You are on page 1of 19

review

Venous thromboembolism in multiple myeloma – choice


of prophylaxis, role of direct oral anticoagulants and
special considerations

1
Dawn Swan, Alberto Rocci2,3 Charlotte Bradbury,4,5 and Jecko Thachil,2
1
Department of Haematology, University Hospital Galway, Galway, Republic of Ireland, 2Department of Haematology, Manchester
University Hospitals NHS Foundation Trust, Manchester, United Kingdom, 3Faculty of Biology, Medicine and Health, School of
Medical Science, Division of Cancer Science, University of Manchester, Manchester, United Kingdom, 4Cellular and Molecular
Medicine, University of Bristol, Bristol, United Kingdom and 5Bristol Haematology and Oncology Centre, University Hospitals
Bristol NHS Foundation Trust, Bristol, United Kingdom

et al, 2007; Schoen et al, 2018). Amongst haematological


Summary
malignancies, multiple myeloma confers an especially high
Multiple myeloma is associated with a significant risk of risk, with at least 10% of patients developing VTE during
venous thromboembolism (VTE), causing substantial levels their disease history (Barlogie et al, 1999; Eby, 2009; Falanga
of morbidity and mortality. The thrombogenicity of mye- & Marchetti, 2009; Kristinsson et al, 2010). A recent review
loma is multifactorial, with disease- and treatment-related of nearly 5000 myeloma patients showed VTE to be signifi-
factors playing important roles. Immunomodulatory drugs cantly associated with increased mortality at 2 and 5 years
(IMiDs) and high-dose dexamethasone, in particular, are after diagnosis, independent of other known prognostic fac-
known to enhance the thrombotic potential of myeloma. For tors (Schoen et al, 2018). Although VTE risk is highest with
this reason, assessment of the VTE risk has long been advo- active myeloma, it also extends to some degree to those with
cated prior to treatment initiation in patients with myeloma monoclonal gammopathy of uncertain significance (MGUS).
requiring IMiD-based regimens. However, despite routine A large retrospective study of over 4 million US veterans
use of thromboprophylaxis, these patients can still develop found a three-fold increased risk of VTE in MGUS cases,
VTE and its sequelae. The optimum choice and dose of and a nine-fold increased risk in myeloma cases (Kristinsson
thromboprophylactic drug is not entirely clear, and with this, et al, 2008). As VTE is associated with an increased mortality
there is growing interest regarding use of the direct oral anti- (Sanfilippo et al, 2014), appropriate identification of VTE
coagulants in this setting. In this review we discuss the risk factors and subsequent stratification of patients is of
pathogenesis of thrombosis in multiple myeloma, its relation paramount importance in the optimal care of patients with
to some of the commonly used chemotherapeutic regimens, myeloma.
current risk stratification and the evidence supporting the Our aim was to review the pathophysiology of thrombosis
different anticoagulants used as thromboprophylaxis. We in myeloma, how this is affected by common anti-myeloma
propose an amended risk stratification, and consider man- treatments, and the efficacy of the thromboprophylactic
agement of challenging patients, including those with renal agents available. Finally, we propose practical algorithms for
impairment and recurrent thrombosis. assessment of thrombotic risk, choice of prophylaxis, and
management of VTE recurrence. MEDLINE, Embase and
Keywords: anticoagulant, myeloma, thrombosis, heparin, NHS EVIDENCE were searched systematically for publica-
embolism. tions in English using the key words ‘thrombosis’, ‘anticoag-
ulation’ and ‘multiple myeloma’. References from relevant
The risk of venous thromboembolism (VTE) is significantly publications were also searched. Editorials, studies with <8
increased in patients with cancer (43-fold increased inci- cases and letters were excluded. Conference abstracts have
dence) (Blom et al, 2005), and VTE is the second common- been included if deemed to be of particular relevance.
est cause of death in these patients, other than the
malignancy itself (Ambrus et al, 1975; Donati, 1994; Khorana
Pathogenesis of thrombosis in myeloma
Correspondence: Dr D Swan, Department of Haematology, Several disease-related, treatment-based and patient-related
University Hospital Galway, Galway H91 YR71, Ireland. factors can lead to thrombosis in myeloma (Bellotti et al,
E-mail: dawnswan123@gmail.com 1989; Amrani, 1990; O’Kane et al, 1994; Stirling et al, 1998;

First published online 18 November 2018 ª 2018 British Society for Haematology and John Wiley & Sons Ltd
doi: 10.1111/bjh.15684 British Journal of Haematology, 2018, 183, 538–556
Review

Mechtcheriakova et al, 1999; Yasin et al, 1999; Gado et al, supportive evidence for the thrombogenic potential of high-
2000; Thiagarajan et al, 2000; Esmon, 2001; Yagci et al, 2003; dose dexamethasone (Rajkumar et al, 2010).
Zangari et al, 2003; Palumbo et al, 2004; Elice et al, 2006;
Auwerda et al, 2007; Kotlin et al, 2008; Dong et al, 2018;
Immunomodulatory agents
Guo et al, 2018). The effect of plasma cell neoplasms on
thrombotic risk is paramount and the various ways they can Thalidomide is the first in the class of immunomodulatory
promote clot formation are given in Table I. Risk of VTE is drugs (IMiDs) used for myeloma treatment. Its mode of
well documented to be increased during the active phase of action is multifactorial, affecting angiogenesis, adhesion of
disease, particularly during the first 6 months of treatment. myeloma plasma cells, and regulation of the immune system
This risk can be further augmented by certain therapeutic (Palumbo & Palladino, 2012). VTE can be associated with
agents, in particular the immunomodulatory drugs, and single-agent thalidomide use, but is seen more often when
especially in combination with high doses of corticosteroids thalidomide is combined with glucocorticoids, and is a fre-
and chemotherapy. quent complication of treatment with high-dose dexametha-
sone or combination chemotherapy (Barlogie et al, 2001;
Bennett et al, 2002; Rajkumar et al, 2002; Tosi et al, 2002;
Dexamethasone
Weber, 2002; Li et al, 2017). Rates of VTE seen with thalido-
Corticosteroids have been associated with VTE in different mide with the addition of dexamethasone range from 2% to
diseases, including myeloma (Johannesdottir et al, 2013). 17% (Dimopoulos et al, 2001; Cavo et al, 2004; Rajkumar
High doses of dexamethasone have been shown to stimulate et al, 2006) while rates of 10–58% have been reported
increased expression of tissue factor (TF), cellular adhesion with combination chemotherapy (Zangari et al, 2002; Zervas
molecules (ICAM-1, VCAM-1 and E-selectin) and von Wille- et al, 2004; Baz et al, 2005; Schutt et al, 2005; Barlogie et al,
brand factor (VWF), and decreased expression of thrombo- 2006). The highest incidence (58%) was reported by Baz et al
modulin (TM) and plasminogen activator inhibitor-1 (PAI-1) (2005) in their trial evaluating the use of thalidomide in
by human umbilical vein endothelial cells in vitro (Kerachian association with doxorubicin, vincristine and dexamethasone.
et al, 2009), with similar antifibrinolytic effects seen in rats Thrombosis rates are higher in newly diagnosed patients than
(van Giezen & Jansen, 1992). Dexamethasone may act indi- those with relapsed/refractory disease (2–15% vs. 3–34%)
rectly, by sensitizing cells to cytokine stimulation e.g. although the exact reasons for this difference is unclear
tumour necrosis factor alpha (TNF-a), but there is limited (Palumbo et al, 2008). The incidence is maximal during the
definitive data available to confirm its mode of action. first 3 months of treatment and decreases after about
Indeed, at low doses, glucocorticoids may even have a pro- 12 months (Zangari et al, 2002, 2004a), possibly due to
tective effect against inflammation and thrombosis. In the release of prothrombotic factors from apoptotic myeloma
context of orthopaedic surgery, administration of hydrocor- cells when burden of disease is at its highest (Zangari et al,
tisone pre-operatively was associated with a significant 2003). In vitro studies have shown that application of
reduction in markers of thrombin generation, and a non- thalidomide to endothelial cells damaged by doxorubicin
significant increase in fibrinolysis markers (McLawhorn exposure leads to altered protease activated receptor-1 (PAR-1)
et al, 2015). expression, indicating endothelial dysfunction (Baz et al,
What is known however, is that clinically, the addition of 2005). This may partly explain the extremely high rate of
steroids, particularly at higher doses, is associated with a sig- thrombosis seen by Baz et al (2005). Association studies
nificant elevation in thrombosis risk. Rajkumar and Blood have also suggested a link between IMiDs and increased
(2006) reported a comparison between lenalidomide with endothelial TF expression (Li et al, 2017). A transient
high- or low-dose dexamethasone. In this study, high-dose reduction in levels of soluble TM was reported during the
was referred to as 40 mg dexamethasone on days 1–4, 9–12 first month of therapy in a group of 13 relapsed/refractory
and 17–20 of a 28-day cycle, versus low-dose where 40 mg of patients, one of whom developed VTE (Corso et al, 2004).
dexamethasone was administered once weekly. The total dose Interestingly, IMiDs themselves have not been shown
of dexamethasone received in the ‘high-dose’ group was to cause endothelial damage (Streetly et al, 2005). The
480 mg/month, in line with the international Myeloma thrombogenicity of thalidomide may therefore be potenti-
Working Group’s (IMWG’s) later definition of ‘high-dose’ ated by endothelial damage from combination chemother-
glucocorticoids (Palumbo et al, 2008). In the initial part of apy, and dexamethasone-induced sensitization of cells to
the study, VTE prophylaxis was recommended but not man- cytokine stimulation, which is known to be upregulated
dated. Of the first 266 enrolled patients, 182% developed by IMiDs.
VTE in the high-dose group and 37% in the low-dose Lenalidomide is a second-generation immunomodulatory
group, after which thromboprophylaxis became mandatory agent with increased in vitro efficacy compared with thalido-
(Rajkumar & Blood, 2006). At 1 year from study initiation, mide, and less toxicity in data pooled from different trials
the VTE rate in the high-dose group was over double that of (no head-to-head comparison of thalidomide and lenalido-
the low-dose group (26% vs. 12%), providing substantial mide exists in the literature). Like thalidomide, single agent

ª 2018 British Society for Haematology and John Wiley & Sons Ltd 539
British Journal of Haematology, 2018, 183, 538–556
540
Review

Table I. Thrombotic risk factors in plasma cell neoplasms.

Disease-related Treatment-related Patient-related

Immunoglobulin dependent Drugs


• Hyperviscosity, e.g. serum viscosity >4 cp (serum IgG usually ≥40 g/l, or IgA ≥60 g/l) • Thalidomide/lenalidomide • Fractures and other causes of immobility
*(Zangari et al, 2003) • Dexamethasone • Severe infections
• Defective fibrin polymerisation and fibrinolysis (O’Kane et al, 1994; Thiagarajan et al, • Multiagent chemotherapy • Elevated body mass index
2000; Yagci et al, 2003; Kotlin et al, 2008) • Proteasome inhibitors- bortezomib exerts a • Comorbidities, e.g. cardiac disease, chronic renal
• Lupus anticoagulant activity of the paraprotein, antibodies against Protein S and C, protective effect, carfilzomib is a risk failure, diabetes mellitus, inflammatory bowel dis-
acquired APC resistance (Bellotti et al, 1989; Yasin et al, 1999) factor for VTE ease, autoimmune disease
• Erythropoietin-stimulating agents • Surgery, anaesthesia, trauma
• Indwelling central catheters (Palumbo • History of VTE or inherited thrombophilias (not
et al, 2008) myeloma- specific) (Palumbo et al, 2008)

Immunoglobulin independent
• Elevated PAI-1
• Increased proinflammatory cytokines IL6 and VEGF (Amrani, 1990; Stirling et al, 1998;
Mechtcheriakova et al, 1999; Gado et al, 2000)
• Increased endothelial tissue factor expression (Dong et al, 2018)
• Elevated FVIII, VWF and fibrinogen levels (Stirling et al, 1998; Auwerda et al, 2007)
• Increased cell surface phosphatidylserine expression (Dong et al, 2018; Guo et al, 2018)
• Acquired APC resistance due to reduced levels of thrombomodulin (Esmon, 2001; Elice
et al, 2006)

APC, activated protein C; FVIII, factor VIII; IL6, interleukin 6; PAI-1, plasminogen activator inhibitor-1; VEGF, vascular endothelial growth factor; VTE, venous thromboembolism; VWF, von Wille-
brand factor.
*IgM myeloma is uncommon. Hyperviscosity predisposing to increased risk of thrombosis is more likely to occur if serum IgM is >30 g/l.

ª 2018 British Society for Haematology and John Wiley & Sons Ltd
British Journal of Haematology, 2018, 183, 538–556
Review

lenalidomide has a modest thrombogenic potential (reported transcription factor production results in enhanced expres-
incidence 4%) (Richardson et al, 2009), which is enhanced sion of the natural anticoagulant endothelial TM via induc-
by the addition of glucocorticoids, particularly high-dose tion of Kruppel-like transcription factors. Bortezomib can
dexamethasone (VTE 26% vs. 12% high i.e. 480 mg/month also prevent TM downregulation by inflammatory cytokines
vs. low-dose dexamethasone) (Zonder et al, 2005; Dimopou- (Lonial et al, 2008; Hiroi et al, 2009; Nayak et al, 2014).
los et al, 2007; Weber et al, 2007; Rajkumar et al, 2010), and Data relating to bortezomib use suggests a far lower, or pos-
combination chemotherapy (14% when used with cyclophos- sibly even absent, thrombogenic potential. Addition of borte-
phamide and 9% with doxorubicin) (Baz et al, 2006; Knop zomib to melphalan-prednisolone in the phase 3 VISTA
et al, 2006; Morgan et al, 2007). VTE rates are again higher (Velcade as Initial Standard Therapy in Multiple Myeloma)
in newly diagnosed patients. A meta-analysis of 125 patients trial was not associated with increased VTE rates (San Miguel
enrolled in three clinical trials stratified the patients into high et al, 2008). The APEX (Assessment of Proteasome Inhibition
and low-VTE risk groups based on the concomitant dose of for Extending Remissions) trial of single-agent bortezomib
dexamethasone administered (40 mg weekly, or 40 mg on versus high-dose dexamethasone in relapsed patients reported
12 days/month, total 480 mg/month). Most of the patients differential VTE rates of 06% for bortezomib vs. 27% in the
(n = 110) received thromboprophylaxis, which in the major- dexamethasone arm (Lonial et al, 2008). Addition of borte-
ity was 325 mg aspirin once daily. The VTE rate was 12% zomib to the DT-PACE regimen (dexamethasone, thalido-
for the high-dose dexamethasone group, 6% in the low-dose mide, cisplatin, doxorubicin, cyclophosphamide and
group, 7% in those on aspirin and 13% in those not receiv- etoposide) was associated with a significant reduction in
ing thromboprophylaxis. Seven of the 10 VTE episodes thrombotic episodes, from 10% to 0% (Zangari et al,
occurred during the initial 6 months of therapy in keeping 2004b). In the frontline setting, Zangari et al (2011) reviewed
with the known increased thrombogenic potential associated phase 3 trials of bortezomib and/or immunomodulatory
with high disease burden (Menon et al, 2008). Consistent agent-based therapy. Regimens including bortezomib had
with these results, the Greek Myeloma Study Group analysed overall VTE rates of ≤5% whereas IMiD-based treatment
212 relapsed/refractory patients treated with lenalidomide without bortezomib was associated with higher rates, further
and low-dose dexamethasone outside of clinical trials, and corroborating the low thrombogenicity associated with borte-
reported a similar VTE incidence of 57% overall (Katodritou zomib (Zangari et al, 2011).
et al, 2014). A small study involving 10 patients with relapsed multiple
There is less evidence again regarding the third-generation myeloma described in vivo effects of bortezomib on routine
immunomodulatory agent, pomalidomide. A small phase I coagulation tests and impairment of platelet function (Zan-
study of 24 relapsed/refractory patients receiving single agent gari et al, 2008). Platelet aggregation with different agonists
pomalidomide reported VTE in 17% without thrombopro- was decreased after bortezomib infusion, with statistically sig-
phylaxis (Schey et al, 2004). Subsequent studies have incor- nificant results with ADP on days one (20% decrease,
porated varying doses of thromboprophylaxis into treatment P = 0033) and four (29% decrease, P = 0009). Similar
regimens. Of 60 relapsed/refractory patients receiving poma- results were also obtained with epinephrine-induced platelet
lidomide with low-dose weekly dexamethasone, only one aggregation and ristocetin-induced agglutination. Expression
thromboembolic event was reported (16%). All patients level of the platelet surface marker, P-selectin, which has
received thromboprophylaxis with a high dose of aspirin roles in adhesion and thrombosis, was also decreased after
(325 mg), therapeutic dose low molecular weight heparin bortezomib treatment (Zangari et al, 2008). This anti-platelet
(LMWH) or warfarin with a target International Normalised effect and alteration in adhesion properties may explain the
Ratio (INR) of 2–3 (of note, none of these thromboprophy- low VTE rates seen with proteasome inhibition.
laxis strategies would be routinely chosen in current clinical There is less available data on the second-generation pro-
practice) (Lacy et al, 2009). A recent study of pomalidomide teasome inhibitor, carfilzomib, which, unlike bortezomib,
in combination with low-dose dexamethasone and borte- causes irreversible inhibition of the 26S proteasome. The
zomib in 50 patients reported a higher VTE rate of 10%, phase 3 study ASPIRE (Carfilzomib, lenalidomide and dex-
despite using the high doses of thromboprophylactic agents amethasone versus lenalidomide and dexamethasone for the
detailed in the previous study (Paludo et al, 2017). A sum- treatment of patients with relapsed multiple myeloma) com-
mary of trial data pertaining to VTE risk associated with the pared the triplet of carfilzomib, lenalidomide and dexam-
immunomodulatory drugs either alone, with dexamethasone, ethasone, with lenalidomide and dexamethasone. VTE
or with combination chemotherapy, is provided in Table II. incidence during the first year was 13% vs. 6% against the
carfilzomib arm (Stewart et al, 2015), with significant rates
of carfilzomib-related cardiovascular disease [hypertension,
Proteasome inhibitors
arrhythmias, myocardial infarction (MI), and congestive car-
The first in-class proteasome inhibitor, bortezomib, reversibly diac failure] noted in this and other studies. These effects,
inhibits the 20S subunit of the 26S proteasome. Downstream suspected to be a reflection of endothelial toxicity possibly
suppression of nuclear factor jB (NF-jB)-mediated related to the irreversible 26S proteasome inhibition, appear

ª 2018 British Society for Haematology and John Wiley & Sons Ltd 541
British Journal of Haematology, 2018, 183, 538–556
Table II. Primary experimental trials using IMiDs in multiple myeloma (meta-analyses excluded).

542
Review

Study reference Regimen and patients Exclusions Thromboprophylaxis VTE rate

Thalidomide
Barlogie et al 169 R/R patients – single-agent thalidomide Renal/liver impairment NOT Not described <2%
(2001) excluded
Tosi et al 65 R/R patients – single agent thalidomide None stated None 15%
(2002) (maximum 800 mg/day)
Thalidomide-dexamethasone
Cavo et al 71 newly diagnosed patients – thalidomide with high-dose Age >65 years, history of First 19 patients- no 26% without warfarin;
(2004) dexamethasone prior to ASCT thrombosis thromboprophylaxis; remaining 52 13% with warfarin
– 125 mg prophylactic warfarin
Rajkumar et al 207 patients with newly diagnosed myeloma randomized Severe renal impairment, liver None 17% in the thalidomide
(2006) to high dose dexamethasone  thalidomide impairment, cytopenias, or arm vs. 3% without
past/current thrombosis
Rajkumar et al 50 patients with newly diagnosed myeloma – thalidomide Severe cytopenias, poor None Single agent arm 4%;
(2002) with dexamethasone at alternating high and low doses performance status combination 15%
Weber et al 28 newly diagnosed asymptomatic myeloma patients – Newly diagnosed patients; those None given in single-agent arm. In 34% vs. 18% with or
(2003) single agent thalidomide (maximum dose 600 mg);40 with CRAB criteria were excluded combination arm, first 24 patients without thalidomide
patients – thalidomide with high-dose dexamethasone – 1 mg o.d. coumadin; remaining
16 – treatment dose LMWH or
coumadin
Thalidomide combination therapy
Barlogie et al Newly diagnosed myeloma patients randomized to Adequate performance status, renal First 162 patients – no 24% without LMWH vs.
(2006) melphalan-ASCT  thalidomide failure not excluded thromboprophylaxis; remaining 15% with LMWH
163 – prophylactic LMWH
Baz et al (2005) 105 newly diagnosed or relapsed patients – thalidomide, Life-expectancy <3 months, severely Aspirin 81 mg o.d. p.o., from the 19% (aspirin from the
pegylated doxorubicin and vincristine deranged liver function, start of treatment, after 1 cycle or start); 15% (from cycle
impaired LV function not at all 2); 58% (no aspirin)
Palumbo et al 667 patients treated with thalidomide-containing regimen: Previous venous or arterial Aspirin 100 mg o.d. p.o., warfarin At 6 months: 64% for
(2011) if aged <65 years, bortezomib, thalidomide, and thrombosis or high risk of 125 mg o.d. p.o., or enoxaparin aspirin; 5% for LMWH;
dexamethasone, or thalidomide and bleeding 40 mg o.d s.c. 82% for warfarin
dexamethasone with ASCT; if aged >65 years,
melphalan, prednisolone, and thalidomide with
maintenance, or bortezomib, melphalan, thalidomide
and prednisolone
without maintenance
Schutt et al 31 patients – thalidomide 400 mg with vincristine, None stated Not specified 26%
(2005) epirubicin and dexamethasone  ASCT
Zangari et al Comparison between 98 patients receiving DT-PACE and None stated Not specified 10% in DT-PACE trial
(2004b) 68 receiving VDT-PACE in the UARK 2001–12 and vs. 0% for VDT-PACE
2003–33 trials respectively (

ª 2018 British Society for Haematology and John Wiley & Sons Ltd
British Journal of Haematology, 2018, 183, 538–556
Table II. (Continued)

Study reference Regimen and patients Exclusions Thromboprophylaxis VTE rate

Zangari et al 192 R/R patients received DT-PACE and 40 received None stated Not specified DVT occurred in 165%
(2002) DCEP-T in the DT-PACE arm
vs. 25% in the DCEP-T arm
Zervas et al 39 newly diagnosed patients – thalidomide (200 mg), None stated Not specified DVT in 10%
(2004) vincristine, liposomal doxorubicin and dexamethasone
Lenalidomide
Richardson 222 R/R patients – lenalidomide 30 mg days 1–21 Non-myeloma related severe Not specified DVT 4%; PE 1%
et al (2009) cytopenias, severe renal
impairment, liver
enzymes >3 times ULN
Lenalidomide-dexamethasone

British Journal of Haematology, 2018, 183, 538–556


Zonder et al 12 newly diagnosed patients – 25 mg lenalidomide with None stated No prophylaxis LD arm 75%; placebo 0%
(2005) high-dose dexamethasone (LD), and 9 received
dexamethasone with placebo
Weber et al 177 R/R patients – lenalidomide 25 mg days 1–21 with Non-myeloma related severe Thrombo-prophylaxis- physician 147% vs. 34% in

ª 2018 British Society for Haematology and John Wiley & Sons Ltd
(2007) high dose dexamethasone, vs. 176 patients on a placebo cytopenias, severe renal discretion treatment arm vs. placebo
arm impairment, liver
enzymes >3 times ULN
Dimopoulos 351 R/R patients – lenalidomide days 21 days per 28-day Non-myeloma related severe Not specified 114% in placebo/
et al (2007) cycle and high-dose dexamethasone, or placebo/ cytopenias, Liver enzymes >2 times dexamethasone arm vs. 46% in
dexamethasone ULN, severe renal impairment lenalido
mide- o.d. p.o.arm
Rajkumar et al 34 newly diagnosed patients – lenalidomide 25 mg days 1 Cytopenias, severe renal Aspirin 80 mg or 325 mg at 9%
(2005) –21 and high dose dexamethasone impairment, unanticoagulated VTE clinician’s discretion o.d. p.o.
Rajkumar et al 445 newly diagnosed patients – randomized to Severe cytopenias, renal impairment Thrombo-prophylaxis was 26% in high dose o.d. p.o.arm
(2010) lenalidomide 25 mg days 1–21 with either high or low or liver impairment, current recommended initially then and 12% in the low dose arm
dose dexamethasone  ASCT or past thrombosis, poor mandated after the first 266
performance status patients due to high VTE rates
Lenalidomide combination therapy
Baz et al (2006) 62 newly diagnosed or relapsed patients -lenalidomide, Life-expectancy <3 months, Non- Aspirin 81 mg o.d. p.o. 3%
peglylated doxorubicin and vincristine myeloma related cytopenias, liver
enzymes >2 times ULN, severe
renal impairment, impaired LV
Knop et al 41 R/R patients received lenalidomide, doxorubicin and Adequate organ and bone marrow Not specified 0%
(2006) dexamethasone function required

543
Review
544
Review

Table II. (Continued)

Study reference Regimen and patients Exclusions Thromboprophylaxis VTE rate

Larocca et al, 342 newly diagnosed patients – melphalan, prednisolone Excluded if history of arterial Assigned to aspirin 100 mg o.d. p.o. 227% in aspirin group, 12% in
2012) and lenalidomide plus ASCT thrombosis or VTE in past or enoxaparin 40 mg o.d. s.c. enoxaparin group
12 months,
contraindication to aspirin or
LMWH, or active bleeding
Morgan et al 21 heavily pre-treated R/R patients – lenalidomide 25 mg Not specified Not specified 14%
(2007) o.d. days 1–21, cyclophosphamide weekly, and 40 mg
dexamethasone days 1–4 and 12–15
Palumbo et al 54 newly diagnosed patients – oral melphalan and Creatinine clearance <20 ml/min, Aspirin 100 mg o.d. p.o. 48%
(2007) prednisolone days 1–4 and lenalidomide days 1–21 severely deranged LFTs, severe
cytopenias, amyloidosis,
psychiatric illness
Stewart et al 792 R/R patients – carfilzomib with lenalidomide and Moderate renal impairment, All patients received 1-year rate 13% in the carfilzomib
(2015) dexamethasone, or lenalidomide and dexamethasone cytopenias, heart failure, grade 3–4 thromboprophylaxis- not specified arm vs. 6% without
(low-dose) peripheral neuropathy
Thalidomide or lenalidomide
Bradbury et al 3838 newly diagnosed patients – cyclophosphamide, Grade 2 or worse peripheral Thrombo-prophylaxis as per the 118%, no significant difference
(2017) dexamethasone + thalidomide or lenalidomide  ASCT neuropathy, acute renal failure IMWG 2008 guidelines (Palumbo between thalidomide or
if eligible unresponsive to 72 h of et al, 2008) lenalidomide arms
intravenous fluids
Pomalidomide
Schey et al 24 R/R patients – escalating doses of CC-4047 Severe cytopenias, moderate renal None 1 case - due to undiagnosed
(2004) (pomalidomide) impairment melanoma
Streetly et al 15 R/R patients received escalating doses of CC-4047 None specified None 20%
(2005) (pomalidomide)
Pomalidomide combination therapy
Paludo et al 50 R/R patients – pomalidomide 4 mg days 1–21, weekly DVT not anticoagulated, poor 325 mg aspirin o.d. p.o., 10%
(2017) bortezomib and weekly dexamethasone 40 mg performance status, pregnancy therapeutic LMWH or warfarin

ASCT, autologous stem cell transplantation; CRAB, (elevated) calcium, renal failure, anaemia, bone lesions; DCEP-T, dexamethasone, thalidomide, cisplatin, cyclophosphamide, etoposide; DT-PACE,
dexamethasone, thalidomide, cisplatin, doxorubicin, cyclophosphamide, etoposide; DVT, deep venous thrombosis; IMiD, immunomodulatory drug; IMWG, International Myeloma Working Group;
LFT, liver function test; LMWH, low molecular weight heparin; LV, left ventricular; o.d., every day; PE, pulmonary embolism; p.o., orally; R/R, relapsed/refractory; s.c. , subcutaneously; ULN, upper
limit of normal; VDT-PACE, velcade, dexamethasone, thalidomide, cisplatin, doxorubicin, cyclophosphamide, etoposide; VTE, venous thromboembolism.

ª 2018 British Society for Haematology and John Wiley & Sons Ltd
British Journal of Haematology, 2018, 183, 538–556
Review

to be specific to carfilzomib and are not seen with borte- (Preventive treatment of venous thrombo-embolic disease in
zomib. patients with multiple myeloma receiving chemotherapy with
thalidomide or lenalinomide) study was a large, multicentric,
observational study that sought to evaluate VTE incidence,
Prophylaxis – risk stratification
risk assessment and prophylaxis chosen in real-life practice in
Assessment of VTE risk in myeloma has been advocated for France. Of 513 patients on lenalidomide- or thalidomide-
many years (Palumbo et al, 2008), the mainstay of which based regimens, 47% were deemed to be at low VTE risk,
requires a thorough clinical history, with close attention 39% intermediate and 14% high. Patients were managed with
being paid to the features described in Table I. Re-assessment aspirin, LMWH or vitamin K antagonist (VKA). At
is suggested at diagnosis, relapse, or when otherwise clinically 12 months follow-up, VTE rates were 7% for aspirin, 3%
indicated e.g. if the patient develops VTE, or at instigation of LMWH and 0% in the VKA-treated group with no statistical
a new non myeloma-related therapy with increased thrombo- significance identified. However, risk stratification was not
genic potential. standardised; 47% of patients were thought to be low risk,
Optimal choice of pharmacological prophylaxis and however only 15% had fewer than 3 VTE risk factors. Choice
appropriate patient VTE risk-stratification in patients with of prophylaxis was also not standardised and did not follow
myeloma requires ongoing attention. Recent results from the current international recommendations (Palumbo et al,
large, phase 3 Myeloma XI trial highlight this fact. The 2008), with 19% of high-risk individuals receiving low-dose
updated analysis showed an overall VTE rate of 124%, aspirin, and LMWH or VKA only prescribed for 40% of
111% of which was during the first 6 months of therapy, intermediate- and high-risk patients. A lack of consensus and
with no differences seen between the thalidomide and clarity appeared evident with respect to both risk stratifica-
lenalidomide-containing regimens. The majority of patients tion of patients and subsequent choice of thromboprophy-
(876%) were on some type of thromboprophylaxis prior to laxis (Leleu et al, 2013; Chalayer et al, 2016a).
VTE with only 124% of patients not receiving thrombopro- Attempts to improve VTE risk stratifications have been
phylaxis. However, the types and doses of thrombopro- made, and various groups are investigating markers of
phyaxis were variable and not accounted for by the thrombosis that could be incorporated into practical, vali-
preceding VTE risk assessment. Over 10% were on therapeu- dated risk assessment tools. Elevated levels of soluble P-selec-
tic dose LMWH prior to their thrombotic event, further tin, an adhesion protein which mediates interactions between
emphasising the strong prothrombotic potential of selected platelets, leucocytes and cancer cells, have been shown to be
patients (Bradbury et al, 2017, 2018). associated with an increased risk of VTE, and incorporated
In 2008, the IMWG recommended that a VTE risk assess- into the Vienna risk prediction model (Ay et al, 2008). Other
ment should be performed in all multiple myeloma patients markers under investigation include D-dimers, microparticles
treated with thalidomide or lenalidomide (Palumbo et al, and TF, which was shown to be associated with recurrence
2008). They suggested aspirin prophylaxis (dose 81–325 mg of VTE in the CATCH [Long-term Innohep treatment ver-
once daily) for those with no or one risk factor, LMWH or sus a vitamin K antagonist (warfarin) for the treatment of
therapeutic dose warfarin if two or more risk factors are pre- VTE in Cancer] trial (Khorana et al, 2017). Very recently, a
sent, and the same for those receiving high-dose dexametha- group analysed conventional clinical myeloma risk factors,
sone, doxorubicin or multi-agent chemotherapy irrespective of such as dexamethasone use, thalidomide, previous VTE, cen-
preceding risk factors (Palumbo et al, 2008). The recommen- tral venous catheter in situ and obesity, and developed a new
dations were provided with the caveat that they were based on risk model (the myeloma clot score, MCS), which they vali-
the limited available data alongside expert opinion, and do not dated (Sanfilippo et al, 2018).
provide a definitive model for this issue. The British Society of Point-of-care tests of global haemostasis may play a future
Haematology guidelines on the supportive care of multiple role in identifying patients with prothrombotic haemostatic
myeloma, 2011, concur with this advice (Snowden et al, 2011), profiles, and perhaps in highlighting those with features of
as do the 2016 National Institute of Clinical Excellence (NICE) resistance to heparin or other anticoagulation (see Recurrent
guidelines, once again acknowledging the relative lack of rigor- thrombosis section). Given their increasing importance in
ous evidence on the topic (NICE 2016). However, the most other areas, such as management of major haemorrhage, it is
recent NICE guidance, (NICE 2018), merely suggests that likely they may also be of use in this field, however more
either aspirin or prophylactic dose LMWH may be considered research is required.
equivalent for myeloma patients receiving chemotherapy with
IMiDs and a corticosteroid.
Prophylaxis – choice of agent
Although risk assessment tools are available, there is evi-
dence to suggest that clinicians are often stratifying patients
Aspirin versus LMWH
according to personal perception of strength of certain risk
factors, e.g. placing more weight on a family history than The IMWG have advised the use of aspirin in low VTE risk
treatment with high-dose dexamethasone. The MELISSE patients receiving IMiD-based regimens, concluding from the

ª 2018 British Society for Haematology and John Wiley & Sons Ltd 545
British Journal of Haematology, 2018, 183, 538–556
Review

available evidence that it is of clinical utility in this subgroup dexamethasone (103%) or melphalan, lenalidomide, pred-
(Palumbo et al, 2008). Due to its oral route and lack of moni- nisolone (MPR) (41%). They concluded that aspirin may be
toring, it is an appealing option for patients, however most of insufficient for regimens incorporating high doses of dexam-
the current evidence is not based on randomized trials, and ethasone but may be a safe option for lower risk treatments,
published results do not uniformly show benefit. A study of such as MPR (Al-Ani et al, 2016). In contrast to these find-
patients receiving combination chemotherapy with thalido- ings, a large retrospective cohort study of 4892 multiple mye-
mide, pegylated doxorubicin, vincristine and dexamethasone loma patients in whom 586 developed VTE did not find
without pharmacological thromboprophylaxis yielded a VTE evidence that aspirin reduced the risk of VTE, after adjusting
incidence of 58% which was reduced to 18% in subsequent for risk factors including IMiD use and past history of VTE.
patients by the addition of aspirin 81 mg daily (Baz et al, They suggested that aspirin may be insufficient for patients
2005). A small study of 34 patients receiving lenalidomide, on immunomodulatory agent-based treatments and also
dexamethasone and aspirin reported a 3% VTE rate (Rajkumar those with a personal history of VTE (Sanfilippo et al, 2017).
et al, 2005). A subsequent, larger, randomized study of From an economic standpoint, the limited cost associated
lenalidomide with high or low dose dexamethasone found the with low-dose aspirin is advantageous. Chalayer et al (2016b)
addition of low-dose aspirin reduced VTE rates from 23% to performed a cost-effectiveness analysis of LMWH versus
14% for high-dose dexamethasone patients and from 14% to aspirin in newly diagnosed multiple myeloma patients, based
5% for low-dose patients (Rajkumar et al, 2006). A study of on the work reported by Palumbo et al (2011). In their
melphalan, prednisolone and lenalidomide with aspirin model, aspirin use was associated with a higher frequency of
100 mg reported a 5% VTE rate (Palumbo et al, 2007), and VTE, stroke and major bleeding, but reduced incidence of
another of lenalidomide, doxorubicin and dexamethasone with acute MI, and, due to the difference in route of administra-
81 mg aspirin reported a 9% rate (Baz et al, 2006). tion, a slightly higher quality-adjusted life years (QALY) than
More recently, 342 patients treated with lenalidomide- LMWH (0300 vs. 0299). Over 6 months of treatment, using
based induction and consolidation therapy (lenalidomide aspirin in place of LMWH was calculated to save an average
and low-dose dexamethasone induction; melphalan-predniso- of €1245 per patient, with a slight improvement in quality of
lone-lenalidomide consolidation) were randomized between life. Of course, these results do not apply to patients who
aspirin 100 mg/day and enoxaparin 40 mg/day. This develop VTE with its associated complications, and no
prospective, open-label, randomized sub-study of a phase 3 assessment of VTE risk was made in this model (Chalayer
trial showed that during the first 6 months post-randomisa- et al, 2016b).
tion, VTE incidence was 227% in the aspirin group and Data supports low-dose aspirin as a reasonable option for
120% in the LMWH group, with an absolute difference of patients on IMiD-containing regimens who are otherwise at
107% [95% confidence interval (CI), 169 to 383; low-risk of thromboembolic complications, although the
P = 0452] in favour of enoxaparin. Importantly, high-risk number of patients in this category is likely to be small.
patients were excluded including those with recent orthopae- Given the conflicting results available, we would generally
dic surgery, vertebroplasty, immobilisation, thrombophilia, recommend offering prophylactic LMWH to all patients
known ischaemic heart disease or previous atrial fibrillation, receiving initial multiple myeloma treatment with an IMiD
and none of the cohort had a past history of VTE (Larocca in combination with corticosteroid or chemotherapy, at least
et al, 2012). Another larger randomised trial of 667 patients during the initial 6 months of treatment when risk of throm-
receiving thalidomide-based first-line myeloma therapy com- bosis is at its highest. Aspirin could be considered beyond
pared aspirin 100 mg, fixed low-dose warfarin 125 mg and 6 months in the absence of additional significant patient-
enoxaparin 40 mg. Reported rates of serious thromboembolic related VTE risk factors. However, these recommendations
events, acute cardiovascular events or sudden death during are not based on robust evidence or any formal guideline.
the first 6 months were 64% for aspirin, 82% for warfarin Prophylactic dose LMWH confers the greatest protection out
and 50% for LMWH. Three major and 10 minor bleeding of the conventional agents available, and is therefore the drug
episodes were recorded. High risk patients were excluded in of choice for ‘high-risk’ individuals. There is some evidence
this trial. The authors concluded from their data that aspirin that it may be inadequate for particularly high-risk patients
was an effective prophylactic agent in low-risk patients, who may benefit from more intensive VTE prophylaxis.
although there was no placebo arm for comparison. Of note, However, there is a lack of evidence that a higher dose of
in this trial, the risk of VTE was 138 times greater in anticoagulation is effective and safe, and risk of thrombosis
patients treated with thalidomide without bortezomib and haemorrhage must be balanced if intensification of anti-
(Palumbo et al, 2011). coagulation is to be considered.
In terms of lenalidomide, a systematic review of over 1126
patients found overall VTE occurrence rates of 107% in
Warfarin
patients receiving aspirin prophylaxis vs. 14% for those on
LMWH. The highest risk was in patients taking lenalidomide The little data available on warfarin as thromboprophylaxis
with high-dose steroids (266%), compared with low-dose suggests that, at therapeutic doses aiming for an INR

546 ª 2018 British Society for Haematology and John Wiley & Sons Ltd
British Journal of Haematology, 2018, 183, 538–556
Review

between 2 and 3, it isn’t superior to prophylactic dose events in the DOAC arm (13% vs. 4%) (Young et al, 2018).
LMWH (Palumbo et al, 2011), and that at low fixed doses, A significant proportion of bleeds were upper GI in source,
e.g. 1–125 mg/day, it has poor efficacy. One study of and more common amongst patients with gastric tract malig-
thalidomide, dexamethasone and low dose warfarin saw a nancies (Raskob et al, 2018; Young et al, 2018), which would
VTE incidence of 25% (Weber et al, 2003), and another of be of limited relevance in the setting of myeloma. These
thalidomide, dexamethasone and chemotherapy reported a studies did include some myeloma patients, however num-
rate of 31% with fixed dose warfarin prophylaxis versus 15% bers were very small e.g. one patient in the DOAC arm in
with 40 mg enoxaparin (Zangari et al, 2004a). Warfarin also SELECT-D. They also did not address the issue of VTE pro-
has the potential for drug interactions and of course the need phylaxis in cancer.
for frequent blood tests. Fluctuations in the INR due to Data regarding prophylactic use of DOACs in the cancer
issues with absorption [from vomiting and other gastroin- setting is presently sparser, and based mainly on small case
testinal (GI) issues], drug interactions, low albumin, systemic series and case reports, but with promising results seen. A
illnesses and interruptions for procedures can make warfarin retrospective review of patients on IMiD-based regimens
a less attractive option. An additional practical issue to con- receiving therapeutic dose warfarin (16 patients) or therapeu-
sider is the fluctuating thrombocytopenia which often devel- tic or prophylactic doses of dabigatran, rivaroxaban or apixa-
ops in patients on proteasome inhibitors. The increased ban (21 patients) as thromboprophylaxis reported four non-
bleeding risk associated with a platelet count <50 9 109/l major bleeds in the DOAC group versus a total of six bleeds
also needs consideration in these patients. in the warfarin group, two of which were major, and no
VTE events in either group (Man et al, 2017). A larger
review of 70 patients receiving apixaban 25 mg b.d. during
The role of direct oral anticoagulants (DOACs)
front-line therapy with IMiD-containing regimens reported
The DOACs, either inhibitors of factor Xa (apixaban, no episodes of VTE within the first 6 months, one ischaemic
rivaroxaban, edoxaban, betrixaban) or IIa (dabigatran), are stroke, one non-ST-elevation MI in a patient with known
an attractive option for VTE prophylaxis in myeloma, requir- ischaemic heart disease and one episode of major bleeding in
ing no monitoring at routine doses, and sparing patients a patient with concomitant severe thrombocytopenia. VTE
from daily subcutaneous injections. None of the DOACs are risk assessment scores for the cohort are not available, how-
currently licensed for this use. A meta-analysis of trials com- ever two patients had previous pulmonary embolism (PE),
paring DOACs to VKAs for treatment of acute VTE in over two had a history of stroke or transient ischaemic attack and
27 000 patients, without a specific diagnosis of myeloma, three had documented prior MI (Storrar et al, 2018). A
reported overall lower rates of VTE recurrence, major bleed- prospective randomized controlled trial of apixaban 25 mg
ing (including fatal bleeds and intracranial haemorrhage) and b.d. versus placebo in myeloma patients on IMiD-based ther-
clinically relevant non-major bleeds. Subgroup analysis of apy, is ongoing (NCT02958969), and a large, multi-centre
participants with cancer reported a hazard ratio (HR) of 057 study comparing DOACs, LMWH and warfarin in VTE pro-
for VTE recurrence in favour of the DOACs (van Es et al, phylaxis in cancer patients (CANVAS trial, NCT02744092) is
2014). There is also evidence for the use of DOACs in the also in recruitment.
setting of secondary VTE prevention, at reduced prophylactic Lack of routine monitoring at prophylactic doses is an
doses. This is provided by EINSTEIN CHOICE, where low attractive quality for patients, however may be an issue in
dose rivaroxaban (10 mg) was superior to aspirin (100 mg) patients where GI absorption is altered (e.g. chemotherapy-
at prevention of VTE recurrence with equally low rates of induced vomiting and diarrhoea), when drug clearance is
bleeding (Weitz et al, 2017), and by AMPLIFY EXT, which reduced in renal failure, or when drug interactions with
found 25 mg twice daily apixaban to be as effective as 5 mg chemotherapeutic agents and supportive treatments are
twice daily, without increased bleeding compared with pla- unknown. Drug level monitoring may be a solution to these
cebo (Agnelli et al, 2013). problems but isn’t currently widely available and there is no
Two recent trials have evaluated use of a DOAC in cancer evidence to guide thromboprophylaxis drug level targets for
patients for the treatment of VTE, one comparing edoxaban this indication, or dose alterations based on drug levels. In
to dalteparin and the other rivaroxaban to dalteparin addition to this, in comparison with solid cancers, bleeding
[HOKUSAI (Raskob et al, 2018) and SELECT-D (Young risk in myeloma is likely to be higher due to older age of the
et al, 2018) respectively]. Both studies obtained similar patients, co-existing thrombocytopenia and higher prevalence
results. The HOKUSAI trial reported a 12-month VTE recur- of renal dysfunction. Currently, a reversal agent is only avail-
rence rate favouring edoxaban (79% vs. 113%), but with a able for dabigatran but not yet for rivaroxaban, apixaban or
slight increase in major bleeding (69% vs. 4%) (Raskob edoxaban. Bleeding episodes related to the direct anti-Xa
et al, 2018). Similarly, the SELECT-D saw a reduction in 6- inhibitors require treatment with pro-haemostatic products
month VTE recurrence with rivaroxaban (4% vs. 11%), a (e.g. prothrombin complex concentrate).
slight increase in major bleeding (6% vs. 4%) and a signifi- The problems that may arise with DOAC use are pre-
cant increase in clinically significant non-major bleeding sented in Table III.

ª 2018 British Society for Haematology and John Wiley & Sons Ltd 547
British Journal of Haematology, 2018, 183, 538–556
Review

Pragmatic approach for VTE prophylaxis in • Very high-risk patients – after assessment of bleeding
myeloma risk and adjustment of any modifiable risk factors, a
high prophylactic dose of LMWH (target anti-Xa of
Here we provide a pragmatic approach to managing a
04–06 iu/ml) or therapeutic dose LMWH may be cho-
patient with myeloma who requires pharmacological throm-
sen until the period of greatest thromboembolic risk
boprophylaxis, taking into consideration the risk factors as
has passed. Given the evidence provide by Myeloma XI
proposed by the IMWG (Palumbo et al, 2008), current avail-
that the vast majority of VTEs occurred within the ini-
able evidence for the use of different anticoagulants in this
tial 6 month induction period, this may be a practical
setting, practicalities of managing the type of anticoagulant
timeframe to apply (Bradbury et al, 2018).
chosen, and situations where additional thrombotic risks
may arise, such as the need for surgery, hospitalization and The duration of thromboprohylaxis has not been ade-
other reasons for poor mobility. quately studied but at least 6 months from the initial diagno-
This approach is based on the available, albeit limited sis is preferable. If IMiD-based treatment is being continued
evidence, and would require future review and/or modifica- in the absence of high-dose dexamethasone or other
tion as new evidence becomes available. It represents the chemotherapies, continued thromboprophylaxis should be
authors opinions only and not a formal guideline or discussed with the patient. This may be with aspirin, a
recommendation. DOAC or even continued prophylactic LMWH. In all cases,
We recommend risk stratification based on the IMWG the dosing may need altering based on the patient’s initial
model (Palumbo et al, 2008), with classification of patients risks of bleeding or thrombosis, and at regular intervals. For
into low-and high-risk groups. Given that there are example, if the patient’s mobility is restricted following a
patients who may fail standard dose thromboprophylaxis, myeloma-related fracture, a higher dose may need considera-
we propose an additional group, which would be consid- tion, whereas renal impairment or thrombocytopenia may
ered as very high-risk. This would include patients who necessitate dose reduction.
have had a previous thrombosis, and those known to have
anti-thrombin deficiency. The choice of anticoagulant Special considerations
would be (see Fig 1):
Renal disease
• Low-risk patients – prophylactic LMWH based on
the current available data, but aspirin may be chosen Renal disease in multiple myeloma is common and variable,
if patients do not prefer daily injections. Once evi- ranging from patients with mild proteinuria, to the nephrotic
dence is available, aspirin may be substituted with a syndrome, with or without renal impairment. Renal failure
DOAC. occurs in 50% of patients during the course of their disease
• High-risk patients – prophylactic weight-based LMWH (Clark et al, 1999), yet patients with end-stage renal failure
or warfarin with an INR target of 2–3 are the drugs of are poorly represented in trials. It is a prothrombotic state,
choice. A prophylactic DOAC may be chosen in this with a multifactorial aetiology, due to prothrombotic changes
group in the context of a clinical trial or according to in the vascular endothelium, increased levels of inflammatory
patient wishes after appropriate counselling and consent procoagulant factors and alterations in platelet physiology
regarding the unlicensed indication. If a DOAC is pre- (Hughes et al, 2014). In patients with myeloma, and espe-
ferred, meticulous review of concomitant interacting cially those with AL amyloidosis, proteinuria can contribute
medications and contraindications such as renal failure to increased thrombotic tendency, and this is most marked
should be performed. in those with nephrotic-grade proteinuria. Bever et al (2016)

Table III. Practical issues with direct oral anticoagulant (DOAC) use in patients with myeloma.

• Dosing – We lack definitive evidence that prophylactic doses of DOACs are adequate in all patients who require thromboprophylaxis with
myeloma. This is especially of concern since it is known that prophylactic doses of low molecular weight heparin do not prevent thrombosis in
very high-risk patients. Escalated doses of DOACs are likely to reduce recurrence rates of venous thromboembolism, but possibly at the
expense of increased haemorrhagic complications. Well-designed randomized controlled trials are needed to clarify this issue.
• Drug interactions – Concomitant administration of DOACs with myeloma-specific treatment, and supportive medications (e.g. azole antifun-
gals etc.) is yet to be evaluated.
• Extremes of body weight – Steroid use may cause weight gain in myeloma patients while cachexia may be an issue in some others. There is
insufficient data for DOACs in extremes of body weight.
• Renal impairment – Optimal dosing strategies depending upon severity of renal impairment are not yet known. The various DOACs have dif-
ferent licenses based upon creatinine clearance or estimated glomerular filtration rate threshold, and levels are further affected by hypoalbu-
minaemia. This is discussed further in the renal impairment section.

548 ª 2018 British Society for Haematology and John Wiley & Sons Ltd
British Journal of Haematology, 2018, 183, 538–556
Review

Fig 1. Algorithm for risk stratification and choice of anticoagulants for patients with myeloma. DOAC, direct oral anticoagulant; INR, Interna-
tional Normalised Ratio; LMWH, low molecular weight heparin.

reviewed 929 patients with AL amyloidosis presenting to a thrombotic issues, and involve defective vascular integrity,
single-centre over a 10-year period, of whom 7% had acquired factor X deficiency and acquired von Willebrand
documented VTE. A non-significant increase in VTE risk was syndrome (Bever et al, 2016), and clearly have to be
observed if serum albumin was 3–4 g/l (HR 216, CI borne in mind when managing thrombotic risk in affected
080–581). Patients with serum albumin of less than 3 g/l, individuals.
however had a HR of 430 for the development of thrombo- Separate to proteinuria and the nephrotic syndrome,
sis, which was statistically significant (CI 160–1155, chronic renal impairment is not only a risk factor for throm-
P = 00038). Increasing levels of proteinuria were also associ- bosis, but subsequent anticoagulation also poses challenges in
ated with increased VTE risk, although this only reached this demographic, who are at greater risk of haemorrhagic
statistical significance at levels of more than 8 g/day. Of 382 complications (Parker et al, 2018). Most bleeding risk assess-
cases with nephrotic-grade proteinuria, VTE rate was 97%. ment scores include renal impairment as a risk factor. Analysis
Serum albumin level is inversely correlated with urinary pro- of over 14 000 patients enrolled in the Registro Informatizado
tein loss, not only of albumin but also important endoge- de Enfermedad Thrombo Embolica reported that, of the 20%
nous antithrombotic proteins, particularly antithrombin III who had active cancer, rates of fatal PE and fatal bleeding were
(AT). Overall, the studies on AL amyloidosis patients have increased compared to patients without malignancy, and renal
found similar rates of thrombosis to those seen in myeloma, insufficiency was identified as one of several independent risk
with high associated mortality noted (one-month mortality factors for mortality (Trujillo-Santos et al, 2009).
20% in one study) (Srkalovic et al, 2005; Halligan et al, In relation to anticoagulant treatment in patients with
2006). Given the association between nephrotic-grade pro- cancer and co-existing renal impairment, data from the
teinuria and clinically significant antithrombin deficiency, CLOT trial (Lee et al, 2003) was reviewed in patients with
assessment of urine is important in myeloma patients not moderate [creatinine clearance (CrCl) 30–60 ml/min] or
purely for the detection of M-proteins and light chains. severe (CrCl < 30 ml/min) renal impairment. Of the total
Patients with documented AT deficiency and thrombosis group, 24% had some degree of renal impairment, but
provide an even greater management challenge (they are at only 2% met the criteria for ‘severe’ impairment. Major
extremely high risk of thrombosis and may require LMWH and minor bleeding episodes occurred in 20% in the dal-
dosing alterations, as LMWH works by potentiating AT). We teparin group vs. 24% of VKA-treated patients with signif-
would suggest AT deficiency is worth excluding in those with icantly more VTE recurrences in the VTE group (17% vs.
thrombosis in the context of significant proteinuria (Kumar 3%). Although based on this data, LMWH was concluded
et al, 2012), and concurrent AL amyloidosis should be to be superior to warfarin in patients with cancer and
considered in these patients. This is of additional relevance renal impairment; patients with a creatinine three times or
due to the haemorrhagic deficiencies seen in amyloidosis. more above the upper limit of normal were excluded from the
These have been more extensively studied than the study. If LMWH is chosen in this setting, it also needs to be

ª 2018 British Society for Haematology and John Wiley & Sons Ltd 549
British Journal of Haematology, 2018, 183, 538–556
Review

borne in mind that the various LMWHs are not identical in any form e.g. in the HOKUSAI trial, 79% of edoxaban or
terms of their pharmacokinetic profiles in renal insufficiency. 113% of dalteparin patients suffered a further VTE (Raskob
Prophylactic doses of enoxaparin have been shown to accu- et al, 2018). These rates are similar to those seen in patients
mulate in patients with CrCl <30 ml/min, whereas tinzaparin with unprovoked events who discontinue anticoagulation.
and dalteparin do not. For this reason, dose reductions or Similar findings have been seen in myeloma trials. In a
monitoring might be required if enoxaparin is chosen (Atiq cohort of 256 newly diagnosed myeloma patients, where
et al, 2015). Monitoring using an anti-Xa assay calibrated to LMWH, (enoxaparin 40 mg daily) was instituted as deep
LMWH is essential and is recommended in all antithrom- venous thrombosis (DVT) prophylaxis in the thalidomide-
botic therapy guidelines, with a target trough level of 01– treated patients (n = 68), DVT recurred in four patients
03 iu/ml and peak of 05–10 iu/ml for treatment, while a (11%) (Zangari et al, 2004a), and the majority of patients in
peak level of 01–03 iu/ml would be ideal for prophylaxis Myeloma XI (over 4000 patients in all) were on thrombopro-
(Hughes et al, 2014). phyaxis at the time of VTE (Bradbury et al, 2017, 2018). This
VKAs are not renally excreted, being metabolised by cyto- means that prophylactic LMWH does not fully eliminate the
chrome P450 enzymes. However, renal failure causes downreg- thrombotic risk in myeloma. The International Society on
ulation of enzymatic activity and can therefore still alter the Thrombosis and Haemostasis (ISTH) issued recommenda-
pharmacokinetic profile of warfarin (Limdi et al, 2010). This tions for patients with malignancy who suffer recurrent
may partly explain the poor INR control seen in patients with thrombosis on anticoagulation, giving the following options
severe renal failure on warfarin (Chaaban et al, 2015; Kai et al, – switching from a VKA to LMWH, increasing the dose of
2017; Yang et al, 2017), providing additional evidence against LMWH by 20–25% if already in use, or to consider insertion
the use of VKAs in this setting. Furthermore, alongside the evi- of an inferior vena cava filter (Farge et al, 2016). These rec-
dence of inferiority to LMWH, meta-analysis of studies com- ommendations were based on results of a small, 70-patient,
paring DOACs to VKAs has shown fewer haemorrhagic retrospective cohort study of patients with VTE on anticoag-
complications with DOACs in patients with moderate renal ulation, alongside expert opinion in lieu of robust evidence.
impairment (CrCl 30–50 ml/min) (van Es et al, 2014). In this study, those on warfarin or prophylactic dose LMWH
The DOACs in common usage have different licenses with switched to therapeutic dose LMWH, whereas those on treat-
respect to treatment of VTE in renal impairment. This has ment dose LMWH received a 20–25% dose escalation for at
recently been reviewed (Parker et al, 2018). Specifically in least 4 weeks. Despite this, 86% of the cohort had a further
relation to myeloma, rivaroxaban and apixaban are highly VTE during a minimum of only 3 months follow-up, and it
protein-bound in circulation, and therefore affected by should be noted that the group’s median survival was less
hypoalbuminaemia, which may develop in renal failure than 1 year (Carrier et al, 2009). General measures, including
patients with proteinuria or nutritional deficiency. Given that assessment of compliance to pharmacological thrombopro-
patients with renal impairment may require other drugs in phylaxis, consideration of other prothrombotic conditions
addition to those specific for myeloma, and the fact that all (e.g. heparin-induced thrombocytopenia) and presence of
four DOACs are, to some extent, metabolized by cytochrome mechanical compression from tumour masses are also rou-
P450 enzymes, careful drug selection is required. If a DOAC tinely advised (Romualdi & Ageno, 2016). For those on oral
is chosen, close attention to drug monitoring may be neces- agents, GI absorption should be reviewed alongside possible
sary in this setting (Parker et al, 2018). Betrixaban, the most interactions with other medications being administered.
recent anti-Xa inhibitor on the market, has the lowest renal Most of the available information with respect to ‘failure
clearance at only 11% and is not metabolized by the cyto- of anticoagulation’ pertains to VKAs and LMWH. The
chrome 450 system. It has been approved by the US Food HOKUSAI (Raskob et al, 2018) and SELECT-D (Young et al,
and Drug Administration for VTE prophylaxis in those with 2018) trials provide evidence in support of DOAC use in the
CrCl ≥ 15 ml/min without severe liver impairment (Huis- therapeutic setting but are not specific to myeloma, nor VTE
man & Klok, 2018). Betrixaban has not yet been approved recurrence. 9% of the HOKUSAI group had a prior history
for prophylaxis by the European Medicines Agancy, and of VTE, but whether this was in the context of malignancy is
while it offers a potentially attractive future option for use in not known, and patients on therapeutic anticoagulation at
patients with renal failure, it should be noted that its long time of thrombosis are specifically excluded. A case report of
half-life (approximately 37 h) may be of concern in those successful management of a myeloma patient with VTE on
with a high bleeding risk. prophylactic aspirin and erratic INR control on warfarin was
recently published (Oka et al, 2017), and it is likely that
DOACs are being used more frequently in clinical practice in
Recurrent thrombosis
this setting.
There is a paucity of data on how best to manage recurrent The ISTH set up an international registry to increase the
VTE in cancer patients, and even less in those with myeloma available data regarding this issue. A cohort of 212 cancer
specifically. What is abundantly clear is that patients with patients with a recurrence of VTE despite anticoagulation
cancer have high recurrence rates despite anticoagulation of with unfractionated heparin, LMWH, fondaparinux or a

550 ª 2018 British Society for Haematology and John Wiley & Sons Ltd
British Journal of Haematology, 2018, 183, 538–556
Review

VKA were monitored for 3 months. A small proportion (8%) remained hypercoagulable, suggesting that a significant pro-
had leukaemia, lymphoma or myeloma. 70% of the cohort had portion of the patients assessed had evidence of heparin
been therapeutically or supra-therapeutically anticoagulated at resistance. If this finding were reproduced in a larger sample
the time of VTE. 11% had a further event during follow-up, size, particularly in patients receiving LMWH, it would pro-
8% had major bleeding and 27% died. Increasing anticoagula- vide concerning evidence of lack of efficacy of current
tion intensity had no beneficial effect on recurrence rate, or a thromboprophylactic measures, at least in a subset of
significant association with increased haemorrhagic complica- patients, which may be contributing to ongoing rates of VTE
tions. As seen previously, rate of recurrence was higher with despite anticoagulation. It is likely that anti-Xa levels may
VKAs than LMWH (Schulman et al, 2015). not be fully reflective of global haemostasis in patients with
Evaluation of whether anticoagulation is in the therapeutic recurrent VTE despite seemingly therapeutic LMWH antico-
range is traditionally achieved using standard laboratory agulation, and more information is required regarding the
investigations e.g. the INR for VKA and anti-Xa levels for practical utility of global tests of haemostasis in managing
LMWH. However, use of global measures of coagulation these challenging patients.
may provide further, relevant information regarding the The findings from the ISTH registry, along with evidence of
patient’s overall haemostatic state. One group used throm- heparin resistance in a significant minority of patients, suggests
boelastography (TEG), thrombin generation test (TGT), that therapeutic doses of LMWH do not offer sufficient pro-
thrombodynamics (TD) and some standard laboratory tection for a subset of myeloma patients, and that mere dose
parameters, such as D-dimer levels, fibrinogen, activated par- escalation may not be an adequate strategy. Our approach in
tial prothrombin time (APTT) and prothrombin time (PT) these patients is summarized in Fig 2, with suggestions for
in MM patients at diagnosis, during stem cell mobilisation patients who may have been on warfarin, LMWH or a DOAC.
and in remission (Gracheva et al, 2015). D-dimer was signifi- Given that no better approaches are currently available in
cantly elevated, and global haemostasis tests showed patients patients on therapeutic doses of LMWH who developed recur-
to be hypercoagulable compared with normal controls, rent clots; we would choose the split dose strategy (i.e. the full
whether at diagnosis or in remission. However, there was sig- weight-based dose divided into two doses, given at 12-h inter-
nificant heterogeneity of results, with some patients also vals) in addition to modifying thrombotic risk factors (mobi-
demonstrated to be hypocoagulable. Patients undergoing lization, temporary interruption of IMiDs etc.). Some of the
stem cell mobilisation received infusional unfractionated hep- rare causes of recurrent thrombosis on anticoagulation include
arin as VTE prophylaxis. Importantly, in 22%, at least 2 of antiphospholipid syndrome, vasculitis, vascular malformations
the global tests of haemostasis showed no hypocoagulable and hyperhomocysteanemia, which require due consideration
features despite APTT levels in the target range, and some (Thachil, 2012).

Fig 2. Management of recurrent thrombosis while on anticoagulant therapy in patients with myeloma. DOAC, direct oral anticoagulant; LMWH,
low molecular weight heparin.

ª 2018 British Society for Haematology and John Wiley & Sons Ltd 551
British Journal of Haematology, 2018, 183, 538–556
Review

Spinal cord compression low-dose aspirin if at low risk of thrombosis and LMWH
if at high-risk, as per the IMWG 2008 model (Palumbo
Spinal cord compression may occur in patients with mye-
et al, 2008). If future models allow improved identification
loma as a result of vertebral lytic lesions or growth of com-
of those at the highest risk, either through use of novel
pressive soft tissue masses. VTE risk in these patients is
thrombosis biomarkers, point-of-care testing, or presence
markedly increased by the combination of immobility, and
of numerous clinical risk factors, thromboprophylaxis mea-
need for surgical interventions, venous stasis due to loss of
sures will need to adapt to reflect this, following confirma-
contraction of lower limb muscles, inflammatory prothrom-
tion of safety and efficacy in prospective randomized
botic changes stimulated by tissue damage, alongside all the
controlled trials. Data from the HOKUSAI trial suggests
usual myeloma-related risk factors. Reported rates of VTE in
that DOACs may possibly confer greater protection than
spinal cord injury are as high as 50% (Teasell et al, 2009).
LMWH (Raskob et al, 2018), based on the reduction of
Specific guidance relating to spinal cord compression in mye-
VTE seen, however this evidence does not pertain to pri-
loma patients is lacking, however the NICE recommendations
mary VTE prevention, nor is it specific to myeloma, and
on management of metastatic spinal cord compression suggest
further information is needed in order to improve man-
mechanical thromboprophylaxis with thigh-high graduated
agement of these patients. Similarly, more work can only
compression stockings, unless contraindication by, for exam-
provide us with better knowledge on how to deal with the
ple, sensory loss, or intermittent pneumatic compression
special situations of renal impairment and spinal cord
devices with concomitant LMWH prophylaxis. Duration of
compression, where the risk of both thrombosis and haem-
thromboprophylaxis should be based on individual risk assess-
orrhage are increased and carefully weighing the risks and
ment, overall clinical condition, and whether the patient has
benefits are crucial.
regained normal mobility (NICE 2008).

Future directions Author contributions

Management of multiple myeloma has developed signifi- DS performed the literature review, wrote the first draft and
cantly in recent years, with patients surviving for many made the edits for the final version. AR and CB critically
years more than previously reported (Kumar et al, 2008). reviewed the manuscript. JT conceived the review and criti-
However, in this era of novel agents, patients are still cally reviewed the manuscript. All the authors approved the
dying from VTE-related complications and, despite current final version submitted.
thromboprophylactic approaches, a significant proportion
of patients are still expected to develop thromboembolic
Conflicts of interest
disease as a sequelae of myeloma or its treatment, with
those at highest risk not fully protected by the pharmaco- JT has received honoraria from BMS-Pfizer, Boehringer,
logical agents administered as part of modern treatment Bayer and Daichii Sankyo, whose products are mentioned in
protocols. The current recommendation for thrombopro- the paper. CB has received honoraria from BMS-Pfizer and
phylaxis in patients receiving IMiD-based regimens is Novartis.

References review on the accumulation of prophylactic J., Epstein, J., Spoon, D., Ayers, D., Cheson, B.
dosages of low-molecular-weight heparins & Crowley, J. (1999) Total therapy with tandem
Agnelli, G., Buller, H.R., Cohen, A., Curto, M., (LMWHs) in patients with renal insufficiency. transplants for newly diagnosed multiple mye-
Gallus, A.S., Johnson, M., Porcari, A., Raskob, European Journal of Clinical Pharmacology, 71, loma. Blood, 93, 55–65.
G.E. & Weitz, J.I. (2013) Apixaban for extended 921–929. Barlogie, B., Desikan, R., Eddlemon, P., Spencer,
treatment of venous thromboembolism. New Auwerda, J.J., Sonneveld, P., de Maat, M.P. & Lee- T., Zeldis, J., Munshi, N., Badros, A., Zangari,
England Journal of Medicine, 368, 699–708. beek, F.W. (2007) Prothrombotic coagulation M., Anaissie, E., Epstein, J., Shaughnessy, J.,
Al-Ani, F., Bermejo, J.M., Mateos, M.V. & Lou- abnormalities in patients with newly diagnosed Ayers, D., Spoon, D. & Tricot, G. (2001)
zada, M. (2016) Thromboprophylaxis in multi- multiple myeloma. Haematologica, 92, 279–280. Extended survival in advanced and refractory
ple myeloma patients treated with lenalidomide Ay, C., Simanek, R., Vormittag, R., Dunkler, D., multiple myeloma after single-agent thalido-
- A systematic review. Thrombosis Research, 141, Alguel, G., Koder, S., Kornek, G., Marosi, C., mide: identification of prognostic factors in a
84–90. Wagner, O., Zielinski, C. & Pabinger, I. (2008) phase 2 study of 169 patients. Blood, 98, 492–
Ambrus, J.L., Ambrus, C.M., Mink, I.B. & Pickren, High plasma levels of soluble P-selectin are pre- 494.
J.W. (1975) Causes of death in cancer patients. dictive of venous thromboembolism in cancer Barlogie, B., Tricot, G., Anaissie, E., Shaughnessy,
Journal of Medicine, 6, 61–64. patients: results from the Vienna Cancer and J., Rasmussen, E., van Rhee, F., Fassas, A., Zan-
Amrani, D.L. (1990) Regulation of fibrinogen Thrombosis Study (CATS). Blood, 112, 2703– gari, M., Hollmig, K., Pineda-Roman, M., Lee,
biosynthesis: glucocorticoid and interleukin-6 2708. C., Talamo, G., Thertulien, R., Kiwan, E.,
control. Blood Coagulation & Fibrinolysis, 1, Barlogie, B., Jagannath, S., Desikan, K.R., Mattox, Krishna, S., Fox, M. & Crowley, J. (2006)
443–446. S., Vesole, D., Siegel, D., Tricot, G., Munshi, N., Thalidomide and hematopoietic-cell transplanta-
Atiq, F., van den Bemt, P.M., Leebeek, F.W., van Fassas, A., Singhal, S., Mehta, J., Anaissie, E., tion for multiple myeloma. New England Journal
Gelder, T. & Versmissen, J. (2015) A systematic Dhodapkar, D., Naucke, S., Cromer, J., Sawyer, of Medicine, 354, 1021–1030.

552 ª 2018 British Society for Haematology and John Wiley & Sons Ltd
British Journal of Haematology, 2018, 183, 538–556
Review

Baz, R., Li, L., Kottke-Marchant, K., Srkalovic, G., autologous stem cell transplantation for multiple van Es, N., Coppens, M., Schulman, S., Middel-
McGowan, B., Yiannaki, E., Karam, M.A., Fai- myeloma. Haematologica, 89, 826–831. dorp, S. & Buller, H.R. (2014) Direct oral anti-
man, B., Jawde, R.A., Andresen, S., Zeldis, J. & Chaaban, A., Gebran, N., Abdelmegid, K., Abou- coagulants compared with vitamin K antagonists
Hussein, M.A. (2005) The role of aspirin in the chacra, S., Soliman, K., Agha, L. & Butt, L for acute venous thromboembolism: evidence
prevention of thrombotic complications of (2015) Effectiveness and safety of warfarin use from phase 3 trials. Blood, 124, 1968–1975.
thalidomide and anthracycline-based chemother- for cardiac indications in hemodialysis patients. Esmon, C.T. (2001) Protein C anticoagulant path-
apy for multiple myeloma. Mayo Clinic Proceed- Nephrology Dialysis Transplantation, 30, 563– way and its role in controlling microvascular
ings, 80, 1568–1574. 564. thrombosis and inflammation. Critical Care
Baz, R., Walker, E., Karam, M.A., Choueiri, T.K., Chalayer, E., Chapelle, C., Leleu, X., Elalamy, I., Medicine, 29, S48–S51; discussion 51-42.
Jawde, R.A., Bruening, K., Reed, J., Faiman, Laporte, S. & Tardy, B. (2016a) Does the choice Falanga, A. & Marchetti, M. (2009) Venous
B., Ellis, Y., Brand, C., Srkalovic, G., of thrombotic prophylactic drug depend on the thromboembolism in the hematologic malignan-
Andresen, S., Knight, R., Zeldis, J. & Hussein, known risk factors of patients with multiple cies. Journal of Clinical Oncology, 27, 4848–4857.
M.A. (2006) Lenalidomide and pegylated lipo- myeloma in clinical practice? Thrombosis Farge, D., Bounameaux, H., Brenner, B., Cajfinger,
somal doxorubicin-based chemotherapy for Research, 143, 101–102. F., Debourdeau, P., Khorana, A.A., Pabinger, I.,
relapsed or refractory multiple myeloma: safety Chalayer, E., Bourmaud, A., Tinquaut, F., Chau- Solymoss, S., Douketis, J. & Kakkar, A. (2016)
and efficacy. Annals of Oncology, 17, 1766– vin, F. & Tardy, B. (2016b) Cost-effectiveness International clinical practice guidelines includ-
1771. analysis of low-molecular-weight heparin versus ing guidance for direct oral anticoagulants in
Bellotti, V., Gamba, G., Merlini, G., Montani, N., aspirin thromboprophylaxis in patients newly the treatment and prophylaxis of venous throm-
Bucciarelli, E., Stoppini, M. & Ascari, E. (1989) diagnosed with multiple myeloma. Thrombosis boembolism in patients with cancer. The Lancet
Study of three patients with monoclonal gam- Research, 145, 119–125. Oncology, 17, e452–e466.
mopathies and ‘lupus-like’ anticoagulants. Bri- Clark, A.D., Shetty, A. & Soutar, R. (1999) Renal Gado, K., Domjan, G., Hegyesi, H. & Falus, A.
tish Journal of Haematology, 73, 221–227. failure and multiple myeloma: pathogenesis (2000) Role of INTERLEUKIN-6 in the patho-
Bennett, C.L., Schumock, G.T., Desai, A.A., and treatment of renal failure and manage- genesis of multiple myeloma. Cell Biology Inter-
Kwaan, H.C., Raisch, D.W., Newlin, R. & Sta- ment of underlying myeloma. Blood Reviews, national, 24, 195–209.
dler, W. (2002) Thalidomide-associated deep 13, 79–90. van Giezen, J.J. & Jansen, J.W. (1992) Correlation
vein thrombosis and pulmonary embolism. Corso, A., Lorenzi, A., Terulla, V., Airo, F., Varet- of in vitro and in vivo decreased fibrinolytic
American Journal of Medicine, 113, 603–606. toni, M., Mangiacavalli, S., Zappasodi, P., Rus- activity caused by dexamethasone. Annals of the
Bever, K.M., Masha, L.I., Sun, F., Stern, L., Havasi, coni, C. & Lazzarino, M. (2004) Modification of New York Academy of Sciences, 667, 199–201.
A., Berk, J.L., Sanchorawala, V., Seldin, D.C. & thrombomodulin plasma levels in refractory Gracheva, M.A., Urnova, E.S., Sinauridze, E.I.,
Sloan, J.M. (2016) Risk factors for venous myeloma patients during treatment with Tarandovskiy, I.D., Orel, E.B., Poletaev, A.V.,
thromboembolism in immunoglobulin light thalidomide and dexamethasone. Annals of Mendeleeva, L.P., Ataullakhanov, F.I. & Baland-
chain amyloidosis. Haematologica, 101, 86–90. Hematology, 83, 588–591. ina, A.N. (2015) Thromboelastography, throm-
Blom, J.W., Doggen, C.J., Osanto, S. & Rosendaal, Dimopoulos, M.A., Zervas, K., Kouvatseas, G., bin generation test and thrombodynamics reveal
F.R. (2005) Malignancies, prothrombotic muta- Galani, E., Grigoraki, V., Kiamouris, C., Verves- hypercoagulability in patients with multiple
tions, and the risk of venous thrombosis. JAMA, sou, E., Samantas, E., Papadimitriou, C., Econo- myeloma. Leukaemia & Lymphoma, 56, 3418–
293, 715–722. mou, O., Gika, D., Panayiotidis, P., Christakis, 3425.
Bradbury, C.A., Jenner, M.W., Striha, A., Cook, I. & Anagnostopoulos, N. (2001) Thalidomide Guo, L., Tong, D., Yu, M., Zhang, Y., Li, T.,
G., Pawlyn, C., Jones, J.R., Cairns, D., Hocka- and dexamethasone combination for refractory Wang, C., Zhou, P., Jin, J., Li, B., Liu, Y., Liu,
day, A., Paterson, A., Drayson, M.T., Owen, multiple myeloma. Annals of Oncology, 12, 991– R., Novakovic, V.A., Dong, Z., Tian, Y., Kou, J.,
R.G., Kaiser, M.F., Gregory, W.M., Davies, F.E., 995. Bi, Y., Zhou, J. & Shi, J. (2018) Phosphatidylser-
Morgan, G.J. & Jackson, G.H. (2017) Throm- Dimopoulos, M., Spencer, A., Attal, M., Prince, ine-exposing cells contribute to the hypercoagu-
botic events in patients with myeloma treated H.M., Harousseau, J.L., Dmoszynska, A., San lable state in patients with multiple myeloma.
with immunomodulatory drugs; results of the Miguel, J., Hellmann, A., Facon, T., Foa, R., International Journal of Oncology, 52, 1981–1990.
myeloma XI study. Blood, 130, 553. Corso, A., Masliak, Z., Olesnyckyj, M., Yu, Z., Halligan, C.S., Lacy, M.Q., Vincent Rajkumar, S.,
Bradbury, C.A., Jenner, M., Striha, A., Cook, G., Patin, J., Zeldis, J.B. & Knight, R.D. (2007) Dispenzieri, A., Witzig, T.E., Lust, J.A., Fonseca,
Pawlyn, C., Jones, J., Cairns, D., Hockaday, A., Lenalidomide plus dexamethasone for relapsed R., Gertz, M.A., Kyle, R.A. & Pruthi, R.K.
Paterson, A., Drayson, M., Owen, R., Kaiser, M., or refractory multiple myeloma. New England (2006) Natural history of thromboembolism in
Gregory, W., Davies, F., Morgan, G. & Jackson, Journal of Medicine, 357, 2123–2132. AL amyloidosis. Amyloid, 13, 31–36.
G. (2018) Thrombotic events in patients with Donati, M.B. (1994) Cancer and thrombosis. Hae- Hiroi, T., Deming, C.B., Zhao, H., Hansen, B.S.,
Myeloma treated with Immunomodulatory mostasis, 24, 128–131. Arkenbout, E.K., Myers, T.J., McDevitt, M.A. &
drugs; results of the myeloma XI study. British Dong, R., Ma, G., Zhang, S., Gao, Y., Liu, S. & Su, Rade, J.J. (2009) Proteasome inhibitors enhance
Journal of Haematology, 181, p19. abstract Y. (2018) Simvastatin reverses multiple myeloma endothelial thrombomodulin expression via
BSH18-OR-014. serum-induced prothrombotic phenotype in induction of Kruppel-like transcription factors.
Carrier, M., Le Gal, G., Cho, R., Tierney, S., endothelial cells via ERK 1/2 signalling pathway. Arteriosclerosis, Thrombosis, and Vascular Biology,
Rodger, M. & Lee, A.Y. (2009) Dose escalation Blood Coagulation & Fibrinolysis, 29, 501–508. 29, 1587–1593.
of low molecular weight heparin to manage Eby, C. (2009) Pathogenesis and management of Hughes, S., Szeki, I., Nash, M.J. & Thachil, J.
recurrent venous thromboembolic events bleeding and thrombosis in plasma cell dyscra- (2014) Anticoagulation in chronic kidney dis-
despite systemic anticoagulation in cancer sias. British Journal of Haematology, 145, 151– ease patients-the practical aspects. Clinical Kid-
patients. Journal of Thrombosis and Haemosta- 163. ney Journal, 7, 442–449.
sis, 7, 760–765. Elice, F., Fink, L., Tricot, G., Barlogie, B. & Zan- Huisman, M.V. & Klok, F.A. (2018) Pharmacologi-
Cavo, M., Zamagni, E., Tosi, P., Cellini, C., Can- gari, M. (2006) Acquired resistance to activated cal properties of betrixaban. European Heart
gini, D., Tacchetti, P., Testoni, N., Tonelli, M., protein C (aAPCR) in multiple myeloma is a Journal Supplements, 20, E12–E15.
de Vivo, A., Palareti, G., Tura, S. & Baccarani, transitory abnormality associated with an Johannesdottir, S.A., Horvath-Puh o, E., Dekkers,
M. (2004) First-line therapy with thalidomide increased risk of venous thromboembolism. Bri- O.M., Cannegieter, SC., Jørgensen, JO., Ehren-
and dexamethasone in preparation for tish Journal of Haematology, 134, 399–405. stein, V., Vandenbroucke, JP., Pedersen, L. &

ª 2018 British Society for Haematology and John Wiley & Sons Ltd 553
British Journal of Haematology, 2018, 183, 538–556
Review

Sørensen, HT (2013) Use of glucocorticoids and Landgren, O. (2010) Arterial and venous throm- Beasley, T.M. (2010) Warfarin dosing in patients
risk of venous thromboembolism: a nationwide bosis in monoclonal gammopathy of undeter- with impaired kidney function. American Jour-
population-based case-control study. JAMA mined significance and multiple myeloma: a nal of Kidney Diseases, 56, 823–831.
Internal Medicine, 173, 743–752. population-based study. Blood, 115, 4991–4998. Lonial, S., Richardson, P.G., San Miguel, J., Sonn-
Kai, B., Bogorad, Y., Nguyen, L.N., Yang, S.J., Kumar, S.K., Rajkumar, S.V., Dispenzieri, A., Lacy, eveld, P., Schuster, M.W., Blade, J., Cavenagh,
Chen, W., Spencer, H.T., Shen, A.Y. & Lee, M.S. M.Q., Hayman, S.R., Buadi, F.K., Zeldenrust, J., Rajkumar, S.V., Jakubowiak, A.J., Esseltine,
(2017) Warfarin use and the risk of mortality, S.R., Dingli, D., Russell, S.J., Lust, J.A., Greipp, D.L., Anderson, K.C. & Harousseau, J.L. (2008)
stroke, and bleeding in hemodialysis patients P.R., Kyle, R.A. & Gertz, M.A. (2008) Improved Characterisation of haematological profiles and
with atrial fibrillation. Heart Rhythm: the Official survival in multiple myeloma and the impact of low risk of thromboembolic events with borte-
Journal of the Heart Rhythm Society, 14, 645– novel therapies. Blood, 111, 2516–2520. zomib in patients with relapsed multiple mye-
651. Kumar, S., Chapagain, A., Nitsch, D. & Yaqoob, loma. British Journal of Haematology, 143, 222–
Katodritou, E., Vadikolia, C., Lalagianni, C., Kot- M.M. (2012) Proteinuria and hypoalbuminemia 229.
sopoulou, M., Papageorgiou, G., Kyrtsonis, are risk factors for thromboembolic events in Man, L., Morris, A., Brown, J., Palkimas, S. &
M.C., Matsouka, P., Giannakoulas, N., Kyriakou, patients with idiopathic membranous nephropa- Davidson, K. (2017) Use of direct oral anticoag-
D., Karras, G., Anagnostopoulos, N., Michali, E., thy: an observational study. BMC Nephrology, ulants in patients on immunomodulatory
Briasoulis, E., Hatzimichael, E., Spanoudakis, E., 13, 107. agents. Journal of Thrombosis and Thrombolysis,
Zikos, P., Tsakiridou, A., Tsionos, K., Anargy- Lacy, M.Q., Hayman, S.R., Gertz, M.A., Dispen- 44, 298–302.
rou, K., Symeonidis, A., Maniatis, A. & Terpos, zieri, A., Buadi, F., Kumar, S., Greipp, P.R., McLawhorn, A.S., Beathe, J., YaDeau, J., Buschi-
E. (2014) “Real-world” data on the efficacy and Lust, J.A., Russell, S.J., Dingli, D., Kyle, R.A., azzo, V., Purdue, P.E., Ma, Y., Sculco, T.P. &
safety of lenalidomide and dexamethasone in Fonseca, R., Bergsagel, P.L., Roy, V., Mikhael, Jules-Elysee, K. (2015) Effects of steroids on
patients with relapsed/refractory multiple mye- J.R., Stewart, A.K., Laumann, K., Allred, J.B., thrombogenic markers in patients undergoing
loma who were treated according to the stan- Mandrekar, S.J. & Rajkumar, S.V. (2009) unilateral total knee arthroplasty: a prospective,
dard clinical practice: a study of the Greek Pomalidomide (CC4047) plus low-dose dexam- double-blind, randomized controlled trial. Jour-
Myeloma Study Group. Annals of Hematology, ethasone as therapy for relapsed multiple mye- nal of Orthopaedic Research, 33, 412–416.
93, 129–139. loma. Journal of Clinical Oncology, 27, 5008– Mechtcheriakova, D., Wlachos, A., Holzmuller, H.,
Kerachian, M.A., Cournoyer, D., Harvey, E.J., 5014. Binder, B.R. & Hofer, E. (1999) Vascular
Chow, T.Y., Neagoe, P.E., Sirois, M.G. & Seguin, Larocca, A., Cavallo, F., Bringhen, S., Di Rai- endothelial cell growth factor-induced tissue fac-
C. (2009) Effect of high-dose dexamethasone on mondo, F., Falanga, A., Evangelista, A., Cavalli, tor expression in endothelial cells is mediated by
endothelial haemostatic gene expression and M., Stanevsky, A., Corradini, P., Pezzatti, S., EGR-1. Blood, 93, 3811–3823.
neutrophil adhesion. Journal of Steroid Biochem- Patriarca, F., Cavo, M., Peccatori, J., Catalano, Menon, S.P., Rajkumar, S.V., Lacy, M., Falco, P. &
istry and Molecular Biology, 116, 127–133. L., Carella, A.M., Cafro, A.M., Siniscalchi, A., Palumbo, A. (2008) Thromboembolic events
Khorana, A.A., Francis, C.W., Culakova, E., Kud- Crippa, C., Petrucci, M.T., Yehuda, D.B., Beg- with lenalidomide-based therapy for multiple
erer, N.M. & Lyman, G.H. (2007) Thromboem- giato, E., Di Toritto, T.C., Boccadoro, M., Nag- myeloma. Cancer, 112, 1522–1528.
bolism is a leading cause of death in cancer ler, A. & Palumbo, A. (2012) Aspirin or Morgan, G.J., Schey, S.A., Wu, P., Srikanth, M.,
patients receiving outpatient chemotherapy. enoxaparin thromboprophylaxis for patients Phekoo, K.J., Jenner, M. & Davies, F.E. (2007)
Journal of Thrombosis and Haemostasis, 5, 632– with newly diagnosed multiple myeloma treated Lenalidomide (Revlimid), in combination with
634. with lenalidomide. Blood, 119, 933–939; quiz cyclophosphamide and dexamethasone (RCD),
Khorana, A.A., Kamphuisen, P.W., Meyer, G., 1093. is an effective and tolerated regimen for mye-
Bauersachs, R., Janas, M.S., Jarner, M.F. & Lee, Lee, A.Y., Levine, M.N., Baker, R.I., Bowden, C., loma patients. British Journal of Haematology,
A.Y. (2017) Tissue factor as a predictor of recur- Kakkar, A.K., Prins, M., Rickles, F.R., Julian, 137, 268–269.
rent venous thromboembolism in malignancy: J.A., Haley, S., Kovacs, M.J. & Gent, M. (2003) Nayak, L., Shi, H., Atkins, G.B., Lin, Z., Schmaier,
biomarker analyses of the CATCH trial. Journal Low-molecular-weight heparin versus a cou- A.H. & Jain, M.K. (2014) The thromboprotec-
of Clinical Oncology, 35, 1078–1085. marin for the prevention of recurrent venous tive effect of bortezomib is dependent on the
Knop, S., Gerecke, C., Topp, M.S., Liebisch, P., thromboembolism in patients with cancer. New transcription factor Kruppel-like factor 2
Hess, G., Kotkiewitz, S., Einsele, H. & Bargou, England Journal of Medicine, 349, 146–153. (KLF2). Blood, 123, 3828–3831.
R. (2006) Lenalidomide (RevlimidTM), adri- Leleu, X., Rodon, P., Hulin, C., Daley, L., Dauriac, NICE. (2008) Metastatic spinal cord compression in
amycin and dexamethasone chemotherapy C., Hacini, M., Decaux, O., Eisemann, J.C., adults: risk assessment, diagnosis and manage-
(RAD) is safe and effective in treatment of Fitoussi, O., Lioure, B., Voillat, L., Slama, B., Al ment. Clinical guideline [CG75]. National Insti-
relapsed multiple myeloma - first results of a Jijakli, A., Benramdane, R., Chaleteix, C., Cost- tute for Health and Care Excellence, London,
german multicenter phase I/II trial. Blood, 108, ello, R., Thyss, A., Mathiot, C., Boyle, E., Maloi- UK. https://www.nice.org.uk/guidance/cg75.
408–408. sel, F., Stoppa, A.M., Kolb, B., Michallet, M., NICE. (2016) Myeloma: diagnosis and manage-
Kotlin, R., Sobotkova, A., Riedel, T., Salaj, P., Sut- Lamblin, A., Natta, P., Facon, T., Elalamy, I., ment NICE guideline [NG35]. National Institute
tnar, J., Reicheltova, Z., Majek, P., Khaznadar, Fermand, J.P. & Moreau, P. (2013) MELISSE, a for Health and Care Excellence, London, UK.
T. & Dyr, J.E. (2008) Acquired dysfibrinogene- large multicentric observational study to deter- https://www.nice.org.uk/guidance/ng35
mia secondary to multiple myeloma. Acta Hae- mine risk factors of venous thromboembolism NICE. (2018) Venous thromboembolism in over
matologica, 120, 75–81. in patients with multiple myeloma treated with 16s: reducing the risk of hospital-acquired deep
Kristinsson, S.Y., Fears, T.R., Gridley, G., Tures- immunomodulatory drugs. Thrombosis and Hae- vein thrombosis or pulmonary embolism. NICE
son, I., Mellqvist, U.H., Bjorkholm, M. & Land- mostasis, 110, 844–851. guideline [NG89]. National Institute for Health
gren, O. (2008) Deep vein thrombosis after Li, W., Garcia, D., Cornell, R.F., Gailani, D., Lau- and Care Excellence, London, UK. https://www.
monoclonal gammopathy of undetermined sig- bach, J., Maglio, M.E., Richardson, P.G. & Mos- nice.org.uk/guidance/ng89.
nificance and multiple myeloma. Blood, 112, lehi, J. (2017) Cardiovascular and thrombotic Oka, S., Takeuchi, S., Shiragami, H., Hamahata, K.
3582–3586. complications of novel multiple myeloma thera- & Nohgawa, M. (2017) Successful management
Kristinsson, S.Y., Pfeiffer, R.M., Bjorkholm, M., pies: a review. JAMA Oncology, 3, 980–988. of venous thromboembolism with apixaban in a
Goldin, L.R., Schulman, S., Blimark, C., Mel- Limdi, N.A., Limdi, M.A., Cavallari, L., Anderson, multiple myeloma patient on lenalidomide ther-
lqvist, U.H., Wahlin, A., Turesson, I. & A.M., Crowley, M.R., Baird, M.F., Allon, M. & apy. Rinsho Ketsueki, 58, 37–41.

554 ª 2018 British Society for Haematology and John Wiley & Sons Ltd
British Journal of Haematology, 2018, 183, 538–556
Review

O’Kane, M.J., Wisdom, G.B., Desai, Z.R. & Arch- valvular atrial fibrillation too risky? British Jour- patients with multiple myeloma. Blood, 124,
bold, G.P. (1994) Inhibition of fibrin monomer nal of Haematology, 181, 725–736. 2176–2176.
polymerisation by myeloma immunoglobulin. Rajkumar, S.V. & Blood, E. (2006) Lenalidomide Sanfilippo, K.M., Luo, S., Carson, K.R. & Gage,
Journal of Clinical Pathology, 47, 266–268. and venous thrombosis in multiple myeloma. B.F. (2017) Aspirin may be inadequate throm-
Paludo, J., Mikhael, J.R., LaPlant, B.R., Halvorson, New England Journal of Medicine, 354, 2079– boprophylaxis in multiple myeloma. Blood, 130,
A.E., Kumar, S., Gertz, M.A., Hayman, S.R., 2080. 3419–3419.
Buadi, F.K., Dispenzieri, A., Lust, J.A., Kapoor, Rajkumar, S.V., Hayman, S., Gertz, M.A., Dispen- Sanfilippo, K.M., Wang, T.F., Luo, S., Carson,
P., Leung, N., Russell, S.J., Dingli, D., Go, R.S., zieri, A., Lacy, M.Q., Greipp, P.R., Geyer, S., K.R., Thomas, T.S. & Gage, B. (2018) Prediction
Lin, Y., Gonsalves, W.I., Fonseca, R., Bergsagel, Iturria, N., Fonseca, R., Lust, J.A., Kyle, R.A. & of venous thromboembolism (VTE) in multiple
P.L., Roy, V., Sher, T., Chanan-Khan, A.A., Witzig, T.E. (2002) Combination therapy with myeloma (MM): myeloma clot score (MCS).
Ailawadhi, S., Stewart, A.K., Reeder, C.B., thalidomide plus dexamethasone for newly diag- Journal of Clinical Oncology, 36(suppl), abstr
Richardson, P.G., Rajkumar, S.V. & Lacy, M.Q. nosed myeloma. Journal of Clinical Oncology, 20, 6585.
(2017) Pomalidomide, bortezomib, and dexam- 4319–4323. Schey, S.A., Fields, P., Bartlett, J.B., Clarke, I.A.,
ethasone for patients with relapsed lenalido- Rajkumar, S.V., Hayman, S.R., Lacy, M.Q., Dis- Ashan, G., Knight, R.D., Streetly, M. & Dal-
mide-refractory multiple myeloma. Blood, 130, penzieri, A., Geyer, S.M., Kabat, B., Zeldenrust, gleish, A.G. (2004) Phase I study of an
1198–1204. S.R., Kumar, S., Greipp, P.R., Fonseca, R., Lust, immunomodulatory thalidomide analog, CC-
Palumbo, A. & Palladino, C. (2012) Venous and J.A., Russell, S.J., Kyle, R.A., Witzig, T.E. & 4047, in relapsed or refractory multiple mye-
arterial thrombotic risks with thalidomide: evi- Gertz, M.A. (2005) Combination therapy with loma. Journal of Clinical Oncology, 22, 3269–
dence and practical guidance. Therapeutic lenalidomide plus dexamethasone (Rev/Dex) for 3276.
Advances in Drug Safety, 3, 255–266. newly diagnosed myeloma. Blood, 106, 4050– Schoen, M.W., Luo, S., Gage, B., Carson, K.R. &
Palumbo, A., Bertola, A., Falco, P., Rosato, R., 4053. Sanfilippo, K.M. (2018) Association of venous
Cavallo, F., Giaccone, L., Bringhen, S., Musto, Rajkumar, S.V., Blood, E., Vesole, D., Fonseca, R. thromboembolism with increased mortality in
P., Pregno, P., Caravita, T., Ciccone, G. & Boc- & Greipp, P.R. (2006) Phase III clinical trial of patients with multiple myeloma. Journal of Clin-
cadoro, M. (2004) Efficacy of low-dose thalido- thalidomide plus dexamethasone compared with ical Oncology, 36, 8051–8051.
mide and dexamethasone as first salvage dexamethasone alone in newly diagnosed multi- Schulman, S., Zondag, M., Linkins, L., Pasca, S.,
regimen in multiple myeloma. The Hematology ple myeloma: a clinical trial coordinated by the Cheung, Y.W., de Sancho, M., Gallus, A.,
Journal, 5, 318–324. Eastern Cooperative Oncology Group. Journal of Lecumberri, R., Molnar, S., Ageno, W., Le Gal,
Palumbo, A., Falco, P., Corradini, P., Falcone, A., Clinical Oncology, 24, 431–436. G., Falanga, A., Hulegardh, E., Ranta, S., Kam-
Di Raimondo, F., Giuliani, N., Crippa, C., Cic- Rajkumar, S.V., Jacobus, S., Callander, N.S., Fon- phuisen, P., Debourdeau, P., Rigamonti, V.,
cone, G., Omede, P., Ambrosini, M.T., Gay, F., seca, R., Vesole, D.H., Williams, M.E., Abonour, Ortel, T.L. & Lee, A. (2015) Recurrent venous
Bringhen, S., Musto, P., Foa, R., Knight, R., R., Siegel, D.S., Katz, M. & Greipp, P.R. (2010) thromboembolism in anticoagulated patients
Zeldis, J.B., Boccadoro, M. & Petrucci, M.T. Lenalidomide plus high-dose dexamethasone with cancer: management and short-term prog-
(2007) Melphalan, prednisone, and lenalidomide versus lenalidomide plus low-dose dexametha- nosis. Journal of Thrombosis and Haemostasis,
treatment for newly diagnosed myeloma: a sone as initial therapy for newly diagnosed mul- 13, 1010–1018.
report from the GIMEMA–Italian Multiple Mye- tiple myeloma: an open-label randomised Schutt, P., Ebeling, P., Buttkereit, U., Brandhorst,
loma Network. Journal of Clinical Oncology, 25, controlled trial. The Lancet Oncology, 11, 29–37. D., Opalka, B., Hoiczyk, M., Flasshove, M.,
4459–4465. Raskob, G.E., Buller, H.R. & Segers, A. (2018) Hense, J., Bojko, P., Metz, K., Moritz, T., See-
Palumbo, A., Rajkumar, S.V., Dimopoulos, M.A., Edoxaban for cancer-associated venous throm- ber, S. & Nowrousian, M.R. (2005) Thalidomide
Richardson, P.G., San Miguel, J., Barlogie, B., boembolism. New England Journal of Medicine, in combination with vincristine, epirubicin and
Harousseau, J., Zonder, J.A., Cavo, M., Zangari, 379, 95–96. dexamethasone (VED) for previously untreated
M., Attal, M., Belch, A., Knop, S., Joshua, D., Richardson, P., Jagannath, S., Hussein, M., Beren- patients with multiple myeloma. European Jour-
Sezer, O., Ludwig, H., Vesole, D., Blade, J., Kyle, son, J., Singhal, S., Irwin, D., Williams, S.F., nal of Haematology, 74, 40–46.
R., Westin, J., Weber, D., Bringhen, S., Nies- Bensinger, W., Badros, A.Z., Vescio, R., Kenvin, Snowden, J.A., Ahmedzai, S.H., Ashcroft, J., D’Sa,
vizky, R., Waage, A., von Lilienfeld-Toal, M., L., Yu, Z., Olesnyckyj, M., Zeldis, J., Knight, R. S., Littlewood, T., Low, E., Lucraft, H., Maclean,
Lonial, S., Morgan, G.J., Orlowski, R.Z., Shi- & Anderson, K.C. (2009) Safety and efficacy of R., Feyler, S., Pratt, G. & Bird, J.M. (2011)
mizu, K., Anderson, K.C., Boccadoro, M., Durie, single-agent lenalidomide in patients with Guidelines for supportive care in multiple mye-
B.G., Sonneveld, P. & Hussein, M.A. (2008) relapsed and refractory multiple myeloma. loma 2011. British Journal of Haematology, 154,
Prevention of thalidomide- and lenalidomide- Blood, 114, 772–778. 76–103.
associated thrombosis in myeloma. Leukemia, Romualdi, E. & Ageno, W. (2016) Management of Srkalovic, G., Cameron, M.G., Deitcher, S.R., Kat-
22, 414–423. recurrent venous thromboembolism in cancer tke-Marchant, K. & Hussein, M.A. (2005) Inci-
Palumbo, A., Cavo, M., Bringhen, S., Zamagni, E., patients. Thrombosis Research, 140, S128–S131. dence and risk factors of venous
Romano, A., Patriarca, F., Rossi, D., Gentilini, San Miguel, J.F., Schlag, R., Khuageva, N.K., thromboembolism (VTD) in patients with amy-
F., Crippa, C., Galli, M., Nozzoli, C., Ria, R., Dimopoulos, M.A., Shpilberg, O., Kropff, M., loidosis. International Seminars in Surgical
Marasca, R., Montefusco, V., Baldini, L., Elice, Spicka, I., Petrucci, M.T., Palumbo, A., Samoi- Oncology, 2, 17–17.
F., Callea, V., Pulini, S., Carella, A.M., Zambello, lova, O.S., Dmoszynska, A., Abdulkadyrov, Stewart, A.K., Rajkumar, S.V., Dimopoulos,
R., Benevolo, G., Magarotto, V., Tacchetti, P., K.M., Schots, R., Jiang, B., Mateos, M.V., M.A., Masszi, T., Spicka, I., Oriol, A., Hajek,
Pescosta, N., Cellini, C., Polloni, C., Evangelista, Anderson, K.C., Esseltine, D.L., Liu, K., R., Rosinol, L., Siegel, D.S., Mihaylov, G.G.,
A., Caravita, T., Morabito, F., Offidani, M., Cakana, A., van de Velde, H. & Richardson, Goranova-Marinova, V., Rajnics, P., Suvorov,
Tosi, P. & Boccadoro, M. (2011) Aspirin, war- P.G. (2008) Bortezomib plus melphalan and A., Niesvizky, R., Jakubowiak, A.J., San-Miguel,
farin, or enoxaparin thromboprophylaxis in prednisone for initial treatment of multiple J.F., Ludwig, H., Wang, M., Maisnar, V., Min-
patients with multiple myeloma treated with myeloma. New England Journal of Medicine, arik, J., Bensinger, W.I., Mateos, M.V., Ben-
thalidomide: a phase III, open-label, randomized 359, 906–917. Yehuda, D., Kukreti, V., Zojwalla, N., Tonda,
trial. Journal of Clinical Oncology, 29, 986–993. Sanfilippo, K.M., Luo, S., Gage, B.F., O’Brian, K. M.E., Yang, X., Xing, B., Moreau, P. &
Parker, K., Mitra, S. & Thachil, J. (2018) Is antico- & Carson, K.R. (2014) Venous thromboem- Palumbo, A. (2015) Carfilzomib, lenalidomide,
agulating haemodialysis patients with non- bolism is associated with increased mortality in and dexamethasone for relapsed multiple

ª 2018 British Society for Haematology and John Wiley & Sons Ltd 555
British Journal of Haematology, 2018, 183, 538–556
Review

myeloma. New England Journal of Medicine, thalidomide-dexamethasone (TD) (abstract). doxorubicin in myeloma patients receiving
372, 142–152. Blood, 100, 787. thalidomide: implications for therapy. Blood,
Stirling, D., Hannant, W.A. & Ludlam, C.A. Weber, D., Rankin, K., Gavino, M., Delasalle, K. & 100, 1168–1171.
(1998) Transcriptional activation of the factor Alexanian, R. (2003) Thalidomide alone or with Zangari, M., Saghafifar, F., Mehta, P., Barlogie, B.,
VIII gene in liver cell lines by interleukin-6. dexamethasone for previously untreated multiple Fink, L. & Tricot, G. (2003) The blood coagula-
Thrombosis and Haemostasis, 79, 74–78. myeloma. Journal of Clinical Oncology, 21, 16– tion mechanism in multiple myeloma. Seminars
Storrar, N.P.F., Mathur, A., Johnson, P.R.E. & 19. in Thrombosis and Hemostasis, 29, 275–282.
Roddie, P.H. (2018) Safety and efficacy of apix- Weber, D.M., Chen, C., Niesvizky, R., Wang, M., Zangari, M., Barlogie, B., Anaissie, E., Saghafifar,
aban for routine thromboprophylaxis in mye- Belch, A., Stadtmauer, E.A., Siegel, D., Borrello, F., Eddlemon, P., Jacobson, J., Lee, C.K., Ther-
loma patients treated with thalidomide- and I., Rajkumar, S.V., Chanan-Khan, A.A., Lonial, tulien, R., Talamo, G., Thomas, T., Van Rhee,
lenalidomide-containing regimens. British Jour- S., Yu, Z., Patin, J., Olesnyckyj, M., Zeldis, J.B. F., Fassas, A., Fink, L. & Tricot, G. (2004a) Deep
nal of Haematology. https://doi.org/10.1111/bjh. & Knight, R.D. (2007) Lenalidomide plus dex- vein thrombosis in patients with multiple mye-
15392 amethasone for relapsed multiple myeloma in loma treated with thalidomide and chemother-
Streetly, M., Hunt, B.J., Parmar, K., Jones, R., North America. New England Journal of Medi- apy: effects of prophylactic and therapeutic
Zeldis, J. & Schey, S. (2005) Markers of cine, 357, 2133–2142. anticoagulation. British Journal of Haematology,
endothelial and haemostatic function in the Weitz, J.I., Lensing, A.W.A., Prins, M.H., Bauer- 126, 715–721.
treatment of relapsed myeloma with the sachs, R., Beyer-Westendorf, J., Bounameaux, Zangari, M., Barlogie, B., Lee, C.-K., Tricot,
immunomodulatory agent Actimid (CC-4047) H., Brighton, T.A., Cohen, A.T., Davidson, B.L., A.E., Fassas, A., Anaissie, E., Hollmig, K.,
and their relationship with venous thrombosis. Decousus, H., Freitas, M.C.S., Holberg, G., Kak- Thertulien, R., Talamo, G., van Rhee, F.,
European Journal of Haematology, 74, 293–296. kar, A.K., Haskell, L., van Bellen, B., Pap, A.F., Stover, J.M., Fink, L. & Tricot, G.J. (2004b)
Teasell, R.W., Hsieh, J.T., Aubut, J.A., Eng, J.J., Berkowitz, S.D., Verhamme, P., Wells, P.S. & Protective effect of VELCADE on thalido-
Krassioukov, A. & Tu, L. (2009) Venous throm- Prandoni, P. (2017) Rivaroxaban or aspirin for mide-associated deep vein thrombosis (DVT).
boembolism after spinal cord injury. Archives of extended treatment of venous thromboem- Blood, 104, 4914–4914.
Physical Medicine and Rehabilitation, 90, 232– bolism. New England Journal of Medicine, 376, Zangari, M., Guerrero, J., Cavallo, F., Prasad,
245. 1211–1222. H.K., Esseltine, D. & Fink, L. (2008) Hemostatic
Thachil, J. (2012) Recurrent venous thromboem- Yagci, M., Sucak, G.T. & Haznedar, R. (2003) Fib- effects of bortezomib treatment in patients with
bolism while on anticoagulant therapy. Blood rinolytic activity in multiple myeloma. American relapsed or refractory multiple myeloma. Hae-
Reviews, 26, 175–181. Journal of Hematology, 74, 231–237. matologica, 93, 953–954.
Thiagarajan, P., Dannenbring, R., Matsuura, K., Yang, F., Hellyer, J.A., Than, C., Ullal, A.J., Kaiser, Zangari, M., Fink, L., Zhan, F. & Tricot, G. (2011)
Tramontano, A., Gololobov, G. & Paul, S. D.W., Heidenreich, P.A., Hoang, D.D., Winkel- Low venous thromboembolic risk with borte-
(2000) Monoclonal antibody light chain with mayer, W.C., Schmitt, S., Frayne, S.M., Phibbs, zomib in multiple myeloma and potential pro-
prothrombinase activity. Biochemistry, 39, 6459– C.S. & Turakhia, M.P. (2017) Warfarin utilisa- tective effect with thalidomide/lenalidomide-
6465. tion and anticoagulation control in patients with based therapy: review of data from phase 3 trials
Tosi, P., Zamagni, E., Cellini, C., Ronconi, S., atrial fibrillation and chronic kidney disease. and studies of novel combination regimens.
Patriarca, F., Ballerini, F., Musto, P., Di Rai- Heart, 103, 818–826. Clinical Lymphoma Myeloma and Leukemia, 11,
mondo, F., Ledda, A., Lauria, F., Masini, L., Yasin, Z., Quick, D., Thiagarajan, P., Spoor, D., 228–236.
Gobbi, M., Vacca, A., Ria, R., Cangini, D., Tura, Caraveo, J. & Palascak, J. (1999) Light-chain Zervas, K., Dimopoulos, M.A., Hatzicharissi, E.,
S., Baccarani, M. & Cavo, M. (2002) Salvage paraproteins with lupus anticoagulant activity. Anagnostopoulos, A., Papaioannou, M., Mit-
therapy with thalidomide in patients with American Journal of Hematology, 62, 99–102. souli, C., Panagiotidis, P., Korantzis, J., Tzil-
advanced relapsed/refractory multiple myeloma. Young, A.M., Marshall, A., Thirlwall, J., Chapman, ianos, M. & Maniatis, A. (2004) Primary
Haematologica, 87, 408–414. O., Lokare, A., Hill, C., Hale, D., Dunn, J.A., treatment of multiple myeloma with thalido-
Trujillo-Santos, J., Ruiz-Gamietea, A., Luque, J.M., Lyman, G.H., Hutchinson, C., MacCallum, P., mide, vincristine, liposomal doxorubicin and
Samperiz, A.L., Garcia-Bragado, F., Todoli, J.A. Kakkar, A., Hobbs, F.D.R., Petrou, S., Dale, J., dexamethasone (T-VAD doxil): a phase II mul-
& Monreal, M. (2009) Predicting recurrences or Poole, C.J., Maraveyas, A. & Levine, M. (2018) ticenter study. Annals of Oncology, 15, 134–138.
major bleeding in women with cancer and Comparison of an oral factor Xa inhibitor with Zonder, J.A., Durie, B.G.M., McCoy, J., Crowley,
venous thromboembolism. Findings from the low molecular weight heparin in patients with J., Zeldis, J.B., Ghannam, L. & Barlogie, B.
RIETE Registry. Thrombosis Research, 123(Suppl cancer with venous thromboembolism: results of (2005) High incidence of thrombotic events
2), S10–S15. a randomized trial (SELECT-D). Journal of Clin- observed in patients receiving Lenalidomide
Weber, D. (2002) Correlation of thrombotic/em- ical Oncology, 36, 2017–2023. (L) + Dexamethasone (D) (LD) as first-line
bolic events (T/E) with features of hypercagula- Zangari, M., Siegel, E., Barlogie, B., Anaissie, E., therapy for multiple myeloma (MM) without
bility in previously treated patients beore and Saghafifar, F., Fassas, A., Morris, C., Fink, L. & aspirin (ASA) prophylaxis. Blood, 106, 3455–
after treatment with thalidmide (T) or Tricot, G. (2002) Thrombogenic activity of 3455.

556 ª 2018 British Society for Haematology and John Wiley & Sons Ltd
British Journal of Haematology, 2018, 183, 538–556

You might also like