You are on page 1of 5

S58

Thrombosis Research 191S1 (2020) S58–S62

Contents lists available at ScienceDirect

Thrombosis Research
j o u r n a l h o m e p a g e : w w w. e l s e v i e r . c o m / l o c a t e / t h r o m r e s

Thrombosis in hematological malignancies: mechanisms and implications


Netanel A. Horowitza,b,*, Benjamin Brennera,b
a
Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
b
Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel

ARTICLE INFO ABSTRACT

Keywords: Thrombotic events are a major cause of morbidity and mortality in cancer. While the association of venous
Hematological malignancies thromboembolic events with cancer is well documented, in recent years arterial events (i.e. acute myocardial
Venous thromboembolism infarction and ischemic strokes) have also emerged as relatively common complications among cancer patients.
Arterial thrombotic events
In hematological malignancies incorporating a heterogeneous group of diseases, the prediction of thrombosis
Risk assessment
Thrombocytopenia
occurrence and/or recurrence is challenging, due to unique disease characteristics. Furthermore, the treatment
of thrombosis in these patients is often complicated because of disease- or therapy-related thrombocytopenia. In
addition, patients with hematological cancers are poorly represented in randomized control clinical trials; hence,
evidence-based guidelines are limited. This review will discuss the incidence of venous and arterial thrombotic
events in common myeloid and lymphoproliferative diseases. Several new mechanisms contributing to cancer-
associated thrombosis will be elaborated. The complicated issue of risk assessment and management of venous
thrombosis in patients with hematological malignancies will be delineated.

Highlights controlled clinical trials, at least in some hematological malignancies,


challenging. Recently, a large Danish population-based cohort study
• Arterial and venous thrombotic events are common in assessed the risks of myocardial infarction, ischemic stroke (IS),
hematological malignancies. venous thromboembolism (VTE), and bleeding (requiring hospital
• Clonal hematopoiesis of indeterminate potential can promote contact) in patients with hematological cancers [1]. Among 32,141
arterial thrombosis. evaluated patients (2000–2013), the 10‐year absolute risk of any
• Risk assessment tools for thrombosis and bleeding in these thromboembolic or bleeding events following hematological cancers
patients are limited. was 19%, with the percentage for myocardial infarction, IS, VTE and
• Prospective trials are needed to define anticoagulation strategies bleeding amounting to 3.3%, 3.5%, 5.2%, and 8.5%, respectively. Of
in this setting. note, the risk of thromboembolic events overcame that of bleeding in
all the patients, except for those with acute myeloid leukemia (AML),
1. Introduction acute lymphoid leukemia (ALL), or myelodysplastic syndrome (MDS).
Overall, patients with hematological cancer were at increased risk
Thrombosis is a leading cause of morbidity and mortality in cancer for acute myocardial infarction (AMI) (hazard ratio (HR) 1.36, 95%
patients. During recent years, efforts have been made to identify among confidence interval (CI) 1.25–1.49), IS (HR 1.22, 95% CI 1.12–1.33),
patients with cancer those who are at high risk of developing venous VTE (HR 3.37, 95% CI 3.13–3.64), and bleeding (HR 2.39, 95% CI
and arterial thrombotic events. Clinical trials have been conducted to 2.26–2.53), relative to the general population [1]. The findings of this
evaluate the efficacy and safety of thrombo-prophylaxis and treatment large, well-conducted epidemiological study emphasize the magnitude
in this high-risk population. However, data regarding the epidemiology of the problem and highlight the need for trials targeting these clinical
of thrombotic events in patients with hematological malignancies and, settings. The present review will address the issue of thrombotic risk
particularly, their clinical management are limited, as such patients are (arterial and venous) in hematological malignancies, focusing on the
under-represented in clinical trials. In addition, disease- or treatment- diseases with limited available data. In addition, pathophysiological
related thrombocytopenia increases the risk of bleeding associated mechanisms contributing to VTE occurrence, approaches to VTE
with anticoagulation, which renders the performance of large-scale management, and most challenging clinical scenarios will be discussed.

* Corresponding author at: Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, 8, Ha’Aliya Street, Haifa 3109601,
Israel.
E-mail address: n_horowitz@rambam.health.gov.il (N.A. Horowitz).

Received 2 October 2019; Received in revised form 12 January 2020; Accepted 13 January 2020
This article was published as part of a supplement sponsored by the International Conference on Thrombosis and Hemostasis Issues in Cancer.
0049-3848/ © 2020 Published by Elsevier Ltd.
N.A. Horowitz and B. Brenner Thrombosis Research 191S1 (2020) S58–S62

2. Acute leukemia In the analysis of the US Veterans Affairs Hospital database, the crude
incidence of deep vein thrombosis in more than 6,000 MM patients
In a population-based cohort study from California (1993–1999), was about 6% at an 8-year follow-up, which was higher than among
including 5,934 AML patients, the 2-year cumulative VTE incidence was patients without MM [8].
5.2%. Sixty-four percent of events occurred within 3 months of AML The use of immunomodulatory drugs was shown to increase plasma
diagnosis. Of note, the VTE diagnosis was not associated with reduced levels of factor VIII, von Willebrand factor and protein C resistance,
survival at 1 year. The same study also included 2,482 ALL patients. and to reduce thrombomodulin concentration [9]. In a meta-analysis
The 2-year VTE incidence was 4.5% and VTE was associated with a of MM patients treated with immunomodulatory drugs, those with
40% increase in the risk of death at 1 year. Overall, the incidence of newly diagnosed MM receiving thalidomide alone or in combination
VTE among patients with acute leukemia was similar to that observed with dexamethasone without thrombo-prophylaxis, had a 3-month
in patients with solid tumors [2]. In the Danish study, the 10-year VTE risk of around 4% and 12%, respectively. Likewise, patients with
incidence of VTE was 12.4 % among AML patients, with an HR of 4.49 newly diagnosed MM receiving lenalidomide in combination with
(6.14–6.42), while among ALL patients the incidence was 11.24%, dexamethasone had a 3-month risk of VTE of about 3% [10]. Of note,
with an HR of 7.42 (3.07–17.97) [1]. In a prospective evaluation of the proteasome inhibitor bortezomib was not found to increase the
the incidence, risk factors and outcomes of thromboembolism (TE) risk of VTE or arterial TE [11,12]. The thrombo-protective effect of
in 1038 children with ALL, TE occurred in 6.1% of children, with bortezomib was suggested to be mediated by an increased expression
the highest incidence (20.5%) observed those aged 15–17 years. The of the transcription factor KLF2 [13].
TE-associated case fatality rate of 6.4% indicates that TE is a severe VTE risk assessment should be performed in all MM patients
complication of ALL treatment [3]. treated with thalidomide or lenalidomide [14]. Recently, two new
risk assessment models have been reported [15,16]. The International
3. Lymphoma Myeloma Working Group (IMWG) recommends aspirin prophylaxis for
patients with no or one VTE risk factor and low molecular weight
The rate of arterial and venous thrombotic events in lymphoma heparin (LMWH) or therapeutic-dose warfarin if two or more risk
patients was estimated in a meta-analysis that included 29 cohort factors are present. The same approach is proposed for those receiving
studies and 18,018 patients. Among 1,149 recorded events 83.5% high‐dose dexamethasone, doxorubicin or multi‐agent chemotherapy,
were venous; the majority of them occurred prior to or within the first irrespective of preceding risk factors [14]. The 2016 National Institute
3 months of chemotherapy. The global incidence ratio of thrombosis of Clinical Excellence (NICE) guidelines reiterate this recommendation,
was 6.4%, while patients with non-Hodgkin lymphoma (NHL) had while acknowledging the relative lack of evidence on the subject.
a significantly higher ratio than Hodgkin lymphoma (HL) patients However, the most recent NICE guidance [17] suggests that either
(6.5% versus 4.7%, respectively). Among NHL patients, individuals aspirin or prophylactic dose LMWH may be considered equivalent
with aggressive disease (i.e., diffuse large B cell lymphoma and Burkitt for myeloma patients receiving chemotherapy in combination with
lymphoma) had a greater risk of VTE events than those with indolent immunomodulatory drugs and a corticosteroid.
disease (8.3% versus 6.3%, respectively). A higher risk for VTE in
aggressive NHL was also demonstrated in other studies [4,5]. In the 6. Myeloproliferative neoplasms
Danish population-based cohort study the incidence rate and 10-year
HR for VTE were 5.93 and 4.19 in the HL cohort and 11.84 and 3.79 In this group of hematological disorders, polycythemia vera
in the NHL cohort. It is of interest that no association with myocardial and essential thrombocythemia are associated with increased risk
infarction or IS was found in in patients with aggressive or indolent of thrombosis particularly, with arterial events (e.g., stroke, AMI,
NHL as well as in patients with HL [1]. peripheral arterial occlusive disease) accounting for 60–70% of all
thrombotic events [18–21]. Of interest, splanchnic vein thrombosis
4. Chronic lymphocytic leukemia is a unique subtype of VTE commonly found in myeloproliferative
neoplasms (MPNs). In a large study of 1,062 patients with Budd-
A recent retrospective analysis demonstrated the VTE incidence of Chiari syndrome and 855 patients with portal vein thrombosis, the
11% in a group of 346 patients with chronic lymphocytic leukemia prevalence of MPNs was 40.9% and 31.5%, respectively [22]. Along
(CLL), at a median follow-up of 6 years [6]. In the Danish study, the with age and previous thrombotic events, the high cellular counts and,
10-year incidence rate for thromboembolic complications in CLL moreover, the procoagulant phenotypes of blood cells attributed to
was 7.94% (relative to 6.07 in the comparison cohort) with an HR genetic alterations in these malignancies have been reported as the
of 2.18 (1.80–2.63), whereas the corresponding values for AMI and major determinants of thrombosis occurrence [23]. Guidelines for
IS were 7.68 and 6.20% (HR 1.31 (1.09–1.57) and 0.95 (0.78–1.15), prophylaxis and treatment of arterial and thrombotic events have been
respectively) [1]. These data imply that it is the rate of VTE and not of published and extensively reviewed, but are beyond the scope of this
arterial events (at least IS) that is increased among CLL patients, which review.
could be attributed to the older age of these patients. Of importance,
performance status, poor CLL prognostics factors, concurrent 7. Mechanisms
malignancies, and inherited thrombophilia have all been found to be
associated with VTE occurrence [6]. The three components of the Virchow’s Triad are most pertinent
to cancer patients. The endothelial wall is shifting toward the pro-
5. Multiple myeloma coagulant phenotype and tumor cells invade the blood vessel wall.
Stasis of blood is increased due to frequent immobilization and surgery
In the aforementioned Danish study, the incidence rate for VTE in and blood vessels are often compressed by tumor masses. The third
multiple myeloma (MM) was 18% and a 10-year HR of 5.53 (4.51– component of this triad is of exceptional relevance to hematological
6.77) relative to 4.30 (3.92–4.71) in a comparison cohort of matched malignancies. Thus, leukocytosis, thrombocytosis and elevated hemato-
individuals from the general population. It is of interest that the crit are major contributors to the thrombotic risk in MPNs.
incidence rates for AMI and IS were also high amounting to 9.77% and The complexity of the underlying mechanisms has been reflected
7.4, respectively with a HR of 1.87 (1.50–2.33) and 1.3 (1.02–1.65) in a broad variety of suggested pathways. Tissue factor (TF) is known
relative to the comparison group (6.07 and 6.83, respectively). Higher to have a crucial role in promoting coagulation and angiogenesis
rates of arterial TE were also demonstrated by other study groups [7]. in cancer [24]. In hematological malignancies, AML, MM and

S59
N.A. Horowitz and B. Brenner Thrombosis Research 191S1 (2020) S58–S62

other tumor cells have been found to bear TF and low tissue factor including major ones. When included as an individual covariate along
pathway inhibitor (TFPI) on their membranes and as a result have a with components of the HAS‐BLED score, cancer has emerged as the
procoagulant phenotype [25]. In recent years, the role of extracellular strongest predictor of major and overall bleeding.
vesicles (EVs) in cancer-associated thrombosis (CAT) has emerged. The Khorana score (KRS) has been developed to identify ambulatory
EVs are lipid bilayer-delimited particles that are naturally released cancer patients receiving chemotherapy who have a high risk for
from a variety of cells, including tumor cells. They carry a cargo of VTE [38]. However, its validity for patients with hematological
proteins, nucleic acids, lipids, metabolites, and even organelles from malignancies requires further confirmation. In an attempt to validate
the parent cell. In the cancer setting, tumor-derived EVs have been the KRS applicability in lymphoma, Rupa-Matysek and colleagues
shown to circulate in vivo and express procoagulant molecules (i.e., TF assessed 428 HL and diffuse large B-cell lymphoma (DLBCL) patients.
and phospholipids). EVs induce platelet aggregation through PSGL-1 The score failed to discriminate between high- and intermediate-risk
[26] and exert a procoagulant phenotype on endothelial cells exposed patients with respect to VTE occurrence [44]. In a recent analysis
to EVs derived from AML patients [27]. Furthermore, in EVs obtained of 12 clinical trials conducted by the Italian Lymphoma Foundation
from MM cells, the TF expression is enhanced relative to TFPI and (FIL), 40% of patients were characterized as being at ‘high risk’ for
thrombomodulin expression [28]. VTE events. However, the actual rate of VTE was only 2.9% in these
Heparanase is an endo-β-D-glucuronidase which cleaves heparan patients. Of interest, in this study, DLBCL was found to be associated
sulfate chains on cell surfaces and in the extracellular matrix. Hepara- with a higher risk for VTE [45]. Overall, these findings suggest that
nase has been demonstrated to enhance angiogenesis, inflammation the KRS might not provide an accurate estimation of the VTE risk in
and cancer progression [29]. Under physiological conditions, hepara- all lymphoma subtypes and could thus be recommended to be used
nase levels are highest in the placenta, platelets, neutrophils and for patients with more aggressive lymphoma histology only. Another
monocytes. Thrombin is a selective inducer of heparanase release predictive model, specific for lymphoma patients (the ThroLy score),
from platelets and granulocytes via protease-activated receptor-1 was proposed by Antic et al. [46]. The study population included 1,820
[30]. In MPNs, heparanase staining has appeared to be prominent in lymphoma patients (a derivation cohort, n=1,236, and a validation
the bone marrow of ET JAK2-positive patients relative to CML and cohort, n=584). The variables independently associated with risk for
JAK2-negative patients. Furthermore, JAK2 is involved in heparanase VTE were: previous venous/arterial events, mediastinal involvement,
upregulation via the erythropoietin receptor and heparanase levels body mass index >30 kg/m2, reduced mobility, extranodal localization,
and procoagulant activity is reported to be increased by erythropoietin neutropenia and a hemoglobin level <100 g/L. A high-risk score (i.e.,
and decreased by JAK2 inhibitors [31]. >3) had a positive predictive value of 65.2%. The authors conclude
In recent years, the term ‘clonal hematopoiesis of indeterminate that the ThroLy score is more specific for lymphoma patients than
potential’ (CHIP) has been introduced to describe individuals who any other available score targeting thrombosis in cancer patients.
acquire somatic mutations in hematopoietic stem cells in the bone While it looks promising, the ThroLy score needs to be validated in
marrow [32–34]. Only in the minority of patients (0.5–1% per year), other lymphoma cohorts prior to its wide implementation as a tool
these cells can lead to the generation of clones of mutated leukocytes for precise identification of high-risk patients who should be offered
that populate peripheral blood and ultimately develop acute leukemia. thrombo-prophylaxis.
However, CHIP increases the rate of cardiovascular events (AMI and
IS) by 40% [35]. The genes associated with CHIP are DNMT3A, TET2, 9. Management
ASXL1, PPM1D, JAK2, TP53, SF3B1 and SRSF2, genes that are often
implicated in the pathogenesis of myelodysplastic syndrome and Several clinical trials have established the role of LMWH in the
AML. Findings from mouse experiments suggest a direct mechanism treatment of CAT. However, the majority of these trials were conducted
by which a CHIP mutation can accelerate atherosclerosis [35]. For in patients with solid tumors. In the CLOT trial, dalteparin therapy
instance, mice bearing mutations in Tet2 in the presence of low-density was associated with a significant (52%) reduction in the risk for VTE
lipoprotein receptor deficiency (i.e., atherosclerosis susceptibility) recurrence relative to treatment with vitamin K antagonists (VKA)
have demonstrated accelerated lesion formation. Furthermore, cells [47] Moreover, no significant difference in the rate of major bleeding
with loss-of-function mutations in TET2 show enhanced expression between the two arms was observed. Similar results were demonstrated
of pro-inflammatory mediators involved in atherosclerosis, including in the CATCH study, where tinzaparin treatment resulted in a 35% risk
interleukin (IL)-1b and IL-6, when stimulated with a classic cardio- reduction in VTE recurrence compared to VKA therapy. Once again,
vascular risk factor [36]. All these data give rise to intriguing questions the rate of major bleeding was equal in both arms [48].
that will have to be addressed in the future. Should screening for CHIP The International Clinical Practice Guidelines recommend LMWH
be performed and, if so, in whom? How could the cardiovascular for the initial treatment of established VTE in patients with cancer.
risk in patients with CHIP be estimated? Moreover, what approaches Then, a once-daily regimen of LMWH is generally recommended [49]
to the management of the excessive cardiovascular risk could be After 3–6 months, termination or continuation of anticoagulation
recommended in the absence of clinical trials? should be based on individual assessment. The use of direct oral
anticoagulants (DOACs) in the CAT setting continues to be tested in
8. Risk assessment clinical trials. The Hokusai trial demonstrated the non-inferiority of
edoxaban to dalteparin for the composite end point of VTE recurrence
In general, patients with cancer should be evaluated for the risk and major bleeding [50]. Similar findings were reported in the
of VTE occurrence, or recurrence in case of existing CAT [37]. It is SELECT-D trial comparing rivaroxaban and dalteparin in a similar study
also imperative that if pharmacological anticoagulation or thrombo- design [51]. Of note, an increase in gastrointestinal (and potentially
prophylaxis is indicated, the risk of bleeding be assessed. While genitourinary) bleeding events was observed in the DOAC arms. The
several predictive models for CAT development have been proposed ADAM VTE trial compared the use of apixaban and dalteparin in active
[38–41], the models estimating the bleeding risk associated with cancer-associated VTE [52]. Oral apixaban was associated with low
anticoagulation in cancer patients are scarce. The HAS-BLED score is VTE recurrence rates (0.7% versus 6.3%, p=0.028), while there was
designed to determine the risk of major bleeding in atrial fibrillation no significant difference in the risk of major bleeding. In all the trials,
patients on anticoagulation [42]. Lately, the HAS‐BLED score has the number of patients with hematological cancers was low (10.5%,
been evaluated in a large cohort of 132,280 VTE patients during the 2.5% and 10%, respectively).
first 6 months of anticoagulation [43]. In the cancer cohort (20% of When considering the use of DOACS for the treatment of CAT, their
patients) the increase from 3 to 4 points was significant for all bleeds, potential interactions with anti-cancer medications should be taken

S60
N.A. Horowitz and B. Brenner Thrombosis Research 191S1 (2020) S58–S62

into account. For example, imatinib, crizotinib and other tyrosine Conflict of interest statement:
kinase inhibitors are known to inhibit CYP 3A4, and may therefore
reduce the effect of DOACS in the plasma. Vinblastine and doxorubicin The authors declare no conflicts of interest.
are known inducers of P-glycoprotein, and hence may also reduce
DOAC plasma levels and activity [53,54]. Data from other clinical References
trials (e.g., Caravaggio) are expected to solidify the evidence on the [1] K. Adelborg, P. Corraini, B. Darvalics, et al., Risk of thromboembolic and bleeding
efficacy and safety of these agents. outcomes following hematological cancers: a Danish population-based cohort study,
Recently, Soff et al. reported the experience of rivaroxaban treatment J. Thromb. Haemost. 17 (2019) 1305–1318.
in 1,072 CAT patients at the Memorial Sloan Kettering Cancer Center [2] G.H. Ku, R.H. White, H.K. Chew, et al., Venous thromboembolism in patients with
acute leukemia: incidence, risk factors, and effect on survival, Blood 113 (2009)
[55]. Again, just 8% of patients had hematologic malignancies. Six-
3911–3917.
month cumulative incidence of recurrent VTE and major bleeding [3] R. Tuckuviene, S. Ranta, B.K. Albertsen, et al., Prospective study of thromboem-
were 4.2% and 2.2%, respectively, with 73% of major bleeds occurring bolism in 1038 children with acute lymphoblastic leukemia: a Nordic Society of
in the gastrointestinal tract. Of note, the elderly (aged over 75 years) Pediatric Hematology and Oncology (NOPHO) study, J. Thromb. Haemost. 14
(2016) 485–494.
had similar rates of recurrent thrombosis and bleeding as those aged
[4] I.H. Borg, M.D. Bendtsen, M. Bogsted, et al., Incidence of venous thromboembo-
under 75 years. Since patients with hematological malignancies are lism in patients with diffuse large B-cell lymphoma, Leuk. Lymphoma 57 (2016)
under-represented in these studies, real-life information from large 2771–2776.
datasets is crucial for developing guidance on VTE management in [5] I.L. Gade, S. Braekkan, I.A. Naess, et al., Epidemiology of venous thromboembolism
in hematological cancers: the Scandinavian Thrombosis and Cancer (STAC) cohort,
this most challenging clinical setting.
Thromb. Res. 158 (2017) 157–160.
Thrombocytopenia is common in hematological patients as a result [6] M. Simkovic, P. Vodarek, M. Motyckova, et al., Venous thromboembolism in
of bone marrow infiltration by tumor cells, toxicity of chemotherapy patients with chronic lymphocytic leukemia, Thromb. Res. 136 (2015) 1082–1086.
or other targeted therapies, immune mediated, stem cell conditioning [7] A.K. Stewart, S.V. Rajkumar, M.A. Dimopoulos, et al., Carfilzomib, lenalidomide,
regimens or radiation. In a recent observational cohort study of and dexamethasone for relapsed multiple myeloma, N. Engl. J. Med. 372 (2015)
142–152.
patients experiencing VTE during periods of treatment-related [8] S.Y. Kristinsson, T.R. Fears, G. Gridley, et al., Deep vein thrombosis after monoclo-
thrombocytopenia, 67 patients received anticoagulation and 88% of nal gammopathy of undetermined significance and multiple myeloma, Blood 112
them were managed with platelet transfusion support for a goal of (2008) 3582–3586.
50×109/L. Still, 31 patients experienced bleeding events. The majority [9] F.W. Leebeek, Update of thrombosis in multiple myeloma, Thromb. Res. 140 Suppl
1 (2016) S76–S80.
of events were non-major/non-clinically relevant [56]. Similar
[10] M. Carrier, G. Le Gal, J. Tay, et al., Rates of venous thromboembolism in multi-
findings were reported in the study by Houghton et al. [57]. In this ple myeloma patients undergoing immunomodulatory therapy with thalidomide or
study, withholding anticoagulation was associated with a substantial lenalidomide: a systematic review and meta-analysis, J. Thromb. Haemost. 9 (2011)
‘trade off’ of recurrent VTE. 653–663.
[11] P. Richardson, R. Schlag, N. Khuageva, et al., Characterization of haematological
Recently, guidance from the SSC of the ISTH regarding the manage-
parameters with bortezomib-melphalan-prednisone versus melphalan-prednisone
ment of CAT patients with thrombocytopenia have been published in newly diagnosed myeloma, with evaluation of long-term outcomes and risk of
[58]. In thrombocytopenic (i.e., <50,000/μL) patients at high risk thromboembolic events with use of erythropoiesis-stimulating agents: analysis of
for thrombosis progression or recurrence (symptomatic segmental or the VISTA trial, Br. J. Haematol. 153 (2011) 212–221.
[12] W. Li, R.F. Cornell, D. Lenihan, et al., Cardiovascular complications of novel multi-
more proximal pulmonary embolism, proximal deep vein thrombosis,
ple myeloma treatments, Circulation 133 (2016) 908–912.
or a history of or recurrent/progressive thrombosis) full-dose anti- [13] L. Nayak, H. Shi, G.B. Atkins, et al., The thromboprotective effect of bortezomib
coagulation in conjunction with platelet transfusion should be is dependent on the transcription factor Kruppel-like factor 2 (KLF2), Blood 123
administered. For patients at lower risk, a dose‐modification strategy (2014) 3828–3831.
using 50% or prophylactic‐dose LMWH may be considered. In severe [14] A. Palumbo, S.V. Rajkumar, M.A. Dimopoulos, et al., Prevention of thalidomide-
and lenalidomide-associated thrombosis in myeloma, Leukemia 22 (2008) 414–423.
thrombocytopenia (i.e., <25,000/μL) anticoagulation should be [15] A. Li, Q. Wu, S. Luo, et al., Derivation and validation of a risk assessment model
withheld, although prophylactic doses might be reasonable in patients for immunomodulatory drug-associated thrombosis among patients with multiple
with a platelet count >10,000/μL. Inferior vena cava filter placement myeloma, J. Natl. Compr. Cancer Netw. 17 (2019) 840–847.
should be considered only in patients with absolute contraindications [16] K.M. Sanfilippo, S. Luo, T.F. Wang, et al., Predicting venous thromboembolism in
multiple myeloma: development and validation of the IMPEDE VTE score, Am. J.
to anticoagulation. LMWH is currently the preferred anticoagulant
Hematol. 94 (2019) 1176–1184.
among patients with CAT and thrombocytopenia, and DOACs are [17] Venous thromboembolism in over 16s: reducing the risk of hospital-acquired deep
not recommended. Prospective studies are warranted to identify the vein thrombosis or pulmonary embolism, Available at https://www.nice.org.uk/
optimal anticoagulation dose and transfusion strategy in this setting. guidance/ng89/chapter/Recommendations#interventions-for-people-with-cancer,
Accessed in September 2019.
[18] A. Tefferi, M. Elliott, Thrombosis in myeloproliferative disorders: prevalence, prog-
10. Conclusions nostic factors, and the role of leukocytes and JAK2V617F, Semin. Thromb. Hemost.
33 (2007) 313–320.
Large datasets and registries provide real-world evidence regarding [19] A. Falanga, M. Marchetti, Thrombosis in myeloproliferative neoplasms, Semin.
Thromb. Hemost. 40 (2014) 348–358.
the epidemiology of thrombosis in hematological malignancies.
[20] A. Falanga, M. Marchetti, Thrombotic disease in the myeloproliferative neoplasms,
Mechanisms of thrombosis in hematological malignancies are Hematology Am. Soc. Hematol. Educ. Program 2012 (2012) 571–581.
closely related to tumor biology. Personalized risk assessment of [21] M. Sekhar, Prevention and management of thrombosis in myeloproliferative neo-
thrombosis and bleeding is mandatory in patients with hematological plasms, Clin. Adv. Hematol. Oncol. 15 (2017) 178–181.
malignancies. Risk-assessment tools based on disease-, patient- and [22] M.A. Elliott, A. Tefferi, Thrombosis and haemorrhage in polycythaemia vera and
essential thrombocythaemia, Br. J. Haematol. 128 (2005) 275–290.
therapy-associated risk factors need to be developed. Clinical trials [23] T. Barbui, G. Finazzi, A. Falanga, Myeloproliferative neoplasms and thrombosis,
targeting thrombocytopenic patients with hematological malignancies Blood 122 (2013) 2176–2184.
should be designed. These studies should also explore the role of low- [24] F.R. Rickles, A. Falanga, Molecular basis for the relationship between thrombosis
intensity anticoagulation and DOACs in these complicated clinical and cancer, Thromb. Res. 102 (2001) V215–V224.
[25] Y. Nadir, T. Katz, G. Sarig, et al., Hemostatic balance on the surface of leuke-
scenarios.
mic cells: the role of tissue factor and urokinase plasminogen activator receptor,
Haematologica 90 (2005) 1549–1556.
[26] N.S. Key, P. Chantrathammachart, P.W. Moody, J.Y. Chang, Membrane microparti-
Funding cles in VTE and cancer, Thromb. Res. 125 Suppl 2 (2010) S80–S83.
[27] I. Tzoran, A. Rebibo-Sabbah, B. Brenner, A. Aharon, Disease dynamics in patients
None. with acute myeloid leukemia: new biomarkers, Exp. Hematol. 43 (2015) 936–943.

S61
N.A. Horowitz and B. Brenner Thrombosis Research 191S1 (2020) S58–S62

[28] M. Zarfati, I. Avivi, B. Brenner, et al., Extracellular vesicles of multiple myeloma [44] J. Rupa-Matysek, M. Lembicz, E.K. Rogowska, et al., Evaluation of risk factors and
cells utilize the proteasome inhibitor mechanism to moderate endothelial angiogen- assessment models for predicting venous thromboembolism in lung cancer patients,
esis, Angiogenesis 22 (2019) 185–196. Med. Oncol. 35 (2018) 63.
[29] Y. Nadir, B. Brenner, L. Fux, et al., Heparanase enhances the generation of activated [45] R.M. Santi, M. Ceccarelli, G. Catania, et al., PO-03 – Khorana score and histotype
factor X in the presence of tissue factor and activated factor VII, Haematologica 95 predict the incidence of early venous thromboembolism (VTE) in non-hodgkin lym-
(2010) 1927–1934. phoma (NHL). A pooled data analysis of twelve clinical trials of Fondazione Italiana
[30] M. Tatour, M. Shapira, E. Axelman, et al., Thrombin is a selective inducer of hep- Linfomi (FIL), Thromb. Res. 140 Suppl 1 (2016) S177.
aranase release from platelets and granulocytes via protease-activated receptor-1, [46] D. Antic, N. Milic, S. Nikolovski, et al., Development and validation of multivari-
Thromb. Haemost. 117 (2017) 1391–1401. able predictive model for thromboembolic events in lymphoma patients, Am. J.
[31] I. Kogan, D. Chap, R. Hoffman, et al., JAK-2 V617F mutation increases heparanase Hematol. 91 (2016) 1014–1019.
procoagulant activity, Thromb. Haemost. 115 (2016) 73–80. [47] A.Y. Lee, M.N. Levine, R.I. Baker, et al., Low-molecular-weight heparin versus a
[32] S. Jaiswal, P. Fontanillas, J. Flannick, et al., Age-related clonal hematopoiesis asso- coumarin for the prevention of recurrent venous thromboembolism in patients with
ciated with adverse outcomes, N. Engl. J. Med. 371 (2014) 2488–2498. cancer, N. Engl. J. Med. 349 (2003) 146–153.
[33] D.P. Steensma, R. Bejar, S. Jaiswal, et al., Clonal hematopoiesis of indeterminate [48] A.Y.Y. Lee, P.W. Kamphuisen, G. Meyer, et al., Tinzaparin vs warfarin for treatment
potential and its distinction from myelodysplastic syndromes, Blood 126 (2015) of acute venous thromboembolism in patients with active cancer: a randomized
9–16. clinical trial, JAMA 314 (2015) 677–686.
[34] L. Busque, M. Buscarlet, L. Mollica, R.L. Levine, Concise review: age-related clonal [49] D. Farge, C. Frere, J.M. Connors, et al., 2019 International clinical practice guide-
hematopoiesis: stem cells tempting the devil, Stem Cells 36 (2018) 1287–1294. lines for the treatment and prophylaxis of venous thromboembolism in patients with
[35] S. Jaiswal, P. Natarajan, A.J. Silver, et al., Clonal hematopoiesis and risk of athero- cancer, Lancet Oncol. 20 (2019) e566–e581.
sclerotic cardiovascular disease, N. Engl. J. Med. 377 (2017) 111–121. [50] G.E. Raskob, N. van Es, P. Verhamme, et al., Edoxaban for the treatment of cancer-
[36] J.J. Fuster, S. MacLauchlan, M.A. Zuriaga, et al., Clonal hematopoiesis associated associated venous thromboembolism, N. Engl. J. Med. 378 (2018) 615–624.
with TET2 deficiency accelerates atherosclerosis development in mice, Science 355 [51] A.M. Young, A. Marshall, J. Thirlwall, et al., Comparison of an oral factor Xa
(2017) 842–847. Inhibitor with low molecular weight heparin in patients with cancer with venous
[37] N.S. Key, A.A. Khorana, N.M. Kuderer, et al., Venous thromboembolism prophylaxis thromboembolism: results of a randomized trial (SELECT-D), J. Clin. Oncol. 36
and treatment in patients with cancer: ASCO clinical practice guideline update, J. (2018) 2017–2023.
Clin. Oncol. (2019) doi:10.1200/JCO.19.01461. [52] R.D. McBane 2nd, W.E. Wysokinski, J.G. Le-Rademacher, et al., Apixaban and
[38] A.A. Khorana, N.M. Kuderer, E. Culakova, et al., Development and validation of dalteparin in active malignancy-associated venous thromboembolism: the ADAM
a predictive model for chemotherapy-associated thrombosis, Blood 111 (2008) VTE trial, J. Thromb. Haemost. (2019) doi:10.1111/jth.14662.
4902–4907. [53] N.J. Short, J.M. Connors, New oral anticoagulants and the cancer patient, Oncologist
[39] R. Patell, L. Rybicki, K.R. McCrae, A.A. Khorana, Predicting risk of venous throm- 19 (2014) 82–93.
boembolism in hospitalized cancer patients: utility of a risk assessment tool, Am. J. [54] A.Y. Lee, E.A. Peterson, Treatment of cancer-associated thrombosis, Blood 122
Hematol. 92 (2017) 501–507. (2013) 2310–2317.
[40] N.M. Kuderer, M.S. Poniewierski, E. Culakova, et al., Predictors of venous thrombo- [55] G.A. Soff, J. Mones, C. Wilkins, et al., Rivaroxaban treatment of cancer-associated
embolism and early mortality in lung cancer: results from a global prospective study venous thromboembolism: Memorial Sloan Kettering Cancer Center institutional
(CANTARISK), Oncologist 23 (2018) 247–255. experience, Res. Pract. Thromb. Haemost. 3 (2019) 349–356.
[41] G.T. Gerotziafas, A. Taher, H. Abdel-Razeq, et al., A predictive score for throm- [56] B.T. Samuelson Bannow, R.B. Walter, T.B. Gernsheimer, D.A. Garcia, Patients
bosis associated with breast, colorectal, lung, or ovarian cancer: the prospective treated for acute VTE during periods of treatment-related thrombocytopenia have
COMPASS-Cancer-Associated Thrombosis Study, Oncologist 22 (2017) 1222–1231. high rates of recurrent thrombosis and transfusion-related adverse outcomes, J.
[42] R. Pisters, D.A. Lane, R. Nieuwlaat, et al., A novel user-friendly score (HAS-BLED) Thromb. Thrombolysis. 44 (2017) 442–447.
to assess 1-year risk of major bleeding in patients with atrial fibrillation: the Euro [57] D.E. Houghton, N.S. Key, N.A. Zakai, et al., Analysis of anticoagulation strategies for
Heart Survey, Chest 138 (2010) 1093–1100. venous thromboembolism during severe thrombocytopenia in patients with hemato-
[43] J.D. Brown, A.J. Goodin, G.Y.H. Lip, V.R. Adams, Risk stratification for bleeding logic malignancies: a retrospective cohort, Leuk. Lymphoma 58 (2017) 2573–2581.
complications in patients with venous thromboembolism: application of the HAS- [58] B.T. Samuelson Bannow, A. Lee, A.A. Khorana, et al., Management of cancer-asso-
BLED bleeding score during the first 6 months of anticoagulant treatment, J. Am. ciated thrombosis in patients with thrombocytopenia: guidance from the SSC of the
Heart. Assoc. 7 (2018) e007901. ISTH, J. Thromb. Haemost. 16 (2018) 1246–1249.

S62

You might also like