You are on page 1of 20

Ann. N.Y. Acad. Sci.

ISSN 0077-8923

A N N A L S O F T H E N E W Y O R K A C A D E M Y O F SC I E N C E S
MARROW

Review

Multiple myeloma in the marrow: pathogenesis


and treatments
Heather Fairfield,1,∗ Carolyne Falank,1,∗ Lindsey Avery,2 and Michaela R. Reagan1,2
1 2
Maine Medical Center Research Institute, Scarborough, Maine. University of Maine, Orono, Maine

Address for correspondence: Michaela R. Reagan, 81 Research Drive, Scarborough, ME 04074. mreagan@mmc.org

Multiple myeloma (MM) is a B cell malignancy resulting in osteolytic lesions and fractures. In the disease state,
bone healing is limited owing to increased osteoclastic and decreased osteoblastic activity, as well as an MM-induced
forward-feedback cycle where bone-embedded growth factors further enhance tumor progression as bone is resorbed.
Recent work on somatic mutation in MM tumors has provided insight into cytogenetic changes associated with this
disease; the initiating driver mutations causing MM are diverse because of the complexity and multitude of mutations
inherent in MM tumor cells. This manuscript provides an overview of MM pathogenesis by summarizing cytogenic
changes related to oncogenes and tumor suppressors associated with MM, reviewing risk factors, and describing the
disease progression from monoclonal gammopathy of undetermined significance to overt MM. It also highlights the
importance of the bone marrow microenvironment (BMM) in the establishment and progression of MM, as well as
associated MM-induced bone disease, and the relationship of the bone marrow to current and future therapeutics.
This review highlights why understanding the basic biology of the healthy and diseased BMM is crucial in the quest
for better treatments and work toward a cure for genetically diverse diseases such as MM.

Keywords: multiple myeloma; bone marrow niche; treatments; bone marrow

Preferred citation:
Fairfield, H., C. Falank, L. Avery & M. R. Reagan. 2016. Multiple myeloma in the marrow: pathogenesis and treatments.
In “MARROW,” ed. by M. Zaidi. Ann. N.Y. Acad. Sci. 1364: 32–51.

Introduction a hallmark of MM is heterogeneous chromosomal


aberrations and numerous mutations in a range of
Multiple myeloma (MM) is a fatal, malignant B cell
genes, both of which make the disease very difficult
neoplasm characterized by uncontrolled, destruc-
to target therapeutically.2
tive growth of mutated plasma cells within the bone
MM begins as monoclonal gammopathy of unde-
marrow (BM). Patients over the age of 65 years are
termined significance (MGUS), progresses to smol-
most commonly affected by this disease1 and, as in-
dering (asymptomatic) myeloma, and finally be-
dicated by its name, MM is characterized by dissem-
comes overt (symptomatic) myeloma, resulting in
ination of multiple tumor cells throughout the BM.
BM infiltration and osteolytic lesions. During MM
A mnemonic sometimes used for the common MM
progression, the normal equilibrium between os-
pathologies is CRAB: C (calcium, elevated), R (renal
teoblastic (bone building) and osteoclastic (bone
failure), A (anemia), B (bone lesions). Additionally,
breakdown and resorption) activities is skewed to-
ward net bone loss. MM-induced osteolysis releases
growth factors embedded inside the bone matrix,

These authors contributed equally to the manuscript. fueling MM progression and expansion in the BM

doi: 10.1111/nyas.13038
32 Ann. N.Y. Acad. Sci. 1364 (2016) 32–51 
C 2016 New York Academy of Sciences.
Fairfield et al. Understanding the pathogenesis of multiple myeloma

niche and resulting in greater osteoclastic activity in patients have MGUS during the 2 years before they
a process known as the vicious cycle.3,4 During this develop MM, most patients have MGUS 4 years be-
MM-induced cycle, osteoclastic bone breakdown re- fore MM, and 82% of MM patients have MGUS 8
leases bone-embedded growth factors to promote years before MM develops.16 Other known risk fac-
tumor growth, which, in turn, stimulates greater tors for MM include being of male sex, older age, and
osteoclast (OC) activity. MM progression and drug African American racial background.17 However,
resistance depend on interactions with the local mi- as the number of MM cases continues to increase
croenvironment; therapies that target the marrow annually,1 stratification of secondary risk factor as-
microenvironment rather than the tumor cells di- sessment to encourage early diagnosis will become
rectly, such as bisphosphonates, have proven to be clinically significant. These secondary risk factors
effective in inhibiting tumor growth and osteolysis, include exposure to chronic, low-grade inflamma-
suggesting that targeting of the marrow can be an tion, preexisting and/or chronic immunodeficiency,
effective therapeutic avenue to pursue.5–10 In addi- and, potentially, the presence of known inflamma-
tion to osteoblasts (OBs) and OCs, other BM niche tory diseases or conditions (e.g., obesity, cardiovas-
cells, including mesenchymal stem cells (MSCs), cular disease, or type II diabetes mellitus).1,18–20 Of
BM adipocytes, immune cells, and osteocytes, also growing interest are the pathogenic links between
likely affect MM progression.7 BM cells exist in a MM and obesity-related chronic, low-grade inflam-
dynamic, stress-sensing environment where they re- mation, BM/tumor microenvironment hypoxia,
lease soluble signaling molecules to communicate and genetic instability.21–23 Research into how each
with one another; how these signals are processed of these components support malignant transition
by MM cells invading the BM niche remains a key is ongoing.24,25
area of investigation. Overt MM is often characterized by mutations in
Recent advances in genetic technologies have pro- KRAS (particularly in previously treated patients),
vided additional insight into mutations and chro- NRAS, BRAF, FAM46C, TP53, and DIS3. Often
mosomal abnormalities associated with MM.11 We mutations affecting proteins within the same
now know that the majority of MM cases are as- pathway are also observed (e.g., KRAS, NRAS, and
sociated with an initiating somatic mutation and BRAF).26 Common mutations include loss at 1p and
are frequently accompanied by additional oncogenic inactivation of the tumor suppressor p53, resulting
mutations. This genetic complexity makes the treat- in an abundance of immunoglobulin production.11
ment of MM problematic. Thus far, MM treatments Somatic mutations associated with the devel-
primarily rely on chemotherapies in conjunction opment of MM can be characterized as either
with proteasome inhibitors (PIs),12,13 antiresorptive non-hyperdiploid disease (NHD) or hyperdiploid
agents such as bisphosphonates,14 corticosteroids, disease (HD).27 Both classes of mutations involve
and BM transplantation.12–14 Immunostimulatory large chromosomal structural events that result in
agents are also being investigated as potential treat- the dysregulation of G1 /S cell cycle and cyclin D gene
ments for MM; however, a better understanding of transcription.11,28,29 NHD is characterized by chro-
the MM BM niche and cell–cell interactions is cru- mosomal translocations at 14q32, including t(4;14),
cial in providing novel, more effective treatments t(11;14), t(14;16), and t(14;20), which involve early
for MM patients. This review aims to describe the translocation of immunoglobulin heavy chain
nature of somatic mutations as they relate to MM (IgH) genes.30 This results in increased expression
disease prognosis. It also outlines the interactions of IgH in the clinical presentation of MM.11,28,29
of MM with the various cell types in the BM niche NHD is linked with an increased likelihood of dis-
and describes current treatments as they relate to ease progression and poor overall prognosis.11,28,31
microenvironmental components. By contrast, HD is characterized by the presence of
trisomies, specifically chromosomes 3, 5, 7, 9, 11,
Epidemiology, cytogenetics, and risk
15, 19, and 21. Excess cyclin D in the absence of ele-
stratification in MM
vated IgH is the main signature of HD.1 Additional
Over 22,000 new cases of MM were diagnosed in cytogenetic aberrations in MM patients with refrac-
2013, and the number of cases of MM increases tory disease include 13q deletion and 1q21 gains in
steadily each year.15 It appears that nearly all 47.4% and 52.2% of MM patients, respectively.29

Ann. N.Y. Acad. Sci. 1364 (2016) 32–51 


C 2016 New York Academy of Sciences. 33
Understanding the pathogenesis of multiple myeloma Fairfield et al.

MM may also be characterized through molec- “low-intermediate risk” (21%), and the absence
ular genetic (gene expression) profiling.2 High- of risk factors is considered “low risk” (5%).16
throughput sequencing of MM patient samples has Approximately 3% of the adult population over the
identified novel driver mutations beyond classical age of 50 is expected to have MGUS.16 However,
cancer mutations (NRAS, KRAS, and TP53). These owing to the absence of symptoms and the lack
include mutations in transcription factors BRAF, of standardized MGUS/MM screening procedures,
FAM46C, DIS3, XBP1, IRF4, and PRDM1, as well MGUS is often underdiagnosed. Progression
as various histone-modifying enzymes MLL, MLL2, of MGUS to MM is discussed in further detail
MLL3, UTX, MMSET (WHSC1), WHSC1L1, and throughout the subsequent sections.
recent inference toward HOXA-9.30 Whole genome
Pathogenesis of MM
sequencing of MM patient samples from the COS-
MIC database also identified mutations in AFF1, The progression of MM begins with the asymp-
c12orf42, CSMD3, LRRC4C, PCDH7, PTPRD, tomatic, precursor pathogenic state of MGUS.
PPFIBP1, RB1, and ZKSCAN3.30,32 Identification Evidence indicates that MGUS, previously char-
and investigation of these mutations may aid in the acterized by myeloma cell growth without bone
diagnosis of MM cases and potentially identify pa- destruction or other organ involvement, is in fact
tients who may be genetically predisposed or sus- associated with alterations in the bone. Epidemio-
ceptible to development of this disease. logic evidence has shown that patients with MGUS
MM clonality and tumor heterogeneity also limit suffer from a significantly increased fracture risk,
the identification of effective therapeutic drugs that and that the prevalence of MGUS is increased in
target this array of mutations. Secondary genetic patients with osteoporosis.36 Recent findings by
mutations are associated with, and thought to Drake36 have demonstrated that the onset of MGUS
determine, the longevity of mutated MM cells, is concurrent with the deterioration of both aux-
where cells with the greatest resilience and survival iliary and appendicular microarchitecture leading
likelihood are selectively expanded.2 Notably, intra- to skeletal fragility. The relationship between bone
clonal heterogeneity is considered to be nonuniform loss in MGUS and the progression to MM is an area
among individual cases as well as within MM cell of current interest, as a correlation may suggest
populations,2,5 presenting a particular clinical that treating bone loss in MGUS could not only
challenge in the context of chemosensitivity and decrease fracture risk in these patients, but could
drug resistance. The focus of a substantial amount also delay the onset of MM, a theory currently
of research effort is therefore aimed at identifying under investigation in our and others’ laboratories.
critical BM microenvironmental and systemic In the absence of clinical symptoms, MGUS
variables contributing to intraclonal, heterogeneic is diagnosed by quantifying the amount of im-
malignancy and subsequent refractory disease. munoglobulin present in both the bloodstream and
While the above mutations and phenotypic BM, specifically with a plasma cell population of
correlations have been identified as primary risk 10% in the BM. With a plasma cell content ex-
factors for MM, additional risk stratification has ceeding 10%, the disease state transitions into ei-
been implemented in populations with MGUS. Risk ther smoldering MM (SMM) or MM (if clinically
factors for the progression of MGUS to MM include manifested).16 MGUS progresses to MM at a rate
(1) increased serum myeloma protein (M-protein); of 1–2% of patients per year, and this transition
(2) type of M-protein present (e.g., IgG, IgM, is likely influenced by the presence of mutational
IgA);33 (3) abnormal ratio of free kappa to lambda diversity or clonality of MM cell populations as
light chain immunoglobulins (<0.26, >1.26);33 (4) well as changes in the local BM and other systemic
presence of >95% (clonal) plasma cells present in factors.17,31,37,38 MM cells are thought to initially
the BM;34 and (5) presence of circulating plasma create a plasmacytoma, a single tumor, and then de-
cells.16,35 In patients with MGUS, the presence velop into multiple lesions to form the disease of
of three or more risk factors is considered “high MM.39 Smoldering SMM is an intermediate clini-
risk” (58% 20-year progression within MGUS cal stage between MGUS and MM. It is classified as
patient population), two risk factors represent having high serum or urinary monoclonal protein
“high-intermediate risk” (37%), one risk factor is as well as clonal BM plasma cells in the range of

34 Ann. N.Y. Acad. Sci. 1364 (2016) 32–51 


C 2016 New York Academy of Sciences.
Fairfield et al. Understanding the pathogenesis of multiple myeloma

10–60%, in the absence of additional myeloma- mice,53 demonstrating the complexities of under-
defining events40 such as hypercalcemia, renal in- standing oncogenesis in the BMM. Additionally,
sufficiency, anemia, or bone lesions.41 other BM inflammatory components such as SDF1,
With the evasion techniques of clonal evolution a chemoattractant for C-X-C chemokine receptor
and drug resistance, MM may progress to an ag- type 4 (CXCR4), expressed on MM cells, are also
gressive, bone-marrow independent disease known known to promote MM tumor cell BM homing
as plasma cell leukemia (PCL).23,37 In PCL, MM and engraftment;23 increased expression of CXCR4
cells proliferate and spread into circulation, caus- in clonal cells supported epithelial-to-mesenchymal
ing an increase in plasma cells (20%) in the blood transition-like transcriptional patterns.23,54 A hy-
and often creating plasmacytomas at other places poxic microenvironment can further enhance the
outside the BM throughout the body.42 Efforts to genomic instability for cells within the BM, select
fully understand the pathogenesis of both precur- for dormant clones, and also contribute to MM cell
sor, symptomatic, and terminal-stage MM should drug resistance.23,55,56 Still, the effects of hypoxia
aim to identify mechanisms that cause somatic mu- on MM remain under investigation as novel model
tations, drug resistance, immune evasion, or relapse systems to measure or induce hypoxia and quantify
as potential drug targets. Treatments that interfere cellular responses in the BMM are being engineered
with MM growth and osteolysis in the bone mar- and optimized.
row microenvironment (BMM) to slow disease pro- The tumor microenvironment is ever evolving
gression and improve patient quality of life and life in response to changes in the molecular biochem-
expectancy are already in use with additional treat- istry, genetic profile, and diversity in type and num-
ments in development.43 ber of cell populations. Tumors and the surround-
ing microenvironment are thought to communi-
Pathogenesis of MM within the BMM
cate in a highly bidirectional, parasitic manner, each
The contributions of the BMM to MM are a focal with the potential to alter inherent characteristics
area of research. The BMM is a highly dynamic of the other.57 A typical MM tumor microenviron-
niche, capable of renewing damage and responding ment contains several cellular mediators/cell sub-
to systemic energy levels, inflammatory mediators, types (Fig. 1), including MM mesenchymal stromal
and endocrine signals.44–47 The BMM is a primary cells, OB and OC bone cells, BM adipocytes, and
modulator of both malignant transformation and a variety of immunomodulatory cell types (e.g.,
MM disease progression.6,48 Along with activated macrophages, natural killer (NK) cells, regulatory
inflammatory agents, reactive oxygen species (ROS) T cells, etc.). This type of diverse microenviron-
and reactive nitrogen intermediates are also present ment contributes to the dynamic characteristics of
in the hypoxic state within the BMM.49–51 Prop- healthy BMM as well as to the growth and survival
erties of the BMM allow for infiltration, growth, of a tumor.6 The roles of these cell types in MM
proliferation, adhesion, and migration of MM progression are outlined below.5
cells, while providing the structural and nutritional
sustenance to harbor quiescent, drug-resistant MM Mesenchymal stem cells
cells.2 In addition to providing an optimal substrate MSCs are of key importance in the BM as they serve
for MM initiation and progression, the BMM as the common progenitor to a variety of BMM
can also provide activated inflammatory agents, cells, including OBs, OCs, and adipocytes. MSCs
including cytokines, chemokines, adipokines (e.g., found in BM are a dynamic cell type with many
adiponectin and leptin), and growth factors (e.g., capabilities, including self-renewal, differentiation,
IL-6, IGF-1, VEGF, TNF-␣, and SDF-1) secreted cell signaling, injury- and tumor-homing, and
by macrophages, neutrophils, and other cells in immunomodulation.7,58 MSC function is highly in-
the BM. These factors then support malignant cell fluenced by the surrounding BMM and is thus
growth, drug resistance, and cytotoxicity of healthy altered in many disease states.6 MSCs from MM
cells.23,52 patients (MM-MSCs)7 interact with MM tumor
Interestingly, ROS formation via cells in the cell clusters6,59 and are altered by this interaction
BM niche has been implicated as a potential pri- but are not genetically compromised. MM-MSCs
mary, tumor-initiating event in leukemogenesis in have also been shown to produce exosomes that

Ann. N.Y. Acad. Sci. 1364 (2016) 32–51 


C 2016 New York Academy of Sciences. 35
Understanding the pathogenesis of multiple myeloma Fairfield et al.

Figure 1. The bone marrow microenvironment (BMM): osteoclastic and vascular niches. Schematic of the BMM and its role
in multiple myeloma. Pathogenesis and progression of multiple myeloma (MM) is carried out via pathological bone resorption,
resulting in the formation of lytic bone lesions and the degradation of bone density. Additionally, cells within the BMM may
contribute to MM cell migration, adhesion, quiescence, and tumor formation, which triggers the release of soluble growth factors,
collectively promoting invasion of the microvasculature by the MM tumor.

specifically contain proteins and microRNAs that bortezomib resistance in MM cells through secre-
promote tumor growth. The role of pharmacologi- tion of IL-8 and other factors that increase MM
cal treatments in targeting MM-MSCs is under cur- NF-␬B signaling.63 IL-6, IGF-1, VEGF, TNF-␣,
rent investigation, as previously reviewed.60 SDF-1, and BAFF constitute other stromal elements
Twenty years ago, it was first shown that MM- that promote the survival, migration, and drug resis-
MSCs were altered and “hijacked” by myeloma tance of MM cells.64 IL-6 is of particular importance,
cells to change their expression of certain cytokines as it acts both in paracrine and autocrine manners,65
(TGF-␤1, MIP-1␤, and IL-7);61 this led to a feed- and targeting MSC-derived IL-6 using siRNAs has
back loop enhancing tumor growth by inhibiting recently been proven effective preclinically to inhibit
pre-B cells to create a more immunosuppressed en- MM cell growth.66
vironment for MM cells to grow. Since then, many
abnormalities in MM-MSCs in relation to gene and Osteoblasts
microRNA expression have been found, some of OBs line the bone surface in basic multicellular
which enhance drug-resistance of MM cells in the units, separated from the BM by a layer of canopy
BMM or contribute to decreases in osteoblastic cells in what has been termed a “bone remodeling
activity.62 MM-MSCs have been shown to induce compartment” (in the trabeculae) or behind a

36 Ann. N.Y. Acad. Sci. 1364 (2016) 32–51 


C 2016 New York Academy of Sciences.
Fairfield et al. Understanding the pathogenesis of multiple myeloma

cutting zone led by OCs (in the cortex).67 Within dormant MM cells by remodeling the endosteal
this compartment, OBs produce and mineralize niche, releasing MM cells from their physical and
bone osteoid as OCs resorb bone.68 In healthy con- temporal hibernation.73 Moreover, OCs, as well
ditions, once activated by coupling factors released as stromal cells, have been shown to protect MM
during OC maturation and activity, OBs mineralize cells from dexamethasone-induced apoptosis.79
and rebuild healthy bone matrices.69 In MM, OB OCs also support angiogenesis through secretion
growth is significantly repressed, resulting in unbal- of proangiogenic factors;80 hence, targeting OCs
anced osteoclastic bone resorption and abnormal through bisphosphonates is efficacious through
bone turnover.4,6 The role of OBs in MM and MM numerous avenues.
drug resistance is still unclear, but OBs appear The RANK/RANKL/OPG signaling pathway,
to suppress MM growth or induce dormancy via deregulated in MM, is one of the important regula-
paracrine signaling.70–73 Decorin is one proteogly- tory pathways involved in maintaining the balance
can produced by OBs that has been demonstrated between bone remodeling and resorption.81–83 Re-
to directly inhibit growth and survival of MM ceptor activator of NF-␬B ligand (RANKL) is pro-
cells in vitro by inducing apoptosis and subsequent duced by OB-progenitor cells as well as MM cells,
arrest of MM cell cycle.6,74,75 By contrast, OBs may and signals by binding to its receptor on the surface
also promote growth and survival of MM cells of the OC-progenitors to stimulate OC differenti-
through the production of various growth factors ation. RANKL also binds osteoprotegerin (OPG),
or providing a niche to harbor quiescent MM which inhibits RANK/RANKL signaling and, in
cells, inducing chemotherapy resistance. Factors turn, osteoclastogenesis.74 In metastatic bone dis-
such as osteocalcin, osteopontin, FGF, and TGF-␤, ease, such as MM, this system is dysregulated,
which are synthesized and secreted by OBs, have leading to increased osteolysis and bone resorp-
been identified as factors that modulate the growth tion. MM cells directly stimulate osteoclastogene-
and survival of MM cells.6 Direct OB support, sis through their own production of RANKL but
inhibition, or effects on drug resistance of MM also indirectly manipulate this system by driving
cells may be dependent on the characteristics of the increased RANKL expression and decreased OPG
surrounding BMM. More remains to be examined in both BM stromal cells and OB.74 Denosumab,
in this realm, and three-dimensional models of a RANKL neutralizing antibody, was found to be
OBs and MM cells may help us explore these in- noninferior and trending toward superior versus
teractions in a more physiologically relevant model zoledronic acid (a bisphosphonate) in one random-
than possible in two-dimensional cocultures.6,70,74 ized, double-blind study in delaying time to first
on-study skeletal-related event in the treatment of
Osteoclasts bone metastases in patients with advanced cancer
OCs, the major target of bisphosphonate therapies, (excluding breast and prostate cancer) or MM.84
are the primary mediators of bone resorption in However, overall survival and disease progression
both healthy and pathological bone turnover.4,74 were similar between groups and hypocalcemia oc-
Clinically, the presence of lytic bone lesions in curred more frequently with denosumab. Another
MM manifests with severe bone pain, pathological clinical study, in the recruiting stage, will compare
fractures, increased bone turnover serum markers, denosumab to zoledronic acid in the treatment of
and hypercalcemia.6 In MM, OCs are increasingly bone disease in subjects with MM.
prominent in the BMM because of their activation
by various cytokines and signaling pathways.75–77 Osteocytes
With excessive bone resorption overpowering the A subset of OBs that slow in their building bone
protective “rebuilding” effects of OBs, the integrity activities become buried by their more active OB
of bone structure becomes compromised by the neighbors until they are fully embedded in bone
presence of lytic bone lesions. Pathological lysis matrix and differentiate into osteocytes. OCs sit
of bone tissue activates a vicious cycle, resulting within lacunae, tiny crevices within bone and make
in the ongoing release of calcium, growth factors, up 95% of bone cells.85 These cells regulate the
and ECM proteins, all of which enhance tumor response of bone to mechanical stress through the
growth and survival.78 OCs may also reactivate secretion of factors including sclerostin (SOST) and

Ann. N.Y. Acad. Sci. 1364 (2016) 32–51 


C 2016 New York Academy of Sciences. 37
Understanding the pathogenesis of multiple myeloma Fairfield et al.

RANKL into the BMM to modulate OB and OC pathways, including cyclic AMP-dependent protein
activity.86–88 Sclerostin, a Wnt inhibitor,89 decreases kinase A, signal transducer and signal activator 3,
OB differentiation from MSCs,90,91 and is also se- mitogen-activated protein kinase, ␤-catenin, and
creted from some MM cells to suppress OB dif- phosphatidylinositol 3-kinase.104 This results in the
ferentiation in vitro.92 Interestingly, OC number is antiproliferative and antitumorigenic effects seen
decreased and OC apoptosis is increased in MM,93 in MM, breast, prostate, colon, and liver epithelial
while circulating sclerostin levels are positively cor- cell cancers.18,104 From a mechanistic perspective,
related with bone disease severity in MM patients.94 deficiency in adiponectin (as seen in obesity)
Sclerostin is also upregulated in the BMM of MM limits the biological actions of several signaling
patients versus healthy people, and sclerostin is over- pathways that are essential to preventing growth,
expressed by plasma cells from MM patients.95,96 proliferation, migration, and drug resistance in
These findings implicate sclerostin as a novel tar- MM cells.22 Additionally, leptin may play a role
get corrupting the BMM, skewing the balance away in the transition from MGUS to MM through the
from osteoblastogenesis and toward osteolysis. regulation of the OPG/RANKL signaling pathway.
Elevated serum leptin levels have been documented
Adipocytes in MM patient populations when compared to
In addition to bone cells, adipocytes in the BMM healthy individuals.105,106 Together these results
also interact with MM cells. In the classical model suggest that adipocytes may support advancement
of lineage commitment, adipocytes arise from MSCs of MM in the BM niche; however, additional studies
and are the major cell type found in adipose tissue. are required to evaluate this hypothesis.
Lipid-laden adipocytes serve as an energy reserve
in the BM and also serve an endocrine function Immunomodulatory factors
through the expression and secretion of hormones In addition to OCs, other HSC-derived cells
known as adipokines (e.g., leptin and adiponectin) populate the BM niche, including tissue-specific
and growth factors (e.g., IL-6, TNF-␣, MCP-1, and macrophages known as osteomacs and other im-
insulin), which may promote myelomagenesis or mune cells.48 Osteomacs are responsible for the reg-
disease progression.18 Bone marrow adipose tissue ulation of HSCs in the BM niche by directing their
(BMAT) has been characterized as yellow adipose homing and colonization, as well as their transition
tissue, which has gene expression patterns that over- between active stem cell status and dormancy.107
lap with both white and brown fat.97 This sug- Other immune cells that populate the BM include
gests that BMAT can serve as an important reg- additional macrophages, neutrophils, and myeloid-
ulator of energy maintenance in the BM niche derived suppressor cells, all of which may respond to
during times of stress.97 Clinical studies have sug- stress, affect tumor growth, and affect bone turnover
gested an association between BMAT and an in- and other BM cells.108,109 Regulatory T cells in
creased likelihood of MM98,99 but the direct rela- the BM niche create an immune-privileged110 or
tionship and potential mechanisms remain largely immunosuppressive environment that can be cor-
unexplained. Adipocytes isolated from MM pa- rupted by the colonization of foreign entities includ-
tient femoral biopsies can support myeloma growth ing cancer cell types.48,109 Importantly, immune cells
in vitro,100 and a recent study suggests that they in the microenvironment can be modulated by the
may protect MM cells from chemotherapy-induced tumor to provide favorable conditions for MM tu-
apoptosis.101 mors. Specifically, myeloid-lineage progenitor cells
Obesity has been established as a physiological have been demonstrated to support tumor cells both
risk factor for MM development, and obese in vitro and in vivo.111,112 Immune cell populations
individuals have shown a 1.5–2-fold elevated risk are often skewed by MM cells, with increased reg-
for developing MM when compared to individuals ulatory T cells (immune-inhibiting) and decreased
of normal weight.18,22 Circulating adiponectin, activity and/or number of effector T cells (tumor
which is inversely proportional to fat mass,102 has killing) present in the BM.113 Upon tumor invasion,
been shown to inhibit proliferation of MM and mononuclear cells begin to produce and secrete
reduce tumorigenic angiogenesis.103,104 Inhibition VEGF, which stimulates mobilization of endothe-
via adiponectin occurs through several signaling lial cells and subsequent tumor vascularization.114

38 Ann. N.Y. Acad. Sci. 1364 (2016) 32–51 


C 2016 New York Academy of Sciences.
Fairfield et al. Understanding the pathogenesis of multiple myeloma

Endothelial cells treatment of overt MM, as patients with asymp-


The BMM is conducive to MM cell homing and tomatic, smoldering MM do not benefit from
proliferation, in part, because of unique BM en- chemotherapy.16,118 Chemotherapeutics are often
dothelial cells that express adhesion proteins that used in conjunction with ASCT, which is able to
specifically enable the rolling and intravasation of both replenish stem cells and potentially induce BM
MM cells through fenestrated BM capillaries.57 An- remodeling.119 This treatment combination has
giogenic cytokines and growth factors are primarily been shown to increase progression-free survival
modulated through BM stromal cells.6,54 Endothe- and overall survival in MM patients, and may
lial cells and endothelial progenitor cells, which are be used in the treatment of relapsed, refractory
associated with BM stromal cells, mobilize in re- MM.120,121 Additional combination treatments that
sponse to microenvironment stress, such as MM utilize the cancer targeting aspects of chemotherapy
invasion, to stimulate vasculogenesis of the BM drugs as well as agents that target and modulate
niche.115 In addition to stimulating VEGF secre- bone remodeling such as PIs and bisphosphonates
tion from mononuclear cells, tumor cells also se- are now used to treat MM. Specifically, doxorubicin
crete extremely high amounts of VEGF to stimu- has been combined with the PI, bortezomib, to treat
late the production and release of IL-6 from en- patients with refractory MM. This dual treatment
dothelial cells and BM stromal cells.114 This creates resulted in a greater myeloma-suppressive effect
a feedback stimulus of IL-6 signals from the endoge- than bortezomib alone.122,123 Combination thera-
nous endothelial cells to increase VEGF, indirectly pies utilizing chemotherapy and bisphosphonates
promoting vascularization of MM-infiltrated bone. have response rates as high as 80% in newly
Studies suggest that vasculogenesis as mediated by diagnosed patients124 and up to 75% in patients
endothelial and endothelial progenitor cells may be with relapsed disease.125
an early disease event that takes place during the
transition from either MGUS or smoldering MM to Proteasome inhibitors
overt MM.116 PIs such as bortezomib or the second-generation
carfilzomib block the proteasome, leading to the
Treatments accumulation of proteins in MM cells, causing
While MM remains incurable, new therapies have their destruction.126,127 Specifically, bortezomib
substantially improved patient quality of life and has been shown to promote apoptosis in MM cells
survival rates. Treatment of MM has substantially as well as OCs (attenuating bone loss) and induce
progressed in the last decade, with the development OB differentiation.78,128,129 Bortezomib has been
of novel pharmaceutical agents and the use of bis- shown to enhance the activity of certain concurrent
phosphonate drugs in conjunction with autologous therapies (e.g., lenalidomide and dexamethasone)
stem cell transplantation (ASCT). New approaches and is often prescribed in combination with other
have focused on the cotreatment of specific PIs anti-MM drugs owing to its chemosensitizing
with immunomodulatory drugs (IMiDs), the use of effects.130–136 However, there are clinical challenges
monoclonal antibodies, and histone deacetylase in- associated with bortezomib therapy, including drug
hibitors (HDACi). Nonetheless, chemotherapy and resistance and off-target effects including peripheral
steroids remain effective treatments for most MM neuropathy.137,138 Carfilzomib has been demon-
patients. Highlighted below and in Table 1 is a com- strated to also be effective when used with lenalido-
pilation of the most common treatments currently mide and dexamethasone by significantly reducing
used for MM. the minimal residual disease in patients who are
newly diagnosed with MM.139–141 Because of the
Chemotherapy effectiveness of bortezomib, next-generation PIs are
MM is often treated with classical chemotherapies currently being developed (e.g., oprozomib, ixa-
to target and destroy cancer cells.93 Chemother- zomib, marizomib, and delanzomib). Many of these
apeutics commonly used for MM include new PIs are being used after treatment with borte-
melphalan, vincristine, doxorubicin, and liposomal zomib, where MM progresses to bortezomib resis-
doxorubicin.117 These are used primarily in the tance or becomes relapsed and refractory MM.142

Ann. N.Y. Acad. Sci. 1364 (2016) 32–51 


C 2016 New York Academy of Sciences. 39
Understanding the pathogenesis of multiple myeloma Fairfield et al.

Table 1. Current treatments used to treat multiple myeloma


Clinical trial References and/or trial
Drug Type of drug phase identification numbers
BORT PI FDA approved
BORT/LEN/DEX PI/IMiD/steroid III NCI-2010-02211, IFM/DFCI 2009,
NCT01208662
BORT/LEN/DEX/LEN PI/IMiD/steroid/IMiD II Roussel et al.225
CARF PI FDA approved
CARF/LEN/DEX PI/IMiD/steroid III Stewart et al.226 ; E1A11,
NCI-2012-02608,
ECOG-E1A11, NCT01863550
CARF/POM/DEX PI/IMiD/steroid I/II AMyC 10-MM-01,
NCI-2012-01279,
IST-CAR-521,
PO-MM-PI-0034,
NCT01464034
THAL/DEX IMiD/steroid III MT2003-13, NCI-2010-01413,
0312M54569, 2004LS001,
NCT00293306,
UMN-2004LS001,
UMN-MT2003-13,
UMN-0312M54569,
NCT00177047
Oprozomib PI Preclinical Hurchla et al.227
Ixazomib/LEN/DEX PI/IMiD/steroid III NCI-2012-02752, 2011-005468-10,
C16011-CTIL, NL41603.028.12,
U1111-1164-7621,
NCT01659658
Delanzomib PI I Gallerani et al.228
Melphalan/THAL/PRED/DEF Chemo/IMiD/steroid/antic I/II Palumbo et al.229
Vincristine Chemo III Kyle et al.230
CYCLO/THAL/DEX Chemo/IMiD/steroid Garcı́a-Sanz et al.125
DOX/LEN/BORT Chemo/IMiD/PI I/II Jakubowiak et al.166
Liposomal DOX/DEX/LEN Chemo II Baz et al.231
Bendamustine Chemo I Michael et al.232
Etoposide Chemo Preclinical Wood et al.233
LEN IMiD FDA approved
POM IMiD FDA approved
POM/DEX IMiD/steroid III San Miguel et al.203
ELO IMiD FDA approved Starr234
LEN/BORT/DEX/ELO IMiD/PI/steroid/Ab I/II Usmani et al.235
Daratumumab Ab FDA approved
BI-505 Ab I/II Hansson et al.236
Rituximab Ab II Baz et al.,237 Bergua et al.,238 Korte
et al.239
SAHA HDACi I Mitsiades et al.,240
Richardson et al.241
LBH589 HDACi III San-Miguel et al.205
AR-42 HDACi Preclinical Canella et al.198
Vorinostat HDACi I Vesole et al.242
Ricolinostat (ACY-1215) HDACi Ib/II Yee et al.243
BORT, bortezomib; LEN, lenalidomide; DEX, dexamethasone; CARF, carfilzomib; POM, pomalidomide; ELO,
elotuzumab; SAHA, suberoylanilide hydroxamic acid; Ab, antibody; PI, proteasome inhibitor; IMiD, immunomod-
ulatory drug; HDACi, histone deacetylase inhibitor; Chemo, chemotherapy; CYCLO, cyclophosphamide; DOX, dox-
orubicin; PRED, prednisone; DEF, defibrotide; antic, anticoagulant; THAL, thalidomide.

40 Ann. N.Y. Acad. Sci. 1364 (2016) 32–51 


C 2016 New York Academy of Sciences.
Fairfield et al. Understanding the pathogenesis of multiple myeloma

Bisphosphonates combined with low-dose dexamethasone to be safe


Bisphosphonates are a class of drugs that inhibit re- and effective in patients with relapsed MM disease,
sorption of bone by blocking OCs and decrease os- who had previously been treated with bortezomib
teolytic bone pain.143,144 It is well known that MM or thalidomide.167 Richardson et al.163 have re-
is able to increase the number of OCs within the cently published phase II clinical trial outcomes
BMM,145,146 leading to bone pain, hypercalcemia, exploring the maximum tolerated dose (MTD) of
and increased bone fractures in patients.147–149 lenalidomide in patients with relapsed or refractory
Therefore, treatment with bisphosphonates is com- MM disease. Findings from this investigation
monly used in conjunction with PI drugs150–152 or revealed that the MTD of lenalidomide (15 mg/day)
chemotherapy to kill tumor cells and inhibit bone administered alongside low-dose dexamethasone
destruction.153,154 Several studies suggest that bis- (40 mg/week) on the first 21 of 28 total days of treat-
phosphonates may not have direct antitumor effects, ment was tolerable and resulted in favorable results
despite their bone-protective effects.155–157 Specif- in response and 2-year survival rates.163 Thalido-
ically, nitrogen-containing bisphosphonates alen- mide, the first-generation IMiD, is still commonly
dronate, pamidronate, and zoledronate are all U.S. used for managing MM by inhibiting angiogenesis,
Food and Drug Administration (FDA) approved inducing apoptosis of MM cells, and inhibiting
and are often administered as a crucial aspect of the secretion of IL-6 and VEGF by MM cells
MM patient pain and bone disease management.158 during adhesion to BM stromal cells.168 Like other
IMiDs, thalidomide is currently being used with
IMiDs and monoclonal antibodies PIs (bortezomib or carfilzomib) and corticosteroids
Endogenous immunomodulatory actions and/or (e.g., dexamethasone, prednisone, prednisolone,
immunodeficiency can significantly affect the and methylprednisolone).169–173 Developments in
dynamics of the BMM to either promote or MM drug therapies will likely continue to maximize
suppress MM tumor cell growth.23,113 Recent drug selectivity through antibody-mediated mechanisms
therapies for many cancer types, including MM, combined with PIs, IMiDs, bisphosphonates, and
continue to support a paradigm shift in using an traditional chemotherapy.115,159
immunomodulatory approach for the management Monoclonal antibodies are increasingly being uti-
and treatment of malignant disease. As recently lized to treat myeloma patients, both to target the
reviewed by Kawano et al.,113 MM tumor cells myeloma cells directly and to modulate the im-
can induce pronounced immunosuppression mune response and microenvironment.174 Recent
both systemically as well as locally at the BMM. work with daratumumab, a human monoclonal
Immunodeficiency in the BMM is characterized by antibody that binds CD38,175 has shown target-
an increase in immunosuppressive cell populations, cell killing of tumor cells.176 CD38 is a trans-
including regulatory T cells and myeloid derived membrane glycoprotein that is highly expressed by
suppressor cells (MDSCs).159 In the pathogenesis myeloma cells, making this a desirable, myeloma-
of MM, the ability of MDSCs to differentiate into specific target.177 Importantly, single-agent dara-
immune-protective macrophages and granulocytes tumumab treatment, which has recently been ap-
is inhibited.113,160 Thus, restoration of tumor- proved by the FDA, was effective in patients with re-
suppressor immunomodulation is a crucial compo- fractory disease, including those who had previously
nent in the development of anti-MM drug therapies. been treated with lenalidomide and bortezomib.176
Lenalidomide and pomalidomide, second- Radioimmunotherapy (RIT), which combines radi-
generation IMiDs, enhance the sensitivity of MM ation with immunotherapy, is in preclinical stages
cells to both bortezomib and dexamethasone. as a treatment for MM. Perhaps the most promis-
They also promote the tumor suppressor actions ing RIT in respect to MM treatments is the use
of host immune defense mechanisms.161 For this of indatuximab ravtansine, an antibody targeting
reason, multiple combination therapies are often CD138 antigen, which is expressed in greater than
utilized in clinical practice to improve treatment 95% of MM cells.178,179 Recent work has shown
response by delaying progression or recurrence that coupling this antibody to radioactive isotopes
and improving symptom management.162–166 significantly increased survival in animal models
A phase III trial found pomalidomide therapy of MM.179 The targeting of antigens and other

Ann. N.Y. Acad. Sci. 1364 (2016) 32–51 


C 2016 New York Academy of Sciences. 41
Understanding the pathogenesis of multiple myeloma Fairfield et al.

cell-surface proteins in the treatment of cancers antibodies in the treatment of myeloma, please refer
is rapidly expanding through the use of antibody- to the review by Sondergeld et al.195
drug conjugates, which combine the specificity of
a monoclonal antibody with a small anticancer Histone deacetylase inhibitors
drug molecule. Preclinical in vitro and in vivo work Epigenetic modifications have been shown to play
with these conjugates has demonstrated promise a major role in cancer development resulting in
in the treatment of MM and other hematological aberrant histone modifications.196 Histone acety-
cancers.180 lation is a major regulatory mechanism of gene
Another promising new approach to MM treat- transcription, where histone deacetylases remove
ment is the addition of an immunostimula- acetyl groups from the histone lysine residues,
tory agent, elotuzumab, to the now standard resulting in repressed transcription.197 HDACi
treatment of lenalidomide and dexamethasone have been shown to inhibit cell growth and induce
therapy.1,30 Elotuzumab, an FDA-approved human- apoptosis in MM cells as both a single treatment or
ized immunoglobulin G1 monoclonal antibody, tar- in conjunction with bortezomib treatment.198,199
gets signaling lymphocytic activation molecule F7 Furthermore, HDACi are able to overcome the
(SLAMF7).181 SLAMF7, expressed by both NK and bortezomib resistance that is commonly seen in
MM cells, plays an integral role in the activation of MM cells.200 There are several HDACi that are cur-
NK cells through binding with the EAT-2 adapter rently under evaluation.201 The first FDA-approved
protein (EAT-2 is now known as SH2 domain con- HDACi was suberoylanilide hydroxamic acid, which
taining 1B (SH2D1B)).159,182 In MM cells, EAT-2 is commonly used in combination with bortezomib
is significantly underexpressed, resulting in poor or lenalidomide.202–204 MM treatment with the
SLAMF7 signaling. Elotuzumab is able to reduce HDACi vorinostat or panobinostat, with borte-
tumor burden through the activation of NK cells zomib or bortezomib/dexamethasone, has been
that induce antibody-mediated apoptosis of MM shown to prolong MM patient survival for 0.8 and
cells, with little effect on healthy cells.159,182 3.9 months, respectively.202,205 Yet, further studies
In addition to bisphosphonates, other bone an- are needed in order to fully elucidate the epigenetic
abolic and antiresorption agents such as anti-DKK1, modifications and interactions in MM and the role
anti-sclerostin, and anti-activin A antibodies are of epigenetics in MM progression and pathogenesis.
being explored for stimulating bone healing and
Future directions and conclusions
regrowth in MM patients, and may also have anti-
tumor effects.92,94–96,183–190 Specifically, antibodies The pathogenesis of MM is influenced by alter-
used to target and suppress DKK1 in preclinical ations, aberrations, and/or dysregulation in en-
trials have been shown to prevent the development docrine, vascular, genetic, and metabolic factors.
of MM-induced osteolysis and increased bone Thus, identifying and targeting the corresponding
formation in animal models.191 SOST-neutralizing molecular pathways are crucial in working toward
antibodies (anti-SOST), which have been shown the optimal delivery of individualized pharmaco-
to increase bone formation and bone volume in logic therapies in MM. Increases or excess in adipos-
both experimental models of, and patients with, ity may contribute to MM risk via systemic proin-
osteoporosis, are also under development as a po- flammatory effects or BM-specific adiposity; this re-
tential treatment for MM.192,193 Two recent studies quires further investigation in patient populations
have indicated the efficacy of anti-SOST antibody as well as in both in vitro and in vivo models. MM
to prevent MM-induced bone loss and inhibit MM is characterized by the presence of osteolytic bone
growth in mouse myeloma models.190,194 lesions, resulting from prolonged declines in OB-
The use of monoclonal antibodies and their ef- mediated bone mineralization, increases in bone
fectiveness to treat MM both in early experimental resorption by OCs, and propagation of the vicious
models and in clinical trials has demonstrated the cycle.206 While this review has focused mainly on the
importance of understanding the molecular mech- effects of the BM niche on MM pathogenesis, neu-
anisms of MM progression, including MM interac- roendocrine regulation of bone metabolism could
tion with other cell types within the BM niche. For also play a role in MM progression. For example,
more information on the potential of monoclonal sympathetic nervous system (SNS) fibers, known to

42 Ann. N.Y. Acad. Sci. 1364 (2016) 32–51 


C 2016 New York Academy of Sciences.
Fairfield et al. Understanding the pathogenesis of multiple myeloma

innervate cortical bone, modulate a number of out- owing to challenges in proving effectiveness. The in-
comes, including regulating homeostatic hormonal tegration of multisystem regulatory factors through
control of bone turnover.206 Please refer to the the use of better disease models is crucial for
elegant review by Elefteriou et al.207 for more infor- continued understanding of MM pathogenesis for
mation on the effects of the SNS on bone remodeling better prevention, as well as treatment approaches.
in response to many factors including oncogenesis. In the meantime, more thorough characterization
The complex regulation of MM from multiple sys- of patient tumors through genetic and epigenetic se-
tems (the BMM and beyond) makes understanding quencing, and other profiling techniques, as well as
and combatting disease initiation and progression increased sharing of patient data among centers, will
difficult. Integration of these effects is necessary to provide critical information about potential causa-
provide better therapeutics and work toward a cure. tions linking systemic health, genetics, epigenetics,
Myeloma is now being treated by therapies aimed and environment, to cancers including myeloma.
at either directly killing tumor cells or normalizing Last, we suggest one of the greatest strategies in
the aberrant tumor microenvironment. Treatments accelerating effective myeloma research may be a
such as bisphosphonates, which inhibit osteolysis, collaborative, transparent “precompetitive” space
and IMiDs that stimulate immune cell activation for researchers and pharmaceutical companies alike.
have been implemented with success. As we gain We envision that transparent sharing of data and
better understanding of the bone, BM, and blood results will push the myeloma research field forward
biology disrupted by MM, we are developing safer, in a novel, patient-centric manner toward a cure,
more efficacious treatment strategies. Exciting, or, more realistically, patient-specific cures. Such a
novel strategies to inhibit the vicious cycle and space and cooperative agreement would optimize
osteolysis and heal osteolytic lesions on the basis of our global scientific journey toward a greater under-
a better understanding of the basic biology of the standing of this and other diseases, and accelerate
disease and the BMM are in development.6,208–210 research and discoveries relevant for those who rely
These microenvironment-targeting treatments, in on rapid development of effective therapies.
conjunction with ASCT, will induce better patient
Acknowledgments
outcomes, and are leading the way in the treatment
of relapsed, refractory MM.121 Interesting new di- The authors thank Dr. Michael Erard, scientific edi-
rections may target OCs and OBs, as we learn more tor and writing consultant, at Maine Medical Center
about the roles of these and other cells in the BM. Research Institute for editorial assistance. The au-
Novel strategies are also being explored in the thors’ work is supported by Start-up funds, a pilot
preclinical setting targeting NF-␬B, JAK, MEK, Pim, project Grant from NIH/NIGMS (P30GM106391),
PYK2, Src, mTor, PDPK1/RSK2, PERK, and other and the NIH/NIDDK (R24 DK092759-01) at Maine
kinases and pathways.211–221 Targeting hypoxia (e.g., Medical Center Research Institute. The authors’
HIF-1␣) and double-strand break repairs are other work is also supported by the Department of
avenues being explored. Moreover, many novel Defense (DoD) Visionary Postdoctoral Fellowship
strategies are being employed using chimeric anti- Award, through the Peer Reviewed Cancer Re-
gen receptor T cells222–224 and other engineered im- search Program, under Award FY14 DoD Congres-
mune responses against MM in preclinical models sionally Directed Medical Research Programs 30
that may soon make their way to the clinic. Combi- (No. W81XWH-13-1-0390). Opinions, interpreta-
natorial strategies, as with all cancers, are inevitably tions, conclusions, and recommendations are those
required for the most efficacious treatments. More of the authors and are not necessarily endorsed by
targetable and prognostically significant genomic the DoD.
alterations are continually arising as our bioinfor-
matics, sequencing, and preclinical validation ca- Conflicts of interest
pacities grow, and high-throughput drug screens The authors declare no conflicts of interests.
are enabling more rapid, accurate identification of
novel natural and synthetic anti-MM therapies. References
A crucial aspect in the fight against MM and 1. Alexander, D.D., P.J. Mink, H.O. Adami, et al. 2007. Multi-
other cancers is the prevention of the disease, an ple myeloma: a review of the epidemiologic literature. Int.
issue arguably underrepresented in cancer research J. Cancer 120: 40–61.

Ann. N.Y. Acad. Sci. 1364 (2016) 32–51 


C 2016 New York Academy of Sciences. 43
Understanding the pathogenesis of multiple myeloma Fairfield et al.

2. Morgan, G.J., B.A. Walker & F.E. Davies. 2012. The genetic 22. Greenfield, D.M., E. Boland, Y. Ezaydi, et al. 2014. En-
architecture of multiple myeloma. Nat. Rev. Cancer 12: docrine, metabolic, nutritional and body composition ab-
335–348. normalities are common in advanced intensively-treated
3. Kingsley, L.A., P.G.J. Fournier, J.M. Chirgwin & T.A. Guise. (transplanted) multiple myeloma. Bone Marrow Trans-
2007. Molecular biology of bone metastasis. Mol. Cancer plant. 49: 907–912.
Ther. 6: 2609–2617. 23. Grivennikov, S.I., F.R. Greten & M. Karin. 2010. Immunity,
4. Fowler, J.A., C.M. Edwards & P.I. Croucher. 2011. Tumor– inflammation, and cancer. Cell 140: 883–899.
host cell interactions in the bone disease of myeloma. Bone 24. Azab, A.K., J. Hu, P. Quang, et al. 2012. Hypoxia promotes
48: 121–128. dissemination of multiple myeloma through acquisition of
5. Roccaro, A.M., A. Sacco, P. Maiso, et al. 2013. BM mes- epithelial to mesenchymal transition-like features. Blood
enchymal stromal cell-derived exosomes facilitate multiple 119: 5782–5794.
myeloma progression. J. Clin. Invest. 123: 1542–1555. 25. Lwin, S.T., S.W.Z. Olechnowicz, J.A. Fowler & C.M.
6. Reagan, M.R., L. Liaw, C.J. Rosen & I.M. Ghobrial. 2015. Edwards 2015. Diet-induced obesity promotes a myeloma-
Dynamic interplay between bone and multiple myeloma: like condition in vivo. Leukemia 29: 507–510.
emerging roles of the osteoblast. Bone 75: 161–169. 26. Lohr, J.G., P. Stojanov, S.L. Carter, et al. 2014. Widespread
7. Reagan, M.R. & I.M. Ghobrial. 2012. Multiple myeloma genetic heterogeneity in multiple myeloma: implications
mesenchymal stem cells: characterization, origin, and for targeted therapy. Cancer Cell 25: 91–101.
tumor-promoting effects. Clin. Cancer Res. 18: 342–349. 27. Chng, W.J., R. Santana-Dávila, S.A. Van Wier, et al.
8. Alegre, A., M. Gironella, A. Bailén & P. Giraldo. 2014. Zole- 2006. Prognostic factors for hyperdiploid-myeloma: ef-
dronic acid in the management of bone disease as a con- fects of chromosome 13 deletions and IgH translocations.
sequence of multiple myeloma: a review. Eur. J. Haematol. Leukemia 20: 807–813.
92: 181–188. 28. Chubb, D., N. Weinhold, P. Broderick, et al. 2013. Com-
9. Pozzi, S. & N. Raje. 2011. The role of bisphosphonates mon variation at 3q26.2, 6p21.33, 17p11.2 and 22q13.1
in multiple myeloma: mechanisms, side effects, and the influences multiple myeloma risk. Nat. Genet. 45: 1221–
future. Oncologist 16: 651–662. 1225.
10. Guise, Ta., K.S. Mohammad, G. Clines, et al. 2006. Ba- 29. Walker, B.A., P.E. Leone, L. Chiecchio, et al. 2010. A com-
sic mechanisms responsible for osteolytic and osteoblastic pendium of myeloma associated chromosomal copy num-
bone metastases. Clin. Cancer Res. 12: 6213–6217. ber abnormalities and their prognostic value. Blood 116:
11. Furukawa, Y. & J. Kikuchi. 2015. Molecular pathogenesis 56–65.
of multiple myeloma. Int. J. Clin. Oncol. 20: 413–422. 30. Bergsagel, P.L., W.M. Kuehl, M. Ferrarini, et al. 2013. De-
12. Bergsagel, D.E. 1971. Total body irradiation for myelomato- coding the pathophysiology and the genetics of multiple
sis. Br. Med. J. 2: 325. myeloma to identify new therapeutic targets. J. Clin. Oncol.
13. Swami, A., M.R. Reagan, P. Basto, et al. 2014. Engineered 23: 2997.
nanomedicine for myeloma and bone microenvironment 31. Pawlyn, C., L. Melchor, A. Murison, et al. 2015. Coexis-
targeting. Proc. Natl. Acad. Sci. U.S.A. 111: 10287–10292. tent hyperdiploidy does not abrogate poor prognosis in
14. Schmiedel, B.J., C.A. Scheible, T. Nuebling, et al. 2013. myeloma with adverse cytogenetics and may precede IGH
RANKL expression, function, and therapeutic targeting translocations. Blood 125: 831–841.
in multiple myeloma and chronic lymphocytic leukemia. 32. Amend, S.R., W.C. Wilson, L. Chu, et al. 2015.
Cancer Res. 73: 683–694. Whole genome sequence of multiple myeloma-prone
15. Siegel, R., D. Naishadham & A. Jemal. 2013. Cancer statis- C57BL/KaLwRij mouse strain suggests the origin of dis-
tics, 2013. CA Cancer J. Clin. 63: 11–30. ease involves multiple cell types. PLoS One 10: e0127828.
16. Berenson, J.R., K.C. Anderson, R.A. Audell, et al. 2010. 33. Rajkumar, S.V., R.A. Kyle, T.M. Therneau, et al. 2005.
Monoclonal gammopathy of undetermined significance: a Serum free light chain ratio is an independent risk factor for
consensus statement: guideline. Br. J. Haematol. 150: 28– progression in monoclonal gammopathy of undetermined
38. significance. Blood 106: 812–817.
17. Jemal, A., R. Siegel, E. Ward, et al. 2009. Cancer statistics, 34. Pérez-Persona, E., M.B. Vidriales, G. Mateo, et al. 2007. New
2009. CA Cancer J Clin. 59: 225–249. criteria to identify risk of progression in monoclonal gam-
18. Medina, E.A., K. Oberheu, S.R. Polusani, et al. 2014. mopathy of uncertain significance and smoldering mul-
PKA/AMPK signaling in relation to adiponectin’s antipro- tiple myeloma based on multiparameter flow cytometry
liferative effect on multiple myeloma cells. Leukemia 28: analysis of bone marrow plasma cells. Blood 110: 2586–
2080–2089. 2592.
19. Blair, C.K., J.R. Cerhan, A.R. Folsom & J.A. Ross. 2005. An- 35. Kumar, S., S.V. Rajkumar, R.A. Kyle, et al. 2005. Prognos-
thropometric characteristics and risk of multiple myeloma. tic value of circulating plasma cells in monoclonal gam-
Epidemiology 16: 691–694. mopathy of undetermined significance. J. Clin. Oncol. 23:
20. Renehan, A.G., M. Tyson, M. Egger, et al. 2008. Body-mass 5668–5674.
index and incidence of cancer: a systematic review and 36. Drake, M.T. 2014. Unveiling skeletal fragility in patients
meta-analysis of prospective observational studies. Lancet diagnosed with MGUS: no longer a condition of undeter-
371: 569–578. mined significance? J. Bone Miner. Res. 29: 2529–2533.
21. De Pergola, G. & F. Silvestris. 2013. Obesity as a major risk 37. Barlogie, B., J. Shaughnessy, G. Tricot, et al. 2004. Treatment
factor for cancer. J. Obes. 2013: 291546. of multiple myeloma. Blood 103: 20–32.

44 Ann. N.Y. Acad. Sci. 1364 (2016) 32–51 


C 2016 New York Academy of Sciences.
Fairfield et al. Understanding the pathogenesis of multiple myeloma

38. Röllig, C., S. Knop & M. Bornhäuser. 2015. Multiple 55. Asosingh, K., H. De Raeve, M. de Ridder, et al. 2005. Role of
myeloma. Lancet 385: 2197–2208. the hypoxic bone marrow microenvironment in 5T2MM
39. Lorsbach, R.B., E.D. Hsi, A. Dogan & F. Fend. 2011. Plasma murine myeloma tumor progression. Haematologica 90:
cell myeloma and related neoplasms. Am. J. Clin. Pathol. 810–817.
136: 168–182. 56. Martin, S.K., P. Diamond, S.A. Williams, et al. 2010.
40. Glavey, S.V. & I.M. Ghobrial. 2015. American Society of Hypoxia-inducible factor-2 is a novel regulator of aber-
Hematology annual meeting 2014: highlights in multiple rant CXCL12 expression in multiple myeloma plasma cells.
myeloma. Expert Rev. Hematol. 8: 273–275. Haematologica 95: 776–784.
41. Rajkumar, S.V., M.A. Dimopoulos, A. Palumbo, et al. 2014. 57. De Raeve, H.R. & K. Vanderkerken. 2005. The role of
International Myeloma Working Group updated criteria the bone marrow microenvironment in multiple myeloma.
for the diagnosis of multiple myeloma. Lancet Oncol. 15: Histol. Histopathol. 20: 1227–1250.
e538–e548. 58. Reagan, M.R. & D.L. Kaplan. 2011. Concise review: mes-
42. Simeon, V., K. Todoerti, F. La Rocca, et al. 2015. Molecular enchymal stem cell tumor-homing: detection methods in
classification and pharmacogenetics of primary plasma cell disease model systems. Stem Cells 29: 920–927.
leukemia: an initial approach toward precision medicine. 59. Abe, M. 2011. Targeting the interplay between myeloma
Int. J. Mol. Sci. 16: 17514–17534. cells and the bone marrow microenvironment in myeloma.
43. Podar, K., D. Chauhan & K.C. Anderson. 2009. Bone mar- Int. J. Hematol. 94: 334–343.
row microenvironment and the identification of new tar- 60. Garcia-Gomez, A. 2014. Multiple myeloma mesenchymal
gets for myeloma therapy. Leukemia 23: 10–24. stromal cells: contribution to myeloma bone disease and
44. Ecklund, K., S. Vajapeyam, H.A. Feldman, et al. 2010. Bone therapeutics. World J. Stem Cells 6: 322–343.
marrow changes in adolescent girls with anorexia nervosa. 61. Tsujimoto, T., I.A. Lisukov, N. Huang, et al. 1996. Plasma
J. Bone Miner. Res. 25: 298–304. cells induce apoptosis of pre-B cells by interacting with
45. Rosen, C.J., C. Ackert-Bicknell, J.P. Rodriguez & A.M. Pino. bone marrow stromal cells. Blood 87: 3375–3383.
2009. Marrow fat and the bone microenvironment: devel- 62. Reagan, M.R., Y. Mishima, S.V. Glavey, et al. 2014. In-
opmental, functional, and pathological implications. Crit. vestigating osteogenic differentiation in multiple myeloma
Rev. Eukaryot. Gene Expr. 19: 109–124. using a novel 3D bone marrow niche model. Blood 124:
46. Kawai, M., F.J. de Paula & C.J. Rosen. 2012. New insights 3250–3259.
into osteoporosis: the bone–fat connection. J. Intern. Med. 63. Markovina, S., N.S. Callander, S.L. O’Connor, et al. 2010.
272: 317–329. Bone marrow stromal cells from multiple myeloma patients
47. Hardaway, A.L., M.K. Herroon, E. Rajagurubandara & uniquely induce bortezomib resistant NF-kappaB activity
I. Podgorski. 2014. Bone marrow fat: linking adipocyte- in myeloma cells. Mol. Cancer 9: 176.
induced inflammation with skeletal metastases. Cancer 64. Kastritis, E., A. Palumbo & M.A. Dimopoulos. 2009. Treat-
Metastasis Rev. 33: 527–543. ment of relapsed/refractory multiple myeloma. Semin.
48. Reagan, M.R. & C.J. Rosen. 2015. Navigating the bone mar- Hematol. 46: 143–157.
row niche: translational insights and cancer-driven dys- 65. Kawano, M., T. Hirano, T. Matsuda, et al. 1988. Autocrine
function. Nat. Rev. Rheumatol. 12:154–168. generation and requirement of BSF-2/IL-6 for human mul-
49. Ibrahim, M., M. Jang, M. Park, et al. 2015. Capsaicin tiple myelomas. Nature 332: 83–85.
inhibits the adipogenic differentiation of bone marrow 66. Teoh, H.K., P.P. Chong, M. Abdullah, et al. 2016. Small
mesenchymal stem cells by regulating cell proliferation, interfering RNA silencing of interleukin-6 in mesenchymal
apoptosis, oxidative and nitrosative stress. Food Funct. 6: stromal cells inhibits multiple myeloma cell growth. Leuk.
2165–2178. Res. 40: 44–53.
50. Yagi, H., J. Tan & R.S. Tuan. 2013. Polyphenols suppress hy- 67. Sims, N.A. & T.J. Martin. 2014. Coupling the activities
drogen peroxide-induced oxidative stress in human bone- of bone formation and resorption: a multitude of sig-
marrow derived mesenchymal stem cells. J. Cell. Biochem. nals within the basic multicellular unit. Bonekey Rep. 3:
114: 1163–1173. 481.
51. Hole, P.S., R.L. Darley & A. Tonks. 2011. Do reactive oxygen 68. Feng, X. & J.M. McDonald. 2011. Disorders of bone re-
species play a role in myeloid leukemias? Blood 117: 5816– modeling. Annu. Rev. Pathol. 6: 121–145.
5826. 69. Andersen, T.L., K. Søe, T.E. Sondergaard, et al. 2010.
52. Gilbert, L.C., H. Chen, X. Lu & M.S. Nanes. 2013. Chronic Myeloma cell-induced disruption of bone remodelling
low dose tumor necrosis factor-alpha (TNF) suppresses compartments leads to osteolytic lesions and generation
early bone accrual in young mice by inhibiting osteoblasts of osteoclast-myeloma hybrid cells: research paper. Br. J.
without affecting osteoclasts. Bone 56: 174–183. Haematol. 148: 551–561.
53. Raaijmakers, M.H.G.P., S. Mukherjee, S. Guo, et al. 2010. 70. Chen, Z., R.Z. Orlowski, M. Wang, et al. 2014. Osteoblastic
Bone progenitor dysfunction induces myelodysplasia and niche supports the growth of quiescent multiple myeloma
secondary leukaemia. Nature 464: 852–857. cells. Blood 123: 2204–2208.
54. Roccaro, A.M., Y. Mishima, A. Sacco, et al. 2015. CXCR4 71. Yaccoby, S., M.J. Wezeman, M. Zangari, et al. 2006. In-
regulates extra-medullary myeloma through epithelial- hibitory effects of osteoblasts and increased bone formation
mesenchymal-transition-like transcriptional activation. on myeloma in novel culture systems and a myelomatous
Cell Rep. 12: 622–635. mouse model. Haematologica 91: 192–199.

Ann. N.Y. Acad. Sci. 1364 (2016) 32–51 


C 2016 New York Academy of Sciences. 45
Understanding the pathogenesis of multiple myeloma Fairfield et al.

72. Li, X., A. Pennisi & S. Yaccoby. 2008. Role of decorin in the 89. Baron, R. & M. Kneissel. 2013. WNT signaling in bone
antimyeloma effects of osteoblasts. Blood 112: 159–168. homeostasis and disease: from human mutations to treat-
73. Lawson, M.A., M.M. McDonald, N. Kovacic, et al. 2015. ments. Nat. Med. 19: 179–192.
Osteoclasts control reactivation of dormant myeloma cells 90. Semenov, M., K. Tamai & X. He. 2005. SOST is a ligand for
by remodelling the endosteal niche. Nat. Commun. 6: LRP5/LRP6 and a Wnt signaling inhibitor. J. Biol. Chem.
8983. 280: 26770–26775.
74. Kovacic, N., P.I. Croucher & M.M. McDonald. 2013. Sig- 91. Niedźwiedzki, T. & J. Filipowska. 2015. Bone remodeling
naling between tumor cells and the host bone marrow mi- in the context of cellular and systemic regulation: the role
croenvironment. Calcif. Tissue Int. 94: 125–139. of osteocytes and the nervous system. J. Mol. Endocrinol.
75. Tian, E., F. Zhan, R. Walker, et al. 2003. The role of the Wnt- 55: R23–R36.
signaling antagonist DKK1 in the development of osteolytic 92. Colucci, S., G. Brunetti, A. Oranger, et al. 2011. Myeloma
lesions in multiple myeloma. N. Engl. J. Med. 349: 2483– cells suppress osteoblasts through sclerostin secretion.
2494. Blood Cancer J. 1: e27.
76. Zannettino, A.C.W., A.N. Farrugia, A. Kortesidis, et al. 93. Giuliani, N., M. Ferretti, M. Bolzoni, et al. 2012. In-
2005. Elevated serum levels of stromal-derived factor- creased osteocyte death in multiple myeloma patients: role
1alpha are associated with increased osteoclast activity and in myeloma-induced osteoclast formation. Leukemia 26:
osteolytic bone disease in multiple myeloma patients. Can- 1391–1401.
cer Res. 65: 1700–1709. 94. Terpos, E., D. Christoulas, E. Katodritou, et al. 2012. Ele-
77. Choi, S.J., J.C. Cruz, F. Craig, et al. 2000. Macrophage vated circulating sclerostin correlates with advanced disease
inflammatory protein 1-alpha is a potential osteoclast features and abnormal bone remodeling in symptomatic
stimulatory factor in multiple myeloma. Blood 96: 671– myeloma: reduction post-bortezomib monotherapy. Int. J.
675. Cancer 131: 1466–1471.
78. Obeng, E.A., D.M. Gutman, L.M. Carlson, et al. 2006. 95. Brunetti, G., A. Oranger, G. Mori, et al. 2011. Sclerostin
Proteasome inhibitors induce a terminal unfolded pro- is overexpressed by plasma cells from multiple myeloma
tein response in multiple myeloma cells. Blood 107: 4907– patients. Ann. N. Y. Acad. Sci. 1237: 19–23.
4916. 96. Wang, X.T., Y.C. He, S.Y. Zhou, et al. 2014. Bone mar-
79. Ramasamy, K., H. Khatun, L. Macpherson, et al. 2012. row plasma macrophage inflammatory protein protein-1
Fluorescence-based experimental model to evaluate the alpha(MIP-1 alpha) and sclerostin in multiple myeloma:
concomitant effect of drugs on the tumour microenviron- relationship with bone disease and clinical characteristics.
ment and cancer cells. Br. J. Haematol. 157: 564–579. Leuk. Res. 38: 525–531.
80. Tanaka, Y., M. Abe, M. Hiasa, et al. 2007. Myeloma cell– 97. Lecka-Czernik, B. 2012. Marrow fat metabolism is linked
osteoclast interaction enhances angiogenesis together with to the systemic energy metabolism. Bone 50: 534–539.
bone resorption: a role for vascular endothelial cell growth 98. Vogl, D.T., T. Wang, W.S. Pérez, et al. 2011. Effect of obe-
factor and osteopontin. Clin. Cancer Res. 13: 816–823. sity on outcomes after autologous hematopoietic stem cell
81. Lacey, D.L., E. Timms, H.L. Tan, et al. 1998. Osteoprote- transplantation for multiple myeloma. Biol. Blood Marrow
gerin ligand is a cytokine that regulates osteoclast differen- Transplant. 17: 1765–1774.
tiation and activation. Cell 93: 165–176. 99. Islam, R., K. Altundag, M. Kurt, et al. 2005. Association
82. Kong, Y.Y., H. Yoshida, I. Sarosi, et al. 1999. OPGL is a key between obesity and multiple myeloma in postmenopausal
regulator of osteoclastogenesis, lymphocyte development women may be attributed to increased aromatization of
and lymph-node organogenesis. Nature 397: 315–323. androgen in adipose tissue. Med. Hypotheses 65: 1001–1002.
83. Roodman, G.D. & W.C. Dougall. 2008. RANK ligand as 100. Caers, J., S. Deleu, Z. Belaid, et al. 2007. Neighboring
a therapeutic target for bone metastases and multiple adipocytes participate in the bone marrow microenviron-
myeloma. Cancer Treat. Rev. 34: 92–101. ment of multiple myeloma cells. Leukemia 21: 1580–1584.
84. Henry, D.H., L. Costa, F. Goldwasser, et al. 2011. Random- 101. Liu, Z., J. Xu, J. He, et al. 2015. Mature adipocytes in
ized, double-blind study of denosumab versus zoledronic bone marrow protect myeloma cells against chemother-
acid in the treatment of bone metastases in patients with apy through autophagy activation. Oncotarget 6: 34329–
advanced cancer (excluding breast and prostate cancer) or 34341.
multiple myeloma. J. Clin. Oncol. 29: 1125–1132. 102. Arita, Y., S. Kihara, N. Ouchi, et al. 1999. Paradoxical de-
85. Dallas, S.L., M. Prideaux & L.F. Bonewald. 2013. The os- crease of an adipose-specific protein, adiponectin, in obe-
teocyte: an endocrine cell . . . and more. Endocr. Rev. 34: sity. Biochem. Biophys. Res. Commun. 257: 79–83.
658–690. 103. Bråkenhielm, E., N. Veitonmäki, R. Cao, et al. 2004.
86. van Bezooijen, R.L. 2004. Sclerostin is an osteocyte- Adiponectin-induced antiangiogenesis and antitumor ac-
expressed negative regulator of bone formation, but not tivity involve caspase-mediated endothelial cell apoptosis.
a classical bmp antagonist. J. Exp. Med. 199: 805–814. Proc. Natl. Acad. Sci. U.S.A. 101: 2476–2481.
87. Kogianni, G. & B.S. Noble. 2007. The biology of osteocytes. 104. Dalamaga, M., K.N. Diakopoulos & C.S. Mantzoros. 2012.
Curr. Osteoporos. Rep. 5: 81–86. The role of adiponectin in cancer: a review of current evi-
88. Nakashima, T., M. Hayashi, T. Fukunaga, et al. 2011. Evi- dence. Endocr. Rev. 33: 547–594.
dence for osteocyte regulation of bone homeostasis through 105. Esheba, N.E., A. Shahba & O. El Shora. 2014. Assessment
RANKL expression. Nat. Med. 17: 1231–1234. of leptin and resistin levels in non-obese multiple myeloma

46 Ann. N.Y. Acad. Sci. 1364 (2016) 32–51 


C 2016 New York Academy of Sciences.
Fairfield et al. Understanding the pathogenesis of multiple myeloma

patients and their relation with Ig level and disease stage. J. of the International Myeloma Working Group. Leukemia
Egypt. Natl. Canc. Inst. 26: 61–66. DOI: 10.1038/leu.2015.356.
106. Hofmann, J.N., L.M. Liao, M.N. Pollak, et al. 2012. A 122. Orlowski, R.Z., A. Nagler, P. Sonneveld, et al. 2007. Ran-
prospective study of circulating adipokine levels and risk domized phase III study of pegylated liposomal doxoru-
of multiple myeloma. Blood 120: 4418–4421. bicin plus bortezomib compared with bortezomib alone
107. Winkler, I.G., N.A. Sims, A.R. Pettit, et al. 2010. Bone in relapsed or refractory multiple myeloma: combination
marrow macrophages maintain hematopoietic stem cell therapy improves time to progression. J. Clin. Oncol. 25:
(HSC) niches and their depletion mobilizes HSCs. Blood 3892–3901.
116: 4815–4828. 123. Sonneveld, P., R. Hajek, A. Nagler, et al. 2008. Combined
108. Casanova-Acebes, M., N. A-González, L.A. Weiss & A. Hi- pegylated liposomal doxorubicin and bortezomib is highly
dalgo. 2014. Innate immune cells as homeostatic regulators effective in patients with recurrent or refractory multi-
of the hematopoietic niche. Int. J. Hematol. 99: 685–694. ple myeloma who received prior thalidomide/lenalidomide
109. Baschuk, N., J. Rautela & B.S. Parker. 2015. Bone specific therapy. Cancer 112: 1529–1537.
immunity and its impact on metastasis. Bonekey Rep. 4: 124. Palumbo, A., A. Bertola, P. Musto, et al. 2005. Oral mel-
665. phalan, prednisone, and thalidomide for newly diagnosed
110. Reya, T. 2011. Illuminating immune privilege–a role for patients with myeloma. Cancer 104: 1428–1433.
regulatory T cells in preventing rejection. N. Engl. J. Med. 125. Garcı́a-Sanz, R., M.I. González-Fraile, M. Sierra, et al. 2002.
365: 956–957. The combination of thalidomide, cyclophosphamide and
111. Dawson, M.R., S.S. Chae, R.K. Jain & D.G. Duda. 2011. dexamethasone (ThaCyDex) is feasible and can be an op-
Direct evidence for lineage-dependent effects of bone mar- tion for relapsed/refractory multiple myeloma. Hematol. J.
row stromal cells on tumor progression. Am. J. Cancer Res. 3: 43–48.
1: 144–154. 126. Kiziltepe, T., T. Hideshima, L. Catley, et al. 2007. 5-
112. Asimakopoulos, F., J. Kim, R.A. Denu, et al. 2013. Azacytidine, a DNA methyltransferase inhibitor, induces
Macrophages in multiple myeloma: emerging concepts and ATR-mediated DNA double-strand break responses, apop-
therapeutic implications. Leuk. Lymphoma 54: 2112–2121. tosis, and synergistic cytotoxicity with doxorubicin and
113. Kawano, Y., M. Moschetta, S. Manier, et al. 2015. Targeting bortezomib against multiple myeloma cells. Mol. Cancer
the bone marrow microenvironment in multiple myeloma. Ther. 6: 1718–1727.
Immunol. Rev. 263: 160–172. 127. Ciechanover, A., A. Orian & A.L. Schwartz. 2000. The
114. Dankbar, B., T. Padró, R. Leo, et al. 2000. Vascular endothe- ubiquitin-mediated proteolytic pathway: mode of action
lial growth factor and interleukin-6 in paracrine tumor– and clinical implications. J. Cell. Biochem. 77: 40–51.
stromal cell interactions in multiple myeloma. Blood 95: 128. Giuliani, N., F. Morandi, S. Tagliaferri, et al. 2007. The
2630–2636. proteasome inhibitor bortezomib affects osteoblast differ-
115. Moschetta, M., Y. Mishima, I. Sahin, et al. 2014. Role of entiation in vitro and in vivo in multiple myeloma patients.
endothelial progenitor cells in cancer progression. Biochim. Blood 110: 334–338.
Biophys. Acta 1846: 26–39. 129. von Metzler, I., H. Krebbel, M. Hecht, et al. 2007. Borte-
116. Mishima, Y., Y. Aljawai, I. Sahin, et al. 2013. Bone marrow zomib inhibits human osteoclastogenesis. Leukemia 21:
mobilization of endothelial progenitor cells represents an 2025–2034.
early pathogenic event during multiple myeloma progres- 130. Popat, R., H. Oakervee, C. Williams, et al. 2009. Borte-
sion. Blood 122: 680. zomib, low-dose intravenous melphalan, and dexametha-
117. Kyle, R.A. & S.V. Rajkumar. 2004. Multiple myeloma. N. sone for patients with relapsed multiple myeloma. Br. J.
Engl. J. Med. 351: 1860–1873. Haematol. 144: 887–894.
118. Kyle, R.A., E.D. Remstein, T.M. Therneau, et al. 2007. 131. Kropff, M., G. Bisping, E. Schuck, et al. 2007. Bortezomib in
Clinical course and prognosis of smoldering (asymp- combination with intermediate-dose dexamethasone and
tomatic) multiple myeloma. N. Engl. J. Med. 356: 2582– continuous low-dose oral cyclophosphamide for relapsed
2590. multiple myeloma. Br. J. Haematol. 138: 330–337.
119. Hussein, K., A. Stucki-Koch, H. Alchalby, et al. 2015. Cy- 132. Ludwig, H., H. Kasparu, C. Leitgeb, et al. 2014.
tokine expression pattern in bone marrow microenviron- Bendamustine–bortezomib–dexamethasone is an active
ment after allogeneic stem cell transplantation in primary and well-tolerated regimen in patients with relapsed or
myelofibrosis. Biol. Blood Marrow Transplant. pii: S1083- refractory multiple myeloma. Blood 123: 985–91.
8791(15)01873-X. 133. Offidani, M., L. Corvatta, L. Maracci, et al. 2013. Efficacy
120. Gay, F., S. Oliva, M.T. Petrucci, et al. 2015. Chemother- and tolerability of bendamustine, bortezomib and dex-
apy plus lenalidomide versus autologous transplantation, amethasone in patients with relapsed-refractory multiple
followed by lenalidomide plus prednisone versus lenalido- myeloma: a phase II study. Blood Cancer J. 3: e162.
mide maintenance, in patients with multiple myeloma: a 134. Harousseau, J.L., M.A. Dimopoulos, M. Wang, et al. 2010.
randomised, multicentre, phase 3 trial. Lancet Oncol. 16: Better quality of response to lenalidomide plus dexam-
1617–1629. ethasone is associated with improved clinical outcomes
121. Laubach, J., L. Garderet, A. Mahindra, et al. 2015. Man- in patients with relapsed or refractory multiple myeloma.
agement of relapsed multiple myeloma: recommendations Haematologica 95: 1738–1744.

Ann. N.Y. Acad. Sci. 1364 (2016) 32–51 


C 2016 New York Academy of Sciences. 47
Understanding the pathogenesis of multiple myeloma Fairfield et al.

135. Mitsiades, N., C.S. Mitsiades, P.G. Richardson, et al. 2003. 151. Levy, J. & G.D. Roodman. 2009. The role of bisphospho-
The proteasome inhibitor PS-341 potentiates sensitivity of nates in multiple myeloma. Curr. Hematol. Malig. Rep. 4:
multiple myeloma cells to conventional chemotherapeutic 108–112.
agents: therapeutic applications. Blood 101: 2377–2380. 152. Merin, N.M. & K.R. Kelly. 2014. Clinical use of proteasome
136. Ma, M.H., H.H. Yang, K. Parker, et al. 2003. The proteasome inhibitors in the treatment of multiple myeloma. Pharma-
inhibitor PS-341 markedly enhances sensitivity of multiple ceuticals (Basel). 8: 1–20.
myeloma tumor cells to chemotherapeutic agents. Clin. 153. Berenson, J.R., A. Lichtenstein, L. Porter, et al. 1996. Effi-
Cancer Res. 9: 1136–1144. cacy of pamidronate in reducing skeletal events in patients
137. Richardson, P.G., B. Barlogie, J. Berenson, et al. 2003. with advanced multiple myeloma. Myeloma Aredia Study
A phase 2 study of bortezomib in relapsed, refractory Group. N. Engl. J. Med. 334: 488–493.
myeloma. N. Engl. J. Med. 348: 2609–2617. 154. Berenson, J.R., A. Lichtenstein, L. Porter, et al. 1998.
138. Petrucci, M.T., P. Giraldo, P. Corradini, et al. 2013. A Long-term pamidronate treatment of advanced multiple
prospective, international phase 2 study of bortezomib re- myeloma patients reduces skeletal events. Myeloma Aredia
treatment in patients with relapsed multiple myeloma. Br. Study Group. J. Clin. Oncol. 16: 593–602.
J. Haematol. 160: 649–659. 155. Chlebowski, R.T., Z. Chen, J.A. Cauley, et al. 2010. Oral
139. Jakubowiak, A.J., D. Dytfeld, K.A. Griffith, et al. 2012. bisphosphonate use and breast cancer incidence in post-
A phase 1/2 study of carfilzomib in combination with menopausal women. J. Clin. Oncol. 28: 3582–3590.
lenalidomide and low-dose dexamethasone as a frontline 156. Holen, I. & R.E. Coleman. 2010. Bisphosphonates as treat-
treatment for multiple myeloma. Blood 120: 1801–1809. ment of bone metastases. Curr. Pharm. Des. 16: 1262–
140. Wang, M. & J. Cheng. 2013. Overview and manage- 1271.
ment of cardiac and pulmonary adverse events in patients 157. Gnant, M. & H. Eidtmann. 2010. The anti-tumor effect of
with relapsed and/or refractory multiple myeloma treated bisphosphonates ABCSG-12, ZO-FAST and more . . . Crit.
with single-agent carfilzomib. Oncology (Williston Park) Rev. Oncol. Hematol. 74(Suppl. 1): S2–S6.
27(Suppl. 3): 24–30. 158. Van Acker, H.H., S. Anguille, Y. Willemen, et al. 2016. Bis-
141. Siegel, D.S., T. Martin, M. Wang, et al. 2012. A phase 2 study phosphonates for cancer treatment: mechanisms of action
of single-agent carfilzomib (PX-171-003-A1) in patients and lessons from clinical trials. Pharmacol. Ther. 158: 24–
with relapsed and refractory multiple myeloma. Blood 120: 40.
2817–2825. 159. Glavey, S., M. Reagan, S. Manier, et al. 2014. Dissecting the
142. Shah, J.J., E.A. Stadtmauer, R. Abonour, et al. 2015. Carfil- mechanisms of activity of SLAMF7 and the targeting anti-
zomib, pomalidomide, and dexamethasone for relapsed or body elotuzumab in multiple myeloma. Blood 124: 3431.
refractory myeloma. Blood 126: 2284–2290. 160. Gabrilovich, D.I. & S. Nagaraj. 2009. Myeloid-derived sup-
143. Averbuch, S.D. 1993. New bisphosphonates in the treat- pressor cells as regulators of the immune system. Nat. Rev.
ment of bone metastases. Cancer 72: 3443–3452. Immunol. 9: 162–174.
144. Baron, R., S. Ferrari & R.G.G. Russell. 2011. Denosumab 161. Zhu, Y.X., K.M. Kortuem & A.K. Stewart. 2012. Molec-
and bisphosphonates: different mechanisms of action and ular mechanism of action of immune-modulatory drugs
effects. Bone 48: 677–692. thalidomide, lenalidomide and pomalidomide in multiple
145. Mundy, G.R., K.J. Ibbotson, S.M. D’Souza, et al. 1984. myeloma. Leuk. Lymphoma 54: 1–5.
The hypercalcemia of cancer. Clinical implications and 162. Dimopoulos, M.A., A.S. Swern, J.S. Li, et al. 2014. Efficacy
pathogenic mechanisms. N. Engl. J. Med. 310: 1718–1727. and safety of long-term treatment with lenalidomide and
146. Croucher, P.I. & J.F. Apperley. 1998. Bone disease in multi- dexamethasone in patients with relapsed/refractory multi-
ple myeloma. Br. J. Haematol. 103: 902–910. ple myeloma. Blood Cancer J. 4: e257.
147. Bataille, R., P.D. Delmas, D. Chappard & J. Sany. 1990. Ab- 163. Richardson, P.G., W. Xie, S. Jagannath, et al. 2014. A phase
normal serum bone Gla protein levels in multiple myeloma. 2 trial of lenalidomide, bortezomib, and dexamethasone
Crucial role of bone formation and prognostic implica- in patients with relapsed and relapsed/refractory myeloma.
tions. Cancer 66: 167–172. Blood 123: 1461–1469.
148. Bataille, R. & B. Klein. 1991. Serum beta-2-microglobulin 164. Santo, L., T. Hideshima, D. Cirstea, et al. 2011. An-
(beta 2m) in myeloma: toward a simple prognostic stratifi- timyeloma activity of a multitargeted kinase inhibitor,
cation using beta 2M and acute-phase proteins? Blood 77: AT9283, via potent Aurora kinase and STAT3 inhibition
1616–1617. either alone or in combination with lenalidomide. Clin.
149. Abildgaard, N., J. Rungby, H. Glerup, et al. 1998. Long-term Cancer Res. 17: 3259–3271.
oral pamidronate treatment inhibits osteoclastic bone re- 165. van der Veer, M.S., M. de Weers, B. van Kessel, et al. 2011.
sorption and bone turnover without affecting osteoblastic Towards effective immunotherapy of myeloma: enhanced
function in multiple myeloma. Eur. J. Haematol. 61: 128– elimination of myeloma cells by combination of lenalido-
134. mide with the human CD38 monoclonal antibody daratu-
150. Jagannath, S., B.G.M. Durie, J. Wolf, et al. 2005. Bortezomib mumab. Haematologica 96: 284–290.
therapy alone and in combination with dexamethasone for 166. Jakubowiak, A.J., K.A. Griffith, D.E. Reece, et al. 2011.
previously untreated symptomatic multiple myeloma. Br. Lenalidomide, bortezomib, pegylated liposomal doxoru-
J. Haematol. 129: 776–783. bicin, and dexamethasone in newly diagnosed multiple

48 Ann. N.Y. Acad. Sci. 1364 (2016) 32–51 


C 2016 New York Academy of Sciences.
Fairfield et al. Understanding the pathogenesis of multiple myeloma

myeloma: a phase 1/2 Multiple Myeloma Research Con- row biopsies of multiple myeloma: shed syndecan-1 accu-
sortium trial. Blood 118: 535–543. mulates in fibrotic regions. Mod. Pathol. 14: 1052–1058.
167. Weisel, K., M. Dimopoulos, K.W. Song, et al. 2015. Poma- 179. Fichou, N., S. Gouard, C. Maurel, et al. 2015. Single-
lidomide and low-dose dexamethasone improves health- dose anti-CD138 radioimmunotherapy: bismuth-213 is
related quality of life and prolongs time to worsening in re- more efficient than lutetium-177 for treatment of multi-
lapsed/refractory patients with multiple myeloma enrolled ple myeloma in a preclinical model. Front. Med. 2: 76.
in the MM-003 randomized phase III trial. Clin. Lymphoma 180. Kim, S.Y., J.-W. Theunissen, J. Balibalos, et al. 2015. A novel
Myeloma Leuk. 15: 519–530. antibody–drug conjugate targeting SAIL for the treatment
168. Mitsiades, N., C.S. Mitsiades, V. Poulaki, et al. 2002. Apop- of hematologic malignancies. Blood Cancer J. 5: e316.
totic signaling induced by immunomodulatory thalido- 181. Glavey, S.V., S. Manier, A. Natoni, et al. 2014. The sialyl-
mide analogs in human multiple myeloma cells: therapeutic transferase ST3GAL6 influences homing and survival in
implications. Blood 99: 4525–4530. multiple myeloma. Blood 124: 1765–1777.
169. Prince, H.M., M. Adena, D.K. Smith & J. Hertel. 2007. Effi- 182. Lonial, S., M. Dimopoulos, A. Palumbo, et al. 2015.
cacy of single-agent bortezomib vs. single-agent thalido- Elotuzumab therapy for relapsed or refractory multiple
mide in patients with relapsed or refractory multiple myeloma. N. Engl. J. Med. 373: 621–631.
myeloma: a systematic comparison. Eur. J. Haematol. 79: 183. Jin, H., B. Wang, J. Li, et al. 2015. Anti-DKK1 antibody
93–99. promotes bone fracture healing through activation of ␤-
170. Pineda-Roman, M., M. Zangari, F. van Rhee, et al. 2008. catenin signaling. Bone 71: 63–75.
VTD combination therapy with bortezomib–thalidomide– 184. Feng, X., S. Deng, G. An, et al. 2015. [Detection of serum
dexamethasone is highly effective in advanced and refrac- DKK1 in multiple myeloma and myeloma bone disease].
tory multiple myeloma. Leukemia 22: 1419–1427. Zhonghua Xue Ye Xue Za Zhi 36: 682–685.
171. Garrison, L.P., S.-T. Wang, H. Huang, et al. 2013. The 185. Ščudla, V., P. Petrová, T. Pika, et al. 2015. [Analysis of serum
cost-effectiveness of initial treatment of multiple myeloma levels of Dickkopf-1 (DKK-1) in monoclonal gammopathy
in the U.S. with bortezomib plus melphalan and pred- of undetermined significance and multiple myeloma]. Cas.
nisone versus thalidomide plus melphalan and prednisone Lek. Cesk. 154: 181–188.
or lenalidomide plus melphalan and prednisone with con- 186. Pozzi, S., M. Fulciniti, H. Yan, et al. 2013. In vivo and in
tinuous lenalidomide maintenance treatment. Oncologist vitro effects of a novel anti-Dkk1 neutralizing antibody in
18: 27–36. multiple myeloma. Bone 53: 487–496.
172. Garderet, L., S. Iacobelli, P. Moreau, et al. 2012. Su- 187. Scullen, T., L. Santo, S. Vallet, et al. 2013. Lenalidomide
periority of the triple combination of bortezomib– in combination with an activin A-neutralizing antibody:
thalidomide–dexamethasone over the dual combination of preclinical rationale for a novel anti-myeloma strategy.
thalidomide–dexamethasone in patients with multiple Leukemia 27: 1715–1721.
myeloma progressing or relapsing after autologous trans- 188. Vallet, S., S. Mukherjee, N. Vaghela, et al. 2010. Activin
plantation: the MMVAR/IFM 2005-04 randomized phase A promotes multiple myeloma-induced osteolysis and is
III trial from the Chronic Leukemia Working Party of the a promising target for myeloma bone disease. Proc. Natl.
European Group for Blood and Marrow Transplantation. Acad. Sci. U.S.A. 107: 5124–5129.
J. Clin. Oncol. 30: 2475–2482. 189. Kristensen, I.B., J.H. Christensen, M.B. Lyng, et al. 2014.
173. Sonneveld, P., I.G.H. Schmidt-Wolf, B. van der Holt, et al. Expression of osteoblast and osteoclast regulatory genes in
2012. Bortezomib induction and maintenance treatment in the bone marrow microenvironment in multiple myeloma:
patients with newly diagnosed multiple myeloma: results of only up-regulation of Wnt inhibitors SFRP3 and DKK1 is
the randomized phase III HOVON-65/ GMMG-HD4 trial. associated with lytic bone disease. Leuk. Lymphoma 55:
J. Clin. Oncol. 30: 2946–2955. 911–919.
174. Laubach, J.P., P.M. Voorhees, H. Hassoun, et al. 2014. Cur- 190. Reagan, M.R., M.M. McDonald, R. Terry, et al. 2015. Anti-
rent strategies for treatment of relapsed/refractory multiple sclerostin treatment prevents multiple myeloma induced
myeloma. Expert Rev. Hematol. 7: 97–111. bone loss and reduces tumor burden [abstract]. In 57th
175. de Weers, M., Y.-T. Tai, M.S. van der Veer, et al. 2011. Dara- Annual Meeting & Exposition of American Society of Hema-
tumumab, a novel therapeutic human CD38 monoclonal tology, Orlando, FL, 119.
antibody, induces killing of multiple myeloma and other 191. Heath, D.J., A.D. Chantry, C.H. Buckle, et al. 2009. In-
hematological tumors. J. Immunol. 186: 1840–1848. hibiting Dickkopf-1 (Dkk1) removes suppression of bone
176. Lokhorst, H.M., T. Plesner, J.P. Laubach, et al. 2015. Tar- formation and prevents the development of osteolytic bone
geting CD38 with daratumumab monotherapy in multiple disease in multiple myeloma. J. Bone Miner. Res. 24: 425–
myeloma. N. Engl. J. Med. 373: 1207–1219. 436.
177. Santonocito, A.M., U. Consoli, S. Bagnato, et al. 2004. Flow 192. van Dinther, M., J. Zhang, S.E. Weidauer, et al. 2013. Anti-
cytometric detection of aneuploid CD38(++) plasma cells sclerostin antibody inhibits internalization of sclerostin and
and CD19(+) B-lymphocytes in bone marrow, peripheral sclerostin-mediated antagonism of Wnt/LRP6 signaling.
blood and PBSC harvest in multiple myeloma patients. PLoS One 8: e62295.
Leuk. Res. 28: 469–477. 193. Mazziotti, G., J. Bilezikian, E. Canalis, et al. 2012. New
178. Bayer-Garner, I.B., R.D. Sanderson, M.V. Dhodapkar, et al. understanding and treatments for osteoporosis. Endocrine
2001. Syndecan-1 (CD138) immunoreactivity in bone mar- 41: 58–69.

Ann. N.Y. Acad. Sci. 1364 (2016) 32–51 


C 2016 New York Academy of Sciences. 49
Understanding the pathogenesis of multiple myeloma Fairfield et al.

194. Eda, H., L. Santo, M.N. Wein, et al. 2016. Regulation of 210. Ayed, A.O., L.-J. Chang & J.S. Moreb. 2015. Immunother-
sclerostin expression in multiple myeloma by Dkk-1: a po- apy for multiple myeloma: current status and future direc-
tential therapeutic strategy for myeloma bone disease. J. tions. Crit. Rev. Oncol. Hematol. 96: 399–412.
Bone Miner. Res. DOI: 10.1002/jbmr.2789. 211. Lorenz, J., J. Waldschmidt, D. Wider, et al. 2015. From
195. Sondergeld, P., N.W.C.J. van de Donk, P.G. Richardson & CLL to multiple myeloma – spleen tyrosine kinase (SYK)
T. Plesner. 2015. Monoclonal antibodies in myeloma. Clin. influences multiple myeloma cell survival and migration.
Adv. Hematol. Oncol. 13: 599–609. Br. J. Haematol. DOI: 10.1111/bjh.13825.
196. Bolden, J.E., M.J. Peart & R.W. Johnstone. 2006. Anticancer 212. Brown, R., S. Yang, C. Weatherburn, et al. 2015. Phospho-
activities of histone deacetylase inhibitors. Nat. Rev. Drug flow detection of constitutive and cytokine-induced
Discov. 5: 769–784. pSTAT3/5, pAKT and pERK expression highlights novel
197. Grunstein, M. 1997. Histone acetylation in chromatin prognostic biomarkers for patients with multiple myeloma.
structure and transcription. Nature 389: 349–352. Leukemia 29: 483–490.
198. Canella, A., H.C. Nieves, D.W. Sborov, et al. 2015. HDAC 213. Pemmaraju, N., H. Kantarjian, T. Kadia, et al. 2015. A phase
inhibitor AR-42 decreases CD44 expression and sensi- I/II study of the Janus kinase (JAK)1 and 2 inhibitor ruxoli-
tizes myeloma cells to lenalidomide. Oncotarget 6: 31134– tinib in patients with relapsed or refractory acute myeloid
31150. leukemia. Clin. Lymphoma Myeloma Leuk. 15: 171–176.
199. Dimopoulos, K., P. Gimsing & K. Grønbæk. 2014. The role 214. Nur, H., L. Rao, M.A. Frassanito, et al. 2014. Stimulation
of epigenetics in the biology of multiple myeloma. Blood of invariant natural killer T cells by ␣-galactosylceramide
Cancer J. 4: e207. activates the JAK-STAT pathway in endothelial cells and
200. Chauhan, D., Z. Tian, B. Nicholson, et al. 2012. A small reduces angiogenesis in the 5T33 multiple myeloma model.
molecule inhibitor of ubiquitin-specific protease-7 induces Br. J. Haematol. 167: 651–663.
apoptosis in multiple myeloma cells and overcomes borte- 215. Duan, W., S. Chen, Y. Zhang, et al. 2015. Protein C-terminal
zomib resistance. Cancer Cell 22: 345–358. enzymatic labeling identifies novel caspase cleavages during
201. Qian, D.Z., Y. Kato, S. Shabbeer, et al. 2006. Targeting tu- the apoptosis of multiple myeloma cells induced by kinase
mor angiogenesis with histone deacetylase inhibitors: the inhibition. Proteomics 16: 60–69.
hydroxamic acid derivative LBH589. Clin. Cancer Res. 12: 216. Suzuki, R., S. Kikuchi, T. Harada, et al. 2015. Combination
634–642. of a selective HSP90␣/␤ inhibitor and a RAS-RAF-MEK-
202. Dimopoulos, M., D.S. Siegel, S. Lonial, et al. 2013. Vorinos- ERK signaling pathway inhibitor triggers synergistic cyto-
tat or placebo in combination with bortezomib in patients toxicity in multiple myeloma cells. PLoS One 10: e0143847.
with multiple myeloma (VANTAGE 088): a multicentre, 217. Burger, M.T., G. Nishiguchi, W. Han, et al. 2015. Iden-
randomised, double-blind study. Lancet Oncol. 14: 1129– tification of N-(4-((1R,3S,5S)-3-amino-5-methylcyclo-
1140. hexyl)pyridin-3-yl)-6-(2,6-difluorophenyl)-5-fluoropico-
203. San Miguel, J., K. Weisel, P. Moreau, et al. 2013. Pomalido- linamide (PIM447), a potent and selective proviral
mide plus low-dose dexamethasone versus high-dose dex- insertion site of moloney murine leukemia (PIM) 1, 2,
amethasone alone for patients with relapsed and refractory and 3 kinase inhibitor in clinical trials for hematological
multiple myeloma (MM-003): a randomised, open-label, malignancies. J. Med. Chem. 58: 8373–8386.
phase 3 trial. Lancet Oncol. 14: 1055–1066. 218. Meads, M.B., B. Fang, L. Mathews, et al. 2015. Targeting
204. Biran, N., S. Shahnaz, S. Jagannath, et al. 2013. A phase PYK2 mediates microenvironment-specific cell death in
II, single-center, open-label study of oral panobinostat in multiple myeloma. Oncogene. DOI: 10.1038/onc.2015.334.
combination with lenalidomide and weekly dexametha- 219. Zhang, Y., M. Moschetta, D. Huynh, et al. 2014. Pyk2 pro-
sone in patients with multiple myeloma. Blood 122: 5392. motes tumor progression in multiple myeloma. Blood 124:
205. San-Miguel, J.F., V.T.M. Hungria, S.-S. Yoon, et al. 2014. 2675–2686.
Panobinostat plus bortezomib and dexamethasone ver- 220. Han, K., X. Xu, Z. Xu, et al. 2015. SC06, a novel small
sus placebo plus bortezomib and dexamethasone in pa- molecule compound, displays preclinical activity against
tients with relapsed or relapsed and refractory multiple multiple myeloma by disrupting the mTOR signaling path-
myeloma: a multicentre, randomised, double-blind phase way. Sci. Rep. 5: 12809.
3 trial. Lancet Oncol. 15: 1195–1206. 221. Chinen, Y., J. Kuroda, Y. Shimura, et al. 2014. Phos-
206. Olechnowicz, S.W.Z. & C.M. Edwards. 2014. Contributions phoinositide protein kinase PDPK1 is a crucial cell sig-
of the host microenvironment to cancer-induced bone dis- naling mediator in multiple myeloma. Cancer Res. 74:
ease. Cancer Res. 74: 1625–1631. 7418–7429.
207. Elefteriou, F., P. Campbell & Y. Ma. 2014. Control of bone 222. Garfall, A.L., J.A. Fraietta & M.V. Maus. 2014. Immunother-
remodeling by the peripheral sympathetic nervous system. apy with chimeric antigen receptors for multiple myeloma.
Calcif. Tissue Int. 94: 140–151. Discov. Med. 17: 37–46.
208. Ashjian, E. & K. Redic. 2015. Multiple myeloma: updates 223. Maus, M.V. & C.H. June. 2014. CARTs on the road for
for pharmacists in the treatment of relapsed and refractory myeloma. Clin. Cancer Res. 20: 3899–3901.
disease. J. Oncol. Pharm. Pract. 22: 289–302. 224. Guo, B., M. Chen, Q. Han, et al. 2015. CD138-directed
209. Auner, H.W. & S. Cenci. 2015. Recent advances and future adoptive immunotherapy of chimeric antigen receptor
directions in targeting the secretory apparatus in multiple (CAR)-modified T cells for multiple myeloma. J. Cell. Im-
myeloma. Br. J. Haematol. 168: 14–25. munother. DOI: 10.1016/j.jocit.2014.11.001.

50 Ann. N.Y. Acad. Sci. 1364 (2016) 32–51 


C 2016 New York Academy of Sciences.
Fairfield et al. Understanding the pathogenesis of multiple myeloma

225. Roussel, M., V. Lauwers-Cances, N. Robillard, et al. 2014. multiple myeloma and overcomes the effects of age and
Front-line transplantation program with lenalidomide, prior therapy. Biol. Blood Marrow Transplant. 17: 141–146.
bortezomib, and dexamethasone combination as induction 234. Starr, P. 2015. Elotuzumab, first-in-class monoclonal anti-
and consolidation followed by lenalidomide maintenance body immunotherapy, improves outcomes in patients with
in patients with multiple myeloma: a phase II study by multiple myeloma. Am. Health Drug Benefits 8: 17.
the Intergroupe Francophone du Myélo. J. Clin. Oncol. 32: 235. Usmani, S.Z., R. Sexton, S. Ailawadhi, et al. 2015. Phase I
2712–2717. safety data of lenalidomide, bortezomib, dexamethasone,
226. Stewart, A.K., S.V. Rajkumar, M.A. Dimopoulos, et al. and elotuzumab as induction therapy for newly diagnosed
2015. Carfilzomib, lenalidomide, and dexamethasone for symptomatic multiple myeloma: SWOG S1211. Blood Can-
relapsed multiple myeloma. N. Engl. J. Med. 372: 142– cer J. 5: e334.
152. 236. Hansson, M., P. Gimsing, A. Badros, et al. 2015. A
227. Hurchla, M.A., A. Garcia-Gomez, M.C. Hornick, et al. phase I dose-escalation study of antibody BI-505 in re-
2013. The epoxyketone-based proteasome inhibitors lapsed/refractory multiple myeloma. Clin. Cancer Res. 21:
carfilzomib and orally bioavailable oprozomib have anti- 2730–2736.
resorptive and bone-anabolic activity in addition to anti- 237. Baz, R., S. Fanning, L. Kunkel, et al. 2007. Combination
myeloma effects. Leukemia 27: 430–440. of rituximab and oral melphalan and prednisone in newly
228. Gallerani, E., M. Zucchetti, D. Brunelli, et al. 2013. A first diagnosed multiple myeloma. Leuk. Lymphoma 48: 2338–
in human phase I study of the proteasome inhibitor CEP- 2344.
18770 in patients with advanced solid tumours and multi- 238. Bergua, J.M., C. Cabrera, E.G. Arteta & J. Prieto. 2008.
ple myeloma. Eur. J. Cancer. 49: 290–296. Rituximab in CD20 positive multiple myeloma. Leukemia
229. Palumbo, A., A. Larocca, M. Genuardi, et al. 2010. 22: 1082–1083.
Melphalan, prednisone, thalidomide and defibrotide in re- 239. Korte, W., C. Jost, S. Cogliatti, et al. 1999. Accelerated pro-
lapsed/refractory multiple myeloma: results of a multicen- gression of multiple myeloma during anti-CD20 (Ritux-
ter phase I/II trial. Haematologica 95: 1144–1149. imab) therapy. Ann. Oncol. 10: 1249–1250.
230. Kyle, R.A., S. Jacobus, W.R. Friedenberg, et al. 2009. The 240. Mitsiades, C.S., N.S. Mitsiades, C.J. McMullan, et al. 2004.
treatment of multiple myeloma using vincristine, car- Transcriptional signature of histone deacetylase inhibition
mustine, melphalan, cyclophosphamide, and prednisone in multiple myeloma: biological and clinical implications.
(VBMCP) alternating with high-dose cyclophosphamide Proc. Natl. Acad. Sci. U.S.A. 101: 540–545.
and alpha(2)beta interferon versus VBMCP: results of a 241. Richardson, P., C. Mitsiades, K. Colson, et al. 2008. Phase
phase III Eastern Cooperative Oncology. Cancer 115: 2155– I trial of oral vorinostat (suberoylanilide hydroxamic acid,
2164. SAHA) in patients with advanced multiple myeloma. Leuk.
231. Baz, R.C., K.H. Shain, M.A. Hussein, et al. 2014. Lymphoma 49: 502–507.
Phase II study of pegylated liposomal doxorubicin, low- 242. Vesole, D.H., E. Bilotti, J.R. Richter, et al. 2015. Phase I study
dose dexamethasone, and lenalidomide in patients with of carfilzomib, lenalidomide, vorinostat, and dexametha-
newly diagnosed multiple myeloma. Am. J. Hematol. 89: sone in patients with relapsed and/or refractory multiple
62–67. myeloma. Br. J. Haematol. 171: 52–59.
232. Michael, M., I. Bruns, E. Bölke, et al. 2010. Bendamustine 243. Yee, A.J., P.M. Voorhees, W. Bensinger, et al. 2014. Ricoli-
in patients with relapsed or refractory multiple myeloma. nostat (ACY-1215), a selective HDAC6 inhibitor, in com-
Eur. J. Med. Res. 15: 13–19. bination with lenalidomide and dexamethasone: results of
233. Wood, W.A., J. Whitley, D. Moore, et al. 2011. Chemomo- a phase 1b trial in relapsed and relapsed refractory multiple
bilization with etoposide is highly effective in patients with myeloma. Blood 124: 4772.

Ann. N.Y. Acad. Sci. 1364 (2016) 32–51 


C 2016 New York Academy of Sciences. 51

You might also like