117
Views
1
CrossRef citations to date
0
Altmetric
Review

New developments in the management of Waldenström macroglobulinemia

, &
Pages 73-83 | Published online: 10 Mar 2017

Abstract

Waldenström macroglobulinemia (WM) is a rare, immunoglobulin M -associated lymphoplasmacytic lymphoma. With the recent discoveries of CXCR warts, hypogammaglobulinemia, infections, and myelokathexis (WHIM) and MYD88 mutations, our understanding of the biology of WM has expanded substantially. While WM still remains incurable, the field is rapidly evolving, and a number of promising agents with significant activity in this malignancy are being evaluated currently. In this review, we discuss the new developments that have occurred in WM over the past 15 years, with a focus on the role of ibrutinib, an oral Bruton’s tyrosine kinase inhibitor that has recently been approved for WM in the United States, Europe, and Canada.

Introduction

The report of two patients by WaldenströmCitation1 in the year 1944, presenting with epistaxis, hematemesis, lymphadenopathy, decreased fibrinogen level, increased blood viscosity, and elevated macroglobulins, was the first account of Waldenström macroglobulinemia (WM). WM is a rare, indolent lymphoma with lymphoplasmacytic infiltration of the bone marrow (BM) and monoclonal immunoglobulin M (IgM) gammopathy. The management of WM is evolving, with a deeper understanding of the disease pathophysiology and introduction of newer drugs. WM remains an incurable disease, with a median overall survival (OS) of ~8 years from diagnosis.Citation2,Citation3 In this review, we discuss new developments in the management of WM based on the data published over the past 15 years, with an emphasis on the role of Bruton’s tyrosine kinase (BTK) inhibitor, ibrutinib, that has been recently approved for WM in the US, Canada, and European Union. To better serve our patients, a holistic understanding of this fascinating malignancy and its current and emerging therapeutic options remains paramount, and this review has been written with the intention of making our patients the ultimate beneficiaries.

Epidemiology

Approximately 1,500 new cases of WM are diagnosed in the US each year.Citation4 The median age at diagnosis is ~70 years, with an incidence rate of 3.4 per million among men and 1.7 per million among women in the US.Citation2 Caucasians are predominantly affected.Citation5

Although no proven inheritance patterns have been observed in WM to date, a population-based study was remarkable for demonstrating a 20-fold increased risk of development of lymphoplasmacytic lymphoma (LPL)/WM in the first-degree relatives of the patients.Citation6 Moreover, the first-degree relatives were noted to have a three- to fivefold increased risk of developing another non-Hodgkin’s lymphoma, chronic lymphocytic leukemia, or monoclonal gammopathy of undetermined significance.Citation6

Diagnosis

The Mayo Clinic criteria that must be met for the diagnosis of active WM are

  • serum IgM monoclonal protein of any size

  • BM lymphoplasmacytic infiltration of at least 10%.

Besides the BM, the lymphoplasmacytic cells can infiltrate extramedullary sites, predominantly the lymph nodes and the spleen, with a spectrum of clonal B cells, including small lymphocytes, plasmacytoid lymphocytes (showing features of both plasma cells and lymphocytes), and plasma cells. In contrast to the Mayo Clinic criteria, the International Workshop on Waldenström’s Macroglobulinemia (IWWM) Consensus criteria merely require the presence of a lymphoplasmacytic infiltrate in the marrow and serum IgM monoclonal protein of any size to establish a diagnosis of WM. The typical immunophenotypic signature of the infiltrate is surface IgM+, CD5±, CD10, CD19+, CD20+, CD22+, CD23, CD25+, CD27+, FMC7+, CD103, and CD138+.Citation7,Citation8

Pathogenesis

Although the exact etiology of WM remains unclear to date, the whole genome sequencing (WGS) has identified important mutations that appear to be pathogenic. Recent studies have partially unraveled the role of myeloid differentiation factor 88 (MYD88) mutations in the pathogenesis of WM. The MYD88 protein is involved in the signal transduction pathways activating nuclear factor kappa B (NFκβ) and mitogen-activated protein kinase (MAPK).Citation9 Its activation is enhanced by the mutations occurring in the MYD88 gene, which induce tumorigenesis.Citation9 The WGS in WM has identified a somatic variant (T → C) at position 38182641 in chromosome 3p22.2 that harbors the MYD88 gene.Citation10 The MYD88L265P mutation leads to an amino-acid change from leucine to proline (L265P) in the MYD88 protein.Citation10 The protein is capable of hyperactivating NFκβ via interleukin-1 receptor-associated kinase (IRAK) and BTK pathways through phosphorylation, thereby increasing cell survival.Citation11 Treon et alCitation10 reported that MYD88L265P is a common mutation in WM with a prevalence of 91%. Patients with MYD88L265P mutation are noted to have distinct disease profile, including higher serum IgM levels, hyperviscosity, and higher BM involvement compared to their counterparts harboring the MYD88 wild-type gene.Citation12

Another seminal discovery is that of the somatic mutations involving the C-X-C chemokine receptor type 4 (CXCR4, also known as Fusin or CD184), encoded by the CXCR4 gene.Citation13,Citation14 The association in WM bears a striking similarity to the finding observed in the warts, hypogammaglobulinemia, infections, and myelokathexis (WHIM) syndrome that also results from a mutation in the CXCR4 gene.Citation12,Citation15 The CXCR4 mutations have been observed in 27–40% of patients with WM and were found to be involved with its pathogenesis.Citation12,Citation15,Citation16 These mutations lead to the formation of a truncated receptor protein associated with a high expression of CXCR4 receptor and could be either nonsense (CXCR4WHIM/NS) type or frameshift (CXCR4WHIM/FS) type.

Genotypic–phenotypic association

A single retrospective study by the Dana–Farber Cancer InstituteCitation12 noted that the MYD88L265P with CXCR4WHIM/NS genotype was associated with severe disease, greater BM involvement, and increased likelihood of developing hyperviscosity-related complications with higher serum IgM levels.

The patients with MYD88L265P and CXCR4WHIM/FS genotypes also exhibit an aggressive disease course. However, the disease severity of patients with MYD88L265P and CXCR4WHIM/FS genotypes appears milder compared to their MYD88L265P and CXCR4WHIM/NS counterparts ().Citation12,Citation15 The patients with MYD88WTCXCR4WT show the lowest marrow involvement.Citation12 Despite the association of severe disease with MYD88L265P and CXCR4WHIM/NS or FS mutations compared to MYD88WTCXCR4WT, the survival outcomes do not appear to be affected by the presence of CXCR4 mutations. Rather, the outcomes appear to be impacted by the MYD88 mutation status and were found to be surprisingly better for the patients harboring MYD88L265P gene than the patients harboring MYD88WT gene in one study.Citation12 Importantly, we now know that the presence of MYD88 and CXCR4 mutations will affect the degree of response to ibrutinib (discussed in the subsequent section). Another recent article on the transcriptional profiling (RNAseq) and comparison of the WM patients to the normal population without B-cell disorders attempted to shed more light on this matter. Four genotypic groups as indicated in are currently identifiable.Citation16,Citation17

Table 1 Genotypic–phenotypic association in WM

Clinical presentation

The clinical presentation and the disease characteristics of WM could be attributed to tumor/B-lymphocyte infiltration or monoclonal immunoglobulins as summarized in .

Table 2 Disease characteristics of WM

Presentation of WM could be heterogenous. Some of the most common presenting features are hyper viscosity, constitutional symptoms, bleeding, and neurologic symptoms.Citation18,Citation19 Lymphadenopathy, hepatomegaly, splenomegaly, and funduscopic abnormalities may be detected.Citation18,Citation19 Anemia is a common presenting feature, and type I cryoglobulinemia may be infrequently encountered.Citation18,Citation19

Prognosis and natural history

A large study involving >5,000 patients with WM from 1991 to 2010 showed that the median OS for the entire cohort was 7 years and the 5- and 10-year OS were 62% and 39%, respectively.Citation2 The 5-year OS for patients who were diagnosed prior to the age of 70 years compared to those older than 70 years was 71% and 39%, respectively.Citation4 The OS has increased over the last two decades; the median OS for the patients diagnosed between 1991 and 2000 was 6 vs 8.2 years for those diagnosed between 2001 and 2010, P<0.05.Citation2,Citation20

Smoldering WM

At the time of diagnosis, ~25% of patients with WM do not have symptoms or signs that are attributable to an increased monoclonal protein or infiltration by malignant cells. This population of patients is considered to have smoldering WM, and there are no indications for the initiation of treatment until the development of symptoms or significant anemia/thrombocytopenia in this patient population given the risks involved and the lack of survival advantage with early treatment.Citation21 The OS rate of patients with smoldering WM can approximate that of the normal age-matched population.Citation8 A Mayo Clinic study demonstrated that the rate of progression of patients with smoldering WM to symptomatic WM during a 15-year follow-up was 71%, and the cumulative risk of progression of smoldering WM to symptomatic WM, amyloidosis, or a related disorder was 6% at 1 year, 39% at 3 years, 59% at 5 years, and 68% at 10 years.Citation22

Prognosis

The International Prognostic Symptom Score (IPSS) for WM was created to assess the disease prognosis and survival risk in patients with active WM who require treatment ().Citation23

Table 3 Disease prognosis based on IPSS

Treatment

The optimal initial approach requires assessment of the symptomatology, the genetic signature, ie, MYD88 status (CXCR4 status is not routinely used in practice at present), the monoclonal protein size, age of the patient, and certain laboratory parameters. The decision to commence therapy typically should not be based on the size of serum IgM size.Citation24 The treatment indications for symptomatic patients are outlined in .Citation21,Citation24,Citation25

Table 4 Treatment indications for symptomatic patients

Furthermore, the therapy-related decisions depend on the rapidity with which the symptom control is necessary. Hyperviscosity-related symptoms warrant urgent plasmapheresis.Citation25,Citation26 Other indications that require immediate reduction in IgM protein include moderate-to-severe hemolytic anemia, bulky lymphadenopathy, and symptomatic cryoglobulinemia.

Immunotherapy

Rituximab, a chimeric anti-CD20 monoclonal antibody, serves as a backbone of therapy directed against CD20+ WM cells.Citation27Citation29 The two important side effects include IgM flare and late-onset neutropenia (LON). IgM flare (defined as a transient increase in IgM levels by at least 25% from the baseline pretreatment levels) is generally seen in patients with serum IgM levels >4 g/dL. It is typically encountered in the first month of rituximab therapy but at times can persist for several months.Citation30 In patients with hyperviscosity symptoms and IgM >4 g/dL, preemptive plasmapheresis and avoidance of rituximab during the first 1–2 cycles is recommended.Citation31,Citation32 LON is a poorly understood complication of rituximab with a speculated correlation to FcRγIIIa-V158*F polymorphism secondary to profound antibody-dependent cell-mediated cytotoxicity activity and pronounced B-cell depletion associated with this polymorphism, causing neutrophil destruction by the release of granzyme and lysozyme.Citation33Citation36 Ofatumumab, a fully human anti-CD20 antibody, targets an epitope that is different from that of rituximab. It may be used in patients who are intolerant of rituximab, but higher cost and the lack of data suggesting superiority over rituximab restrict its use.Citation37,Citation38

Chemoimmunotherapy

Alkylator-based combination therapy has been considered highly effective in WM. Dexamethasone and rituximab in combination with cyclophosphamide (DRC) is a commonly prescribed regimen. Adriamycin and vincristine are avoided in WM owing to significant side effects without substantial additional benefit.Citation39 The DRC regimen is unsuitable for rapid control of symptoms as the median time to response is 4.1 months.Citation40 The 8-year OS rates in IPSS for WM low-, intermediate- and high-risk patients have been reported to be 100%, 55%, and 27%, respectively. The median progression- free survival (PFS) with DRC is 35 months (95% CI: 22–48 months). Patients who relapse after DRC are found to be still sensitive to rituximab. The regimen appears to be safe in both short and long terms.Citation40,Citation41

A subset analysis of a Phase III, open-label trial by Rummel et al, comparing bendamustine in combination with rituximab (BR) with rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP), showed a markedly improved median PFS with BR (69.5 [36.6–73] months vs 28.1 [17.8–51] months with R-CHOP). Both regimens led to an overall response rate (ORR) that approximated 95%. The patients who received BR tolerated therapy better, with lower rates of high-grade neutropenia, infectious complications, peripheral neuropathy, and absence of alopecia.Citation42 Of note, patients with severe functional defects of the organ dysfunction (NYHA III or IV, creatinine >2 mg/dL, aspartate aminotransferase/alanine aminotransferase/bilirubin >3× upper limit of normal) were excluded.Citation42 Due to the superior tolerability of BR, the Mayo Clinic group suggests using BR as the front-line option in patients with symptomatic WM.Citation7 Comparable results, with an ORR of 80% and 83%, were reported in two retrospective studies involving relapsed/refractory WM patients treated with BR.Citation43,Citation44 Prolonged cytopenias are frequent in patients receiving BR who have previously received nucleoside analog therapy.Citation44 Truncation of BR therapy from six to four cycles to avoid the prolonged cytopenias is not uncommon owing to comparable efficacy.Citation45 Reduction of the dose from 90 to 60–70 mg/m2 in the elderly and those with compromised renal function is also a common practice.Citation39

A survival benefit with nucleoside analog in WM was demonstrated in a multicenter Phase III randomized WM1 trial (trial comparing chlorambucil with fludarabine in patients with advanced WM). Oral fludarabine in comparison to chlorambucil resulted in a significantly longer PFS (36 vs 27 months; P=0.012) and OS (not reached vs 70 months; P=0.014), with a longer duration of response (38 vs 20 months; P<0.001).Citation46 In this study, myelodysplastic syndrome and acute myeloid leukemia (MDS/AML) developed in ~1.5% in the chlorambucil arm, but surprisingly, this complication was not observed in the fludarabine-treated patients.Citation46 Despite being highly effective, nucleoside analogs are less preferred now in the frontline setting owing to the risk of second malignancies.Citation39,Citation47 Because nucleoside analogs can also adversely impact stem-cell mobilization, they are best avoided as frontline therapies in the transplant-eligible patients. We typically reserve the use of nucleoside analogs for the relapsed–refractory population.

The proteasome inhibitors are among the most effective therapies in WM, particularly when combined with a steroid and rituximab.Citation48,Citation49 In the initial trial by the Waldenström’s Macroglobulinemia Clinical Trials Group (WMCTG) using this combination in 23 treatment-naive WM patients, the ORR was 96%, with a median time to response of 1.4 months. The rapid response prompted physicians to use this combination in patients with hyperviscosity syndromes.Citation50 Strikingly, despite substantial response, 60% of patients discontinued therapy after four cycles due to severe neuropathy. Investigators have attempted to reduce the incidence of treatment-emergent neuropathy by switching to weekly administration (three doses per cycle, D1,8,15) or using subcutaneous doses (NCT01788020). Preliminary data show that subcutaneous bortezomib in combination with rituximab and cyclophosphamide is quite effective.Citation51 Bortezomib-based therapy may be considered in patients with severe symptoms requiring rapid decrease in the monoclonal protein. Bortezomib is nonmyelotoxic and, therefore, can potentially be also used for maintenance therapy in patients without neuropathy, although clinicians should be vigilant about treatment-emergent neuropathy. An antiviral prophylaxis is required for the prevention of herpes zoster. A novel combination of carfilzomib, rituximab, and dexamethasone (CaRD) used as a neuropathy-sparing regimen in patients with WM led to 87% ORR and 36% VGPR when used as frontline therapy.Citation52 Other novel proteosome inhibitors that are being investigated include oprozomib (an oral epoxyketone irreversible proteasome inhibitor) and ixazomib (an oral boronic acid-based reversible proteasome inhibitor).Citation39,Citation53Citation55

Immunomodulatory drugs (IMiDs)

The use of IMiDs has been attempted in WM, given their impressive activity in MM. Thalidomide has been shown to be quite effective in WM.Citation56Citation59 Coleman et alCitation57 reported its use in patients previously treated with a purine analog or alkylating agent in combination with clarithromycin and dexamethasone. This combination was suggested as a salvage regimen in heavily pretreated patients.Citation59 Thalidomide in combination with rituximab led to an ORR of 72%.Citation58 The major disadvantage was the high incidence of neurotoxicity and poor tolerability by elderly patients.Citation56Citation59 Lenalidomide has been evaluated in Phase I and II trials in the setting of WM. Due to the dose limiting toxicity at 20 mg, a dose of 15 mg was used for 21 of 28 days. At a median follow-up of 36 months, the time to progression (TTP) was 16 months and the 5-year OS was 91%. Combination of lenalidomide and rituximab led to abrupt decrease in hemoglobin in 88% of the patients.Citation60 Investigators also evaluated the combination of pomalidomide with dexamethasone and rituximab (NCT01078974) in a dose-escalating Phase I study in seven patients, with a median time to response of 2.1 months and a median response duration of 15 months. A substantial proportion of patients (3/7) required plasmapheresis for IgM flare due to which the study was prematurely terminated. Given these issues, IMiDs have not been considered particularly attractive in WM.

Other agents

Everolimus, an inhibitor of mechanistic (formerly mammalian) target of rapamycin (mTOR) was studied in 60 relapsed/refractory patients with WM. The median time to remission was 2 months with a median PFS of 4 months. Major remission and partial remission were noted in 23% and 50% of the patients, respectively, with 23% developing grade 3/4 cytopenias.Citation61 The use of everolimus was also attempted in treatment-naive patients of WM with an ORR of 66.7% and major responses of 42.4%.Citation62 Severe adverse events included significant cytopenias, oral ulceration (prevented by dexamethasone swashes), and pulmonary toxicity. Everolimus has been tried in combination with bortezomib and rituximab (RVR), demonstrating a median PFS of 21 months.Citation63

Idelalisib targets the PI3K pathway, which is activated in patient with MYD88 mutation. de Rooij et al described the inhibition of WM cells’ proliferation by idelalisib. They also suggested that both idelalisib and ibrutinib dislodge the WM cells from the microenvironment into the circulation, which leads to the death of these cells in the absence of a supportive niche.Citation64 Recently, a Phase II study evaluating the safety and efficacy of idelalisib in patients with relapsed and/or refractory symptomatic WM was prematurely closed owing to the high incidence of hepatotoxicity.Citation65 Sildenafil, a phosphodiesterase inhibitor, was serendipitously found to be effective after an unusual response in the disease status of five WM patients following the prescription of sildenafil for erectile dysfunction.Citation66 summarizes the dosing schedules and outcomes of rituximab treatment as monotherapy and as combination therapy when combined with various abovementioned treatment regimens.

Table 5 Various regimens with rituximab in WM

Role of ibrutinib in the management of WM

Ibrutinib, an oral BTK inhibitor, acts by inhibiting downstream signaling of B-cell receptor pathway. By impairing crosstalk between MYD88 and BTK, it induces apoptosis of WM cells.Citation67 Additionally, it has a role in inhibiting hematopoietic cell kinase (HCK) as described below in detail. The important pharmacological aspects of ibrutinib and the various clinical trials conducted to date to evaluate its role in WM are tabulated in and , respectively.

Table 6 Aide memoirs on ibrutinib

Table 7 Studies using ibrutinib monotherapy in WM

Ibrutinib as monotherapy

The first Phase II multicenter clinical trial to show the efficacy of ibrutinib in relapsed–refractory WM (≥1 prior therapy) was NCT01614821 that led to this agent’s approval in WM in the US, Europe, and Canada. The population studied had received a median of two lines of therapy (range: 1–9 lines), with a median BM involvement of 60% (3–95%). IgM decreased rapidly as suggested by a median time to partial response of 8 weeks and the achievement of minimal responses as early as 4 weeks from the start of therapy.Citation68 The best clinical responses to ibrutinib at a median duration of treatment of 19 months (0.5–29.7 months) were an ORR of 91% and a major response rate of 73%. Complete response (CR) with ibrutinib mono-therapy was strikingly absent. The short follow-up showed a 2-year PFS and an OS of 69.1% and 95.2%, respectively.Citation68 Ibrutinib monotherapy has also been evaluated as a part of the iNNOVATE™ study in patients refractory to rituximab. The therapy was effective with a high ORR of 90% and an estimated 18-month OS of 97% and PFS of 86% (). The median time to best response was 2 months.Citation69 Adverse events included grade 1–2 thrombocytopenia (13%) and diarrhea (36%) and grade 3 neutropenia (10%) and hypertension (10%). A vast majority of serious adverse events were related to infections.Citation69

Effect of mutations on the efficacy of ibrutinib

CXCR4 mutations confer resistance to ibrutinib. In vitro studies of the CXCR4WHIM-engineered WM cells demonstrated that the presence of a mutation in the CXCR4 gene of the cells decreases their apoptosis following CXCL12 stimulation owing to the persistent activation of AKT and extracellular signal-regulated kinases.Citation70 These survival effects of CXCR4WHIM mutation are abrogated by the inhibition of simultaneous MYD88L265P signaling.Citation71 Treon et alCitation72 have demonstrated significant differences in response rates (ORR and major response rate) in patients with MYD88L265P/CXCR4WT, MYD88L265P/CXCR4WHIM, and MYD88WTmutations; the major response rate was 62% in patients with mutated MYD88 and CXCR4WHIM gene and 92% in those with mutated MYD88 and CXCR4WT gene, whereas no major response was observed in patients with MYD88WTCXCR4WT genotype. Of the arm C study population (n=31) of the iNNOVATE trial, 25 patients underwent mutation analysis, of which MYD88L265P/CXCR4WT mutation status and MYD88L265P/CXCR4WHIM mutation status were seen in 17 and 7 patients, respectively.Citation69 Though this study was not powered to assess the differences of treatment response with respect to mutation status, major response was seen in 82% of patients with MYD88L265P/CXCR4WT genotype and 71% of patients with MYD88L265P/CXCR4WHIM genotype.Citation69 IgM reduction was greater and achieved earlier in patients with MYD88L265P/CXCR4WT mutation status than in patients with MYD88L265P/CXCR4WHIM mutation status.Citation69 Therefore, for ibrutinib-treated patients with WM, the MYD88L265P mutation serves as a favorable prognostic marker, while the CXCR4 mutation is a predictive marker, and it is reasonable to check for the presence of MYD88L265P mutation prior to subjecting a patient to indefinite ibrutinib therapy, ie, until progression or intolerable side effects.Citation7

Adverse effects of ibrutinib

Adverse events include cytopenias, infections, arrhythmias, and bleeding (particularly epistaxis). Previously treated patients with WM were more likely to experience cytopenias with ibrutinib.

A tenfold increase in the risk of atrial fibrillation (AFib) in the ibrutinib arm was first noticed in the randomized Phase III open-label RESONATE study comparing ibrutinib and ofatumumab in 391 refractory CLL/SLL patients.Citation73 A meta-analysis to evaluate the risk of AFib in patients on ibrutinib vs the comparator drug/arm in random and fixed effects models showed a relative risk of 3.5 and 3.9, respectively. The rate of AFib on pooled analysis of the 20 studies was 3.3 per 100 person years.Citation74 Gustine et alCitation75 reported the cumulative risk of AFib of 5.4, 7.1, and 8.9% in 112 WM patients on ibrutinib at 1, 2, and 3 years, respectively. A past history of AFib leads to a shorter recurrence time of 3.9 vs 33.4 months in otherwise asymptomatic patients, and dose reduction and cardiologic intervention allowed the patients to continue therapy.Citation75 Ibrutinib should be avoided in patients with AFib on anticoagulation.Citation25 Animal experiments have suggested a role of ibrutinib in the inhibition of the cardiac PI3K–AKT signaling as the cause of the development of AFib.Citation76 Although a decrease in QTc interval is another potential cardiac side effect with ibrutinib, its significance is unclear. Ibrutinib also leads to platelet aggregation abnormalities and is best avoided in patients concurrently on other antiplatelet agents that have a potential to cause bleeding diathesis/platelet dysfunction.Citation77 This off-target effect was first observed in CLL trials. Acquired von Willebrand’s disease (aVWD) can be infrequently encountered in WM patients (~5%) due to elevated IgM levels.Citation78 Therefore, it is reasonable to check aVWD levels/activity prior to commencing ibrutinib in WM patients with a history of bleeding.Citation25

Experimental combinations of ibrutinib with novel agents

The aforementioned multicenter study (iNNOVATE; NCT02165397) began enrollment in July 2014 in 51 centers across the world to study the role of ibrutinib in combination with rituximab. This is primarily a randomized controlled trial wherein patients receive rituximab (4 weekly doses of 375 mg/m2 IV, with repeat 4 weekly doses after 3 months), with or without ibrutinib 420 mg daily until progression or unacceptable toxicity. WM patients who are treatment naive as well as those with documented progression and no response to their most recent treatment regimen were included in the study. Those patients who were refractory to rituximab-containing regimen were excluded from randomization but studied separately in an open-label arm with single-agent ibrutinib, the results of which were discussed earlier ().

IRAK1 is another protein downstream of BTK, which can contribute to the WM cell survival after exposure to ibrutinib. WM cells with MYD88 mutation show preferential IRAK1 rather than IRAK4 signaling. A combination therapy of ibrutinib with IRAK inhibitors can potentially augment the NFκβ blockade and theoretically overcome ibrutinib resistance.Citation79

Yang et al have demonstrated the overexpression of hemopoietic cell kinase (HCK) in WM cell lines with MYD88L265P mutation based on transcriptome profiling. The efficacy of ibrutinib in WM cell lines with MYD88L265P mutation was directly correlated to its binding to HCK, whereas mutated HCK blocked ibrutinib-related tumor cell killing. This study implicated the HCK pathway for ibrutinib resistance and a novel target in patients with MYD88 mutation.Citation80,Citation81

B-cell CLL/lymphoma 2 (BCL2) antagonist, venetoclax, is effective in WM with CXCR4WHIM mutation that is known to cause resistance to BTK and PI3K inhibitors. A study on the cell lines derived from WM patients with CXCR4WHIM mutations showed enhanced apoptosis with ibrutinib and idelalisib in the presence of venetoclax. Activity of venetoclax as a single agent has also been demonstrated in cell lines with CXCR4WT mutations and is postulated to be due to overexpression of BCL2 by the WM cells.Citation82 Aurora kinase inhibitors have also been proposed for therapy in patients exhibiting ibrutinib resistance.Citation83

Role of stem cell transplant in WM

Autologous stem cell transplant (ASCT) is a therapeutic consideration for the young patients with relapsed disease. Patients who are considered eligible for transplantation should avoid induction with drugs that are stem cell toxic.Citation7,Citation39 The European Bone Marrow Transplant Registry (EBMTR) data on 158 patients who underwent ASCT for WM suggest a 5-year OS of 69% and a PFS of 49%, with nonrelapse mortality (NRM) of 5.6%.Citation86 Patients undergoing transplant in the first remission had a significantly superior outcome as compared to those undergoing ASCT at a later time (5-year disease-free survival [DFS] of 50% vs 40%; P=0.004, and 5-year OS of 71% vs 63%; P=0.033).Citation84,Citation85

Allogeneic hematopoietic stem cell transplant (allo-SCT) has a very limited role, given the high transplant-related mortality, and we do not recommend this approach outside of clinical trials. Some experts suggest restricting its use primarily to young patients with multiple relapsed–refractory disease and particularly to those with early relapse post-ASCT. The reported ORR was 76%, the 5-year PFS was 56%, and the OS was 62% in a cohort of 86 patients. Twenty percent of patients achieved CR with allo-SCT. The 3-year NRM was 33% in patients receiving full myeloablative conditioning and 23% in patients receiving reduced-intensity conditioning.Citation86

Conclusion

With the introduction of molecular prognostic markers and improved understanding of the role of MYD88 and CXCR mutations in the pathophysiology of WM, the field appears to be rapidly evolving. The efficacy of multiple new agents including the second-generation BTK inhibitor (acalabrutinib), BCL2 inhibitors, IRAK inhibitors, and monoclonal antibodies, such as belimumab (anti-Blys) and ulocuplumab (anti-CXCR4), is being evaluated. Given the remarkable strides that have been made recently in our understanding of this rare malignancy, we suspect that the treatment options are bound to improve in the coming years, albeit at the cost of making its management increasingly complex.

Disclosure

Research funding has been provided to the institution from Celgene, Takeda and Amgen for studies, in which Prashant Kapoor is a principal investigator. The authors report no other conflicts of interest in this work.

References

  • WaldenströmJIncipient myelomatosis or essential hyperglobulinemia with fibrinogenopenia — a new syndrome?J Intern Med19441173–4216247
  • CastilloJJOlszewskiAJKananSMeidKHunterZRTreonSPOverall survival and competing risks of death in patients with Waldenstrom macroglobulinaemia: an analysis of the Surveillance, Epidemiology and End Results databaseBr J Haematol20151691818925521528
  • KristinssonSYElorantaSDickmanPWPatterns of survival in lymphoplasmacytic lymphoma/Waldenstrom macroglobulinemia: a population-based study of 1,555 patients diagnosed in Sweden from 1980 to 2005Am J Hematol2013881606523165980
  • SekharJSanfilippoKZhangQTrinkausKVijRMorgenszternDWaldenstrom macroglobulinemia: a Surveillance, Epidemiology, and End Results database review from 1988 to 2005Leuk Lymphoma20125381625162622239669
  • OlszewskiAJTreonSPCastilloJJEvolution of management and outcomes in Waldenstrom macroglobulinemia: a population-based analysisOncologist Epub2016729
  • KristinssonSYBjorkholmMGoldinLRMcMasterMLTuressonILandgrenORisk of lymphoproliferative disorders among first-degree relatives of lymphoplasmacytic lymphoma/Waldenstrom macroglobulinemia patients: a population-based study in SwedenBlood200811283052305618703425
  • KapoorPDiagnosis and management of Waldenstrom macroglobulinemia mayo stratification of macroglobulinemia and risk-adapted therapy (mSMART) guidelinesJAMA Oncol Epub201715
  • AnsellSMKyleRAReederCBDiagnosis and management of Waldenstrom macroglobulinemia: mayo stratification of macroglobulinemia and risk-adapted therapy (mSMART) guidelinesMayo Clin Proc201085982483320702770
  • LandgrenOTagejaNMYD88 and beyond: novel opportunities for diagnosis, prognosis and treatment in Waldenstrom’s macroglobulinemiaLeukemia20142891799180324573383
  • TreonSPXuLYangGMYD88 L265P somatic mutation in Waldenstrom’s macroglobulinemiaN Engl J Med2012367982683322931316
  • YangGZhouYLiuXA mutation in MYD88 (L265P) supports the survival of lymphoplasmacytic cells by activation of Bruton tyrosine kinase in Waldenstrom macroglobulinemiaBlood201312271222123223836557
  • TreonSPCaoYXuLYangGLiuXHunterZRSomatic mutations in MYD88 and CXCR4 are determinants of clinical presentation and overall survival in Waldenstrom macroglobulinemiaBlood2014123182791279624553177
  • MoriuchiMMoriuchiHTurnerWFauciASCloning and analysis of the promoter region of CXCR4, a coreceptor for HIV-1 entryJ Immunol19971599432243299379028
  • CaruzASamsomMAlonsoJMGenomic organization and promoter characterization of human CXCR4 geneFEBS Lett199842622712789599023
  • HunterZRXuLYangGThe genomic landscape of Waldenstrom macroglobulinemia is characterized by highly recurring MYD88 and WHIM-like CXCR4 mutations, and small somatic deletions associated with B-cell lymphomagenesisBlood2014123111637164624366360
  • XuLHunterZRTsakmaklisNClonal architecture of CXCR4 WHIM-like mutations in Waldenstrom macroglobulinaemiaBr J Haematol2016172573574426659815
  • HunterZRXuLYangGTranscriptome sequencing reveals a profile that corresponds to genomic variants in Waldenstrom macroglobulinemiaBlood2016128682783827301862
  • Garcia-SanzRMontotoSTorrequebradaAWaldenstrom macroglobulinaemia: presenting features and outcome in a series with 217 casesBr J Haematol2001115357558211736938
  • GhobrialIMAre you sure this is Waldenstrom macroglobulinemia?Hematology Am Soc Hematol Educ Program2012201258659423233639
  • KapoorPPaludoJVallumsetlaNGreippPRWaldenstrom macroglobulinemia: what a hematologist needs to knowBlood Rev201529530131925882617
  • KyleRATreonSPAlexanianRPrognostic markers and criteria to initiate therapy in Waldenstrom’s macroglobulinemia: consensus panel recommendations from the Second International Workshop on Waldenstrom’s macroglobulinemiaSemin Oncol200330211612012720119
  • GertzMAReederCBKyleRAAnsellSMStem cell transplant for Waldenstrom macroglobulinemia: an underutilized techniqueBone Marrow Transplant20124791147115321874060
  • LouwVJWebbMJPrognosis and treatment of Waldenstrom’s macroglobulinemiaTransfus Apher Sci201042219319720117052
  • JohnsonSABirchallJLuckieCOscierDGOwenRGGuidelines on the management of Waldenstrom macroglobulinaemiaBr J Haematol2006132668369716487169
  • LeblondVKastritisEAdvaniRTreatment recommendations for Waldenstrom macroglobulinemia from the Eighth International Workshop on WMBlood2016128101321132827432877
  • SzczepiorkowskiZMWintersJLBandarenkoNGuidelines on the use of therapeutic apheresis in clinical practice – evidence-based approach from the Apheresis Applications Committee of the American Society for apheresisJ Clin Apher20102538317720568098
  • TreonSPAgusDBLinkBCD20-directed antibody-mediated immunotherapy induces responses and facilitates hematologic recovery in patients with Waldenstrom’s macroglobulinemiaJ Immunother (1991)200124327227911395644
  • TreonSPAgusTBLinkBCD20-directed antibody-mediated immunotherapy induces responses and facilitates hematologic recovery in patients with Waldenstrom’s macroglobulinemiaJ Immunother2001243272279
  • GertzMARueMBloodEKaminerLSVesoleDHGreippPRMulticenter phase 2 trial of rituximab for Waldenstrom macroglobulinemia (WM): an Eastern Cooperative Oncology Group Study (E3A98)Leuk Lymphoma200445102047205515370249
  • TreonSPBranaganARHunterZSantosDTournhilacOAndersonKCParadoxical increases in serum IgM and viscosity levels following rituximab in Waldenstrom’s macroglobulinemiaAnn Oncol200415101481148315367407
  • TreonSPEmmanouilidesCKimbyEExtended rituximab therapy in Waldenstrom’s macroglobulinemiaAnn Oncol200516113213815598950
  • GhobrialIMFonsecaRGreippPRInitial immunoglobulin M ‘flare’ after rituximab therapy in patients diagnosed with Waldenstrom macroglobulinemia: an Eastern Cooperative Oncology Group StudyCancer2004101112593259815493038
  • LiSCChenYCEvensAMRituximab-induced late-onset neutropenia in newly diagnosed B-cell lymphoma correlates with Fc receptor FcgammaRIIIa 158(V/F) polymorphismAm J Hematol2010851081081220730791
  • WolachOBaireyOLahavMLate-onset neutropenia after rituximab treatment: case series and comprehensive review of the literatureMedicine (Baltimore)201089530831820827108
  • WolachOShpilbergOLahavMNeutropenia after rituximab treatment: new insights on a late complicationCurr Opin Hematol2012191323822080846
  • WengWKNegrinRSLavoriPHorningSJImmunoglobulin G Fc receptor FcgammaRIIIa 158 V/F polymorphism correlates with rituximab- induced neutropenia after autologous transplantation in patients with non-Hodgkin’s lymphomaJ Clin Oncol201028227928419933905
  • CastilloJJKananSMeidKManningRHunterZRTreonSPRituximab intolerance in patients with Waldenstrom macroglobulinaemiaBr J Haematol2016174464564826523929
  • FurmanRREradatHSwitzkyJCA phase II trial of ofatumumab in subjects with Waldenstrom’s macroglobulinemiaBlood2010116211795179520530284
  • TreonSPHow I treat Waldenstrom macroglobulinemiaBlood2015126672173226002963
  • DimopoulosMAAnagnostopoulosAKyrtsonisMCPrimary treatment of Waldenstrom macroglobulinemia with dexamethasone, rituximab, and cyclophosphamideJ Clin Oncol200725223344334917577016
  • KastritisEGavriatopoulouMKyrtsonisMCDexamethasone, rituximab, and cyclophosphamide as primary treatment of Waldenstrom macroglobulinemia: final analysis of a phase 2 studyBlood2015126111392139426359434
  • RummelMJNiederleNMaschmeyerGBendamustine 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 trialLancet201338198731203121023433739
  • TedeschiAPicardiPFerreroSBendamustine and rituximab combination is safe and effective as salvage regimen in Waldenstrom macroglobulinemiaLeuk Lymphoma20155692637264225651423
  • TreonSPHanzisCTripsasCBendamustine therapy in patients with relapsed or refractory Waldenstrom’s macroglobulinemiaClin Lymphoma Myeloma Leuk201111113313521454214
  • CastilloJJGustineJMeidKDubeauTGhobrialIMTreonSPLower doses of bendamustine are not associated with lower response rates in previously untreated patients with Waldenström macroglobulinemiaBlood20161282229692969
  • LeblondVJohnsonSChevretSResults of a randomized trial of chlorambucil versus fludarabine for patients with untreated Waldenstrom macroglobulinemia, marginal zone lymphoma, or lymphoplasmacytic lymphomaJ Clin Oncol201331330130723233721
  • DimopoulosMAKastritisEOwenRGTreatment recommendations for patients with Waldenstrom macroglobulinemia (WM) and related disorders: IWWM-7 consensusBlood201412491404141125027391
  • TreonSPProteasome inhibitors in Waldenstrom macroglobulinemiaBlood2013122193243324424203925
  • KapoorPAnother bidder (BDR) revisitsBlood2017129439840028126954
  • TreonSPIoakimidisLSoumeraiJDPrimary therapy of Waldenstrom macroglobulinemia with bortezomib, dexamethasone, and rituximab: WMCTG clinical trial 05-180J Clin Oncol200927233830383519506160
  • GhobrialIMXieWPadmanabhanSPhase II trial of weekly bortezomib in combination with rituximab in untreated patients with Waldenstrom macroglobulinemiaAm J Hematol201085967067420652865
  • TreonSPTripsasCKMeidKCarfilzomib, rituximab, and dexamethasone (CaRD) treatment offers a neuropathy-sparing approach for treating Waldenstrom’s macroglobulinemiaBlood2014124450351024859363
  • ChauhanDTianZZhouBIn vitro and in vivo selective antitumor activity of a novel orally bioavailable proteasome inhibitor MLN9708 against multiple myeloma cellsClin Cancer Res201117165311532121724551
  • RichardsonPGBazRWangMPhase 1 study of twice-weekly ixazomib, an oral proteasome inhibitor, in relapsed/refractory multiple myeloma patientsBlood201412471038104624920586
  • HurchlaMAGarcia-GomezAHornickMCThe epoxyketone-based proteasome inhibitors carfilzomib and orally bioavailable oprozomib have anti-resorptive and bone-anabolic activity in addition to anti-myeloma effectsLeukemia201327243044022763387
  • DimopoulosMAZomasAViniouNATreatment of Waldenstrom’s macroglobulinemia with thalidomideJ Clin Oncol200119163596360111504741
  • ColemanMLeonardJLyonsLSzelenyiHNiesvizkyRTreatment of Waldenstrom’s macroglobulinemia with clarithromycin, low-dose thalidomide, and dexamethasoneSemin Oncol200330227027412720151
  • TreonSPSoumeraiJDBranaganARThalidomide and rituximab in Waldenstrom macroglobulinemiaBlood2008112124452445718713945
  • DimopoulosMATsatalasCZomasATreatment of Waldenstrom’s macroglobulinemia with single-agent thalidomide or with the combination of clarithromycin, thalidomide and dexamethasoneSemin Oncol200330226526912720150
  • TreonSPSoumeraiJDBranaganARLenalidomide and rituximab in Waldenstrom’s macroglobulinemiaClin Cancer Res200915135536019118065
  • GhobrialIMWitzigTEGertzMLong-term results of the phase II trial of the oral mTOR inhibitor everolimus (RAD001) in relapsed or refractory Waldenstrom macroglobulinemiaAm J Hematol201489323724224716234
  • TreonSPTripsasCKIoakimidisLProspective, multicenter study of the MTOR inhibitor everolimus (RAD001) as primary therapy in Waldenstrom’s macroglobulinemiaBlood2011118212951295121734234
  • GhobrialIMReddRArmandPPhase I/II trial of everolimus in combination with bortezomib and rituximab (RVR) in relapsed/refractory Waldenstrom macroglobulinemiaLeukemia201529122338234626139427
  • de RooijMFMKuilAKraanWIbrutinib and idelalisib target B cell receptor- but not CXCL12/CXCR4-controlled integrin-mediated adhesion in Waldenström macroglobulinemiaHaematologica20161013e111e11526635033
  • CastilloJJGustineJNMeidKIdelalisib in Waldenstrom macroglobulinemia: high incidence of hepatotoxicityLeuk Lymphoma20175841002100427562445
  • TreonSPTournilhacOBranaganARClinical responses to sildenafil in Waldenstrom’s macroglobulinemiaClin Lymphoma20045320520715636699
  • PalsSTKerstenMJSpaargarenMTargeting cell adhesion and homing as strategy to cure Waldenström’s macroglobulinemiaBest Pract Res Clin Haematol201629216116827825462
  • TreonSPTripsasCKMeidKIbrutinib in previously treated Waldenstrom’s macroglobulinemiaN Engl J Med2015372151430144025853747
  • DimopoulosMATrotmanJTedeschiAIbrutinib for patients with rituximab-refractory Waldenström’s macroglobulinaemia (iNNOVATE): an open-label substudy of an international, multicentre, phase 3 trialLancet Oncol Epub2016129
  • CaoYHunterZLiuXThe WHIM-like CXCR4S338X somatic mutation activates AKT and ERK, and promotes resistance to ibrutinib and other agents used in the treatment of Waldenstrom’s macroglobulinemiaLeukemia201529116917624912431
  • CaoYHunterZRLiuXCXCR4 WHIM-like frameshift and nonsense mutations promote ibrutinib resistance but do not supplant MYD88L265P-directed survival signalling in Waldenström macroglobulinaemia cellsBr J Haematol2015168570170725371371
  • TreonSPXuLHunterZMYD88 mutations and response to ibrutinib in Waldenstrom’s macroglobulinemiaN Engl J Med20153736584586
  • ByrdJCBrownJRO’BrienSIbrutinib versus ofatumumab in previously treated chronic lymphoid leukemiaN Engl J Med2014371321322324881631
  • LeongDPCaronFHillisCThe risk of atrial fibrillation with ibrutinib use: a systematic review and meta-analysisBlood2016128113814027247135
  • GustineJNMeidKDubeauTETreonSPCastilloJJAtrial fibrillation associated with ibrutinib in Waldenstrom macroglobulinemiaAm J Hematol2016916E312E31326994323
  • McMullenJRBoeyEJOoiJYSeymourJFKeatingMJTamCSIbrutinib increases the risk of atrial fibrillation, potentially through inhibition of cardiac PI3K-Akt signalingBlood2014124253829383025498454
  • LevadeMDavidEGarciaCIbrutinib treatment affects collagen and von Willebrand factor-dependent platelet functionsBlood2014124263991399525305202
  • HivertBCaronCPetitSClinical and prognostic implications of low or high level of von Willebrand factor in patients with Waldenstrom macroglobulinemiaBlood2012120163214322122896002
  • YangGLiuXChenJTargeting IRAK1/IRAK4 signaling in Waldenstrom’s macroglobulinemiaBlood20151262340044004
  • YangGBuhrlageSJTanLHCK is a survival determinant trans-activated by mutated MYD88, and a direct target of ibrutinibBlood2016127253237325227143257
  • YangGBuhrlageSTanLHCK is a highly relevant target of ibrutinib in MYD88 mutated Waldenstrom’s macroglobulinemia and diffuse large B-cell lymphomaBlood201512623705705
  • CaoYYangGHunterZRThe BCL2 antagonist ABT-199 triggers apoptosis, and augments ibrutinib and idelalisib mediated cytotoxicity in CXCR4Wild-type and CXCR4WHIM mutated Waldenstrom macroglobulinaemia cellsBr J Haematol2015170113413825582069
  • PaulusAChittaKSAkhtarSAurora kinase is a therapeutic target in ibrutinib-resistant Waldenstrom macroglobulinemia: in-silico target identification and in-vitro validationBlood20151262327542754
  • KyriakouCCanalsCSibonDHigh-dose therapy and autologous stem-cell transplantation in Waldenstrom macroglobulinemia: the Lymphoma Working Party of the European Group for Blood and Marrow TransplantationJ Clin Oncol201028132227223220368570
  • KyriakouCBoumendilAFinelHAutologous stem cell transplantation (ASCT) for the treatment of patients with Waldenstrom’s macroglobulinemia/lymphoplasmacytic lymphoma (WM/LPL). A risk factor analysis by the European Society for Blood and Marrow Transplantation (EBMT) Lymphoma Working PartyBlood20141242167867825221805
  • KyriakouCCanalsCCornelissenJJAllogeneic stem-cell transplantation in patients with Waldenstrom macroglobulinemia: report from the Lymphoma Working Party of the European Group for Blood and Marrow TransplantationJ Clin Oncol201028334926493420956626
  • GertzMAKyleRAHyperviscosity syndromeJ Intensive Care Med199510312814110155178
  • LetendreLKyleRAMonoclonal cryoglobulinemia with high thermal insolubilityMayo Clin Proc198257106296337121069
  • MichaelABLawesMKamalarajanMHuissoonAPrattGCryoglobulinaemia as an acute presentation of Waldenstrom’s macroglobulinaemiaBr J Haematol2004124556514871241
  • Nobile-OrazioEMarmiroliPBaldiniLPeripheral neuropathy in macroglobulinemia: incidence and antigen-specificity of M proteinsNeurology1987379150615142442666
  • AdvaniRHBuggyJJSharmanJPBruton tyrosine kinase inhibitor ibrutinib (PCI-32765) has significant activity in patients with relapsed/refractory B-cell malignanciesJ Clin Oncol2013311889423045577
  • FurmanRBilottiEGraefTSingle-agent ibrutinib demonstrates long-term activity and safety in patients with relapsed/refractory Waldenstrom’s macroglobulinemiaPaper presented at: HaematologicaVienna, Austria2015