You are on page 1of 10

Seminars in Cell and Developmental Biology 112 (2021) 49–58

Contents lists available at ScienceDirect

Seminars in Cell and Developmental Biology


journal homepage: www.elsevier.com/locate/semcdb

Review

Multiple myeloma—A painful disease of the bone marrow


Marta Diaz-delCastillo a, b, c, d, *, Andrew D. Chantry b, c, d, Michelle A. Lawson b, c,
Anne-Marie Heegaard a
a
Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 160, Copenhagen Ø DK-2100, Denmark
b
Sheffield Myeloma Research Team, Department of Oncology and Metabolism, Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK
c
Mellanby Centre for Bone Research, University of Sheffield Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK
d
Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Glossop Road, Sheffield S10 2JF, UK

A R T I C L E I N F O A B S T R A C T

Keywords: Multiple myeloma is a bone marrow neoplasia with an incidence of 6/100,000/year in Europe. While the disease
Multiple myeloma remains incurable, the development of novel treatments such as autologous stem cell transplantation, protea­
Bone marrow some inhibitors and monoclonal antibodies has led to an increasing subset of patients living with long-term
Quality of life
myeloma. However, more than two thirds of patients suffer from bone pain, often described as severe, and
Bone pain
knowledge on the pain mechanisms and its effect on their health-related quality of life (HRQoL) is limited. In this
review, we discuss the mechanisms of myeloma bone disease, the currently available anti-myeloma treatments
and the lessons learnt from clinical studies regarding HRQoL in myeloma patients. Moreover, we discuss the
mechanisms of cancer-induced bone pain and the knowledge that animal models of myeloma-induced bone pain
can provide to identify novel analgesic targets. To date, information regarding bone pain and HRQoL in myeloma
patients is still scarce and an effort should be made to use standardised questionnaires to assess patient-reported
outcomes that allow inter-study comparisons of the available clinical data.

US National Cancer Institute register were analysed, demonstrating that


myeloma is a disease of the elderly, with the mean patient age at diag­
1. Introduction nosis being almost 70 years [14]. These data are in line with other Eu­
ropean multicentre studies showing mean age at diagnosis between 60
Multiple myeloma (MM) is a haematological condition characterised and 80 years [15–18]. While survival expectancy is lower in patients
by the abnormal proliferation of clonal, terminally differentiated B above 65 years of age [14], autologous stem cell transplantation, a
lymphocytes in the bone marrow. The estimated worldwide incidence of common treatment approach among developed nations e.g. European
MM is 2 per 100,000 people [1] and up to 6 per 100,000 in Europe [2,3]. countries, Australia or North America [1], has shown encouraging re­
While the aetiology of the disease remains unknown, a genetic sults even in patients above 75 years of age [19].
component is suspected, as an increased risk among first-degree rela­
tives of myeloma patients has been identified [4–6]. Whether the spike
in prevalence among relatives has an underlying genetic cause or is a 1.1. Monoclonal gammopathy of undetermined significance (MGUS) and
consequence of shared environmental factors, such as exposure to toxic MM: what is the difference?
chemicals, remains unclear. Several reports have suggested that expo­
sure to pesticides is correlated with higher risk of myeloma development Formal diagnosis of MM requires the detection of elevated mono­
[7–11]. Similarly, it has been reported that clean-up workers of the clonal paraprotein (also known as monoclonal M protein) in serum or
Chernobyl disaster [12] and survivors of the Nagasaki atomic bomb [13] urine, the presence of at least 10% myeloma cells in the bone marrow,
present a higher incidence of the disease than the general population, and the display of end-organ damage, often referred to as the CRAB
suggesting a role for ionising radiation in myelomagenesis. criteria: hypercalcemia, renal failure, anaemia, and bone lesions
Age is an important risk factor for disease development. In a 2012 [20,21]. The clinical presentation of the disease comprises a constella­
study on myeloma prevalence, data from over 40,000 patients from the tion of symptoms, with bone pain, fatigue and infections being the main

* Corresponding author at: Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 160,
Copenhagen Ø DK-2100, Denmark.
E-mail address: marta.castillo@sund.ku.dk (M. Diaz-delCastillo).

https://doi.org/10.1016/j.semcdb.2020.10.006
Received 3 February 2020; Received in revised form 13 October 2020; Accepted 15 October 2020
Available online 4 November 2020
1084-9521/© 2020 Elsevier Ltd. All rights reserved.
M. Diaz-delCastillo et al. Seminars in Cell and Developmental Biology 112 (2021) 49–58

Nomenclature MMP Matrix metalloproteinase


NGF Nerve growth factor
ASIC Acid sensing ion channel NMDA N-methyl-D-aspartate
BDNF Brain derived nerve factor PGE2 Prostaglandin E
CCL Chemokine C-C motif Ligand PTHrP Parathyroid hormone-related protein
DKK-1 Dickkopf WNT signalling pathway inhibitor 1; RANKL Receptor activator of nuclear factor kappa-В ligand
HRQoL Health related quality of life SFRP Secreted frizzled-related protein
IL Interleukin TGFβ Transforming growth factor β
IMiDs Immunomodulatory drugs TrkA Tropomyosin receptor kinase A
QoL Quality of life TNF-α Tumour necrosis factor α
M-CSF Macrophage colony-stimulating factor TRPV1 Transient receptor potential cation channel subfamily V
MGUS Monoclonal gammopathy of undetermined significance member 1
MM Multiple myeloma V-ATPase Vacuolar-type H+ATPase

cause of initial physician consultation [22]. of osteolytic lesions, which frequently result in fracture in myeloma
Interestingly, MM is preceded in virtually all patients by monoclonal patients. Myeloma-induced bone disease occurs via several mechanisms,
gammopathy of undetermined significance (MGUS) [23], an asymp­ leading to an imbalance in normal bone remodelling, where bone
tomatic disorder characterised by paraprotein levels > 3 g/dL and less resorption is dramatically increased and bone formation is suppressed
than 10% clonal myeloma cells in the bone marrow without end-organ (Fig. 1). Myeloma cells can induce these changes to bone remodelling by
damage features [20]. In a prospective study, Landgren et al. [24] the secretion of factors that can directly modulate osteoclasts or osteo­
screened a population of almost 78,000 people and identified a total of blasts. In addition, myeloma cells can indirectly cause alterations in
71 patients who developed MM during the course of the study. By bone remodelling by stimulating other cells in the bone marrow
measuring paraprotein levels in serum samples collected annually from microenvironment to release bone modulating factors.
8 to 2 years before MM diagnosis, they found that all myeloma patients There are numerous osteoclast-activating factors secreted by
had previously developed MGUS. The estimated risk of MGUS evolving myeloma cells or released from the bone matrix following resorption,
to MM or related disorders is approximately 1% per year, and remains including receptor activator of nuclear factor kappa-В ligand (RANKL)
stable over time [25,26]. Several reports have indicated that the prev­ [29,30], chemokine (C-C motif) ligands (CCL3 [31–34], CCL4 [32] and
alence of MGUS ranges from 2% to 4% in the general population over 50 CCL20 [34,35]), various interleukins (IL-1β [36,37], IL-3 [38], IL-6
years of age, with men presenting a slightly higher prevalence than [39,40] and IL-17 [36,41]), transforming growth factor beta (TGFβ)
women, and ethnical heterogeneity, with Black and Hispanic men pre­ [42–44] and tumour-necrosis factor alpha (TNFα) [45,46]. More
senting higher prevalence of the disorder [26–28]. recently, this pool has expanded to include microRNA-21 (miR-21) [47],
X-box-binding protein 1 (XBP1) [48,49], B cell activating factor (BAFF)
2. Myeloma-induced bone disease [50,51], syndecan-1 [52], notch signalling mediators [53,54], osteo­
pontin (OPN) [55–57], sequestrome 1 (p62) [58] and C-X-C motif che­
Myeloma-induced bone disease, which is one of the main compli­ mokine 12 (CXCL12) [59,60]. Many of the more recently identified
cations of MM, is characterised by severe bone loss and the development osteoclast-activating factors act as upstream or downstream mediators

Fig. 1. Pathophysiology of myeloma-induced


bone disease. Osteoclast-activating factors (e.g.
RANKL, CCLs, ILs, PTHrP, TGFß, MMPs, miR­
6 NAs, TNFα, M-CSF, PGE2) and osteoblast
inhibitory factors (e.g. DKK-1, SFRPs, ILs, acti­
1 vin A, TGFß) lead to an imbalance in normal
2 bone function in ~90% of myeloma patients
resulting in significant bone loss. These factors
are released by myeloma cells (1 and 2) and
5 other cells in the bone marrow microenviron­
ment (3, 4, 5 and 6) causing the upregulation of
osteoclastic bone resorption, whilst simulta­
neously inhibiting osteoblastic bone formation.
3 4 Myeloma cells also interact with osteoblasts or
bone lining cells leading to tumour cell
dormancy (7), which can result in drug resis­
tance, or later reactivation by osteoclasts.
Decreased Increased (Figure adapted from Green et al., 2019).
osteoblast osteoclast RANKL: Receptor activator of nuclear factor
formation
7 number kappa-В ligand; CCL: chemokine C-C motif
Bone lining ligand; IL: interleukin; PTHrP: parathyroid
cells hormone-related protein; TGFβ: Transforming
growth factor ß MMP: Matrix metalloproteinase;
miRNA: micro RNA; TNFα: Tumour Necrosis
Factor α M-CSF: Macrophage colony-stimulating
factor; PGE2: Prostaglandin E; DKK-1: Dickkopf
WNT signalling pathway inhibitor 1; SFRP:
secreted Frizzled-related protein.

50
M. Diaz-delCastillo et al. Seminars in Cell and Developmental Biology 112 (2021) 49–58

of signalling pathways previously known to stimulate osteoclastogenesis relapse, frequently suppressing disease for many years; further infor­
and osteolysis (e.g. RANKL), and offer new therapeutic targets for the mation on the effect of these therapies on the patient’s quality of life
treatment of myeloma-induced bone disease. (QoL) will be discussed in Section 4 of this review.
Myeloma-induced bone disease is further exacerbated by the simul­ Fourth line and subsequent lines of therapy include daratumumab
taneous release of osteoblast-inhibitory factors from myeloma cells and monotherapy (which is NICE approved in the UK for treatment as a
the bone marrow microenvironment, which prevent osteoblast differ­ monotherapy in daratumumab-naïve patients [85]), the thalidomide
entiation and subsequently impair bone formation and repair. These analogue pomalidamide and dexamethasone [86], as well as re-
factors include dickoff-1 (DKK-1) [61,62], activin A [63–65], sclerostin treatment with velcade and dexamethasone with the addition of the
[66,67], secreted frizzled-related protein 2 (sFRP-2) [68,69], IL-3 HDAC inhibitor panobinostat [87]. Other more hard hitting regimens, if
[70,71], IL-7 [63,72,73] and hepatocyte growth factor (HGF) [74,75]. the patient is robust enough to tolerate them, include a triplet consisting
Moreover, bone resorption also causes release of TGFβ from the bone of the dual alkylator, purine analogue bendamustine, thalidomide and
matrix, which further increases myeloma-induced bone disease by pro­ dexamethasone [88].
moting osteoclastogenesis and preventing osteoblast progenitor differ­ Choice of fourth line and subsequent line therapies depend in part on
entiation into mature osteoblasts. In addition, TGFβ acts on osteoblasts response to previous lines of therapy, as well as how robust the patient
and stromal cells to stimulate the release of pro-tumorigenic factors (e.g. remains in the face of progressive disease and whether there is a decline
IL-6) [76], creating what is termed the ‘vicious cycle’ of bone destruc­ in organ function. Eventually most patients succumb even though
tion and myeloma tumour growth [77]. multiple novel therapies are being introduced, often trialled first at these
Myeloma cells can also interact directly with cells in the bone latter stages of disease. It should also be noted that a cohort of patients
marrow microenvironment leading to tumour cell dormancy. This can have myeloma for 20 or more years, tolerating multiple lines of treat­
result in drug-resistant populations of myeloma cells, which can later be ment, sometimes enjoying prolonged remission phases, and may even­
reactivated into proliferating tumour cells. Myeloma cell dormancy has tually die of other pathologies. At present, the only therapy considered
been demonstrated to be a reversible state that is switched ‘on’ by capable of completely eliminating MM is allogeneic stem cell trans­
engagement with bone-lining cells or osteoblasts, and switched ‘off’ by plantation. However, treatment-related mortality for patients enduring
osteoclastic bone resorption [78]. Therefore, increased osteoclastic bone this therapy is high, currently quoted around 15–20% and is therefore
resorption is likely to lead to increased tumour cells and increased reserved for high risk patients with aggressive disease [89].
myeloma-induced bone disease.
4. Quality of life (QoL) in MM patients
3. Current treatments for MM patients
MM patients have been shown to report more problems and symp­
Over the last twenty years major advances have been made in the toms than patients suffering from other haematological malignancies,
treatment of MM, leading to improved overall survival. Typically, such as leukaemia or lymphoma [90], with more than two thirds of
myeloma patients undergo a cycle of induction chemotherapy followed patients being symptomatic at time of diagnosis [91]. Among the
by autologous stem cell transplantation, which induces a remission symptoms that impair the patient’s health-related QoL (HRQoL), bone
phase of variable duration. In most cases, this phase is followed by pain and fatigue are the commonest, with pain affecting 51–61% of
disease relapse, leading to the use of second line therapy and the patients [22,79,92,93], followed by sleepiness, hypo- or paraesthesia,
consequent treatment-free remission phase. This cycle generally con­ muscle cramps, constipation, oedema and insomnia [92].
tinues, with each remission phase progressively shortening until anti- As discussed in Section 3, induction chemotherapy followed by
myeloma drugs are unable to stop disease progression [79]. autologous stem cell transplantation is the commonest treatment for
As discussed in Section 2, myeloma-induced bone disease is a MM, and several studies have addressed its clinical efficacy and its effect
recurrent complication of MM. To reduce the risk of skeletal-related on HRQoL, showing that even during a stable plateau phase (in the
events, patients are often treated with bisphosphonates (specific anti­ absence of active myeloma disease), bone aches and/or neuropathic
resorptive agents that promote osteoclast apoptosis) in addition to their pain continues to affect 50% of patients, with 10% describing it as
anti-myeloma treatments. “severe” [94]. Interestingly, both loss of weight and exposure to steroids
In the UK and Europe, induction therapy in the form of triplet regi­ prior to autologous stem cell transplantation have been correlated with
mens is becoming the norm for newly diagnosed patients, typically worse physical function following treatment [95].
comprising a proteasome inhibitor (e.g. bortezomib or velcade), an On the other hand, patients who are ineligible for a stem cell trans­
immune-modulatory imide drug (IMiD, e.g. thalidomide) and a corti­ plant are often treated with a combination of chemotherapy and IMiDs.
costeroid (typically dexamethasone). This VTD regimen often involves A recent phase III clinical study evaluated the HRQoL of transplant
cycles that last 21 days, with four to six cycles typically given [80]. For ineligible myeloma patients treated with melphalan-prednisolone
those patients considered ineligible for autologous stem cell trans­ (chemotherapy) in combination with either the IMiDs thalidomide or
plantation, either due to frailty or co-morbidity, induction chemo­ lenalidomide, as well as the effect of continuing treatment with the
therapy triplets are favoured as they are slightly gentler in intensity and IMiDs as maintenance therapy [96]. The results showed an improve­
typically comprise either velcade, the alkylating agent melphalan and ment in global QoL, future perspective and role and emotional func­
prednisolone (VMP) [81], or the alkylator cyclophosphamide, thalido­ tioning, as well as a decrease in pain (regardless of the number of bone
mide and dexamethasone-attenuated regimen (CTDa) [82]. lesions) for both IMiDs, while thalidomide caused peripheral neuropa­
Second line therapy offered on evidence of relapse generally com­ thy in the majority of patients and lenalidomide caused diarrhoea.
prises re-induction with a regimen such as CTD when a second autolo­ In another study, QoL was assessed in patients with relapsed or re­
gous stem cell transplantation is considered. Alternatively, the newly UK fractory myeloma who were treated with eight cycles of ixazomib-
National Institute of Clinical Excellence (NICE) approved regimen dar­ thalidomide-dexamethasone, followed by 12 months maintenance
atumumab, a monoclonal antibody directed at CD38, in combination therapy with ixazomib [97]. A clinically significant improvement in
with velcade and dexamethasone (DVd), offers an excellent long term re- pain was observed following induction therapy, but a worsening of
induction programme [83]. neuropathy was also detected (neuropathy was present in 41% of pa­
Third line therapy in the UK and frequently in Europe, often com­ tients at baseline, with a mean score of 22, which increased by 23 points
prises the IMiD lenalidamide (Revlimid, Celgene) and dexamethasone, at the end of the study; scale 0–100, where 0 indicates less or no
plus or minus the novel oral proteasome inhibitor ixazomib [84]. This is symptom). Moreover, myeloma patients who went on maintenance
an attractive regimen being well tolerated and given indefinitely until therapy showed non-significant improvements in HRQoL and pain,

51
M. Diaz-delCastillo et al. Seminars in Cell and Developmental Biology 112 (2021) 49–58

without worsening of side effects. These studies highlight the impor­ 5.1. Mechanisms of myeloma-induced bone pain
tance of addressing the effect of novel therapies on pain and QoL,
especially as it has been shown that gains in QoL can be more valuable Despite the widespread use of animal models of MM over the last few
than increases in survival for elderly cancer patients [98]. decades, which has significantly contributed towards our understanding
To date, most studies addressing HRQoL are intervention studies in of the pathophysiology of the disease, there is a lack of preclinical pain
which the effect of a novel drug or treatment is evaluated, like the ones research of this condition. Animal models of myeloma mainly include
describe above. A 2017 systematic review on HRQoL in longitudinal syngeneic models, in which myeloma cell lines are inoculated in
studies of myeloma patients concluded that newly diagnosed patients immunocompetent, strain-matched mice, and xenograft models, in
are generally more symptomatic than those in relapse (possibly due to which human myeloma cell lines or patient-derived cells are inoculated
the close monitoring of diagnosed patients by their clinicians), but in immunosuppressed rodents [109].
generally report an improvement in pain following first line treatment, To date, only a few publications have addressed myeloma-induced
while pain levels were mostly unchanged during relapse treatment [99]. bone pain in preclinical models, and most knowledge on cancer-
However, as the study highlights, information on pain and HRQoL in induced bone pain has been generated in models of bone metastases
patients who are not undergoing experimental cancer treatment (older (breast and prostate cancer) and osteosarcoma, revealing that cancer-
and frailer patients who are often not included in studies) is limited [99]. induced bone pain shares characteristics and mechanisms with inflam­
As the development of novel therapies is leading to a growing subset of matory and neuropathic pain, but presents its own neurochemical
myeloma patients for whom the disease becomes long-lasting, under­ signature [110]. From these studies, three main drivers of cancer-
standing and improving the pain and consequently the HRQoL of these induced bone pain have been identified: (1) the activation of periph­
patients is becoming increasingly important, and the use of standardised eral nerve fibres by pro-inflammatory agents and protons released in the
tools to asses patient-reported outcomes, including pain, has been sug­ bone cancer microenvironment, (2) the damage, denervation or
gested as the next step into personalised medicine for cancer patients sprouting of peripheral sensory nerves innervating the bone, and (3) the
[100,101]. deregulation of different proteins and molecular pathways in the dorsal
root ganglia and central nervous system. These findings are summarised
5. Pain in myeloma patients in Table 2.

Pain and fatigue are the main symptoms reported by myeloma pa­ 5.1.1. Peripheral mechanisms: activation of sensory fibres
tients, with the lower back and the arm/shoulder being the commonest As cancer cells proliferate in the bone, they often form a tumour mass
sites of pain reported by patients (Table 1); interestingly, average pain built of cancer, stromal and inflammatory cells, such as macrophages,
intensity seems to be maintained at the different phases of disease
progression and treatment. Myeloma pain can be classified into bone
pain as a consequence of myeloma-induced bone disease (including Table 2
skeletal-related events such as pathological fractures), procedural pain Summary of the proposed peripheral and central mechanisms of bone pain in
(as patients are exposed to a range of invasive and non-invasive pro­ multiple myeloma. NGF = nerve growth factor.
cedures) or neuropathic pain, often caused by treatment-related toxicity Proposed mechanisms of Animal model Reference
[102]. Chemotherapy-induced peripheral neuropathy is characterised myeloma-induced bone
by numbness, tingling and pain in a stocking/glove pattern, and can be pain

caused by neurotoxicity derived from chemotherapeutic drugs, as well Peripheral Activation of afferent Intratibial Hiasa et al.
as proteasome inhibitors such as bortezomib or IMiDs like thalidomide nociceptors due to innoculation of JJN3 [117]
tumour-induced cells in C57BL/6
[103,104]. The grade of neuropathy depends on the drug, dose and
acidification of neuronal SCID mice (xenograft
duration of the treatment, and it is more prevalent in myeloma patients microenvironment. model).
with baseline neuropathy [105]. Chemotherapy-induced peripheral Tumour-induced
neuropathy can often resolve [105] or be treated with calcium channel sprouting of sensory nerve
blockers such as gabapentin or pregabalin [106]; however, poly­ fibres.
Tumour-induced injury of Intrafemoral Diaz-
neuropathy remains the commonest treatment-related adverse effect
sensory nerve fibres. innoculation of delCastillo
[79]. 5TGM1-GFP cells in et al. [129]
Myeloma-induced bone pain is generally treated following the syngeneic C57BL/
analgesic ladder for the management of cancer pain proposed by the KaLwRijHsd mice.
Increased NGF production Intravenous Olechnowicz
World Health Organization [107]. This three-step ladder recommends
by bone marrow stromal inoculation of et al. [128]
the use of non-steroidal anti-inflammatory drugs (NSAIDs) for the cells. 5TGM1-GFP cells in
treatment of mild pain, followed by increasingly strong opioids as the Central Spinal microglia syngeneic C57BL/
pain progresses. The use of bisphosphonates is also recommended for the activation. KaLwRijHsd mice.
treatment of skeletal disease derived pain, even though their analgesic Spinal astrocyte
activation.
effect on bone pain remains to be firmly established [108].

Table 1
Pain prevalence and average pain intensity in different body sites in myeloma patients during disease progression. Average pain intensity reported in the standardised
QLQ-MY20 questionnaire, based on the following scale: 1 = “Not at all”, 2 = “A little”, 3 = “Quite a bit” and 4 = “Very much”. The average pain intensity at diagnosis
reported in this table was calculated by extrapolating the data published by Mols et al. [153] Data on average pain intensity during/on completion of first line
treatment and at advanced but stable disease phases were extracted from Cocks et al. [154] and Boland et al. [94], respectively.
Pain

Bone Back Hip Arm/ shoulder Chest

At diagnosis [138] Presence of pain (% patients) 52.7 61.3 34.6 47.6 16.54
Average pain intensity (Mean ± SD) 1.9 ± 1.0 2.0 ± 1.0 1.5 ± 0.8 1.7 ± 0.8 1.2 ± 0.5
During or on completion of first-line treatment [139] Average pain intensity (Mean ± SD) 2.1 ± 0.9 2.0 ± 0.9 1.6 ± 0.8 1.5 ± 0.7 1.3 ± 0.6
At advanced (> 2 treatment lines), stable phase [94] Average pain intensity (Mean ± SD) 2.4 ± 1.0 2.0 ± 1.0 1.6 ± 1.0 1.2 ± 0.4 2.2 ± 1.0

52
M. Diaz-delCastillo et al. Seminars in Cell and Developmental Biology 112 (2021) 49–58

neutrophils, leucocytes and mast cells [111,112]. Often the tumour cells NGF) have shown promising preliminary results [126]. These data
release neurotrophines i.e. nerve growth factor (NGF) and brain-derived suggest that an important driver in cancer-induced bone pain is the
neurotrophic factor (BDNF), which can then bind to their receptors in disorganised, pathological sprouting of sensory and sympathetic nerve
the macrophages, causing the release of pro-inflammatory cytokines (e. fibres innervating the periosteum. Paradoxically, tumour-induced nerve
g. TNFα, IL6 and IL1ß) and other proalgesic inflammatory mediators (e. damage has also been reported in the mouse model of osteosarcoma as
g. prostaglandin E) that can directly sensitise the sensory primary af­ an important mechanism of bone pain [127]. It seems, thus, that both
ferents innervating the bone [112]. nerve sprouting and nerve damage may be caused by bone cancer and
As discussed in Section 2 of this review, cancer cells also cause al­ involved in cancer-induced bone pain.
terations in bone remodelling process, and in MM an increase in oste­ In 2017, Hiasa et al. [117] proposed that myeloma cells populating
oclastic bone resorption and a suppression of osteoblastic bone the bone marrow express proton pumps, which induce an acidification
formation [113], resulting in osteolytic lesion development [114]. As of the tumour microenvironment, as discussed in Section 5.1.1. Their
both osteoclasts and cancer cells liberate protons and lactate, the bone research suggested that such acidification of the bone marrow could, in
microenvironment can become pathologically acidic, possibly inducing turn, stimulate nerve sprouting and activate the acid-sensing noci­
the activation of acid-sensing nociceptors such as acid-sensing ion ceptors of the sensory neurons, consequently causing nociception.
channels (ASICs) or transient receptor potential cation channel sub­ Immunohistological examination of the femurs of myeloma-bearing
family member 1 (TRPV1) [115], resulting in the depolarisation and mice showed increased density of calcitonin gene-related peptide
sensitisation of sensory fibres and consequent generation of nociception. (CGRP)-positive neurons parallel to disease progression, and inhibition
In 2007, Nagae et al. showed that ASIC1a, ASIC1b and TRVP1 are of both the V-ATPase or ASIC3 was efficient in reducing pain-like be­
transcriptionally up-regulated in the ipsilateral dorsal root ganglia of a haviours in these mice, as well as CGRP sprouting and overexpression of
rat model of painful breast cancer bone metastases, parallel to the excitation neuronal markers in the dorsal root ganglia.
development of pain-like behaviour [116]. Moreover, they showed that Another recent publication [128] has studied pain-related mecha­
daily systemic treatment with the bisphosphonate zolendronic acid was nisms in a systemic, immunocompetent mouse model of myeloma,
analgesic, and reduced the mRNA expression of these nociceptors. suggesting that factors released by myeloma cells in the marrow induce
Similarly, a xenograft mouse model of myeloma-induced bone pain the production of NGF by bone marrow stromal cells populating the
displayed a significant improvement in pain-like behaviours following tumour niche, and that NGF may also mediate myeloma-induced bone
the inhibition of either the vacuolar-type H+ ATPase (V-ATPase) or pain.
ASIC3 [117]. Moreover, these treatments also reduced the over­ Our group has evaluated nociception in a syngeneic mouse model of
expression of neuronal excitation markers suggestive of nociception, myeloma established by direct intrafemoral inoculation of a murine
such as phosphorylated ERK and cAMP response element binding pro­ myeloma cell line (5TGM1) in strain-matched immunocompetent BKAL
tein (pCREB) [117]. In this model, bisphosphonate treatment also mice [129]. These mice presented high levels of systemic serum para­
ameliorated myeloma-induced bone pain, and the analgesic effect was protein and developed osteolytic lesions, in agreement with the clinical
potentiated when combined with the V-ATPase inhibitor. Based on these presentation of the disease. Moreover, the mice presented pain-related
data, the authors propose that the acidification of the tumour micro­ behaviours over time, which could be completely reversed by systemic
environment caused by proton pumps located in myeloma cells may be morphine administration. Biweekly bisphosphonate treatment in these
one of the mechanisms involved in myeloma-induced bone pain mice prevented the development of osteolytic lesions, increased survival
development. and decreased pain-like behaviours, though the treatment did not fully
reverse the nociceptive phenotype. In the clinic, conflicting data has
5.1.2. Peripheral mechanisms: disturbances of sensory fibres been reported regarding the analgesic effect of bisphosphonate treat­
Under physiological conditions, sympathetic and sensory neurons ment on myeloma-induced bone pain; while several studies have shown
innervate all bone compartments: mineralised bone, bone marrow and an analgesic effect, data from double-blinded randomised clinical trials
periosteum. While the density of sympathetic fibres is clearly higher in have shown no clinical significance [108]. Interestingly, immunohisto­
cortical pores [118], the ratio of sensory neurons in mineralised bone, chemical characterisation of the sensory neurons in the myeloma-
bone marrow and periosteum has been described as 0.1:2:100 (density, bearing femurs of these mice revealed complete bone marrow dener­
mm/mm3), respectively [119]. Interestingly, mouse studies of bone vation at end-stage disease, suggesting alternative pathways contrib­
innervation have shown a decrease in sympathetic innervation of the uting to the development of myeloma-induced bone pain, which may
cortical bone with aging, while the density of sensory neurons remained arise from a combination of osteolytic lesion development and periph­
unchanged, suggesting that nociceptors transmitting bone pain are eral nerve injury (Fig. 2). However, the timeline of this myeloma-
intact in the elderly [120]. Most sensory neurons innervating the adult induced peripheral nerve injury and how it might impact nociception
bone express tropomyosin receptor kinase A (TrkA+) [119] and can be is unknown. As mentioned above, tumour-induced nerve damage of the
classified as Aδ or peptide-rich C-fibres: Aδ fibres are fast-conducting, bone marrow and mineralised bone was reported in a mouse model of
myelinated neurons that convey acute, localised noxious stimuli, while osteosarcoma-induced bone pain. In this model, nerve damage was
C-fibres are small and unmyelinated, transmitting slow, poorly localised accompanied by an increase in activating transcription factor 3 (ATF3)
noxious stimuli [121,122]. The sensory fibres innervating the bone are and the neuropeptide galanin in the cell bodies of afferent sensory
often considered as silent nociceptors, which can be activated by me­ neurons innervating the femur [127]. This model also showed increased
chanical, acidic or chemical stimuli to convey the nociceptive signal. It is glial fibrillary acidic protein (GFAP) and macrophage infiltration and
generally believed that the Aδ fibres transmit the fast, acute pain upon activation in the dorsal root ganglia, all of it indicative of neuropathic
activation (e.g. bone fracture), while the C-fibres are responsible for the mechanisms contributing to osteosarcoma-induced bone pain following
dull, constant, secondary bone pain [122]. tumour-induced bone denervation [127]; it remains to be elucidated
In the last two decades, several reports have acknowledged an in­ whether these neuropathic features also play a role in myeloma-induced
crease in the density of TrkA+ nerve fibres (sprouting) innervating the bone pain.
periosteum of cancer-bearing bones in rodent models of painful breast The observation of myeloma-induced bone denervation is in contrast
[123] and prostate [124] cancer metastases to the bone, as well as in the to the results by Hiasa et al. [117] who showed myeloma-induced nerve
NCTC2472 mouse model of osteosarcoma [125]. In all cases, treatment sprouting as a main driver for myeloma-induce bone pain. These con­
with an antibody targeting NGF (ligand of the TrkA receptor) attenuated tradictory data may be explained by the different animal models
nerve sprouting and induced preventive treatment-induced analgesia. (immunosuppressed vs. immunocompetent) or disease time-course, and
Similarly, clinical trials with tanezumab (monoclonal antibody targeting highlight the challenges of using animal models in pain research.

53
M. Diaz-delCastillo et al. Seminars in Cell and Developmental Biology 112 (2021) 49–58

GAP-43 TH CGRP

am
Sha

20µm 20µm 20µm


MM

20µm 20µm 20µm

Fig. 2. Myeloma induces disturbances in bone innervation. Representative pictures of the bone marrow innervation of sham and myeloma-bearing mice displaying
pain-related behaviours. Adapted from Diaz-delCastillo et al. [129]. GAP-43: Growth-associated protein 43; TH: Tyrosine Hydroxylase; CGRP: Calcitonin gene-
related peptide.

Importantly, none of the studies described above report on the effect of excitability of spinal neurons, known as central sensitisation, in which
multiple myeloma on periosteal bone innervation. As discussed, peri­ low-stimulus nociceptive inputs result in increased amplitude and
osteal nerve sprouting has been implicated in nociception in several duration (hyperalgesia). Central sensitisation is, to a great extent,
rodent models of painful bone disorders including models of cancer- mediated by N-methyl-D-aspartate (NMDA) receptors [135], and their
induced bone pain [123–125]. Thus, a further characterisation of the involvement in the maintenance of cancer-induced bone pain has been
effect of myeloma on periosteal bone innervation is warranted. described [136,137]. However, no studies on NMDA-mediated central
sensitisation in myeloma-induced bone pain are available to date. Taken
5.1.3. Central mechanisms: role of microglia and astrocytes together, it seems that diverse mechanisms may be involved in driving
Upon noxious activation of afferent sensory neurons innervating the myeloma-induced bone pain, including bone disturbances, changes in
bone, the nociceptive signal is transmitted towards the dorsal horn of the the peripheral nervous system and central mechanisms that remain
spinal cord, where it is integrated and further projected towards undiscovered.
suprathalamic brain structures. This extra modulation in the central
nervous system probably explains why there is no direct correlation
between the number and volume of bone tumours and the pain levels 5.2. Mechanisms of treatment-derived pain in multiple myeloma
reported by cancer patients [110,130].
Cancer-induced bone pain is often characterised by changes in the As discussed in Section 3, multiple myeloma patients are typically
expression of different factors in spinal cord and brain that lead to treated with triple regimens containing chemotherapy agents.
hyperalgesia (perception of excessive pain following a noxious stimulus) Chemotherapy-induced peripheral neuropathy (CIPN) is a common
and allodynia (perception of pain following a non-noxious stimulus). complication in cancer survivors that significantly impairs their quality
Rodent models of cancer-induced bone pain have shown multiple of life [138], with 68% patients reporting symptoms within the first
neurochemical changes in the dorsal horn of the spinal cord, mainly the month following chemotherapy treatment, and 30% reporting persistent
upregulation of c-Fos [131,132] and dynorphin [127,132], as well as symptoms after 6 months of end of chemotherapy [139]. Clinical CIPN
hypertrophy of astrocytes [132,133], which may secrete cytokines and frequently manifests as decreased touch perception, which is believed to
other proalgesic molecules that in turn sensitise spinal neurons involved be caused by loss of epidermal nerve fibres (affecting both Aβ, Aδ and C-
in the processing of noxious signals. Similarly, an increase in ionised fibres), as well as Meissner’s corpuscles [140].
calcium binding adaptor molecule 1 (Iba1) and glial fibrillary acidic To better understand the underlying pathophysiology of CIPN, many
protein (GFAP), specific markers of microglia/macrophages and astro­ animal models have been developed by systemic administration of the
cytes, respectively, has been detected in dorsal horn of the spinal cords specific chemotherapeutic agents (platinum compounds such as
of myeloma-bearing mice, indicating that myeloma may induce spinal cisplatin or oxiplatin, taxanes like paclitaxel or docetaxel, vinca alka­
glia activation [128]. Moreover, activation of glial cells in suprathalamic loids like vinscritine, proteasome inhibitors, etc.) to mouse or rat [141].
brain centres involved in pain signalling, such as the rostral ventrome­ These models generally display mechanical allodynia and hyperalgesia,
dial medulla, has also been described in cancer-induced bone pain and occasionally thermal alterations, providing a useful insight into the
[134]. nociceptive mechanisms of CIPN and the potential analgesics for its
An important phenomenon in chronic pain is the increased treatment [141].
In the case of multiple myeloma, patients often receive the first-

54
M. Diaz-delCastillo et al. Seminars in Cell and Developmental Biology 112 (2021) 49–58

generation proteasome inhibitor bortezomib, and it has been reported [8] D. Baris, D.T. Silverman, L.M. Brown, G.M. Swanson, R.B. Hayes, A.G. Schwartz,
J.M. Liff, J.B. Schoenberg, L.M. Pottern, R.S. Greenberg, P.A. Stewart,
that the incidence of sensory peripheral neuropathy in this patient
Occupation, pesticide exposure and risk of multiple myeloma, Scand. J. Work
population ranges from 8.4% to 80.5% [142]. Animal models of Environ. Health 30 (3) (2004) 215–222.
bortezomib-induced peripheral neuropathy include mice displaying [9] P. Pahwa, C.P. Karunanayake, J.A. Dosman, J.J. Spinelli, H.H. McDuffie, J.
disturbed thermal sensitivity [143], and rat models displaying me­ R. McLaughlin, Multiple myeloma and exposure to pesticides: a Canadian case-
control study, J. Agromed. 17 (1) (2012) 40–50.
chanical and cold allodynia, but no thermal hyperalgesia [144,145]. The [10] R. Presutti, S.A. Harris, L. Kachuri, J.J. Spinelli, M. Pahwa, A. Blair, S.H. Zahm, K.
differences in behavioural outcomes across models may be explained by P. Cantor, D.D. Weisenburger, P. Pahwa, J.R. McLaughlin, J.A. Dosman, L.
the myriad of bortezomib-induced alterations that have been identified. B. Freeman, Pesticide exposures and the risk of multiple myeloma in men: an
analysis of the North American Pooled Project, Int. J. Cancer 139 (8) (2016)
These include a reduction in amplitude of sensory action potentials 1703–1714.
[143], axonal degeneration [145,146], nociceptor sensitisation as a [11] L. Kachuri, P.A. Demers, A. Blair, J.J. Spinelli, M. Pahwa, J.R. McLaughlin,
result of decreased astrocyte-mediated glutamate clearance [147,148] P. Pahwa, J.A. Dosman, S.A. Harris, Multiple pesticide exposures and the risk of
multiple myeloma in Canadian men, Int. J. Cancer 133 (8) (2013) 1846–1858.
and increased TRPV1 expression in the spinal cord and cell bodies of [12] D. Gluzman, N. Imamura, L. Sklyarenko, V. Nadgornaya, M. Zavelevich,
sensory neurons [149]. Moreover, bortezomib-induced damage of V. Machilo, Malignant diseases of hematopoietic and lymphoid tissues in
mitochondria [150] and mRNA processing in the cell bodies of primary Chernobyl clean-up workers, Hematol. J. Off. J. Eur. Haematol. Assoc. 5 (7)
(2005) 565–571.
sensory neurons have also been identified [151]. The complicated and [13] M. Iwanaga, M. Tagawa, K. Tsukasaki, T. Matsuo, K. Yokota, Y. Miyazaki,
additive mechanisms by which chemotherapeutic agents induce CIPN T. Fukushima, T. Hata, Y. Imaizumi, D. Imanishi, J. Taguchi, S. Momita,
have hindered the development of effective analgesic options for this S. Kamihira, M. Tomonaga, Relationship between monoclonal gammopathy of
undetermined significance and radiation exposure in Nagasaki atomic bomb
patient population; however, it is expected that the next generations of
survivors, Blood 113 (8) (2009) 1639–1650.
proteasome inhibitors will induce significantly less side effects [152]. [14] H. Kaya, B. Peressini, I. Jawed, D. Martincic, A.L. Elaimy, W.T. Lamoreaux, R.
K. Fairbanks, K.A. Weeks, C.M. Lee, Impact of age, race and decade of treatment
on overall survival in a critical population analysis of 40,000 multiple myeloma
6. Conclusion patients, Int. J. Hematol. 95 (1) (2012) 64–70.
[15] S.Y. Kristinsson, O. Landgren, P.W. Dickman, Å.R. Derolf, M. Björkholm, Patterns
Multiple myeloma is a highly heterogeneous disease that causes bone of survival in multiple myeloma: a population-based study of patients diagnosed
in Sweden from 1973 to 2003, J. Clin. Oncol. 25 (15) (2007) 1993–1999.
pain in the majority of patients. While current advances in the field have [16] K. Remes, P. Anttila, R. Silvennoinen, M. Putkonen, H. Ollikainen, V. Terävä,
led to increased survival, further research is needed to understand the M. Sinisalo, K. Kananen, F. Schain, P. Castren-Kortegangas, T.M. Järvinen,
aetiology of the disease and its symptoms, especially in regards to bone M. Pisini, F. Wahl, T. Dixon, A. Leval, Real-world treatment outcomes in multiple
myeloma: multicenter registry results from Finland 2009-2013, PLoS One 13 (12)
pain. Animal models of MM and myeloma-induced bone pain can be a (2018), e0208507.
useful tool to identify the underlying mechanisms of bone pain and [17] H.M. Lin, K.L. Davis, J.A. Kaye, K. Luptakova, S.P. Nagar, M. Mohty, Real-world
potential novel targets for its treatment. treatment patterns, outcomes, and healthcare resource utilization in relapsed or
refractory multiple myeloma: evidence from a medical record review in France,
Adv. Hematol. 2019 (2019), 4625787.
[18] M.S. Raab, M. Cavo, M. Delforge, C. Driessen, L. Fink, A. Flinois, S. Gonzalez-
Conflicts of interest McQuire, R. Safaei, L. Karlin, M.-V. Mateos, P. Schoen, K. Yong, Multiple
myeloma: practice patterns across Europe, Br. J. Haematol. 175 (1) (2016) 66–76.
[19] Q. Bashir, K. Chamoun, D.R. Milton, M. Khan, S. Ahmed, R. Mehta, U.R. Popat,
The authors report no conflicts of interest. P. Kebriaei, Y. Nieto, B. Oran, S.O. Ciurea, C. Hosing, I. Khouri, K. Patel, E.
E. Manasanch, H.C. Lee, R.Z. Orlowski, R.E. Champlin, M.H. Qazilbash, Outcomes
Acknowledgements of autologous hematopoietic cell transplantation in myeloma patients aged ≥ 75
years, Leuk. Lymphoma 60 (2019) 1–8.
[20] S.V. Rajkumar, M.A. Dimopoulos, A. Palumbo, J. Blade, G. Merlini, M.V. Mateos,
This work was supported by the IMK Fonden. The funding source was S. Kumar, J. Hillengass, E. Kastritis, P. Richardson, O. Landgren, B. Paiva,
not involved in planning, writing or submitting this manuscript. A. Dispenzieri, B. Weiss, X. LeLeu, S. Zweegman, S. Lonial, L. Rosinol, E. Zamagni,
S. Jagannath, O. Sezer, S.Y. Kristinsson, J. Caers, S.Z. Usmani, J.J. Lahuerta, H.
E. Johnsen, M. Beksac, M. Cavo, H. Goldschmidt, E. Terpos, R.A. Kyle, K.
References C. Anderson, B.G. Durie, J.F. Miguel, International Myeloma Working Group
updated criteria for the diagnosis of multiple myeloma, Lancet Oncol. 15 (12)
(2014) e538–e548.
[1] A.J. Cowan, C. Allen, A. Barac, H. Basaleem, I. Bensenor, M.P. Curado,
[21] T.I.M.W. Group, Criteria for the classification of monoclonal gammopathies,
K. Foreman, R. Gupta, J. Harvey, H.D. Hosgood, M. Jakovljevic, Y. Khader,
multiple myeloma and related disorders: a report of the International Myeloma
S. Linn, D. Lad, L. Mantovani, V.M. Nong, A. Mokdad, M. Naghavi, M. Postma,
Working Group, Br. J. Haematol. 121 (5) (2003) 749–757.
G. Roshandel, K. Shackelford, M. Sisay, C.T. Nguyen, T.T. Tran, B.T. Xuan, K.
[22] R.A. Kyle, M.A. Gertz, T.E. Witzig, J.A. Lust, M.Q. Lacy, A. Dispenzieri,
N. Ukwaja, S.E. Vollset, E. Weiderpass, E.N. Libby, C. Fitzmaurice, Global burden
R. Fonseca, S.V. Rajkumar, J.R. Offord, D.R. Larson, M.E. Plevak, T.M. Therneau,
of multiple myeloma: a systematic analysis for the global burden of disease study
P.R. Greipp, Review of 1027 patients with newly diagnosed multiple myeloma,
2016, JAMA Oncol. 4 (9) (2018) 1221–1227.
Mayo Clin. Proc. 78 (1) (2003) 21–33.
[2] M. Sant, C. Allemani, C. Tereanu, R. De Angelis, R. Capocaccia, O. Visser,
[23] R.A. Kyle, S.V. Rajkumar, Criteria for diagnosis, staging, risk stratification and
R. Marcos-Gragera, M. Maynadié, A. Simonetti, J.-M. Lutz, F. Berrino, Incidence
response assessment of multiple myeloma, Leukemia 23 (2008) 3–9.
of hematologic malignancies in Europe by morphologic subtype: results of the
[24] O. Landgren, R.A. Kyle, R.M. Pfeiffer, J.A. Katzmann, N.E. Caporaso, R.B. Hayes,
HAEMACARE project, Blood 116 (19) (2010) 3724–3734.
A. Dispenzieri, S. Kumar, R.J. Clark, D. Baris, R. Hoover, S.V. Rajkumar,
[3] A. Smith, D. Howell, R. Patmore, A. Jack, E. Roman, Incidence of haematological
Monoclonal gammopathy of undetermined significance (MGUS) consistently
malignancy by sub-type: a report from the Haematological Malignancy Research
precedes multiple myeloma: a prospective study, Blood 113 (22) (2009)
Network, Br. J. Cancer 105 (11) (2011) 1684–1692.
5412–5417.
[4] C.M. Vachon, R.A. Kyle, T.M. Therneau, B.J. Foreman, D.R. Larson, C.L. Colby, T.
[25] R.A. Kyle, T.M. Therneau, S.V. Rajkumar, J.R. Offord, D.R. Larson, M.F. Plevak, L.
K. Phelps, A. Dispenzieri, S.K. Kumar, J.A. Katzmann, S.V. Rajkumar, Increased
J. Melton 3rd, A long-term study of prognosis in monoclonal gammopathy of
risk of monoclonal gammopathy in first-degree relatives of patients with multiple
undetermined significance, N. Engl. J. Med. 346 (8) (2002) 564–569.
myeloma or monoclonal gammopathy of undetermined significance, Blood 114
[26] A. Dispenzieri, J.A. Katzmann, R.A. Kyle, D.R. Larson, L.J. Melton, C.L. Colby, T.
(4) (2009) 785–790.
M. Therneau, R. Clark, S.K. Kumar, A. Bradwell, R. Fonseca, D.F. Jelinek, S.
[5] O. Landgren, S.Y. Kristinsson, L.R. Goldin, N.E. Caporaso, C. Blimark, U.
V. Rajkumar, Prevalence and risk of progression of light-chain monoclonal
H. Mellqvist, A. Wahlin, M. Bjorkholm, I. Turesson, Risk of plasma cell and
gammopathy of undetermined significance: a retrospective population-based
lymphoproliferative disorders among 14621 first-degree relatives of 4458
cohort study, Lancet 375 (9727) (2010) 1721–1728.
patients with monoclonal gammopathy of undetermined significance in Sweden,
[27] R.A. Kyle, T.M. Therneau, S.V. Rajkumar, D.R. Larson, M.F. Plevak, J.R. Offord,
Blood 114 (4) (2009) 791–795.
A. Dispenzieri, J.A. Katzmann, L.J. Melton, Prevalence of monoclonal
[6] A. Altieri, B. Chen, J.L. Bermejo, F. Castro, K. Hemminki, Familial risks and
gammopathy of undetermined significance, N. Engl. J. Med. 354 (13) (2006)
temporal incidence trends of multiple myeloma, Eur. J. Cancer 42 (11) (2006)
1362–1369.
1661–1670.
[28] O. Landgren, B.I. Graubard, J.A. Katzmann, R.A. Kyle, I. Ahmadizadeh, R. Clark,
[7] O. Landgren, R.A. Kyle, J.A. Hoppin, L.E. Beane Freeman, J.R. Cerhan, J.
S.K. Kumar, A. Dispenzieri, A.J. Greenberg, T.M. Therneau, L.J. Melton Iii,
A. Katzmann, S.V. Rajkumar, M.C. Alavanja, Pesticide exposure and risk of
N. Caporaso, N. Korde, M. Roschewski, R. Costello, G.M. McQuillan, S.
monoclonal gammopathy of undetermined significance in the Agricultural Health
V. Rajkumar, Racial disparities in the prevalence of monoclonal gammopathies: a
Study, Blood 113 (25) (2009) 6386–6391.

55
M. Diaz-delCastillo et al. Seminars in Cell and Developmental Biology 112 (2021) 49–58

population-based study of 12 482 persons from the National Health and [50] P. Neri, S. Kumar, M.T. Fulciniti, S. Vallet, S. Chhetri, S. Mukherjee, Y. Tai,
Nutritional Examination Survey, Leukemia 28 (2014) 1537–1542. D. Chauhan, P. Tassone, S. Venuta, N.C. Munshi, T. Hideshima, K.C. Anderson,
[29] E. Terpos, R. Szydlo, J.F. Apperley, E. Hatjiharissi, M. Politou, J. Meletis, N. Raje, Neutralizing B-cell activating factor antibody improves survival and
N. Viniou, X. Yataganas, J.M. Goldman, A. Rahemtulla, Soluble receptor activator inhibits osteoclastogenesis in a severe combined immunodeficient human
of nuclear factor kappaB ligand-osteoprotegerin ratio predicts survival in multiple multiple myeloma model, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 13 (19)
myeloma: proposal for a novel prognostic index, Blood 102 (3) (2003) (2007) 5903–5909.
1064–1069. [51] F. Hemingway, R. Taylor, H.J. Knowles, N.A. Athanasou, RANKL-independent
[30] O. Sezer, U. Heider, C. Jakob, I. Zavrski, J. Eucker, K. Possinger, C. Sers, V. Krenn, human osteoclast formation with APRIL, BAFF, NGF, IGF I and IGF II, Bone 48 (4)
Immunocytochemistry reveals RANKL expression of myeloma cells, Blood 99 (12) (2011) 938–944.
(2002) 4646–4647. [52] M.V. Dhodapkar, E. Abe, A. Theus, M. Lacy, J.K. Langford, B. Barlogie, R.
[31] S.J. Choi, J.C. Cruz, F. Craig, H. Chung, R.D. Devlin, G.D. Roodman, M. Alsina, D. Sanderson, Syndecan-1 is a multifunctional regulator of myeloma
Macrophage inflammatory protein 1-alpha is a potential osteoclast stimulatory pathobiology: control of tumor cell survival, growth, and bone cell
factor in multiple myeloma, Blood 96 (2) (2000) 671–675. differentiation, Blood 91 (8) (1998) 2679–2688.
[32] M. Abe, K. Hiura, J. Wilde, K. Moriyama, T. Hashimoto, S. Ozaki, S. Wakatsuki, [53] M. Colombo, K. Thummler, L. Mirandola, S. Garavelli, K. Todoerti, L. Apicella,
M. Kosaka, S. Kido, D. Inoue, T. Matsumoto, Role for macrophage inflammatory E. Lazzari, M. Lancellotti, N. Platonova, M. Akbar, M. Chiriva-Internati, R. Soutar,
protein (MIP)-1alpha and MIP-1beta in the development of osteolytic lesions in A. Neri, C.S. Goodyear, R. Chiaramonte, Notch signaling drives multiple myeloma
multiple myeloma, Blood 100 (6) (2002) 2195–2202. induced osteoclastogenesis, Oncotarget 5 (21) (2014) 10393–10406.
[33] E. Terpos, M. Politou, R. Szydlo, J.M. Goldman, J.F. Apperley, A. Rahemtulla, [54] A. Skrtic, P. Korac, D.R. Kristo, R. Ajdukovic Stojisavljevic, D. Ivankovic,
Serum levels of macrophage inflammatory protein-1 alpha (MIP-1alpha) correlate M. Dominis, Immunohistochemical analysis of NOTCH1 and JAGGED1 expression
with the extent of bone disease and survival in patients with multiple myeloma, in multiple myeloma and monoclonal gammopathy of undetermined significance,
Br. J. Haematol. 123 (1) (2003) 106–109. Hum. Pathol. 41 (12) (2010) 1702–1710.
[34] B.D. Palma, D. Guasco, M. Pedrazzoni, M. Bolzoni, F. Accardi, F. Costa, [55] Y. Saeki, T. Mima, T. Ishii, A. Ogata, H. Kobayashi, S. Ohshima, T. Ishida,
G. Sammarelli, L. Craviotto, M. De Filippo, L. Ruffini, P. Omede, R. Ria, F. Aversa, Y. Tabunoki, H. Kitayama, M. Mizuki, Y. Katada, H. Asaoku, M. Kitano,
N. Giuliani, Osteolytic lesions, cytogenetic features and bone marrow levels of N. Nishimoto, K. Yoshizaki, M. Maeda, S. Kon, N. Kinoshita, T. Uede, I. Kawase,
cytokines and chemokines in multiple myeloma patients: Role of chemokine (C-C Enhanced production of osteopontin in multiple myeloma: clinical and
motif) ligand 20, Leukemia 30 (2) (2016) 409–416. pathogenic implications, Br. J. Haematol. 123 (2) (2003) 263–270.
[35] N. Giuliani, G. Lisignoli, S. Colla, M. Lazzaretti, P. Storti, C. Mancini, S. Bonomini, [56] A. Sfiridaki, S. Miyakis, C. Pappa, G. Tsirakis, A. Alegakis, V. Kotsis,
C. Manferdini, K. Codeluppi, A. Facchini, V. Rizzoli, CC-chemokine ligand 20/ E. Stathopoulos, M. Alexandrakis, Circulating osteopontin: a dual marker of bone
macrophage inflammatory protein-3alpha and CC-chemokine receptor 6 are destruction and angiogenesis in patients with multiple myeloma, J. Hematol.
overexpressed in myeloma microenvironment related to osteolytic bone lesions, Oncol. 4 (2011) 22.
Cancer Res. 68 (16) (2008) 6840–6850. [57] T. Standal, H. Hjorth-Hansen, T. Rasmussen, I.M. Dahl, S. Lenhoff, A.T. Brenne,
[36] K. Noonan, L. Marchionni, J. Anderson, D. Pardoll, G.D. Roodman, I. Borrello, C. Seidel, V. Baykov, A. Waage, M. Borset, A. Sundan, O. Hjertner, Osteopontin is
A novel role of IL-17-producing lymphocytes in mediating lytic bone disease in an adhesive factor for myeloma cells and is found in increased levels in plasma
multiple myeloma, Blood 116 (18) (2010) 3554–3563. from patients with multiple myeloma, Haematologica 89 (2) (2004) 174–182.
[37] I. Yamamoto, M. Kawano, T. Sone, K. Iwato, H. Tanaka, H. Ishikawa, N. Kitamura, [58] Y. Hiruma, T. Honjo, D.F. Jelinek, J.J. Windle, J. Shin, G.D. Roodman,
K. Lee, C. Shigeno, J. Konishi, et al., Production of interleukin 1 beta, a potent N. Kurihara, Increased signaling through p62 in the marrow microenvironment
bone resorbing cytokine, by cultured human myeloma cells, Cancer Res. 49 (15) increases myeloma cell growth and osteoclast formation, Blood 113 (20) (2009)
(1989) 4242–4246. 4894–4902.
[38] J.W. Lee, H.Y. Chung, L.A. Ehrlich, D.F. Jelinek, N.S. Callander, G.D. Roodman, S. [59] Y. Alsayed, H. Ngo, J. Runnels, X. Leleu, U.K. Singha, C.M. Pitsillides, J.
J. Choi, IL-3 expression by myeloma cells increases both osteoclast formation and A. Spencer, T. Kimlinger, J.M. Ghobrial, X. Jia, G. Lu, M. Timm, A. Kumar,
growth of myeloma cells, Blood 103 (6) (2004) 2308–2315. D. Cote, I. Veilleux, K.E. Hedin, G.D. Roodman, T.E. Witzig, A.L. Kung,
[39] O. Kudo, A. Sabokbar, A. Pocock, I. Itonaga, Y. Fujikawa, N.A. Athanasou, T. Hideshima, K.C. Anderson, C.P. Lin, I.M. Ghobrial, Mechanisms of regulation of
Interleukin-6 and interleukin-11 support human osteoclast formation by a CXCR4/SDF-1 (CXCL12)-dependent migration and homing in multiple myeloma,
RANKL-independent mechanism, Bone 32 (1) (2003) 1–7. Blood 109 (7) (2007) 2708–2717.
[40] L. Danks, S. Workman, D. Webster, N.J. Horwood, Elevated cytokine production [60] A.C. Zannettino, A.N. Farrugia, A. Kortesidis, J. Manavis, L.B. To, S.K. Martin,
restores bone resorption by human Btk-deficient osteoclasts, J. Bone Miner. Res. P. Diamond, H. Tamamura, T. Lapidot, N. Fujii, S. Gronthos, Elevated serum
Off. J. Am. Soc. Bone Miner. Res. 26 (1) (2011) 182–192. levels of stromal-derived factor-1alpha are associated with increased osteoclast
[41] R.H. Prabhala, M. Fulciniti, D. Pelluru, N. Rashid, A. Nigroiu, P. Nanjappa, C. Pai, activity and osteolytic bone disease in multiple myeloma patients, Cancer Res. 65
S. Lee, N.S. Prabhala, R.L. Bandi, R. Smith, S.B. Lazo-Kallanian, S. Valet, N. Raje, (5) (2005) 1700–1709.
J.S. Gold, P.G. Richardson, J.F. Daley, K.C. Anderson, S.A. Ettenberg, F. Di [61] E. Tian, F. Zhan, R. Walker, E. Rasmussen, Y. Ma, B. Barlogie, J.
Padova, N.C. Munshi, Targeting IL-17A in multiple myeloma: a potential novel D. Shaughnessy Jr., The role of the Wnt-signaling antagonist DKK1 in the
therapeutic approach in myeloma, Leukemia 30 (2) (2016) 379–389. development of osteolytic lesions in multiple myeloma, N. Engl. J. Med. 349 (26)
[42] K.S. Mohammad, C.G. Chen, G. Balooch, E. Stebbins, C.R. McKenna, H. Davis, (2003) 2483–2494.
M. Niewolna, X.H. Peng, D.H. Nguyen, S.S. Ionova-Martin, J.W. Bracey, W. [62] X.T. Wang, Y.C. He, S.Y. Zhou, J.Z. Jiang, Y.M. Huang, Y.Z. Liang, Y.R. Lai, Bone
R. Hogue, D.H. Wong, R.O. Ritchie, L.J. Suva, R. Derynck, T.A. Guise, T. Alliston, marrow plasma macrophage inflammatory protein protein-1 alpha(MIP-1 alpha)
Pharmacologic inhibition of the TGF-beta type I receptor kinase has anabolic and and sclerostin in multiple myeloma: relationship with bone disease and clinical
anti-catabolic effects on bone, PLoS One 4 (4) (2009), e5275. characteristics, Leuk. Res. 38 (5) (2014) 525–531.
[43] J.S. Nyman, A.R. Merkel, S. Uppuganti, B. Nayak, B. Rowland, A.J. Makowski, B. [63] A. Oranger, C. Carbone, M. Izzo, M. Grano, Cellular mechanisms of multiple
O. Oyajobi, J.A. Sterling, Combined treatment with a transforming growth factor myeloma bone disease, Clin. Dev. Immunol. 2013 (2013), 289458.
beta inhibitor (1D11) and bortezomib improves bone architecture in a mouse [64] S. Vallet, S. Mukherjee, N. Vaghela, T. Hideshima, M. Fulciniti, S. Pozzi, L. Santo,
model of myeloma-induced bone disease, Bone 91 (2016) 81–91. D. Cirstea, K. Patel, A.R. Sohani, A. Guimaraes, W. Xie, D. Chauhan, J.
[44] A. Lu, M.A. Pallero, W. Lei, H. Hong, Y. Yang, M.J. Suto, J.E. Murphy-Ullrich, A. Schoonmaker, E. Attar, M. Churchill, E. Weller, N. Munshi, J.S. Seehra,
Inhibition of transforming growth factor-beta activation diminishes tumor R. Weissleder, K.C. Anderson, D.T. Scadden, N. Raje, Activin A promotes multiple
progression and osteolytic bone disease in mouse models of multiple myeloma, myeloma-induced osteolysis and is a promising target for myeloma bone disease,
Am. J. Pathol. 186 (3) (2016) 678–690. Proc. Natl. Acad. Sci. USA 107 (11) (2010) 5124–5129.
[45] J. Lam, S. Takeshita, J.E. Barker, O. Kanagawa, F.P. Ross, S.L. Teitelbaum, TNF- [65] E. Terpos, E. Kastritis, D. Christoulas, M. Gkotzamanidou, E. Eleutherakis-
alpha induces osteoclastogenesis by direct stimulation of macrophages exposed to Papaiakovou, N. Kanellias, A. Papatheodorou, M.A. Dimopoulos, Circulating
permissive levels of RANK ligand, J. Clin. Investig. 106 (12) (2000) 1481–1488. activin-A is elevated in patients with advanced multiple myeloma and correlates
[46] X. Filella, J. Blade, A.L. Guillermo, R. Molina, C. Rozman, A.M. Ballesta, with extensive bone involvement and inferior survival; no alterations post-
Cytokines (IL-6, TNF-alpha, IL-1alpha) and soluble interleukin-2 receptor as lenalidomide and dexamethasone therapy, Ann. Oncol. Off. J. Eur. Soc. Med.
serum tumor markers in multiple myeloma, Cancer Detect. Prev. 20 (1) (1996) Oncol. 23 (10) (2012) 2681–2686.
52–56. [66] H. Eda, L. Santo, M.N. Wein, D.Z. Hu, D.D. Cirstea, N. Nemani, Y.-T. Tai, S.
[47] M.R. Pitari, M. Rossi, N. Amodio, C. Botta, E. Morelli, C. Federico, A. Gulla, E. Raines, S.A. Kuhstoss, N.C. Munshi, H.M. Kronenberg, N.S. Raje, Regulation of
D. Caracciolo, M.T. Di Martino, M. Arbitrio, A. Giordano, P. Tagliaferri, sclerostin expression in multiple myeloma by Dkk-1: a potential therapeutic
P. Tassone, Inhibition of miR-21 restores RANKL/OPG ratio in multiple myeloma- strategy for myeloma bone disease, J. Bone Miner. Res. Off. J. Am. Soc. Bone
derived bone marrow stromal cells and impairs the resorbing activity of mature Miner. Res. 31 (6) (2016) 1225–1234.
osteoclasts, Oncotarget 6 (29) (2015) 27343–27358. [67] E. Terpos, D. Christoulas, E. Katodritou, C. Bratengeier, M. Gkotzamanidou,
[48] N. Mimura, M. Fulciniti, G. Gorgun, Y.T. Tai, D. Cirstea, L. Santo, Y. Hu, C. Fabre, E. Michalis, S. Delimpasi, A. Pouli, J. Meletis, E. Kastritis, K. Zervas, M.
J. Minami, H. Ohguchi, T. Kiziltepe, H. Ikeda, Y. Kawano, M. French, A. Dimopoulos, Elevated circulating sclerostin correlates with advanced disease
M. Blumenthal, V. Tam, N.L. Kertesz, U.M. Malyankar, M. Hokenson, T. Pham, features and abnormal bone remodeling in symptomatic myeloma: reduction
Q. Zeng, J.B. Patterson, P.G. Richardson, N.C. Munshi, K.C. Anderson, Blockade of post-bortezomib monotherapy, Int. J. Cancer 131 (6) (2012) 1466–1471.
XBP1 splicing by inhibition of IRE1alpha is a promising therapeutic option in [68] R.E. Walker, M.A. Lawson, C.H. Buckle, J.A. Snowden, A.D. Chantry, Myeloma
multiple myeloma, Blood 119 (24) (2012) 5772–5781. bone disease: pathogenesis, current treatments and future targets, Br. Med. Bull.
[49] G. Xu, K. Liu, J. Anderson, K. Patrene, S. Lentzsch, G.D. Roodman, H. Ouyang, 111 (1) (2014) 117–138.
Expression of XBP1s in bone marrow stromal cells is critical for myeloma cell [69] T. Oshima, M. Abe, J. Asano, T. Hara, K. Kitazoe, E. Sekimoto, Y. Tanaka,
growth and osteoclast formation, Blood 119 (18) (2012) 4205–4214. H. Shibata, T. Hashimoto, S. Ozaki, S. Kido, D. Inoue, T. Matsumoto, Myeloma

56
M. Diaz-delCastillo et al. Seminars in Cell and Developmental Biology 112 (2021) 49–58

cells suppress bone formation by secreting a soluble Wnt inhibitor, sFRP-2, Blood [90] A.T. Johnsen, D. Tholstrup, M.A. Petersen, L. Pedersen, M. Groenvold, Health
106 (9) (2005) 3160–3165. related quality of life in a nationally representative sample of haematological
[70] L.A. Ehrlich, G.D. Roodman, The role of immune cells and inflammatory patients, Eur. J. Haematol. 83 (2) (2009) 139–148.
cytokines in Paget’s disease and multiple myeloma, Immunol. Rev. 208 (2005) [91] D.A. Howell, A.G. Smith, A. Jack, R. Patmore, U. Macleod, E. Mironska,
252–266. E. Roman, Time-to-diagnosis and symptoms of myeloma, lymphomas and
[71] R. Silbermann, M. Bolzoni, P. Storti, D. Guasco, S. Bonomini, D. Zhou, J. Wu, J. leukaemias: a report from the Haematological Malignancy Research Network,
L. Anderson, J.J. Windle, F. Aversa, G.D. Roodman, N. Giuliani, Bone marrow BMC Hematol. 13 (1) (2013), 9-9.
monocyte-/macrophage-derived activin A mediates the osteoclastogenic effect of [92] K. Jordan, I. Proskorovsky, P. Lewis, J. Ishak, K. Payne, N. Lordan, C. Kyriakou, C.
IL-3 in multiple myeloma, Leukemia 28 (4) (2014) 951–954. D. Williams, S. Peters, F.E. Davies, Effect of general symptom level, specific
[72] N. Giuliani, S. Colla, F. Morandi, M. Lazzaretti, R. Sala, S. Bonomini, M. Grano, adverse events, treatment patterns, and patient characteristics on health-related
S. Colucci, M. Svaldi, V. Rizzoli, Myeloma cells block RUNX2/CBFA1 activity in quality of life in patients with multiple myeloma: results of a European,
human bone marrow osteoblast progenitors and inhibit osteoblast formation and multicenter cohort study, Support Care Cancer 22 (2) (2014) 417–426.
differentiation, Blood 106 (7) (2005) 2472–2483. [93] C. Ramsenthaler, P. Kane, W. Gao, R.J. Siegert, P.M. Edmonds, S.A. Schey, I.
[73] S. D’Souza, D. del Prete, S. Jin, Q. Sun, A.J. Huston, F.E. Kostov, B. Sammut, C. J. Higginson, Prevalence of symptoms in patients with multiple myeloma: a
S. Hong, J.L. Anderson, K.D. Patrene, S. Yu, C.S. Velu, G. Xiao, H.L. Grimes, G. systematic review and meta-analysis, Eur. J. Haematol. 97 (5) (2016) 416–429.
D. Roodman, D.L. Galson, Gfi1 expressed in bone marrow stromal cells is a novel [94] E. Boland, C. Eiser, Y. Ezaydi, D.M. Greenfield, S.H. Ahmedzai, J.A. Snowden,
osteoblast suppressor in patients with multiple myeloma bone disease, Blood 118 Living with advanced but stable multiple myeloma: a study of the symptom
(26) (2011) 6871–6880. burden and cumulative effects of disease and intensive (hematopoietic stem cell
[74] M. Grano, F. Galimi, G. Zambonin, S. Colucci, E. Cottone, A.Z. Zallone, P. transplant-based) treatment on health-related quality of life, J. Pain Symptom
M. Comoglio, Hepatocyte growth factor is a coupling factor for osteoclasts and Manag. 46 (5) (2013) 671–680.
osteoblasts in vitro, Proc. Natl. Acad. Sci. USA 93 (1996) 7644–7648. [95] L.J. Anderson, C. Yin, R. Burciaga, J. Lee, S. Crabtree, D. Migula, K. Geiss-Wessel,
[75] T. Standal, N. Abildgaard, U.M. Fagerli, B. Stordal, O. Hjertner, M. Borset, H.M. Liu, S.A. Graf, T.R. Chauncey, J.M. Garcia, Assessing cachexia acutely after
A. Sundan, HGF inhibits BMP-induced osteoblastogenesis: possible implications autologous stem cell transplant, Cancers 11 (9) (2019) 1300.
for the bone disease of multiple myeloma, Blood 109 (7) (2007) 3024–3030. [96] L.K. Nielsen, C. Stege, B. Lissenberg-Witte, B. van der Holt, U.-H. Mellqvist,
[76] J.T. Buijs, K.R. Stayrook, T.A. Guise, The role of TGF-β in bone metastasis: novel M. Salomo, G. Bos, M.-D. Levin, H. Visser-Wisselaar, M. Hansson, A. van der
therapeutic perspectives, Bone Rep. 1 (2012), 96-96. Velden, W. Deenik, J. Coenen, M. Hinge, S. Klein, B. Tanis, D. Szatkowski,
[77] G.R. Mundy, T.A. Guise, Hypercalcemia of malignancy, Am. J. Med. 103 (2) R. Brouwer, M. Westerman, R. Leys, H. Sinnige, E. Haukås, K. van der Hem,
(1997) 134–145. M. Durian, P. Gimsing, N. van de Donk, P. Sonneveld, A. Waage, N. Abildgaard,
[78] M.A. Lawson, M.M. McDonald, N. Kovacic, W. Hua Khoo, R.L. Terry, J. Down, S. Zweegman, Health-related quality of life in transplant ineligible newly
W. Kaplan, J. Paton-Hough, C. Fellows, J.A. Pettitt, T. Neil Dear, E. Van diagnosed multiple myeloma patients treated with either thalidomide or
Valckenborgh, P.A. Baldock, M.J. Rogers, C.L. Eaton, K. Vanderkerken, A. lenalidomide-based regimen until progression: a prospective, open-label,
R. Pettit, J.M. Quinn, A.C. Zannettino, T.G. Phan, P.I. Croucher, Osteoclasts multicenter, randomized, phase 3 study, Haematologica (2019), 222299
control reactivation of dormant myeloma cells by remodelling the endosteal haematol. 2019.
niche, Nat. Commun. 6 (2015) 8983. [97] H. Ludwig, W. Ponisch, S. Knop, A. Egle, A. Hinke, M. Schreder, D. Lechner,
[79] K. Yong, M. Delforge, C. Driessen, L. Fink, A. Flinois, S. Gonzalez-McQuire, R. Hajek, E. Gunsilius, A. Petzer, K. Weisel, D. Niederwieser, H. Einsele,
R. Safaei, L. Karlin, M.-V. Mateos, M.S. Raab, P. Schoen, M. Cavo, Multiple W. Willenbacher, H. Rumpold, L. Pour, T. Jelinek, K.J. Krenosz, A. Meckl,
myeloma: patient outcomes in real-world practice, Br. J. Haematol. 175 (2) S. Nolte, T. Melchardt, R. Greil, N. Zojer, Quality of life in patients with relapsed/
(2016) 252–264. refractory multiple myeloma during ixazomib-thalidomide-dexamethasone
[80] P. Moreau, C. Hulin, M. Macro, D. Caillot, C. Chaleteix, M. Roussel, L. Garderet, induction and ixazomib maintenance therapy and comparison to the general
B. Royer, S. Brechignac, M. Tiab, M. Puyade, M. Escoffre, A.M. Stoppa, T. Facon, population, Leuk. Lymphoma (2019) 1–10.
B. Pegourie, D. Chaoui, A. Jaccard, B. Slama, G. Marit, K. Laribi, P. Godmer, [98] U. Wedding, L. Pientka, K. Hoffken, Quality-of-life in elderly patients with cancer:
O. Luycx, J.C. Eisenmann, O. Allangba, M. Dib, C. Araujo, J. Fontan, K. Belhadj, a short review, Eur. J. Cancer 43 (15) (2007) 2203–2210.
M. Wetterwald, V. Dorvaux, J.P. Fermand, P. Rodon, B. Kolb, S. Glaisner, J. [99] L.K. Nielsen, M. Jarden, C.L. Andersen, H. Frederiksen, N. Abildgaard,
V. Malfuson, P. Lenain, L. Biron, L. Planche, H. Caillon, H. Avet-Loiseau, A systematic review of health-related quality of life in longitudinal studies of
T. Dejoie, M. Attal, VTD is superior to VCD prior to intensive therapy in multiple myeloma patients, Eur. J. Haematol. 99 (1) (2017) 3–17.
myeloma: results of the prospective IFM2013-04 trial, Blood 127 (21) (2016) [100] F. Efficace, G. Gaidano, F. Lo-Coco, Patient-reported outcomes in hematology: is it
2569–2574. time to focus more on them in clinical trials and hematology practice? Blood 130
[81] M.V. Mateos, A. Oriol, J. Martinez-Lopez, A.I. Teruel, E. Bengoechea, L. Palomera, (7) (2017) 859–866.
F. de Arriba, D.L. Esseltine, A. Cakana, L. Pei, H. van de Velde, J.S. Miguel, [101] P.G. Kluetz, A. Slagle, E.J. Papadopoulos, L.L. Johnson, M. Donoghue, V.
Outcomes with two different schedules of bortezomib, melphalan, and prednisone E. Kwitkowski, W.H. Chen, R. Sridhara, A.T. Farrell, P. Keegan, G. Kim, R. Pazdur,
(VMP) for previously untreated multiple myeloma: matched pair analysis using Focusing on core patient-reported outcomes in cancer clinical trials: symptomatic
long-term follow-up data from the phase 3 VISTA and PETHEMA/GEM05 trials, adverse events, physical function, and disease-related symptoms, Clin. Cancer
Ann. Hematol. 95 (12) (2016) 2033–2041. Res. Off. J. Am. Assoc. Cancer Res. 22 (7) (2016) 1553–1558.
[82] M.V. Mateos, J. San-Miguel, Treatment of newly diagnosed myeloma in patients [102] S. Rome, K. Noonan, P. Bertolotti, J.D. Tariman, T. Miceli, Bone health, pain, and
not eligible for transplantation, Curr. Hematol. Malig. Rep. 6 (2) (2011) 113–119. mobility: evidence-based recommendations for patients with multiple myeloma,
[83] T.T. Zhang, S. Wang, N. Wan, L. Zhang, Z. Zhang, J. Jiang, Cost-effectiveness of Clin. J. Oncol. Nurs. 21 (5 Suppl) (2017) 47–59.
daratumumab-based triplet therapies in patients with relapsed or refractory [103] B. Islam, M. Lustberg, N.P. Staff, N. Kolb, P. Alberti, A.A. Argyriou, Vinca
multiple myeloma, Clin. Ther. 40 (7) (2018) 1122–1139. alkaloids, thalidomide and eribulin-induced peripheral neurotoxicity: from
[84] M.V. Mateos, T. Masszi, N. Grzasko, M. Hansson, I. Sandhu, L. Pour, L. Viterbo, S. pathogenesis to treatment, J. Peripher. Nerv. Syst. JPNS 24 Suppl 2 (2019)
R. Jackson, A.M. Stoppa, P. Gimsing, M. Hamadani, G. Borsaru, D. Berg, J. Lin, S63–s73.
A. Di Bacco, H. van de Velde, P.G. Richardson, P. Moreau, Impact of prior therapy [104] R. Velasco, P. Alberti, J. Bruna, D. Psimaras, A.A. Argyriou, Bortezomib and other
on the efficacy and safety of oral ixazomib-lenalidomide-dexamethasone vs. proteosome inhibitors-induced peripheral neurotoxicity: from pathogenesis to
placebo-lenalidomide-dexamethasone in patients with relapsed/refractory treatment, J. Peripher. Nerv. Syst. JPNS 24 Suppl 2 (2019) S52–s62.
multiple myeloma in TOURMALINE-MM1, Haematologica 102 (10) (2017) [105] M.A. Dimopoulos, M.-V. Mateos, P.G. Richardson, R. Schlag, N.K. Khuageva,
1767–1775. O. Shpilberg, M. Kropff, I. Spicka, A. Palumbo, K.L. Wu, D.-L. Esseltine, K. Liu,
[85] Y. Khagi, T.M. Mark, Potential role of daratumumab in the treatment of multiple W. Deraedt, A. Cakana, H. Van De Velde, J.F. San Miguel, Risk factors for, and
myeloma, Onco Targets Ther. 7 (2014) 1095–1100. reversibility of, peripheral neuropathy associated with
[86] M.A. Dimopoulos, X. Leleu, A. Palumbo, P. Moreau, M. Delforge, M. Cavo, bortezomib–melphalan–prednisone in newly diagnosed patients with multiple
H. Ludwig, G.J. Morgan, F.E. Davies, P. Sonneveld, S.A. Schey, S. Zweegman, myeloma: subanalysis of the phase 3 VISTA study, Eur. J. Haematol. 86 (1) (2011)
M. Hansson, K. Weisel, M.V. Mateos, T. Facon, J.F. Miguel, Expert panel 23–31.
consensus statement on the optimal use of pomalidomide in relapsed and [106] J.L.M. Jongen, A. Broijl, P. Sonneveld, Chemotherapy-induced peripheral
refractory multiple myeloma, Leukemia 28 (8) (2014) 1573–1585. neuropathies in hematological malignancies, J. Neuro-Oncol. 121 (2) (2015)
[87] J.F. San-Miguel, H. Einsele, P. Moreau, The role of panobinostat plus bortezomib 229–237.
and dexamethasone in treating relapsed or relapsed and refractory multiple [107] WHO, Guidelines for the Pharmacological and Radiotherapeutic Management of
myeloma: a European perspective, Adv. Ther. 33 (11) (2016) 1896–1920. Cancer Pain in Adults and Adolescents, WHO Guidelines Approved by the
[88] B.D. Cheson, W. Brugger, G. Damaj, M. Dreyling, B. Kahl, E. Kimby, M. Ogura, Guidelines Review Committee, (c) World Health Organization 2018., Geneva,
E. Weidmann, C.M. Wendtner, P.L. Zinzani, Optimal use of bendamustine in 2018.
hematologic disorders: treatment recommendations from an international [108] R. Mhaskar, A. Kumar, B. Miladinovic, B. Djulbegovic, Bisphosphonates in
consensus panel - an update, Leuk. Lymphoma 57 (4) (2016) 766–782. multiple myeloma: an updated network meta-analysis, Cochrane Database Syst.
[89] T. Ikeda, K. Mori, K. Kawamura, T. Mori, S. Hagiwara, Y. Ueda, K. Kahata, Rev. 12 (2017), Cd003188.
N. Uchida, N. Tsukada, S. Murakami, M. Yamamoto, T. Takahashi, T. Ichinohe, [109] J. Paton-Hough, A.D. Chantry, M.A. Lawson, A review of current murine models
M. Onizuka, Y. Atsuta, Y. Kanda, S. Okamoto, K. Sunami, H. Takamatsu, of multiple myeloma used to assess the efficacy of therapeutic agents on tumour
Comparison between autologous and allogeneic stem cell transplantation as growth and bone disease, Bone 77 (2015) 57–68.
salvage therapy for multiple myeloma relapsing/progressing after autologous [110] S. Falk, A.H. Dickenson, Pain and nociception: mechanisms of cancer-induced
stem cell transplantation, Hematol. Oncol. 37 (5) (2019) 586–594. bone pain, J. Clin. Oncol. 32 (16) (2014) 1647–1654.

57
M. Diaz-delCastillo et al. Seminars in Cell and Developmental Biology 112 (2021) 49–58

[111] J.M. Jimenez-Andrade, W.G. Mantyh, A.P. Bloom, A.S. Ferng, C.P. Geffre, P. mechanical allodynia in a rat model of cancer-induced bone pain, J. Huazhong
W. Mantyh, Bone cancer pain, Ann. N. Y. Acad. Sci. 1198 (2010) 173–181. Univ. Sci. Technol. Med. Sci. 32 (2) (2012) 291–298.
[112] S.H. Park, M.R. Eber, D.B. Widner, Y. Shiozawa, Role of the bone [135] R.M. Gordon-Williams, A.H. Dickenson, Central neuronal mechanisms in cancer-
microenvironment in the development of painful complications of skeletal induced bone pain, Curr. Opin. Support. Palliat. Care 1 (1) (2007) 6–10.
metastases, in: Cancers (Basel), 10, 2018. [136] C.E. Urch, T. Donovan-Rodriguez, A.H. Dickenson, Alterations in dorsal horn
[113] K.G. Halvorson, M.A. Sevcik, J.R. Ghilardi, T.J. Rosol, P.W. Mantyh, Similarities neurones in a rat model of cancer-induced bone pain, Pain 106 (3) (2003)
and differences in tumor growth, skeletal remodeling and pain in an osteolytic 347–356.
and osteoblastic model of bone cancer, Clin. J. Pain 22 (7) (2006) 587–600. [137] G. Xiaoping, Z. Xiaofang, Z. Yaguo, Z. Juan, W. Junhua, M. Zhengliang,
[114] T.L. Andersen, K. Søe, T.E. Sondergaard, T. Plesner, J.M. Delaisse, Myeloma cell- Involvement of the spinal NMDA receptor/PKCgamma signaling pathway in the
induced disruption of bone remodelling compartments leads to osteolytic lesions development of bone cancer pain, Brain Res. 1335 (2010) 83–90.
and generation of osteoclast-myeloma hybrid cells, Br. J. Haematol. 148 (4) [138] F. Mols, T. Beijers, G. Vreugdenhil, L. van de Poll-Franse, Chemotherapy-induced
(2010) 551–561. peripheral neuropathy and its association with quality of life: a systematic review,
[115] T. Yoneda, M. Hiasa, Y. Nagata, T. Okui, F.A. White, Acidic microenvironment Support Care Cancer 22 (8) (2014) 2261–2269.
and bone pain in cancer-colonized bone, Bone Rep. 4 (2015). [139] M. Seretny, G.L. Currie, E.S. Sena, S. Ramnarine, R. Grant, M.R. MacLeod, L.
[116] M. Nagae, T. Hiraga, T. Yoneda, Acidic microenvironment created by osteoclasts A. Colvin, M. Fallon, Incidence, prevalence, and predictors of chemotherapy-
causes bone pain associated with tumor colonization, J. Bone Miner. Metab. 25 induced peripheral neuropathy: a systematic review and meta-analysis, Pain 155
(2) (2007) 99–104. (12) (2014) 2461–2470.
[117] M. Hiasa, T. Okui, Y.M. Allette, M.S. Ripsch, G.H. Sun-Wada, H. Wakabayashi, G. [140] J.A. Boyette-Davis, J.P. Cata, H. Zhang, L.C. Driver, G. Wendelschafer-Crabb, W.
D. Roodman, F.A. White, T. Yoneda, Bone pain induced by multiple myeloma is R. Kennedy, P.M. Dougherty, Follow-up psychophysical studies in bortezomib-
reduced by targeting V-ATPase and ASIC3, Cancer Res. 77 (6) (2017) 1283–1295. related chemoneuropathy patients, J. Pain 12 (9) (2011) 1017–1024.
[118] M. Sayilekshmy, R.B. Hansen, J.M. Delaisse, L. Rolighed, T.L. Andersen, A. [141] G.L. Currie, H.N. Angel-Scott, L. Colvin, F. Cramond, K. Hair, L. Khandoker,
M. Heegaard, Innervation is higher above bone remodeling surfaces and in J. Liao, M. Macleod, S.K. McCann, R. Morland, N. Sherratt, R. Stewart,
cortical pores in human bone: lessons from patients with primary E. Tanriver-Ayder, J. Thomas, Q. Wang, R. Wodarski, R. Xiong, A.S.C. Rice, E.
hyperparathyroidism, Sci. Rep. 9 (1) (2019) 5361. S. Sena, Animal models of chemotherapy-induced peripheral neuropathy: a
[119] G. Castaneda-Corral, J.M. Jimenez-Andrade, A.P. Bloom, R.N. Taylor, W. machine-assisted systematic review and meta-analysis, PLoS Biol. 17 (5) (2019),
G. Mantyh, M.J. Kaczmarska, J.R. Ghilardi, P.W. Mantyh, The majority of e3000243.
myelinated and unmyelinated sensory nerve fibers that innervate bone express [142] T. Li, H.C. Timmins, T. King, M.C. Kiernan, D. Goldstein, S.B. Park, Characteristics
the tropomyosin receptor kinase A, Neuroscience 178 (2011) 196–207. and risk factors of bortezomib induced peripheral neuropathy: a systematic
[120] S.R. Chartier, S.A.T. Mitchell, L.A. Majuta, P.W. Mantyh, The changing sensory review of phase III trials, Hematol. Oncol. 38 (3) (2020) 229–243.
and sympathetic innervation of the young, adult and aging mouse femur, [143] J. Bruna, E. Udina, A. Alé, J.J. Vilches, A. Vynckier, J. Monbaliu, L. Silverman,
Neuroscience 387 (2018) 178–190. X. Navarro, Neurophysiological, histological and immunohistochemical
[121] A.I. Basbaum, D.M. Bautista, G. Scherrer, D. Julius, Cellular and molecular characterization of bortezomib-induced neuropathy in mice, Exp. Neurol. 223 (2)
mechanisms of pain, Cell 139 (2) (2009) 267–284. (2010) 599–608.
[122] P.W. Mantyh, The neurobiology of skeletal pain, Eur. J. Neurosci. 39 (3) (2014) [144] S. Yamamoto, T. Kawashiri, H. Higuchi, K. Tsutsumi, S. Ushio, T. Kaname,
508–519. M. Shirahama, N. Egashira, Behavioral and pharmacological characteristics of
[123] A.P. Bloom, J.M. Jimenez-Andrade, R.N. Taylor, G. Castaneda-Corral, M. bortezomib-induced peripheral neuropathy in rats, J. Pharmacol. Sci. 129 (1)
J. Kaczmarska, K.T. Freeman, K.A. Coughlin, J.R. Ghilardi, M.A. Kuskowski, P. (2015) 43–50.
W. Mantyh, Breast cancer-induced bone remodeling, skeletal pain, and sprouting [145] C. Meregalli, A. Canta, V.A. Carozzi, A. Chiorazzi, N. Oggioni, A. Gilardini,
of sensory nerve fibers, J. Pain 12 (6) (2011) 698–711. C. Ceresa, F. Avezza, L. Crippa, P. Marmiroli, G. Cavaletti, Bortezomib-induced
[124] J.M. Jimenez-Andrade, J.R. Ghilardi, G. Castañeda-Corral, M.A. Kuskowski, P. painful neuropathy in rats: a behavioral, neurophysiological and pathological
W. Mantyh, Preventive or late administration of anti-NGF therapy attenuates study in rats, Eur. J. Pain 14 (4) (2010) 343–350.
tumor-induced nerve sprouting, neuroma formation, and cancer pain, Pain 152 [146] G. Cavaletti, A. Gilardini, A. Canta, L. Rigamonti, V. Rodriguez-Menendez,
(11) (2011) 2564–2574. C. Ceresa, P. Marmiroli, M. Bossi, N. Oggioni, M. D’Incalci, R. De Coster,
[125] W.G. Mantyh, J.M. Jimenez-Andrade, J.I. Stake, A.P. Bloom, M.J. Kaczmarska, R. Bortezomib-induced peripheral neurotoxicity: a neurophysiological and
N. Taylor, K.T. Freeman, J.R. Ghilardi, M.A. Kuskowski, P.W. Mantyh, Blockade pathological study in the rat, Exp. Neurol. 204 (1) (2007) 317–325.
of nerve sprouting and neuroma formation markedly attenuates the development [147] C.R. Robinson, P.M. Dougherty, Spinal astrocyte gap junction and glutamate
of late stage cancer pain, Neuroscience 171 (2) (2010) 588–598. transporter expression contributes to a rat model of bortezomib-induced
[126] M. Sopata, N. Katz, W. Carey, M.D. Smith, D. Keller, K.M. Verburg, C.R. West, peripheral neuropathy, Neuroscience 285 (2015) 1–10.
G. Wolfram, M.T. Brown, Efficacy and safety of tanezumab in the treatment of [148] K. Stockstill, T.M. Doyle, X. Yan, Z. Chen, K. Janes, J.W. Little, K. Braden,
pain from bone metastases, Pain 156 (9) (2015) 1703–1713. F. Lauro, L.A. Giancotti, C.M. Harada, R. Yadav, W.H. Xiao, J.M. Lionberger, W.
[127] C.M. Peters, J.R. Ghilardi, C.P. Keyser, K. Kubota, T.H. Lindsay, N.M. Luger, D. L. Neumann, G.J. Bennett, H.R. Weng, S. Spiegel, D. Salvemini, Dysregulation of
B. Mach, M.J. Schwei, M.A. Sevcik, P.W. Mantyh, Tumor-induced injury of sphingolipid metabolism contributes to bortezomib-induced neuropathic pain,
primary afferent sensory nerve fibers in bone cancer pain, Exp. Neurol. 193 (1) J. Exp. Med. 215 (5) (2018) 1301–1313.
(2005) 85–100. [149] M. Quartu, V.A. Carozzi, S.G. Dorsey, M.P. Serra, L. Poddighe, C. Picci, M. Boi,
[128] S.W.Z. Olechnowicz, M.M. Weivoda, S.T. Lwin, S.K. Leung, S. Gooding, G. Nador, T. Melis, M. Del Fiacco, C. Meregalli, A. Chiorazzi, C.L. Renn, G. Cavaletti,
M.K. Javaid, K. Ramasamy, S.R. Rao, J.R. Edwards, C.M. Edwards, Multiple P. Marmiroli, Bortezomib treatment produces nocifensive behavior and changes
myeloma increases nerve growth factor and other pain-related markers through in the expression of TRPV1, CGRP, and substance P in the rat DRG, spinal cord,
interactions with the bone microenvironment, Sci. Rep. 9 (1) (2019) 14189. and sciatic nerve, Biomed. Res. Int. 2014 (2014), 180428.
[129] M. Diaz-delCastillo, D. Kamstrup, R.B. Olsen, R.B. Hansen, T. Pembridge, [150] H. Zheng, W.H. Xiao, G.J. Bennett, Mitotoxicity and bortezomib-induced chronic
B. Simanskaite, J.M. Andrade-Jimenez, M.A. Lawson, A.-M. Heegaard, painful peripheral neuropathy, Exp. Neurol. 238 (2) (2012) 225–234.
Differential pain-related behaviours and bone disease in immunocompetent [151] I. Casafont, M.T. Berciano, M. Lafarga, Bortezomib Induces the Formation of
mouse models of myeloma, JBMR Plus 4 (2020), e10252. Nuclear poly(A) RNA Granules Enriched in Sam68 and PABPN1 in Sensory
[130] S. Mercadante, Malignant bone pain: pathophysiology and treatment, Pain 69 Ganglia Neurons, Neurotox. Res. 17 (2) (2009) 167–178.
(1–2) (1997) 1–18. [152] D.-L. Esseltine, L. Dick, E. Kupperman, M. Williamson, K.C. Anderson, Second-
[131] P. Honore, N.M. Luger, M.A. Sabino, M.J. Schwei, S.D. Rogers, D.B. Mach, Generation Proteasome Inhibitors, in: I.M. Ghobrial, P.G. Richardson, K.
F. O’Keefe P, M.L. Ramnaraine, D.R. Clohisy, P.W. Mantyh, Osteoprotegerin C. Anderson (Eds.), Bortezomib in the Treatment of Multiple Myeloma, Springer
blocks bone cancer-induced skeletal destruction, skeletal pain and pain-related Basel, Basel, 2011, pp. 149–176.
neurochemical reorganization of the spinal cord, Nat. Med. 6 (5) (2000) 521–528. [153] F. Mols, S. Oerlemans, A.H. Vos, A. Koster, S. Verelst, P. Sonneveld, L.V. van de
[132] M.J. Schwei, P. Honore, S.D. Rogers, J.L. Salak-Johnson, M.P. Finke, M. Poll-Franse, Health-related quality of life and disease-specific complaints among
L. Ramnaraine, D.R. Clohisy, P.W. Mantyh, Neurochemical and cellular multiple myeloma patients up to 10 yr after diagnosis: results from a population-
reorganization of the spinal cord in a murine model of bone cancer pain, based study using the PROFILES registry, Eur. J. Haematol. 89 (4) (2012)
J. Neurosci. 19 (24) (1999) 10886–10897. 311–319.
[133] W. Shen, X.M. Hu, Y.N. Liu, Y. Han, L.P. Chen, C.C. Wang, C. Song, CXCL12 in [154] K. Cocks, D. Cohen, F. Wisloff, O. Sezer, S. Lee, E. Hippe, P. Gimsing, I. Turesson,
astrocytes contributes to bone cancer pain through CXCR4-mediated neuronal R. Hajek, A. Smith, L. Graham, A. Phillips, M. Stead, G. Velikova, J. Brown, An
sensitization and glial activation in rat spinal cord, J. Neuroinflamm. 11 (2014) international field study of the reliability and validity of a disease-specific
75. questionnaire module (the QLQ-MY20) in assessing the quality of life of patients
[134] X. Liu, H. Bu, C. Liu, F. Gao, H. Yang, X. Tian, A. Xu, Z. Chen, F. Cao, Y. Tian, with multiple myeloma, Eur. J. Cancer 43 (11) (2007) 1670–1678.
Inhibition of glial activation in rostral ventromedial medulla attenuates

58

You might also like