You are on page 1of 11

M ET ABOL I SM CL IN I CA L A N D E XP E RI ME N TAL 80 ( 20 1 8 ) 8 0– 90

Available online at www.sciencedirect.com

Metabolism
www.metabolismjournal.com

Biology and treatment of myeloma related


bone disease

Evangelos Terpos⁎, Dimitrios Christoulas, Maria Gavriatopoulou


Department of Clinical Therapeutics, University of Athens School of Medicine, Alexandra General Hospital, Athens, Greece

A R T I C LE I N FO AB S T R A C T

Article history: Myeloma bone disease (MBD) is the most common complication of multiple myeloma (MM),
Received 20 July 2017 resulting in skeleton-related events (SREs) such as severe bone pain, pathologic fractures,
Accepted 18 November 2017 vertebral collapse, hypercalcemia, and spinal cord compression that cause significant
morbidity and mortality. It is due to an increased activity of osteoclasts coupled to the
Keywords: suppressed bone formation by osteoblasts. Novel molecules and pathways that are
Multiple myeloma implicated in osteoclast activation and osteoblast inhibition have recently been described,
Bone disease including the receptor activator of nuclear factor-kB ligand/osteoprotegerin pathway,
Receptor activator of nuclear factor- activin-A and the wingless-type signaling inhibitors, dickkopf-1 (DKK-1) and sclerostin.
kB ligand (RANKL) These molecules interfere with tumor growth and survival, providing possible targets for the
Wnt pathway development of novel drugs for the management of lytic disease in myeloma but also for the
treatment of MM itself. Currently, bisphosphonates are the mainstay of the treatment of
myeloma bone disease although several novel agents such as denosumab and sotatercept
appear promising. This review focuses on recent advances in MBD pathophysiology and
treatment, in addition to the established therapeutic guidelines.
© 2017 Elsevier Inc. All rights reserved.

1. Introduction tumor expansion and bone destruction. New insights into the
pathophysiology have enhanced our understanding of myeloma
Multiple myeloma (MM) is a relatively common hematological cell growth and bone destruction. The biologic pathway of the
malignancy characterized by the accumulation of abnormal receptor activator of nuclear factor-kappa B (RANK), its ligand
plasma cells in the bone marrow, the production of a monoclonal (RANKL), and osteoprotegerin (OPG) which is the decoy receptor
protein present in the blood or urine, and associated organ of RANKL is of major importance for the increased osteoclast
dysfunction presenting with osteolytic bone destruction, hyper- activity observed in MM [2]. More recently, activin-A has been
calcemia, renal failure, anemia, and an increased risk for implicated in MM bone disease, through stimulating RANK
infections. Myeloma Bone disease (MBD), in the form of lytic expression and inducing osteoclastogenesis [3]. On the other
lesions or osteopenia due to increased osteoclast activity, hand, Wnt signaling pathway inhibitors, such as dickkopf-1
accompanied by suppressed osteoblast function is one of the (DKK-1) and sclerostin, are important inhibitors of osteoblast
devastating consequences of myeloma [1]. MM cells inhibit function [4,5]. These insights will probably lead to better bone-
osteoblast differentiation and stimulate osteoclast function, directed therapies aimed at restoring bone homeostasis by
resulting in bone resorption and consequent MBD. In turn, targeting either osteoclast or osteoblast activity. This review
growth factors released by the increased bone resorptive process, summarizes the latest available data for the mechanisms of bone
also increase the growth of MM cells, creating a vicious cycle of destruction in MM and novel agents targeting MBD. (See Fig. 1.)

⁎ Corresponding author at: Department of Clinical Therapeutics, University of Athens School of Medicine, Alexandra General Hospital, 80 Vas.
Sofias Avenue, 11528 Athens, Greece.
E-mail addresses: eterpos@med.uoa.gr, eterpos@hotmail.com (E. Terpos).

https://doi.org/10.1016/j.metabol.2017.11.012
0026-0495/© 2017 Elsevier Inc. All rights reserved.
M ET ABOL I SM CL IN I CA L A N D E XP E RI ME N TAL 8 0 ( 20 1 8 ) 8 0– 9 0 81

Fig. 1 – Schematic overview of myeloma bone disease pathophysiology. Novel therapeutic targets are highlighted.

downstream signaling pathways required for osteoclast


2. Normal Bone Remodeling development, differentiation, and maturation. RANK is a
transmembrane signaling receptor, member of the tumor
Normal bone consists of a mineralized part and an organic necrosis receptor superfamily, which is mainly expressed on
part, made of collagen and non-collagen proteins. Osteocytes, the surface of osteoclast precursors, produced by bone marrow
osteoclasts, and osteoblasts maintain homeostasis in normal stromal cells (BMSCs), osteoblasts, and activated T-lymphocytes
physiologic states by balancing bone formation and bone [11,12]. Its expression is induced by cytokines that stimulate
resorption. Osteocytes make up 90% to 95% of all bone cells, bone resorption [13] such as parathyroid hormone (PTH), 1,25-
and osteoclasts and osteoblasts make up fewer than 10% [6]. dihydroxyvitamin D3 and prostaglandins [14,15]. Interestingly,
Osteoclasts are multinucleated cells originating from hemato- apoptotic osteocytes express RANKL stimulating osteoclast
poietic stem cells committed to monocyte–macrophage lineage. differentiation [16] and recruiting them to sites of remodeling.
They contain certain proteins, such as tartrate-resistant acid The important role of RANKL in normal osteoclastogenesis has
phosphatase (TRAP), tartrate-resistant trinucleotide phospha- been clearly shown in RANKL or RANK gene knockout mice.
tase, carbonic anhydrase II, calcitonin receptors, and a few These animals lack osteoclasts, and as result develop severe
cathepsins (lysosomal proteases) [7], whose main function is osteopetrosis [17–19]. OPG is a soluble decoy receptor for RANKL,
bone resorption. Osteoblasts are mononuclear cells originating member of the tumor necrosis factor receptor superfamily
from mesenchymal stem cells. They contain the enzyme [20]. It is produced by osteoblasts, as well as BMSCs and blocks
alkaline phosphatase, which is used as a marker of osteoblastic the interactions of RANKL with RANK, thereby limiting
activity [8]. Their normal location is near the bone surface where osteoclastogenesis. It is regulated by interleukin 1 beta (IL-1b),
new bone is laid down and their main function is bone tumor necrosis factor alpha (TNF-a), transforming growth factor
formation, by collagen synthesis, osteocalcin production, and beta (TGF-b), estradiol and 17b-estriol. OPG-deficient mice
mineralization [9]. Osteoblasts that become a part of mineralized develop severe osteopenia and osteoporosis [21–23]. In normal
matrix are called osteocytes. Bone remodeling is a continuous subjects, the RANKL/OPG ratio is very low. However, an
process, consisting of old bone resorption (osteoclastic activity) abnormal RANKL/OPG ratio is found both in benign and
and new bone formation (osteoblastic activity). This process is malignant bone diseases [24].
well balanced in a normal person to keep the bones in healthy Osteocytes also regulate osteoblasts by secreting
form [10]. Osteocytes serve as the main regulators of bone sclerostin and DKK1, which block canonical Wnt signaling
homeostasis between osteoclasts and osteoblasts by secreting pathway via binding to low-density lipoprotein receptor–
several cytokines that regulate the activity of these cells, such as related proteins 5 and 6 (LRP5/LRP6) on the surface of
sclerostin, DKK1, RANKL, and OPG [6]. osteoblasts [6]. Binding of Wnt glycoproteins to the Wnt
Osteoclasts are regulating RANK, its ligand RANKL, and receptor and its co-receptors LRP5/LRP6 leads to stabilization
OPG signaling pathway. RANK-RANKL signaling activates of β-catenin. In turn, β-catenin accumulates in cytoplasm,
82 M ET ABOL I SM CL IN I CA L A N D E XP E RI ME N TAL 80 ( 20 1 8 ) 8 0– 90

translocates into the nucleus and stimulates expression from patients with MM show a correlation among tumor
of osteoblastic target genes [25]. If the Wnt signal is blocked, burden, osteoclast numbers, and resorptive surface [33,34].
β-catenin is phosphorylated and degraded by the proteasome. Osteoclast activity also reflects disease activity [2,35]. The
There are many ways of Wnt signaling pathway inhibition: adherence of abnormal myeloma cells in the BMSCs, through
Members of the dickkopf (DKK) family bind to the LRP5/LRP6 the binding of vascular cell adhesion molecule-1 (VCAM-1) on
component of the Wnt receptor complex, while secreted stromal cells and α4β1 integrin on MM cells, leads to the
frizzled-related proteins (sFRP), such as sFRP-2 and sFRP-3, upregulation of a broad spectrum of factors which stimulate
bind to Wnt proteins. Sclerostin inhibits the Wnt pathway by osteoclast formation, differentiation and activity, as well as
antagonizing Wnt proteins for the binding to LRP-5/6 receptor. suppression of negative regulators of osteoclastogenesis. These
All result in a suppression of Wnt-signaling and a reduced cytokines and chemokines include macrophage-colony stimu-
osteoblast function. lating factor (M-CSF), interleukin-6 (IL-6), IL-1α, IL-1β, IL-11, C-C
Besides the central role of osteocytes, osteoblasts and motif ligand 3 (CCL3, also known as macrophage inflammatory
BMSCs also express OPG and RANKL and regulate osteoclast protein 1-alpha, MIP-1-α), tumor necrosis factor-alpha and beta
differentiation. Because OPG is a Wnt canonical signaling target (TNF-α, -β), parathyroid hormone-related peptide (PTHrP), and
[26], osteocytes also regulate osteoclast differentiation via vascular endothelial growth factor (VEGF) [36–38]. However,
regulation of Wnt signaling pathway activity in osteoblasts. over the last years it has been established that the RANKL and
However, osteoclasts express semaphorin 4D (SEMA4D), which OPG system plays the major role in osteoclastogenesis in MM.
inhibits osteoblast differentiation [27].
3.1.1. The RANK/RANKL Signaling Pathway
An increase in the RANKL/OPG ratio results in bone loss in
3. Bone Disease in MM inflammatory diseases, including rheumatoid arthritis and in
several cancers [39–41]. Myeloma cells promote an imbalance in
In MM, the osteocyte-osteoclast-osteoblast axis is disrupted, the RANKL/OPG system in the tumor microenvironment: OPG
leading to an uncoupled bone remodeling, in which bone expression is decreased, whereas RANKL expression is upregu-
construction decreases and bone resorption increases [28]. lated in bone marrow biopsies of MM patients [42]. Several
Dysregulation of osteoblasts and osteoclasts serves as the studies show that myeloma cells both induce the RANKL
key process in the pathogenesis of myeloma bone disease. expression by stromal cells within the bone microenvironment
Increased osteoclasts’ activity in MM patients will promote through direct cell to cell contact [43] and directly express RANKL
bone resorption, while suppression of osteoblasts’ activity [44–46]. On the other hand, myeloma cells decrease the
can lead to impaired bone formation, with the consequent OPG availability within the bone microenvironment through
formation of pathognomonic osteolytic lesions. In turn, reducing OPG secretion by osteoblasts and stromal cells [47].
growth factors released by the increased bone resorptive They also produce syndecan-1 (CD138), a transmembrane proteo-
process, also increase the growth of MM cells, creating a glycan that binds to the heparin-binding domain of OPG and
vicious cycle of tumor expansion and bone destruction. mediates its internalization and consecutively lysosomal degra-
MDB is distinct from other metastatic diseases as there is dation by myeloma cells [48]. The combination of these effects
no bone formation whereas in cancers like prostate and results in an increased RANKL/OPG ratio in the bone marrow
breast, the osteoclastic, as well as osteoblastic activity, is microenvironment that favors the formation and activation of
increased [29]. osteoclasts. A positive correlation is seen between the number of
In a prospective, single center study on myeloma patients osteolytic lesions and increasing levels of serum RANKL [45].
using femoral neck dual energy X-ray absorptiometry (DXA) Among patients with MM, those with lower levels of total
and lumbar spine quantitative computed tomography (QCT), RANKL exhibit a significantly longer progression-free survival
66% of patients had osteoporosis defined by either lumbar spine (PFS) compared with other patients [49]. Levels of OPG and
or femoral neck bone density (T-score <−2.5). The severity RANKL correlate with poor prognosis and severity of bone
of osteoporosis, as defined by initial BMD T-score values, disease [2]. Therefore, the RANKL-OPG axis is an important
significantly impacted on patient survival [30]. In another target in the development of novel therapeutic strategies for
population-based retrospective cohort study, 165 patients MM bone disease.
with myeloma were followed for 537 person-years. In the year
before diagnosis, 16 times more fractures were observed than 3.1.2. Activin-A
expected, mostly pathologic fractures of the vertebrae and ribs. Activin-A is a dimeric multifunctional glycoprotein, member of
Subsequently, there was a 9-fold increase in fracture risk [31]. the transforming growth factor-β (TGF-β) superfamily. Activin-A
The clinical and economic impact of the associated bone pain, regulates a broad spectrum of biological functions, including
pathological fractures requiring surgery and/or radiation to hormonal homeostasis, gonadal function, inflammation, muscle
bone, spinal cord compression and hypercalcaemia can be growth and immunity [50,51]. Osteoblasts produce follistatin (an
devastating [32]. activin-A antagonist) and activin-A in a differentiation-
dependent manner, leading to autocrine regulation of extracel-
3.1. Pathogenesis of the Increased Osteoclast Activity lular matrix formation and mineralization [52]. Activin-A stimu-
in Myeloma lates osteoclasts and has possibly inhibitory effects on osteoblast
function [53,54]. It is elevated in myeloma patients with advanced
The pathogenesis of bone disease in MM is primarily the result disease and correlates with adverse disease features, including
of generalized osteoclast activation. Bone marrow biopsies inferior survival and extensive bone disease [55].
M ET ABOL I SM CL IN I CA L A N D E XP E RI ME N TAL 8 0 ( 20 1 8 ) 8 0– 9 0 83

3.2. Osteoblast Function is Also Impaired in MM


4. Treatment
In the early phase of MM there is an up-regulation of osteoblast
precursors [28]. The interaction of MM cells with BMSCs initiates 4.1. Bisphosphonates – The Mainstay of Therapy
the production of IL-1 and TNFα; in turn, these cytokines recruit
osteoblasts that produce IL-6, a potent myeloma cell growth Bisphosphonates are the standard of care for MBD.
factor. Nevertheless, as disease progresses, osteoblast function Bisphosphonates are pyrophosphate analogues that inhibit
and maturation are inhibited, resulting in net osteolysis, as the farnesyl pyrophosphate synthase [67]. They readily bind the
reducing bone formation rate fails to counter increased exposed mineralized bone and are taken up by osteoclast
resorption [56]. In bone biopsies from MM patients, during bone resorption, inhibiting osteoclast recruitment and
histomorphometric evaluation of osteoblast activity has maturation, preventing the development of monocytes into
revealed osteoblast inhibition, since no evidence of bone osteoclasts, inducing osteoclast apoptosis and interrupting
regeneration was detectable within the skeleton lesions or in their attachment to the bone [68]. Before the advent of
their vicinity [57]. Osteoblast apoptosis is markedly increased effective bisphosphonate therapy, bone healing occurred
due to high cytokine levels and physical interaction between uncommonly in myeloma and was delayed in treated
osteoblasts and MM cells. In vitro studies revealed that patients, despite responses to chemotherapy.
osteoblast growth and function are inhibited when cocultured Oral clodronate, intravenous pamidronate, and intrave-
with myeloma cells or in medium conditioned by myeloma nous zoledronic acid have been licensed for the management
cells, suggesting that this effect is due to soluble osteoblast of myeloma bone disease [69–72]. Etidronate and ibandronate
inhibiting factors [57]. Once deregulated by these inhibitory were found to be ineffective in myeloma patients [73,74].
factors, the osteoblast function remains almost permanently
ineffective after conventional chemotherapy even in responding 4.1.1. Bisphosphonates Head to Head
patients. In recent years, more studies are focusing on the A large phase III, randomized, double-blind, study was
osteoblast-related inhibitory pathways [58]. Functional exhaus- performed to compare pamidronate to zoledronic acid [75].
tion of osteoblasts has been also postulated by the inverse There was no difference in time to the first skeletal related
relation between biochemical indicators of osteoid production, event (SRE) between treatment groups, but the skeletal
namely serum osteocalcin and bone-specific alkaline phospha- morbidity rate was slightly higher in patients treated with
tase, and the presence of osteolytic lesions. pamidronate. However, use of radiation to bone was signifi-
cantly higher in patients treated with pamidronate compared
3.2.1. Dickkopf-1 (DKK-1) Protein with zoledronic acid (p = 0.018). A long-term extension phase
DKK-1 is expressed by osteoblasts and BMSCs and it antagonizes of this study confirmed the equivocal findings that zoledronic
the Wnt pathway resulting in inhibition of osteoblasts matura- acid and pamidronate had similar efficacy in reducing the risk
tion and new bone formation. DKK-1 is also secreted by myeloma of skeletal-related events in MM patients [76]. In the Myeloma
cells and has been shown to inhibit differentiation of osteoblast IX trial (conducted by the Medical Research Council [MRC] of
precursor cells in vitro. In MM patients with lytic lesions, the United Kingdom), zoledronic acid (4 mg IV every 3 to
immunohistochemical analysis of bone marrow biopsies 4 weeks, dose adjusted based on creatinine clearance rates)
showed that myeloma cells overexpress DKK-1. In fact, bone significantly reduced the proportion of patients with on-study
marrow plasma from newly diagnosed MM patients contains SREs compared with clodronate (1600 mg orally daily) in
nearly 3 times more DKK-1 protein compared to control subjects patients with newly diagnosed, symptomatic MM [77]. After
and DKK-1 is increased in the serum of MM patients as well [59]. a median follow-up of 3.7 years, 35% of patients receiving
Furthermore, gene expression levels and serum levels of DKK-1 clodronate had experienced SREs versus 27% of patients
correlate with the extent of bone disease [60,61]. As a result of receiving zoledronic acid (p = 0.004). Zoledronic acid also
high expression of DKK1, mesenchymal stem cells do not reduced mortality by 16% (95% CI 4–26) versus clodronate
differentiate to osteoblasts [60]. On the other hand, since Wnt (hazard ratio [HR] 0.84, 95% CI 0.74–0.96; p = 0.0118), extended
signaling pathway in osteoblasts increases the expression of OPG median overall survival by 5.5 months (50 months, vs.
and downregulates the expression of RANKL [62,63], inhibition of 44.5 months, p = 0.04) and increased median progression-
Wnt signaling pathway promotes osteoclastogenesis. free survival by 2 months (19.5 months, vs. 17.5 months; p =
0.07). These results confirm preclinical studies suggesting
3.2.2. Sclerostin direct anti-myeloma effect of zoledronic acid [78]. Possible
Patients with active myeloma have elevated circulating mechanisms include the expansion of gamma/delta T-cells
sclerostin, which correlates with advanced disease features with possible anti-MM activity and the reduction of IL-6
including severe bone disease. MM patients who present with secretion by bone marrow stromal cells.
fractures at diagnosis have very high levels of circulating
sclerostin compared with all others, whereas sclerostin 4.1.2. Bisphosphonates Adverse-Events
correlates negatively with bone specific alkaline phosphatase Intravenous bisphosphonates have the potential to cause
and positively with C-telopeptide of collagen type-1 (a bone acute or chronic renal dysfunction: approximately 12% of
resorption marker) [64]. The source of sclerostin in MBD patients with MM, breast cancer, or other solid tumors
remains to be defined, since very little or no sclerostin or evidenced renal deterioration (changes in serum creatinine)
SOST messenger RNA expression in primary MM cells from at 24 months of treatment, although in the MRC Myeloma IX
patients or MM cell lines has been detected [65,66]. trial [77] there was no difference regarding renal impairment
84 M ET ABOL I SM CL IN I CA L A N D E XP E RI ME N TAL 80 ( 20 1 8 ) 8 0– 90

between intravenous zoledronic acid and oral clodronate arms. recent study suggests that decreasing bone turnover through
Kidneys, being responsible for 40% of bisphosphonates' excre- long-term antiresorptive treatment not only changes bone’s
tion via glomerular filtration and active tubular excretion, are nanoscale material properties but also affects toughness on the
particularly sensitive to bisphosphonates. The nephrotoxicity is length scale of hundreds of micrometers through reductions in
related to the dose, infusion time, and maximum plasma extrinsic and intrinsic toughening mechanisms [91].
concentration that affect the intracellular bisphosphonate
concentration. Zoledronic acid dosages should be reduced in 4.1.3. Optimal use of Bisphosphonates
patients who have preexisting renal impairment (creatinine Numerous guidelines have been published by different associa-
clearance of 30–60 mL/min). Zoledronic acid has not been tions and study groups for the optimal use of bisphosphonates in
studied in patients with severe renal impairment; and it is not MM patients. The common resultant of all these, with little
recommended in this setting. Pamidronate 90 mg infused over variation, is that bisphosphonates should be administered
4–6 h is recommended for patients with extensive bone disease monthly over a 2-year period in patients with lytic bone disease
and existing severe renal impairment [79]. After withholding on plain radiographs and patients with osteopenia or osteopo-
zoledronic acid or pamidronate in patients who develop renal rosis. The International Myeloma Working Group (IMWG)
deterioration during therapy, bisphosphonate therapy can be recommends that bisphosphonates should be given until disease
resumed, at previous dosage, when serum creatinine returns to progression in patients without a complete response or very
within 10% of baseline. good partial response [87] based on the MRC Myeloma IX trial
Osteonecrosis of the jaw (ONJ) is a rather uncommon (varying findings of improvements in overall survival and reductions in
from 2 to 10%), but potentially serious complication of SREs in patients who received bisphosphonate treatment for
intravenous bisphosphonates, characterized by the presence more than 2 years [92]. However, for patients with a complete
of exposed bone in the mouth [80,81]. Cumulative incidence of response or a very good partial response, the optimal duration of
ONJ increases with longer exposure. Invasive dental procedures bisphosphonates is an active area of investigation, given that
are a major risk factor for the development of ONJ [80]. Other prolonged exposure to bisphosphonates may increase the risk
risk factors include poor oral hygiene, age, and duration of for side effects. Currently, the IMWG panel recommends 12 to
myeloma. Zoledronic acid was associated with a higher 24 months of treatment (timed from the start of treatment) and
incidence of ONJ in retrospective evaluations [82]. This was then continuation at the discretion of the provider. For those
confirmed in the MRC Myeloma IX trial [77] in which zoledronic patients who discontinue bisphosphonate use after 2 years, the
acid was associated with higher rates of confirmed ONJ drug should be re-administered if relapse occurs.
(4%) than was clodronate (less than 1%; p < 0.0001). In Data from retrospective studies suggest that a reduced
approximately one half of patients, ONJ lesions will heal [83], schedule (monthly administration of zoledronic acid during the
but approximately one half of patients who restart bisphos- first year and then every 3 months) may be safer than the
phonate therapy after having stopped it will develop recurrence standard monthly schedule, while maintaining antiresorptive
of ONJ. Before initiation of bisphosphonates and during efficacy [93]. Similar studies focusing in lowering bisphosphonate
treatment, dental status should be monitored and good dose have promising results: 30 mg of pamidronate vs. 90 mg did
oral hygiene maintained. Dental conditions should be treated not show any difference in skeletal related events between the
before starting bisphosphonate therapy. Unnecessary invasive two doses tested [94]. Measurement of serum C-telopeptide
dental procedures should be avoided. Where dental procedures pyridinoline crosslinks of type I collagen (ICTP), serum carboxy-
are required, patients should be treated conservatively, terminal cross-linking telopeptide of type I collagen (CTX) and
minimizing invasive procedures. Temporary suspension of urinary NTX (all markers of bone resorption) is recommended in
bisphosphonates treatment should be considered if invasive monitoring bone turnover during anti-myeloma therapies [95]
dental procedures are necessary. Preventive measures during to establish their efficacy in bone disease. In this context, the
bisphosphonate treatment have the potential to reduce the Z-MARK study evaluated efficacy and safety of less-frequent
incidence of ONJ about 75% [84]. Antibiotic prophylaxis may zoledronic acid dosing based on bone turnover markers in
prevent ONJ occurrence after dental procedures [85]. Management patients with 1 to 2 years of prior bisphosphonate therapy [96].
of patients depends on ONJ stage, ranging from oral antimicrobial Patients received zoledronic acid (4 mg) every 4 or 12 weeks
rinses to culture-directed long-term antimicrobial therapy and based on urinary N-telopeptide of type 1 collagen (uNTX) levels
surgical debridement/resection to reduce the volume of necrotic (every 4 weeks if uNTX ≥50 nmol/mmol creatinine, every
bone [86]. Initial therapy of ONJ should include discontinuation 12 weeks if uNTX <50). Patients in whom an SRE or disease
of bisphosphonates until healing occurs [87]. progression developed were treated on the every-4-weeks
There is evidence that patients with osteoporosis using schedule thereafter. SRE occurred in only 5.8% of patients in
nitrogen-containing bisphosphonates (pamidronate and zoledro- year 1 and 4.9% of patients in year 2, suggesting that less
nic acid) therapy have an increased risk of congestive heart failure frequent dosing of zoledronic acid beyond 1 to 2 years
and atrial fibrillation. This potential risk should be weighed maintains a low SRE rate and can be safely administered.
against the reduction in the risk of osteoporotic fractures [88,89].
Bisphosphonates have been associated with atypical femoral 4.2. RANKL/RANK Pathway Regulators – Targeting
fractures (AFFs), rare fractures with a transverse, brittle mor- the Osteoclast
phology [90]. The underlying cause of AFFs and their causal
relationship to bisphosphonates is unknown and prospective 4.2.1. RANKL Inhibitors
studies investigating fracture pattern, microscopic bone pathol- Preclinical models of MM demonstrated that RANKL inhibition
ogy and pharmacologic exposures should be conducted. A can prevent bone destruction caused by myeloma cells. RANKL
M ET ABOL I SM CL IN I CA L A N D E XP E RI ME N TAL 8 0 ( 20 1 8 ) 8 0– 9 0 85

inhibition with recombinant RANK-Fc protein not only reduced groups; however, an ad hoc overall survival analysis in the
MM-induced osteolysis, but also caused a marked decline in MM subset of patients (n = 180) favored zoledronic acid
tumor burden [44,97]. Similar results were obtained using (hazard ratio (HR) 2.26; 95% confidence interval (CI) 1.13-4.50;
recombinant OPG for the treatment of MM-bearing animals p = 0.014) [106]. Nevertheless, for MM, imbalances in baseline
[98]. These data gave the rationale for using RANKL inhibition in disease characteristics, stem cell transplant, and withdrawals
the clinical setting. (due to consent withdrawal or loss to follow-up) favored
Denosumab is a fully human monoclonal antibody that binds zoledronic acid. These baseline and on-study imbalances may
RANKL with high affinity and specificity and inhibits RANKL- partially account for the difference observed between treat-
RANK interaction, mimicking the endogenous effects of OPG. It is ment groups [107]. To address the discrepancies definitively, a
highly specific because it binds only to RANKL and not to other randomized, double-blind, multicenter phase 3 study is
members of the TNF family, including TNF-α, TNF-β, TNF-related currently comparing denosumab with zoledronic acid in the
apoptosis-inducing ligand, and CD40 ligand [99]. Denosumab treatment of bone disease in patients with newly diagnosed
inhibits bone resorption, increases cortical and cancellous bone MM. A total of 1718 pts were randomized, 859 to each arm. The
mass, and improves trabecular microarchitecture in knock-in study was presented only as abstracts to-date. Denosumab
mice that express chimeric (murine/human) RANKL [100]. demonstrated non-inferiority to zoledronic acid in delaying
Denosumab has been approved for increasing bone density in time to first on-study SRE in myeloma patients. A landmark
patients with osteoporosis and for preventing SREs in patients analysis at 15 months suggested a significant benefit for
with metastatic bone disease [101]. denosumab with respect to time to first SRE. The rates of renal
In a phase I trial, 25 patients with MM with radiologically adverse events were significantly lower in denosumab patients
confirmed bone lesions, received a single dose of either [108,109]. A phase II study of denosumab in MM patients with
denosumab or pamidronate. Denosumab decreased bone renal insufficiency is ongoing (NCT02833610).
resorption within 24 h of administration, as reflected by levels Denosumab appears to have little toxicity, mainly asthenia,
of urinary and serum NTX. That was similar in magnitude but and multiple phase III trials of denosumab in patients with solid
more sustained than with intravenous pamidronate [102]. tumors and bone metastasis are ongoing. However it is crucial
These results were confirmed in another phase I trial, in to mention that RANKL is involved in dendritic cell survival
which denosumab was given at multiple doses [103]. [110] and that the anti-RANKL strategy may have an effect on
In a phase II trial, the ability of denosumab to affect bone the immune system and a possible increase in infection rate,
resorption markers or to reduce serum M-protein levels in especially in cancer patients who have already had severe
relapsed or plateau-phase myeloma subjects was evaluated. All immunodeficiency.
subjects received denosumab monthly (120 mg given as a
subcutaneous injection), with loading doses on days 8 and 15 4.2.2. Activin Inhibitors
of the first month, until disease progression or subject discon- Activin is an osteoclast agonist that is involved in osteoclast
tinuation. Results of this study demonstrated that no subjects in development and differentiation through stimulation of RANK
either cohort met the protocol-defined objective response expression, as well as an osteoblast antagonist [3]. It is secreted
criteria of complete response (CR) or partial response (PR), but by BMSCs and osteoclasts in MBD. In the preclinical setting,
denosumab effectively inhibited the RANKL pathway regardless RAP-011, a fusion protein of the extracellular domain of the
of previous exposure to bisphosphonates, as evidenced by high-affinity activin receptor IIA (ActRIIA) and human immu-
suppressed levels of the bone turnover marker, serum CTX, by noglobulin G (IgG) Fc domain with potent inhibitory effect on
more than 50% [104]. In another phase II trial Fizazi et al activin, prevented the formation of osteolytic lesions in a
evaluated the effect of denosumab in patients with bone murine MM model by stimulating bone formation through
metastases and elevated urinary NTX levels despite ongoing osteoblasts, while having no effect on osteoclast activity [111].
intravenous bisphosphonate therapy. Patients were stratified by In ovariectomized mice with prior bone loss, RAP-011 increased
tumor type (total 111 patients; 9 patients with MM, 50 patients bone formation, mass and strength [112].
with prostate cancer, 46 patients with breast cancer and 6 Sotatercept (ACE-011) is the humanized counterpart of
patients with other solid tumor) and screening NTX levels and RAP-011. In non-human primates sotatercept has been shown
randomly assigned to continue intravenous bisphosphonates to increase bone mass and enhance bone strength [113,114].
every 4 weeks or receive subcutaneous denosumab 180 mg In a phase 1 study in healthy postmenopausal volunteers,
every 4 weeks or every 12 weeks. Denosumab normalized single-dose sotatercept was associated with increased serum
urinary NTX levels more frequently than the continuation of levels of the bone formation marker bone-specific alkaline
intravenous bisphosphonate (64% vs. 37% respectively; p = 0.01), phosphatase (bALP) and decreased bone resorption markers
while fewer patients receiving denosumab experienced on- CTX and TRACP-5b, reflecting an increase in bone formation
study SREs than those receiving intravenous bisphosphonate and a decrease in bone resorption respectively [115]. No safety
(8% vs. 17% respectively) [105]. This study showed that concerns were noted in this study.
denosumab inhibits bone resorption and prevents SREs even in In a multicenter phase 2 trial, patients with osteolytic bone
patients who are refractory to bisphosphonate therapy. lesions due to MM were randomized to receive either four 28-day
In a phase 3 trial of denosumab vs zoledronic acid in cycles of sotatercept or placebo as subcutaneous injection with
patients (n = 1776) with bone metastases and solid tumors or concomitant anticancer therapy consisting of oral melphalan,
MM, denosumab was superior to zoledronic acid for the prednisolone and thalidomide (MPT). Sotatercept treatment
primary end point of prevention of skeletal-related events. demonstrated clinically significant increases in biomarkers of
There was no difference in overall survival between the two bone formation, decreases in bone pain, and anti-tumor activity,
86 M ET ABOL I SM CL IN I CA L A N D E XP E RI ME N TAL 80 ( 20 1 8 ) 8 0– 90

as well as increase in haemoglobin levels [116], but further bisphosphonates should follow approved indications, with
research is needed to support these findings. adjustments if necessary.
It has been shown in vitro that lenalidomide stimulates Over the last few years, study of normal bone metabolism
secretion on BMSCs via an AKT-mediated increase in JNK signaling and mechanisms of myeloma bone disease, has identified
[117]. A clinical trial with sotatercept in combination with several factors, which provide novel targets for treating patients
lenalidomide/dexamethasone or pomalidomide/dexamethasone with MM bone disease. The identification of the role of RANKL/
is ongoing in MM (NCT01562405). OPG pathway in MM has led to the development of the RANKL
inhibitor denosumab which may be incorporated as therapy
4.3. Wnt Pathway Regulators – Helping the Osteoblast for bone disease in the near future, after its first approval
for osteoporosis patients. Sotatercept, an activin inhibitor,
4.3.1. DKK-1 Antagonists enhances the deposition of new bone tissue and prevents bone
DKK-1 plays an important role in the dysfunction of osteoblasts loss in addition to antitumor activity. Antibodies to DKK-1 and
observed in MM. The effect of anti-DKK1 therapy on tumor sclerostin are currently under investigation in clinical trials in
growth and bone metabolism has been tested in mice. patients with MM and the first results are highly anticipated.
Antibodies neutralizing DKK-1 restored the bone mineral Advances in the treatment of MBD will translate into a better
density of the implanted myelomatous bone in mice, reduced quality of life for patients.
number of multinucleated TRAP-expressing osteoclasts and
increased the numbers of osteocalcin-expressing osteoblasts.
Furthermore, the anti-DKK1–treated mice showed reduced
tumor burden [118]. In addition, inhibition of DKK1 reduced Conflicts of Interest Statement
tumor growth, as an indirect effect via modification of the
tumor microenvironment [119]. ET discloses research support from Amgen and Janssen;
BHQ880, an IgG antibody, the first-in-class, fully human honoraria from Amgen, Janssen, Celgene and Novartis.
anti-DKK-1 neutralizing antibody seems to promote bone DC and MG have nothing to disclose.
formation and thus it inhibits tumor-induced osteolytic disease No grants are related to this paper.
in preclinical studies [119]. Inhibiting DKK-1 with BHQ880 in the
5T2MM murine model of myeloma reduced the development of
osteolytic bone lesions and in vivo growth of MM cells [120]. A
phase I/II study of BHQ880 in combination with zoledronic acid REFERENCES
in relapsed or refractory myeloma patients has demonstrated
increases in bone strength at the spine and hip [121]. [1] Christoulas D, Terpos E, Dimopoulos MA. Pathogenesis and
management of myeloma bone disease. Expert Rev Hematol
4.3.2. Sclerostin Antagonists 2009;2:385–98.
Inhibition of sclerostin could be an important strategy in the [2] Terpos E, Szydlo R, Apperley JF, Hatjiharissi E, Politou M,
treatment of bone diseases with a high catabolic rate. Several Meletis J, et al. Soluble receptor activator of nuclear factor
kappaB ligandosteoprotegerin ratio predicts survival in
studies have already demonstrated the importance of sclerostin
multiple myeloma: proposal for a novel prognostic index.
in osteoporosis [122,123]. Clinical trials with the experimental Blood 2003;102:1064–9.
sclerostin-neutralizing antibody romosozumab for the treat- [3] Sugatani T, Alvarez UM, Hruska KA. Activin A stimulates
ment of postmenopausal osteoporosis are ongoing. Preliminary IkappaB-alpha/NFkappaB and RANK expression for osteoclast
results have shown an increase in bone mineral density [124]. differentiation, but not AKT survival pathway in osteoclast
precursors. J Cell Biochem 2003;90:59–67.
[4] Gavriatopoulou M, Dimopoulos MA, Christoulas D, Migkou
M, Iakovaki M, Gkotzamanidou M, et al. Dickkopf-1: a
5. Conclusion
suitable target for the management of myeloma bone
disease. Expert Opin Ther Targets 2009;13:839–48.
Osteolytic bone disease is a central feature of MM and is caused [5] Moester MJ, Papapoulos SE, Löwik CW, van Bezooijen RL.
by factors produced by tumor cells that stimulate osteoclasts to Sclerostin: current knowledge and future perspectives.
resorb bone and inhibit osteoblast activity. The resulting Calcif Tissue Int 2010;87:99–107.
osteolytic lesions and/or osteoporosis and their clinical conse- [6] Bonewald LF. The amazing osteocyte. J Bone Miner Res 2011;
26:229–38.
quences, including fracture and severe pain, diminish both
[7] Janckila AJ, Takahashi K, Sun SZ, Yam LT. Tartrate-resistant
survival and quality of life, with major economic impact.
acid phosphatase isoform 5b as serum marker for osteoclastic
Bisphosphonates remain the cornerstone of therapeutic activity. Clin Chem 2001;47:74–80.
management of myeloma bone disease, offering considerable [8] George J, Kuboki Y, Miyata T. Differentiation of mesenchymal
benefit in preventing or delaying skeletal-related events and stem cells into osteoblasts on honeycomb collagen scaffolds.
relieving pain. Pamidronate and zoledronic acid seem to be Biotechnol Bioeng 2006;95:404–11.
equally potent in treating bone disease manifestations, but the [9] McSheehy PM, Chambers TJ. Osteoblast-like cells in the
presence of parathyroid hormone release soluble factor that
latest MRC-IX data supporting survival benefit for patients
stimulates osteoclastic bone resorption. Endocrinology
treated with zoledronic acid versus clodronate have revealed 1986;119:1654–9.
zoledronic acid as the preferred bisphosphonate in myeloma [10] Phill PA. Bone remodelling. Br J Orthod 1998;25:101–7.
patients. However preventative steps should be taken to avoid [11] Hsu H, Lacey DL, Dunstan CR, Solovyev I, Colombero A,
renal impairment and osteonecrosis of the jaw while dosing of Timms E, et al. Tumor necrosis factor receptor family
M ET ABOL I SM CL IN I CA L A N D E XP E RI ME N TAL 8 0 ( 20 1 8 ) 8 0– 9 0 87

member RANK mediates osteoclast differentiation and survival in myeloma patients: a 13-year prospective study.
activation induced by osteoprotegerin ligand. Proc Natl Am J Hematol 2011;86:617–9.
Acad Sci U S A 1999;96:3540–5. [31] Melton III LJ, Kyle RA, Achenbach SJ, Oberg AL, Rajkumar SV.
[12] Nakagawa N, Kinosaki M, Yamaguchi K, Shima N, Yasuda H, Fracture risk with multiple myeloma: a population-based
Yano K, et al. RANK is the essential signaling receptor for study. J Bone Miner Res 2005 Mar;20(3):487–93.
osteoclast differentiation factor in osteoclastogenesis. [32] Bruce NJ, McCloskey EV, Kanis JA, Guest JF. Economic impact
Biochem Biophys Res Commun 1998;253:395–400. of using clodronate in the management of patients with
[13] Boyle WJ, Simonet WS, Lacey DL. Osteoclast differentiation multiple myeloma. Br J Haematol 1999;104:358–64.
and activation. Nature 2003;423:337–42. [33] Valentin-Opran A, Charhon SA, Meunier PJ, Edouard CM,
[14] Yasuda H, Shima N, Nakagawa N, Yamaguchi K, Kinosaki M, Arlot ME. Quantitative histology of myeloma-induced bone
Mochizuki S, et al. Osteoclast differentiation factor is a changes. Br J Haematol 1982;52:601–10.
ligand for osteoprotegerin/osteoclastogenesis-inhibitory [34] Taube T, Beneton MN, McCloskey EV, Rogers S, Greaves M,
factor and is identical to TRANCE/RANKL. Proc Natl Acad Sci Kanis JA. Abnormal bone remodelling in patients with
U S A 1998;95:3597–602. myelomatosis and normal biochemical indices of bone
[15] Hofbauer LC, Heufelder AE. Osteoprotegerin and its cognate resorption. Eur J Haematol 1992;49:192–8.
ligand: a new paradigm of osteoclastogenesis. Eur J [35] Politou M, Terpos E, Anagnostopoulos A, Szydlo R, Laffan M,
Endocrinol 1998;139:152–4. Layton M, et al. Role of receptor activator of nuclear factor-kappa
[16] Kogianni G, Mann V, Noble BS. Apoptotic bodies convey B ligand (RANKL), osteoprotegerin and macrophage protein
activity capable of initiating osteoclastogenesis and localized 1-alpha (MIP-1a) in monoclonal gammopathy of undetermined
bone destruction. J Bone Miner Res 2008;23:915–27. significance (MGUS). Br J Haematol 2004;126:686–9.
[17] Dougall W-C, Glaccum M, Charrier K, Rohrbach K, Brasel K, [36] Sati HI, Greaves M, Apperley JF, Russell RG, Croucher PI.
De Smedt T, et al. RANK is essential for osteoclast and Expression of interleukin-1beta and tumour necrosis
lymph node development. Genes Dev 1999;13:2412–24. factor-alpha in plasma cells from patients with multiple
[18] Tsukii K, Shima N, Mochizuki S, Yamaguchi K, Kinosaki M, myeloma. Br J Haematol 1999;104:350–7.
Yano K, et al. Osteoclast differentiation factor mediates an [37] Oyajobi BO, Franchin G, Williams PJ, Pulkrabek D, Gupta A,
essential signal for bone resorption induced by 1 alpha,25- Munoz S, et al. Dual effects of macrophage inflammatory
dihydroxyvitamin D3, prostaglandin E2, or parathyroid protein-1 alpha on osteolysis and tumor burden in the murine
hormone in the microenvironment of bone. Biochem 5TGM1 model of myeloma bone disease. Blood 2003;102:311–9.
Biophys Res Commun 1998;246:337–41. [38] Mitsiades CS, Mitsiades NS, Munshi NC, Richardson PG,
[19] Kong YY, Yoshida H, Sarosi I, Tan HL, Timms E, Capparelli C, Anderson KC. The role of the bone microenvironment in the
et al. OPGL is a key regulator of osteoclastogenesis, pathophysiology and therapeutic management of multiple
lymphocyte development and lymph-node organogenesis. myeloma: interplay of growth factors, their receptors and
Nature 1999;397:315–23. stromal interactions. Eur J Cancer 2006;42:1564–73.
[20] Lacey DL, Timms E, Tan HL, Kelley MJ, Dunstan CR, Burgess T, [39] Mancino AT, Klimberg VS, Yamamoto M, Manolagas SC, Abe
et al. Osteoprotegerin ligand is a cytokine that regulates E. Breast cancer increases osteoclastogenesis by secreting
osteoclast differentiation and activation. Cell 1998;93:165–76. M-CSF and upregulating RANKL in stromal cells. J Surg Res
[21] Bucay N, Sarosi I, Dunstan CR, Morony S, Tarpley J, 2001;100:18–24.
Capparelli C, et al. Osteoprotegerin-deficient mice develop [40] Moschen AR, Kaser A, Enrich B, Ludwiczek O, Gabriel M,
early onset osteoporosis and arterial calcification. Genes Obrist P, et al. The RANKL/OPG system is activated in
Dev 1998;12:1260–8. inflammatory bowel disease and relates to the state of bone
[22] Mizuno A, Amizuka N, Irie K, Murakami A, Fujise N, Kanno T, loss. Gut 2005;54:479–87.
et al. Severe osteoporosis in mice lacking osteoclastogenesis [41] Romas E, Gillespie MT, Martin TJ. Involvement of receptor
inhibitory factor/osteoprotegerin. Biochem Biophys Res activator of NFkappaB ligand and tumor necrosis factor-alpha
Commun 1998;247:610–5. in bone destruction in rheumatoid arthritis. Bone 2002;30:
[23] Simonet WS, Lacey DL, Dunstan CR, Kelley M, Chang MS, Lüthy 340–6.
R, et al. Osteoprotegerin: a novel secreted protein involved in [42] Pearse RN, Sordillo EM, Yaccoby S, Wong BR, Liau DF, Colman N,
the regulation of bone density. Cell 1997;89:309–19. et al. Multiple myeloma disrupts the TRANCE/osteoprotegerin
[24] Hofbauer LC, Khosla S, Dunstan CR, Lacey DL, Boyle WJ, cytokine axis to trigger bone destruction and promote tumor
Riggs BL. The roles of osteoprotegerin and osteoprotegerin progression. Proc Natl Acad Sci U S A 2001;98:11581–6.
ligand in the paracrine regulation of bone resorption. J Bone [43] Giuliani N, Bataille R, Mancini C, Lazzaretti M, Barillé S.
Miner Res 2000;15:2–12. Myeloma cells induce imbalance in the osteoprotegerin/
[25] Westendorf JJ, Kahler RA, Schroeder TM. Wnt signaling in osteoprotegerin ligand system in the human bone marrow
osteoblasts and bone diseases. Gene 2004;341:19–39. environment. Blood 2001;98:3527–33.
[26] Baron R, Kneissel M. WNT signaling in bone homeostasis [44] Croucher PI, Shipman CM, Lippitt J, Perry M, Asosingh K, Hijzen
and disease: from human mutations to treatments. Nat Med A, et al. Osteoprotegerin inhibits the development of osteolytic
2013;19:179–92. bone disease in multiple myeloma. Blood 2001;98:3534–40.
[27] Negishi-Koga T, Shinohara M, Komatsu N, Bito H, Kodama T, [45] Heider U, Langelotz C, Jakob C, Zavrski I, Fleissner C, Eucker
Friedel RH, et al. Suppression of bone formation J, et al. Expression of receptor activator of nuclear factor
by osteoclastic expression of semaphorin 4D. kappa B ligand on bone marrow plasma cells correlates with
Nat Med 2011;17:1473–80. osteolytic bone disease in patients with multiple myeloma.
[28] Bataille R, Chappard D, Marcelli C, Dessauw P, Baldet P, Sany Clin Cancer Res 2003;9:1436–40.
J, et al. Recruitment of new osteoblasts and osteoclasts is [46] Farrugia AN, Atkins GJ, To LB, Pan B, Horvath N, Kostakis P,
the earliest critical event in the pathogenesis of human et al. Receptor activator of nuclear factor-kappa B ligand
multiple myeloma. J Clin Invest 1991;88:62–6. expression by human myeloma cells mediates osteoclast
[29] Roodman GD. Pathogenesis of myeloma bone disease. Blood formation in vitro and correlates with bone destruction in
Cells Mol Dis 2004;32:290–2. vivo. Cancer Res 2003;63:5438–45.
[30] Diamond T, Golombick T, Manoharan A, Kwan Y, Bryant C. [47] Qiang YW, Chen Y, Stephens O, Brown N, Chen B, Epstein J,
The impact of osteoporosis (as measured by lumbar spine et al. Myeloma-derived Dickkopf-1 disrupts Wnt-regulated
quantitative computed tomography) on disease activity and osteoprotegerin and RANKL production by osteoblasts: a
88 M ET ABOL I SM CL IN I CA L A N D E XP E RI ME N TAL 80 ( 20 1 8 ) 8 0– 90

potential mechanism underlying osteolytic bone lesions in [65] Colucci S, Brunetti G, Oranger A, Mori G, Sardone F, Specchia
multiple myeloma. Blood 2008;112:196–207. G, et al. Myeloma cells suppress osteoblasts through
[48] Standal T, Seidel C, Hjertner Ø, Plesner T, Sanderson RD, sclerostin secretion. Blood Cancer J 2011;1:e27.
Waage A, et al. Osteoprotegerin is bound, [66] Delgado-Calle J, Bellido T, Roodman GD. Role of osteocytes
internalized, and degraded by multiple myeloma cells. in multiple myeloma bone disease. Curr Opin Support
Blood 2002;100:3002–7. Palliat Care 2014;8:407–13.
[49] Jakob C, Goerke A, Terpos E, Sterz J, Heider U, Kühnhardt D, [67] Favus MJ. Bisphosphonates for osteoporosis. N Engl J Med
et al. Serum levels of total-RANKL in multiple myeloma. Clin 2010;363:2027–35.
Lymphoma Myeloma 2009;9:430–5. [68] Coleman RE. Bisphosphonates: clinical experience. Oncologist
[50] Luisi S, Florio P, Reis FM, Petraglia F. Expression and 2004;9:14–27.
secretion of activin A: possible physiological and clinical [69] Lahtinen R, Laakso M, Palva I, Virkkunen P, Elomaa I.
implications. Eur J Endocrinol 2001;145:225–36. Randomised, placebo-controlled multicentre trial of
[51] Hedger MP, Winnall WR, Phillips DJ, de Kretser DM. The clodronate in multiple myeloma. Lancet 1992;40:1049–52.
regulation and functions of activin and follistatin in [70] McCloskey EV, Dunn JA, Kanis JA, MacLennan IC, Drayson
inflammation and immunity. Vitam Horm 2011;85:255–97. MT. Long-term follow-up of a prospective, double-blind,
[52] Eijken M, Swagemakers S, Koedam M, Steenbergen C, Derkx P, placebo-controlled randomized trial of clodronate in multi-
Uitterlinden AG, et al. The activin A-follistatin system: potent ple myeloma. Br J Haematol 2001;113:1035–43.
regulator of human extracellular matrix mineralization. [71] Berenson JR, Lichtenstein A, Porter L, Dimopoulos MA,
FASEB J 2007;21:2949–60. Bordoni R, George S, et al. Long-term pamidronate treatment
[53] Kawabata N, Kamiya N, Suzuki N, Matsumoto M, Takagi M. of advanced multiple myeloma patients reduces skeletal
Changes in extracellular activin A: follistatin ratio during events. Myeloma Aredia Study Group. J Clin Oncol 1998;16:
differentiation of a mesenchymal progenitor cell line, ROB-C26 593–602.
into osteoblasts and adipocytes. Life Sci 2007;81:8–18. [72] Berenson JR, Rosen LS, Howell A, Porter L, Coleman RE, Morley
[54] Fuller K, Bayley KE, Chambers TJ. Activin A is an essential W, et al. Zoledronic acid reduces skeletal-related events in
cofactor for osteoclast induction. Biochem Biophys Res patients with osteolytic metastases. Cancer 2001;91:1191–200.
Commun 2000;268:2–7. [73] Daragon A, Humez C, Michot C, Le Loet X, Grosbois B, Pouyol F,
[55] Terpos E, Kastritis E, Christoulas D, Gkotzamanidou M, et al. Treatment of multiple myeloma with etidronate: results
Eleutherakis-Papaiakovou E, Kanellias N, et al. Circulating of a multicentre double-blind study. Eur J Med 1993;2:449–52.
activin-A is elevated in patients with advanced multiple [74] Menssen HD, Sakalova A, Fontana A, Herrmann Z, Boewer C,
myeloma and correlates with extensive bone involvement Facon T, et al. Effects of long-term intravenous ibandronate
and inferior survival; no alterations post-lenalidomide and therapy on skeletal-related events, survival, and bone
dexamethasone therapy. Ann Oncol 2012;23:2681–6. resorption markers in patients with advanced multiple
[56] Abildgaard N, Glerup H, Rungby J, Bendix-Hansen K, Kassem myeloma. J Clin Oncol 2002;20:2353–9.
M, Brixen K, et al. Biochemical markers of bone metabolism [75] Rosen LS, Gordon D, Kaminski M, Howell A, Belch A, Mackey
reflect osteoclastic and osteoblastic activity in multiple J, et al. Zoledronic acid versus pamidronate in the treatment
myeloma. Eur J Haematol 2000;64:121–9. of skeletal metastases in patients with breast cancer or
[57] Evans CE, Ward C, Rathour L, Galasko CB. Myeloma affects osteolytic lesions of multiple myeloma: a phase III, double-
both the growth and function of human osteoblast-like blind, comparative trial. Cancer J 2001;7:377–87.
cells. Clin Exp Metastasis 1992;10:33–8. [76] Rosen LS, Gordon D, Kaminski M, Howell A, Belch A, Mackey
[58] Terpos E, Berenson J, Raje N, Roodman GD. Management of J, et al. Long-term efficacy and safety of zoledronic acid
bone disease in multiple myeloma. Expert Rev Hematol compared with pamidronate disodium in the treatment of
2014;7:113–25. skeletal complications in patients with advanced multiple
[59] Politou MC, Heath DJ, Rahemtulla A, Szydlo R, Anagnostopoulos myeloma or breast carcinoma: a randomized, double-blind,
A, Dimopoulos MA, et al. Serum concentrations of Dickkopf-1 multicenter, comparative trial. Cancer 2003;98:1735–44.
protein are increased in patients with multiple myeloma and [77] Morgan GJ, Davies FE, Gregory WM, Cocks K, Bell SE, Szubert AJ,
reduced after autologous stem cell transplantation. Int J Cancer et al. First-line treatment with zoledronic acid as compared
2006;119:1728–31. with clodronic acid in multiple myeloma (MRC Myeloma IX): a
[60] Tian E, Zhan F, Walker R, Rasmussen E, Ma Y, Barlogie B, randomised controlled trial. Lancet 2010;376:1989–99.
et al. The role of the Wnt-signaling antagonist DKK1 in the [78] Croucher PI, De Hendrik R, Perry MJ, Hijzen A, Shipman CM,
development of osteolytic lesions in multiple myeloma. Lippitt J, et al. Zoledronic acid treatment of 5T2MM-bearing
N Engl J Med 2003;349:2483–94. mice inhibits the development of myeloma bone disease:
[61] Kaiser M, Mieth M, Liebisch P, Oberländer R, Rademacher J, evidence for decreased osteolysis, tumor burden and
Jakob C, et al. Serum concentrations of DKK-1 correlate with angiogenesis, and increased survival. J Bone Miner Res 2003;
the extent of bone disease in patients with multiple 18:482–92.
myeloma. Eur J Haematol 2008;80:490–4. [79] Kyle RA, Yee GC, Somerfield MR, Flynn PJ, Halabi S, Jagannath
[62] Glass II DA, Bialek P, Ahn JD, Starbuck M, Patel MS, Clevers S, et al. American Society of Clinical Oncology 2007 clinical
H, et al. Canonical Wnt signaling in differentiated practice guideline update on the role of bisphosphonates in
osteoblasts controls osteoclast differentiation. Dev Cell multiple myeloma. J Clin Oncol 2007;25:2464–72.
2005;8:751–64. [80] Bamias A, Kastritis E, Bamia C, Moulopoulos LA,
[63] Spencer GJ, Utting JC, Etheridge SL, Arnett TR, Genever PG. Melakopoulos I, Bozas G, et al. Osteonecrosis of the jaw in
Wnt signalling in osteoblasts regulates expression of the cancer after treatment with bisphosphonates: incidence
receptor activator of NF kappaB ligand and inhibits and risk factors. J Clin Oncol 2005;23:8580–7.
osteoclastogenesis in vitro. J Cell Sci 2006;119:1283–96. [81] Dimopoulos MA, Kastritis E, Anagnostopoulos A,
[64] Terpos E, Christoulas D, Katodritou E, Bratengeier C, Melakopoulos I, Gika D, Moulopoulos LA, et al. Osteonecrosis
Gkotzamanidou M, Michalis E, et al. Elevated circulating of the jaw in patients with multiple myeloma treated with
sclerostin correlates with advanced disease features and bisphosphonates: evidence of increased risk after treatment
abnormal bone remodeling in symptomatic myeloma: with zoledronic acid. Haematologica 2006;91:968–71.
reduction post-bortezomib monotherapy. Int J Cancer 2012; [82] Zervas K, Verrou E, Teleioudis Z, Vahtsevanos K, Banti A,
131:1466–71. Mihou D, et al. Incidence, risk factors and management of
M ET ABOL I SM CL IN I CA L A N D E XP E RI ME N TAL 8 0 ( 20 1 8 ) 8 0– 9 0 89

osteonecrosis of the jaw in patients with multiple myeloma: [98] Vanderkerken K, De Leenheer E, Shipman C, Asosingh K,
a single-centre experience in 303 patients. Br J Haematol Willems A, Van Camp B, et al. Recombinant osteoprotegerin
2006;134:620–3. decreases tumor burden and increases survival in a murine
[83] Badros A, Terpos E, Katodritou E, Goloubeva O, Kastritis E, model of multiple myeloma. Cancer Res 2003;63:287–9.
Verrou E, et al. Natural history of osteonecrosis of the jaw in [99] Elliott P, Kostenuik P, Chen C, Kelley M, Hawkins N, Housman
patients with multiple myeloma. J Clin Oncol 2008;26:5904–9. J, et al. Denosumab is a selective inhibitor of human receptor
[84] Dimopoulos MA, Kastritis E, Bamia C, Melakopoulos I, Gika activator of NF-KB ligand that blocks osteoclast formation in
D, Roussou M, et al. Reduction of osteonecrosis of the jaw vitro and in vivo. Eur J Cancer Suppl 2006;4:62.
(ONJ) after implementation of preventive measures in [100] Kostenuik PJ, Nguyen HQ, McCabe J, Warmington KS,
patients with multiple myeloma treated with zoledronic Kurahara C, Sun N, et al. Denosumab, a fully human
acid. Ann Oncol 2009;20:117–20. monoclonal antibody to RANKL, inhibits bone resorption
[85] Montefusco V, Gay F, Spina F, Miceli R, Maniezzo M, Teresa and increases BMD in knock-in mice that express chimeric
Ambrosini M, et al. Antibiotic prophylaxis before dental (murine/human) RANKL. J Bone Miner Res 2009;24:182–95.
procedures may reduce the incidence of osteonecrosis of [101] Yee AJ, Raje NS. Denosumab, a RANK ligand inhibitor, for
the jaw in patients with multiple myeloma treated with the management of bone loss in cancer patients. Clin Interv
bisphosphonates. Leuk Lymphoma 2008;49:2156–62. Aging 2012;7:331–8.
[86] Migliorati CA, Casiglia J, Epstein J, Jacobsen PL, Siegel MA, [102] Body JJ, Facon T, Coleman RE, Lipton A, Geurs F, Fan M, et al.
Woo SB. Managing the care of patients with A study of the biological receptor activator of nuclear factor-
bisphosphonate-associated osteonecrosis: an American kappaB ligand inhibitor, denosumab, in patients with
Academy of Oral Medicine position paper. J Am Dent Assoc multiple myeloma or bone metastases from breast cancer.
2005;136:1658–68. Clin Cancer Res 2006;12:1221–8.
[87] Terpos E, Morgan G, Dimopoulos MA, Drake MT, Lentzsch S, [103] Yonemori K, Fujiwara Y, Minami H, Kitagawa K, Fujii H, Arai T,
Raje N, et al. International Myeloma Working Group recom- et al. Phase 1 trial of denosumab safety, pharmacokinetics,
mendations for the treatment of multiple myeloma-related and pharmacodynamics in Japanese women with breast
bone disease. J Clin Oncol 2013;31:2347–57. cancer-related bone metastases. Cancer Sci 2008;99:1237–42.
[88] Wang JC, Chien WC, Chung CH, Liao WI, Tsai SH. Adverse [104] Vij R, Horvath N, Spencer A, Taylor K, Vadhan-Raj S, Vescio
cardiovascular effects of nitrogen-containing bisphosphonates R, et al. An open-label, phase 2 trial of denosumab in the
in patients with osteoporosis: A nationwide population-based treatment of relapsed or plateau-phase multiple myeloma.
retrospective study. Int J Cardiol 2016;215:232–7. Am J Hematol 2009;84:650–6.
[89] Herrera L, Leal I, Lapi F, Schuemie M, Arcoraci V, Cipriani F, [105] Fizazi K, Lipton A, Mariette X, Body JJ, Rahim Y, Gralow JR,
et al. Risk of atrial fibrillation among bisphosphonate users: et al. Randomized phase II trial of denosumab in patients
a multicenter, population-based, Italian study. Osteoporos with bone metastases from prostate cancer, breast cancer,
Int 2015;26:1499–506. or other neoplasms after intravenous bisphosphonates.
[90] Chang ST, Tenforde AS, Grimsrud CD, O'Ryan FS, Gonzalez JR, J Clin Oncol 2009;27:1564–71.
Baer DM, et al. Atypical femur fractures among breast cancer [106] Henry DH, Costa L, Goldwasser F, Hirsh V, Hungria V,
and multiple myeloma patients receiving intravenous Prausova J, et al. Randomized, double-blind study of
bisphosphonate therapy. Bone 2012;51:524–7. denosumab versus zoledronic acid in the treatment of bone
[91] Lloyd AA, Gludovatz B, Riedel C, Luengo EA, Saiyed R, Marty metastases in patients with advanced cancer (excluding
E, et al. Atypical fracture with long-term bisphosphonate breast and prostate cancer) or multiple myeloma. J Clin
therapy is associated with altered cortical composition and Oncol 2011;29:1125–32.
reduced fracture resistance. Proc Natl Acad Sci U S A 2017; [107] Raje N, Vadhan-Raj S, Willenbacher W, Terpos E, Hungria V,
114:8722–7. Spencer A, et al. Evaluating results from the multiple myeloma
[92] Morgan GJ, Child JA, Gregory WM, Szubert AJ, Cocks K, Bell SE, patient subset treated with denosumab or zoledronic acid in a
et al. Effects of zoledronic acid versus clodronic acid on randomized phase 3 trial. Blood Cancer J 2016;6:e378.
skeletal morbidity in patients with newly diagnosed multiple [108] Raje N, Terpos E, Willenbacher W, Shimizu K, Sanz RG, Durie
myeloma (MRC Myeloma IX): secondary outcomes from a B, et al. An international, randomized, double blind trial
randomized controlled trial. Lancet Oncol 2011;12:743–52. comparing denosumab with zoledronic acid (ZA) for the
[93] Corso A, Varettoni M, Zappasodi P, Klersy C, Mangiacavalli S, treatment of bone disease in patients (Pts) with newly
Pica G, et al. A different schedule of zoledronic acid can diagnosed multiple myeloma. OP-046. Presented at 16th
reduce the risk of the osteonecrosis of the jaw in patients International Myeloma Workshop, March 1-4; 2017.
with multiple myeloma. Leukemia 2007;21:1545–8. [109] Terpos E, Roodman GD, Willenbacher W, Shimizu K, Sanz RG,
[94] Gimsing P, Carlson K, Turesson I, Fayers P, Waage A, Vangsted Durie B, et al. Comparison of denosumab with zoledronic acid
A, et al. Effect of pamidronate 30 mg versus 90 mg on physical for the treatment of bone disease in patients with newly
function in patients with newly diagnosed multiple myeloma diagnosed multiple myeloma; an international, randomized,
(Nordic Myeloma Study Group): a double-blind, randomised double blind trial. Haematologica 2017;102(s1):318.
controlled trial. Lancet Oncol 2010;11:973–82. [110] Steinman RM, Bonifaz L, Fujii S, Liu K, Bonnyay D, Yamazaki
[95] Terpos E, Dimopoulos MA, Sezer O, Roodman D, Abildgaard N, S, et al. The innate functions of dendritic cells in peripheral
Vescio R, et al. The use of biochemical markers of bone lymphoid tissues. Adv Exp Med Biol 2005;560:83–97.
remodeling in multiple myeloma: a report of the International [111] Chantry AD, Heath D, Mulivor AW, Pearsall S, Baud'huin M,
Myeloma Working Group. Leukemia 2010;24:1700–12. Coulton L, et al. Inhibiting activin-A signaling stimulates
[96] Raje N, Vescio R, Montgomery CW, Badros A, Munshi N, bone formation and prevents cancer-induced bone de-
Orlowski R, et al. Bone marker-directed dosing of zoledronic struction in vivo. J Bone Miner Res 2010;25:2633–46.
acid for the prevention of skeletal complications in patients [112] Pearsall SR, Canalis E, Cornwall-Brady M, Underwood KW,
with multiple myeloma: results of the Z-MARK Study. Clin Haigis B, Ucran J, et al. A soluble activin Type IIA receptor
Cancer Res 2016;22:1378–84. induces bone formation and improves skeletal integrity.
[97] Yaccoby S, Pearse RN, Johnson CL, Barlogie B, Choi Y, Epstein J. Proc Natl Acad Sci U S A 2008;105:7082–7.
Myeloma interacts with the bone marrow microenvironment [113] Fajardo RJ, Manoharan RK, Pearsall RS, Davies MV, Marvell
to induce osteoclastogenesis and is dependent on osteoclast T, Monnell TE, et al. Treatment with a soluble receptor for
activity. Br J Haematol 2002;116:278–90. activin improves bone mass and structure in the axial and
90 M ET ABOL I SM CL IN I CA L A N D E XP E RI ME N TAL 80 ( 20 1 8 ) 8 0– 90

appendicular skeleton of female cynomolgus macaques therapeutic agent for multiple myeloma. Blood 2009;114:
(Macaca fascicularis). Bone 2010;46:64–71. 371–9.
[114] Lotinum S, Pearsall RS, Davies MV, Marvell TH, Monnell TE, [120] Heath DJ, Chantry AD, Buckle CH, Coulton L, Shaughnessy Jr
Ucran J, et al. A soluble activin receptor Type IIA fusion protein JD, Evans HR, et al. Inhibiting dickkopf-1 (Dkk1) removes
(ACE-011) increases bone mass via dual anabolic-antiresorptive suppression of bone formation and prevents the develop-
effect in Cynomolgus monkeys. Bone 2010;46:1082–8. ment of osteolytic bone disease in multiple myeloma. J Bone
[115] Ruckle J, Jacobs M, Kramer W, Pearsall AE, Kumar R, Miner Res 2009;24:425–36.
Underwood KW, et al. Single-dose, randomized, double- [121] Iyer SP, Beck JT, Stewart AK, Shah J, Kelly KR, Isaacs R, et al.
blind, placebo-controlled study of ACE-011 (ActRIIA-IgG1) in A phase IB multicentre dose-determination study of
postmenopausal women. J Bone Miner Res 2009;24:744–52. BHQ880 in combination with anti-myeloma therapy and
[116] Abdulkadyrov KM, Salogub GN, Khuazheva NK, Sherman ML, zoledronic acid in patients with relapsed or refractory
Laadem A, Barger R, et al. Sotatercept in patients with osteolytic multiple myeloma and prior skeletal-related events. Br J
lesions of multiple myeloma. Br J Haematol 2014;165:814–23. Haematol 2014;167:366–75.
[117] Scullen T, Santo L, Vallet S, Fulciniti M, Eda H, Cirstea D, [122] Hoeppner LH, Secreto FJ, Westendorf JJ. Wnt signaling as a
et al. Lenalidomide in combination with an activin therapeutic target for bone diseases. Expert Opin Ther
A-neutralizing antibody: preclinical rationale for a novel Targets 2009;13:485–96.
anti-myeloma strategy. Leukemia 2013;27:1715–21. [123] Lewiecki EM. New targets for intervention in the treatment
[118] Yaccoby S, Ling W, Zhan F, Walker R, Barlogie B, of postmenopausal osteoporosis. Nat Rev Rheumatol 2011;7:
Shaughnessy Jr JD. Antibody-based inhibition of DKK1 631–8.
suppresses tumor-induced bone resorption and multiple [124] McClung MR, Grauer A, Boonen S, Bolognese MA, Brown JP,
myeloma growth in vivo. Blood 2007;109:2106–11. Diez-Perez A, et al. Romosozumab in postmenopausal
[119] Fulciniti M, Tassone P, Hideshima T, Vallet S, Nanjappa P, women with low bone mineral density. N Engl J Med 2014;
Ettenberg SA, et al. Anti-DKK1 mAb (BHQ880) as a potential 370:412–20.

You might also like