CC BY-NC-ND 4.0 · Thromb Haemost 2022; 122(11): 1843-1857
DOI: 10.1055/a-1896-7092
Review Article

Bleeding Propensity in Waldenström Macroglobulinemia: Potential Causes and Evaluation

1   Australian Cancer Research Foundation, Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
,
2   Department of Haematology, Canberra Hospital, Canberra, Australian Capital Territory, Australia
,
2   Department of Haematology, Canberra Hospital, Canberra, Australian Capital Territory, Australia
3   College of Health and Medicine, The Australian National University, Canberra, Australian Capital Territory, Australia
,
1   Australian Cancer Research Foundation, Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
› Author Affiliations
 

Abstract

Waldenström macroglobulinemia (WM) is a rare, incurable, low-grade, B cell lymphoma. Symptomatic disease commonly results from marrow or organ infiltration and hyperviscosity secondary to immunoglobulin M paraprotein, manifesting as anemia, bleeding and neurological symptoms among others. The causes of the bleeding phenotype in WM are complex and involve several intersecting mechanisms. Evidence of defects in platelet function is lacking in the literature, but factors impacting platelet function and coagulation pathways such as acquired von Willebrand factor syndrome, hyperviscosity, abnormal hematopoiesis, cryoglobulinemia and amyloidosis may contribute to bleeding. Understanding the pathophysiological mechanisms behind bleeding is important, as common WM therapies, including chemo-immunotherapy and Bruton's tyrosine kinase inhibitors, carry attendant bleeding risks. Furthermore, due to the relatively indolent nature of this lymphoma, most patients diagnosed with WM are often older and have one or more comorbidities, requiring treatment with anticoagulant or antiplatelet drugs. It is thus important to understand the origin of the WM bleeding phenotype, to better stratify patients according to their bleeding risk, and enhance confidence in clinical decisions regarding treatment management. In this review, we detail the evidence for various contributing factors to the bleeding phenotype in WM and focus on current and emerging diagnostic tools that will aid evaluation and management of bleeding in these patients.


#

Introduction

Waldenström macroglobulinemia (WM) is the clinical manifestation of lymphoplasmacytic lymphoma, which is a rare, low-grade, B cell lymphoma. WM is characterized by bone marrow infiltration with malignant cells and hypersecretion of immunoglobulin (Ig) M paraprotein. It constitutes less than 5% of all non-Hodgkin lymphomas, with an incidence of approximately 0.3/100,000 cases/year.[1] Many patients are asymptomatic at the time of their initial diagnosis and do not require treatment.[2] [3] However, approximately 30% of these patients are likely to need therapy within 2 years of diagnosis, and 80% within 10 years.[4] Indications for treatment are varied, but most commonly include constitutional symptoms, bone marrow or organ dysfunction, hyperviscosity and neuropathy. With an evolving treatment landscape in WM involving Bruton's tyrosine kinase (BTK) inhibitors, proteasome inhibitors and newer agents targeting BCL2 and CXCR4, understanding potential mechanisms of bleeding is important, as some of these therapies increase bleeding risk and necessitate the use of alternative agents in a personalized medicine approach.


#

Molecular Basis of WM

WM patients carry one or more somatic genetic mutations within malignant lymphoplasmacytoid cells ([Fig. 1]), which have been reported to be present in less mature lymphoid and hematopoietic progenitor cells in some cases.[5] Whether common WM mutations can be detected in megakaryocytes and platelets remains an open research question. Understanding the WM genomic landscape is important, not only because specific mutations can influence disease presentation and treatment options,[3] [6] but also because they can potentially affect platelet function (see below). The most common genetic defect in WM is a gain-of-function mutation within the myeloid differentiation factor (MYD) 88 gene resulting in a leucine-265 to proline (L265P) substitution within the cytoskeletal adaptor protein Myd88, detected in lymphoplasmacytoid cells in over 90% of WM patients.[7] [8] [9] [10] [11] [12] [13] Of note, the MYD88L265P transcript has also been detected in WM plasma cells, mature B lymphocytes, phenotypically normal B cell precursors and CD34+ hematopoietic precursor cells.[5] There are several other less common mutations now identified[14] [15] and at least two distinct WM signature DNA methylation profiles specific for memory B cells or plasma cells.[16]

Zoom Image
Fig. 1 Pathways of development of lymphoplasmacytoid B cells in healthy individuals and in WM patients. In WM patients, mutations and loss of DNA methylation occur to the lymphoplasmacytoid cells, with enrichment in B memory cells at an earlier stage of differentiation. The most common mutation, MYD88L265P, results in increased BTK phosphorylation and faster MYD88 complexing with IRAK1/4 in response to low or absent TLR or IL-1R stimulus, which results in increased NF-κB translocation to the nucleus, increased target gene transcription, uncontrolled proliferation of WM lymphoplasmacytoid cells and overproduction of IgM. BTK, Bruton's tyrosine kinase; IgM, immunoglobulin M; IL-1R, interleukin-1 receptor; IRAK, IL-1 receptor-associated kinase; MYD88, myeloid differentiation primary response 88; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; TLR, toll-like receptor; WM, Waldenström macroglobulinemia.

Through association with toll-like receptors (TLR) and the interleukin-1 receptor (IL-1R), Myd88 has an important role in coordinating innate immune cell responses. Cells expressing Myd88L265P protein exhibit constitutive activation of TLR and IL-1R pathways, leading to nuclear translocation of transcription factor nuclear factor (NF)-κB and enhanced B cell proliferation and survival.[17] [18] MYD88L265P has also been detected in approximately 61% of IgM monoclonal gammopathy of undetermined significance (IgM-MGUS) patients.[9] [10] [12] [19] This mutation is likely to be an early oncogenic event in WM development, with IgM-MGUS acting as a precursor condition. However, additional genetic mutations likely contribute to WM onset.[14] [20] Of note, Myd88 is expressed in platelets and megakaryocytes and is essential for appropriate TLR-driven platelet responses to viremia.[21] A link between Myd88 activation and platelet function will be discussed below.

Gain-of-function mutations in the gene encoding the C-X-C chemokine receptor (CXCR) 4 also occur in approximately 30% of WM patients.[22] [23] [24] CXCR4 engages with stromal-derived factor (SDF) 1 to mediate the homing of cells to the bone marrow. A common activating mutation involving serine-338 in the C-terminal region of CXCR4 prevents CXCR4 internalization following SDF-1 stimulation.[25] This leads to persistent CXCR4 activation and signaling via AKT, ERK and BTK pathways, and bone marrow myeloid cell migration, adhesion, proliferation, and survival.[26] [27]

The mutations in WM lymphoplasmacytoid cells, within genes involved in cell proliferation and survival, cause an over-proliferation of these cells, resulting in overproduction of IgM ([Fig. 1]). Increased IgM contributes to the neurological and bleeding symptoms observed in WM, as correction of blood IgM levels often resolves symptoms.[24] WM malignant cell infiltration of the bone marrow leads to cytopenias, which increase bleeding risk and predispose to infections. While some of these mutations have been detected in lymphoid and hematopoietic precursors, it is not evident that the mutations arise in platelet-producing megakaryocytes or contribute to a bleeding phenotype. This review will explore the multifactorial nature of bleeding encountered in WM and explore considerations that may aid clinical decisions around therapy for these patients.


#

Waldenström Macroglobulinemia Patients Can Present with a Bleeding Phenotype

When Jan Waldenström first described WM in 1944, the symptoms in the two patients included oronasal bleeding.[28] While bleeding is often a feature of the initial presentation of WM, bleeding symptoms usually resolve after therapeutic intervention, implying that WM disease etiology does not directly affect platelet production and function. However, four studies have evaluated the frequency of bleeding in patients with WM before treatment intervention ([Table 1]). When considered together, approximately 17% of patients displayed bleeding symptoms,[145] [146] [2] [30] with limited description of magnitude. Most other studies describe easy bruising and chronic oronasal bleeding as a common symptom of WM. It is worth noting that due to a paucity of large studies on this issue, evidence-based guidelines and standard of care management for treatment of bleeding in these patients are lacking. Thus, reported treatment approaches are variable, with inconsistent reporting of key diagnostic information. Standardized reporting of laboratory findings and outcomes is also lacking.

Table 1

Studies reporting the frequency of bleeding in treatment-naïve WM patients

Study

Sample size

% Bleeding

Nature of bleeding

Perkins et al 1970[145]

62

36

Not specified

Merlini et al 2003[146]

215

7

Hemorrhagic manifestations

García-Sanz et al 2001[2]

217

23

Not specified

Merchionne et al 2011[30]

121

12.3

Epistaxis and gum bleeding

Abbreviation: WM, Waldenström macroglobulinemia.


Laboratory Test Abnormalities Associated with Bleeding in WM

The cause of the WM bleeding phenotype is unknown, but laboratory studies have identified several vascular and platelet abnormalities that often co-occur ([Table 2]). All of these sequelae are likely to contribute to bleeding symptoms in WM patients prior to initiation of treatment; however several of these findings could occur as a result of IgM protein binding to and/or inhibition of coagulation factor(s) function.

Table 2

Laboratory test abnormalities associated with bleeding in WM patients

Reference

Sample size

Finding

Comments and observations that may explain bleeding symptoms

Hivert et al 2012[133]

43/72 (59%)

Reduced VWF levels

- Possibly due to IgM-mediated inhibition.[48]

- Possibly due to specific autoantibody-mediated destruction, absorption onto malignant cells, altered blood rheology resulting in VWF multimer proteolysis by ADAMTS-13.[36]

- Results in reduced VWF-GPIb-IX-V-mediated platelet activation and bleeding.

Gavriatopoulou et al 2019[134]

Hivert et al 2012[133]

6/42 (14%)

10/72 (14%)

Increased VWF levels

- Correlated with poor prognosis and low circulating ADAMTS-13 levels.[134]

- Reflects greater engagement between WM lymphoplasmacytoid cells and endothelium.[133] [134]

- Could trigger thrombosis due to increased VWF-GPIb-IX-V-mediated platelet activation.

- Could result in increased bleeding due to low numbers of VWF multimers.[135]

Castillo et al 2019[38]

49/320 (15%)

Reduced FVIII levels

- FVIII is produced in the liver. Low FVIII levels possibly caused by liver dysfunction.[136]

- FVIII is bound to VWF in plasma. Low FVIII levels possibly coincide with AVWS.

- Possibly due to IgM-mediated inhibition of FVIII or VWF.[48] [49] [50]

- Results in decelerated FX activation and bleeding.[137]

Saraya et al 1972[138]

Kasturi et al 1978[139]

3/3 (100%)

4/4 (100%)

Reduced platelet adhesion

- Reduced in vivo platelet adhesion to a wound.[138]

- Results in bleeding

Kasturi et al 1978[139]

4/4 (100%)

Reduced platelet activation

- Platelets take up microparticles containing tissue factor. Platelet activation measured via release of tissue factor in response to ADP.[140] [141]

- Results in bleeding

Saraya et al 1972[138]

Kasturi et al 1978[139]

2/3 (67%)

3/4 (75%)

Reduced platelet aggregation

- Reduced aggregation in response to ADP and adrenaline. Within two standard deviations for noradrenaline, thrombin, and collagen.[138]

- Results in bleeding

Abbreviations: ADAMTS-13; a disintegrin and metalloprotease; ADP, adenosine diphosphate; aPTT; activated partial thromboplastin time; AVWS; acquired von Willebrand syndrome; FVIII, factor VIII; PT; prothrombin time; TF; tissue factor; VWF; von Willebrand factor.


Note: Sample size indicates number of patients and percentage of cohort with clinically significant bleeding.



#

Thrombocytopenia

Thrombocytopenia is a common occurrence in WM, often coincident with anemia. One large WM study (n = 454) classified 18% of WM patients with thrombocytopenia.[32] Mechanisms by which thrombocytopenia arises are likely to be complex, potentially resulting from combinations that are autoimmune- and drug-mediated, as well as marrow infiltration which can cause overcrowding of hematopoietic stem cells and progenitors, resulting in disturbed megakaryopoiesis and thrombopoiesis.[33] Occasionally thrombocytopenia is secondary to peripheral platelet sequestration within the spleen (splenomegaly).[34] In isolation, thrombocytopenia rarely explains the observed bleeding, and the degree of bleeding is often out of step with the platelet count.


#

Acquired von Willebrand Syndrome

Von Willebrand factor (VWF) is a biorheological shear sensitive glycoprotein (GP) produced by the vascular endothelium and megakaryocytes. VWF plays a vital role in primary hemostasis by triggering GPIb-IX-V-mediated platelet activation and formation of an adhesive bridge between platelets and the vasculature at sites of endothelial injury. VWF is also a carrier protein for factor VIII (FVIII) and contributes to fibrin clot formation.[35] Acquired von Willebrand syndrome (AVWS) is an uncommon disorder caused by a loss of high-molecular-weight VWF multimers, either by specific autoantibody-mediated destruction, absorption onto malignant cells, or increased fluid shear stress resulting in VWF multimer unfolding and proteolysis by ADAMTS-13.[36] In hematological cancer, AVWS is reported in lymphoproliferative neoplasms and several myeloproliferative disorders.[37] AVWS occurs in 6% of WM patients, where incidence is strongly correlated with elevated IgM levels (30–60 g/L).[38] Symptoms include mucosal and GI bleeding, which generally resolve following WM therapy.[38] To mitigate bleeding, specific therapeutic approaches aim to increase VWF antigen levels (treatment with desmopressin and/or transfusion of FVIII/VWF concentrate), remove an offending autoantibody (plasmapheresis), or disturb destructive autoantibody functions, via transfusion of intravenous immunoglobulin (IVIg).[39] [40] IVIg has been reported to successfully increase VWF/FVIII levels and reduce bleeding times in AVWS linked to IgGMGUS.[41] [42] Although the mechanism of action of IVIg is unclear, isolated case reports clearance of VWF in the setting of IgM-MGUS may also be ablated by IVIg therapy, implying that IVIg could be a favourable therapeutic option for AVWS associated with WM.[39] [41]


#

Hyperviscosity

Hyperviscosity, caused by the accumulation of large (approximately 925 kDa), pentameric, positively charged IgM paraprotein in the blood, is a classic manifestation of WM. The IgM proteins electrostatically interact with sialic acid-rich red blood cells, resulting in an agglutinating effect and contributing to increased viscosity.[43] Healthy individuals have around 1.5 g/L of IgM, of which 80% is intravascular,[44] and hyperviscosity emerges when IgM levels exceed 50 to 60 g/L.[44] Hyperviscosity causes the physical tearing of small venules from increased rheological drag,[45] and the suspected inhibitory coating of platelets by IgM protein, resulting in reduced platelet adhesion and aggregation.[46] Symptoms include bleeding, vision problems, and neurological symptoms, which occur in 13% of WM patients.[47] Acute management of hyperviscosity involves plasmapheresis, while longer term management with chemo-immunotherapy or targeted agents works by depleting the IgM-producing cells in the marrow.


#

Hemostasis-Inhibiting Paraproteins

Circulating paraproteins have been reported to have VWF and FVIII-inhibitory activity in WM in vivo, caused by IgM[48] [49] [50] or IgG[51] [52] antibodies. This has also been observed in other paraproteinemias, including multiple myeloma, MGUS, lymphoma, chronic lymphocytic leukemia, and amyloidosis.[53] [54] Reports showed that the monoclonal IgM isolated from a WM patient demonstrated antiplatelet activity and immune thrombocytopenia in vivo,[55] [56] implying a derangement causing autoimmunity. Additionally, WM IgM cryoglobulins can suppress erythroid and granulocyte progenitor cells grown in culture in vitro,[57] and possibly megakaryocyte progenitor cell maturation, which could alter platelet quality and function.


#

Cryoglobulinemia

Cryoglobulinemia, where temperature-sensitive Igs form concentration-dependent insoluble aggregates that precipitate below 37°C, can occur in WM.[58] These cryoglobulins may form immune complexes, where monoclonal IgM antibodies bind to the Fc region of polyclonal IgG antibodies.[44] The symptoms of cryoglobulinemia occur at varied cryoglobulin concentrations depending on the individual. These include purpura and mucosal bleeding, caused by the tearing of small blood vessels by the aggregates, and are observed in approximately 5% of WM patients.[44]


#

Amyloidosis

Amyloidosis can be associated with potentially life-threatening hemorrhage, by causing coagulation factor deficiency, hyperfibrinolysis, platelet dysfunction, angiopathy, and/or vascular fragility.[59] Amyloidosis is characterized by the production of misfolded proteins, often monoclonal Ig light and/or heavy chains, which form insoluble amyloid fibrils, that accumulate and form plaques, leading to tissue and organ dysfunction. IgM amyloidosis occurs in 7.5% of WM patients and is associated with a dramatic reduction in overall survival, from 12.1 to 2.5 years.[60] Bleeding occurs in 5 to 41% of amyloidosis patients, ranging from ecchymoses and purpura, to gastrointestinal (GI) and postprocedural bleeding.[61] [62] [63] [64] [65] Bleeding results from increased vessel-wall fragility from perivascular amyloid deposition, and/or from acquired factor X (FX) deficiencies. Acquired FX deficiencies occur in 5 to 10% of amyloidosis patients and are caused by the absorption of FX and pentraxin-2 onto amyloid fibrils, particularly in the spleen, resulting in direct FX removal from circulation and indirect FX internalization by macrophages.[66] This can be corrected by splenectomy, chemotherapy treatment, or autologous hematopoietic cell transplant.[66]

To summarize, WM patients display significantly elevated concentrations of serum IgM and one or more symptoms of blood hyperviscosity, cryoglobulinemia, coagulation irregularities, thrombocytopenia, amyloidosis and bleeding. The factor(s) responsible for bleeding in untreated WM remains to be fully defined, but bleeding is likely to be the result of a combination of all of the above observations.


#
#

Platelet Dysfunction in WM

Platelets circulate throughout the vasculature and are the primary mediators of hemostasis ([Fig. 2]). These are produced in the bone marrow by megakaryocytes, via controlled endomitosis. A healthy individual generally has a very stable platelet count; however, the numbers of circulating platelets can range from 150 to 400 × 109 platelets/L. Thrombocytopenic individuals (<100 × 109 platelets/L) can have a heightened risk of bleeding, which significantly increases if the platelet count falls below 20 × 109 platelets/L. However, as mentioned earlier, bleeding can also occur in the absence of thrombocytopenia. Thus, the prediction of an imminent bleeding event should not rely solely on a low platelet count.[67] [68] [69] Besides the platelet count, platelet quality and functionality are critical components of an effective hemostatic response. This involves detection of injury-exposed collagen and other matrix proteins, and sensing alterations to local blood rheology. Platelets respond by adhering to a site of vascular injury and undergoing platelet activation. Platelet surface receptors coordinate this response[70] as well as the subsequent formation of a thrombus (platelet aggregate or blood clot), which acts to seal the blood vessel, reduce blood loss and begin the process of wound repair ([Fig. 2]).[71] [72]

Zoom Image
Fig. 2 Simplified hemostasis. (A) Platelet plug formation. Following an injury, GPIb-IX-V and GPVI bind exposed extracellular matrix ligands such as von Willebrand factor (vWF) and collagen respectively to enable platelet adhesion to the endothelium. Engagement of these receptors triggers platelet signaling, and platelets undergo changes to the cytoskeleton and release granules. The platelet-specific integrin, αIIbβ3, becomes active and binds fibrinogen, bridging adjacent platelets. Platelets aggregate forming a platelet plug at the site of the injury. (B) Blood coagulation and the securing of the platelet plug. Tissue factor (TF) is exposed at the site of the injury triggering the extrinsic coagulation cascade, resulting in thrombin production. Thrombin converts fibrinogen into insoluble fibrin which secures the platelet plug in place. Blood hyperviscosity and high levels of IgM are likely to interfere with these hemostatic pathways, and potentially underpin bleeding events. GPVI, glycoprotein VI; IgM, immunoglobulin M.

Platelet Function Is Controlled by Surface Receptors and Signaling Pathway Proteins

Levels of platelet receptors and their attendant surface densities mediate platelet responsiveness to molecular cues in the vasculature. Low receptor numbers and densities have been associated with bleeding in patients receiving mechanocirculatory support[73] and in trauma patients.[74] Further, a loss of platelet receptors, including GPIbα and GPVI,[75] and a diminution of platelet function prior to therapy[76] have been demonstrated in leukemia patients. The molecular explanation for these losses remains undefined, but may be linked to disturbances in bone marrow cellularity and megakaryocyte maturation.[77] As GPIb-IX-V and the collagen/fibrin receptor GPVI also contribute to efficient thrombus generation by binding thrombin and other coagulation proteins,[78] [79] [80] [81] any alteration to normal levels of platelet adhesion receptors due to changes in megakaryocyte maturity could disrupt efficient thrombin generation at the platelet surface.

Bleeding has been observed in WM patients who do not have hyperviscosity, thrombocytopenia, cryoglobulinemia or AVWS,[30] [82] implying that there are other potential causes of bleeding in WM. It is possible that many WM patients do not display chronic bleeding symptoms but possess an underlying platelet lesion. When these patients experience vascular and hemostatic challenges such as surgery or trauma, or when the platelet lesion is present in combination with one or more common bleeding causes as outlined above, the platelet lesion can become more evident and unexplained bleeding complications ensue.[83] [84] Studies specifically evaluating platelet function in WM would help identify patients with reduced hemostatic capacity and enhanced bleeding risk, and this information could aid in clinical decisions regarding therapeutic approaches.


#
#

Standard WM Therapies May Accentuate the Bleeding Phenotype

WM is an incurable disease where the treatments aim to alleviate the symptoms and achieve prolonged remissions.[1] [2] [3] Treatment decisions in this regard are generally based on the symptoms, diagnostic laboratory profile and the availability of drugs and clinical trials.[85] For asymptomatic patients, a “wait and watch” approach is routinely implemented.[3] Options for treatment of symptomatic WM patients include alkylating chemotherapy agents (bendamustine, cyclophosphamide), proteosome inhibitors (bortezomib, carfilzomib, ixazomib), or the first-generation BTK inhibitor (ibrutinib), alone or in combination with rituximab. Newer therapeutic options include new, irreversible and more selective BTK inhibitors (acalabrutinib, zanubrutinib) administered as a monotherapy, and emerging options include BCL2 antagonists (venetoclax).[85] Many of these treatments carry an attendant bleeding risk which can be enhanced in WM patients and will be discussed in this context below.

Rituximab

Rituximab is an anti-CD20 monoclonal antibody that specifically causes B cell depletion and thus acts to reduce IgM production.[86] Although rituximab is commonly used in combination with chemotherapy agents, if used as monotherapy, it can be associated with >25% rises in IgM (an IgM flare), which can exacerbate hyperviscosity-related bleeding symptoms.[87] This risk is reduced when used in combination with other drugs. Alternatively, rituximab can be associated with acute thrombocytopenia, linked with cytokine release syndrome and complement activation.[88]


#

Alkylating Chemotherapies

Alkylating chemotherapies, such as bendamustine and cyclophosphamide, have been used effectively as a frontline therapy in WM to kill rapidly dividing cells.[89] However, hematopoietic stem cells and their progenitor lineages are also sensitive to these therapies, resulting in cytopenias, particularly thrombocytopenia, and increasing the risk of bleeding.


#

BTK Inhibitors

The B cell receptor (BCR) signaling pathway is a central determinant of B cell fate and function. This pathway is activated in WM, particularly in patients bearing the Myd88 mutation.[14] When an antigen binds to the BCR, this triggers BCR clustering and initiation of signal transduction via phosphorylation of BCR cytoplasmic tyrosine-based activation motifs (ITAMs). ITAM clustering enables recruitment of Src-family kinases, which serve to phosphorylate Syk and activate phosphoinositide 3-kinase (PI3K) δ. PI3Kδ mediates the conversion of phosphatidylinositol 4,5-bisphosphate to phosphatidylinositol 3,4,5-triphosphate, which engages BTK. BTK phosphorylates phospholipase C (PLC) γ2, activating NF-κB, NF of activated T cells, and mitogen-activated protein (MAP) kinase pathways ([Fig. 3]). These are all key elements of survival, development and cell proliferation pathways.

Zoom Image
Fig. 3 Platelets and B lymphocytes utilize common signaling pathways. BTK is differentially involved in the downstream signaling pathways triggered by ligand engagement of the major platelet adhesion/signaling receptors (αIIbβ3, GPVI, and GPIbα of the GPIb-IX-V complex) and the BCR. In receptors with ITAMs, following ligand binding and receptor clustering, phosphorylation of the cytoplasmic ITAMs and recruitment of SFKs ensue, resulting in phosphorylation of Syk and activation of PI3K. PI3K mediates the conversion of phosphatidylinositol 4,5-bisphosphate to phosphatidylinositol 3,4,5-triphosphate, which engages BTK, resulting in phosphorylation of PLCγ. In platelets, this leads to activation and aggregation. In B cells, activation of MAPKs, NF-κB, and NFAT leads to B cell proliferation, development, and survival. BCR and GPVI ITAM signaling are more reliant on the BTK pathway compared with GPIbα and αIIbβ3, thus signaling downstream of these receptors is more sensitive to BTK inhibition. BCR, B cell receptor; BTK, Bruton's tyrosine kinase; GP, glycoprotein; ITAM, immunoreceptor tyrosine-based activation motif; MAPK, mitogen-activated protein kinase; NF-κB, nuclear factor κB; NFAT, nuclear factor of activated T cells ; PI3K, phosphoinositide 3-kinase; PLCγ, phospholipase C γ; SFK, Src-family kinases.

The clinical use of BTK inhibitors for the treatment of B cell malignancies has grown remarkably, resulting in improved outcomes. At present, three different covalent irreversible BTK inhibitors are approved for clinical use. These inhibitors (ibrutinib, zanubrutinib and acalabrutinib) all bind cysteine 481 within the ATP-binding pocket of BTK with different avidities and selectivities, and serve to inhibit the phosphorylation of downstream kinases in the BCR signaling pathway, blocking B cell activation. As many as 20 new BTK inhibitors are under development.[90] Nonetheless, bleeding remains a significant side effect that is associated with the use of these therapies.[91]

Platelet activation requires intra-platelet signaling, triggered by platelet receptor–ligand interactions, leading to enhanced platelet aggregation. Several of these pathways involve activation of BTK[92] [93] ([Fig. 3]), which is important in receptor signaling. Importantly there is redundancy in this pathway, as studies of genetically engineered mice or patients with X-linked agammaglobulinemia with loss-of-function mutations to the BTK gene did not reveal any bleeding propensity, most likely due to compensatory signaling by Tec and other kinases.[94] BTK inhibitors interfere with BTK activity by irreversibly and covalently binding to the kinase domain,[95] preventing autophosphorylation of BTK and phosphorylation of PLCγ2 and MAP kinases. Ibrutinib is less specific for BTK and can inhibit other tyrosine kinases such as TEC (tyrosine kinase expressed in hepatocellular carcinoma), ITK (interleukin-2-inducible T cell kinase), TXK (T and X cell expressed kinase), and EGFR (epidermal growth factor receptor), while the second-generation therapeutics zanubrutinib and acalabrutinib are more specific to BTK ([Table 3]). As BTK is critical for BCR signaling, there is a level of specificity achieved by targeting BTK. However, because of the broad kinase target spectrum of most tyrosine kinase inhibitors, they inevitably have off-target adverse events ([Table 3]). Platelets rely on tyrosine kinase activity for their activation. BTK is involved in platelet signaling pathways, mediated by GPVI, CLEC-2, GPIb, and αIIbβ3, that enable platelet adhesion in flowing blood.[96] Platelet function has been shown to be inhibited in patients being treated with ibrutinib,[97] [98] as well as several other tyrosine kinase inhibitors.[99] Platelets from ibrutinib- but not zanubrutinib-treated patients showed reduced levels of GPIb-IX-V and αIIbβ3 and an ablation of platelet aggregate formation.[100]

Table 3

Inhibited kinases and adverse events associated with various Bruton's tyrosine kinase inhibitors

BTK inhibitor

Number of studies evaluated[142]

Total number of patients[142]

Type of hematological malignancies included[142]

Inhibited kinases:

IC50 ± SD (nM)[143]

% Adverse events:

grade 1–2 (≥ grade 3)[142]

Ibrutinib

12

1,263

TN + R/R CLL/SLL with P53 aberrations or del(17)p

TN + pretreated WM

R/R MCL

Pretreated GvHD

BTK: 1.5 ± 0.2

TEC: 10 ± 12

ITK: 4.9 ± 1.2

TXK: 2.0 ± 0.3

EGFR: 5.3 ± 1.3

Bleeding: 35 (4.4)[144]

AF: 12.4 (5.9)

Hypertension: 19.6 (15.6)

Rash: 15.8 (1.1)

Diarrhea: 47.3 (3.8)

Acalabrutinib

9

937

TN + R/R + ibrutinib-intolerant CLL/SLL

TN + R/R WM

R/R MCL

BTK: 5.1 ± 1.0

TEC: 126 ± 11

ITK: >1,000

TXK: 368 ± 141

EGFR: >1,000

Bleeding: 40.7 (2.5)

AF: 4.3 (1.1)

Hypertension: 8.6 (3.9)

Rash: 16.1 (0.3)

Diarrhea: 37.7 (1.9)

Zanubrutinib

6

756

TN + R/R CLL/SLL with del(17)p

TN + R/R + symptomatic WM

R/R MCL

BTK: 0.5 ± 0.0

TEC: 44 ± 19

ITK: 50 ± 5

TXK: 2.2 ± 0.6

EGFR: 21 ± 1

Bleeding: 39.2 (3.9)

AF: 2.0 (0.7)

Hypertension: 10.9 (2.8)

Rash: 17.7 (0)

Diarrhea: 20.8 (1.4)

Tirabrutinib

5

178

R/R CLL

TN + R/R WM

R/R MCL

R/R B cell malignancies or NHLs

BTK: 5.6 ± 1.0

TEC: 77 ± 7

ITK: >1,000

TXK: 116 ± 35

EGFR: >1,000

Bleeding: 17.8 (1.8)

AF: 0 (0)

Hypertension: 0 (0)

Rash: 24.7 (3.7)

Diarrhea: 19.8 (1.1)

Abbreviations: AF, atrial fibrillation; BTK, Bruton's tyrosine kinase; CLL, chronic lymphocytic leukemia; EGFR, epidermal growth factor receptor; GvHD, graft-versus-host disease; ITK, interleukin-2-inducible T cell kinase; MCL, mantle cell lymphoma; NHL, non-Hodgkin lymphoma; R/R, relapsed/refractory; SD, standard deviation; SLL, small lymphocytic lymphoma; TEC, tyrosine kinase expressed in hepatocellular carcinoma; TN, treatment-naïve; TXK, T and X cell expressed kinase; WM, Waldenström mcroglobulinemia.


Consistent with an off-target effect of BTK inhibitors on platelets, a meta-analysis by Brown and colleagues found that approximately 40% of patients with B cell malignancies receiving ibrutinib experienced bleeding, with 4.4% experiencing major hemorrhage (> grade III) ([Table 3]).[101] Reports of bleeding in WM patients as a result of ibrutinib treatment were slightly lower than calculated for all B cell malignancy patients,[96] where approximately 23% of WM patients on ibrutinib experienced bleeding ([Table 4]). While differences in study design and definition of what constitutes a major or minor bleed may account for some of the disparity, it is likely that differences in disease pathogenesis may also contribute to incidence of bleeding across these different malignancies. Ibrutinib discontinuation reverses major toxicities observed in WM patients (bleeding, GI toxicity), despite causing IgM rebound in 73% and withdrawal symptoms (fever, body aches, night sweats, arthralgia, headaches) in 19%.[102] [103]

Table 4

Reports of bleeding frequency in Waldenström macroglobulinemia patients treated with ibrutinib

Reference

Sample size

Number of WM patients

Study type

n, minor bleeding

(% grade 1–2)

n, major bleeding (% grade 3–5)

Thrombocytopenia

Bleeding description

Abeykoon et al 2020

80

80 (100%)

Nonclinical trial (84% previously treated, 16% treatment naïve)

3 (4%) hemorrhage,

1 (1%) petechiae

2 (3%) hematuria

1 (1%)

8 (10%)

Unspecified hemorrhage, petechiae, hematuria; intracranial hemorrhage due to CNS involvement

Ali et al 2017

45

8 (18%)

Retrospective observational cohort analysis

Previously treated

14 (30.5%)

0 (0%)

NA

Bruising, epistaxis; gastrointestinal (rectal) bleeding

Dimopoulos et al 2017

31

31 (100%)

Previously treated

12 (39%) bleeding

7 (23%) bruising

0 (0%)

5 (16%)

Grade 1–2 bleed, bruising

Dimopoulos et al 2018

75

75 (100%)

Phase 3 trial of ibrutinib plus rituximab

51% in ibrutinib + rituximab cf. 31% placebo + rituximab

4% incidence in each arm

0 (0%)

Unspecified low-grade bleeding; unspecified major hemorrhage

Treon et al 2018

30

30 (100%)

Treatment-naïve

2 (7%) bruising,

1 (3) postprocedural hemorrhage

1 (3%)

1 (3%)

Bruising, postprocedural hemorrhage; rectal bleeding

Treon et al 2015

63

63 (100%)

Previously treated

2 (3%)

0 (0%)

9 (14%)

Epistaxis, postprocedural hemorrhage

Abbreviations: CNS, central nervous system.


Note: n: number of individuals; minor bleeding consisted of grade 1–2 bleeds; major bleeding comprised grade 3–5 bleeds.



#

Antiplatelet and Anticoagulant Therapies

WM affects an older demographic, many of whom have existing comorbidities. Treatments of these comorbidities can include antiplatelet or anticoagulant drugs, such as aspirin, clopidogrel, warfarin or direct oral anticoagulants, which carry an attendant bleeding risk. Treatment of WM patients with antiplatelet or anticoagulant therapies in combination with ibrutinib is common because of a significantly increased risk (approximately 10%) of atrial fibrillation with ibrutinib.[24] [104] [105] [106] [107] [108] [109] [110] The use of these therapeutics concomitantly elevates the bleeding risk. In one study of B cell malignancy patients, major bleeding occurred in 3.7% of patients receiving ibrutinib monotherapy compared with 5.1% receiving ibrutinib in combination with antiplatelet reagents or anticoagulants.[101]

Taken together, WM treatments increase bleeding risk, and treatments for WM comorbidities can enhance this risk. Therefore, it is important to strengthen our understanding of the molecular basis underlying the WM bleeding phenotype to accurately estimate bleeding risk, to adjust clinical management plans accordingly, minimize bleeding potential and improve patient quality of life.


#
#

Evaluating the Bleeding Phenotype in the Diagnostic Laboratory

Current approaches to WM patients with bleeding include coagulation testing, measurement of plasma viscosity and specialized blood and platelet testing ([Fig. 4]). Further platelet quality and function tests utilizing research tools are emerging ([Fig. 4]). However, these tests present several challenges which will be discussed below.

Zoom Image
Fig. 4 Recommended pathway to evaluate bleeding phenotypes in Waldenström macroglobulinemia, with treatment options. Blue: asymptomatic WM; red: symptomatic WM; green: therapy recommendations[1] (Castillo et al 2019). ADAM10, a disintegrin and metalloproteinase 10; Ag, antigen; aPTT, activated partial thromboplastin time; BM, bone marrow; CBA, collagen binding assay; DOAC, direct oral anticoagulant; FBC, full blood count; FVIII, factor VIII; IgM, immunoglobulin M; LTA, light transmission aggregometry; PFA, platelet function analyzer; PT, prothrombin time; RBC, red blood cell; R:Co: ristocetin cofactor assay; ROTEM, rotational thromboelastometry; SE, side effect; sGPVI, soluble glycoprotein VI; TEG, thromboelastography; TKI, tyrosine kinase inhibitor; VWD, von Willebrand disease; VWF, von Willebrand factor.

Laboratory Tests Assessing Blood Coagulation

Coagulation assays measuring prothrombin time (PT)/international normalized ratio and activated partial thromboplastin time (aPTT) are routinely performed.[15] It should be noted that these tests are influenced by several preanalytical variables which can result in considerable intra- and inter-laboratory variation.[111] Variables include phlebotomy technique, anticoagulant volume based on patient hematocrit, sample mixing and centrifugation, sample transport conditions, delays in transport (over 4 hours), patient age, gender (females have increased levels of certain coagulation factors and antithrombin, and reduced Protein S), physiological states (postsurgery, pregnancy), and drugs (anticoagulants, antiplatelets, anti-inflammatories).

VWF antigen and cofactor binding assays have value in the appropriate clinical context (bleeding phenotype).[15] VWF antigen assays measure VWF levels using a monoclonal antibody in a sandwich-based enzyme-linked immunosorbent assay (ELISA). The original VWF ristocetin cofactor binding assay (VWF:RCo) remains the gold standard method to measure VWF activity in plasma, by evaluating donor platelet agglutination following ristocetin-mediated VWF unfolding and binding to platelet-GPIbα, using light transmission aggregometry (LTA). Unfortunately, this method is insensitive at VWF levels below 20 U/dL, and subject to variation based on the source of ristocetin, variation in donor platelets, and the presence of VWF A1 domain mutations resulting in poor ristocetin–VWF binding.[112] Newer versions of the VWF:RCo assay address sensitivity limitations, for example through a chemiluminescence-based method which directly evaluates VWF binding to magnetic particles coated with recombinant GPIbα in an active configuration (removing the requirement for ristocetin). Additionally, the VWF collagen-binding assay analyses VWF multimers by measuring the preferential binding of high-molecular-weight VWF multimers to collagen, using an ELISA.[113] This assay has been shown to be more sensitive, reproducible, and less variable than the VWF:RCo assay, improving discrimination between functional and nonfunction VWF.[114] VWF multimer analysis by gel electrophoresis is clinically informative but challenging to perform and not widely available as a diagnostic assay.

Thrombin generation assays measure the rate and “hemostatic potential” of plasma for thrombin generation, via the cleavage of a quenched synthetic fluorogenic or chromogenic substrate. Assays require calibration, and although several commercial kits exist, limitations including a lack of standardization and reference values across laboratories have been highlighted and are being addressed.[115] [116] Thrombin generation assays are becoming more prevalent, but will require additional clinical trials with well-defined endpoints to fully determine utility. To date, these assays have been used to monitor anticoagulant or antiplatelet therapy,[117] and abnormal measurements have been associated with bleeding in patients with rare inherited coagulation disorders such as hemophilia[118] and the risk of venous thromboembolism recurrence.[119] Capturing the contribution of blood cells to thrombin potential, however, remains a clear gap in clinical applications of this assay.[120] Platelets accelerate the initiation of thrombin production via provision of membranes bearing phosphatidylserine and receptors that interact with coagulation proteins (GPIb-IX-V complex, GPVI and αIIbβ3 amongst others),[72] as well as the release of granule contents. Platelets, erythrocytes, leukocytes and the endothelium are all likely to modulate thrombin potential in vivo. Further, in the setting of pathologies with high paraprotein levels such as WM, the degree to which high levels of plasma proteins interfere with normal production of thrombin remains to be determined.


#

Laboratory Tests Assessing Platelet Function

Both LTA and the platelet function analyzer (PFA-100 and PFA-200)[121] are established diagnostic platelet function tests. Whole blood impedance aggregometry (Multiplate) is an emerging tool,[122] and together with the PFA-100/200, these techniques enable rapid screening of platelet responses to physiological agonists (adenosine diphosphate, thrombin, collagen). These tests are influenced by the same preanalytical variables as the PT and aPTT tests and require platelet counts to be above 100 × 109/L, so are not suitable for thrombocytopenic patients. The LTA also requires a high sample volume, and elevated levels of bilirubin and lipids increase plasma turbidity and affect LTA data. The PFA-100/200 has the advantage of incorporating fluid shear stress into the assay, and so provides a readout that is more physiological, but remains dependent on an aperture closure time and does not evaluate platelet secretion defects.[123] All of these tests lack sensitivity to changes in receptor levels and can discriminate only certain platelet function disorders.[124]


#

Laboratory Tests Assessing both Blood Coagulation and Platelet Function

Thromboelastography (TEG) and rotational thromboelastometry (ROTEM) are simple, automated, highly sensitive, emerging viscoelastic tests that provide a global assessment of hemostasis and are widely utilized in massive transfusion and acute bleeding scenarios.[125] Parameters including time to clot initiation, rate of clot formation, clot firmness and strength and clot lysis time are quantified, and the contribution of platelets to clot parameters can also be gleaned.[126] Oncologic diseases can cause coagulopathic states that may be identifiable by TEG or ROTEM; however, more work is required to evaluate the utility of these tests for assessing hemostasis beyond surgical bleeding.[125] Further, ROTEM and TEG are not yet standardized for evaluation of thrombocytopenic samples, which may be relevant to patients with hematological diseases, particularly those receiving treatment. There are no data to date specifically evaluating whole blood clotting using TEG or ROTEM in WM.

In summary, evaluating platelet function and coagulation in WM patients presents several challenges. First, none of the standardized platelet functional assays can evaluate platelets in thrombocytopenic WM patients. Second, evaluation of platelet receptor levels, which govern platelet function and are often diminished in hematological malignancies,[84] is not routinely evaluated by flow cytometry. This is due to lack of standardized routine protocols for platelet flow cytometry and lack of clear understanding of its clinical implications. Finally, none of these assays evaluate platelet function under conditions that replicate vascular shear rates found in flowing blood.


#
#

Future Directions

In the research laboratory setting, several additional techniques can be used to evaluate platelet function. The bone marrow microenvironment is disrupted in WM, contributing to the initiation and propagation of WM lymphoplasmacytoid cells and likely disturbing megakaryocyte maturation and platelet production. Therefore, megakaryocyte and platelet flow cytometry can be used to evaluate the levels of receptors (αIIbβ3, GPIbα, GPVI), as well as extent of platelet activation (P-selectin, active αIIbβ3) on circulating platelets. Platelet flow cytometry has the advantage of remaining viable even when the platelet count is extremely low, and useful data can be gathered on chemically fixed samples, meaning samples can be stored for short periods. Levels of shed receptor ectodomains can be quantified by ELISA[127] and microfluidic systems can be used to quantify platelet adhesion to immobilized substrates (collagen, fibrinogen, fibrin) under shear, providing direct readouts of platelet function under conditions that mimic a range of vascular rheological conditions.[128] [129] [130]

The generation of thrombin and fibrin as part of the coagulation pathway plays a crucial role in the securing of platelet aggregates across sites of blood vessel injury. Insufficient levels or defective coagulation factors can lead to formation of an unstable thrombus. Whole blood coagulation, evaluated by ROTEM or TEG, assesses coagulation throughout all phases of clot formation, triggered via extrinsic or intrinsic coagulation pathways. While these parameters have not been previously evaluated in WM patients, it would be of interest to compare samples from newly diagnosed individuals with those on different therapies and ascertain whether a platelet defect can be determined. Additionally, it might be of value to evaluate the effect of WM plasma, particularly from patients with high levels of IgM and hyperviscosity, in mixing experiments using healthy donor plasma-depleted blood to assess the effect of elevated IgM on whole blood coagulation.

Besides enhancing the availability of these tests, it will also be important to define the situations in which these tests will be most helpful. As information on the platelet lesion and more broadly the hemostatic defects in WM emerge, existing diagnostic tools such as ROTEM or TEG may become more widely applied. While platelet flow cytometry and testing for platelet activation markers using ELISAs remain distant from the diagnostic laboratory, these additional new approaches can potentially be developed and incorporated into diagnostic algorithms and may help guide therapy decisions in patients with compromised hemostatic pathways.

Finally, evaluation of WM-related genes within the megakaryocytic progenitor populations has not yet been explored. As platelets and megakaryocytes express both Myd88 and CXCR4,[131] the prevalence of WM-related gene mutations in the megakaryocytes and megakaryocyte progenitor populations should be evaluated. Platelet α-granules possess functional membrane-bound CXCR4 as well as SDF-1. Activating CXCR4 mutations in megakaryocytes, like the CXCR4 S338X mutation, prevents CXCR4 internalization by platelets, following SDF-1 stimulation.[25] This may impair platelet aggregation, thromboxane A2 production and dense granule secretion,[132] and could contribute to a bleeding phenotype.


#

Concluding Remarks

With the advent of BTK inhibitors as efficacious and routine therapies for WM, it is important that patients are evaluated and monitored continually for bleeding propensity. By applying new approaches complemented by sensitive research-based techniques (flow cytometry, thrombin generation assays, platelet spreading assays), in combination with megakaryocyte-specific genetic approaches to evaluate common WM mutations, it may be possible to stratify WM patients for bleeding risk based on platelet functionality. One or more of these techniques could be integrated into routine testing for WM patients at diagnosis and then during treatment, particularly in patients who present with an elevated risk for bleeding.


#
#

Conflict of Interest

None declared.

Acknowledgments

The authors thank Dr. Philip J Crispin for helpful comments. This work was supported by the National Health and Medical Research Council of Australia, the Australian Research Council, the National Blood Authority of Australia, and the Australian Capital Territory Department of Health.

Author Contributions

S.A.B., D.T., and E.E.G. planned and drafted the manuscript. All authors contributed to the review of the manuscript. Images were created using Smart Servier (https://smart.servier.com/).


* Equal senior authors.


  • References

  • 1 Talaulikar D, Tam CS, Joshua D. et al. Treatment of patients with Waldenström macroglobulinaemia: clinical practice guidelines from the Myeloma Foundation of Australia Medical and Scientific Advisory Group. Intern Med J 2017; 47 (01) 35-49
  • 2 García-Sanz R, Montoto S, Torrequebrada A. et al; Spanish Group for the Study of Waldenström Macroglobulinaemia and PETHEMA (Programme for the Study and Treatment of Haematological Malignancies). Waldenström macroglobulinaemia: presenting features and outcome in a series with 217 cases. Br J Haematol 2001; 115 (03) 575-582
  • 3 Treon SP. How I treat Waldenström macroglobulinemia. Blood 2015; 126 (06) 721-732
  • 4 Bustoros M, Sklavenitis-Pistofidis R, Kapoor P. et al. Progression risk stratification of asymptomatic Waldenström macroglobulinemia. J Clin Oncol 2019; 37 (16) 1403-1411
  • 5 Rodriguez S, Celay J, Goicoechea I. et al. Preneoplastic somatic mutations including MYD88 L265P in lymphoplasmacytic lymphoma. Sci Adv 2022; 8 (03) eabl4644
  • 6 Treon SP, Meid K, Hunter ZR. et al. Phase 1 study of ibrutinib and the CXCR4 antagonist ulocuplumab in CXCR4-mutated Waldenström macroglobulinemia. Blood 2021; 138 (17) 1535-1539
  • 7 Treon SP, Xu L, Yang G. et al. MYD88 L265P somatic mutation in Waldenström's macroglobulinemia. N Engl J Med 2012; 367 (09) 826-833
  • 8 Gachard N, Parrens M, Soubeyran I. et al. IGHV gene features and MYD88 L265P mutation separate the three marginal zone lymphoma entities and Waldenström macroglobulinemia/lymphoplasmacytic lymphomas. Leukemia 2013; 27 (01) 183-189
  • 9 Xu L, Hunter ZR, Yang G. et al. MYD88 L265P in Waldenström macroglobulinemia, immunoglobulin M monoclonal gammopathy, and other B-cell lymphoproliferative disorders using conventional and quantitative allele-specific polymerase chain reaction. Blood 2013; 121 (11) 2051-2058
  • 10 Varettoni M, Arcaini L, Zibellini S. et al. Prevalence and clinical significance of the MYD88 (L265P) somatic mutation in Waldenstrom's macroglobulinemia and related lymphoid neoplasms. Blood 2013; 121 (13) 2522-2528
  • 11 Landgren O, Staudt L. MYD88 L265P somatic mutation in IgM MGUS. N Engl J Med 2012; 367 (23) 2255-2256 , author reply 2256–2257
  • 12 Jiménez C, Sebastián E, Chillón MC. et al. MYD88 L265P is a marker highly characteristic of, but not restricted to, Waldenström's macroglobulinemia. Leukemia 2013; 27 (08) 1722-1728
  • 13 Ansell SM, Hodge LS, Secreto FJ. et al. Activation of TAK1 by MYD88 L265P drives malignant B-cell Growth in non-Hodgkin lymphoma. Blood Cancer J 2014; 4: e183
  • 14 Hunter ZR, Xu L, Tsakmaklis N. et al. Insights into the genomic landscape of MYD88 wild-type Waldenström macroglobulinemia. Blood Adv 2018; 2 (21) 2937-2946
  • 15 Maqbool MG, Tam CS, Morison IM. et al. A practical guide to laboratory investigations at diagnosis and follow up in Waldenström macroglobulinaemia: recommendations from the Medical and Scientific Advisory Group, Myeloma Australia, the Pathology Sub-committee of the Lymphoma and Related Diseases Registry and the Australasian Association of Clinical Biochemists Monoclonal Gammopathy Working Group. Pathology 2020; 52 (02) 167-178
  • 16 Roos-Weil D, Giacopelli B, Armand M. et al. Identification of 2 DNA methylation subtypes of Waldenström macroglobulinemia with plasma and memory B-cell features. Blood 2020; 136 (05) 585-595
  • 17 Yang G, Zhou Y, Liu X. et al. A mutation in MYD88 (L265P) supports the survival of lymphoplasmacytic cells by activation of Bruton tyrosine kinase in Waldenström macroglobulinemia. Blood 2013; 122 (07) 1222-1232
  • 18 Deguine J, Barton GM. MyD88: a central player in innate immune signaling. F1000Prime Rep 2014; 6: 97
  • 19 Landgren O, Tageja N. MYD88 and beyond: novel opportunities for diagnosis, prognosis and treatment in Waldenström's Macroglobulinemia. Leukemia 2014; 28 (09) 1799-1803
  • 20 Sewastianik T, Guerrera ML, Adler K. et al. Human MYD88L265P is insufficient by itself to drive neoplastic transformation in mature mouse B cells. Blood Adv 2019; 3 (21) 3360-3374
  • 21 Banerjee M, Huang Y, Joshi S. et al. Platelets endocytose viral particles and are activated via TLR (Toll-like receptor) signaling. Arterioscler Thromb Vasc Biol 2020; 40 (07) 1635-1650
  • 22 Hunter ZR, Xu L, Yang G. et al. The genomic landscape of Waldenstrom macroglobulinemia is characterized by highly recurring MYD88 and WHIM-like CXCR4 mutations, and small somatic deletions associated with B-cell lymphomagenesis. Blood 2014; 123 (11) 1637-1646
  • 23 Poulain S, Roumier C, Venet-Caillault A. et al. Genomic landscape of CXCR4 mutations in Waldenstrom Macroglobulinemia. Clin Cancer Res 2016; 22 (06) 1480-1488
  • 24 Treon SP, Tripsas CK, Meid K. et al. Ibrutinib in previously treated Waldenström's macroglobulinemia. N Engl J Med 2015; 372 (15) 1430-1440
  • 25 Dotta L, Tassone L, Badolato R. Clinical and genetic features of warts, hypogammaglobulinemia, infections and myelokathexis (WHIM) syndrome. Curr Mol Med 2011; 11 (04) 317-325
  • 26 Cao Y, Hunter ZR, Liu X. et al. The WHIM-like CXCR4(S338X) somatic mutation activates AKT and ERK, and promotes resistance to ibrutinib and other agents used in the treatment of Waldenstrom's macroglobulinemia. Leukemia 2015; 29 (01) 169-176
  • 27 Roccaro AM, Sacco A, Jimenez C. et al. C1013G/CXCR4 acts as a driver mutation of tumor progression and modulator of drug resistance in lymphoplasmacytic lymphoma. Blood 2014; 123 (26) 4120-4131
  • 28 Waldenström J. Incipient myelomatosis or «essential« hyperglobulinemia with fibrinogenopenia — a new syndrome?. Acta Med Scand 1944; 117: 216-247
  • 29 Gertz MA, Merlini G, Treon SP. Amyloidosis and Waldenström's macroglobulinemia. Hematology (Am Soc Hematol Educ Program) 2004; •••: 257-282
  • 30 Merchionne F, Procaccio P, Dammacco F. Waldenström's macroglobulinemia. An overview of its clinical, biochemical, immunological and therapeutic features and our series of 121 patients collected in a single center. Crit Rev Oncol Hematol 2011; 80 (01) 87-99
  • 31 Zangari M, Elice F, Fink L, Tricot G. Hemostatic dysfunction in paraproteinemias and amyloidosis. Semin Thromb Hemost 2007; 33 (04) 339-349
  • 32 Buske C, Sadullah S, Kastritis E. et al; European Consortium for Waldenström's Macroglobulinemia. Treatment and outcome patterns in European patients with Waldenström's macroglobulinaemia: a large, observational, retrospective chart review. Lancet Haematol 2018; 5 (07) e299-e309
  • 33 Kuter DJ. Managing thrombocytopenia associated with cancer chemotherapy. Oncology (Williston Park) 2015; 29 (04) 282-294
  • 34 Kolikkat N, Moideen S, Khader A, Mohammed TP, Uvais NA. Waldenstrom's macroglobulinemia: a case report. J Family Med Prim Care 2020; 9 (03) 1768-1771
  • 35 Gardiner EE, Andrews RK. Structure and function of platelet receptors initiating blood clotting. Adv Exp Med Biol 2014; 844: 263-275
  • 36 Mital A. Acquired von Willebrand Syndrome. Adv Clin Exp Med 2016; 25 (06) 1337-1344
  • 37 Federici AB, Rand JH, Bucciarelli P. et al; Subcommittee on von Willebrand Factor. Acquired von Willebrand syndrome: data from an international registry. Thromb Haemost 2000; 84 (02) 345-349
  • 38 Castillo JJ, Gustine JN, Meid K. et al. Low levels of von Willebrand markers associate with high serum IgM levels and improve with response to therapy, in patients with Waldenström macroglobulinaemia. Br J Haematol 2019; 184 (06) 1011-1014
  • 39 Franchini M, Mannucci PM. Acquired von Willebrand syndrome: focused for hematologists. Haematologica 2020; 105 (08) 2032-2037
  • 40 Boros P, Gondolesi G, Bromberg JS. High dose intravenous immunoglobulin treatment: mechanisms of action. Liver Transpl 2005; 11 (12) 1469-1480
  • 41 Abou-Ismail MY, Rodgers GM, Bray PF, Lim MY. Acquired von Willebrand syndrome in monoclonal gammopathy - a scoping review on hemostatic management. Res Pract Thromb Haemost 2021; 5 (02) 356-365
  • 42 Dicke C, Schneppenheim S, Holstein K. et al. Distinct mechanisms account for acquired von Willebrand syndrome in plasma cell dyscrasias. Ann Hematol 2016; 95 (06) 945-957
  • 43 Javadi E, Deng Y, Karniadakis GE, Jamali S. In silico biophysics and hemorheology of blood hyperviscosity syndrome. Biophys J 2021; 120 (13) 2723-2733
  • 44 Stone MJ. Waldenström's macroglobulinemia: hyperviscosity syndrome and cryoglobulinemia. Clin Lymphoma Myeloma 2009; 9 (01) 97-99
  • 45 Gertz MA. Acute hyperviscosity: syndromes and management. Blood 2018; 132 (13) 1379-1385
  • 46 van Breugel HF, de Groot PG, Heethaar RM, Sixma JJ. Role of plasma viscosity in platelet adhesion. Blood 1992; 80 (04) 953-959
  • 47 Castillo JJ, Treon SP. Initial evaluation of the patient with Waldenström macroglobulinemia. Hematol Oncol Clin North Am 2018; 32 (05) 811-820
  • 48 Mayerhofer M, Haushofer A, Kyrle PA. et al. Mechanisms underlying acquired von Willebrand syndrome associated with an IgM paraprotein. Eur J Clin Invest 2009; 39 (09) 833-836
  • 49 McKelvey EM, Kwaan HC. An IgM circulating anticoagulant with factor VIII inhibitory activity. Ann Intern Med 1972; 77 (04) 571-575
  • 50 Castaldi PA, Penny R. A macroglobulin with inhibitory activity against coagulation factor VIII. Blood 1970; 35 (03) 370-376
  • 51 Mazurier C, Parquet-Gernez A, Descamps J, Bauters F, Goudemand M. Acquired von Willebrand's syndrome in the course of Waldenström's disease. Thromb Haemost 1980; 44 (03) 115-118
  • 52 Endo T, Yatomi Y, Amemiya N. et al. Antibody studies of factor VIII inhibitor in a case with Waldenström's macroglobulinemia. Am J Hematol 2000; 63 (03) 145-148
  • 53 Loftus LS, Arnold WN. Acquired hemophilia in a patient with myeloma. West J Med 1994; 160 (02) 173-176
  • 54 Taher A, Abiad R, Uthman I. Coexistence of lupus anticoagulant and acquired haemophilia in a patient with monoclonal gammopathy of unknown significance. Lupus 2003; 12 (11) 854-856
  • 55 Varticovski L, Pick AI, Schattner A, Shoenfeld Y. Anti-platelet and anti-DNA IgM in Waldenström macroglobulinemia and ITP. Am J Hematol 1987; 24 (04) 351-355
  • 56 Owen RG, Lubenko A, Savage J, Parapia LA, Jack AS, Morgan GJ. Autoimmune thrombocytopenia in Waldenström's macroglobulinemia. Am J Hematol 2001; 66 (02) 116-119
  • 57 Zago-Novaretti M, Khuri F, Miller KB, Berkman EM. Waldenström's macroglobulinemia with an IgM paraprotein that is both a cold agglutinin and a cryoglobulin and has a suppressive effect on progenitor cell growth. Transfusion 1994; 34 (10) 910-914
  • 58 Stone MJ, Pascual V. Pathophysiology of Waldenström's macroglobulinemia. Haematologica 2010; 95 (03) 359-364
  • 59 Nicol M, Siguret V, Vergaro G. et al. Thromboembolism and bleeding in systemic amyloidosis: a review. ESC Heart Fail 2022; 9 (01) 11-20
  • 60 Zanwar S, Abeykoon JP, Ansell SM. et al. Primary systemic amyloidosis in patients with Waldenström macroglobulinemia. Leukemia 2019; 33 (03) 790-794
  • 61 Sundaram S, Rathod R. Gastric amyloidosis causing nonvariceal upper gastrointestinal bleeding. ACG Case Rep J 2019; 6 (05) 3-4
  • 62 Osman K, Comenzo R, Rajkumar SV. Deep venous thrombosis and thalidomide therapy for multiple myeloma. N Engl J Med 2001; 344 (25) 1951-1952
  • 63 Mitrani LR, De Los Santos J, Driggin E. et al. Anticoagulation with warfarin compared to novel oral anticoagulants for atrial fibrillation in adults with transthyretin cardiac amyloidosis: comparison of thromboembolic events and major bleeding. Amyloid 2021; 28 (01) 30-34
  • 64 Gamba G, Montani N, Anesi E. et al. Abnormalities in thrombin-antithrombin pathway in AL amyloidosis. Amyloid 1999; 6 (04) 273-277
  • 65 Cowan AJ, Skinner M, Seldin DC. et al. Amyloidosis of the gastrointestinal tract: a 13-year, single-center, referral experience. Haematologica 2013; 98 (01) 141-146
  • 66 Patel G, Hari P, Szabo A. et al. Acquired factor X deficiency in light-chain (AL) amyloidosis is rare and associated with advanced disease. Hematol Oncol Stem Cell Ther 2019; 12 (01) 10-14
  • 67 Hicks SM, Coupland LA, Jahangiri A, Choi PY, Gardiner EE. Novel scientific approaches and future research directions in understanding ITP. Platelets 2020; 31 (03) 315-321
  • 68 Neunert C, Noroozi N, Norman G. et al. Severe bleeding events in adults and children with primary immune thrombocytopenia: a systematic review. J Thromb Haemost 2015; 13 (03) 457-464
  • 69 Vinholt PJ, Hvas AM, Nybo M. An overview of platelet indices and methods for evaluating platelet function in thrombocytopenic patients. Eur J Haematol 2014; 92 (05) 367-376
  • 70 Hansen CE, Qiu Y, McCarty OJT, Lam WA. Platelet mechanotransduction. Annu Rev Biomed Eng 2018; 20: 253-275
  • 71 Ruggeri ZM. Platelet adhesion under flow. Microcirculation 2009; 16 (01) 58-83
  • 72 Andrews RK, Gardiner EE, Shen Y, Berndt MC. Platelet interactions in thrombosis. IUBMB Life 2004; 56 (01) 13-18
  • 73 Muthiah K, Connor D, Ly K. et al. Longitudinal changes in hemostatic parameters and reduced pulsatility contribute to non-surgical bleeding in patients with centrifugal continuous-flow left ventricular assist devices. J Heart Lung Transplant 2016; 35 (06) 743-751
  • 74 Vulliamy P, Montague SJ, Gillespie S. et al. Loss of GPVI and GPIbα contributes to trauma-induced platelet dysfunction in severely injured patients. Blood Adv 2020; 4 (12) 2623-2630
  • 75 Qiao J, Schoenwaelder SM, Mason KD. et al. Low adhesion receptor levels on circulating platelets in patients with lymphoproliferative diseases before receiving Navitoclax (ABT-263). Blood 2013; 121 (08) 1479-1481
  • 76 Kamel S, Horton L, Ysebaert L. et al. Ibrutinib inhibits collagen-mediated but not ADP-mediated platelet aggregation. Leukemia 2015; 29 (04) 783-787
  • 77 Thomas S, Krishnan A. Platelet heterogeneity in myeloproliferative neoplasms. Arterioscler Thromb Vasc Biol 2021; 41 (11) 2661-2670
  • 78 Kaplan ZS, Zarpellon A, Alwis I. et al. Thrombin-dependent intravascular leukocyte trafficking regulated by fibrin and the platelet receptors GPIb and PAR4. Nat Commun 2015; 6: 7835
  • 79 Mammadova-Bach E, Ollivier V, Loyau S. et al. Platelet glycoprotein VI binds to polymerized fibrin and promotes thrombin generation. Blood 2015; 126 (05) 683-691
  • 80 Dumas JJ, Kumar R, Seehra J, Somers WS, Mosyak L. Crystal structure of the GpIbalpha-thrombin complex essential for platelet aggregation. Science 2003; 301 (5630): 222-226
  • 81 Byzova TV, Plow EF. Networking in the hemostatic system. Integrin alphaiibbeta3 binds prothrombin and influences its activation. J Biol Chem 1997; 272 (43) 27183-27188
  • 82 Haider S, Latif T, Hochhausler A, Lucas F, Abdel Karim N. Waldenstrom's macroglobulinemia and peripheral neuropathy, organomegaly, endocrinopathy, monoclonal gammopathy, and skin changes with a bleeding diathesis and rash. Case Rep Oncol Med 2013; 2013: 890864
  • 83 Zangari M, Elice F, Tricot G, Fink L. Bleeding disorders associated with cancer dysproteinemias. Cancer Treat Res 2009; 148: 295-304
  • 84 Luu S, Gardiner EE, Andrews RK. Bone marrow defects and platelet function: a focus on MDS and CLL. Cancers (Basel) 2018; 10 (05) 147
  • 85 Castillo JJ, Advani RH, Branagan AR. et al. Consensus treatment recommendations from the tenth International Workshop for Waldenström Macroglobulinaemia. Lancet Haematol 2020; 7 (11) e827-e837
  • 86 Salles G, Barrett M, Foà R. et al. Rituximab in B-cell hematologic malignancies: a review of 20 years of clinical experience. Adv Ther 2017; 34 (10) 2232-2273
  • 87 Ghobrial IM, Fonseca R, Greipp PR. et al; Eastern Cooperative Oncology Group. 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
  • 88 Ram R, Bonstein L, Gafter-Gvili A, Ben-Bassat I, Shpilberg O, Raanani P. Rituximab-associated acute thrombocytopenia: an under-diagnosed phenomenon. Am J Hematol 2009; 84 (04) 247-250
  • 89 Cheson BD, Leoni L. Bendamustine: mechanism of action and clinical data. Clin Adv Hematol Oncol 2011; 9 (08, Suppl 19): 1-11
  • 90 Robak E, Robak T. Bruton's kinase inhibitors for the treatment of immunological diseases: current status and perspectives. J Clin Med 2022; 11 (10) 11
  • 91 von Hundelshausen P, Siess W. Bleeding by Bruton tyrosine kinase-inhibitors: dependency on drug type and disease. Cancers (Basel) 2021; 13 (05) 13
  • 92 Liu J, Fitzgerald ME, Berndt MC, Jackson CW, Gartner TK. Bruton tyrosine kinase is essential for botrocetin/VWF-induced signaling and GPIb-dependent thrombus formation in vivo. Blood 2006; 108 (08) 2596-2603
  • 93 Li Z, Delaney MK, O'Brien KA, Du X. Signaling during platelet adhesion and activation. Arterioscler Thromb Vasc Biol 2010; 30 (12) 2341-2349
  • 94 Atkinson BT, Ellmeier W, Watson SP. Tec regulates platelet activation by GPVI in the absence of Btk. Blood 2003; 102 (10) 3592-3599
  • 95 Burger JA, Buggy JJ. Bruton tyrosine kinase inhibitor ibrutinib (PCI-32765). Leuk Lymphoma 2013; 54 (11) 2385-2391
  • 96 Shatzel JJ, Olson SR, Tao DL, McCarty OJT, Danilov AV, DeLoughery TG. Ibrutinib-associated bleeding: pathogenesis, management and risk reduction strategies. J Thromb Haemost 2017; 15 (05) 835-847
  • 97 Levade M, David E, Garcia C. et al. Ibrutinib treatment affects collagen and von Willebrand factor-dependent platelet functions. Blood 2014; 124 (26) 3991-3995
  • 98 Bye AP, Unsworth AJ, Desborough MJ. et al. Severe platelet dysfunction in NHL patients receiving ibrutinib is absent in patients receiving acalabrutinib. Blood Adv 2017; 1 (26) 2610-2623
  • 99 Tullemans BME, Heemskerk JWM, Kuijpers MJE. Acquired platelet antagonism: off-target antiplatelet effects of malignancy treatment with tyrosine kinase inhibitors. J Thromb Haemost 2018; 16 (09) 1686-1699
  • 100 Dobie G, Kuriri FA, Omar MMA. et al. Ibrutinib, but not zanubrutinib, induces platelet receptor shedding of GPIb-IX-V complex and integrin αIIbβ3 in mice and humans. Blood Adv 2019; 3 (24) 4298-4311
  • 101 Brown JR, Moslehi J, Ewer MS. et al. Incidence of and risk factors for major haemorrhage in patients treated with ibrutinib: an integrated analysis. Br J Haematol 2019; 184 (04) 558-569
  • 102 Castillo JJ, Gustine JN, Meid K, Dubeau T, Severns P, Treon SP. Ibrutinib withdrawal symptoms in patients with Waldenström macroglobulinemia. Haematologica 2018; 103 (07) e307-e310
  • 103 Gustine JN, Meid K, Dubeau T. et al. Ibrutinib discontinuation in Waldenström macroglobulinemia: etiologies, outcomes, and IgM rebound. Am J Hematol 2018; 93 (04) 511-517
  • 104 Gustine JN, Meid K, Dubeau TE, Treon SP, Castillo JJ. Atrial fibrillation associated with ibrutinib in Waldenström macroglobulinemia. Am J Hematol 2016; 91 (06) E312-E313
  • 105 Ali N, Malik F, Jafri SIM, Naglak M, Sundermeyer M, Pickens PV. Analysis of efficacy and tolerability of Bruton tyrosine kinase inhibitor ibrutinib in various B-cell malignancies in the general community: a single-center experience. Clin Lymphoma Myeloma Leuk 2017; 17S: S53-S61
  • 106 Dimopoulos MA, Tedeschi A, Trotman J. et al; iNNOVATE Study Group and the European Consortium for Waldenström's Macroglobulinemia. Phase 3 trial of ibrutinib plus rituximab in Waldenström's macroglobulinemia. N Engl J Med 2018; 378 (25) 2399-2410
  • 107 Dimopoulos MA, Trotman J, Tedeschi A. et al; iNNOVATE Study Group and the European Consortium for Waldenström's Macroglobulinemia. Ibrutinib for patients with rituximab-refractory Waldenström's macroglobulinaemia (iNNOVATE): an open-label substudy of an international, multicentre, phase 3 trial. Lancet Oncol 2017; 18 (02) 241-250
  • 108 Treon SP, Gustine J, Meid K. et al. Ibrutinib monotherapy in symptomatic, treatment-naive patients with Waldenstrom macroglobulinemia. J Clin Oncol 2018; 36 (27) 2755-2761
  • 109 Fradley MG, Gliksman M, Emole J. et al. Rates and risk of atrial arrhythmias in patients treated with ibrutinib compared with cytotoxic chemotherapy. Am J Cardiol 2019; 124 (04) 539-544
  • 110 Abeykoon JP, Zanwar S, Ansell SM. et al. Ibrutinib monotherapy outside of clinical trial setting in Waldenström macroglobulinaemia: practice patterns, toxicities and outcomes. Br J Haematol 2020; 188 (03) 394-403
  • 111 Favaloro EJ, Funk DM, Lippi G. Pre-analytical variables in coagulation testing associated with diagnostic errors in hemostasis. Lab Med 2012; 43: 1-10
  • 112 Sharma R, Haberichter SL. New advances in the diagnosis of von Willebrand disease. Hematology (Am Soc Hematol Educ Program) 2019; 2019 (01) 596-600
  • 113 Favaloro EJ, Oliver S, Mohammed S, Vong R. Comparative assessment of von Willebrand factor multimers vs activity for von Willebrand disease using modern contemporary methodologies. Haemophilia 2020; 26 (03) 503-512
  • 114 Laporte P, Tuffigo M, Ryman A. et al. HemosIL VWF:GPIbR assay has a greater sensitivity than VWF:RCo technique to detect acquired von Willebrand syndrome in myeloproliferative neoplasms. Thromb Haemost 2022; 122 (10) 1673-1682
  • 115 Lim HY, Donnan G, Nandurkar H, Ho P. Global coagulation assays in hypercoagulable states. J Thromb Thrombolysis 2022; 54 (01) 132-144
  • 116 Ninivaggi M, de Laat-Kremers R, Tripodi A. et al. Recommendations for the measurement of thrombin generation: communication from the ISTH SSC Subcommittee on Lupus Anticoagulant/Antiphospholipid Antibodies. J Thromb Haemost 2021; 19 (05) 1372-1378
  • 117 de Breet CPDM, Zwaveling S, Vries MJA. et al. Thrombin generation as a method to identify the risk of bleeding in high clinical-risk patients using dual antiplatelet therapy. Front Cardiovasc Med 2021; 8: 679934
  • 118 Beltrán-Miranda CP, Khan A, Jaloma-Cruz AR, Laffan MA. Thrombin generation and phenotypic correlation in haemophilia A. Haemophilia 2005; 11 (04) 326-334
  • 119 Tripodi A, Martinelli I, Chantarangkul V, Battaglioli T, Clerici M, Mannucci PM. The endogenous thrombin potential and the risk of venous thromboembolism. Thromb Res 2007; 121 (03) 353-359
  • 120 Wan J, Konings J, de Laat B, Hackeng TM, Roest M. Added value of blood cells in thrombin generation testing. Thromb Haemost 2021; 121 (12) 1574-1587
  • 121 Favaloro EJ, Bonar R. An update on quality control for the PFA-100/PFA-200. Platelets 2018; 29 (06) 622-627
  • 122 Vinholt PJ. The role of platelets in bleeding in patients with thrombocytopenia and hematological disease. Clin Chem Lab Med 2019; 57 (12) 1808-1817
  • 123 Paniccia R, Priora R, Liotta AA, Abbate R. Platelet function tests: a comparative review. Vasc Health Risk Manag 2015; 11: 133-148
  • 124 Moenen FCJI, Vries MJA, Nelemans PJ. et al. Screening for platelet function disorders with Multiplate and platelet function analyzer. Platelets 2019; 30 (01) 81-87
  • 125 Walsh M, Kwaan H, McCauley R. et al. Viscoelastic testing in oncology patients (including for the diagnosis of fibrinolysis): review of existing evidence, technology comparison, and clinical utility. Transfusion 2020; 60 (Suppl. 06) S86-S100
  • 126 Kay AB, Morris DS, Collingridge DS, Majercik S. Platelet dysfunction on thromboelastogram is associated with severity of blunt traumatic brain injury. Am J Surg 2019; 218 (06) 1134-1137
  • 127 Al-Tamimi M, Arthur JF, Gardiner E, Andrews RK. Focusing on plasma glycoprotein VI. Thromb Haemost 2012; 107 (04) 648-655
  • 128 Lui M, Gardiner EE, Arthur JF. et al. Novel stenotic microchannels to study thrombus formation in shear gradients: influence of shear forces and human platelet-related factors. Int J Mol Sci 2019; 20 (12) 20
  • 129 Mangin PH, Gardiner EE, Nesbitt WS. et al; Subcommittee on Biorheology. In vitro flow based systems to study platelet function and thrombus formation: recommendations for standardization: Communication from the SSC on Biorheology of the ISTH. J Thromb Haemost 2020; 18 (03) 748-752
  • 130 de Witt SM, Swieringa F, Cavill R. et al. Identification of platelet function defects by multi-parameter assessment of thrombus formation. Nat Commun 2014; 5: 4257
  • 131 Burkhart JM, Vaudel M, Gambaryan S. et al. The first comprehensive and quantitative analysis of human platelet protein composition allows the comparative analysis of structural and functional pathways. Blood 2012; 120 (15) e73-e82
  • 132 Chatterjee M, Rath D, Gawaz M. Role of chemokine receptors CXCR4 and CXCR7 for platelet function. Biochem Soc Trans 2015; 43 (04) 720-726
  • 133 Hivert B, Caron C, Petit S. et al. Clinical and prognostic implications of low or high level of von Willebrand factor in patients with Waldenstrom macroglobulinemia. Blood 2012; 120 (16) 3214-3221
  • 134 Gavriatopoulou M, Terpos E, Ntanasis-Stathopoulos I. et al. Elevated vWF antigen serum levels are associated with poor prognosis, and decreased circulating ADAMTS-13 antigen levels are associated with increased IgM levels and features of WM but not increased vWF levels in patients with symptomatic WM. Clin Lymphoma Myeloma Leuk 2019; 19 (01) 23-28
  • 135 Stockschlaeder M, Schneppenheim R, Budde U. Update on von Willebrand factor multimers: focus on high-molecular-weight multimers and their role in hemostasis. Blood Coagul Fibrinolysis 2014; 25 (03) 206-216
  • 136 Shahani T, Covens K, Lavend'homme R. et al. Human liver sinusoidal endothelial cells but not hepatocytes contain factor VIII. J Thromb Haemost 2014; 12 (01) 36-42
  • 137 Federici AB. The factor VIII/von Willebrand factor complex: basic and clinical issues. Haematologica 2003; 88 (06) EREP02
  • 138 Saraya AK, Kasturi J, Kishan R. A study of haemostasis in macroglobulinaemia. Acta Haematol 1972; 47: 33-42
  • 139 Kasturi J, Saraya AK. Platelet functions in dysproteinaemia. Acta Haematol 1978; 59 (02) 104-113
  • 140 Camera M, Brambilla M, Toschi V, Tremoli E. Tissue factor expression on platelets is a dynamic event. Blood 2010; 116 (23) 5076-5077
  • 141 Siddiqui FA, Desai H, Amirkhosravi A, Amaya M, Francis JL. The presence and release of tissue factor from human platelets. Platelets 2002; 13 (04) 247-253
  • 142 Estupiñán HY, Berglöf A, Zain R, Smith CIE. Comparative analysis of BTK inhibitors and mechanisms underlying adverse effects. Front Cell Dev Biol 2021; 9: 630942
  • 143 Kaptein A, de Bruin G, Emmelot-van Hoek M. et al. Potency and selectivity of BTK inhibitors in clinical development for B-cell malignancies. Blood 2018; 132: 1871-1871
  • 144 Brown JR. Ibrutinib in chronic lymphocytic leukemia and B cell malignancies. Leuk Lymphoma 2014; 55 (02) 263-269
  • 145 Perkins HA, MacKenzie MR, Fudenberg HH. Hemostatic defects in dysproteinemias. Blood 1970; 35 (05) 695-707
  • 146 Merlini G, Baldini L, Broglia C. et al. Prognostic factors in symptomatic Waldenstrom's macroglobulinemia. Semin Oncol 2003; 30 (02) 211-215

Address for correspondence

Elizabeth E. Gardiner, PhD
ACRF Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research, Australian National University
131 Garran Rd, Canberra, ACT 2601 Australia

Publication History

Received: 08 March 2022

Accepted: 22 June 2022

Accepted Manuscript online:
11 July 2022

Article published online:
17 October 2022

© 2022. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution-NonDerivative-NonCommercial License, permitting copying and reproduction so long as the original work is given appropriate credit. Contents may not be used for commercial purposes, or adapted, remixed, transformed or built upon. (https://creativecommons.org/licenses/by-nc-nd/4.0/)

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

  • References

  • 1 Talaulikar D, Tam CS, Joshua D. et al. Treatment of patients with Waldenström macroglobulinaemia: clinical practice guidelines from the Myeloma Foundation of Australia Medical and Scientific Advisory Group. Intern Med J 2017; 47 (01) 35-49
  • 2 García-Sanz R, Montoto S, Torrequebrada A. et al; Spanish Group for the Study of Waldenström Macroglobulinaemia and PETHEMA (Programme for the Study and Treatment of Haematological Malignancies). Waldenström macroglobulinaemia: presenting features and outcome in a series with 217 cases. Br J Haematol 2001; 115 (03) 575-582
  • 3 Treon SP. How I treat Waldenström macroglobulinemia. Blood 2015; 126 (06) 721-732
  • 4 Bustoros M, Sklavenitis-Pistofidis R, Kapoor P. et al. Progression risk stratification of asymptomatic Waldenström macroglobulinemia. J Clin Oncol 2019; 37 (16) 1403-1411
  • 5 Rodriguez S, Celay J, Goicoechea I. et al. Preneoplastic somatic mutations including MYD88 L265P in lymphoplasmacytic lymphoma. Sci Adv 2022; 8 (03) eabl4644
  • 6 Treon SP, Meid K, Hunter ZR. et al. Phase 1 study of ibrutinib and the CXCR4 antagonist ulocuplumab in CXCR4-mutated Waldenström macroglobulinemia. Blood 2021; 138 (17) 1535-1539
  • 7 Treon SP, Xu L, Yang G. et al. MYD88 L265P somatic mutation in Waldenström's macroglobulinemia. N Engl J Med 2012; 367 (09) 826-833
  • 8 Gachard N, Parrens M, Soubeyran I. et al. IGHV gene features and MYD88 L265P mutation separate the three marginal zone lymphoma entities and Waldenström macroglobulinemia/lymphoplasmacytic lymphomas. Leukemia 2013; 27 (01) 183-189
  • 9 Xu L, Hunter ZR, Yang G. et al. MYD88 L265P in Waldenström macroglobulinemia, immunoglobulin M monoclonal gammopathy, and other B-cell lymphoproliferative disorders using conventional and quantitative allele-specific polymerase chain reaction. Blood 2013; 121 (11) 2051-2058
  • 10 Varettoni M, Arcaini L, Zibellini S. et al. Prevalence and clinical significance of the MYD88 (L265P) somatic mutation in Waldenstrom's macroglobulinemia and related lymphoid neoplasms. Blood 2013; 121 (13) 2522-2528
  • 11 Landgren O, Staudt L. MYD88 L265P somatic mutation in IgM MGUS. N Engl J Med 2012; 367 (23) 2255-2256 , author reply 2256–2257
  • 12 Jiménez C, Sebastián E, Chillón MC. et al. MYD88 L265P is a marker highly characteristic of, but not restricted to, Waldenström's macroglobulinemia. Leukemia 2013; 27 (08) 1722-1728
  • 13 Ansell SM, Hodge LS, Secreto FJ. et al. Activation of TAK1 by MYD88 L265P drives malignant B-cell Growth in non-Hodgkin lymphoma. Blood Cancer J 2014; 4: e183
  • 14 Hunter ZR, Xu L, Tsakmaklis N. et al. Insights into the genomic landscape of MYD88 wild-type Waldenström macroglobulinemia. Blood Adv 2018; 2 (21) 2937-2946
  • 15 Maqbool MG, Tam CS, Morison IM. et al. A practical guide to laboratory investigations at diagnosis and follow up in Waldenström macroglobulinaemia: recommendations from the Medical and Scientific Advisory Group, Myeloma Australia, the Pathology Sub-committee of the Lymphoma and Related Diseases Registry and the Australasian Association of Clinical Biochemists Monoclonal Gammopathy Working Group. Pathology 2020; 52 (02) 167-178
  • 16 Roos-Weil D, Giacopelli B, Armand M. et al. Identification of 2 DNA methylation subtypes of Waldenström macroglobulinemia with plasma and memory B-cell features. Blood 2020; 136 (05) 585-595
  • 17 Yang G, Zhou Y, Liu X. et al. A mutation in MYD88 (L265P) supports the survival of lymphoplasmacytic cells by activation of Bruton tyrosine kinase in Waldenström macroglobulinemia. Blood 2013; 122 (07) 1222-1232
  • 18 Deguine J, Barton GM. MyD88: a central player in innate immune signaling. F1000Prime Rep 2014; 6: 97
  • 19 Landgren O, Tageja N. MYD88 and beyond: novel opportunities for diagnosis, prognosis and treatment in Waldenström's Macroglobulinemia. Leukemia 2014; 28 (09) 1799-1803
  • 20 Sewastianik T, Guerrera ML, Adler K. et al. Human MYD88L265P is insufficient by itself to drive neoplastic transformation in mature mouse B cells. Blood Adv 2019; 3 (21) 3360-3374
  • 21 Banerjee M, Huang Y, Joshi S. et al. Platelets endocytose viral particles and are activated via TLR (Toll-like receptor) signaling. Arterioscler Thromb Vasc Biol 2020; 40 (07) 1635-1650
  • 22 Hunter ZR, Xu L, Yang G. et al. The genomic landscape of Waldenstrom macroglobulinemia is characterized by highly recurring MYD88 and WHIM-like CXCR4 mutations, and small somatic deletions associated with B-cell lymphomagenesis. Blood 2014; 123 (11) 1637-1646
  • 23 Poulain S, Roumier C, Venet-Caillault A. et al. Genomic landscape of CXCR4 mutations in Waldenstrom Macroglobulinemia. Clin Cancer Res 2016; 22 (06) 1480-1488
  • 24 Treon SP, Tripsas CK, Meid K. et al. Ibrutinib in previously treated Waldenström's macroglobulinemia. N Engl J Med 2015; 372 (15) 1430-1440
  • 25 Dotta L, Tassone L, Badolato R. Clinical and genetic features of warts, hypogammaglobulinemia, infections and myelokathexis (WHIM) syndrome. Curr Mol Med 2011; 11 (04) 317-325
  • 26 Cao Y, Hunter ZR, Liu X. et al. The WHIM-like CXCR4(S338X) somatic mutation activates AKT and ERK, and promotes resistance to ibrutinib and other agents used in the treatment of Waldenstrom's macroglobulinemia. Leukemia 2015; 29 (01) 169-176
  • 27 Roccaro AM, Sacco A, Jimenez C. et al. C1013G/CXCR4 acts as a driver mutation of tumor progression and modulator of drug resistance in lymphoplasmacytic lymphoma. Blood 2014; 123 (26) 4120-4131
  • 28 Waldenström J. Incipient myelomatosis or «essential« hyperglobulinemia with fibrinogenopenia — a new syndrome?. Acta Med Scand 1944; 117: 216-247
  • 29 Gertz MA, Merlini G, Treon SP. Amyloidosis and Waldenström's macroglobulinemia. Hematology (Am Soc Hematol Educ Program) 2004; •••: 257-282
  • 30 Merchionne F, Procaccio P, Dammacco F. Waldenström's macroglobulinemia. An overview of its clinical, biochemical, immunological and therapeutic features and our series of 121 patients collected in a single center. Crit Rev Oncol Hematol 2011; 80 (01) 87-99
  • 31 Zangari M, Elice F, Fink L, Tricot G. Hemostatic dysfunction in paraproteinemias and amyloidosis. Semin Thromb Hemost 2007; 33 (04) 339-349
  • 32 Buske C, Sadullah S, Kastritis E. et al; European Consortium for Waldenström's Macroglobulinemia. Treatment and outcome patterns in European patients with Waldenström's macroglobulinaemia: a large, observational, retrospective chart review. Lancet Haematol 2018; 5 (07) e299-e309
  • 33 Kuter DJ. Managing thrombocytopenia associated with cancer chemotherapy. Oncology (Williston Park) 2015; 29 (04) 282-294
  • 34 Kolikkat N, Moideen S, Khader A, Mohammed TP, Uvais NA. Waldenstrom's macroglobulinemia: a case report. J Family Med Prim Care 2020; 9 (03) 1768-1771
  • 35 Gardiner EE, Andrews RK. Structure and function of platelet receptors initiating blood clotting. Adv Exp Med Biol 2014; 844: 263-275
  • 36 Mital A. Acquired von Willebrand Syndrome. Adv Clin Exp Med 2016; 25 (06) 1337-1344
  • 37 Federici AB, Rand JH, Bucciarelli P. et al; Subcommittee on von Willebrand Factor. Acquired von Willebrand syndrome: data from an international registry. Thromb Haemost 2000; 84 (02) 345-349
  • 38 Castillo JJ, Gustine JN, Meid K. et al. Low levels of von Willebrand markers associate with high serum IgM levels and improve with response to therapy, in patients with Waldenström macroglobulinaemia. Br J Haematol 2019; 184 (06) 1011-1014
  • 39 Franchini M, Mannucci PM. Acquired von Willebrand syndrome: focused for hematologists. Haematologica 2020; 105 (08) 2032-2037
  • 40 Boros P, Gondolesi G, Bromberg JS. High dose intravenous immunoglobulin treatment: mechanisms of action. Liver Transpl 2005; 11 (12) 1469-1480
  • 41 Abou-Ismail MY, Rodgers GM, Bray PF, Lim MY. Acquired von Willebrand syndrome in monoclonal gammopathy - a scoping review on hemostatic management. Res Pract Thromb Haemost 2021; 5 (02) 356-365
  • 42 Dicke C, Schneppenheim S, Holstein K. et al. Distinct mechanisms account for acquired von Willebrand syndrome in plasma cell dyscrasias. Ann Hematol 2016; 95 (06) 945-957
  • 43 Javadi E, Deng Y, Karniadakis GE, Jamali S. In silico biophysics and hemorheology of blood hyperviscosity syndrome. Biophys J 2021; 120 (13) 2723-2733
  • 44 Stone MJ. Waldenström's macroglobulinemia: hyperviscosity syndrome and cryoglobulinemia. Clin Lymphoma Myeloma 2009; 9 (01) 97-99
  • 45 Gertz MA. Acute hyperviscosity: syndromes and management. Blood 2018; 132 (13) 1379-1385
  • 46 van Breugel HF, de Groot PG, Heethaar RM, Sixma JJ. Role of plasma viscosity in platelet adhesion. Blood 1992; 80 (04) 953-959
  • 47 Castillo JJ, Treon SP. Initial evaluation of the patient with Waldenström macroglobulinemia. Hematol Oncol Clin North Am 2018; 32 (05) 811-820
  • 48 Mayerhofer M, Haushofer A, Kyrle PA. et al. Mechanisms underlying acquired von Willebrand syndrome associated with an IgM paraprotein. Eur J Clin Invest 2009; 39 (09) 833-836
  • 49 McKelvey EM, Kwaan HC. An IgM circulating anticoagulant with factor VIII inhibitory activity. Ann Intern Med 1972; 77 (04) 571-575
  • 50 Castaldi PA, Penny R. A macroglobulin with inhibitory activity against coagulation factor VIII. Blood 1970; 35 (03) 370-376
  • 51 Mazurier C, Parquet-Gernez A, Descamps J, Bauters F, Goudemand M. Acquired von Willebrand's syndrome in the course of Waldenström's disease. Thromb Haemost 1980; 44 (03) 115-118
  • 52 Endo T, Yatomi Y, Amemiya N. et al. Antibody studies of factor VIII inhibitor in a case with Waldenström's macroglobulinemia. Am J Hematol 2000; 63 (03) 145-148
  • 53 Loftus LS, Arnold WN. Acquired hemophilia in a patient with myeloma. West J Med 1994; 160 (02) 173-176
  • 54 Taher A, Abiad R, Uthman I. Coexistence of lupus anticoagulant and acquired haemophilia in a patient with monoclonal gammopathy of unknown significance. Lupus 2003; 12 (11) 854-856
  • 55 Varticovski L, Pick AI, Schattner A, Shoenfeld Y. Anti-platelet and anti-DNA IgM in Waldenström macroglobulinemia and ITP. Am J Hematol 1987; 24 (04) 351-355
  • 56 Owen RG, Lubenko A, Savage J, Parapia LA, Jack AS, Morgan GJ. Autoimmune thrombocytopenia in Waldenström's macroglobulinemia. Am J Hematol 2001; 66 (02) 116-119
  • 57 Zago-Novaretti M, Khuri F, Miller KB, Berkman EM. Waldenström's macroglobulinemia with an IgM paraprotein that is both a cold agglutinin and a cryoglobulin and has a suppressive effect on progenitor cell growth. Transfusion 1994; 34 (10) 910-914
  • 58 Stone MJ, Pascual V. Pathophysiology of Waldenström's macroglobulinemia. Haematologica 2010; 95 (03) 359-364
  • 59 Nicol M, Siguret V, Vergaro G. et al. Thromboembolism and bleeding in systemic amyloidosis: a review. ESC Heart Fail 2022; 9 (01) 11-20
  • 60 Zanwar S, Abeykoon JP, Ansell SM. et al. Primary systemic amyloidosis in patients with Waldenström macroglobulinemia. Leukemia 2019; 33 (03) 790-794
  • 61 Sundaram S, Rathod R. Gastric amyloidosis causing nonvariceal upper gastrointestinal bleeding. ACG Case Rep J 2019; 6 (05) 3-4
  • 62 Osman K, Comenzo R, Rajkumar SV. Deep venous thrombosis and thalidomide therapy for multiple myeloma. N Engl J Med 2001; 344 (25) 1951-1952
  • 63 Mitrani LR, De Los Santos J, Driggin E. et al. Anticoagulation with warfarin compared to novel oral anticoagulants for atrial fibrillation in adults with transthyretin cardiac amyloidosis: comparison of thromboembolic events and major bleeding. Amyloid 2021; 28 (01) 30-34
  • 64 Gamba G, Montani N, Anesi E. et al. Abnormalities in thrombin-antithrombin pathway in AL amyloidosis. Amyloid 1999; 6 (04) 273-277
  • 65 Cowan AJ, Skinner M, Seldin DC. et al. Amyloidosis of the gastrointestinal tract: a 13-year, single-center, referral experience. Haematologica 2013; 98 (01) 141-146
  • 66 Patel G, Hari P, Szabo A. et al. Acquired factor X deficiency in light-chain (AL) amyloidosis is rare and associated with advanced disease. Hematol Oncol Stem Cell Ther 2019; 12 (01) 10-14
  • 67 Hicks SM, Coupland LA, Jahangiri A, Choi PY, Gardiner EE. Novel scientific approaches and future research directions in understanding ITP. Platelets 2020; 31 (03) 315-321
  • 68 Neunert C, Noroozi N, Norman G. et al. Severe bleeding events in adults and children with primary immune thrombocytopenia: a systematic review. J Thromb Haemost 2015; 13 (03) 457-464
  • 69 Vinholt PJ, Hvas AM, Nybo M. An overview of platelet indices and methods for evaluating platelet function in thrombocytopenic patients. Eur J Haematol 2014; 92 (05) 367-376
  • 70 Hansen CE, Qiu Y, McCarty OJT, Lam WA. Platelet mechanotransduction. Annu Rev Biomed Eng 2018; 20: 253-275
  • 71 Ruggeri ZM. Platelet adhesion under flow. Microcirculation 2009; 16 (01) 58-83
  • 72 Andrews RK, Gardiner EE, Shen Y, Berndt MC. Platelet interactions in thrombosis. IUBMB Life 2004; 56 (01) 13-18
  • 73 Muthiah K, Connor D, Ly K. et al. Longitudinal changes in hemostatic parameters and reduced pulsatility contribute to non-surgical bleeding in patients with centrifugal continuous-flow left ventricular assist devices. J Heart Lung Transplant 2016; 35 (06) 743-751
  • 74 Vulliamy P, Montague SJ, Gillespie S. et al. Loss of GPVI and GPIbα contributes to trauma-induced platelet dysfunction in severely injured patients. Blood Adv 2020; 4 (12) 2623-2630
  • 75 Qiao J, Schoenwaelder SM, Mason KD. et al. Low adhesion receptor levels on circulating platelets in patients with lymphoproliferative diseases before receiving Navitoclax (ABT-263). Blood 2013; 121 (08) 1479-1481
  • 76 Kamel S, Horton L, Ysebaert L. et al. Ibrutinib inhibits collagen-mediated but not ADP-mediated platelet aggregation. Leukemia 2015; 29 (04) 783-787
  • 77 Thomas S, Krishnan A. Platelet heterogeneity in myeloproliferative neoplasms. Arterioscler Thromb Vasc Biol 2021; 41 (11) 2661-2670
  • 78 Kaplan ZS, Zarpellon A, Alwis I. et al. Thrombin-dependent intravascular leukocyte trafficking regulated by fibrin and the platelet receptors GPIb and PAR4. Nat Commun 2015; 6: 7835
  • 79 Mammadova-Bach E, Ollivier V, Loyau S. et al. Platelet glycoprotein VI binds to polymerized fibrin and promotes thrombin generation. Blood 2015; 126 (05) 683-691
  • 80 Dumas JJ, Kumar R, Seehra J, Somers WS, Mosyak L. Crystal structure of the GpIbalpha-thrombin complex essential for platelet aggregation. Science 2003; 301 (5630): 222-226
  • 81 Byzova TV, Plow EF. Networking in the hemostatic system. Integrin alphaiibbeta3 binds prothrombin and influences its activation. J Biol Chem 1997; 272 (43) 27183-27188
  • 82 Haider S, Latif T, Hochhausler A, Lucas F, Abdel Karim N. Waldenstrom's macroglobulinemia and peripheral neuropathy, organomegaly, endocrinopathy, monoclonal gammopathy, and skin changes with a bleeding diathesis and rash. Case Rep Oncol Med 2013; 2013: 890864
  • 83 Zangari M, Elice F, Tricot G, Fink L. Bleeding disorders associated with cancer dysproteinemias. Cancer Treat Res 2009; 148: 295-304
  • 84 Luu S, Gardiner EE, Andrews RK. Bone marrow defects and platelet function: a focus on MDS and CLL. Cancers (Basel) 2018; 10 (05) 147
  • 85 Castillo JJ, Advani RH, Branagan AR. et al. Consensus treatment recommendations from the tenth International Workshop for Waldenström Macroglobulinaemia. Lancet Haematol 2020; 7 (11) e827-e837
  • 86 Salles G, Barrett M, Foà R. et al. Rituximab in B-cell hematologic malignancies: a review of 20 years of clinical experience. Adv Ther 2017; 34 (10) 2232-2273
  • 87 Ghobrial IM, Fonseca R, Greipp PR. et al; Eastern Cooperative Oncology Group. 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
  • 88 Ram R, Bonstein L, Gafter-Gvili A, Ben-Bassat I, Shpilberg O, Raanani P. Rituximab-associated acute thrombocytopenia: an under-diagnosed phenomenon. Am J Hematol 2009; 84 (04) 247-250
  • 89 Cheson BD, Leoni L. Bendamustine: mechanism of action and clinical data. Clin Adv Hematol Oncol 2011; 9 (08, Suppl 19): 1-11
  • 90 Robak E, Robak T. Bruton's kinase inhibitors for the treatment of immunological diseases: current status and perspectives. J Clin Med 2022; 11 (10) 11
  • 91 von Hundelshausen P, Siess W. Bleeding by Bruton tyrosine kinase-inhibitors: dependency on drug type and disease. Cancers (Basel) 2021; 13 (05) 13
  • 92 Liu J, Fitzgerald ME, Berndt MC, Jackson CW, Gartner TK. Bruton tyrosine kinase is essential for botrocetin/VWF-induced signaling and GPIb-dependent thrombus formation in vivo. Blood 2006; 108 (08) 2596-2603
  • 93 Li Z, Delaney MK, O'Brien KA, Du X. Signaling during platelet adhesion and activation. Arterioscler Thromb Vasc Biol 2010; 30 (12) 2341-2349
  • 94 Atkinson BT, Ellmeier W, Watson SP. Tec regulates platelet activation by GPVI in the absence of Btk. Blood 2003; 102 (10) 3592-3599
  • 95 Burger JA, Buggy JJ. Bruton tyrosine kinase inhibitor ibrutinib (PCI-32765). Leuk Lymphoma 2013; 54 (11) 2385-2391
  • 96 Shatzel JJ, Olson SR, Tao DL, McCarty OJT, Danilov AV, DeLoughery TG. Ibrutinib-associated bleeding: pathogenesis, management and risk reduction strategies. J Thromb Haemost 2017; 15 (05) 835-847
  • 97 Levade M, David E, Garcia C. et al. Ibrutinib treatment affects collagen and von Willebrand factor-dependent platelet functions. Blood 2014; 124 (26) 3991-3995
  • 98 Bye AP, Unsworth AJ, Desborough MJ. et al. Severe platelet dysfunction in NHL patients receiving ibrutinib is absent in patients receiving acalabrutinib. Blood Adv 2017; 1 (26) 2610-2623
  • 99 Tullemans BME, Heemskerk JWM, Kuijpers MJE. Acquired platelet antagonism: off-target antiplatelet effects of malignancy treatment with tyrosine kinase inhibitors. J Thromb Haemost 2018; 16 (09) 1686-1699
  • 100 Dobie G, Kuriri FA, Omar MMA. et al. Ibrutinib, but not zanubrutinib, induces platelet receptor shedding of GPIb-IX-V complex and integrin αIIbβ3 in mice and humans. Blood Adv 2019; 3 (24) 4298-4311
  • 101 Brown JR, Moslehi J, Ewer MS. et al. Incidence of and risk factors for major haemorrhage in patients treated with ibrutinib: an integrated analysis. Br J Haematol 2019; 184 (04) 558-569
  • 102 Castillo JJ, Gustine JN, Meid K, Dubeau T, Severns P, Treon SP. Ibrutinib withdrawal symptoms in patients with Waldenström macroglobulinemia. Haematologica 2018; 103 (07) e307-e310
  • 103 Gustine JN, Meid K, Dubeau T. et al. Ibrutinib discontinuation in Waldenström macroglobulinemia: etiologies, outcomes, and IgM rebound. Am J Hematol 2018; 93 (04) 511-517
  • 104 Gustine JN, Meid K, Dubeau TE, Treon SP, Castillo JJ. Atrial fibrillation associated with ibrutinib in Waldenström macroglobulinemia. Am J Hematol 2016; 91 (06) E312-E313
  • 105 Ali N, Malik F, Jafri SIM, Naglak M, Sundermeyer M, Pickens PV. Analysis of efficacy and tolerability of Bruton tyrosine kinase inhibitor ibrutinib in various B-cell malignancies in the general community: a single-center experience. Clin Lymphoma Myeloma Leuk 2017; 17S: S53-S61
  • 106 Dimopoulos MA, Tedeschi A, Trotman J. et al; iNNOVATE Study Group and the European Consortium for Waldenström's Macroglobulinemia. Phase 3 trial of ibrutinib plus rituximab in Waldenström's macroglobulinemia. N Engl J Med 2018; 378 (25) 2399-2410
  • 107 Dimopoulos MA, Trotman J, Tedeschi A. et al; iNNOVATE Study Group and the European Consortium for Waldenström's Macroglobulinemia. Ibrutinib for patients with rituximab-refractory Waldenström's macroglobulinaemia (iNNOVATE): an open-label substudy of an international, multicentre, phase 3 trial. Lancet Oncol 2017; 18 (02) 241-250
  • 108 Treon SP, Gustine J, Meid K. et al. Ibrutinib monotherapy in symptomatic, treatment-naive patients with Waldenstrom macroglobulinemia. J Clin Oncol 2018; 36 (27) 2755-2761
  • 109 Fradley MG, Gliksman M, Emole J. et al. Rates and risk of atrial arrhythmias in patients treated with ibrutinib compared with cytotoxic chemotherapy. Am J Cardiol 2019; 124 (04) 539-544
  • 110 Abeykoon JP, Zanwar S, Ansell SM. et al. Ibrutinib monotherapy outside of clinical trial setting in Waldenström macroglobulinaemia: practice patterns, toxicities and outcomes. Br J Haematol 2020; 188 (03) 394-403
  • 111 Favaloro EJ, Funk DM, Lippi G. Pre-analytical variables in coagulation testing associated with diagnostic errors in hemostasis. Lab Med 2012; 43: 1-10
  • 112 Sharma R, Haberichter SL. New advances in the diagnosis of von Willebrand disease. Hematology (Am Soc Hematol Educ Program) 2019; 2019 (01) 596-600
  • 113 Favaloro EJ, Oliver S, Mohammed S, Vong R. Comparative assessment of von Willebrand factor multimers vs activity for von Willebrand disease using modern contemporary methodologies. Haemophilia 2020; 26 (03) 503-512
  • 114 Laporte P, Tuffigo M, Ryman A. et al. HemosIL VWF:GPIbR assay has a greater sensitivity than VWF:RCo technique to detect acquired von Willebrand syndrome in myeloproliferative neoplasms. Thromb Haemost 2022; 122 (10) 1673-1682
  • 115 Lim HY, Donnan G, Nandurkar H, Ho P. Global coagulation assays in hypercoagulable states. J Thromb Thrombolysis 2022; 54 (01) 132-144
  • 116 Ninivaggi M, de Laat-Kremers R, Tripodi A. et al. Recommendations for the measurement of thrombin generation: communication from the ISTH SSC Subcommittee on Lupus Anticoagulant/Antiphospholipid Antibodies. J Thromb Haemost 2021; 19 (05) 1372-1378
  • 117 de Breet CPDM, Zwaveling S, Vries MJA. et al. Thrombin generation as a method to identify the risk of bleeding in high clinical-risk patients using dual antiplatelet therapy. Front Cardiovasc Med 2021; 8: 679934
  • 118 Beltrán-Miranda CP, Khan A, Jaloma-Cruz AR, Laffan MA. Thrombin generation and phenotypic correlation in haemophilia A. Haemophilia 2005; 11 (04) 326-334
  • 119 Tripodi A, Martinelli I, Chantarangkul V, Battaglioli T, Clerici M, Mannucci PM. The endogenous thrombin potential and the risk of venous thromboembolism. Thromb Res 2007; 121 (03) 353-359
  • 120 Wan J, Konings J, de Laat B, Hackeng TM, Roest M. Added value of blood cells in thrombin generation testing. Thromb Haemost 2021; 121 (12) 1574-1587
  • 121 Favaloro EJ, Bonar R. An update on quality control for the PFA-100/PFA-200. Platelets 2018; 29 (06) 622-627
  • 122 Vinholt PJ. The role of platelets in bleeding in patients with thrombocytopenia and hematological disease. Clin Chem Lab Med 2019; 57 (12) 1808-1817
  • 123 Paniccia R, Priora R, Liotta AA, Abbate R. Platelet function tests: a comparative review. Vasc Health Risk Manag 2015; 11: 133-148
  • 124 Moenen FCJI, Vries MJA, Nelemans PJ. et al. Screening for platelet function disorders with Multiplate and platelet function analyzer. Platelets 2019; 30 (01) 81-87
  • 125 Walsh M, Kwaan H, McCauley R. et al. Viscoelastic testing in oncology patients (including for the diagnosis of fibrinolysis): review of existing evidence, technology comparison, and clinical utility. Transfusion 2020; 60 (Suppl. 06) S86-S100
  • 126 Kay AB, Morris DS, Collingridge DS, Majercik S. Platelet dysfunction on thromboelastogram is associated with severity of blunt traumatic brain injury. Am J Surg 2019; 218 (06) 1134-1137
  • 127 Al-Tamimi M, Arthur JF, Gardiner E, Andrews RK. Focusing on plasma glycoprotein VI. Thromb Haemost 2012; 107 (04) 648-655
  • 128 Lui M, Gardiner EE, Arthur JF. et al. Novel stenotic microchannels to study thrombus formation in shear gradients: influence of shear forces and human platelet-related factors. Int J Mol Sci 2019; 20 (12) 20
  • 129 Mangin PH, Gardiner EE, Nesbitt WS. et al; Subcommittee on Biorheology. In vitro flow based systems to study platelet function and thrombus formation: recommendations for standardization: Communication from the SSC on Biorheology of the ISTH. J Thromb Haemost 2020; 18 (03) 748-752
  • 130 de Witt SM, Swieringa F, Cavill R. et al. Identification of platelet function defects by multi-parameter assessment of thrombus formation. Nat Commun 2014; 5: 4257
  • 131 Burkhart JM, Vaudel M, Gambaryan S. et al. The first comprehensive and quantitative analysis of human platelet protein composition allows the comparative analysis of structural and functional pathways. Blood 2012; 120 (15) e73-e82
  • 132 Chatterjee M, Rath D, Gawaz M. Role of chemokine receptors CXCR4 and CXCR7 for platelet function. Biochem Soc Trans 2015; 43 (04) 720-726
  • 133 Hivert B, Caron C, Petit S. et al. Clinical and prognostic implications of low or high level of von Willebrand factor in patients with Waldenstrom macroglobulinemia. Blood 2012; 120 (16) 3214-3221
  • 134 Gavriatopoulou M, Terpos E, Ntanasis-Stathopoulos I. et al. Elevated vWF antigen serum levels are associated with poor prognosis, and decreased circulating ADAMTS-13 antigen levels are associated with increased IgM levels and features of WM but not increased vWF levels in patients with symptomatic WM. Clin Lymphoma Myeloma Leuk 2019; 19 (01) 23-28
  • 135 Stockschlaeder M, Schneppenheim R, Budde U. Update on von Willebrand factor multimers: focus on high-molecular-weight multimers and their role in hemostasis. Blood Coagul Fibrinolysis 2014; 25 (03) 206-216
  • 136 Shahani T, Covens K, Lavend'homme R. et al. Human liver sinusoidal endothelial cells but not hepatocytes contain factor VIII. J Thromb Haemost 2014; 12 (01) 36-42
  • 137 Federici AB. The factor VIII/von Willebrand factor complex: basic and clinical issues. Haematologica 2003; 88 (06) EREP02
  • 138 Saraya AK, Kasturi J, Kishan R. A study of haemostasis in macroglobulinaemia. Acta Haematol 1972; 47: 33-42
  • 139 Kasturi J, Saraya AK. Platelet functions in dysproteinaemia. Acta Haematol 1978; 59 (02) 104-113
  • 140 Camera M, Brambilla M, Toschi V, Tremoli E. Tissue factor expression on platelets is a dynamic event. Blood 2010; 116 (23) 5076-5077
  • 141 Siddiqui FA, Desai H, Amirkhosravi A, Amaya M, Francis JL. The presence and release of tissue factor from human platelets. Platelets 2002; 13 (04) 247-253
  • 142 Estupiñán HY, Berglöf A, Zain R, Smith CIE. Comparative analysis of BTK inhibitors and mechanisms underlying adverse effects. Front Cell Dev Biol 2021; 9: 630942
  • 143 Kaptein A, de Bruin G, Emmelot-van Hoek M. et al. Potency and selectivity of BTK inhibitors in clinical development for B-cell malignancies. Blood 2018; 132: 1871-1871
  • 144 Brown JR. Ibrutinib in chronic lymphocytic leukemia and B cell malignancies. Leuk Lymphoma 2014; 55 (02) 263-269
  • 145 Perkins HA, MacKenzie MR, Fudenberg HH. Hemostatic defects in dysproteinemias. Blood 1970; 35 (05) 695-707
  • 146 Merlini G, Baldini L, Broglia C. et al. Prognostic factors in symptomatic Waldenstrom's macroglobulinemia. Semin Oncol 2003; 30 (02) 211-215

Zoom Image
Fig. 1 Pathways of development of lymphoplasmacytoid B cells in healthy individuals and in WM patients. In WM patients, mutations and loss of DNA methylation occur to the lymphoplasmacytoid cells, with enrichment in B memory cells at an earlier stage of differentiation. The most common mutation, MYD88L265P, results in increased BTK phosphorylation and faster MYD88 complexing with IRAK1/4 in response to low or absent TLR or IL-1R stimulus, which results in increased NF-κB translocation to the nucleus, increased target gene transcription, uncontrolled proliferation of WM lymphoplasmacytoid cells and overproduction of IgM. BTK, Bruton's tyrosine kinase; IgM, immunoglobulin M; IL-1R, interleukin-1 receptor; IRAK, IL-1 receptor-associated kinase; MYD88, myeloid differentiation primary response 88; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; TLR, toll-like receptor; WM, Waldenström macroglobulinemia.
Zoom Image
Fig. 2 Simplified hemostasis. (A) Platelet plug formation. Following an injury, GPIb-IX-V and GPVI bind exposed extracellular matrix ligands such as von Willebrand factor (vWF) and collagen respectively to enable platelet adhesion to the endothelium. Engagement of these receptors triggers platelet signaling, and platelets undergo changes to the cytoskeleton and release granules. The platelet-specific integrin, αIIbβ3, becomes active and binds fibrinogen, bridging adjacent platelets. Platelets aggregate forming a platelet plug at the site of the injury. (B) Blood coagulation and the securing of the platelet plug. Tissue factor (TF) is exposed at the site of the injury triggering the extrinsic coagulation cascade, resulting in thrombin production. Thrombin converts fibrinogen into insoluble fibrin which secures the platelet plug in place. Blood hyperviscosity and high levels of IgM are likely to interfere with these hemostatic pathways, and potentially underpin bleeding events. GPVI, glycoprotein VI; IgM, immunoglobulin M.
Zoom Image
Fig. 3 Platelets and B lymphocytes utilize common signaling pathways. BTK is differentially involved in the downstream signaling pathways triggered by ligand engagement of the major platelet adhesion/signaling receptors (αIIbβ3, GPVI, and GPIbα of the GPIb-IX-V complex) and the BCR. In receptors with ITAMs, following ligand binding and receptor clustering, phosphorylation of the cytoplasmic ITAMs and recruitment of SFKs ensue, resulting in phosphorylation of Syk and activation of PI3K. PI3K mediates the conversion of phosphatidylinositol 4,5-bisphosphate to phosphatidylinositol 3,4,5-triphosphate, which engages BTK, resulting in phosphorylation of PLCγ. In platelets, this leads to activation and aggregation. In B cells, activation of MAPKs, NF-κB, and NFAT leads to B cell proliferation, development, and survival. BCR and GPVI ITAM signaling are more reliant on the BTK pathway compared with GPIbα and αIIbβ3, thus signaling downstream of these receptors is more sensitive to BTK inhibition. BCR, B cell receptor; BTK, Bruton's tyrosine kinase; GP, glycoprotein; ITAM, immunoreceptor tyrosine-based activation motif; MAPK, mitogen-activated protein kinase; NF-κB, nuclear factor κB; NFAT, nuclear factor of activated T cells ; PI3K, phosphoinositide 3-kinase; PLCγ, phospholipase C γ; SFK, Src-family kinases.
Zoom Image
Fig. 4 Recommended pathway to evaluate bleeding phenotypes in Waldenström macroglobulinemia, with treatment options. Blue: asymptomatic WM; red: symptomatic WM; green: therapy recommendations[1] (Castillo et al 2019). ADAM10, a disintegrin and metalloproteinase 10; Ag, antigen; aPTT, activated partial thromboplastin time; BM, bone marrow; CBA, collagen binding assay; DOAC, direct oral anticoagulant; FBC, full blood count; FVIII, factor VIII; IgM, immunoglobulin M; LTA, light transmission aggregometry; PFA, platelet function analyzer; PT, prothrombin time; RBC, red blood cell; R:Co: ristocetin cofactor assay; ROTEM, rotational thromboelastometry; SE, side effect; sGPVI, soluble glycoprotein VI; TEG, thromboelastography; TKI, tyrosine kinase inhibitor; VWD, von Willebrand disease; VWF, von Willebrand factor.