You are on page 1of 13

Supportive Care in Multiple Myeloma

31
Simit Mahesh Doshi, Tom T. Noff, and G. David Roodman

Myeloma Bone Disease tion of hypercalcemia or fracture, and suppressing the growth
of MM cells in the bone marrow.
Multiple myeloma (MM) is the second most common hema-
tologic malignancy, affecting more than 60,000 patients in
the United States with 30,000 patients diagnosed in 2016 Pathophysiology of Myeloma Bone Disease
[1, 2]. Multiple myeloma (MM) is the most frequent cancer
to involve bone with almost 70% of patients presenting with During the normal bone-remodeling process, osteoclastic bone
bone lesions at diagnosis [3]. Twenty percent of patients resorption is coupled to osteoblastic bone formation, so that
present with a pathologic fracture at diagnosis and until bone removal and formation are balanced (Fig. 31.1). However,
recently up to 60% of patients developed a pathologic frac- in MM the normal bone-remodeling process is essentially
ture over the course of their disease [7]. This is extremely uncoupled, with markedly increased bone resorption at sites of
important since pathologic fractures increase mortality of MM and absent or severely decreased bone formation. This is
MM patients by 20% [8]. In addition, MM bone disease can due to suppression of osteoblast differentiation that can persist
cause excruciating bone pain that remains undertreated in even when patients are in long-term remission [10]. Thus, little
many patients, and hypercalcemia that can be life threaten- or no repair of lytic lesions occurs in the vast majority of
ing, as well as require surgery and/or radiation to bone to patients with MM, although there have been anecdotal reports
control bone pain, treat impending or repair fractures, and of healing of bone lesions in patients receiving bortezomib-
relieve spinal cord compression. Further, in the vast majority based therapy. However, once a patient’s bone density is below
of patients, MM bone lesions rarely heal even when patients the fracture threshold, the patient continues to be at an increased
are in long-term remission [10], and MM patients continue risk of fracture and experience persistent bone pain.
to suffer from the sequelae of their bone disease, even when A multiplicity of osteoclast stimulating factors are produced
their MM is under excellent control. Tremendous progress by MM cells and induced by MM cells in the bone marrow
has been made in the treatment of MM, with a median sur- microenvironment [10]. These factors include MM cell pro-
vival of patients increasing to more than 6.1 years in the last duction of Rank ligand, a highly potent osteoclast-­stimulating
decade and a much better overall survival for patients over factor, or induction of Rank ligand production by marrow stro-
the age of 65 [9]. Thus, treatment and repair of MM bone mal cells and osteocytes in the MM microenvironment.
disease are increasingly important, both for enhancing the Furthermore, production of MIP-1 α by MM cells, interleukin
quality of life of patients, increasing their survival, preven- 3 production by both MM cells and T-lymphocytes in the MM
microenvironment, and IL-6 production by marrow stromal
cells, osteoclasts, and other cells in the bone microenvironment
S.M. Doshi, M.D., M.P.H.
Department of Nephrology, IUH University Hospital, also occur [10]. In addition, osteoclastic bone resorption and
950 W Walnut Street, Indianapolis, IN 46202, USA increased osteoclast numbers enhance the growth of MM cells.
T.T. Noff, M.D. This process has been described as the vicious cycle hypothe-
Department of Neurology, University of Indiana School of sis, in which MM cells induce osteoclastic bone resorption, and
Medicine, 355 W 16th Street, Indianapolis, IN 46202, USA the bone resorption process releases growth factors, such as
G. David Roodman, M.D., Ph.D. (*) IGF1 and TGF-beta from bone matrix, which in turn stimulate
Department of Hematology and Oncology, Indiana University the growth of MM cells (Fig. 31.2). In addition, MM cells
School of Medicine, 980 W Walnut Street, Suite C312,
induce polarization of T cells in the tumor microenvironment,
Indianapolis, IN 46202, USA
e-mail: groodman@iu.edu which reverses the ratio of Th17 to Th1 T cells to 10:1 com-

© Springer International Publishing AG 2018 595


P.H. Wiernik et al. (eds.), Neoplastic Diseases of the Blood, DOI 10.1007/978-3-319-64263-5_31
596 S.M. Doshi et al.

Fig. 31.1  Bone remodeling


is uncoupled in myeloma. The
normal bone-remodeling
process in which bone
resorption is followed by new
bone at the same site is
uncoupled in myeloma with
increased bone resorption
followed by little or no bone
formation. Adapted by
permission from Macmillan
Publishers Ltd: Hattner R
et al. Nature. 1965;206:489

Fig. 31.2  Reciprocal interactions between myeloma cells and bone bone formation. In addition, myeloma cells directly interact with
microenvironment contribute to progression of myeloma. Myeloma osteocytes in the bone matrix and these interactions increase myeloma
cells colonize the marrow and some of the cells engage the endosteal cell growth and osteoclast formation as well as block osteoblast dif-
niche and become dormant, while other cells remain active myeloma ferentiation. Finally, osteoclastic resorption also releases dormant
cells. Active myeloma cells produce factors and induce factors by myeloma cells to become active myeloma cells, further contributing
cells in the bone microenvironment to increase osteoclast formation to myeloma progression. Adapted from Roodman GD. N Engl J Med.
and resorption. The increased bone resorption in turn releases growth 2004;350(16):1655–1664. Copyright © 2004 Massachusetts Medical
factors from bone matrix to stimulate the growth of myeloma cells. Society. Reprinted with permission
Myeloma cells also suppress osteoblast differentiation to block new

pared to the normal ratio of 1:10. Th17 cells produce IL17 that increased numbers of osteoclasts directly support MM
induces dendritic cell differentiation toward the osteoclast lin- growth. Yaccoby and coworkers demonstrated that primary
eage, as well as enhance induction of Rank ligand. MM cells from patients can be passaged over feeder layers of
MM cells, osteoclasts, and marrow stromal cells in the osteoclasts for long periods of time [12]. Abe and coworkers
microenvironment also produce angiogenic factors that fur- found that osteoclasts support the growth of primary MM
ther increase the growth of MM cells [11]. In addition, the cells through the production of IL-6 and osteopontin [13].
31  Supportive Care in Multiple Myeloma 597

Finally, direct interactions between osteoclasts and osteo- MM bone disease (see below). However, none of these fac-
cytes enhance bidirectional Notch signaling between the tors can explain why osteoblast suppression persists in
cells to increase the growth of MM cells, induce osteocyte patients even in the absence of MM cells. The mechanisms
apoptosis, and increase production of sclerostin and Rank responsible for the long-term suppression of osteoblast dif-
ligand by osteocytes that suppresses osteoblast differentia- ferentiation in MM are just beginning to be understood.
tion and stimulates osteoclastogenesis [14]. In addition, D’Souza and all demonstrated that epigenetic changes in the
osteoclasts produce Annexin II, BAFF, and April that sup- Runx2 promoter in pre-osteoblasts, the master gene control-
port MM cell growth [15, 16]. ling osteoblast differentiation, are induced by MM cells [23].
Recently, osteoclastic bone resorption has also been shown These changes in the Runx2 promoter result from induction
to play an important role in activating dormant MM cells in of Gfi1 in pre-osteoblastic marrow stromal cells exposed to
the MM microenvironment to active MM cells. Activation of MM cells. Gfi1 acts as a transcriptional repressor of Runx2
dormant MM cells results in increased tumor burden, bone and blocks osteoblast differentiation. Increased expression
destruction, and distant colonization of bones. Lawson et al. of Gfi1 persists in marrow stromal cells from MM patients,
recently showed by intravital imaging that treatment of mice even when the stromal cells are passaged for long periods of
with Rank ligand increases osteoclast numbers and decreases time in the absence of MM cells. Furthermore, knockdown
the numbers of dormant MM cells colonizing the osteoblast of Gfi1 in marrow stromal cells from MM patients allows
niche [17]. Thus, osteoclasts play multiple roles in progres- them to undergo osteoblast differentiation. These insights
sion of MM, including activation of dormant MM cells, bone into the mechanisms responsible for long-term suppression
destruction that results in release of matrix-bound growth fac- of osteoblast differentiation have provided potential new
tors to stimulate tumor growth, and enhancement of MM therapeutic targets for repairing bone lesions in MM patients.
growth by factors derived from osteoclasts. Furthermore, Li and coworkers reported that mature osteo-
Studies in preclinical models of MM found that blocking blasts could suppress MM cell growth through the produc-
osteoclastic bone resorption decreases tumor burden in addi- tion of decorin [24]. These results suggest that in addition to
tion to bone destruction [18]. However, until recently this repairing bone lesions, inducing osteoblast differentiation
phenomenon has not been demonstrated in patients. The may also suppress the growth of MM cells in patients.
MM IX trial showed that treating newly diagnosed MM
patients with zoledronate, a potent bisphosphonate that
blocks osteoclastic bone resorption, rather than with clodro- Imaging of Myeloma Bone Disease
nate, a weaker bisphosphonate, enhances survival of MM
patients. This increase in survival occurred regardless if the Imaging of MM bone disease has become increasingly impor-
patients had detectable bone disease at diagnosis [19]. tant with the development of new criteria for the diagnosis of
However, the greatest benefit in survival was seen in patients active MM. These criteria include presence of at least one
with detectable bone disease. Based on preclinical and clini- lytic lesion detected by conventional radiography, whole-
cal studies, the International Myeloma Working Group body low-dose CT or PET/CT, or presence of more than one
(IMWG) guidelines now support targeting osteoclastic bone focal bone marrow lesion greater than or equal to 5 mm on
resorption in all patients with MM [20]. MRI studies [25]. Skeletal surveys have been the gold stan-
dard for detecting MM bone disease. The lesions appear as
punched-out lytic bone lesions with the absence of reactive
Osteoblast Suppression in Myeloma new bone formation and can occur in any bone. However,
conventional radiographs are relatively insensitive for detect-
As noted above, bone formation is markedly suppressed in ing bone disease in MM, since more than 30% of bone must
patients with MM and persists even when the MM is under be removed before a lesion is detectable. Conventional radio-
excellent control. This explains why bone scans, which mea- graphs are also not useful for following disease progression
sure reactive bone formation, frequently under-represent or response to therapy, because lytic lesions rarely heal in
bone disease in MM patients, making bone scans inappropri- these patients and enumerating new lesions can frequently be
ate for imaging bone disease in MM patients [21]. Multiple difficult in patients with extensive bone disease. In addition, it
factors produced by MM cells or induced by MM cells in the is often difficult to distinguish between osteoporotic and
MM microenvironment block osteoblast precursor differen- MM-induced vertebral fractures. Finally, skeletal surveys
tiation to mature osteoblasts. A large number of inhibitors of take a great deal of time to perform, which can be difficult for
osteoblast differentiation have been identified in MM patients who have severe bone pain.
patients, including TNFα, MIP-1 α, IL-3, activin A, scleros- Low-dose whole-body CT has been recently shown to be
tin, DKK1, hepatocyte growth factor, TGF-beta, and IL7 superior to conventional radiographs for detecting osteolytic
[22]. All these factors can directly suppress osteoblast dif- lesions in MM patients, and is especially useful for detecting
ferentiation and are potential therapeutic targets for treating lesions in the spine or pelvis [26]. Low-dose whole-body CT
598 S.M. Doshi et al.

can take less than 5 min to perform, and has the advantage that bone, fractures, radiation to bone, and spinal cord compres-
it can also demonstrate extraosseous lesions. Because of these sion) and bone pain in patients with MM bone disease [33].
advantages, low-dose whole-body CT is becoming the stan- Pamidronate and zoledronic acid are the cornerstone of treat-
dard screening method for MM bone disease in Europe. ment for MM bone disease. Bisphosphonates are non-­
However, although low-dose whole-body CT exposes the hydrolyzable pyrophosphate analogues that block
patients to a higher dose of radiation than standard skeletal osteoclastic bone resorption and inhibit osteoclast formation
surveys, the higher resolution and speed of the examination through their capacity to inhibit protein prenylation by inhib-
outweigh the disadvantages. Many experts predict that low-­ iting farnesyl diphosphate synthase required for normal
dose whole-body CT will replace skeletal surveys as the new osteoclast activity [34]. Pamidronate is usually administered
gold standard for detecting MM bone disease [26]. as a 90 mg intravenous infusion over 2 h, while its more
Magnetic resonance imaging (MRI) is also more sensitive potent analogue, zoledronic acid, is administered as 4 mg
than standard skeletal radiographs for detecting MM bone dis- intravenously over 15 min. Both bisphosphonates have simi-
ease, and also detects bone marrow infiltration by MM cells. A lar efficacy for treating MM bone disease and are routinely
large study from the University of Arkansas showed that MRI given every 3–4 weeks. Bisphosphonates are very safe drugs
detected focal lesions in 74% of MM patients, compared to and their major toxicity is their potential deleterious effects
56% of patients who were examined with whole-body radio- on renal function. Therefore, serum creatinine levels must be
graphs [27]. MRI is particularly useful for distinguishing measured prior to infusion. If creatinine levels in patients
between asymptomatic smoldering MM and symptomatic receiving these agents increase more than 10% from their
MM. Patients who were thought to have smoldering MM but baseline values, pamidronate or zoledronic acid should be
had more than one focal lesion on MRI had a 70% probability withheld until the serum creatinine returns to within 10% of
of progression to active MM within 2 years [28]. These results the previous baseline value. In patients who present with
form the basis for adding the presence of more than one focal impaired renal function, the infusion time for pamidronate
criteria on MRI to the updated criteria for active MM [25]. The can be increased or the dose of zoledronate can be reduced,
International Myeloma Working Group recommends, at a min- since renal toxicity of bisphosphonates is related to peak
imum, MRI examination of the spine and pelvis as part of the dose [35]. In patients who present with severe renal failure,
evaluation for smoldering MM, if whole-body MRI cannot be initial therapy with bortezomib-based regimens should be
done [29]. However, MRI limited to the spine and pelvis will started prior to initiation of bisphosphonate therapy (see
not detect lesions in the peripheral skeleton that occur in 10% below).
of patients who present with focal lesions [30]. Multiple organizations have developed guidelines for use
PET/CT is a very useful albeit expensive imaging technique of bisphosphonates in MM. Each of these guidelines state
for assessing MM bone disease. PET detects the hypermeta- that bisphosphonates can be given intravenously every 3–4
bolic activity of MM cells in both intraosseous and extramedul- weeks for patients with MM bone disease that is identified
lary sites, while CT detects bone-destructive lesions. PET/CT by plain radiographs or MRI. More recent IMWG recom-
has a similar sensitivity to that of MRI, and is better than MRI mendations for treatment of MM-related bone disease state
at detecting bony lesions. More importantly, PET/CT can be that bisphosphonate therapy should be considered for all
used to assess response to therapy [31]. Further, the presence of patients with MM, regardless of the presence of osteolytic
extramedullary lesions or an SUV Max greater than 4.21 on a bone disease by conventional radiography [20]. These rec-
PET/CT performed at diagnosis, persistence of FDG uptake ommendations are based on the results of the MM IX trial
following autologous stem cell transplantation for MM, or that compared clodronate to zoledronic acid therapy in more
detection of extramedullary disease are all associated with a than 1000 newly diagnosed patients receiving treatment for
poor prognosis for patients. However, it is unclear if PET/CT or MM (see above). This study found that patients receiving
MRI is better for determining if patients with solitary plasma- zoledronate achieved a 5-month survival benefit compared to
cytoma have disease restricted to one site or multiple sites. patients receiving clodronate [19]. This survival advantage
PET/CT is also useful for following patients with non-secre- occurred regardless if the patient had demonstrable bone
tory MM [32]. However, PET/CT may also detect false-posi- ­disease at diagnosis. A subsequent secondary analysis of this
tive findings in patients following radiotherapy or infections. study showed that the major impact on survival occurred in
patients with bone disease [36]. However, it’s unclear how
long patients should continue to receive bisphosphonate
Treatment of MM Bone Disease therapy. This question became a major concern with the rec-
ognition that some MM patients receiving bisphosphonates
The landmark studies of Berenson and coworkers in 1996 develop bisphosphonate-associated osteonecrosis of the jaw
demonstrated that the nitrogen- containing bisphosphonate, (BRONJ). This complication of bisphosphonate therapy was
pamidronate, decreased skeletal related events (surgery to first recognized by Marx et al. [37]. The occurrence of
31  Supportive Care in Multiple Myeloma 599

BRONJ is usually dependent on the duration and dose of a superior effect on skeletal related events compared with
bisphosphonate therapy, undergoing invasive dental proce- zoledronate, and patients treated with denosumab who had
dures, age of the patient, and treatment with glucocorticoids. breast cancer bone metastasis had a survival advantage [42].
BRONJ occurs more frequently in patients receiving zole- A large phase 3 study compared denosumab to zoledronic
dronate than pamidronate [37]. Because of concern for acid treatment in patients with bone metastasis that did not
development of BRONJ, most guidelines recommend treat- have prostate cancer or breast cancer, and also included 200
ment with bisphosphonates for approximately 2 years, unless MM patients. This study found a similar decrease in skeletal
active MM persists, and then stopping or increasing the related events in the MM patients with either treatment, but a
interval between administration of bisphosphonates should post hoc analysis found a decreased survival for MM patients
be considered. However, with the regular institution of close receiving denosumab [43]. Further analysis of this trial found
dental monitoring, dental prophylaxis, and performing most that the baseline characteristics of the MM patients receiving
dental procedures prior to initiating bisphosphonate therapy, zoledronic acid or denosumab were not balanced, and that
the incidence of BRONJ has markedly decreased. the patients receiving zoledronic acid treatment had more
Another question that often occurs is should bisphospho- early withdrawals from the study compared to those treated
nate therapy should be stopped prior to invasive dental pro- with denosumab [43]. A large phase 3 study comparing
cedures. The American Dental Association recently denosumab to zoledronic acid treatment in newly diagnosed
recommended not stopping bisphosphonates or other bone-­ patients with MM bone disease has just been completed, and
targeted agents when patients undergo dental procedures, the results of this trial should be reported soon. It is impor-
and states that the decision to withhold bisphosphonate ther- tant to note that the incidence of ONJ is similar in patients
apy should be based primarily on the patient’s risk of devel- receiving denosumab or zoledronic acid so that similar den-
oping a skeletal related event [38]. In contrast, the IMWG tal screening, routine dental prophylaxis, and follow-up are
recommends stopping bisphosphonates for 90 days before also required for patients receiving denosumab. Finally, nei-
and after invasive dental procedures, such as tooth extrac- ther denosumab nor zoledronic acid increases bone forma-
tion, dental implants, and surgery to the jaw [20]. tion in patients with MM, so there is still a great need for
Several studies assessed if bone resorption markers are bone anabolic agents that can increase bone formation and
useful for determining the interval between bisphosphonate are safe for MM patients.
administration in patients with MM bone disease. Patel and Recently, several agents have been developed to enhance
coworkers found in patients who had received bisphospho- bone formation in patients with MM. These include an anti-
nate therapy for at least a year and had stable disease that the body to DKK1, BHQ880, and sotatercept, an activin recep-
level of bone resorption markers could be used to determine tor antagonist [22]. A phase 1B clinical trial of anti-DKK1 in
when to administer bisphosphonate therapy [39]. However, patients with MM did not show any benefit for MM bone
other studies show that bone resorption markers are not suf- disease [44], although the anti-DKK1 did show some bone
ficiently sensitive to guide bisphosphonate therapy [40]. anabolic effects in a phase 2 trial of patients with smoldering
Importantly, the MM IX trial demonstrated that extending MM. No anti-MM activity was found in MM patients or
bisphosphonate treatment beyond 2 years continued to have smoldering MM patients receiving anti-DKK1 therapy.
beneficial effects on skeletal related events [36]. However, Sotatercept is in clinical trial for MM but can also increase
only small numbers of patients received long-term bisphos- hemoglobin levels significantly [45]. Thus, its utility as a
phonate therapy in that study. Another consideration with bone anabolic agent of MM is still unclear. Most recently an
bisphosphonate therapy is whether bisphosphonate therapy anti-sclerostin antibody has been developed and is in clinical
can be restarted in patients who developed BRONJ. A retro- trial for patients with osteoporosis [46]. Anti-sclerostin anti-
spective analysis of over 100 MM patients demonstrated that body had potent bone anabolic effects and was well tolerated
about 50% of patients who developed BRONJ healed their by the patients. Preclinical studies demonstrated that anti-­
lesions and 50% of those who restarted bisphosphonates did sclerostin antibody treatment of murine models of MM can
not develop new lesions [41]. These results suggest that increase bone formation and may affect tumor burden.
patients who have active MM and have healed their osteone- Several agents used to treat MM also have effects on bone.
crosis of the jaw can receive bisphosphonate therapy but Bortezomib has been reported to have bone anabolic effects
should be monitored closely for a reoccurrence of BRONJ. in patients with MM whose MM responded to bortezomib
In addition to bisphosphonates, denosumab, a human therapy [47], and transient increases in alkaline phosphatase
monoclonal antibody that targets Rank ligand, has been activity have been shown in patients receiving bortezomib
developed, and potently inhibits osteoclast formation and therapy. In addition there have been anecdotal reports of
bone resorption. Multiple large phase 3 studies of deno- bone healing in patients receiving bortezomib-based thera-
sumab versus zoledronate treatment for patients with bone pies [48]. Furthermore, in preclinical studies, immunomodu-
metastasis found that denosumab has either a non-inferior or latory agents such as pomalidomide can block osteoclast
600 S.M. Doshi et al.

differentiation [47]. Finally, the role of parathyroid hormone coworkers found that VTE can occur in MM patients at a
as an anabolic agent for MM bone disease remains unclear. median time from diagnosis of 952 days, much longer than
Several groups reported the presence of the PTH receptor 1 the IMWG guidelines recommend for thromboprophylaxis.
on MM cells and found that parathyroid hormone-related Importantly, Lipe et al. found that of 60% of patients at high
protein, which activates the PTH receptor, can increase MM risk for thrombosis, only 16% ever received the recommended
cell growth [49]. Other investigators failed to demonstrate prophylactic therapy. These results suggest that VTE may
PTH receptors on MM cells and preclinical studies have occur later in MM than was previously thought, and that
shown an anabolic effect of PTH in SCID-hu mouse model guidelines for thromboprophylaxis in MM may need to be
of MM [50]. adjusted. Finally, the use of erythropoiesis-­stimulating agents
(ESAs) also increases the risk of thrombosis in patients with
MM. Katodritou and coworkers reported that ESAs reduced
 adiation Therapy and Surgery for Myeloma
R survival in MM patients compared to patients not receiving
Bone Disease ESAs (and increased the risk of thrombosis in patients with
MM) [54]. Thus, ESAs should be used with caution in patients
Radiation therapy is frequently used to treat painful bone with MM.
lesions in myeloma. However, it must be used sparingly
because of its myelosuppressive effects. Thus, radiation
therapy is used primarily to treat painful lesions, impend- Neuropathy in Myeloma
ing fracture and spinal cord compression [20]. Similarly,
vertebroplasty and kyphoplasty are used to treat painful Neurological complications in MM range from peripheral
vertebral fractures in patients with myeloma [51]. In a ran- neuropathy to compression of the nerve roots or spinal cord.
domized study for painful vertebral fractures in patients with Compression of the nerve roots and spinal cord is an onco-
myeloma, kyphoplasty was shown to be superior to conser- logical emergency and can result from compression by verte-
vative management, so that kyphoplasty is currently recom- bral and extramedullary plasmacytomas, respectively. Early
mended by the IMWG guidelines. recognition and treatment of spinal cord or nerve root com-
pression are more important than the type of treatment in
improving outcomes.
Thromboprophylaxis for Myeloma Patients

Patients with MM have an increased risk of a thrombotic Radiculopathy


event during the course of their disease. In the original clini-
cal trial of thalidomide for treatment of patients with MM, a Compression of the nerve root by vertebral plasmacytomas,
30% incidence of DVT was reported in patients receiving tha- neuroforaminal stenosis, or vertebral fracture usually pres-
lidomide [52]. Subsequent studies using combination chemo- ents with back pain and a unilateral radiculopathy that may
therapy regimens containing thalidomide also found a high progress to sensory loss and weakness. At least 5% of MM
rate of deep venous thrombosis (DVT) in these patients. patients suffer from spinal cord or cauda equina compres-
Lenalidomide and pomalidomide are also associated with sion, and MM is responsible for 15% of hospitalization for
increased risk of DVT [52]. These results led to the develop- malignant spinal cord compression [55]. Spinal cord com-
ment of thromboprophylaxis guidelines for patients receiving pression often presents with progressively worsening back
immunomodulatory agents [6]. These guidelines suggest that pain and may have a symmetric radiculopathy. The thoracic
patients should receive thromboprophylaxis for at least the cord is most commonly affected and is associated with a
first four cycles of immunomodulatory agent-based therapy sensory-level deficit. Bowel and bladder dysfunction is a
because the risk of DVT or pulmonary embolism (PE) is late complication of spinal cord compression and is often
greatly increased during that time. The type of DVT prophy- preceded by symmetric pain and weakness. Unilateral
laxis is based on individual risk factors (e.g., age, obesity ­complaints are rare but may be seen with lateralized cord
immobilization, history of DVT). Patients who have no or compression. Complete spinal MRI with contrast is the pre-
only one risk factor should receive prophylaxis with low-­dose ferred method of evaluating spinal cord compression, as CT
or standard-dose aspirin (81–325 mg per day) and patients does not clearly demonstrate the spinal cord or epidural
with two or more risk factors should receive low-­molecular-­ space. Immediate treatment with corticosteroids, followed
weight heparin or the equivalent or full-dose warfarin therapy by radiation therapy, is the mainstay of treatment. MRI
to maintain the INR at 2–3. A recent abstract at the American screening of MM patients for cord compression has not
society of clinical oncology found that the risk of VTE persis- been found to be beneficial for detecting spinal cord com-
tently changes throughout the disease process [53]. Lipe and pression [56].
31  Supportive Care in Multiple Myeloma 601

Clinically detectable peripheral neuropathy is uncommon 21% and 37% of patients at the onset of treatment at doses of
in MM at diagnosis and presents as a mild and slowly pro- 1.0 mg/m2, and 1.3 mg/m2, respectively. The severity typi-
gressive symmetric neuropathy. Occurrence rates vary from cally plateaus by the fifth cycle, with an approximate cumu-
2% [5] to approximately 50% [57] of patients. Peripheral lative dose of 30 mg/m2 [65]. The pathogenesis of BIPN
neuropathy in MM is often caused by accompanying amyloi- remains unclear. The involvement of satellite and Schwann
dosis or chemotherapy, and is a major presenting feature of cells in BIPN in animal models supports the involvement of
POEMS syndrome. Cranial nerve involvement is rare and is the dorsal root ganglia in the pathogenesis of the pain.
usually a sign of progressive disease. Chemotherapy-induced Although the autonomic nervous system is also innervated
peripheral neuropathy is a serious and potentially reversible by unmyelinated fibers, it is very rarely affected. The clinical
side effect of treatment. The dose and duration of treatment presentation of a symmetric distal peripheral neuropathy
are commonly affected by the neurotoxic profiles of these with a temporal correlation to recent bortezomib treatment is
medications, which in severe cases may result in discontinu- suggestive of BIPN. Electrodiagnostic studies can provide
ation of treatment. The clinical history and neurological objective evidence to aid in the diagnosis. However, the diag-
examination are crucial for distinguishing CIPN from neuro- nosis can usually be made clinically. The neurologic exami-
pathic involvement from direct nerve compression by nation is very important for distinguishing the cause of
MM. Neuropathy is a frequent complication of thalidomide- peripheral neuropathy. If signs of motor weakness are pres-
and bortezomib-based therapies. ent, nerve root compression is more likely, since BIPN is
Thalidomide is a potent antiangiogenic therapy that has primarily a sensory neuropathy. In patients with BIPN, nerve
been used for MM since 1998. Severe peripheral neuropathy conduction studies show reduced sensory and motor action
occurs in approximately 30% of patients who were treated potentials, with mild slowing of the distal sensory and motor
with doses of thalidomide greater than 200 mg/day [58]. nerve velocities with increased distal motor latencies.
Thalidomide-induced peripheral neuropathy presents as a Bortezomib neurotoxicity is a potentially reversible com-
symmetric distal loss of light touch and temperature that is plication that must be recognized early. Currently, there is no
painful. The neurotoxic effects of thalidomide are cumula- accepted proven treatment for BIPN. The mainstay of treat-
tive and dose dependent. In approximately 60% of patients, ment remains bortezomib dose modification or discontinua-
neuropathy results in lowering the dose or discontinuation of tion and neuropathic pain medication. Subcutaneous
treatment [59]. Reduced starting doses of thalidomide of administration of bortezomib has dramatically reduced the
100 mg/day for patients greater than 75 years old, and incidence of neuropathy, and newer proteasome antagonists
200 mg/day for patients less than 75 years old, have reduced (carfilzomib and ixazomib) are less neurotoxic and better
the incidence of severe peripheral neuropathy to less than tolerated. Severe peripheral neuropathy was reported in less
10%. However, there has been an overall increase in the inci- than 1% of patients in phase II trials of carfilzomib who were
dence of mild-to-moderate peripheral neuropathy at these treated previously with bortezomib [66, 67]. There is a lack
thalidomide doses. Although peripheral neuropathy is par- of evidence to support prevention of BIPN with neuroprotec-
tially reversible with discontinuation of thalidomide, the tive agents [65].
long-term reversibility of neuropathy with discontinuation of Neurologic involvement is a hallmark of POEMS syn-
thalidomide requires further long-term studies [60, 61]. The drome, a rare osteoscerotic form of MM, and is a major cri-
pathogenesis of thalidomide-induced peripheral neuropathy terion for diagnosis. Approximately 50% of patients present
is poorly understood but antiangiogenic and inflammatory with a distal symmetric neuropathy that ascends proximally.
insults to the dorsal root ganglia may be the cause. This may Eventually the vast majority of POEMS patients suffer from
explain the reduced sensory nerve action potentials with rel- prominent sensorimotor neuropathy. Severe neuropathy has
ative sparing of compound motor action potentials found in been reported in up to 76% of patients [68, 69]. Ocular
thalidomide-treated patients [62]. Other neurotoxic side involvement is common in approximately 2/3 of patients.
effects of thalidomide include tremor (in up to 30%) and Papilledema occurs in approximately half of those patients,
somnolence. and patients with papilledema have a worse prognosis [70].
Bortezomib-associated peripheral neuropathy was recog- There is also an increased risk of thromboembolic disease
nized as an early side effect in the initial phase I trials. The and cerebral infarction in POEMS patients. In a retrospective
incidence of bortezomib-induced peripheral neuropathy cohort study of 208 POEMS patients 19 (9%) developed
(BIPN) in phase II and phase III trials was 30–64%, and cerebral infarction at a relatively young age [71]. The risk of
occurred in up to 46% of patients treated for newly diag- cerebral infarction increased with the degree of thrombocy-
nosed MM [62, 63]. BIPN is a subacute predominately sen- tosis, and of plasma cell proliferation in the BM was also
sory neuropathy of the feet and hands that is often painful. associated with increased stroke risk [71]. Electrodiagnostic
Small unmyelinated fibers are affected, causing prominent studies are important to distinguish neuropathy due to
neuropathic pain in 25–80% of cases [64]. BIPN occurs in POEMS from CIDP; in contrast to CIDP, prominent axonal
602 S.M. Doshi et al.

loss in the lower limbs rather than slow conduction of the urine collection for proteinuria and urine electrophoresis.
distal nerve segments with conduction block occurs with The presence of selective proteinuria composed almost
POEMS. Polyneuropathy is commonly the initial presenting entirely of light chains can be attributed to myeloma cast
symptom of POEMS with up to 60% of patients with POEMS nephropathy (MCN), whereas the presence of significant
initially misdiagnosed with CIDP. Thus, distinguishing albuminuria suggests alternative diagnoses that require fur-
between CIDP and POEMS is critically important for treat- ther investigation [78]. A renal biopsy should be considered
ment and prognosis. Nerve conduction studies in POEMS to exclude monoclonal immunoglobulin deposition disease,
show predominate slowing of the nerve trunk when com- amyloidosis, or other underlying causes of renal impairment
pared to distal nerve involvement. in the presence of nonselective proteinuria [78–80]. If amy-
loidosis is strongly suspected, a fat pad aspirate should be
done prior to kidney biopsy, since a diagnosis of amyloidosis
Renal Dysfunction in Multiple Myeloma can be obtained in about 70% of cases with the less invasive
procedure [80].
The characteristic M paraprotein in MM, specifically the light-
chain component, is nephrotoxic to the kidney, and frequently
the kidney is the major target organ in MM. Up to 50% of Pathogenesis of Myeloma Kidney Disease
patients have evidence of renal impairment at the time of diag-
nosis and about 10–15% of patients require renal replacement Free light chains circulating as monomers (predominantly Κ,
therapy [72]. Multiple studies found that renal impairment 25KD) or dimers (predominantly λ, 50KD) are freely filtered
alone is a strong negative predictor of overall survival (OS) [4, by the glomerulus. In the proximal tubule cells (PTC) they
73]. In one study, the difference in median OS between patients are reabsorbed by receptor-mediated endocytosis [81]. This
with and without renal impairment was as high as 8.6 vs. 34.5 glycoprotein receptor cubilin-mediated endocytosis incites a
months (P < 0.001) The same study also showed markedly proinflammatory response that induces release of IL-6, IL-8,
improved OS if renal impairment was reversible [4]. The and monocyte chemoattractant protein 1 (MCP-1) via activa-
Nordic MM study group also found that severity of renal tion of nuclear factor kappa light-chain enhancer of activated
impairment was a determinant of OS MM [73]. Hypercalcemia, B-cells (NF-kB) in the PTC [82]. Similar to other proteinuric
lower proteinuria (<1 g/day), and low serum creatinine were diseases, excess light chains induce factors that promote
positive prognostic factors. Thus, early detection of interstitial injury and fibrosis, as well as direct DNA injury
MM-induced kidney disease is extremely important. and induction of apoptosis [83].
The IMWG defines renal injury as either an elevation of Once the PTC is injured, LCs overflow into the distal
serum creatinine >2 mg/dL or a reduced creatinine clearance lumen and interact with the Tamm-Horsfall protein (THP)
(CrCl) of <40 mL/min/1.73m2, due to MM [74]. CrCl should secreted by the thick ascending limb (TAL). This leads to
be assessed using the Modification of Diet in Renal Disease formation of the MM cast that is responsible for the presen-
(MDRD) formula or the Chronic Kidney Disease Epidemiology tation of MCN. The variability in cast formation by this
Collaboration (CKD-EPI) equation as these most accurately interaction can be explained by the variation in the
predict the clearance obtained from inulin-­based glomerular complementarity-­determining region 3 (CDR3) of different
filtration rate (GFR) estimation [75, 76]. The CKD-EPI group LCs [84]. While urinary light chains are required for the for-
further suggests that an equation based on creatinine and cys- mation of casts, the type or quantity of LC does not correlate
tatin C, which also reflects tumor burden, is more accurate, but with the severity of cast formation. Urinary LC quantity,
this remains to be validated in larger studies [75]. The use of however, is predictive of response to therapy and risk for
eGFR should be restricted to stable patients and not for those renal failure. Tubular solute composition and flow rates are
presenting with an acute kidney injury (AKI). RIFLE (Risk, other factors that also influence cast formation. Obstructed
Injury, Failure, Loss and End-Stage Kidney Disease) criteria tubules in turn induce an intense inflammatory response
and Acute Kidney Injury Network classification should be probably related to urine leak into the interstitium [85].
used for assessing the severity of AKI [77].

 ommon Patterns of Renal Insufficiency


C
I nitial Diagnostic Workup of Renal Injury in Myeloma
in Myeloma
Myeloma cast nephropathy (MCN) is the most common his-
All patients with symptomatic MM should have an initial tologic finding with autopsy studies reporting rates between
laboratory panel that includes serum creatinine, GFR estima- 30 and 50% of all MM patients with renal failure [86–88].
tion, electrolytes, serum free light chains (SFLC), and 24-h MCN almost always occurs in the presence of high free LC
31  Supportive Care in Multiple Myeloma 603

burden and typically when serum FLC levels are >100 mg/ bortezomib-­based regimen. A prior study examining the
dL [74, 89]. Almost all MM patients with significant renal use of HCO-HD in combination with chemotherapy in 67
impairment will also have high urine FLC levels [90]. The MM patients showed encouraging results, with a sustained
serum and urine FLC levels can be prognostic indicators in reduction in FLCs by day 12 in 67% of patients and dialysis
MCN [91, 92]. The hallmark of MCN is tubular obstruction independency in 63% [101]. However, sustained reduction
due to formation of LC casts, as described previously, from in FLC requires effective chemotherapy. The choice of regi-
the interaction of Tamm-Horsfall protein (THP) and mono- men is oftentimes guided by the presence of renal failure
clonal LCs. Factors that can influence formation of casts and nonrenally cleared drugs like bortezomib and thalido-
include urinary concentration of THP, sodium, calcium, pH, mide are preferred.
urine flow rate, and diuretic use [85].
MCN typically presents with acute renal failure; however,
oliguric AKI is a feature in about half of the cases even with  eneral Principles of Treatment Renal
G
very high levels of serum creatinine [93]. The progression of Insufficiency in Myeloma
AKI is rapid [94], with volume depletion and hypercalcemia
being the most common precipitating factors. Other common Initial treatment should include administration of intrave-
risk factors for AKI MM include use of IV contrast media, nous fluids to achieve high urine, reduction of calcium lev-
presence of infection, and nonsteroidal anti-inflammatory els in patients with hypercalcemia, treatment of infection,
drugs (NSAIDs) used for bone pain. One of the initial indica- and avoidance of nephrotoxic agents. However, there is no
tors for MCN is the presence of heavy proteinuria that is dis- established role for urine alkalization in treatment of RI
proportionate to the albuminuria detected on a urine dipstick. [102]. Bisphosphonates can be used for hypercalcemia;
The median amount of proteinuria is about 2 g/24 h and is however, pamidronate and zoledronic acid should be
almost exclusively Bence Jones protein with albuminuria avoided in patients with CrCl <30 mL/min. Pamidronate
contributing <10%. has been associated with development of collapsing variant
Early identification of MCN is essential for the treat- of focal and segmental glomerulosclerosis [103], whereas
ment of MCN since delay in decreasing light-chain burden zoledronic acid is associated with development of ATN
is associated with lower kidney recovery rates [92]. [104]. Denosumab may be helpful in this subgroup with
Treatment is focused on elimination of the precipitating close monitoring of serum calcium levels. The role of
agent and rapid reduction of the paraprotein. This can be mechanical treatments, which include therapeutic plasma
achieved with chemotherapy or extracorporeal removal via exchange (TPE) or high-cutoff hemodialysis (HCO-HD),
therapeutic plasma exchange (TPE). While renal replace- in treatment of MM is limited to MM cast nephropathy
ment therapy (RRT) may be required in certain cases, the (MCN) (see above).
indication for initiation of RRT is similar to other AKI Systemic chemotherapy should be started immediately
states. Role of TPE in the management of MCN and reduc- to reduce FLC burden and provides the best chance of renal
tion of FLC burden remains controversial. Three random- recovery, with bortezomib in combination with dexametha-
ized controlled trials have published conflicting results sone usually employed [72]. Bortezomib-based therapy has
[95–97]. The largest trial showed no benefit but was limited significantly higher rates of renal recovery (≥partial renal
by the absence of histologic diagnosis of MCN. A meta- response) compared to thalidomide- or lenalidomide-based
analysis of the three trials showed higher rates of dialysis regimens (77% vs. 55% vs. 43%, respectively) in a large
independence at 6 months for patients when TPE was com- retrospective analysis of 133 patients [105]. Carfilzomib, a
bined with chemotherapy but no change in overall survival second-generation proteasome inhibitor, has shown good
with use of TPE [98]. Arguments against using TPE include safety and efficacy when used in patients with RI, and
the absence of FLC monitoring in previous randomized tri- progression-­free survival was better with carfilzomib when
als and introduction of newer chemotherapeutic agents compared to bortezomib [106]. High-dose corticosteroids
with high response rates that did not significantly improve (equivalent to dexamethasone 160 mg or greater over 4
with addition of TPE [99, 100]. days) can be effective for rapid recovery of renal function
The role of high-cutoff hemodialysis (HCO-HD) in treat- regardless of the initial chemotherapy regimen [105]. High-
ment of patients with renal insufficiency is being evaluated dose steroids should be used in the initial month of therapy
in two large randomized controlled trials: EuLITE study for MM. Thalidomide can be used in renal insufficiency
(European Trial of Free Light Chain Removal by Extended without dose adjustment; however there have been reports
Hemodialysis in Cast Nephropathy; NCT00700531) of hyperkalemia in patients receiving dialysis [107, 108].
and the French MYRE study (Studies in Patients with The renal recovery rates with use of thalidomide have been
MM and Renal Failure Due To MM Cast Nephropathy; as high as 75% in newly diagnosed MM and up to 60% in
NCT01208818). Both these trials include patients on a patients with relapse/refractory disease [105, 107, 109].
604 S.M. Doshi et al.

Lenalidomide requires dose adjustment in renal insufficiency Conclusion


and response to treatment is not as robust. Autologous stem Thus, supportive care for MM patients becomes increas-
cell transplant (ASCT) can be used in MM even in presence ingly important, if MM patients are to enjoy a higher
of severe RI requiring dialysis but requires dose adjustment quality of life. MM patients are living longer, receiving
of the high-­dose melphalan [110]. RI at the time of trans- more intensive therapy, and are potentially curable.
plantation, however, is associated with higher risk of trans-
plant-related mortality (4% vs. <1%) [110, 111].
References

AL Amyloidosis 1. Ludwig H, Miguel JS, Dimopoulos MA, Palumbo A, Garcia Sanz R,
Powles R, et al. International Myeloma Working Group recommen-
dations for global myeloma care. Leukemia. 2014;28(5):981–92.
Amyloidosis represents the most common glomerular 2. ACS. 2016. http://www.cancer.org/cancer/multiplemyeloma/
lesion in MM. AL amyloidosis is a systemic disease with detailedguide/multiple-myeloma-key-statistics
deposition of congophilic fibrils in soft tissue. The fibrils 3. Greenberg AJ, Rajkumar SV, Therneau TM, Singh PP, Dispenzieri
A, Kumar SK. Relationship between initial clinical presentation and
are composed of light chains (AL), heavy chains (AH), or the molecular cytogenetic classification of myeloma. Leukemia.
intact immunoglobulins (ALH) [112]. AL amyloidosis is 2014;28(2):398–403.
the most common subtype accounting for >95% of cases. 4. Blade J, Fernandez-Llama P, Bosch F, Montoliu J, Lens XM,
Autopsy studies report rates of 5–15% for AL amyloido- Montoto S, et al. Renal failure in multiple myeloma: presenting
features and predictors of outcome in 94 patients from a single
sis [86–88]. MM is diagnosed in <10% of cases of AL institution. Arch Intern Med. 1998;158(17):1889–93.
amyloidosis. About 50% of patients present with renal 5. Dispenzieri A, Kyle RA. Neurological aspects of multiple
failure. Proteinuria is present in >70% of the cases while myeloma and related disorders. Best Pract Res Clin Haematol.
about a fourth of the cases present with nephrotic syn- 2005;18(4):673–88.
6. Palumbo A, Rajkumar SV, Dimopoulos MA, Richardson PG,
drome [113]. As opposed to MCN, the proteinuria is pre- San Miguel J, Barlogie B, et al. Prevention of thalidomide- and
dominantly albumin [78]. Rarely, amyloidosis will lenalidomide-associated thrombosis in myeloma. Leukemia.
present with renal dysfunction in the absence of protein- 2008;22(2):414–23.
uria as in vascular limited amyloidosis or diabetes insipi- 7. Melton LJ 3rd, Kyle RA, Achenbach SJ, Oberg AL, Rajkumar
SV. Fracture risk with multiple myeloma: a population-based study.
dus. Kidney involvement can at times be the only J Bone Miner Res. 2005;20(3):487–93.
presentation of amyloidosis. A fat pad biopsy can pro- 8. Saad F, Lipton A, Cook R, Chen YM, Smith M, Coleman
vide a diagnosis in about 70% of cases, whereas the R. Pathologic fractures correlate with reduced survival in patients
remaining will require kidney biopsy [78]. Kidney with malignant bone disease. Cancer. 2007;110(8):1860–7.
9. Kumar SK, Dispenzieri A, Lacy MQ, Gertz MA, Buadi FK, Pandey
response in amyloidosis is predictive of OS [114, 115]. S, et al. Continued improvement in survival in multiple myeloma:
Kidney transplantation can be considered in combination changes in early mortality and outcomes in older patients.
with chemotherapy and ASCT as one of the therapeutic Leukemia. 2014;28(5):1122–8.
options in patients with AL amyloidosis [116]. 10. Roodman GD. Pathogenesis of myeloma bone disease. Leukemia.
2009;23(3):435–41.
11. Cackowski FC, Anderson JL, Patrene KD, Choksi RJ, Shapiro SD,
Windle JJ, et al. Osteoclasts are important for bone angiogenesis.
Kidney Transplantation in Myeloma Blood. 2010;115(1):140–9.
12. Yaccoby S, Wezeman MJ, Henderson A, Cottler-Fox M,

Yi Q, Barlogie B, et al. Cancer and the microenvironment:
Kidney transplantation is challenging in patients with myeloma-osteoclast interactions as a model. Cancer Res.
MM since the transplantation and ensuing use of immuno- 2004;64(6):2016–23.
suppressive medications are thought to portend a high risk 13. Abe M, Hiura K, Wilde J, Shioyasono A, Moriyama K, Hashimoto
for disease recurrence and infections. Most centers require T, et al. Osteoclasts enhance myeloma cell growth and survival
via cell-cell contact: a vicious cycle between bone destruction and
a treatment-free remission period of 3–5 years. The initial myeloma expansion. Blood. 2004;104(8):2484–91.
lesion can be predictive of graft survival and recurrence. 14. Delgado-Calle J, Anderson J, Cregor MD, Hiasa M, Chirgwin
Patients with MCN have a lower risk of graft recurrence if JM, Carlesso N, et al. Bidirectional notch signaling and osteocyte-­
MM stays in remission [117]. In patients with AL amyloi- derived factors in the bone marrow microenvironment promote
tumor cell proliferation and bone destruction in multiple myeloma.
dosis, specifically those without significant cardiac Cancer Res. 2016;76(5):1089–100.
involvement, kidney transplantation in combination with 15. Alexandrakis MG, Roussou P, Pappa CA, Messaritakis I, Xekalou
chemotherapy and ASCT has shown favorable outcomes A, Goulidaki N, et al. Relationship between circulating BAFF
[116]. Renal transplantation in patients with MIDD is not serum levels with proliferating markers in patients with multiple
myeloma. Biomed Res Int. 2013;2013:389579.
recommended unless complete response is noted, as 16. An G, Acharya C, Feng X, Wen K, Zhong M, Zhang L, et al.
recurrence rates are about 80% in patients who do not Osteoclasts promote immune suppressive microenvironment in mul-
have a complete response to treatment [118]. tiple myeloma: therapeutic implication. Blood. 2016;128:1590–603.
31  Supportive Care in Multiple Myeloma 605

17. Lawson MA, McDonald MM, Kovacic N, Hua Khoo W, Terry RL, 35. Silbermann R, Roodman GD. Current controversies in the

Down J, et al. Osteoclasts control reactivation of dormant myeloma management of myeloma bone disease. J Cell Physiol.
cells by remodelling the endosteal niche. Nat Commun. 2015;6:8983. 2016;231(11):2374–9.
18. Croucher PI, De Hendrik R, Perry MJ, Hijzen A, Shipman CM, 36. Morgan GJ, Davies FE, Gregory WM, Szubert AJ, Bell SE,

Lippitt J, et al. Zoledronic acid treatment of 5T2MM-bearing mice Drayson MT, et al. Effects of induction and maintenance plus long-
inhibits the development of myeloma bone disease: evidence for term bisphosphonates on bone disease in patients with multiple
decreased osteolysis, tumor burden and angiogenesis, and increased myeloma: the Medical Research Council Myeloma IX Trial. Blood.
survival. J Bone Miner Res. 2003;18(3):482–92. 2012;119(23):5374–83.
19. Morgan GJ, Child JA, Gregory WM, Szubert AJ, Cocks K, Bell 37. Marx RE, Sawatari Y, Fortin M, Broumand V. Bisphosphonate-­
SE, et al. Effects of zoledronic acid versus clodronic acid on skel- induced exposed bone (osteonecrosis/osteopetrosis) of the jaws: risk
etal morbidity in patients with newly diagnosed multiple myeloma factors, recognition, prevention, and treatment. J Oral Maxillofac
(MRC Myeloma IX): secondary outcomes from a randomised con- Surg. 2005;63(11):1567–75.
trolled trial. Lancet Oncol. 2011;12(8):743–52. 38. Hellstein JW, Adler RA, Edwards B, Jacobsen PL, Kalmar JR,
20. Terpos E, Morgan G, Dimopoulos MA, Drake MT, Lentzsch S, Koka S, et al. Managing the care of patients receiving antiresorp-
Raje N, et al. International Myeloma Working Group recommenda- tive therapy for prevention and treatment of osteoporosis: execu-
tions for the treatment of multiple myeloma-related bone disease. J tive summary of recommendations from the American Dental
Clin Oncol. 2013;31(18):2347–57. Association Council on Scientific Affairs. J Am Dent Assoc.
21. Dimopoulos M, Terpos E, Comenzo RL, Tosi P, Beksac M, Sezer 2011;142(11):1243–51.
O, et al. International myeloma working group consensus statement 39. Patel CG, Yee AJ, Scullen TA, Nemani N, Santo L, Richardson
and guidelines regarding the current role of imaging techniques PG, et al. Biomarkers of bone remodeling in multiple myeloma
in the diagnosis and monitoring of multiple Myeloma. Leukemia. patients to tailor bisphosphonate therapy. Clin Cancer Res.
2009;23(9):1545–56. 2014;20(15):3955–61.
22.
Roodman GD. Osteoblast function in myeloma. Bone. 40. Coleman RE, Major P, Lipton A, Brown JE, Lee KA, Smith M,
2011;48(1):135–40. et al. Predictive value of bone resorption and formation markers in
23. D'Souza S, del Prete D, Jin S, Sun Q, Huston AJ, Kostov FE, et al. cancer patients with bone metastases receiving the bisphosphonate
Gfi1 expressed in bone marrow stromal cells is a novel osteoblast zoledronic acid. J Clin Oncol. 2005;23(22):4925–35.
suppressor in patients with multiple myeloma bone disease. Blood. 41. Badros A, Terpos E, Katodritou E, Goloubeva O, Kastritis E, Verrou
2011;118(26):6871–80. E, et al. Natural history of osteonecrosis of the jaw in patients with
24. Li X, Pennisi A, Yaccoby S. Role of decorin in the antimyeloma multiple myeloma. J Clin Oncol. 2008;26(36):5904–9.
effects of osteoblasts. Blood. 2008;112(1):159–68. 42. Gul G, Sendur MA, Aksoy S, Sever AR, Altundag K. A compre-
25. Rajkumar SV. Updated diagnostic criteria and staging system for mul- hensive review of denosumab for bone metastasis in patients with
tiple myeloma. Am Soc Clin Oncol Educ Book. 2016;35:e418–23. solid tumors. Curr Med Res Opin. 2016;32(1):133–45.
26. Wolf MB, Murray F, Kilk K, Hillengass J, Delorme S, Heiss C, 43. Raje N, Vadhan-Raj S, Willenbacher W, Terpos E, Hungria V,
et al. Sensitivity of whole-body CT and MRI versus projection radi- Spencer A, et al. Evaluating results from the multiple myeloma
ography in the detection of osteolyses in patients with monoclonal patient subset treated with denosumab or zoledronic acid in a ran-
plasma cell disease. Eur J Radiol. 2014;83(7):1222–30. domized phase 3 trial. Blood Cancer J. 2016;6:e378.
27. Regelink JC, Minnema MC, Terpos E, Kamphuis MH, Raijmakers 44. Iyer SP, Beck JT, Stewart AK, Shah J, Kelly KR, Isaacs R, et al. A
PG, Pieters-van den Bos IC, et al. Comparison of modern and con- Phase IB multicentre dose-determination study of BHQ880 in com-
ventional imaging techniques in establishing multiple myeloma-­ bination with anti-myeloma therapy and zoledronic acid in patients
related bone disease: a systematic review. Br J Haematol. with relapsed or refractory multiple myeloma and prior skeletal-
2013;162(1):50–61. related events. Br J Haematol. 2014;167(3):366–75.
28. Hillengass J, Fechtner K, Weber MA, Bauerle T, Ayyaz S, Heiss C, 45. Abdulkadyrov KM, Salogub GN, Khuazheva NK, Sherman ML,
et al. Prognostic significance of focal lesions in whole-body mag- Laadem A, Barger R, et al. Sotatercept in patients with osteolytic
netic resonance imaging in patients with asymptomatic multiple lesions of multiple myeloma. Br J Haematol. 2014;165(6):814–23.
myeloma. J Clin Oncol. 2010;28(9):1606–10. 46. Feurer E, Chapurlat R. Emerging drugs for osteoporosis. Expert
29. Kyle RA, Durie BG, Rajkumar SV, Landgren O, Blade J, Merlini Opin Emerg Drugs. 2014;19(3):385–95.
G, et al. Monoclonal gammopathy of undetermined significance 47. Roodman GD. Bone building with bortezomib. J Clin Invest.

(MGUS) and smoldering (asymptomatic) multiple myeloma: IMWG 2008;118(2):462–4.
consensus perspectives risk factors for progression and guidelines for 48. Mohty M, Malard F, Mohty B, Savani B, Moreau P, Terpos E. The
monitoring and management. Leukemia. 2010;24(6):1121–7. effects of bortezomib on bone disease in patients with multiple
30. Bauerle T, Hillengass J, Fechtner K, Zechmann CM, Grenacher L, myeloma. Cancer. 2014;120(5):618–23.
Moehler TM, et al. Multiple myeloma and monoclonal gammopa- 49. Cafforio P, Savonarola A, Stucci S, De Matteo M, Tucci M, Brunetti
thy of undetermined significance: importance of whole-body versus AE, et al. PTHrP produced by myeloma plasma cells regulates their
spinal MR imaging. Radiology. 2009;252(2):477–85. survival and pro-osteoclast activity for bone disease progression. J
31. Zamagni E, Tacchetti P, Terragna C, Cavo M. Multiple myeloma: Bone Miner Res. 2014;29(1):55–66.
disease response assessment. Expert Rev Hematol. 2016;9:831–7. 50. Zangari M, Berno T, Yang Y, Zeng M, Xu H, Pappas L, et al.
32. Lonial S, Kaufman JL. Non-secretory myeloma: a clinician’s guide. Parathyroid hormone receptor mediates the anti-myeloma effect of
Oncology (Williston Park). 2013;27(9):924–8. 30 proteasome inhibitors. Bone. 2014;61:39–43.
33. Berenson JR, Lichtenstein A, Porter L, Dimopoulos MA, Bordoni 51. Berenson J, Pflugmacher R, Jarzem P, Zonder J, Schechtman K,
R, George S, et al. Efficacy of pamidronate in reducing skeletal Tillman JB, et al. Balloon kyphoplasty versus non-surgical fracture
events in patients with advanced multiple myeloma. Myeloma management for treatment of painful vertebral body compression
Aredia Study Group. N Engl J Med. 1996;334(8):488–93. fractures in patients with cancer: a multicentre, randomised con-
34. Russell RG, Xia Z, Dunford JE, Oppermann U, Kwaasi A, Hulley trolled trial. Lancet Oncol. 2011;12(3):225–35.
PA, et al. Bisphosphonates: an update on mechanisms of action 52. Fotiou D, Gerotziafas G, Kastritis E, Dimopoulos MA, Terpos E. A
and how these relate to clinical efficacy. Ann N Y Acad Sci. review of the venous thrombotic issues associated with multiple
2007;1117:209–57. myeloma. Expert Rev Hematol. 2016;9(7):695–706.
606 S.M. Doshi et al.

53. Lipe B, Baker H, Weckbaugh B, Webb C, Brown A, Mahnken 71. Dupont SA, Dispenzieri A, Mauermann ML, Rabinstein AA, Brown
J, et al. Validation of a thrombosis risk assessment model in RD. Cerebral infarction in POEMS syndrome: incidence, risk fac-
patients with newly diagnosed multiple myeloma. ASCO Meeting tors, and imaging characteristics. Neurology. 2009;73(16):1308–12.
Abstracts. 2016;34(Suppl. 15):8055. 72. Dimopoulos MA, Terpos E, Chanan-Khan A, Leung N, Ludwig
54. Katodritou E, Verrou E, Hadjiaggelidou C, Gastari V, Laschos K, H, Jagannath S, et al. Renal impairment in patients with multiple
Kontovinis L, et al. Erythropoiesis-stimulating agents are associ- myeloma: a consensus statement on behalf of the International
ated with reduced survival in patients with multiple myeloma. Am Myeloma Working Group. J Clin Oncol. 2010;28(33):4976–84.
J Hematol. 2008;83(9):697–701. 73. Knudsen LM, Hjorth M, Hippe E. Renal failure in multiple

55. Mak KS, Lee LK, Mak RH, Wang S, Pile-Spellman J, Abrahm JL, myeloma: reversibility and impact on the prognosis. Nordic
et al. Incidence and treatment patterns in hospitalizations for malig- Myeloma Study Group. Eur J Haematol. 2000;65(3):175–81.
nant spinal cord compression in the United States, 1998–2006. Int J 74. Dimopoulos MA, Sonneveld P, Leung N, Merlini G, Ludwig H,
Radiat Oncol Biol Phys. 2011;80(3):824–31. Kastritis E, et al. International Myeloma Working Group recom-
56. Wight J, Stillwell A, Morris E, Grant B, Lai HC, Irving I. Screening mendations for the diagnosis and management of myeloma-­related
whole spine magnetic resonance imaging in multiple myeloma. renal impairment. J Clin Oncol. 2016;34(13):1544–57.
Intern Med J. 2015;45(7):762–5. 75. Inker LA, Schmid CH, Tighiouart H, Eckfeldt JH, Feldman HI,
57. Silberman J, Lonial S. Review of peripheral neuropathy in plasma Greene T, et al. Estimating glomerular filtration rate from serum
cell disorders. Hematol Oncol. 2008;26(2):55–65. creatinine and cystatin C. N Engl J Med. 2012;367(1):20–9.
58. Glasmacher A, Hahn C, Hoffmann F, Naumann R, Goldschmidt 76. Masson I, Flamant M, Maillard N, Rule AD, Vrtovsnik F, Peraldi
H, Lilienfeld-Toal M, et al. A systematic review of phase-II trials MN, et al. MDRD versus CKD-EPI equation to estimate glomeru-
of thalidomide monotherapy in patients with relapsed or refractory lar filtration rate in kidney transplant recipients. Transplantation.
multiple myeloma. Br J Haematol. 2006;132(5):584–93. 2013;95(10):1211–7.
59. Kocer B, Sucak G, Kuruoglu R, Aki Z, Haznedar R, Erdogmus 77. Srisawat N, Hoste EE, Kellum JA. Modern classification of acute
NI. Clinical and electrophysiological evaluation of patients kidney injury. Blood Purif. 2010;29(3):300–7.
with thalidomide-induced neuropathy. Acta Neurol Belg. 78. Leung N, Gertz M, Kyle RA, Fervenza FC, Irazabal MV, Eirin
2009;109(2):120–6. A, et al. Urinary albumin excretion patterns of patients with cast
60. Rajkumar SV, Rosinol L, Hussein M, Catalano J, Jedrzejczak W, nephropathy and other monoclonal gammopathy-related kidney
Lucy L, et al. Multicenter, randomized, double-blind, placebo-­ diseases. Clin J Am Soc Nephrol. 2012;7(12):1964–8.
controlled study of thalidomide plus dexamethasone compared 79. Dimopoulos MA, Terpos E. Renal insufficiency and failure.

with dexamethasone as initial therapy for newly diagnosed multiple Hematology Am Soc Hematol Educ Program. 2010;2010:431–6.
myeloma. J Clin Oncol. 2008;26(13):2171–7. 80. Merlini G, Palladini G. Differential diagnosis of monoclonal

61. Hulin C, Facon T, Rodon P, Pegourie B, Benboubker L, Doyen gammopathy of undetermined significance. Hematology Am Soc
C, et al. Efficacy of melphalan and prednisone plus thalidomide Hematol Educ Program. 2012;2012:595–603.
in patients older than 75 years with newly diagnosed multiple 81. Batuman V, Verroust PJ, Navar GL, Kaysen JH, Goda FO, Campbell
myeloma: IFM 01/01 trial. J Clin Oncol. 2009;27(22):3664–70. WC, et al. Myeloma light chains are ligands for cubilin (gp280).
62. Jongen JLM, Broijl A, Sonneveld P. Chemotherapy-induced periph- Am J Physiol. 1998;275(2 Pt 2):F246–54.
eral neuropathies in hematological malignancies. J Neurooncol. 82. Sengul S, Zwizinski C, Simon EE, Kapasi A, Singhal PC, Batuman
2015;121(2):229–37. V. Endocytosis of light chains induces cytokines through activa-
63. Dimopoulos MA, Mateos M-V, Richardson PG, Schlag R, Khuageva tion of NF-kappaB in human proximal tubule cells. Kidney Int.
NK, Shpilberg O, et al. Risk factors for, and reversibility of, periph- 2002;62(6):1977–88.
eral neuropathy associated with bortezomib-­melphalan-­prednisone 83. Pote A, Zwizinski C, Simon EE, Meleg-Smith S, Batuman

in newly diagnosed patients with multiple myeloma: subanalysis of V. Cytotoxicity of myeloma light chains in cultured human kidney
the phase 3 VISTA study. Eur J Haematol. 2010;86(1):23–31. proximal tubule cells. Am J Kidney Dis. 2000;36(4):735–44.
64. Corso A, Mangiacavalli S, Varettoni M, Pascutto C, Zappasodi 84. Ying WZ, Sanders PW. Mapping the binding domain of immu-
P, Lazzarino M. Bortezomib-induced peripheral neuropathy in noglobulin light chains for Tamm-Horsfall protein. Am J Pathol.
multiple myeloma: A comparison between previously treated and 2001;158(5):1859–66.
untreated patients. Leuk Res. 2010;34(4):471–4. 85. Hill GS, Morel-Maroger L, Mery JP, Brouet JC, Mignon F. Renal
65. Rampen AJ, Jongen JL, van Heuvel I, Scheltens-de Boer M,
lesions in multiple myeloma: their relationship to associated pro-
Sonneveld P, van den Bent MJ. Bortezomib-induced polyneuropa- tein abnormalities. Am J Kidney Dis. 1983;2(4):423–38.
thy. Neth J Med. 2013;71(3):128–33. 86. Ivanyi B. Frequency of light chain deposition nephropathy rela-
66. Richardson PG, Briemberg H, Jagannath S, Wen PY, Barlogie tive to renal amyloidosis and Bence Jones cast nephropathy in a
B, Berenson J, et al. Frequency, characteristics, and reversibility necropsy study of patients with myeloma. Arch Pathol Lab Med.
of peripheral neuropathy during treatment of advanced multiple 1990;114(9):986–7.
myeloma with bortezomib. J Clin Oncol. 2006;24(19):3113–20. 87. Kapadia SB. Multiple myeloma: a clinicopathologic study of 62
67. Ceresa C, Avan A, Giovannetti E, Geldof AA, Avan A, Cavaletti consecutively autopsied cases. Medicine. 1980;59(5):380–92.
G, et al. Characterization of and protection from neurotoxicity 88. Oshima K, Kanda Y, Nannya Y, Kaneko M, Hamaki T, Suguro M,
induced by oxaliplatin, bortezomib and epothilone-B. Anticancer et al. Clinical and pathologic findings in 52 consecutively autop-
Res. 2014;34(1):517–23. sied cases with multiple myeloma. Am J Hematol. 2001;67(1):1–5.

68. Dispenzieri A. POEMS syndrome: update on diagnosis, risk-­ 89. Hutchison CA, Bradwell AR, Cook M, Basnayake K, Basu S,
stratification, and management. Am J Hematol. 2015;90(10):951–62. Harding S, et al. Treatment of acute renal failure secondary to mul-
69. Nasu S, Misawa S, Sekiguchi Y, Shibuya K, Kanai K, Fujimaki Y, tiple myeloma with chemotherapy and extended high cut-off hemo-
et al. Different neurological and physiological profiles in POEMS dialysis. Clin J Am Soc Nephrol. 2009;4(4):745–54.
syndrome and chronic inflammatory demyelinating polyneuropa- 90. Drayson M, Begum G, Basu S, Makkuni S, Dunn J, Barth N, et al.
thy. J Neurol Neurosurg Psychiatry. 2012;83(5):476–9. Effects of paraprotein heavy and light chain types and free light
70. Cui R, Yu S, Huang X, Zhang J, Tian C, Pu C. Papilloedema is chain load on survival in myeloma: an analysis of patients receiving
an independent prognostic factor for POEMS syndrome. J Neurol. conventional-dose chemotherapy in Medical Research Council UK
2013;261(1):60–5. multiple myeloma trials. Blood. 2006;108(6):2013–9.
31  Supportive Care in Multiple Myeloma 607

91. Hutchison C, Sanders PW. Evolving strategies in the diagnosis, nosed symptomatic patients with multiple myeloma. Leukemia.
treatment, and monitoring of myeloma kidney. Adv Chronic 2013;27(2):423–9.
Kidney Dis. 2012;19(5):279–81. 106. Dimopoulos MA, Moreau P, Palumbo A, Joshua D, Pour L, Hajek
92. Hutchison CA, Cockwell P, Stringer S, Bradwell A, Cook M, R, et al. Carfilzomib and dexamethasone versus bortezomib and
Gertz MA, et al. Early reduction of serum-free light chains associ- dexamethasone for patients with relapsed or refractory multiple
ates with renal recovery in myeloma kidney. J Am Soc Nephrol. myeloma (ENDEAVOR): a randomised, phase 3, open-label, mul-
2011;22(6):1129–36. ticentre study. Lancet Oncol. 2016;17(1):27–38.
93. Rota S, Mougenot B, Baudouin B, De Meyer-Brasseur M, 107. Fakhouri F, Guerraoui H, Presne C, Peltier J, Delarue R, Muret P,
Lemaitre V, Michel C, et al. Multiple myeloma and severe renal et al. Thalidomide in patients with multiple myeloma and renal
failure: a clinicopathologic study of outcome and prognosis in 34 failure. Br J Haematol. 2004;125(1):96–7.
patients. Medicine. 1987;66(2):126–37. 108. Harris E, Behrens J, Samson D, Rahemtulla A, Russell NH, Byrne
94. Winearls CG. Acute myeloma kidney. Kidney Int. JL. Use of thalidomide in patients with myeloma and renal fail-
1995;48(4):1347–61. ure may be associated with unexplained hyperkalaemia. Br J
95. Clark WF. Correction: Plasma exchange when myeloma presents Haematol. 2003;122(1):160–1.
as acute renal failure. Ann Intern Med. 2007;146(6):471. 109. Tosi P, Zamagni E, Tacchetti P, Ceccolini M, Perrone G, Brioli
96. Johnson WJ, Kyle RA, Pineda AA, O'Brien PC, Holley A, et al. Thalidomide-dexamethasone as induction therapy before
KE. Treatment of renal failure associated with multiple myeloma. autologous stem cell transplantation in patients with newly diag-
Plasmapheresis, hemodialysis, and chemotherapy. Arch Intern nosed multiple myeloma and renal insufficiency. Biol Blood
Med. 1990;150(4):863–9. Marrow Transplant. 2010;16(8):1115–21.
97. Zucchelli P, Pasquali S, Cagnoli L, Ferrari G. Controlled plasma 110. San Miguel JF, Lahuerta JJ, Garcia-Sanz R, Alegre A, Blade J,
exchange trial in acute renal failure due to multiple myeloma. Martinez R, et al. Are myeloma patients with renal failure can-
Kidney Int. 1988;33(6):1175–80. didates for autologous stem cell transplantation? Hematol J.
98. Yu X, Gan L, Wang Z, Dong B, Chen X. Chemotherapy with 2000;1(1):28–36.
or without plasmapheresis in acute renal failure due to mul- 111. Lee CK, Zangari M, Barlogie B, Fassas A, van Rhee F, Thertulien
tiple myeloma: a meta-analysis. Int J Clin Pharmacol Ther. R, et al. Dialysis-dependent renal failure in patients with myeloma
2015;53(5):391–7. can be reversed by high-dose myeloablative therapy and auto-
99. Burnette BL, Leung N, Rajkumar SV. Renal improvement in transplant. Bone Marrow Transplant. 2004;33(8):823–8.
myeloma with bortezomib plus plasma exchange. N Engl J Med. 112. Leung N, Nasr SH, Sethi S. How I treat amyloidosis: the
2011;364(24):2365–6. importance of accurate diagnosis and amyloid typing. Blood.
100. Kastritis E, Anagnostopoulos A, Roussou M, Gika D, Matsouka 2012;120(16):3206–13.
C, Barmparousi D, et al. Reversibility of renal failure in newly 113. Kyle RA, Gertz MA. Primary systemic amyloidosis: clini-
diagnosed multiple myeloma patients treated with high dose cal and laboratory features in 474 cases. Semin Hematol.
dexamethasone-­ containing regimens and the impact of novel 1995;32(1):45–59.
agents. Haematologica. 2007;92(4):546–9. 114. Leung N, Dispenzieri A, Lacy MQ, Kumar SK, Hayman SR,
101. Hutchison CA, Heyne N, Airia P, Schindler R, Zickler D, Cook M, Fervenza FC, et al. Severity of baseline proteinuria predicts renal
et al. Immunoglobulin free light chain levels and recovery from response in immunoglobulin light chain-associated amyloidosis
myeloma kidney on treatment with chemotherapy and high cut-off after autologous stem cell transplantation. Clin J Am Soc Nephrol.
haemodialysis. Nephrol Dial Transplant. 2012;27(10):3823–8. 2007;2(3):440–4.
102. Analysis and management of renal failure in fourth MRC myelo- 115. Leung N, Glavey SV, Kumar S, Dispenzieri A, Buadi FK, Dingli
matosis trial. MRC working party on leukaemia in adults. Br Med D, et al. A detailed evaluation of the current renal response crite-
J (Clin Res Ed). 1984;288(6428):1411–6. ria in AL amyloidosis: is it time for a revision? Haematologica.
103. Markowitz GS, Appel GB, Fine PL, Fenves AZ, Loon NR, 2013;98(6):988–92.
Jagannath S, et al. Collapsing focal segmental glomerulosclero- 116. Herrmann SM, Gertz MA, Stegall MD, Dispenzieri A, Cosio FC,
sis following treatment with high-dose pamidronate. J Am Soc Kumar S, et al. Long-term outcomes of patients with light chain
Nephrol. 2001;12(6):1164–72. amyloidosis (AL) after renal transplantation with or without stem
104. Markowitz GS, Fine PL, Stack JI, Kunis CL, Radhakrishnan J, cell transplantation. Nephrol Dial Transplant. 2011;26(6):2032–6.
Palecki W, et al. Toxic acute tubular necrosis following treatment 117. Heher EC, Spitzer TR, Goes NB. Light chains: heavy burden in
with zoledronate (Zometa). Kidney Int. 2003;64(1):281–9. kidney transplantation. Transplantation. 2009;87(7):947–52.
105. Dimopoulos MA, Roussou M, Gkotzamanidou M, Nikitas 118. Leung N, Lager DJ, Gertz MA, Wilson K, Kanakiriya S, Fervenza
N, Psimenou E, Mparmparoussi D, et al. The role of novel FC. Long-term outcome of renal transplantation in light-chain
agents on the reversibility of renal impairment in newly diag- deposition disease. Am J Kidney Dis. 2004;43(1):147–53.

You might also like