with ibrutinib (2.93, 95% CI: 1.14–7.52).82–84 Ibrutinib may affect platelet aggregation by interfering with integrin signaling and von Willebrand signaling cascade.85,86 For this reason, perioperative interruption of ibrutinib is advised for 3–7 days. Importantly, the combination of ibrutinib with rituximab did not result in an excess of reported toxities.87 However, the adverse events raise concern in the context of long-term, continuous treatment with ibrutinib.65
Ibrutinib therapy may be interrupted to manage or prevent toxicities; however, 20% of the patients may develop withdrawal symptoms within 2 days, which typically resolve with treatment re-initiation.88 Ibrutinib interruption may induce a rebound effect and a hyperactive immune state similar to cytokine-release syndrome.89–91 IgM rebound may be also observed in up to 80% of patients with WM during the first 2 months following ibrutinib discontinuation.92 Re-initiation of treatment usually restores IgM levels and any related symptoms.93 The optimal strategy of interrupting ibrutinib and assuring persistence of disease response has to be determined in future studies.
Resistance to ibrutinib has been associated with poor prognosis.94 Mutations in the binding site of ibrutinib BTKCys481 have been identified in MYD88 mutated WM cells derived from patients progressing while on ibrutinib therapy. Interestingly, these mutations are mainly subclonal and demonstrate a variable clonal distribution. There has been evidence that these mutations occur de novo during treatment with ibrutinib.95 Sustained ERK 1/2 activation has also emerged as a principal mediator of ibrutinib resistance. The activation of ERK 1/2 pathway may also provide protection from BTK inhibition through a paracrine mechanism to BTK wild-type WM cells.48 In this context, ERK1/2 inhibition may overcome resistance. Furthermore, there have been reports of ibrutinib-resistant WM cell lines that lack BTKCys481 mutation or CXC4WHIM-like mutations, which suggest the existence of BTK-independent survival signals. Whole-exome sequencing of ibrutinib-resistant WM cells has revealed a diverse panel of both BTK-related and BTK-independent genomic abnormalities, including chromosomal deletions affecting regulators of BTK signaling, recurring mutations in ubiquitin ligases, innate immune signaling, TLR/MYD88 pathway regulators, AKT and Bcl-2 associated pathways.96,97
Zanubrutinib
The need for more selective BTK inhibitors in order to minimize the off-target effects and the associated toxicity has led to the clinical development of second-generation BTK inhibitors.98
Zanubrutinib (BGB-3111) is an oral, second-generation BTK inhibitor. It binds irreversibly to the Cys481 residue of the adenosine triphosphate (ATP)-binding pocket of the BTK active site. Importantly, it has minimal off-target activity. It has a half-maximal inhibitory concentration for inhibition of off-target kinases, such as ITK, TEC, EGFR, human epidermal growth-factor receptor 2, and Janus kinase 2, 2- to 70-fold higher than ibrutinib.99–101 Zanubrutinib is currently approved by the FDA for the treatment of relapsed/refractory mantle-cell lymphoma. The recommended dose is 160 mg twice daily.102 Table 2 summarizes the results of the most important clinical trials of zanubrutinib in the treatment of WM.
Table 2.
Patients (n) | Design/disease setting | Treatment | Outcomes | Comments | |
---|---|---|---|---|---|
Trotman et al.102, 2020103; Tam et al.101 | 77 (24 TN; 53 RR) | Phase I/II prospective | PO zanubrutinib | ORR: 96% VGPR/CR: 45.2% 3-year PFS: 80.5% 3-year OS: 84.8% |
VGPR/CR rate increased over time: 20.5% at 6 months, 32.9% at 12 months, and 43.8% at 24 months AEs of interest: contusion (32.5%, grade 1), neutropenia (18.2%), major hemorrhage (3.9%), atrial fibrillation/flutter (5.2%), grade 3 diarrhea (2.6%) |
Tam et al.104; Garcia-Sanz et al.105 | 201 MYD88L265P (cohort 1) 26 MYD88WT (cohort 2) |
Phase III prospective, randomized (cohort 1) | PO zanubrutinib versus ibrutinib (cohort 1), PO zanubrutinib (cohort 2) | Cohort 1 VGPR/CR: 28.4% versus 19.2% ORR: 78% versus 78% MRR: 77.5% versus 77.8% 18-month PFS: 85% versus 84% Cohort 2 VGPR:26.9% ORR: 80.8% MRR: 50% 12-month PFS: 72.4% |
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 or higher infection rates were similar in both groups |
Owen et al.106 | 106 (14 TN, 92 RR) | Phase II, prospective | PO acalabrutinib | ORR: 93% MRR: 78% 24-month PFS: 90% TN, 82% RR |
Common AEs: headache, diarrhoea, bruising, fatigue, nausea, arthralgia Most common grade 3 (or higher) AEs: neutropenia and lower respiratory tract infections Atrial fibrillation: 5% MYD88 wild type (n = 14): PR rate = 64% |
Sekiguchi et al.107 | 27 (18 TN, 9 RR) | Phase II, prospective | PO tirabrutinib | TN: ORR, 94.4%, MRR, 88.9% RR: ORR, 100%, MRR, 88.9% Median PFS and OS not reached |
96.2% MYD88L265P mutated Most common AEs: rash (44.4%), neutropenia (25.9%), leukopenia (22.2%) Grade ⩾3 AEs: neutropenia (11.1%), lymphopenia (11.1%), leukopenia (7.4%) |
AEs, adverse events; BTK, Bruton’s tyrosine kinase; CR, complete remission; MRR, major response rate; ORR, overall response rate; OS, overall survival; PFS, progression-free survival; PO, per os (orally); RR, relapsed/refractory; TN, treatment naïve; VGPR, very good partial response; WM, Waldenström’s macroglobulinemia.
The initial phase I trial enrolled 42 patients with WM, of whom nine were treatment-naïve and 33 had relapsed/refractory disease. The overall response rate was 90%, including a 75% of major response rates, whereas 1-year PFS was 91.7%. Four patients harbored MYD88L265CXCR4WHIM and all responded to treatment. Four out of five MYD88wt patients also showed responses. Regarding the safety profile, 20% of the patients experienced grade 1/2 toxicities, 35% purpura/petechia, 31% upper respiratory-tract infections and 25% constipation. Hematological toxicities (anemia and neutropenia, each in 8% of the patients) were the two most frequent grade 3/4 adverse events. Atrial fibrillation grade 2 or less was reported in 6% of the patients.101,102 Interestingly, severe hemolysis and reaction to blood tranfusions have been reported in two patients receiving zanubrutinib in combination with an anti-programmed-cell-death-1 antibody in a phase I clinical trial.108
The previous study was subsequently expanded to a phase I/II trial design and included 77 patients with WM (nine treatment-naïve and 33 previously treated).103 A total of 50 patients received zanubrutinib at 160 mg twice daily and 23 were treated at 320 mg once daily. The overall response rate was 96%, whereas the rate of very good partial response or complete remission (VGPR/CR) was 45.2% after a median follow up of 36 months for the patients with relapsed/refractory disease, and 24 months for the treatment-naïve patients. Interestingly, the depth of response increased over time with 21% of the patients achieving VGPR/CR at 6 months, 33% at 12 months, and 44% at 24 months from treatment initiation. The estimated 3-year PFS rate was 81%, and the overall survival (OS) rate was 85%. The toxicity profile was considered acceptable and included contusion in 32.5% of the patients, neutropenia in 18%, major hemorrhage in 4%, atrial fibrillation/flutter in 5.2%, and diarrhea in 3%.103
Recently, the results of the phase III ASPEN clinical trial comparing zanubrutinib with ibrutinib monotherapy were reported.109,104 The study included 201 previously treated and non-eligible for chemoimmunotherapy treatment-naïve patients with WM, who were randomized to receive one of the two BTK inhibitors. All patients harbored the MYD88L265P mutation. The rate of deep responses (VGPR/CR) was 28% (n = 29) in the zanubrutinib compared with 19% (n = 19) in the ibrutinib patient group (p = 0.09). Therefore, the primary endpoint of statistical superiority in relation to deep responses (VGPR or better) was unmet. Furthermore, the major response rate among patients receiving zanubrutinib was 78% and 80% among those receiving ibrutinib, whereas the overall response rates were over 90% in both treatment groups. The 18-month PFS rate was 85% and 84%, respectively, and the 12-month OS rate was 97% and 94%, respectively. Overall, 58.4% patients experienced grade 3 or higher toxicities with zanubrutinib compared with 63.3% with ibrutinib. Patients with WM receiving zanubrutinib had a lower incidence of atrial fibrillation (3% versus 18% with ibrutinib, p < 0.05), contusion, diarrhea, peripheral edema, hemorrhage, muscle spasms, and pneumonia. The adverse events leading to treatment discontinuation were also fewer among zanubrutinib recipients. Although the incidence of neutropenia was higher with zanubrutinib (32% versus 15% with ibrutinib, p < 0.05), the rates of grade 3 or higher infections were similar in both treatment groups.109,104
The ASPEN study had also a non-randomized patient group who harbored MYD88wt. All 26 WM patients included received zanubrutinib until disease progression or unacceptable toxicity. Five patients were treatment naïve and 23 had received at least one prior line of treatment. Interestingly, the overall response rate reached 81%, including a major response rate of 50%, and a very good partial response rate of 27%. The PFS rate at 12 months was 72%. The most frequently reported toxicities included diarrhea, anemia, contusion, pyrexia, and upper respiratory-tract infection. Major hemorrhage and atrial fibrillation were reported in two patients and one patient, respectively. Following a median follow-up of 17.9 months, two patients discontinued zanubrutinib due to adverse events, whereas there were no cases of disease transformation to high-grade lymphoma.105
Acalabrutinib
Acalabrutinib (ACP-196), is another highly selective, potent covalent BTK inhibitor. Its off-target activity is minimal.110 It has received accelerated FDA approval as monotherapy for the treatment of relapsed/refractory mantle-cell lymphoma and chronic lymphocytic leukemia.111,112 Acalabrutinib monotherapy has showed high response rates of 93% both among treatment-naïve (n = 14) and previously treated (n = 92) patients with WM. Regarding 50 patients with available mutational status, the overall response rate was 94% among MYD88L265P patients compared with 79% among MYD88WT patients, and no very good partial responses were observed in the wild-type patient group. Overall, the responses were similar to those reported with ibrutinib, with a possible superior benefit for the patients with MYD88wt. Acalabrutinib showed a similar toxicity profile with ibrutinib.113,106
Tirabrutinib
Tirabrutinib (GS-4059/ONO) is an irreversible, selective BTK inhibitor. In a phase II trial, tirabrutinib resulted in an overall response rate of 94% and 100% among treatment-naïve and previously treated patients with WM after a median follow up of 6.5 and 8.3 months, respectively.107 Interestingly, the median time to major response was 1.87 months. Among the whole study population, the MYD88L265P mutation was detected in 96% of the patients. The toxicity profile was manageable, whereas the most common adverse effects included rash, neutropenia, and leukopenia.107
Non-covalent BTK inhibitors
Next-generation BTK inhibitors do not rely on covalent binding at the C481 site of the BTK molecule and, thus, they may bypass resistant mechanisms mediated by mutations in the C481 site.114 Two non-covalent, reversible BTK inhibitors that are being currently evaluated in WM are LOXO-305 and ARQ531. Promising preclinical data on the pharmacokinetic properties of LOXO-305 have led to an ongoing phase I/II clinical trial evaluating the maximum tolerated dose in patients with B-cell malignancies including WM [ClinicalTrials.gov identifier: NCT03740529].115 ARQ-531 is another reversible inhibitor that binds to the ATP binding region within the kinase domain of BTK without interacting with the C481 region. Interestingly, ARQ-531 also inhibits downstream effector of B-cell receptor signaling cascade including MEK, ERK, MYC and members of the SRC family kinases.116,117 In preclinical models of ibrutinib-resistant chronic lymphocytic leukemia and Richter’s transformation, ARQ-531 has shown significant activity.116,118 The results of the phase I part of the ongoing phase I/II study [ClinicalTrials.gov identifier: NCT03162536] evaluating ARQ-531 in relapsed/refractory B-cell malignancies indicated an acceptable toxicity profile and promising efficacy. The most common adverse events included gastrointestinal complaints, rash, and hematological toxicity. Ten partial responses were shown in all dose escalation levels (n = 40). A daily dose of 65 mg has been selected as the recommended dose for further evaluation in the phase II part of the study.119
Concluding remarks
The therapeutic approach in WM is increasingly being driven by insights in disease biology and genomic landscape. BTK plays a central role in signaling pathways for the WM clone and is constitutively activated secondary to MYD88 mutations. BTK inhibition has changed the treatment landscape of the disease. Ibrutinib has resulted in deep and durable responses both as an upfront and salvage treatment with a manageable toxicity profile. However, several challenges have emerged in order to optimize the treatment strategy including the development of more selective agents with fewer off-target effects and deeper responses, therapeutic approaches to overcome or even prevent resistance, the potential for a fixed-duration treatment regimen.120 Zanubrutinib has resulted in clinically meaningful antitumor activity, including deep and durable responses, with a low discontinuation rate due to treatment-related toxicities. Cardiovascular adverse events seem to be milder compared with ibrutinib. Interestingly, the efficacy of zanubrutinib in WM is significant both for MYD88L265P and MYD88WT patients. Furthermore, ongoing studies with non-covalent BTK inhibitors [ClinicalTrials.gov identifiers: NCT03740529 and NCT03162536] have shown promising efficacy and a tolerable toxicity profile in B-cell malignancies, whereas they bypass resistance mediated by mutations in the C481 site of the BTK. In an effort to enhance and optimize our current treatment strategies in WM, combinatory regimens including BTK inhibitors with CXCR4 antagonists (ulocuplumab, mavorixafor) [ClinicalTrials.gov identifiers: NCT03225716 and NCT04274738, respectively], or proteasome inhibitors (bortezomib with anti-CD20 monoclonal antibody rituximab, carfilzomib, ixazomib) [ClinicalTrials.gov identifiers: NCT03620903, NCT04263480, and NCT03506373, respectively], or bcl2 antagonists (venetoclax, APG-2575) [ClinicalTrials.gov identifiers: NCT04273139 and NCT04260217, respectively], or anti-CD38 monoclonal antibodies (daratumumab) [ClinicalTrials.gov identifier: NCT03679624], or programmed cell-death ligand-1 inhibitors (pembrolizumab) [ClinicalTrials.gov identifier: NCT02332980], are being currently evaluated in ongoing clinical trials.120 Long-term data will determine whether next-generation BTK inhibitors are more potent, safer, and able to overcome resistance to ibrutinib in WM, either alone or in combination with inhibitors of other interrelated intracellular cascades.
Footnotes
Conflict of interest statement: MAD declares honoraria from Beigene and Janssen. INS, MG, and DF declare that there is no conflict of interest.
Funding: The authors received no financial support for the research, authorship, and/or publication of this article.
ORCID iD: Meletios A. Dimopoulos https://orcid.org/0000-0001-8990-3254
Contributor Information
Ioannis Ntanasis-Stathopoulos, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.
Maria Gavriatopoulou, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.
Despina Fotiou, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.
Meletios A. Dimopoulos, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Alexandra General Hospital, 80 Vas. Sofias Avenue, Athens 11528, Greece.
References
- 1. Owen RG, Treon SP, Al-Katib A, et al. Clinicopathological definition of Waldenstrom’s macroglobulinemia: consensus panel recommendations from the Second International Workshop on Waldenstrom’s macroglobulinemia. Semin Oncol 2003; 30: 110–115. [DOI] [PubMed] [Google Scholar]
- 2. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2017. CA Cancer J Clin 2017; 67: 7–30. [DOI] [PubMed] [Google Scholar]
- 3. Wang H, Chen Y, Li F, et al. Temporal and geographic variations of Waldenstrom macroglobulinemia incidence: a large population-based study. Cancer 2012; 118: 3793–3800. [DOI] [PubMed] [Google Scholar]
- 4. Groves FD, Travis LB, Devesa SS, et al. Waldenstrom’s macroglobulinemia: incidence patterns in the United States, 1988-1994. Cancer 1998; 82: 1078–1081. [PubMed] [Google Scholar]
- 5. Herrinton LJ, Weiss NS. Incidence of Waldenstrom’s macroglobulinemia. Blood 1993; 82: 3148–3150. [PubMed] [Google Scholar]
- 6. Gertz MA. Waldenstrom macroglobulinemia: 2017 update on diagnosis, risk stratification, and management. Am J Hematol 2017; 92: 209–217. [DOI] [PubMed] [Google Scholar]
- 7. Teras LR, DeSantis CE, Cerhan JR, et al. 2016 US lymphoid malignancy statistics by World Health Organization subtypes. CA Cancer J Clin 2016; 66: 443–459. [DOI] [PubMed] [Google Scholar]
- 8. Kastritis E, Kyrtsonis MC, Morel P, et al. Competing risk survival analysis in patients with symptomatic Waldenström macroglobulinemia: the impact of disease unrelated mortality and of rituximab-based primary therapy. Haematologica 2015; 100: e446–e449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. McMaster ML. Familial Waldenstrom’s macroglobulinemia. Semin Oncol 2003; 30: 146–152. [DOI] [PubMed] [Google Scholar]
- 10. Treon SP, Hunter ZR, Aggarwal A, et al. Characterization of familial Waldenstrom’s macroglobulinemia. Ann Oncol 2006; 17: 488–494. [DOI] [PubMed] [Google Scholar]
- 11. Paiva B, Montes MC, Garcia-Sanz R, et al. Multiparameter flow cytometry for the identification of the Waldenstrom’s clone in IgM-MGUS and Waldenstrom’s Macroglobulinemia: new criteria for differential diagnosis and risk stratification. Leukemia 2014; 28: 166–173. [DOI] [PubMed] [Google Scholar]
- 12. Bustoros M, Sklavenitis-Pistofidis R, Kapoor P, et al. Progression risk stratification of asymptomatic Waldenstrom macroglobulinemia. J Clin Oncol 2019; 37: 1403–1411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Kyle RA, Benson JT, Larson DR, et al. Progression in smoldering Waldenstrom macroglobulinemia: long-term results. Blood 2012; 119: 4462–4466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Morel P, Duhamel A, Gobbi P, et al. International prognostic scoring system for Waldenstrom macroglobulinemia. Blood 2009; 113: 4163–4170. [DOI] [PubMed] [Google Scholar]
- 15. Kastritis E, Morel P, Duhamel A, et al. A revised international prognostic score system for Waldenstrom’s macroglobulinemia. Leukemia 2019; 33: 2654–2661. [DOI] [PubMed] [Google Scholar]
- 16. Hunter ZR, Xu L, Yang G, et al. Transcriptome sequencing reveals a profile that corresponds to genomic variants in Waldenstrom macroglobulinemia. Blood 2016; 128: 827–838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Eleutherakis-Papaiakovou E, Kastritis E, Gavriatopoulou M, et al. Circulating soluble receptor activator of nuclear factor kappa B ligand and C-C motif ligand 3 correlate with survival in patients with Waldenstrom macroglobulinemia. Clin Lymphoma Myeloma Leuk 2018; 18: 431–437. [DOI] [PubMed] [Google Scholar]
- 18. Hunter ZR, Treon SP. Epigenomics in Waldenstrom macroglobulinemia. Blood 2020; 136: 527–529. [DOI] [PubMed] [Google Scholar]
- 19. Dhodapkar MV, Hoering A, Gertz MA, et al. Long-term survival in Waldenstrom macroglobulinemia: 10-year follow-up of Southwest Oncology Group-directed intergroup trial S9003. Blood 2009; 113: 793–796. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Pophali PA, Bartley A, Kapoor P, et al. Prevalence and survival of smouldering Waldenstrom macroglobulinaemia in the United States. Br J Haematol 2019; 184: 1014–1017. [DOI] [PubMed] [Google Scholar]
- 21. Kyle RA, Treon SP, Alexanian R, et al. Prognostic markers and criteria to initiate therapy in Waldenstrom’s macroglobulinemia: consensus panel recommendations from the Second International Workshop on Waldenstrom’s macroglobulinemia. Semin Oncol 2003; 30: 116–120. [DOI] [PubMed] [Google Scholar]
- 22. Leblond V, Kastritis E, Advani R, et al. Treatment recommendations from the Eighth International Workshop on Waldenstrom’s macroglobulinemia. Blood 2016; 128: 1321–1328. [DOI] [PubMed] [Google Scholar]
- 23. Castillo JJ, Advani RH, Branagan AR, et al. Consensus treatment recommendations from the tenth International Workshop for Waldenstrom macroglobulinaemia. Lancet Haematol 2020; 7: e827–e837. [DOI] [PubMed] [Google Scholar]
- 24. Castillo JJ, Olszewski AJ, Cronin AM, et al. Survival trends in Waldenstrom macroglobulinemia: an analysis of the surveillance, epidemiology and end results database. Blood 2014; 123: 3999–4000. [DOI] [PubMed] [Google Scholar]
- 25. Buske C, Seymour JF. Immunochemotherapy in Waldenstrom macroglobulinemia - still the backbone of treatment. Leuk Lymphoma 2015; 56: 2489–2490. [DOI] [PubMed] [Google Scholar]
- 26. Gavriatopoulou M, Ntanasis-Stathopoulos I, Kastritis E, et al. How I treat rituximab refractory patients with WM. Oncotarget 2018; 9: 36824–36825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Castillo JJ, Kanan S, Meid K, et al. Rituximab intolerance in patients with Waldenstrom macroglobulinaemia. Br J Haematol 2016; 174: 645–648. [DOI] [PubMed] [Google Scholar]
- 28. Rawlings DJ, Saffran DC, Tsukada S, et al. Mutation of unique region of Bruton’s tyrosine kinase in immunodeficient XID mice. Science 1993; 261: 358–361. [DOI] [PubMed] [Google Scholar]
- 29. Ponader S, Burger JA. Bruton’s tyrosine kinase: from X-linked agammaglobulinemia toward targeted therapy for B-cell malignancies. J Clin Oncol 2014; 32: 1830–1839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Sideras P, Muller S, Shiels H, et al. Genomic organization of mouse and human Bruton’s agammaglobulinemia tyrosine kinase (Btk) loci. J Immunol 1994; 153: 5607–5617. [PubMed] [Google Scholar]
- 31. Stevenson FK, Krysov S, Davies AJ, et al. B-cell receptor signaling in chronic lymphocytic leukemia. Blood 2011; 118: 4313–4320. [DOI] [PubMed] [Google Scholar]
- 32. Satterthwaite AB, Witte ON. The role of Bruton’s tyrosine kinase in B-cell development and function: a genetic perspective. Immunol Rev 2000; 175: 120–127. [PubMed] [Google Scholar]
- 33. de Weers M, Verschuren MC, Kraakman ME, et al. The Bruton’s tyrosine kinase gene is expressed throughout B cell differentiation, from early precursor B cell stages preceding immunoglobulin gene rearrangement up to mature B cell stages. Eur J Immunol 1993; 23: 3109–3114. [DOI] [PubMed] [Google Scholar]
- 34. De Gorter DJ, Beuling EA, Kersseboom R, et al. Bruton’s tyrosine kinase and phospholipase Cgamma2 mediate chemokine-controlled B cell migration and homing. Immunity 2007; 26: 93–104. [DOI] [PubMed] [Google Scholar]
- 35. Varettoni M, Arcaini L, Zibellini S, et al. Prevalence and clinical significance of the MYD88 (L265P) somatic mutation in Waldenstrom’s macroglobulinemia and related lymphoid neoplasms. Blood 2013; 121: 2522–2528. [DOI] [PubMed] [Google Scholar]
- 36. Abeykoon JP, Paludo J, King RL, et al. MYD88 mutation status does not impact overall survival in Waldenstrom macroglobulinemia. Am J Hematol 2018; 93: 187–194. [DOI] [PubMed] [Google Scholar]
- 37. Bagratuni T, Ntanasis-Stathopoulos I, Gavriatopoulou M, et al. Detection of MYD88 and CXCR4 mutations in cell-free DNA of patients with IgM monoclonal gammopathies. Leukemia 2018; 32: 2617–2625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Ntanasis-Stathopoulos I, Gavriatopoulou M, Terpos E, et al. Monitoring plasma cell dyscrasias with cell-free DNA analysis. Clin Lymphoma Myeloma Leuk 2020; 20: e905–e909. [DOI] [PubMed] [Google Scholar]
- 39. Ntanasis-Stathopoulos I, Bagratuni T, Gavriatopoulou M, et al. Cell-free DNA analysis for the detection of MYD88 and CXCR4 mutations in IgM monoclonal gammopathies; an update with clinicopathological correlations. Am J Hematol 2020; 95: E148–E150. [DOI] [PubMed] [Google Scholar]
- 40. Treon SP, Xu L, Yang G, et al. MYD88 L265P somatic mutation in Waldenström’s macroglobulinemia. N Engl J Med 2012; 367: 826–833. [DOI] [PubMed] [Google Scholar]
- 41. Shinners NP, Carlesso G, Castro I, et al. Bruton’s tyrosine kinase mediates NF-kappa B activation and B cell survival by B cell-activating factor receptor of the TNF-R family. J Immunol 2007; 179: 3872–3880. [DOI] [PubMed] [Google Scholar]
- 42. Gray P, Dunne A, Brikos C, et al. MyD88 adapter-like (Mal) is phosphorylated by Bruton’s tyrosine kinase during TLR2 and TLR4 signal transduction. J Biol Chem 2006; 281: 10489–10495. [DOI] [PubMed] [Google Scholar]
- 43. Watters TM, Kenny EF, O’Neill LA. Structure, function and regulation of the Toll/IL-1 receptor adaptor proteins. Immunol Cell Biol 2007; 85: 411–419. [DOI] [PubMed] [Google Scholar]
- 44. Leleu X, Eeckhoute J, Jia X, et al. Targeting NF-kappaB in Waldenstrom macroglobulinemia. Blood 2008; 111: 5068–5077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Lin SC, Lo YC, Wu H. Helical assembly in the MyD88-IRAK4-IRAK2 complex in TLR/IL-1R signalling. Nature 2010; 465: 885–890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Treon SP, Xu L, Hunter Z. MYD88 Mutations and response to ibrutinib in Waldenstrom’s macroglobulinemia. N Engl J Med 2015; 373: 584–586. [DOI] [PubMed] [Google Scholar]
- 47. Yang G, Zhou Y, Liu X, et al. A mutation in MYD88 (L265P) supports the survival of lymphoplasmacytic cells by activation of Bruton tyrosine kinase in Waldenstrom macroglobulinemia. Blood 2013; 122: 1222–1232. [DOI] [PubMed] [Google Scholar]
- 48. Chen JG, Liu X, Munshi M, et al. BTK(Cys481Ser) drives ibrutinib resistance via ERK1/2 and protects BTK(wild-type) MYD88-mutated cells by a paracrine mechanism. Blood 2018; 131: 2047–2059. [DOI] [PubMed] [Google Scholar]
- 49. Yang G, Buhrlage SJ, Tan L, et al. HCK is a survival determinant transactivated by mutated MYD88, and a direct target of ibrutinib. Blood 2016; 127: 3237–3252. [DOI] [PubMed] [Google Scholar]
- 50. Hunter ZR, Xu L, Yang G, 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. Blood 2014; 123: 1637–1646. [DOI] [PubMed] [Google Scholar]
- 51. Castillo JJ, Moreno DF, Arbelaez MI, et al. CXCR4 mutations affect presentation and outcomes in patients with Waldenstrom macroglobulinemia: a systematic review. Expert Rev Hematol 2019; 12: 873–881. [DOI] [PubMed] [Google Scholar]
- 52. Xu L, Hunter ZR, Tsakmaklis N, et al. Clonal architecture of CXCR4 WHIM-like mutations in Waldenstrom macroglobulinaemia. Br J Haematol 2016; 172: 735–744. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Treon SP, Cao Y, Xu L, et al. Somatic mutations in MYD88 and CXCR4 are determinants of clinical presentation and overall survival in Waldenstrom macroglobulinemia. Blood 2014; 123: 2791–2796. [DOI] [PubMed] [Google Scholar]
- 54. Busillo JM, Benovic JL. Regulation of CXCR4 signaling. Biochim Biophys Acta 2007; 1768: 952–963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Cao Y, Hunter ZR, Liu X, et al. CXCR4 WHIM-like frameshift and nonsense mutations promote ibrutinib resistance but do not supplant MYD88(L265P)-directed survival signalling in Waldenstrom macroglobulinaemia cells. Br J Haematol 2015; 168: 701–707. [DOI] [PubMed] [Google Scholar]
- 56. Cao Y, Hunter ZR, Liu X, et al. The WHIM-like CXCR4(S338X) somatic mutation activates AKT and ERK, and promotes resistance to ibrutinib and other agents used in the treatment of Waldenstrom’s macroglobulinemia. Leukemia 2015; 29: 169–176. [DOI] [PubMed] [Google Scholar]
- 57. Tahirovic YA, Pelly S, Jecs E, et al. Small molecule and peptide-based CXCR4 modulators as therapeutic agents. A patent review for the period from 2010 to 2018. Expert Opin Ther Pat 2020; 30: 87–101. [DOI] [PubMed] [Google Scholar]
- 58. Castillo JJ, Treon SP. What is new in the treatment of Waldenstrom macroglobulinemia? Leukemia 2019; 33: 2555–2562. [DOI] [PubMed] [Google Scholar]
- 59. Roccaro AM, Sacco A, Jimenez C, et al. C1013G/CXCR4 acts as a driver mutation of tumor progression and modulator of drug resistance in lymphoplasmacytic lymphoma. Blood 2014; 123: 4120–4131. [DOI] [PubMed] [Google Scholar]
- 60. Hunter ZR, Xu L, Tsakmaklis N, et al. Insights into the genomic landscape of MYD88 wild-type Waldenstrom macroglobulinemia. Blood Adv 2018; 2: 2937–2946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Charalambous A, Schwarzbich MA, Witzens-Harig M. Ibrutinib. Recent Results Cancer Res 2018; 212: 133–168. [DOI] [PubMed] [Google Scholar]
- 62. Imbruvica (ibrutinib) Capsules [prescribing Information]. Sunnyvale, CA: Pharmacyclics, Inc, 2016. [Google Scholar]
- 63. De Jong J, Skee D, Murphy J, et al. Effect of CYP3A perpetrators on ibrutinib exposure in healthy participants. Pharmacol Res Perspect 2015; 3: e00156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Advani RH, Buggy JJ, Sharman JP, et al. Bruton tyrosine kinase inhibitor ibrutinib (PCI-32765) has significant activity in patients with relapsed/refractory B-cell malignancies. J Clin Oncol 2013; 31: 88–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Treon SP, Tripsas CK, Meid K, et al. Ibrutinib in previously treated Waldenstrom’s macroglobulinemia. N Engl J Med 2015; 372: 1430–1440. [DOI] [PubMed] [Google Scholar]
- 66. Treon S, Meid K, Gustine J, et al. Long-term follow-up of previously treated patients who received ibrutinib for symptomatic Waldenstrom’s macroglobulinemia: update of pivotal clinical trial. Blood 2017; 130: 2776. [Google Scholar]
- 67. Buske C, Tedeschi A, Trotman J, et al. Ibrutinib treatment in Waldenström’s macroglobulinemia: follow-up efficacy and safety from the iNNOVATETM study. Blood 2018; 132: 149. [Google Scholar]
- 68. Dimopoulos MA, Trotman J, Tedeschi A, 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: 241–250. [DOI] [PubMed] [Google Scholar]
- 69. Treon SP, Gustine J, Meid K, et al. Ibrutinib monotherapy in symptomatic, treatment-naive patients with Waldenstrom macroglobulinemia. J Clin Oncol 2018; 36: 2755–2761. [DOI] [PubMed] [Google Scholar]
- 70. Dimopoulos MA, Tedeschi A, Trotman J, et al. Phase 3 trial of ibrutinib plus rituximab in Waldenstrom’s macroglobulinemia. N Engl J Med 2018; 378: 2399–2410. [DOI] [PubMed] [Google Scholar]
- 71. Papanota AM, Ntanasis-Stathopoulos I, Kastritis E, et al. Evaluating ibrutinib in the treatment of symptomatic Waldenstrom’s macroglobulinemia. J Blood Med 2019; 10: 291–300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Gavriatopoulou M, Ntanasis-Stathopoulos I, Moulopoulos LA, et al. Treatment of Bing-Neel syndrome with first line sequential chemoimmunotherapy: a case report. Medicine 2019; 98: e17794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Castillo JJ, Itchaki G, Paludo J, et al. Ibrutinib for the treatment of Bing-Neel syndrome: a multicenter study. Blood 2019; 133: 299–305. [DOI] [PubMed] [Google Scholar]
- 74. De Weerdt I, Koopmans SM, Kater AP, et al. Incidence and management of toxicity associated with ibrutinib and idelalisib: a practical approach. Haematologica 2017; 102: 1629–1639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Brown JR, Moslehi J, O’Brien S, et al. Characterization of atrial fibrillation adverse events reported in ibrutinib randomized controlled registration trials. Haematologica 2017; 102: 1796–1805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. McMullen JR, Boey EJ, Ooi JY, et al. Ibrutinib increases the risk of atrial fibrillation, potentially through inhibition of cardiac PI3K-Akt signaling. Blood 2014; 124: 3829–3830. [DOI] [PubMed] [Google Scholar]
- 77. Ganatra S, Sharma A, Shah S, et al. Ibrutinib-associated atrial fibrillation. JACC Clin Electrophysiol 2018; 4: 1491–1500. [DOI] [PubMed] [Google Scholar]
- 78. Alexandre J, Moslehi JJ, Bersell KR, et al. Anticancer drug-induced cardiac rhythm disorders: current knowledge and basic underlying mechanisms. Pharmacol Ther 2018; 189: 89–103. [DOI] [PubMed] [Google Scholar]
- 79. Caldeira D, Alves D, Costa J, et al. Ibrutinib increases the risk of hypertension and atrial fibrillation: systematic review and meta-analysis. PLoS One 2019; 14: e0211228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Lampson BL, Yu L, Glynn RJ, et al. Ventricular arrhythmias and sudden death in patients taking ibrutinib. Blood 2017; 129: 2581–2584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Gustine JN, Meid K, Dubeau TE, et al. Atrial fibrillation associated with ibrutinib in Waldenstrom macroglobulinemia. Am J Hematol 2016; 91: E312–313. [DOI] [PubMed] [Google Scholar]
- 82. Brown JR, Moslehi J, Ewer MS, et al. Incidence of and risk factors for major haemorrhage in patients treated with ibrutinib: an integrated analysis. Br J Haematol 2019; 184: 558–569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Mock J, Kunk PR, Palkimas S, et al. Risk of major bleeding with ibrutinib. Clin Lymphoma Myeloma Leuk 2018; 18: 755–761. [DOI] [PubMed] [Google Scholar]
- 84. Yun S, Vincelette ND, Acharya U, et al. Risk of atrial fibrillation and bleeding diathesis associated with ibrutinib treatment: a systematic review and pooled analysis of four randomized controlled trials. Clin Lymphoma Myeloma Leuk 2017; 17: 31–37.e13. [DOI] [PubMed] [Google Scholar]
- 85. Bye AP, Unsworth AJ, Vaiyapuri S, et al. Ibrutinib inhibits platelet integrin αIIbβ3 outside-in signaling and thrombus stability but not adhesion to collagen. Arterioscler Thromb Vasc Biol 2015; 35: 2326–2335. [DOI] [PubMed] [Google Scholar]
- 86. Levade M, David E, Garcia C, et al. Ibrutinib treatment affects collagen and von Willebrand factor-dependent platelet functions. Blood 2014; 124: 3991–3995. [DOI] [PubMed] [Google Scholar]
- 87. Dimopoulos MA, Salman Z, Buske C. Ibrutinib and rituximab in Waldenstrom’s macroglobulinemia. N Engl J Med 2018; 379: 1975–1976. [DOI] [PubMed] [Google Scholar]
- 88. Castillo JJ, Gustine JN, Meid K, et al. Impact of ibrutinib dose intensity on patient outcomes in previously treated Waldenstrom macroglobulinemia. Haematologica 2018; 103: e466–e468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Niemann CU, Herman SE, Maric I, et al. Disruption of in vivo chronic lymphocytic leukemia tumor-microenvironment interactions by ibrutinib—findings from an investigator-initiated phase II study. Clin Cancer Res 2016; 22: 1572–1582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Ping L, Ding N, Shi Y, et al. The Bruton’s tyrosine kinase inhibitor ibrutinib exerts immunomodulatory effects through regulation of tumor-infiltrating macrophages. Oncotarget 2017; 8: 39218–39229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Ren L, Campbell A, Fang H, et al. Analysis of the effects of the Bruton’s tyrosine kinase (Btk) inhibitor ibrutinib on monocyte Fcγ receptor (FcγR) function. J Biol Chem 2016; 291: 3043–3052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Gustine JN, Meid K, Dubeau T, et al. Ibrutinib discontinuation in Waldenstrom macroglobulinemia: etiologies, outcomes, and IgM rebound. Am J Hematol 2018; 93: 511–517. [DOI] [PubMed] [Google Scholar]
- 93. Castillo JJ, Gustine JN, Meid K, et al. Ibrutinib withdrawal symptoms in patients with Waldenstrom macroglobulinemia. Haematologica 2018; 103: e307–e310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Martin P, Maddocks K, Leonard JP, et al. Postibrutinib outcomes in patients with mantle cell lymphoma. Blood 2016; 127: 1559–1563. [DOI] [PubMed] [Google Scholar]
- 95. Xu L, Tsakmaklis N, Yang G, et al. Acquired mutations associated with ibrutinib resistance in Waldenstrom macroglobulinemia. Blood 2017; 129: 2519–2525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Paulus A, Akhtar S, Yousaf H, et al. Waldenstrom macroglobulinemia cells devoid of BTK(C481S) or CXCR4(WHIM-like) mutations acquire resistance to ibrutinib through upregulation of Bcl-2 and AKT resulting in vulnerability towards venetoclax or MK2206 treatment. Blood Cancer J 2017; 7: e565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Jimenez C, Chan GG, Xu L, et al. Genomic evolution of ibrutinib-resistant clones in Waldenstrom macroglobulinaemia. Br J Haematol 2020; 189: 1165–1170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Wu J, Liu C, Tsui ST, et al. Second-generation inhibitors of Bruton tyrosine kinase. J Hematol Oncol 2016; 9: 80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Tam CS, LeBlond V, Novotny W, et al. A head-to-head Phase III study comparing zanubrutinib versus ibrutinib in patients with Waldenstrom macroglobulinemia. Future Oncol 2018; 14: 2229–2237. [DOI] [PubMed] [Google Scholar]
- 100. Li N, SZ, Liu Y, et al. BGB-3111 is a novel and highly selective Bruton’s tyrosine kinase (BTK) inhibitor. In: AACR 106th annual meeting, Philadelphia, PA, 18–22 April 2015. [Google Scholar]
- 101. Tam C, GA, Opat S, et al. The BTK inhibitor, Bgb-3111, is safe, tolerable, and highly active in patients with relapsed/refractory B-cell malignancies: initial report of a phase 1 first-in-human trial. Blood 2015; 126: 832. [Google Scholar]
- 102. Trotman J, Opat S, Marlton P, et al. Bruton’s Tyrosine Kinase (BTK) inhibitor BGB-3111 demonstrates high very good partial response (VGPR) rate in patients with Waldenström macroglobulinemia (WM). Hematol Oncol 2017; 35: 70–71.28591420 [Google Scholar]
- 103. Trotman J, Opat S, Gottlieb DJ, et al. Zanubrutinib for the treatment of patients with Waldenstrom macroglobulinemia: three years of follow-up. Blood 2020; 136: 2027–2037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Tam CS, Opat S, D’Sa S, et al. A randomized phase 3 trial of zanubrutinib versus ibrutinib in symptomatic Waldenstrom macroglobulinemia:the ASPEN study. Blood 2020; 136: 2038–2050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Garcia-Sanz R, Dimopoulos MA, Lee HP, et al. Updated results of the ASPEN trial from a cohort of patients with MYD88 wild-type (MYD88WT) Waldenström macroglobulinemia (WM). J Clin Oncol 2020; 38(Suppl. 15): e20056. [Google Scholar]
- 106. Owen RG, McCarthy H, Rule S, et al. Acalabrutinib monotherapy in patients with Waldenstrom macroglobulinemia: a single-arm, multicentre, phase 2 study. Lancet Haematol 2020; 7: e112–e121. [DOI] [PubMed] [Google Scholar]
- 107. Sekiguchi N, Rai S, Munakata W, et al. A multicenter, open-label, phase II study of tirabrutinib (ONO/GS-4059) in patients with Waldenstrom’s macroglobulinemia. Cancer Sci 2020; 111: 3327–3337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Othman J, Verner E, Tam CS, et al. Severe hemolysis and transfusion reactions after treatment with BGB-3111 and PD-1 antibody for Waldenstrom macroglobulinemia. Haematologica 2018; 103: e223–e225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. BieGene. BeiGene announces results of phase 3 ASPEN trial of zanubrutinib compared to ibrutinib for the treatment of patients with Waldenström’s macroglobulinemia. Epub ahead of print 2019. https://www.globenewswire.com/news-release/2019/12/16/1960839/0/en/BeiGene-Announces-Results-of-Phase-3-ASPEN-Trial-of-Zanubrutinib-Compared-to-Ibrutinib-for-the-Treatment-of-Patients-with-Waldenstr%C3%B6m-s-Macroglobulinemia.html
- 110. Barf T, Covey T, Izumi R, et al. Acalabrutinib (ACP-196): a covalent Bruton tyrosine kinase inhibitor with a differentiated selectivity and in vivo potency profile. J Pharmacol Exp Ther 2017; 363: 240–252. [DOI] [PubMed] [Google Scholar]
- 111. Byrd JC, Harrington B, O’Brien S, et al. Acalabrutinib (ACP-196) in relapsed chronic lymphocytic leukemia. N Engl J Med 2016; 374: 323–332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Wang M, Rule S, Zinzani PL, et al. Acalabrutinib in relapsed or refractory mantle cell lymphoma (ACE-LY-004): a single-arm, multicentre, phase 2 trial. Lancet 2018; 391: 659–667. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Owen R, McCarthy CA, Rule S, et al. Acalabrutinib in patients (pts) with Waldenström macroglobulinemia (WM). J Clin Oncol 2018; 36: 7501. [Google Scholar]
- 114. Johnson AR, Kohli PB, Katewa A, et al. Battling Btk mutants with noncovalent inhibitors that overcome Cys481 and Thr474 mutations. ACS Chem Biol 2016; 11: 2897–2907. [DOI] [PubMed] [Google Scholar]
- 115. Brandhuber B, Gomez E, Smith S. Abstract CLL-200: LOXO-305, a next generation reversible BTK inhibitor, for overcoming acquired resistance to irreversible BTK inhibitors [Abstract]. Clin Lymphoma Myeloma Leuk 2018; 18: S216. [Google Scholar]
- 116. Reiff SD, Mantel R, Smith LL, et al. The BTK inhibitor ARQ 531 targets ibrutinib-resistant CLL and richter transformation. Cancer Discov 2018; 8: 1300–1315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Soncini D, Orecchioni S, Ruberti S, et al. The new small molecule tyrosine-kinase inhibitor ARQ531 targets acute myeloid leukemia cells by disrupting multiple tumor-addicted programs. Haematologica 2020; 105: 224956. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Bond DA, Woyach JA. Targeting BTK in CLL: beyond ibrutinib. Curr Hematol Malig Rep 2019; 14: 197–205. [DOI] [PubMed] [Google Scholar]
- 119. Woyach J, Stephens D, Flinn I, et al. Final results of phase 1, dose escalation study evaluating ARQ 531 in patients with relapsed or refractory B-cell lymphoid malignancies. Blood 2019; 134: 4298–4298. [Google Scholar]
- 120. Gavriatopoulou M, Fotiou D, Ntanasis-Stathopoulos I, et al. The current role of BTK inhibitors in the treatment of Waldenstrom’s macroglobulinemia. Expert Rev Anticancer Ther 2020; 20: 663–674. [DOI] [PubMed] [Google Scholar]