You are on page 1of 10

Curr Hematol Malig Rep

DOI 10.1007/s11899-017-0370-5

MULTIPLE MYELOMA (P KAPOOR, SECTION EDITOR)

An Evidence-Based Approach to Myeloma Bone Disease


Nicholas Bingham 1 & Antonia Reale 2 & Andrew Spencer 1,2

# Springer Science+Business Media New York 2017

Abstract Multiple myeloma is an incurable clonal plasma Introduction


cell malignancy characterised by osteolytic bone lesions and
the presence of a monoclonal immunoglobulin. The bone dis- Multiple myeloma MM is an incurable haematological malig-
ease caused by myeloma is a major cause of morbidity with nancy of clonal plasma cells, characterised by osteolytic bone
the related complications of pathological fractures, lesions and the presence of a monoclonal immunoglobulin [1].
hypercalcaemia and bone pain affecting both quality of life MM accounts for 1% of all malignancies, with an incidence of
and patient survival. The osteolytic lesions arise due to an approximately 6 cases per 100,000 people, with a median age
imbalance of osteoclast and osteoblast function, arising from at diagnosis of 65–70 years [2].
complex interactions between myeloma cells, bone marrow Osteolytic bone disease in MM develops due to increased
stromal cells and the bone marrow microenvironment. These bone resorption by osteoclasts and reduced bone formation
advances in understanding of the pathophysiology have di- through suppression of osteoblast function [3]. This imbal-
rectly led to improvements in patient management and out- ance arises due to complex interactions between MM cells,
comes. Recent advances in myeloma bone disease are bone marrow stromal cells and osteoclasts [4]. The osteolytic
reviewed, including the role of novel and emerging therapies, lesions predominantly affect the axial skeleton and proximal
and evidence-based management strategies for myeloma bone long bones, although any bone can be affected [5]. Osteolytic
disease are discussed. bone lesions lead to complications known as skeletal-related
events (SREs), which are defined as bone pain, pathological
f r a c t u r e s , n e ed fo r s u rg e r y or ra d i ot h e r a p y a n d
Keywords Multiple myeloma . Myeloma bone disease . hypercalcaemia [6]. Spinal instability and spinal cord or nerve
RANK/RANKL pathway root compression can also occur from bone disease [7].
Treatment of MM bone disease is required to reduce SREs
This article is part of the Topical Collection on Multiple Myeloma and maintain or improve quality of life and reduce treatment
Nicholas Bingham and Antonia Reale contributed equally to this work. cost [8].

* Andrew Spencer
andrew.spencer@monash.edu Prevalence and Impact of Bone Disease
Nicholas Bingham
n.bingham@alfred.org.au Bone disease affects 80–90% of patients with MM during
Antonia Reale
their disease course [3]. Bone disease can take the form of a
antonia.reale@monash.edu single osteolytic lesion (solitary plasmacytoma), diffuse
osteopenia or multiple lytic lesions. Osteopenia is found in
1
Department of Haematology, Alfred Health, Melbourne, Victoria, up to 90% of MM patients [9] while lytic lesions occur in at
Australia least 80% [10]. Untreated patients with bone lesions typically
2
Australian Centre for Blood Disorders, Monash University, have >2 SREs per year [11]. Pathological fractures occur in
Melbourne, Victoria, Australia 40% of patients in the first year after diagnosis and 60% of
Curr Hematol Malig Rep

patients during the disease course [12]. Spinal cord compres- the progression of bone disease in MM by increasing RANKL
sion can affect up to 20% of patients [7]. Symptoms related to expression on BMSCs [24]. The Notch signalling pathway
bone disease and SREs are a major cause of reduced quality of also drives osteoclast activation through the RANK/RANKL
life, mobility and performance status in MM patients [13]. pathway. Activation of the Notch pathway in MM and
Bone pain is the first reported symptom in 70% of cases BMSCs leads to increased secretion of RANKL, and this in
[10]. Aside from reduced quality of life, pathological fractures turn leads to the upregulation of the receptor Notch2 on pre-
also have a negative impact on survival; compared to patients osteoclasts, stimulating osteoclast differentiation via an auto-
without fractures, there is a 20% increase in mortality [3]. crine RANKL signalling pathway [25]. Finally, MM cells
Moreover, the number of bone lesions has been shown to may also secrete parathyroid hormone-related protein
correlate with prognosis [14]. Osteosclerotic lesions in MM (PTHrP) and Annexin II. PTHrP increases expression of
are rare, being found in less than 3% of cases [15], although RANKL through stimulation of osteoblasts and bone marrow
they are seen more frequently in the related plasma cell dys- stromal cells [26]. Annexin II stimulates osteoclast growth and
crasia POEMS syndrome [16]. increases bone resorption by stimulating the expression of
RANK on BMSCs [3].
There are also a number of RANK/RANKL independent
Pathophysiology mechanisms of osteoclast stimulation in MM. BMSCs express
growth differentiation factor 15 (GDF15), a member of the
MM plasma cells accumulate in the bone marrow where they transforming growth factor-beta family that both increases
promote unbalanced bone remodelling through increased os- osteoclast differentiation and inhibits osteoblast differentiation
teoclast activity and suppressed osteoblast activity [3]. Several in vitro. Serum GDF15 levels are elevated in patients with
theories have been proposed to explain this uncoupling of the advanced osteolytic bone disease compared to the levels in
bone remodelling process (see Fig. 1). patients with no lesions [27]. EphrinB2 is a member of the
One key mechanism is the adherence of MM cells to bone Ephrin family and binds to its specific receptor, EphB4.
marrow stromal cells (BMSCs) that stimulate secretion of pro- EphrinB2/EphB4 plays an important role in bone remodel-
osteoclastogenic and anti-osteoblastogenic cytokines. Cell-to- ling; the bidirectional signal transduction allows transition
cell adhesion is mediated by integrins such as VLA-4 from bone resorption to bone formation. MM cells negatively
expressed by MM cells and VCAM-1 expressed on BMSCs regulate EphrinB2/EphB4 expression, leading to uncoupling
[17, 18]. Similarly, ANGPTL4, an extracellular matrix protein, of bone remodelling process [3]. PU.1 is a transcription factor
mediates interaction between MM cells and BMSCs during the that controls early OCL differentiation from myeloid precur-
early stages of MM bone disease and has a role in the sors [28]. PU.1-deficient mice show marked inhibition of
uncoupling of angiogenesis and osteogenesis [19]. MM cells OCL differentiation [29]. In contrast, extracellular signal-
produce cytokines such as IL-6, chemokine (C-C motif) ligand regulated kinase (ERK) mediates the later stages of OCL dif-
3 (CCL3) and dickkopf-1 (DKK1) that directly or indirectly ferentiation and continuing OCL survival [30];
affect bone cell activity [4]. Many of the osteoclastogenic cy- phosphorylated-ERK is critical for MM cell growth [31].
tokines converge on the receptor activator of nuclear factor-κB Inhibition of these pathways induces apoptosis of OCL [30].
(RANK) pathway that is of central importance in osteoclast While some pathways stimulate osteoclast development in
differentiation and activation [20, 21]. RANK is a transmem- MM, other pathways are altered leading to suppression of
brane signalling receptor located on the surface of osteoclast osteoblast maturation and/or function. Runt-related transcrip-
precursors whereas RANK ligand (RANKL) is secreted by tion factor 2 (Runx2) is suppressed in MM by tumour necrosis
activated lymphocytes and also expressed on BMSCs and os- factor-alpha (TNF-α) and Gfi1. TNF-alpha is an inflammatory
teoblasts. Through the NF-κB and JunN terminal kinase path- cytokine that is increased in MM [32, 33] whereas Gfi1 is a
ways, the RANK/RANKL signal enhances osteoclast survival transcriptional repressor of Runx2 and increased levels of
and increases bone resorption [20]. Gfi1 have been reported in MM patients [34]. IL-7 has been
The RANK/RANKL pathway is perturbed in MM. MM implicated in the Runx2-mediated inhibition in osteoblast pro-
cells can disrupt the interplay between RANKL and its soluble genitors and in the consequent suppression of osteoblast for-
decoy receptor osteoprotegerin (OPG) with increased mation [35].
RANKL expression and decreased OPG expression promot- The loss of osteoblast numbers and function as MM pro-
ing the formation and activation of osteoclasts [22]. gresses has been recognised as a reflection of tumour evolu-
Additionally, T cells derived from the peripheral blood mono- tion, with a loss of the MM cell dependence on osteoblast-
nuclear cells (PBMCs) of MM patients with osteolysis have derived factors or the increased compensatory growth factors
been shown to support the development and survival of oste- that are released through bone resorption, e.g., IGF-I [36].
oclasts through expression of high levels of RANKL [23]. CCL3 is a pro-inflammatory cytokine involved in the recruit-
These T cells also produce IL-17, which plays a key role in ment and differentiation of osteoclast precursors. It is
Curr Hematol Malig Rep

Fig. 1 The pathophysiology of bone disease in myeloma. The adherence towards RANKL. On the other hand, OCs produce several growth
of MM cells to bone marrow stromal cells (BMSCs) stimulates the factors (e.g., IL-6) which promote the growth and survival of MM cells.
secretion of pro-osteoclastogenic and anti-osteoblastogenic cytokines in A number of other pathways are altered leading to suppression of OB
the bone marrow microenvironment. Cell-to-cell adhesion is mediated by maturation and/or function. MM cells produce high levels of DKK1
integrins such as VLA-4 expressed by MM cells and VCAM-1 expressed which inhibits OB activity. Runx2 is suppressed by factors secreted by
on BMSCs. MM cells, stromal cells and T cells produce several pro- MM cells such as TNF-α and IL-7. Furthermore, MM cells directly
osteoclastogenic factors (including RANKL, CCL3, IL-3, IL-6, IL-17, induce OB apoptosis via TRAIL. RANKL receptor activator of nuclear
PTHrP) that directly or indirectly promote OC differentiation from OC factor-κB ligand, CCL3 chemokine (C-C motif) ligand 3, IL-3/6/7/17
precursors, hence OC resorptive activity. The expression of OPG, the interleukin 3/6/7/17, OPG osteoprotegerin, OC osteoclast, pOC pre-
RANKL-soluble decoy receptor, by stromal cells is reduced, and osteoclast, OB osteoblast, DKK1 dickkopf-1, TNF-α tumour necrosis
inactivation of OPG through binding to syndecan-1 on the surface of factor-alpha, Runx2 Runt-related transcription factor 2, TRAIL TNF-
MM cells is also observed, thus favouring the RANKL/OPG ratio related apoptosis-inducing ligand, MM Multiple myeloma

upregulated in MM bone marrow, promotes tumour progres- function have been noted prior to the development of overt
sion and bone lysis [3] and impairs osteoblast function but has bone loss in MM [38, 41]. MM cells establish intimate rela-
no effect on osteoblast differentiation [37]. In early MM, there tionships with bone cells and are likely to exert effects soon
is reduced osteoblast function despite preserved numbers, after their arrival in the bone marrow. Abnormal bone metab-
likely driven by elevated CCL3 expression. Later in disease olism has been described also in asymptomatic or smoulder-
evolution, osteoblast suppression is manifested by both re- ing MM (SMM) or MGUS, reinforcing the concept that bone
duced differentiation and impaired function [38]. physiology is affected even while the tumour is in its pre-
Furthermore, CCL3 levels fall while DKK1 levels rise, sug- malignant phase [42]. This provides a rationale for clinical
gesting that disruption of Wnt signalling may play a role at intervention at an early stage of disease when patients are still
this stage [39]. Wnt is a ligand that binds to the low-density asymptomatic, supporting the recent change in recommenda-
lipoprotein receptor-related protein with subsequent stimula- tions regarding treatment of high-risk SMM [43].
tion of osteoblastogenesis; sclerostin, like DKK1, is a Wnt
antagonist, also found at high levels in MM patients [40]. As
the disease progresses, MM cells lose their dependence on Imaging in Myelomatous Bone Disease
BMSCs. Levels of several stromal-derived MM growth/
survival factors (such as BAFF [TNFSF13B], IL6, IL6R, Diagnosis
VEGFA) fall in advanced MM. This reflects a disturbance of
stromal physiology, when other factors become important [3, Plain radiographs of the axial and appendicular skeleton (the
4, 38]. The consistent downregulation of adhesion molecules skeletal survey—SS) are the gold standard for the detection of
LFA-1 (ITGB2), VLA-4 (ITGA4) and VCAM1 in advanced osteolytic lesions in MM [8]. Importantly, however, the SS has
disease reflects reduced dependence of tumour cells on adhe- several limitations: quantification of lesion size is not possible
sive interactions at this stage [38]. and it has low sensitivity, requiring loss of at least 30% of
The typical uncoupling of osteoclast and osteoblast func- cortical bone to be detected [5]. Whole-body low-dose
tion occurs in advanced MM; however, changes in osteoblast computerised tomography (WBLD CT) has a higher
Curr Hematol Malig Rep

sensitivity than plain radiographs [5]. It detects more bone targets. Bone turnover markers can reflect either bone resorp-
lesions and more lesions at risk of fracture and upstaged al- tion or bone formation, but the clinical utility of many of these
most 50% of patients in one study [44]. Moreover, lesions can markers remains unclear. However, the identification of a val-
be quantified more accurately. Functional imaging, such as idated and reliable marker of disease activity could minimise
18F-fluorodeoxyglucose positron emission tomography com- the requirement for invasive procedures, expensive imaging
bined with CT improves upon the sensitivity of CT (reportedly and radiation exposure [54].
between 80 and 90%) with a specificity of 80–100% [5]. PET/ The most commonly used marker is C-terminal type 1 col-
CT identifies more lesions that SS alone [45] and results at lagen telopeptide (CTX-1, C-telopeptide). CTX-1 is a break-
diagnosis correlate with survival [14]. PET/CT may be partic- down product of collagen and is used as a marker of bone
ularly useful in cases of solitary plasmacytoma as available resorption [55]. Levels are high at diagnosis and highest in
evidence suggests that it upgrades the diagnosis in approxi- patients with significant bone involvement; this can reflect
mately 42% of patients, significantly altering management in marrow infiltration rather than purely lytic lesions. CTX-1 is
these cases [46]. renally excreted and is therefore difficult to interpret in severe
Magnetic resonance imaging MRI and Sestamibi scans, renal failure (eGFR <30). Patients who respond to treatment
unlike CT and SS, are used to detect focal infiltration of bone have a reduction in CTX-1 at 3 months. Relapse is similarly
marrow rather than lytic lesions. Sestamibi scans utilise preceded by a rise in CTX-1 3–6 months prior to changes on
technetium-99-m labelled hexakis-2-methoxy-isobutyl- imaging, so CTX-1 has potential as a marker of early relapse.
isonitrite (99mTc-sestamibi) that accumulates in tissues with Bisphosphonate therapy reduces the CTX-1 level, but there is
high mitochondrial activation such as MM. The degree of still a statistically significant change in CTX-1 level prior to
uptake has been shown to correlate with disease severity in relapse, thus CTX-1 retains potential as a biomarker in pa-
MM, but the scan is limited by failure to identify lytic lesions tients on bisphosphonates [55].
at risk of fracture [5]. MRI is useful in MM and complements Other markers reported in the literature include DKK-1 and
SS and WBCT by detecting marrow infiltration in contrast to RANKL (used as markers of MM cell growth and develop-
lytic lesions. MRI is inferior to WBCT in detecting bony le- ment of lytic lesions), TRACP-5b, (a marker of osteoclast
sions but detects bone marrow infiltration in the absence of function) and osteocalcin and bone alkaline phosphatase
lytic lesions. MRI is superior to SS and upgrades diagnosis in (markers of bone formation). However, the use of these
68% of cases. Furthermore, in the evaluation of SMM, the markers is generally limited to research settings.
presence of more than one focal lesion on MRI is associated
with a 70% risk of progression to symptomatic MM in 2 years
[47] and MRI findings have also been shown to correlate with Management of Myeloma Bone Disease
survival and treatment response [14]. The presence of more
than one focal lesion on MRI of at least 5 mm in size is now Systemic control of MM via the modulation of bone disease
considered a myeloma-defining event [48]. pathophysiology is the cornerstone of treatment for MM bone
disease. More specific MM bone disease therapies, includes
Assessment of Disease Response bisphosphonates, radiotherapy, surgery and balloon
kyphoplasty, with newer investigational therapies remaining
Current response criteria are based on laboratory parameters an active area of current research.
as response to therapy cannot be determined with SS or
WBLDCT as osteolytic lesions rarely improve with therapy Bisphosphonates
[49]; additionally, SS do not enable the evaluation of
extramedullary disease. In contrast, improvement on com- Bisphosphonates (BPs) are recommended for all patients with
bined PET/CT imaging after treatment correlates with both MM requiring front-line therapy, regardless of the presence of
clinical improvement and prolonged survival [50, 51]. bone disease at diagnosis. Moreover, BPs are recommended
Alternatively, whole-body MRI can be used if available [52] for those with low- and intermediate-risk SMM if osteoporosis
as can Sestamibi scans, as results after therapy correlate with is identified by dual-energy x-ray absorptiometry scan, at
biochemical markers of disease activity [53]. doses used in patients with osteoporosis. For high-risk
SMM, or if one cannot differentiate between MM-related ver-
sus age-related bone loss, the treating physician should con-
Bone Turnover Markers in Myeloma sider using the dosing and schedule of BPs as for symptomatic
MM, especially in patients with abnormal MRIs [8].
A better understanding of the pathophysiology of MM has led The BPs zoledronate (ZOL), pamidronate (PAM) and
to the discovery of a range of potential bone turnover markers, clodronate (CLO) reduce SREs and control bone pain [8, 56].
many of which have also been investigated as therapeutic There is no single bisphosphonate that shows benefit over
Curr Hematol Malig Rep

others with regard to reduction in SREs, bone pain or overall to moderate renal impairment (CrCl rate of 30 to 60 mL/min)
survival, although ZOL improved overall survival by should receive reduced doses of CLO and ZOL under close
10 months in MM patients with lytic lesions at diagnosis when clinical monitoring. No change to ZOL infusion time is rec-
compared to clodronate [57]. ZOL use in newly diagnosed MM ommended. PAM should be administered via extended infu-
patients also results in a reduced rate of SREs when compared sion duration (>4 h) and clinicians should consider initial dose
with CLO [8], whereas intravenous (IV) ZOL and PAM exhibit reduction in patients with renal impairment. PAM and ZOL
comparable efficacy in reducing SREs [58]. While no data are not recommended for patients with CrCl <30 mL/min.
demonstrate a survival advantage for PAM, both PAM 30 and PAM has been rarely associated with nephrotic syndrome
90 mg have shown comparable effects for preventing SREs and due to focal sclerosing glomerulosclerosis [62] with most
PAM 30 mg is associated with a lower risk of both cases appearing dose related and cessation of PAM was asso-
osteonecrosis of the jaw (ONJ) and nephrotoxicity compared ciated with improvement in renal function [63].
to PAM 90 mg [8]. When feasible, IV administration of BPs is
preferred with infusion times ranging from 15 min for ZOL to 2 Calcium and Vitamin D
to 4 h for PAM. Oral administration remains an option for
patients who cannot receive regular hospital care or in-home Hypocalcaemia is a complication of bisphosphonate therapy.
nursing visits. IV BPs should be administered at 3- to 4-week This can be prevented by the administration of oral calcium
intervals to all patients with active MM [6, 8]. While the rec- and vitamin D3. Patients should routinely receive calcium
ommended dosing schedule for ZOL is that it be given 4 week- (600 mg per day) and vitamin D3 (400 IU per day) supple-
ly, a recent study has shown that the dosing of ZOL at 12- mentation. Calcium supplementation should be used with cau-
weekly intervals is not associated with an increased risk of tion in patients with renal insufficiency [6, 8].
SREs compared to 4 weekly [59]. While in all randomized,
placebo-controlled trials, the bisphosphonates were given for Denosumab
a maximum period of 2 years, in the MRC-IX trial, the
bisphosphonates were given until disease progression [60]. Denosumab is a fully human monoclonal antibody specific to
The subset of patients who received ZOL for more than 2 years RANKL that inhibits the formation, function and survival of
continued to experience a reduction in SREs and an improve- osteoclasts, thus decreasing cancer-mediated bone destruc-
ment of overall survival compared with clodronate. However, tion. In a large randomized trial of patients with evidence of
there was no sub-analysis according to the response status of bone disease including MM, denosumab was not inferior to
the patients, and thus it is not clear if the continuous use of ZOL ZOL and reduced the risk of SREs compared with ZOL; how-
produces similar results in patients who have achieved a CR, ever, overall survival at 1 year was less in MM patients receiv-
sCR, very good partial response (VGPR) or PR. Therefore, ing denosumab compared to patients receiving ZOL (83 vs
ZOL should be administered until disease progression, except 97%) [64, 65]. These results are difficult to interpret as the
in patients who have achieved CR or VGPR [8, 59] and had no study was not powered to ask a question about the two agents
lytic disease at diagnosis. There are no data supporting the specifically in MM with resulting small numbers of MM pa-
continuous use of pamidronate, thus PAM should be adminis- tients and imbalances in baseline disease characteristics con-
tered for 2 years and then at the physician’s discretion in pa- founding any post hoc evaluation. The toxicity of denosumab
tients with active disease. PAM therapy should be resumed is low, most commonly causing asthenia and hypocalcaemia,
after disease relapse [8]. with a rate of ONJ comparable to ZOL. Denosumab may lead
Potential adverse events (AEs) associated with BP admin- to increased infection rates through its anti-RANKL effect so
istration include hypocalcaemia and hypophosphataemia, GI caution is needed in immunocomprised MM patients [64]. A
events after oral administration, inflammatory reactions at the larger international study (NCT01345019) is underway and
injection site and acute-phase reactions after IVadministration will provide further evidence regarding the efficacy and safety
of amino BPs. Renal impairment and ONJ represent infre- of denosumab in MM when compared to ZOL. Denosumab
quent but potentially serious AEs. can currently be given in MM patients only in the rare cases of
Preventive strategies should be adopted to avoid ONJ. hypercalcaemia resistant to BPs [66].
Patients should receive a comprehensive dental examination
and education regarding optimal dental hygiene, and existing Anti-MM Therapies—Proteasome Inhibitors
dental conditions should be treated before initiating BP thera-
py. After BP treatment initiation, unnecessary invasive dental Several studies have confirmed the positive effects of PIs on
procedures should be avoided and dental health status should bone formation and resorption markers [67–70]; moreover, a
be monitored annually [61]. Patients should be monitored for retrospective analysis of bortezomib-treated MM patients
compromised renal function by measuring creatinine clear- demonstrated a correlation between the bone formation mark-
ance (CrCl) prior to each IV BP infusion. Patients with mild er alkaline phosphatase and disease response. A decrease in
Curr Hematol Malig Rep

DKK-1 serum levels and other markers of bone resorption osteoblasts [77]. It is increasingly clear that IMiDs are effec-
(such as TRAP-5b, CTX and soluble RANKL) has also been tive anti-MM agents with separate positive effects on the re-
observed after bortezomib treatment. No significant changes lated bone disease [74, 78].
in these markers were detected in non responders [54].
Histological studies have shown that MM patients who re- Surgery
spond to bortezomib have an increase in the number of oste-
oblastic cells/mm2 of bone tissue compared to non-responders Osteolytic lesions in MM rarely heal due to suppressed oste-
[69]. The positive anabolic effect of bortezomib on bone oblast function. Thus, surgical management of actual or
healing and new matrix deposition was confirmed by bone impending pathological fractures should be considered first
imaging techniques such as dual-energy X-ray absorptiometry line [79]. Impending fracture is suggested by a loss of >50%
(DEXA), technetium-99 m-(99mTc-) methyl-diphosphonate of cortical bone thickness. Orthopaedic consultation should be
(MDP) bone scan and micro-CT measurements on biopsy sought for lesions in weight-bearing bones, bony compression
specimens [33]. of the spinal cord and vertebral body instability, although sur-
In addition to its clinical anti-tumour effect, the second gery is not frequently indicated due to the inherent radiosen-
generation PI carfilzomib (PR-171) has been shown to reverse sitivity of most MM [79]. Technically, the operative interven-
osteoblast inhibition and improve bone disease in an in vivo tion should allow full weight bearing post-operatively and not
MM mouse model [71]. Carfilzomib stimulates mesenchymal rely upon healing of the fracture to achieve stability. Surgery
stromal cell (MSC) differentiation into osteoblasts, and oste- should be avoided in critically ill patients with an expected
oblasts derived from MM patient MSCs when treated with duration of survival less than the time required for recovery
carfilzomib display increased bone formation in vitro [71, [80].
72]. At doses toxic to MM cells, carfilzomib also inhibits
osteoclast differentiation and function without affecting the
precursor viability. This effect is due in part to disruption of Radiotherapy
RANKL-induced NF-κB signalling [33]. A recent retrospec-
tive analysis of 67 relapsed or refractory MM patients treated Radiotherapy has an established role in MM management and
with carfilzomib demonstrated that elevation in ALP levels is used in up to a third of patients during their disease course
correlated with response to the treatment [73]. Likewise, other [81]. Radiotherapy should be considered first-line therapy in
second-generation PIs such as ixazomib and oprozomib inhib- true solitary plasmacytomas, where it can be potentially cura-
it osteoclastogenesis and bone resorption and promote osteo- tive, as well as in extramedullary masses and in spinal cord
blastogenesis and activity [33]. compression [9]. It is also very useful as an adjunct to analge-
sia in the management of bone pain, with response rates of
Anti-MM Therapies—Immunomodulatory Drugs more than 80% reported [81]. It should also be used as an
adjunct to operative fixation of actual and impending patho-
Thalidomide, and its second-generation derivatives logical fractures. In these settings, radiotherapy promotes
lenalidomide and pomalidomide, have direct anti-tumour bone healing and remineralisation, alleviates pain and reduces
effects via several different mechanisms [74]. IMiDs have analgesia use, improves functional status and reduces risk of
been reported to prevent the adhesion of MM cells to non- subsequent fractures. The limitation of radiotherapy is the
MM cells in the BM microenvironment including BMSCs, need to limit the involved field, especially prior to autologous
osteoclasts and immune cells. This breaks the pathophysi- stem cell collection in eligible patients [81].
ological cycle of MM by reducing MM-induced osteoclas-
togenesis and the resultant growth factors released during Balloon Kyphoplasty
bone destruction [74, 75].
Lenalidomide inhibits osteoclast formation and activation Balloon kyphoplasty is a minimally invasive procedure that
by inhibiting key factors, such as PU.1 and pERK, during should be considered in patients with symptomatic vertebral
osteoclastogenesis and also by reducing MM burden [29, compression fractures [6]. The procedure involves the percu-
75]. Additionally, lenalidomide has been shown to decrease taneous injection of bone cement (polymethylmethacrylate)
RANKL secreted by BMSCs derived from MM and down- into the vertebral body after balloon inflation to expand the
regulate cathepsin K (which is secreted by osteoclasts and collapsed vertebra. Kyphoplasty can be considered in patients
induces matrix degradation during bone resorption) [75]. who are not fit for surgical intervention, and major complica-
Pomalidomide (CC-4047) has also been shown to inhibit tions are rare [82]. It is the procedure of choice to improve
PU.1 and therefore osteoclastogenesis [76]. Another study quality of life, physical function and pain control in patients
reported that at a dose that induced apoptosis in MM cells, with symptomatic vertebral compression fractures, with ben-
IMiDs also showed an anti-osteoclast effect without affecting efits persisting for up to 2 years [82]. The role of simple
Curr Hematol Malig Rep

vertebroplasty is less clear due to paucity of data and a failure Compliance with Ethical Standards
to show benefit in osteoporosis trials [6].
Conflict of Interest The authors declare that they have no conflicts of
interest.
Investigational Therapies
Human and Animal Rights and Informed Consent This article does
Investigational therapies are being developed in MM bone not contain any studies with human or animal subjects performed by any
disease [77]. Most therapies to date inhibit osteoclast function, of the authors.
with none promoting bone formation through osteoblast stim-
ulation. BHQ880 is a first-in-class, fully human IgG1 anti-
DKK-1 monoclonal antibody, which increases osteoblast dif- References
ferentiation and inhibits malignant plasma cell growth thus
mitigating against the development of osteolytic lesions 1. Anderson KC, Alsina M, Atanackovic D. Multiple Myeloma. J Natl
Compr Canc Netw. 2016;13(11):1398–1435.
[83]. Recently, in a phase IB multicentre study of BHQ880
2. Moreau P, San Miguel J, Ludwig H, Schouten H, Mohty M,
in combination with anti-MM therapy and ZOL in patients Dimopoulos M, et al. Multiple myeloma: ESMO clinical practice
with RRMM, BHQ880 was well tolerated and a trend toward guidelines for diagnosis, treatment and follow-up. Ann Oncol.
increased bone mineral density over time was observed. 2013;24:vi133–7. doi:10.1093/annonc/mdt297.
3. Silbermann R, Roodman GD. Myeloma bone disease: pathophysi-
However, the relative contribution of BHQ880 could not be
ology and management. J Bone Oncol. 2013;2:59–69. doi:10.1016/
established due to concomitantly administered ZOL and anti- j.jbo.2013.04.001.
MM therapy. Further research into the effects of BHQ880 in 4. García-Gómez A, Sanchez-Guijo F, del Canizo C. Multiple myelo-
MM is warranted [83, 84]. Sclerostin is a Wnt inhibitor, se- ma mesenchymal stromal cells: contribution to myeloma bone dis-
creted by MM cells and found in high levels in patients with ease and therapeutics. WJSC. 2014;6:322. doi:10.4252/wjsc.v6.i3.
322.
advanced bone disease. Monoclonal antibodies against 5. Tan E, Weiss BM, Mena E, Korde N, Choyke PL, Landgren O.
sclerostin have been developed, including AMG785. This Current and future imaging modalities for multiple myeloma and its
agent has been shown to have biochemical anabolic effects precursor states. Leuk Lymphoma. 2011;52:1630–40. doi:10.3109/
and increases bone mineral density, and the results of further 10428194.2011.573036.
6. Terpos E, Kleber M, Engelhardt M, Zweegman S, Gay F, Kastritis
studies are awaited [77]. E, et al. European myeloma network guidelines for the management
of multiple myeloma-related complications. Haematologica.
2015;100:1254–66. doi:10.3324/haematol.2014.117176.
Conclusion 7. Tosi P. Diagnosis and treatment of bone disease in multiple myelo-
ma: spotlight on spinal involvement. Scientifica. 2013;2013:1–12.
doi:10.1155/2013/104546.
Survival outcomes for MM patients have substantially im- 8. Terpos E, Morgan G, Dimopoulos MA, Drake MT, Lentzsch S,
proved over the past decade, but the disease remains incur- Raje N, et al. International myeloma working group recommenda-
able. Bone disease and its complications remain key aspects of tions for the treatment of multiple myeloma-related bone disease. J
Clin Oncol. 2013;31:2347–57. doi:10.1200/JCO.2012.47.7901.
the management of patients, to not only maintain quality of
9. Hameed, Brady J, Dowling P, Clynes M, O’Gorman P. Bone dis-
life but also improve survival. An increased understanding of ease in multiple myeloma: pathophysiology and management.
the biology of MM-related bone disease has not only facilitat- CGM. 2014:33–10. doi: 10.4137/CGM.S16817
ed the rational use of BPs but also the preclinical and clinical 10. Kyle RA, Gertz MA, Witzig TE, Lust JA, Lacy MQ, Dispenzieri A,
et al. Review of 1027 patients with newly diagnosed multiple my-
development of new agents that inhibit osteoclastogenesis or
eloma. Mayo Clin Proc. 2003;78:21–33. doi:10.4065/78.1.21.
bone anabolic agents, such as anti-Dkk-1 and sclerostin. 11. Berenson JR, Porter L, Lichtenstein A. Efficacy of pamidronate in
Overall, a multimodal approach is required to MM bone reducing skeletal events in patients with advanced multiple myelo-
disease. Aggressive systemic therapy has beneficial effects on ma. N Eng J Med. 2000;334(8):1–6.
MM bone disease itself, making it the cornerstone of therapy. 12. Melton III LJ, Kyle RA, Achenbach SJ, Oberg AL, Rajkumar SV.
Fracture risk with multiple myeloma: a population-based study. J
Bisphosphonates are obligatory in all MM patients and in Bone Miner Res. 2004;20:487–93. doi:10.1359/JBMR.041131.
selected SMM patients. Radiotherapy or surgery should be 13. Cocks K, Cohen D, Wisløff F, Sezer O, Lee S, Hippe E, et al. An
considered in patients with or at risk of pathological fractures. international field study of the reliability and validity of a disease-
Radiotherapy is particularly effective in controlling bone pain. specific questionnaire module (the QLQ-MY20) in assessing the
quality of life of patients with multiple myeloma. Eur J Cancer.
Kyphoplasty has a role in vertebral compression fractures. 2007;43:1670–8. doi:10.1016/j.ejca.2007.04.022.
Results of investigational therapies are awaited. 14. Walker R, Barlogie B, Haessler J, Tricot G, Anaissie E,
Finally, the continued development of biomarkers that re- Shaughnessy JD, et al. Magnetic resonance imaging in multiple
flect the activity of MM-related bone disease and predict treat- myeloma: diagnostic and clinical implications. J Clin Oncol.
2007;25:1121–8. doi:10.1200/JCO.2006.08.5803.
ment response will represent a major advancement in the man- 15. Ghosh S, Wadhwa P, Kumar A, Pai KM, Seshadri S, Manohar C.
agement of this common and potentially devastating Abnormal radiological features in a multiple myeloma patient: a
complication. case report and radiological review of myelomas.
Curr Hematol Malig Rep

Dentomaxillofacial Radiol. 2011;40:513–8. doi:10.1259/dmfr/ angiogenesis. Leukemia. 2004;18:628–35. doi:10.1038/sj.leu.


74265829. 2403269.
16. Mulleman D, Gaxatte C, Guillerm G, Leroy X, Cotten A, 32. Gilbert L, He X, Farmer P, Rubin J, Drissi H, van Wijnen AJ, et al.
Duquesnoy B, et al. Multiple myeloma presenting with widespread Expression of the osteoblast differentiation factor RUNX2 (Cbfa1/
osteosclerotic lesions. Joint Bone Spine. 2004;71:79–83. doi:10. AML3/Pebp2 A) Is inhibited by tumor necrosis factor. J Biol Chem.
1016/S1297-319X(03)00152-0. 2002;277:2695–701. doi:10.1074/jbc.M106339200.
17. Mori G, D’Amelio P, Faccio R, Brunetti G. Bone-immune cell 33. Accardi F, Toscani D, Bolzoni M, Dalla Palma B, Aversa F, Giuliani
crosstalk: bone diseases. J Immunol Res. 2015;2015:1–11. doi:10. N. Mechanism of action of bortezomib and the new proteasome
1155/2015/108451. inhibitors on myeloma cells and the bone microenvironment: im-
18. Roodman GD. Pathogenesis of myeloma bone disease. Leukemia. pact on myeloma-induced alterations of bone remodeling. Biomed
2008;23:435–41. doi:10.1038/leu.2008.336. Res Int. 2015;2015:1–13. doi:10.1155/2015/172458.
19. Dotterweich J, Schlegelmilch K, Keller A, Geyer B, Schneider D, 34. D’Souza S, del Prete D, Jin S, Sun Q, Huston AJ, Kostov FE, et al.
Zeck S, et al. Contact of myeloma cells induces a characteristic Gfi1 expressed in bone marrow stromal cells is a novel osteoblast
transcriptome signature in skeletal precursor cells—implications suppressor in patients with multiple myeloma bone disease. Blood.
for myeloma bone disease. Bone. 2016;93:155–66. doi:10.1016/j. 2011;118:6871–80. doi:10.1182/blood-2011-04-346775.
bone.2016.08.006. 35. Giuliani N, Colla S, Morandi F, Lazzaretti M, Sala R, Bonomini S,
20. Roux S, Meignin V, Quillard J, Meduri G, Guiochon-Mantel A, et al. Myeloma cells block RUNX2/CBFA1 activity in human bone
Fermand J-P, et al. RANK (receptor activator of nuclear factor- marrow osteoblast progenitors and inhibit osteoblast formation and
kappaB) and RANKL expression in multiple myeloma. Br J differentiation. Blood. 2005;106:2472–83. doi:10.1182/blood-
Haematol. 2002;117:86–92. doi:10.1046/j.1365-2141.2002. 2004-12-4986.
03417.x. 36. Sprynski AC, Hose D, Caillot L, Reme T, Shaughnessy JD,
21. Schmiedel BJ, Scheible CA, Nuebling T, Kopp HG, Wirths S, Barlogie B, et al. The role of IGF-1 as a major growth factor for
Azuma M, et al. RANKL expression, function, and therapeutic myeloma cell lines and the prognostic relevance of the expression
targeting in multiple myeloma and chronic lymphocytic leukemia. of its receptor. Blood. 2009;113:4614–26. doi:10.1182/blood-
Cancer Res. 2013;73:683–94. doi:10.1158/0008-5472.CAN-12- 2008-07-170464.
2280. 37. Vallet S, Pozzi S, Patel K, Vaghela N, Fulciniti MT, Veiby P, et al. A
22. Sezer O, Heider U, Zavrski I, Kuhne C, Hofbauer L. RANK ligand novel role for CCL3 (MIP-1α) in myeloma-induced bone disease
and osteoprotegerin in myeloma bone disease. Blood. 2002;101: via osteocalcin downregulation and inhibition of osteoblast func-
2094–8. doi:10.1182/blood-2002-09-2684. tion. Leukemia. 2011;25:1174–81. doi:10.1038/leu.2011.43.
23. Colucci S, Brunetti G, Rizzi R, Zonno A, Mori G, Colaianni G,
38. Kassen D, Lath D, Lach A, Evans H, Chantry A, Rabin N, et al.
et al. T cells support osteoclastogenesis in an in vitro model derived
Myeloma impairs mature osteoblast function but causes early ex-
from human multiple myeloma bone disease: the role of the OPG/
pansion of osteo-progenitors: temporal changes in bone physiology
TRAIL interaction. Blood. 2004;104:3722–30. doi:10.1182/blood-
and gene expression in the KMS12BM model. Br J Haematol.
2004-02-0474.
2015;172:64–79. doi:10.1111/bjh.13790.
24. Oranger A, Carbone C, Izzo M, Grano M. Cellular mechanisms of
39. Qiang Y-W, Endo Y, Rubin JS, Rudikoff S. Wnt signaling in B-cell
multiple myeloma bone disease. Clin Dev Immunol. 2013;2013:1–
neoplasia. Oncogene. 2003;22:1536–45. doi:10.1038/sj.onc.
11. doi:10.1155/2013/289458.
1206239.
25. Colombo M, Thümmler K, Mirandola L, Garavelli S, Todoerti K,
Apicella L, et al. Notch signaling drives multiple myeloma induced 40. Colucci S, Brunetti G, Oranger A, Mori G, Sardone F, Specchia G,
osteoclastogenesis. Oncotarget. 2014;5:10393–406. doi:10.18632/ et al. Myeloma cells suppress osteoblasts through sclerostin secre-
oncotarget.2084. tion. Blood Cancer J. 2011;1, e27. doi:10.1038/bcj.2011.22.
26. Cafforio P, Savonarola A, Stucci S, De Matteo M, Tucci M, 41. Manier S, Sacco A, Leleu X, Ghobrial IM, Roccaro AM. Bone
Brunetti AE, et al. PTHrP produced by myeloma plasma cells reg- marrow microenvironment in multiple myeloma progression. J
ulates their survival and pro-osteoclast activity for bone disease Biomed Biotechnol. 2012;2012:1–5. doi:10.1155/2012/157496.
progression. J Bone Miner Res. 2013;29:55–66. doi:10.1002/ 42. Kristinsson SY, Minter AR, Korde N, Tan E, Landgren O. Bone
jbmr.2022. disease in multiple myeloma and precursor disease: novel diagnos-
27. Westhrin M, Moen SH, Holien T, Mylin AK, Heickendorff L, Olsen tic approaches and implications on clinical management. Expert
OE, et al. Growth differentiation factor 15 (GDF15) promotes os- Rev Mol Diagn. 2011;11:593–603. doi:10.1586/erm.11.44.
teoclast differentiation and inhibits osteoblast differentiation and 43. Rajkumar SV, Landgren O, Mateos MV. Smoldering multiple my-
high serum GDF15 levels are associated with multiple myeloma eloma. Blood. 2015;125:3069–75. doi:10.1182/blood-2014-09-
bone disease. Haematologica. 2015;100:e511–4. doi:10.3324/ 568899.
haematol.2015.124511. 44. Gleeson TG, Moriarty J, Shortt CP, Gleeson JP, Fitzpatrick P, Byrne
28. Breitkreutz I, Raab MS, Vallet S, Hideshima T, Raje N, Mitsiades B, et al. Accuracy of whole-body low-dose multidetector CT
C, et al. Lenalidomide inhibits osteoclastogenesis, survival factors (WBLDCT) versus skeletal survey in the detection of myelomatous
and bone-remodeling markers in multiple myeloma. Leukemia. lesions, and correlation of disease distribution with whole-body
2008;22:1973. doi:10.1038/leu.2008.216. MRI (WBMRI). Skeletal Radiol. 2008;38:225–36. doi:10.1007/
29. Anderson G, Gries M, Kurihara N. Thalidomide derivative CC- s00256-008-0607-4.
4047 inhibits osteoclast formation by down-regulation of PU.1. 45. Nanni C, Zamagni E, Farsad M, Castellucci P, Tosi P, Cangini D,
Blood. 2006;107:3098–105. doi:10.1182/blood-2005-08-3450. et al. Role of 18F-FDG PET/CT in the assessment of bone involve-
30. Gingery A, Bradley E, Shaw A, Oursler MJ. Phosphatidylinositol ment in newly diagnosed multiple myeloma: preliminary results.
3-kinase coordinately activates the MEK/ERK and AKT/NFκB Eur J Nucl Med Mol Imaging. 2006;33:525–31. doi:10.1007/
pathways to maintain osteoclast survival. J Cell Biochem. s00259-005-0004-3.
2003;89:165–79. doi:10.1002/jcb.10503. 46. Nanni C, Rubello D, Zamagni E, Castellucci P, Ambrosini V,
31. Giuliani N, Lunghi P, Morandi F, Colla S, Bonomini S, Hojden M, Montini G, Cavo M, Lodi F, Pettinato C, Grassetto G, Franchi R,
et al. Downmodulation of ERK protein kinase activity inhibits Gross MD, Fanti S. 18F-FDG PET/CT in myeloma with presumed
VEGF secretion by human myeloma cells and myeloma-induced solitary plasmocytoma of bone. In Vivo. 2008;22(4):513–7.
Curr Hematol Malig Rep

47. Rajkumar SV. Updated diagnostic criteria and staging system for 63. Dam ten MAGJ, Hilbrands LB, Wetzels JFM. Nephrotic syndrome
multiple myeloma, ASCO Educational Book. 2016. p. 1–6. induced by pamidronate. Med Oncol. 2010;28:1196–200. doi:10.
48. Rajkumar SV, Dimopoulos MA, Palumbo A, Blade J, Merlini G, 1007/s12032-010-9628-7.
Mateos M-V, et al. International Myeloma Working Group updated 64. Henry DH, Costa L, Goldwasser F, Hirsh V, Hungria V, Prausova J,
criteria for the diagnosis of multiple myeloma. Lancet Oncol. et al. Randomized, double-blind study of denosumab versus zole-
2014;15:e538–48. doi:10.1016/S1470-2045(14)70442-5. dronic acid in the treatment of bone metastases in patients with
49. Papamerkouriou YM, Kenanidis E, Gamie Z, Papavasiliou K, advanced cancer (excluding breast and prostate cancer) or multiple
Kostakos T, Potoupnis M, et al. Treatment of multiple myeloma myeloma. J Clin Oncol. 2011;29:1125–32. doi:10.1200/JCO.2010.
bone disease: experimental and clinical data. Expert Opin Biol 31.3304.
Ther. 2014;15:213–30. doi:10.1517/14712598.2015.978853. 65. Terpos E, Moulopoulos LA, Dimopoulos MA. Advances in imag-
50. Bartel TB, Haessler J, Brown TLY, Shaughnessy JD, van Rhee F, ing and the management of myeloma bone disease. J Clin Oncol.
Anaissie E, et al. F18-fluorodeoxyglucose positron emission to- 2011;29:1907–15. doi:10.1200/JCO.2010.32.5449.
mography in the context of other imaging techniques and prognos- 66. Raje N, Santo L. New Clinical and Research Directions in
tic factors in multiple myeloma. Blood. 2009;114:2068–76. doi:10. Myeloma-Related Bone Disease. Hematologist. 2015;12.
1182/blood-2009-03-213280. 67. Zangari M, Esseltine D, Lee C-K, Barlogie B, Elice F, Burns MJ,
51. Dammacco F, Rubini G, Ferrari C, Vacca A, Racanelli V. 18F-FDG et al. Response to bortezomib is associated to osteoblastic activation
PET/CT: a review of diagnostic and prognostic features in multiple in patients with multiple myeloma. Br J Haematol. 2005;131:71–3.
myeloma and related disorders. Clin Exp Med. 2014;15:1–18. doi: doi:10.1111/j.1365-2141.2005.05733.x.
10.1007/s10238-014-0308-3. 68. Heider U, Kaiser M, Muller C, Jakob C, Zavrski I, Schulz C-O,
52. Nishihori T, Song J, Shain KH. Minimal residual disease assess- et al. Bortezomib increases osteoblast activity in myeloma patients
ment in the context of multiple myeloma treatment. Curr Hematol irrespective of response to treatment. Eur J Haematol. 2006;77:
Malig Rep. 2016;11:118–26. doi:10.1007/s11899-016-0308-3. 233–8. doi:10.1111/j.1600-0609.2006.00692.x.
53. Mele A, Offidani M, Visani G, Marconi M, Cambioli F, Nonni M, 69. Giuliani N, Morandi F, Tagliaferri S, Lazzaretti M, Bonomini S,
et al. Technetium-99m sestamibi scintigraphy is sensitive and spe- Crugnola M, et al. The proteasome inhibitor bortezomib affects
cific for the staging and the follow-up of patients with multiple osteoblast differentiation in vitro and in vivo in multiple myeloma
myeloma: a multicentre study on 397 scans. Br J Haematol. patients. Blood. 2007;110:334–8. doi:10.1182/blood-2006-11-
2007;136:729–35. doi:10.1111/j.1365-2141.2006.06489.x. 059188.
54. Terpos E, Dimopoulos MA, Sezer O, Roodman D, Abildgaard N, 70. Terpos E, Heath DJ, Rahemtulla A, Zervas K, Chantry A,
Vescio R, et al. The use of biochemical markers of bone remodeling Anagnostopoulos A, et al. Bortezomib reduces serum dickkopf-1
in multiple myeloma: a report of the International Myeloma and receptor activator of nuclear factor-?B ligand concentrations
Working Group. Leukemia. 2010;24:1700–12. doi:10.1038/leu. and normalises indices of bone remodelling in patients with re-
2010.173. lapsed multiple myeloma. Br J Haematol. 2006;135:688–92. doi:
55. Ting KR, Brady JJ, Hameed A, Le G, Meiller J, Verburgh E, et al. 10.1111/j.1365-2141.2006.06356.x.
Clinical utility of C-terminal telopeptide of type 1 collagen in mul- 71. Hurchla MA, Garcia-Gomez A, Hornick MC, Ocio EM, Li A,
tiple myeloma. Br J Haematol. 2016;173:82–8. doi:10.1111/bjh. Blanco JF, et al. The epoxyketone-based proteasome inhibitors
13928. carfilzomib and orally bioavailable oprozomib have anti-
56. Giuliani N, Dalla Palma B, Bolzoni M. Bisphosphonates in multi- resorptive and bone-anabolic activity in addition to anti-myeloma
ple myeloma: preclinical and clinical data. Clinic Rev Bone Miner effects. Leukemia. 2012;27:430–40. doi:10.1038/leu.2012.183.
Metab. 2013;11:113–21. doi:10.1007/s12018-013-9143-4. 72. Hu B, Chen Y, Usmani SZ, Ye S, Qiang W, Papanikolaou X, et al.
57. Mhaskar R, Redzepovic J, Wheatley K, Clark OAC, Miladinovic Characterization of the molecular mechanism of the bone-anabolic
B, Glasmacher A, Kumar A, Djulbegovic B. Bisphosphonates in activity of carfilzomib in multiple myeloma. PLoS ONE. 2013;8,
multiple myeloma: a network meta-analysis. 2012. doi: 10.1002/ e74191. doi:10.1371/journal.pone.0074191.
14651858.CD003188.pub3 73. Zangari M, Aujay M, Zhan F, Hetherington KL, Berno T, Vij R,
58. Rosen LS, Gordon D, Kaminski M, Howell A, Belch A, Mackey J, et al. Alkaline phosphatase variation during carfilzomib treatment is
et al. Long‐term efficacy and safety of zoledronic acid compared associated with best response in multiple myeloma patients. Eur J
with pamidronate disodium in the treatment of skeletal complica- Haematol. 2011;86:484–7. doi:10.1111/j.1600-0609.2011.01602.x.
tions in patients with advanced multiple myeloma or breast carci- 74. Zhu YX, Kortuem KM, Stewart AK. Molecular mechanism of ac-
noma. Cancer. 2003;98:1735–44. doi:10.1002/cncr.11701. tion of immune-modulatory drugs thalidomide, lenalidomide and
59. Himelstein AL, Foster JC, Khatcheressian JL, Roberts JD, Seisler pomalidomide in multiple myeloma. Leuk Lymphoma. 2012;54:
DK, Novotny PJ, et al. Effect of longer-interval vs standard dosing 683–7. doi:10.3109/10428194.2012.728597.
of zoledronic acid on skeletal events in patients with bone metasta- 75. Breitkreutz I, Raab MS, Vallet S, Hideshima T, Raje N, Mitsiades
ses. JAMA. 2017;317:48. doi:10.1001/jama.2016.19425. C, et al. Lenalidomide inhibits osteoclastogenesis, survival factors
60. Morgan GJ, Davies FE, Gregory WM, Bell SE, Szubert AJ, Cook and bone-remodeling markers in multiple myeloma. Leukemia.
G, et al. Long-term follow-up of MRC myeloma IX trial: survival 2008;22:1925–32. doi:10.1038/leu.2008.174.
outcomes with bisphosphonate and thalidomide treatment. Clin 76. Chanan-Khan AA, Swaika A, Paulus A, Kumar SK, Mikhael JR,
Cancer Res. 2013;19:6030–8. doi:10.1158/1078-0432.CCR-12- Rajkumar SV, et al. Pomalidomide: the new immunomodulatory
3211. agent for the treatment of multiple myeloma. Blood Cancer J.
61. Weitzman R, Sauter N, Eriksen EF, Tarassoff PG, Lacerna LV, Dias 2013;3, e143. doi:10.1038/bcj.2013.38.
R, et al. Critical review: updated recommendations for the preven- 77. Webb SL, Edwards CM. Novel therapeutic targets in myeloma
tion, diagnosis, and treatment of osteonecrosis of the jaw in cancer bone disease. Br J Pharmacol. 2014;171:3765–76. doi:10.1111/
patients—May 2006. Crit Rev Oncol Hematol. 2007;62:148–52. bph.12742.
doi:10.1016/j.critrevonc.2006.12.005. 78. Munemasa S, Sakai A, Kuroda Y, Okikawa Y, Katayama Y, Asaoku
62. Markowitz GS, Appel GB, Fine PL, Fenves AZ, Loon NR, H, et al. Osteoprogenitor differentiation is not affected by immuno-
Jagannath S, et al. Collapsing focal segmental glomerulosclerosis modulatory thalidomide analogs but is promoted by low
following treatment with high-dose pamidronate. J Am Soc bortezomib concentration, while both agents suppress osteoclast
Nephrol. 2001;12:1164–72. differentiation. Int J Oncol. 2008. doi:10.3892/ijo.33.1.129.
Curr Hematol Malig Rep

79. Utzschneider S, Schmidt H, Weber P, Schmidt GP, Jansson V, Dürr 83. Fulciniti M, Tassone P, Hideshima T, Vallet S, Nanjappa P,
HR. Surgical therapy of skeletal complications in multiple myeloma. Ettenberg SA, et al. Anti-DKK1 mAb (BHQ880) as a potential
Int Orthop. 2010;35:1209–13. doi:10.1007/s00264-010-1127-0. therapeutic agent for multiple myeloma. Blood. 2009;114:371–9.
80. Wedin R. Surgical treatment for pathologic fracture. Acta Orthop. doi:10.1182/blood-2008-11-191577.
2001;72:1–29. doi:10.1080/000164701753759546. 84. Iyer SP, Beck JT, Stewart AK, Shah J, Kelly KR, Isaacs R, et al. A
81. Talamo G, Dimaio C, Abbi KKS, Pandey MK, Malysz J, Creer phase IB multicentre dose-determination study of BHQ880 in com-
MH, et al. Current role of radiation therapy for multiple myeloma. bination with anti-myeloma therapy and zoledronic acid in patients
Front Oncol. 2015;5:71. doi:10.3389/fonc.2015.00040. with relapsed or refractory multiple myeloma and prior skeletal-
82. Ontario Health Technology Assessment Series. Vertebral augmen- related events. Br J Haematol. 2014;167:366–75. doi:10.1111/bjh.
tation involving vertebroplasty or kyphoplasty for cancer-related 13056.
vertebral compression fractures: a systematic review. 2016;16(11):
1–202.

You might also like