215
Views
3
CrossRef citations to date
0
Altmetric
Review

Zanubrutinib in Treating Waldenström Macroglobulinemia, the Last Shall Be the First

ORCID Icon &
Pages 657-668 | Published online: 23 Jun 2022

Abstract

In Waldenström macroglobulinemia (WM), a lymphoplasmacytic lymphoma characterized by monoclonal immunoglobulin M (IgM) gammopathy, aberrant Bruton tyrosine kinase (BTK) signaling has been identified as one mechanism of pathogenesis. For this reason, selective BTK inhibiting therapies have emerged as an attractive option for treatment within the therapeutic landscape also comprising chemotherapy, monoclonal antibodies, proteasome inhibitors, and B-cell lymphoma 2 (BCL2) inhibitors. The first BTK inhibiting therapy, ibrutinib, showed great efficacy in treating WM. However, response rates were dependent on whether patients had the CXCR4 mutation, a molecular aberration that may confer resistance to BTK inhibitors. Furthermore, ibrutinib’s toxicities, most notably hypertension and atrial arrhythmia, led to dose reductions or discontinuation. The toxicity profile of ibrutinib can be attributed to the inhibition of additional kinases that are structurally related to BTK. Therefore, the next-generation highly selective zanubrutinib was developed to address the concerns regarding toxicity and tolerance related to ibrutinib therapy. Based on the results of the randomized, open-label Phase 3 ASPEN (NCT03053440) trial, the Food and Drug Administration (FDA) approved zanubrutinib for treating WM. This trial directly compared zanubrutinib to ibrutinib in patients with treatment-naïve or relapsed/refractory WM, and the results showed stronger responses with zanubrutinib. More importantly, patients responded strongly to zanubrutinib therapy regardless of CXCR4 mutation status. Additionally, zanubrutinib was associated with fewer grade 3 or higher toxicities and was generally better tolerated. Another Phase 1/2 study has been conducted with just zanubrutinib in WM showcasing high efficacy with few toxicities as well. Even though zanubrutinib has been the third and last BTK inhibitor to currently penetrate the market for B-cell lymphoproliferative malignancies, we highlight the emergence of zanubrutinib as a key player in the forefront of the therapeutic landscape in WM.

Introduction

B-cell development, survival, and expansion is critically dependent on normally functioning and tightly regulated B-cell receptor (BCR) signaling pathways. Key mediators of the BCR pathway include the proximal kinases – Bruton tyrosine kinase (BTK), phosphatidyl inositol 3 kinase (PI3K), and spleen tyrosine kinase (SYK).Citation1 A member of the Tec protein kinase (TEC) family, BTK is an important regulator of B-cell proliferation and survival.Citation1 In the BCR pathway, BTK gets activated by upstream Src-family kinases; once activated, the enzyme goes on to stimulate downstream cell survival pathways including nuclear factor kappa-light-chain-enhancer of activated B-cells (NF-κB) and mitogen activated protein kinase (MAPK).Citation2 Defective BTK was first identified in 1993 in X-linked hypogammaglobulinemia, an inherited immunodeficiency disease.Citation3 More recently, aberrant BTK signaling has been implicated in the pathogenesis of a variety of B-cell malignancies including chronic lymphocytic leukemia (CLL),Citation4Citation7 mantle cell lymphoma (MCL),Citation5,Citation8,Citation9 diffuse-large B-cell lymphoma (DLBCL),Citation10 marginal zone lymphoma (MZL),Citation11 and Waldenström macroglobulinemia (WM).Citation5,Citation12 For these reasons, BTK inhibitors (BTKi) have emerged as an important and effective treatment option for difficult-to-manage B-cell malignancies. One BTKi available to treat B-cell malignancies is zanubrutinib. This next-generation highly specific BTKi has shown strong efficacy in treating a multitude of B-cell malignancies including CLL, MCL, MZL, and WM. Herein, we will discuss the particularly available data regarding the use of zanubrutinib in WM.

Waldenström Macroglobulinemia Overview

Waldenström macroglobulinemia (WM) is a rare lymphoplasmacytic lymphoma occurring in the bone marrow that is characterized by a monoclonal immunoglobulin M (IgM) gammopathy. This disease is presumed to be primarily sporadic with an unknown cause. However, the genetic landscape of WM has identified genetic mutations in the MYD88 and CXCR4 genes in 90% and 30% of cases, respectively.Citation13,Citation14 A genetic basis for disease pathogenesis has been further supported by analysis of 257 unrelated patients with WM revealing that 48% of patients with WM had a first-degree relative having either WM or another B-cell disorder.Citation15 Additionally, a family history of WM or another plasma disorder led to a diagnosis of WM at a younger age with greater bone marrow involvement.Citation15 Ultimately though, the greatest risk factor for developing WM is a pre-existing monoclonal gammopathy of undetermined significance (MGUS).Citation16

Patients with WM develop symptoms due to disease infiltration of the lymph nodes, bone marrow, or spleen, effects of elevated IgM levels in the blood, or a combination. For disease infiltration, excessive bone marrow involvement and infiltration crowds out the marrow space and creates cytopenias, mainly anemia, thrombocytopenia, and neutropenia.Citation16 Lytic bone disease is highly uncommon in WM, differentiating it from multiple myeloma. However, the disease does usually spread to lymph nodes, the liver, and spleen – creating lymphadenopathy, hepatomegaly, and splenomegaly.Citation16 Some rare cases of pulmonary parenchymal infiltration have been documented, and these have presented as diffuse pulmonary infiltrates, nodules, or effusions.Citation17 Infiltration of the stomach, bowel, skull base, and orbit has also been reported.Citation18 An extremely rare neurologic complication of WM is Bing-Neel syndrome, the manifestation of direct central nervous system disease involvement that presents with a variety of symptoms including headache, ataxia, vertigo, nystagmus, diplopia, and more seriously, coma.Citation19 BTK inhibitors like ibrutinib and zanubrutinib, have demonstrated CNS penetrance, and there are some case reports of BTKi use in Bing-Neel syndrome.Citation20,Citation21

The effects of elevated IgM levels in the blood are due to serum hyperviscosity, tissue deposition of IgM, or the autoantibody activity of IgM. Increased concentration of the monoclonal protein increases serum viscosity, and thus vascular resistance. This leads to the downstream effects of constitutional symptoms, bleeding, high-output cardiac failure, and ocular or neurological complications.Citation16 The IgM can also deposit in a variety of tissues leading to complications such as proteinuria, diarrhea, and macroglobulinemia cutis.Citation16 Furthermore, the immunoglobulin light chains of IgM can precipitate primary amyloidosis in a variety of organs such as the heart, peripheral nerves, kidney, liver, and lungs.Citation16 Finally, autoimmune reactions can be triggered by high levels of IgM in the serum. At cold temperatures, autoimmune hemolytic anemias can occur with elevated cold agglutination titers.Citation16 Symmetric polyneuropathies are also commonly reported.Citation16 Clinical evidence of any of these adverse effects with significant symptoms serves as indication for initiating active treatment.

Therapeutic Landscape

Multiple treatment options are available for WM including chemotherapy, monoclonal antibodies, proteasome inhibitors, BTK inhibitors (like ibrutinib, acalabrutinib, or zanubrutinib), and B-cell lymphoma 2 (BCL2) inhibitors. Treatment is usually initiated when patients present with progressive anemia, hyperviscosity complications, significant fatigue, or other significant constitutional symptoms. The development of novel therapeutic modalities such as BTKis and BCL2is has already altered the therapeutic armamentarium available for B-cell malignancies.Citation22 The selection of an optimal therapeutic regimen is an individualized decision that is tailored by the clinician and the patient while considering a myriad of factors including patient characteristics (eg, age, performance status, other medical problems, other medications), disease characteristics (eg, mutational status, prognostic markers, bulky disease, geographic area of involvement, tumor lysis syndrome risk), and treatment characteristics (eg, cost, toxicity, convenience).Citation22 We prefer enrolling any patient in a clinical trial when available. In the standard of care realm, we are respectful of the label as instructed by the Food and Drug Administration (FDA) approval and/or regimens mentioned in reputable national cancer guidelines after a prolonged discussion with the patient regarding pros and cons. Of note, BTKi appear in National Comprehensive Cancer Network (NCCN) guidelines as preferred options both for primary therapy and for previously treated WM as of the published guidelines accessed on May 03, 2022.Citation23 We align ourselves with NCCN guidelines in the way that we shy away as much as possible from chemotherapeutic regimens in lieu of favoring novel non-chemotherapy agents; nevertheless, the ultimate therapeutic decision is individualized. In NCCN guidelines, both ibrutinib plus or minus rituximab and zanubrutinib appear as category 1 recommendations for primary therapy and for previously treated WM. Acalabrutinib does not currently appear as an option for primary therapy for WM in NCCN; nevertheless, acalabrutinib does appear as a part of other recommended regimens available for previously treated WM. Herein, we provide additional granularity regarding these multiple treatment options.

Plasmapheresis

A notable complication of the monoclonal gammopathy seen in WM is peripheral neuropathy that can be attributed to demyelination and widened myelin lamellae.Citation24 Additionally, elevated IgM levels can create the hyperviscosity syndrome presenting with the classic triad of mucosal bleeding, visual changes, and neurologic complications. For these patients, plasmapheresis has been shown to lower serum IgM levels, improve the symptoms associated with hyperviscosity, and reduce the progression of neuropathy.Citation25Citation27 However, plasmapheresis should only be used for symptom management until systemic therapy decreases tumor burden.Citation28

Rituximab

Rituximab is a chimeric monoclonal antibody that targets the CD20 antigen expressed on all B-cells that leads to lymphocyte lysis after binding. In terms of rituximab monotherapy, one study was conducted on 27 patients with WM who were treated with rituximab intravenously for 4 weeks.Citation29 Twelve patients (44%) achieved a partial response (PR) at a median time to response of 3.3 months.Citation29 Treatment was generally well-tolerated, but 25% of patients experienced a mild infusion reaction mostly manifesting as fever and chills.Citation29 Furthermore, at a median follow-up of 15.7 months, 9 of the 12 patients who responded to treatment remained disease progression free.Citation29 Based on these findings, during the Second International Workshop on WM held in September 2002 in Athens, Greece, a consensus panel recommended using combinations of alkylating agents, nucleoside analogs, and rituximab for treating WM.Citation30 Since then, many clinical studies have been conducted to evaluate the efficacy and safety of rituximab combination regimens in treating WM.Citation31 A pooled meta-analysis found that rituximab combination therapies result in an objective response rate (ORR) of 84%, major response rate (MRR) 71%, and complete response (CR) 7% in patients with symptomatic WM.Citation31 Rituximab plus conventional alkylating agents that containing chemotherapy resulted in an ORR of 86%, MRR 74%, and CRR 8%.Citation31 On the other hand, rituximab plus a purine analog produced an ORR of 85%, MRR 74%, and CRR 9%.Citation31 Finally, rituximab plus a proteasome inhibitor yielded an ORR of 86%, MRR 68%, and CRR 7%.Citation31 In terms of safety, low grade adverse events related to the first infusion of rituximab were the most common; these included dyspnea, hypertension, angioedema, bronchospasm, cough, pyrexia, chills, rash, and vomiting.Citation31 The most common grade 3 or higher adverse events were neutropenia, thrombocytopenia, and anemia; it should be noted that these adverse events generally did not lead to treatment discontinuation.Citation31

One clinical challenge with rituximab therapy in WM is the paradoxical rise in IgM levels after initial infusion. One study evaluating rituximab in 54 patients with WM found that 54% had a rise in IgM levels between infusion and the first scheduled measurement time point.Citation32 Two-months later, 59% of 22 evaluable patients continued to have elevated IgM levels.Citation32 Finally, 4 months after therapy, 4 out of 15 patients still had elevated IgM levels.Citation32 The IgM flare can either precipitate the hyperviscosity syndrome in patients or worsen existing hyperviscosity symptoms, thus creating the need for emergent plasmapheresis. Another challenge is that rituximab intolerance has been identified in a higher proportion of patients with WM when compared to other B-cell malignancies.Citation33 An analysis on 85 patients intolerant to rituximab revealed that 34% developed intolerance during the first-line of treatment while 15% developed intolerance during the first year of exposure to rituximab.Citation33 With both single-agent rituximab and rituximab-containing regimens, the monoclonal antibody had to be discontinued for worsening infusion-related reactions.Citation33 The most common reactions, occurring in greater than 10% of patients, were anaphylaxis, chills and rigors, hives, hypotension, and shortness of breath.Citation33 Thus, rituximab intolerance presents a unique challenge for treating WM. Nevertheless, we are usually able to circumvent these toxicities when using rituximab in B-cell malignancies.

Chemoimmunotherapy

Two commonly prescribed chemoimmunotherapy regimens for treating WM are bendamustine-rituximab (BR) and dexamethasone-rituximab-cyclophosphamide (RCD). For BR, the STiLNHL1–2003 phase 3 trial first highlighted this regimen’s efficacy in treating indolent non-Hodgkin lymphoma (including 41 patients with lymphoplasmacytic lymphoma) when compared to rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP).Citation34 At a median follow-up of 45 months, the median progression-free survival (PFS) was longer in the BR group compared to the R-CHOP group.Citation34 ORRs were 93% and 91% in the BR and R-CHOP groups, respectively.Citation34 Furthermore, BR was better tolerated with lower rates of alopecia, hematological toxicity, infections, peripheral neuropathy, and stomatitis.Citation34 More recently, at longer median 5.9 years follow-up, median PFS with BR was reported as 65.3 months.Citation35 Furthermore, the study compared two groups who had been given BR induction therapy where one group received 2-year rituximab monotherapy afterwards, while the other was just observed.Citation35 No significant difference in PFS or OS was found between the two groups – confirming the highly effective nature of induction with BR therapy.Citation35

For the other regimen, RCD, a Phase II trial on symptomatic treatment-naïve WM patients found that 83% achieved a response (7% complete, 67% partial, and 9% minor).Citation36 For all patients, the 2-year PFS rate was 67%; for patients who responded to RCD treatment, this rate was 80%.Citation36 Thirty-two percent of patients experienced an IgM flare, however, levels dropped below baseline after RCD treatment was completed.Citation36 For toxicities, only 7 of 72 patients experienced a grade 3 or 4 neutropenia with no patients having thrombocytopenia.Citation36 Other adverse events reported were related to the infusion of rituximab and included chills, fevers, and headaches.Citation36 For these reasons, chemoimmunotherapy remains a front-runner in the first-line treatments of WM.

Bortezomib

The proteosome is a 26S nuclear/cytosolic complex that regulates protein degradation, and thus, influences various cellular pathways including the cell cycle. Bortezomib is a small molecule that selectively and reversibly inhibits the proteosome to influence these cellular pathways. In 27 patients with WM who had previously failed at least one line of therapy, bortezomib was found to significantly reduce serum IgM levels from 4660 to 2092 mg/dL.Citation37 An ORR of 85% was achieved with therapy at a median of 1.4 months.Citation37 Within those who responded, 48.1% achieved a MR.Citation37 The most common grade 3 or higher toxicities that occurred were sensory neuropathies, leukopenia, neutropenia, dizziness, and thrombocytopenia – occurring in 22.2%, 18.5%, 14.8%, 11.1%, and 7.4%, respectively.Citation37 All events of neuropathy resolved with cessation of therapy in this particular study.Citation37 Of note, response rates to bortezomib were found to not be negatively impacted by the CXCR4 mutation, a molecular aberration that has conferred resistance to other lines of treatment such as ibrutinib, which will be discussed later.Citation38 Although bortezomib monotherapy has shown improved clinical outcomes in patients with WM, the associated toxicity of sensory neuropathy presents a clinical challenge. In fact, another study showed bortezomib’s efficacy at treating WM, but 74% developed new or worsening neuropathy, and this was the most common cause for dose reduction or therapy cessation.Citation39

Venetoclax

BCL2, a regulatory molecule of the intrinsic apoptosis pathway, is overexpressed in WM, especially in patients with the MYD88L265P mutation, regardless of CXCR4 mutation status.Citation40 Venetoclax is a BCL2 inhibitor that was approved for the treatment of CLL and acute myeloid leukemia.Citation40 A phase 1 study was conducted to evaluate the efficacy and safety of venetoclax in 33 patients with relapsed/refractory (R/R) WM.Citation40 Sixteen (50%) of these patients had previously been treated with BTKi therapy and were either intolerant to the therapy or had disease progression.Citation40 The MYD88L265P mutation was found in all patients, but the CXCR4WHIM mutation was found in 17 patients.Citation40 This study reported an ORR and MRR of 84% and 81%.Citation40 No patient achieved a complete response.Citation40 The median time to achieve either a minor or major response was 1.9 months.Citation40 At a median follow-up of 33 months, 19 patients had progression of disease, and 12 had to start an additional treatment.Citation40 The median progression-free survival (PFS) at 12, 24, and 30 months was 83%, 80%, and 95%.Citation40 No significant difference in PFS was found based on prior BTKi therapy or CXCR4 mutational status.Citation40 Twenty-two percent of patients had to reduce their dose of treatment because of neutropenia (n=2), pancytopenia (n=1), fatigue (n=1), diarrhea (n=1), multiple grade 2 events (n=1), and self-reduction (n=1).Citation40 However, the dose reductions were not associated with any significant difference in response rates.Citation40 Thirty-one (94%) patients experienced a grade 2 or higher adverse event.Citation40 Only 7 patients experienced a grade 4 adverse event, and these were neutropenia (n=6) and febrile neutropenia (n=1).Citation40

Ibrutinib

Ibrutinib is a BTKi that was approved for the treatment of WM by the FDA in January of 2015.Citation41 Ibrutinib, in addition to its active metabolite PCI-45227, covalently and irreversibly binds to Cysteine 481 in the kinase domain of BTK to inhibit its activity.Citation41 In WM, BTK forms a complex with the mutated MYD88 protein to create a constantly activated BTK molecule.Citation41 Thus, ibrutinib has shown significant activity in managing WM. FDA approval followed the landmark phase II trial showing an ORR of 90.5%, MRR of 73%, 2-year PFS of 69.1%, and 2-year overall survival (OS) rates of 95.2% in 63 patients with symptomatic WM refractory to at least one prior treatment.Citation42 The MYD88L265P mutation was present in 89% while the CXCR4WHIM mutation – a mutation that confers resistance to ibrutinib – was present in 34%.Citation42 Responses to treatment were also assessed by genomic group: MYD88L265P CXCR4WT, MYD88L265P CXCR4WHIM, and MYD88WT CXCR4WT. ORR and MRR were 100% and 91.2%, 85.7% and 61.9%, and 71.4% and 28.6% in these groups, respectively.Citation42 At long-term median 59-month follow-up, ORR and MRR were 90.5% and 79.4%, respectively.Citation43 When looking at the best response to treatment, serum IgM levels declined from 3520 to 821 mg/dL, bone marrow disease involvement decreased from 60% to 20%, and hemoglobin levels rose from 10.3 to 14.2 g/dL.Citation43 Patients with the MYD88L265P CXCR4WT genotype showed the best response to treatment with MRR of 97.2% and shortest time to major response duration of 1.8 months.Citation43 For all patients, the 5-year survival rate was 87%.Citation43 For toxicities, 15.9% experienced a grade 3 or higher neutropenia, 11.1% experienced a grade 3 or higher thrombocytopenia, and 3.2% experienced a pneumonia.Citation43 Additionally, 8 patients (12.7%) experienced atrial arrhythmia, and of these, one had a grade 3 arrhythmia.Citation43 A total of 5 patients had to discontinue ibrutinib due to hematoma, thrombocytopenia, influenza-related pneumonia, streptococcal endocarditis, and atrial fibrillation.Citation43 Twelve patients had to reduce the dose of ibrutinib due to cytopenias, dermatitis or rash, stomatitis, leg edema, myalgias, and atrial fibrillation.Citation43 Overall, this study showed the efficacy and durability of response to ibrutinib without any unexpected toxicities.

Since the efficacy of ibrutinib monotherapy showed dependence on MYD88 and CXCR4 mutation status, the iNNOVATE trial was conducted to assess the efficacy and safety of ibrutinib in combination with rituximab in treating WM in 150 patients who had either received no prior treatment (n=68) or who had disease recurrence after prior systemic therapy (n=82).Citation44 Of those who had previously received systemic therapy, 70 patients had received a rituximab-containing regimen.Citation44 In this trial, 150 patients were randomly assigned to receive ibrutinib and rituximab or a placebo and rituximab.Citation44 At follow-up of 30 months, the PFS was higher in the ibrutinib-rituximab group at 82% compared to 28% in the groups who received placebo-rituximab.Citation44 MRR was also higher in the ibrutinib-rituximab group at 73% versus 41% in the placebo-rituximab group.Citation44 Furthermore, the higher response rates noted in the ibrutinib-rituximab group were independent of MYD88 and CXCR4 mutation status.Citation44 In terms of safety, the most common any-grade adverse events reported in the ibrutinib-rituximab group were infusion-related reactions, nausea, diarrhea, and arthralgias, and these occurred more frequently in this group compared to the placebo-rituximab group.Citation44 Grade 3 or higher adverse events that occurred more frequently in the ibrutinib-rituximab group included hypertension (13% vs 4%) and atrial fibrillation (12% vs 1%).Citation44 On the other hand, grade 3 or higher adverse events that occurred more frequently in the placebo-rituximab group included anemia (17% vs 11%) and infusion-related reactions (16% vs 1%).Citation44 The placebo-rituximab group also had higher incidences of any grade of IgM flare (47% vs 8%).Citation44 Major hemorrhage rates were the same in both groups at 4%.Citation44 At longer follow-up of 50 months, the median PFS was longer in the ibrutinib-rituximab group compared to the placebo-rituximab group, and the results showed a 75% reduction in risk of disease progression or death with ibrutinib-rituximab.Citation45 Estimated 54-month PFS rates were 68% and 25% in the ibrutinib-rituximab and placebo-rituximab groups, respectively.Citation45 PRR was also higher in the ibrutinib-rituximab group at 76% versus 31%.Citation45 Seventy-seven percent of patients who received ibrutinib-rituximab had sustained hemoglobin improvement compared to 43% in the placebo-rituximab group.Citation45 For safety, the prevalence of grade 3 or higher toxicities decreased over time.Citation45 A total of 15% experienced a grade 3 or higher hypertension, and 16% experienced a grade 3 or higher atrial fibrillation.Citation45 However, these events mostly occurred in the first 2–3 years of treatment, except for one event of grade 3 or higher hypertension occurring in the fifth year of treatment.Citation45 No major hemorrhage events were reported in the last 2 years of treatment.Citation45 Only one death was reported from a pneumonia, but this was deemed to be unrelated to the study drug.Citation45 Out of 75, a total of 8 patients discontinued ibrutinib and 17 patients reduced the dose of ibrutinib due to adverse events.Citation45 Most importantly, the final results from the iNNOVATE trial highlight that the combination of ibrutinib-rituximab greatly improves clinical outcomes regardless of MYD88 and CXCR4 genotype as well as prior treatment status.Citation45

Acalabrutinib

A next-generation highly selective BTKi, acalabrutinib has less interaction with non-BTK signaling pathways, which contributes to a less severe and different side effect profile compared to ibrutinib.Citation46 For these reasons, a single-arm, multicenter, international, Phase 2 study was conducted to evaluate the efficacy and safety of acalabrutinib monotherapy in WM.Citation46 A total of 122 patients were studied with 14 being treatment naïve and 92 having R/R disease.Citation46 At a median follow-up of 27.4 months, 93% of treatment naïve patients and 93% of R/R disease patients achieved an OR.Citation46 Median duration of response, PFS, and OS were not reached.Citation46 It should be noted that a limitation of this study is that responses were not analyzed based on MYD88 and CXCR4 mutational status via central laboratory assessment.Citation46 Thus, the impact of CXCR4 mutational status regarding acalabrutinib use is currently preliminary.Citation46 In terms of toxicities, grade 3 or 4 adverse events occurred in more than five percent of patients, and this included neutropenia (16%) and pneumonia (7%).Citation46 Only one patient (1%) had grade 3 or 4 atrial fibrillation/flutter and three (3%) had grade 3 or 4 hemorrhage.Citation46 One patient died from an intracranial hematoma that was deemed to be treatment related.Citation46

Zanubrutinib in WM

Overview

Zanubrutinib (BGB-3111) is a next-generation BTKi that was developed to achieve BTK inhibiting efficacy while reducing the off-target adverse events documented with ibrutinib therapy.Citation47,Citation48 With high specificity for the BTK adenosine triphosphate binding site for cysteine 481, zanubrutinib covalently binds to this site, resulting in irreversible, sustainable kinase inhibition.Citation47,Citation48 Compared to ibrutinib, zanubrutinib binds to the EGFR, JAK3, human EGFR2, ITK, and TEC protein kinases with 3.5, 58, 28, 19, and 3.5 times less affinity, respectively.Citation47 Additionally, the IC50 for zanubrutinib is 0.5 nM compared to 1.5 nM for ibrutinib.Citation49 Currently, Zanubrutinib is FDA approved for the treatment of MCL, MZL, and WM.

Efficacy

FDA approval for zanubrutinib in WM was granted on August 31, 2021 following the results of the ASPEN (NCT03053440) trial.Citation50 This trial was a randomized, open-label phase 3 study.Citation51 All patients enrolled had either R/R disease after at least one prior line of therapy or treatment-naïve disease unsuitable for standard chemotherapy based on documented comorbidities or risk factors.Citation51 In the first cohort, the study compared the efficacy and safety of zanubrutinib to ibrutinib in patients with MYD88L265P disease.Citation51 In the second cohort, the study evaluated the efficacy and safety of just zanubrutinib in MYD88WT disease.Citation51 For cohort 1, patients were randomized to receive either 160mg of zanubrutinib BID (n=102) or 420mg of ibrutinib QD (n=99).Citation51 For cohort 2, patients only received 160mg of zanubrutinib BID.Citation51 In cohort 1 within the group receiving zanubrutinib, 91 patients (89%) had the MYD88L265P/CXCR4WT genotype, and 11 (11%) had the MYD88L265P/CXCR4WHIM genotype.Citation51 Within the group receiving ibrutinib, 90 patients (91%) had the MYD88L265P/CXCR4WT genotype, 8 (8%) had the MYD88L265P/CXCR4WHIM genotype, and 1 (1.0%) had the MYD88L265P/CXCR4UNK genotype.Citation51 The two groups in cohort 1 were generally balanced well per baseline characteristics, but more patients assigned to receive zanubrutinib were above the age of 75 (33% vs 22%), and more were anemic (hemoglobin ≤110 g/L in 66% vs 54%).Citation51

In terms of responses, at median follow-up of 19.5 months, no patient achieved a complete response (CR).Citation51 A very good partial response (VPGR) was achieved in 28% of zanubrutinib patients and 19% of ibrutinib patients, but this difference was not statistically different (p=0.09).Citation51 MRRs were 77% and 78% in the zanubrutinib and ibrutinib groups, respectively.Citation51 One patient in each group with the CXCR4WHIM genotype achieved a VGPR.Citation51 On the other hand, 31% of zanubrutinib-treated patients and 20% of ibrutinib-treated patients with the CXCR4WT genotype achieved a VGPR.Citation51 Median time to a major response was 2.8 months for both groups with the CXCR4WT genotype.Citation51 However, in patients with the CXCR4WHIM genotype, median time to major response was 6.6 months with ibrutinib and 3.1 months with zanubrutinib.Citation51 At median follow-up of 18 months, 85% and 84% of zanubrutinib and ibrutinib patients were disease progression free, respectively.Citation51 Median PFS and duration of response (DOR) were not reached.Citation51 Estimated OS was 97% and 93% in the zanubrutinib and ibrutinib groups, respectively.Citation51 The primary end point for the ASPEN trial was the proportion of patients achieving a CR or VGPR by independent review.Citation51 Of note, this trial failed to show statistical superiority of zanubrutinib compared to ibrutinib based on the chosen primary end point.Citation51 Nevertheless, the safety profile seemed to benefit zanubrutinib instead of ibrutinib, hence we will discuss it in detail in the toxicity section.Citation51

For median IgM levels, these were reduced by 79% and 72% with zanubrutinib and ibrutinib, respectively.Citation51 However, it should be noted that zanubrutinib showcased significantly greater efficacy at reducing IgM levels, as well as sustaining lowered IgM levels, based on analysis with the repeated-measures mixed-effect model and area under the IgM × time curve comparisons.Citation51 For the zanubrutinib and ibrutinib groups, 69% and 73% patients had a reduction in bone marrow infiltration, with median maximal reductions of 10% and 15%, respectively.Citation51

Overall, the ASPEN study showed a greater frequency of VGPRs in zanubrutinib-treated patients compared to ibrutinib-treated patients.Citation51 It should be noted that in this study, post hoc analysis using next-generation sequencing for CXCR4 mutation detection instead of the original Sanger sequencing revealed 28% of patients with the CXCR4WHIM genotype compared to the originally reported 19%.Citation51 However, VGPR rates within this analysis were comparable to the originally discussed results.Citation51 Furthermore, this study showed that zanubrutinib therapy resulted in higher response rates among all patients (29% vs 21% in zanubrutinib-treated and ibrutinib-treated patients, respectively), among CXCR4WHIM genotype patients (18% vs 10%, respectively), and among CXCR4WT genotype patients (34% vs 24%, respectively) – showcasing the drug’s efficacy regardless of CXCR4 mutational status.Citation51 At longer follow-up of a median of 24.2 months, the combined CR and VGPR was 30.4% and 18.2% in the zanubrutinib-treated group and ibrutinib-treated group, respectively.Citation52

Another phase 1/2 study on zanubrutinib in 77 patients with treatment-naïve or R/R WM had patients take 160 mg twice daily (n = 50) or 320 mg once daily (n = 23).Citation53 At a long-term median of 36-months follow-up, ORR and VGPR were reported to be 96% and 45%.Citation53 Estimated 3-year PFS and OS were reported as 80.5% and 84.8%.Citation53 In terms of genotype responses, 49.1% and 25% of patients with the MYD88L265 and MYD88WT genotypes achieved a VGPR/CR.Citation53 Additionally, 59% and 27.3% of patients with the MYD88L265P/CXCR4WT and MYD88L265P/CXCR4WHIM genotypes achieved a VGPR/CR.Citation53 However, MRRs were comparable (87.2% vs 90.9%) in the groups with and without the CXCR4 mutation.Citation53 Ultimately, this study showed long-term durable responses with single-agent zanubrutinib in WM.Citation53

Finally, a single-arm, multicenter, phase II study investigated the efficacy and safety of 160mg of zanubrutinib twice daily in 44 Chinese patients with R/R WM.Citation54 The focus on an Asian population for this study was done due to the underrepresentation of this population in prior investigations.Citation54 At a median follow-up of 33 months, MRR and VPGR rates were reported as 69.8% and 32.6%, respectively.Citation54 Additionally, patients with the MYD88L265 and MYD88WT genotypes achieved MRRs of 73% and 50%, respectively.Citation54 Overall, the study found higher response rates in the MYD88L265P/CXCR4WT group when compared to the other groups.Citation54 For all patients, the 24-month PFS and OS were reported as 60.5% and 87.8%, respectively.Citation54 Overall, this study displayed zanubrutinib’s efficacy in treating WM in Asian populations.

Toxicity

In the ASPEN study, in the group who received zanubrutinib, at a median follow-up of 19.4 months, the most common adverse events of any grade were neutropenia (29%), upper respiratory infection (24%), and diarrhea (21%).Citation51 On the other hand, in the ibrutinib-treated group, the most common adverse events of any grade were diarrhea (32%), upper respiratory infection (29%), contusion (24%), and muscle spasms (24%).Citation51 In the zanubrutinib and ibrutinib treated groups, grade 3 or higher adverse rates occurred in 58% and 63% of patients, respectively.Citation51 Grade 3 or higher hypertension and pneumonia occurred more frequently in the ibrutinib-treated patients while a grade 3 or higher neutropenia occurred in the zanubrutinib-treated patients.Citation51 Additionally, 40% and 41% of zanubrutinib and ibrutinib-treated patients experienced at least one serious adverse event, respectively.Citation51 The most common serious adverse events in the zanubrutinib versus ibrutinib groups were pneumonia (seen in 1 vs 9 patients), neutropenia (3 vs 0), influenza (3 vs 1), and sepsis (2 vs 3).Citation51 Atrial fibrillation/flutter occurred within 6 months of treatment in 1 zanubrutinib-treated patient and 7 ibrutinib-treated patients; beyond 12 months of treatment, this occurred in only an additional 4 ibrutinib-treated patients.Citation51 Hypertension also occurred beyond 12 months of treatment in 1 zanubrutinib-treated patient and 6 ibrutinib-treated patients.Citation51 However, zanubrutinib-treated patients had a much greater incidence of any grade neutropenia (25% vs 12%) and grade 3 or higher neutropenia (20% vs 8%) when compared to ibrutinib-treated patients.Citation51 Two ibrutinib-treated patients died from complications related to sepsis, and one zanubrutinib-treated patient died from complications of cardiac arrest.Citation51

At longer median 24.2 months of follow-up, the common adverse events of any grade to occur in the zanubrutinib-treated versus ibrutinib-treated groups were atrial fibrillation/flutter (3.0% vs 18.4%), bleeding (50.5% vs 60.2%), major hemorrhage (5.9% vs 10.2%), diarrhea (21.8% vs 32.7%), hypertension (12.9% vs 20.4%) and neutropenia (31.7% vs 15.3%).Citation52 Zanubrutinib-treated patients had higher rates of grade 3 or higher neutropenia compared to ibrutinib-treated patients (22.8% vs 8.2%).Citation52 However, grade 3 or higher infection rates were comparable between the zanubrutinib-treated and ibrutinib-treated patients (18.8% vs 23.5%).Citation52 No additional patients had any adverse event that led to death.Citation52

In the phase 1/2 study, at long-term median follow-up of 36 months, all patients had experienced at least one grade 1 adverse event, and 58.4% reported at least one grade 3 or higher adverse event.Citation53 The most common adverse events of any grade were upper respiratory tract infection (51.9%), contusion (32.5%), and cough (22.1%).Citation53 For the grade 3 or higher adverse events, the most commonly reported ones were neutropenia (15.6%), anemia (9.1%), basal cell carcinoma (5.2%), cellulitis (5.2%), hypertension (3.9%), pneumonia (3.9%), diarrhea (2.6%), headache (2.6%), fall (2.6%), and actinic keratosis (2.6%).Citation53 A total of nine patients died during the study; two died from progressive disease, two died from unknown causes, and five died from adverse events.Citation53 These adverse events leading to death included abdominal sepsis, bacterial arthritis, Scedosporium infection, gastric adenocarcinoma, and worsening bronchiectasis.Citation53 However, it should be noted, that four of the five adverse events leading to death were deemed to be unrelated to the study drug.Citation53

In the Asian population focused study, at median follow-up of 33 months, the most frequently reported treatment-emergent adverse events were neutropenia (59.1%), lymphopenia (31.8%), upper respiratory tract infection (29.5%), pneumonia (29.5%), and thrombocytopenia (29.5%).Citation54 In terms of grade 3 or higher adverse events, the most reported ones were neutropenia (31.8%), thrombocytopenia (20.5%), pneumonia (20.5%), lymphopenia (11.4%), and hypertension (11.4%).Citation54 It should be noted that none of the patients in this study experienced atrial fibrillation or tumor lysis syndrome.Citation54 Serious adverse events occurred in 56.8% of patients and these were pneumonia (22.7%), upper respiratory tract infection (6.8%), skin infection (4.5%), and pleural effusion (4.5%).Citation54 Seven patients (15.9%) died during the study.Citation54 Three died from disease progression, three from adverse events, and one from severe pneumonia and respiratory failure.Citation54 The adverse events leading to death were acute hepatitis B multi-organ dysfunction syndrome, and respiratory failure.Citation54

Tolerability

In the ASPEN study, at median follow-up of 19.4 months, the median treatment durations for zanubrutinib and ibrutinib were 18.7 months and 18.6 months, respectively.Citation51 For both groups, the median dose intensity was 98%. Due to adverse events, 23% of ibrutinib-treated patients needed to have their dose reduced, while 14% of zanubrutinib-treated patients required a dose reduction. Nine percent of ibrutinib-treated patients discontinued treatment altogether for adverse events of myocardial infarction, sepsis, death, drug-induced liver injury, hepatitis, interstitial lung disease, and pneumonia. On the other hand, 4% of zanubrutinib-treated patients discontinued treatment for adverse events of subdural hemorrhage, cardiac arrest, neutropenia, and IgA multiple myeloma. At a longer follow-up of 24.2 months, no additional zanubrutinib-treated patients discontinued treatment due to adverse events.Citation52 However, 5 additional ibrutinib-treated patients discontinued treatment.Citation52 Thus, these findings illustrated greater tolerability of zanubrutinib compared to ibrutinib.

In the long-term phase 1/2 study evaluating zanubrutinib alone, at a median 36-months of follow-up, only 72.7% of patients remained on treatment.Citation53 Thirteen percent discontinued zanubrutinib because of adverse events, of which only one was attributed to the treatment.Citation53 On the other hand, 10.4% had to discontinue treatment because of disease progression, and 3.9% discontinued for other reasons.Citation53 At long-term follow-up, no patients required dose reduction of zanubrutinib.Citation53

In the Asian population focused study, at a median follow-up of 33 months, seventeen patients (38.6%) had to have treatment interruption rather than reduction due to adverse events including pneumonia (13.6%), thrombocytopenia (6.8%), neutropenia (4.5%), and thrombocytopenia (4.5). After supportive treatment, most of these patients were able to resume treatment. However, six patients (16.3%) had to discontinue treatment altogether because of adverse events.Citation54 These included pneumonia, laryngeal cancer, rapid WM disease progression with suspected transformation to aggressive lymphoma, intracranial mass, and acute hepatitis B with multi-organ dysfunctional syndrome.Citation54 Only one patient in this study had to reduce their dose of the drug because of arthralgias, and these resolved with no recurrence.Citation54

Future Trials

A search was conducted on May 03, 2022, on the current clinical trials on zanubrutinib in WM on clinicaltrials.gov.Citation55 Studies not yet recruiting, recruiting, enrolling by invitation, active (not recruiting), suspended, and completed are summarized in . Studies with a status of terminated, unknown, and withdrawn were excluded.

Table 1 A Comprehensive List of the Current (as of May 03, 2022) Clinical Trials Being Conducted on Zanubrutinib and WM

Beyond the therapies described above, chimeric antigen receptor (CAR) T-cell therapies have shown great efficacy in treating a variety of B-cell malignancies. This therapy involves using a patient’s own T-cells and genetically engineering the cells to express a T-cell receptor that targets a specified tumor antigen.Citation56 The current FDA approved CAR T-cell therapies include axicabtagene ciloleucel, tisagenlecleucel, lisocabtagene maraleucel, brexucabtagene autoleucel, idecabtagene vicleucel, and ciltacabtagene autoleucel. Clinical trials evaluating the efficacy of these therapies have shown strong ORR and CR in R/R B-cell malignancies. Although all the therapies discussed above have shown efficacy in treating WM, the studies eventually have reported disease relapse or progression. Additionally, the studies have discussed dose reduction or treatment cessation due to intolerance. Therefore, to address these issues, studies are looking to evaluate the efficacy and safety of CAR T-cell therapy in WM. So far, one study has reported that three patients with R/R WM treated with anti-CD19 CAR T-cell therapy had a clinical response to treatment.Citation57 However, all three of them had disease recurrence within 3 to 26 months after the initial treatment.Citation57 Of note, CAR T-cell therapy was well tolerated in all patients and no grade 3 or higher events of cytokine release syndrome or neurotoxicity occurred.Citation57 We greatly look forward to further future studies evaluating the efficacy and safety of CAR T-cell therapy in WM.

Conclusions

Within the therapeutic landscape of WM, BTK inhibitors have emerged as greatly effective therapies. The first BTKi used in WM was ibrutinib; however, this treatment as a single agent displayed reduced efficacy in patients with the CXCR4 mutation. Additionally, ibrutinib was associated with many adverse events that led to drug dose reduction or discontinuation. Subsequently, the newer, highly specific BTKi zanubrutinib emerged. This drug showed efficacy in treating WM, regardless of CXCR4 mutation status, with a favorable toxicity profile. summarizes the efficacy and safety data from the main clinical trials evaluating zanubrutinib in WM. Furthermore, randomized trials comparing zanubrutinib directly to ibrutinib in B-cell lymphoproliferative disorders such as CLL or WM showed that zanubrutinib conferred fewer toxicities and greater tolerability. Thus, the last of this drug class to become FDA approved in B-cell lymphoproliferative disorders, zanubrutinib, rises as a major player for the treatment of WM. Certainly, additional non-covalent BTKi are under development which, if proven to be safer and perhaps more efficacious than current options available, will reinforce the paradigm that the last shall be first as the therapeutic field continues to evolve.

Table 2 A Summative Table on the Data Published in the Clinical Trials Evaluating Zanubrutinib in WM

Disclosure

Javier Munoz reports personal fees from consulting for Pharmacyclics/AbbVie, Bayer, Gilead/Kite Pharma, Pfizer, Janssen, Juno/Celgene, BMS, Kyowa, Alexion, Fosunkite, Innovent, Seattle Genetics, Debiopharm, Karyopharm, Genmab, ADC Therapeutics, Epizyme, Beigene, Servier, Novartis, and Morphosys/Incyte, grants from research funding from Bayer, Gilead/Kite Pharma, Celgene, Merck, Portola, Incyte, Genentech, Pharmacyclics, Seattle Genetics, Janssen, Millennium, personal fees from Honoraria from Targeted Oncology, OncView, Curio, Kyowa, Physicians’ Education Resource, and Seattle Genetics, personal fees from speaker’s bureaus for Gilead/Kite Pharma, Kyowa, Bayer, Pharmacyclics/Janssen, Seattle Genetics, Acrotech/Aurobindo, Beigene, Verastem, AstraZeneca, Celgene/BMS, and Genentech/Roche, during the conduct of the study. The authors report no other potential conflicts of interest in relation to this work.

References

  • Fowler N, Davis E. Targeting B-cell receptor signaling: changing the paradigm. Hematology Am Soc Hematol Educ Program. 2013;2013(1):553–560. doi:10.1182/asheducation-2013.1.553
  • Herman SE, Gordon AL, Hertlein E, et al. Bruton tyrosine kinase represents a promising therapeutic target for treatment of chronic lymphocytic leukemia and is effectively targeted by PCI-32765. Blood. 2011;117(23):6287–6296. doi:10.1182/blood-2011-01-328484
  • Vetrie D, Vořechovský I, Sideras P, et al. The gene involved in X-linked agammaglobulinaemia is a member of the src family of protein-tyrosine kinases. Nature. 1993;361(6409):226–233. doi:10.1038/361226a0
  • Palma M, Mulder TA, Osterborg A. BTK inhibitors in chronic lymphocytic leukemia: biological activity and immune effects. Front Immunol. 2021;12:686768. doi:10.3389/fimmu.2021.686768
  • Tam CS, Dimopoulos M, Garcia-Sanz R, et al. Pooled safety analysis of zanubrutinib monotherapy in patients with B-cell malignancies. Blood Adv. 2022;6(4):1296–1308. doi:10.1182/bloodadvances.2021005621
  • Cull G, Burger JA, Opat S, et al. Zanubrutinib for treatment-naive and relapsed/refractory chronic lymphocytic leukaemia: long-term follow-up of the phase I/II AU-003 study. Br J Haematol. 2022;196(5):1209–1218. doi:10.1111/bjh.17994
  • Tam CS, Trotman J, Opat S, et al. Phase 1 study of the selective BTK inhibitor zanubrutinib in B-cell malignancies and safety and efficacy evaluation in CLL. Blood. 2019;134(11):851–859. doi:10.1182/blood.2019001160
  • Wang ML, Rule S, Martin P, et al. Targeting BTK with ibrutinib in relapsed or refractory mantle-cell lymphoma. N Engl J Med. 2013;369(6):507–516. doi:10.1056/NEJMoa1306220
  • Tam CS, Opat S, Simpson D, et al. Zanubrutinib for the treatment of relapsed or refractory mantle cell lymphoma. Blood Adv. 2021;5(12):2577–2585. doi:10.1182/bloodadvances.2020004074
  • Davis RE, Ngo VN, Lenz G, et al. Chronic active B-cell-receptor signalling in diffuse large B-cell lymphoma. Nature. 2010;463(7277):88–92. doi:10.1038/nature08638
  • Noy A, de Vos S, Thieblemont C, et al. Targeting Bruton tyrosine kinase with ibrutinib in relapsed/refractory marginal zone lymphoma. Blood. 2017;129(16):2224–2232. doi:10.1182/blood-2016-10-747345
  • Hunter ZR, Yang G, Xu L, Liu X, Castillo JJ, Treon SP. Genomics, signaling, and treatment of waldenstrom macroglobulinemia. J Clin Oncol. 2017;35(9):994–1001. doi:10.1200/JCO.2016.71.0814
  • Castillo JJ, Moreno DF, Arbelaez MI, Hunter ZR, Treon SP. CXCR4 mutations affect presentation and outcomes in patients with Waldenstrom macroglobulinemia: a systematic review. Expert Rev Hematol. 2019;12(10):873–881. doi:10.1080/17474086.2019.1649132
  • Poulain S, Roumier C, Decambron A, et al. MYD88 L265P mutation in Waldenstrom macroglobulinemia. Blood. 2013;121(22):4504–4511. doi:10.1182/blood-2012-06-436329
  • Treon SP, Hunter ZR, Aggarwal A, et al. Characterization of familial Waldenström’s macroglobulinemia. Ann Oncol. 2006;17(3):488–494. doi:10.1093/annonc/mdj111
  • Vijay A, Gertz MA. Waldenström macroglobulinemia. Blood. 2007;109(12):5096–5103. doi:10.1182/blood-2006-11-055012
  • Fadil A, Taylor DE. The lung and Waldenstrom’s macroglobulinemia. South Med J. 1998;91(7):681–685. doi:10.1097/00007611-199807000-00017
  • Rosenthal JA, Curran WJ Jr., Schuster SJ. Waldenstrom’s macroglobulinemia resulting from localized gastric lymphoplasmacytoid lymphoma. Am J Hematol. 1998;58(3):244–245. doi:10.1002/(SICI)1096-8652(199807)58:3<244::AID-AJH16>3.0.CO;2-9
  • Varettoni M, Defrancesco I, Diamanti L, Marchioni E, Farina LM, Pichiecchio A. Bing-Neel syndrome: illustrative cases and comprehensive review of the literature. Mediterr J Hematol Infect Dis. 2017;9(1):e2017061. doi:10.4084/mjhid.2017.061
  • Castillo JJ, Itchaki G, Paludo J, et al. Ibrutinib for the treatment of Bing-Neel syndrome: a multicenter study. Blood. 2019;133(4):299–305. doi:10.1182/blood-2018-10-879593
  • Hashmi H, Dhanoa JS, Emmons R. Rare case of Bing-Neel syndrome treated successfully with ibrutinib. BMJ Case Rep. 2019;12(6):e230067. doi:10.1136/bcr-2019-230067
  • Rosenthal A, Munoz J. Zanubrutinib for the treatment of B-cell malignancies. touchRev Oncol Haematol. 2022;18:1.
  • NCCN; National Comprehensive Cancer Network. NCCN Guidelines. Available from: https://www.nccn.org/guidelines/guidelines-detail?category=1&id=1475. Accessed May 3, 2022.
  • Vital C, Vital A, Deminiere C, Julien J, Lagueny A, Steck AJ. Myelin modifications in 8 cases of peripheral neuropathy with Waldenstrom’s macroglobulinemia and anti-MAG activity. Ultrastruct Pathol. 1997;21(6):509–516. doi:10.3109/01913129709016367
  • Chaudhry HM, Mauermann ML, Rajkumar SV. Monoclonal gammopathy-associated peripheral neuropathy: diagnosis and management. Mayo Clin Proc. 2017;92(5):838–850. doi:10.1016/j.mayocp.2017.02.003
  • Berkman EM, Orlin JB. Use of plasmapheresis and partial plasma exchange in the management of patients with cryoglobulinemia. Transfusion. 1980;20(2):171–178. doi:10.1046/j.1537-2995.1980.20280169957.x
  • Menke MN, Feke GT, McMeel JW, Treon SP. Effect of plasmapheresis on hyperviscosity-related retinopathy and retinal hemodynamics in patients with Waldenstrom’s macroglobulinemia. Invest Ophthalmol Vis Sci. 2008;49(3):1157–1160. doi:10.1167/iovs.07-1254
  • Stone MJ, Bogen SA. Role of plasmapheresis in Waldenstrom’s macroglobulinemia. Clin Lymphoma Myeloma Leuk. 2013;13(2):238–240. doi:10.1016/j.clml.2013.02.013
  • Dimopoulos MA, Zervas C, Zomas A, et al. Treatment of Waldenstrom’s macroglobulinemia with rituximab. J Clin Oncol. 2002;20(9):2327–2333. doi:10.1200/JCO.2002.09.039
  • Gertz MA, Anagnostopoulos A, Anderson K, et al. Treatment recommendations in Waldenstrom’s macroglobulinemia: consensus panel recommendations from the second International Workshop on Waldenstrom’s Macroglobulinemia. Semin Oncol. 2003;30(2):121–126. doi:10.1053/sonc.2003.50039
  • Zheng YH, Xu L, Cao C, et al. Rituximab-based combination therapy in patients with Waldenstrom macroglobulinemia: a systematic review and meta-analysis. Onco Targets Ther. 2019;12:2751–2766. doi:10.2147/OTT.S191179
  • Ghobrial IM, Fonseca R, Greipp PR, et al. Initial immunoglobulin M ‘flare’ after rituximab therapy in patients diagnosed with Waldenstrom macroglobulinemia: an Eastern Cooperative Oncology Group Study. Cancer. 2004;101(11):2593–2598. doi:10.1002/cncr.20658
  • Castillo JJ, Kanan S, Meid K, Manning R, Hunter ZR, Treon SP. Rituximab intolerance in patients with Waldenstrom macroglobulinaemia. Br J Haematol. 2016;174(4):645–648. doi:10.1111/bjh.13794
  • Rummel MJ, Niederle N, Maschmeyer G, 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):1203–1210. doi:10.1016/S0140-6736(12)61763-2
  • Rummel MJ, Lerchenmüller C, Hensel M, et al. Two years rituximab maintenance vs. observation after first Line treatment with bendamustine plus rituximab (B-R) in patients with Waldenström’s Macroglobulinemia (MW): results of a prospective, randomized, multicenter phase 3 study (the StiL NHL7-2008 MAINTAIN trial). Blood. 2019;134:343.
  • Dimopoulos MA, Anagnostopoulos A, Kyrtsonis M-C, et al. Primary treatment of Waldenström macroglobulinemia with dexamethasone, rituximab, and cyclophosphamide. J Clin Oncol. 2007;25(22):3344–3349. doi:10.1200/JCO.2007.10.9926
  • Treon SP, Hunter ZR, Matous J, et al. Multicenter clinical trial of bortezomib in relapsed/refractory Waldenstrom’s macroglobulinemia: results of WMCTG Trial 03-248. Clin Cancer Res. 2007;13(11):3320–3325. doi:10.1158/1078-0432.CCR-06-2511
  • Sklavenitis-Pistofidis R, Capelletti M, Liu CJ, et al. Bortezomib overcomes the negative impact of CXCR4 mutations on survival of Waldenstrom macroglobulinemia patients. Blood. 2018;132(24):2608–2612. doi:10.1182/blood-2018-07-863241
  • Chen CI, Kouroukis CT, White D, et al. Bortezomib is active in patients with untreated or relapsed Waldenstrom’s macroglobulinemia: a phase II study of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol. 2007;25(12):1570–1575.
  • Castillo JJ, Allan JN, Siddiqi T, et al. Venetoclax in previously treated Waldenström Macroglobulinemia. J Clin Oncol. 2022;40(1):63–71. doi:10.1200/JCO.21.01194
  • Papanota AM, Ntanasis-Stathopoulos I, Kastritis E, Dimopoulos MA, Gavriatopoulou M. Evaluating ibrutinib in the treatment of symptomatic Waldenstrom’s macroglobulinemia. J Blood Med. 2019;10:291–300. doi:10.2147/JBM.S183997
  • Treon SP, Tripsas CK, Meid K, et al. Ibrutinib in previously treated Waldenström’s Macroglobulinemia. N Engl J Med. 2015;372(15):1430–1440. doi:10.1056/NEJMoa1501548
  • Treon SP, Meid K, Gustine J, et al. Long-term follow-up of ibrutinib monotherapy in symptomatic, previously treated patients with Waldenström Macroglobulinemia. J Clin Oncol. 2021;39(6):565–575. doi:10.1200/JCO.20.00555
  • Dimopoulos MA, Tedeschi A, Trotman J, et al. Phase 3 trial of ibrutinib plus rituximab in Waldenström’s Macroglobulinemia. N Engl J Med. 2018;378(25):2399–2410. doi:10.1056/NEJMoa1802917
  • Buske C, Tedeschi A, Trotman J, et al. Ibrutinib plus rituximab versus placebo plus rituximab for Waldenström’s Macroglobulinemia: final analysis from the randomized Phase III iNNOVATE study. J Clin Oncol. 2022;40(1):52–62. doi:10.1200/JCO.21.00838
  • Owen RG, McCarthy H, Rule S, et al. Acalabrutinib monotherapy in patients with Waldenström macroglobulinemia: a single-arm, multicentre, phase 2 study. Lancet Haematol. 2020;7(2):e112–e121. doi:10.1016/S2352-3026(19)30210-8
  • Guo Y, Liu Y, Hu N, et al. Discovery of Zanubrutinib (BGB-3111), a novel, potent, and selective covalent inhibitor of Bruton’s tyrosine kinase. J Med Chem. 2019;62(17):7923–7940. doi:10.1021/acs.jmedchem.9b00687
  • Ou YC, Liu L, Tariq B, et al. Population pharmacokinetic analysis of the BTK inhibitor zanubrutinib in healthy volunteers and patients with B-cell malignancies. Clin Transl Sci. 2021;14(2):764–772. doi:10.1111/cts.12948
  • Tam CS, Ou YC, Trotman J, Opat S. Clinical pharmacology and PK/PD translation of the second-generation Bruton’s tyrosine kinase inhibitor, zanubrutinib. Expert Rev Clin Pharmacol. 2021;14(11):1329–1344. doi:10.1080/17512433.2021.1978288
  • FDA approves zanubrutinib for Waldenström’s macroglobulinemia. U.S. Food & Drug Administration; 2021. Available from: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-zanubrutinib-waldenstroms-macroglobulinemia. Accessed February 22, 2022.
  • Tam CS, Opat S, D’Sa S, et al. A randomized phase 3 trial of zanubrutinib vs ibrutinib in symptomatic Waldenström macroglobulinemia: the ASPEN study. Blood. 2020;136(18):2038–2050. doi:10.1182/blood.2020006844
  • BeiGene presents updated head to head results from phase 3 trial of zanubrutinib vs. ibrutinib in patients with Waldenström’s Macroglobulinemia at the 2020 American Society of Clinical Oncology (ASCO) virtual scientific program. BeiGene, LTD; 2020. Available from: https://ir.beigene.com/news-details/?id=f4dce275-d7c9-4f50-8f0d-2d2ce07db346. Accessed 2022.
  • Trotman J, Opat S, Gottlieb D, et al. Zanubrutinib for the treatment of patients with Waldenstrom macroglobulinemia: 3 years of follow-up. Blood. 2020;136(18):2027–2037. doi:10.1182/blood.2020006449
  • An G, Zhou D, Cheng S, et al. A Phase II trial of the Bruton tyrosine-kinase inhibitor zanubrutinib (BGB-3111) in patients with relapsed/refractory Waldenström Macroglobulinemia. Clin Cancer Res. 2021;27(20):5492–5501. doi:10.1158/1078-0432.CCR-21-0539
  • NIH. ClinicalTrials.gov; 2022. Available from: https://clinicaltrials.gov/ct2/results?cond=Waldenstrom+Macroglobulinemia&term=BGB-3111&cntry=&state=&city=&dist=&Search=Search&recrs=a&recrs=b&recrs=d&recrs=e&recrs=f&recrs=g. Accessed February 20, 2022.
  • Feins S, Kong W, Williams EF, Milone MC, Fraietta JA. An introduction to chimeric antigen receptor (CAR) T-cell immunotherapy for human cancer. Am J Hematol. 2019;94(S1):S3–S9. doi:10.1002/ajh.25418
  • Palomba ML, Qualls D, Monette S, et al. CD19-directed chimeric antigen receptor T cell therapy in Waldenström macroglobulinemia: a preclinical model and initial clinical experience. J Immunother Cancer. 2022;10(2):e004128. doi:10.1136/jitc-2021-004128