Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Jan 1.
Published in final edited form as: Ann N Y Acad Sci. 2016 Jan;1364(1):32–51. doi: 10.1111/nyas.13038

Multiple myeloma in the marrow: pathogenesis and treatments

Heather Fairfield 1,a, Carolyne Falank 1,a, Lindsey Avery 2, Michaela R Reagan 1,2
PMCID: PMC4806534  NIHMSID: NIHMS761730  PMID: 27002787

Abstract

Multiple myeloma (MM) is a B-cell malignancy resulting in osteolytic lesions and fractures. In the disease state, bone healing is limited due 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 due to 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 MGUS 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

Introduction

Multiple myeloma (MM) is a fatal, malignant B-cell neoplasm characterized by uncontrolled, destructive growth of mutated plasma cells within the bone marrow (BM). Patients over age 65 are most commonly affected by this disease1 and, as indicated by its name, MM is characterized by dissemination of multiple tumor cells throughout the BM. A mnemonic sometimes used for the common MM pathologies is CRAB: C (calcium, elevated), R (renal failure), A (anemia), B (bone lesions). Additionally, a hallmark of MM is heterogeneous chromosomal aberrations and numerous mutations in a range of genes, both of which make the disease very difficult to target therapeutically.2

MM begins as monoclonal gammopathy of undetermined significance (MGUS), progresses to smoldering (asymptomatic) myeloma and finally becomes overt (symptomatic) myeloma, resulting in BM infiltration and osteolytic lesions. During MM progression the normal equilibrium between osteoblastic (bone building) and osteoclastic (bone breakdown and resorption) activity is skewed toward net bone loss. MM-induced osteolysis releases growth factors embedded inside the bone matrix, fueling MM progression and expansion in the BM niche and resulting in greater osteoclastic activity in a process known as the vicious cycle.3,4 During this MM-induced cycle, osteoclastic bone breakdown releases bone-embedded growth factors to promote tumor growth which, in turn, stimulates greater osteoclast activity. MM progression and drug resistance depend on interactions with the local microenvironment; therapies that target the marrow microenvironment rather than the tumor cells directly, such as bisphosphonates, have proven to be effective in inhibiting tumor growth and osteolysis, suggesting that targeting of the marrow can be an effective therapeutic avenue to pursue.510 In addition to osteoblasts and osteoclasts, other BM niche cells, including mesenchymal stem cells (MSCs), BM adipocytes, immune cells, and osteocytes, also likely affect MM progression.7 BM cells exist in a dynamic, stress-sensing environment where they release soluble signaling molecules to communicate with one another; how these signals are processed by MM cells invading the BM niche remains a key area of investigation.

Recent advances in genetic technologies have provided additional insight into mutations and chromosomal abnormalities associated with MM.11 We now know that the majority of MM cases are associated with an initiating somatic mutation and are frequently accompanied by additional oncogenic mutations. This genetic complexity makes the treatment of MM problematic. Thus far, MM treatments primarily rely on chemotherapies in conjunction with proteasome inhibitors,12,13 anti-resorptive agents such as bisphosphonates14, corticosteroids and bone marrow transplantation.1214 Immunostimulatory agents are also being investigated as potential treatments for MM; however, a better understanding of the MM BM niche and cell-cell interactions is crucial in providing novel, more effective treatments for MM patients. This review aims to describe the nature of somatic mutations as they relate to MM disease prognosis. It also outlines the interactions of MM with the various cell types in the BM niche and describes current treatments as they relate to microenvironmental components.

Epidemiology, cytogenetics, and risk stratification in MM

Over 22,000 new cases of MM were diagnosed in 2013, and the number of cases of MM increases steadily each year.15 It appears that nearly all patients have MGUS during the 2 years before they develop MM, most patients have MGUS 4 years before MM, and ~82% of MM patients have MGUS 8 years before MM develops.16 Other known risk factors for MM include being of male sex, older age, and African American racial background.17 However, as the number of MM cases continues to increase annually,1 stratification of secondary risk factor assessment to encourage early diagnosis will become clinically significant. These secondary risk factors include exposure to chronic, low-grade inflammation, pre-existing and/or chronic immunodeficiency, and, potentially, the presence of known inflammatory diseases or conditions (eg. obesity, cardiovascular disease, or type II diabetes mellitus).1,1820 Of growing interest are the pathogenic links between MM and obesity-related chronic, low-grade inflammation, BM/tumor microenvironment hypoxia, and genetic instability.2123 Research into how each of these components support malignant transition is ongoing.24,25

Overt MM is often characterized by mutations in KRAS (particularly in previously treated patients), NRAS, BRAF, FAM46C, TP53, and DIS3. Often mutations affecting proteins within the same pathway are also observed (e.g., KRAS, NRAS and BRAF).26 Common mutations include loss at 1p and inactivation of the tumor suppressor p53, resulting in an abundance of immunoglobulin production.11 Somatic mutations associated with the development of MM can be characterized as either non-hyperdiploid disease (NHD) or hyperdiploid disease (HD).27 Both classes of mutations involve large chromosomal structural events which result in the dysregulation of G1/S cell cycle and cyclin D gene transcription.11,28,29 NHD is characterized by chromosomal translocations at 14q32, including t(4;14), t(11;14), t(14;16), and t(14;20), which involve early translocation of immunoglobulin heavy chain (IgH) genes.30 This results in increased expression of IgH in the clinical presentation of MM.11,28,29 NHD is linked with an increased likelihood of disease progression and poor overall prognosis.11,28,31 By contrast, HD is characterized by the presence of trisomies, specifically chromosomes 3, 5, 7, 9, 11, 15, 19 and 21. Excess cyclin D in the absence of elevated IgH is the main signature of HD.1 Additional cytogenetic aberrations in MM patients with refractory disease include 13q deletion and 1q21 gains in approximately 47.4% and 52.2% of MM patients, respectively.29

MM may also be characterized through molecular genetic (gene expression) profiling.2 High-throughput sequencing of MM patient samples has identified novel driver mutations beyond classical cancer mutations (NRAS, KRAS, and TP53). These include mutations in transcription factors BRAF, FAM46C, DIS3, XBP1, IRF4, and PRDM1, as well as various histone modifying enzymes MLL, MLL2, MLL3, UTX, MMSET (WHSC1), WHSC1L1, and recent inference toward HOXA-9.30 Whole genome sequencing of MM patient samples from the COSMIC database also identified mutations in AFF1, c12orf42, CSMD3, LRRC4C, PCDH7, PTPRD, PPFIBP1, RB1, and ZKSCAN3.32,30 Identification and investigation of these mutations may aid in the diagnosis of MM cases and potentially identify patients who may be genetically predisposed or susceptible to development of this disease.

MM clonality and tumor heterogeneity also limit the identification of effective therapeutic drugs that target this array of mutations. Secondary genetic mutations are associated with, and thought to determine, the longevity of mutated MM cells, where cells with the greatest resilience and survival likelihood are selectively expanded.2 Notably, intraclonal heterogeneity is considered to be non-uniform among individual cases as well as within MM cell populations,2,5 presenting a particular clinical challenge in the context of chemosensitivity and drug resistance. The focus of a substantial amount of research effort is therefore aimed at identifying critical BM microenvironmental and systemic variables contributing to intraclonal, heterogeneic malignancy and subsequent refractory disease.

While the above mutations and phenotypic correlations have been identified as primary risk factors for MM, additional risk stratification has been implemented in populations with MGUS. Risk factors for the progression of MGUS to MM include: 1) increased serum myeloma protein (M-protein); 2) type of M-protein present (e.g., IgG, IgM, IgA)33; 3) abnormal ratio of free kappa to lambda light chain immunoglobulins (<0.26, >1.26);33 4) presence of >95% (clonal) plasma cells present in the BM;34 and 5) presence of circulating plasma cells.16,35 In patients with MGUS, the presence of three or more risk factors is considered “high risk” (approximately 58% 20-year progression within MGUS patient population), two risk factors represents “high-intermediate risk” (37%), one risk factor is “low-intermediate risk” (21%) and the absence of risk factors is considered “low risk” (5%).16 Approximately 3% of the adult population over the age of 50 are expected to have MGUS.16 However, due to the absence of symptoms and the lack of standardized MGUS/MM screening procedures, MGUS is often under-diagnosed. Progression of MGUS to MM is discussed in further detail throughout the subsequent sections.

Pathogenesis of MM

The progression of MM begins with the asymptomatic, precursor pathogenic state of MGUS. Evidence indicates that MGUS, previously characterized by myeloma cell growth without bone destruction or other organ involvement, is in fact associated with alterations in the bone. Epidemiologic evidence has shown that patients with MGUS suffer from a significantly increased fracture risk, and that the prevalence of MGUS is increased in patients with osteoporosis.36 Recent findings by Drake et al. have demonstrated that the onset of MGUS is concurrent with the deterioration of both auxiliary and appendicular microarchitecture leading to skeletal fragility.36 The relationship between bone loss in MGUS and the progression to MM is an area of current interest, as a correlation may suggest that treating bone loss in MGUS could not only decrease fracture risk in these patients, but could also delay the onset of MM, a theory currently under investigation in our labs and others.

In the absence of clinical symptoms, MGUS is diagnosed by quantifying the amount of immunoglobulin present in both the bloodstream and BM, specifically with a plasma cell population of <10% in the BM. With a plasma cell content exceeding 10%, the disease state transitions into either smoldering MM (SMM) or MM (if clinically manifested).16 MGUS progresses to MM at a rate of 1–2% of patients per year and this transition is likely influenced by the presence of mutational diversity or clonality of MM cell populations as well as changes in the local BM and other systemic factors.17,31,37,38 MM cells are thought to initially create a plasmacytoma, a single tumor, and then develop into multiple lesions to form the disease of multiple myeloma.39 Smoldering multiple myeloma (SMM) is an intermediate clinical stage between MGUS and MM. It is classified as having high serum or urinary monoclonal protein as well as clonal BM plasma cells in the range of 10–60%, in the absence of additional myeloma-defining events40 such as hypercalcaemia, renal insufficiency, anemia, or bone lesions.41

With the evasion techniques of clonal evolution and drug resistance, MM may progress to an aggressive, bone-marrow independent disease known as plasma cell leukemia (PCL).23,37 In PCL, MM cells proliferate and spread into circulation, causing an increase in plasma cells (≥20%) in the blood and often creating plasmacytomas at other places outside the BM throughout the body.42 Efforts to fully understand the pathogenesis of both precursor, symptomatic, and terminal-stage MM should aim to identify mechanisms that cause somatic mutations, drug resistance, immune evasion, or relapse as potential drug targets. Treatments that interfere with MM growth and osteolysis in the bone marrow microenvironment (BMM) to slow disease progression and improve patient quality of life and life expectancy are already in use with additional treatments in development.43

Pathogenesis of MM within the bone marrow microenvironment

The contributions of the bone marrow microenvironment (BMM) to MM are a focal area of research. The BMM is a highly dynamic niche, capable of renewing damage and responding to systemic energy levels, inflammatory mediators, and endocrine signals.4447 The BMM is a primary modulator of both malignant transformation and MM disease progression.6,48 Along with activated inflammatory agents, reactive oxygen species (ROS) and reactive nitrogen intermediates (RNI) are also present in the hypoxic state within the BMM.4951 Properties of the BMM allow for infiltration, growth, proliferation, adhesion, and migration of MM cells, while providing the structural and nutritional sustenance to harbor quiescent, drug-resistant MM cells.2 In addition to providing an optimal substrate for MM initiation and progression, the BMM can also provide activated inflammatory agents including cytokines, chemokines, adipokines (e.g. adiponectin and leptin), and growth factors (e.g. IL-6, IGF-1, VEGF, TNF-α, and SDF-1) secreted by macrophages, neutrophils, and other cells in the BM. These factors then support malignant cell growth, drug resistance and cytotoxicity of healthy cells.23,52

Interestingly, ROS formation via cells in the BM niche has been implicated as a potential primary, tumor-initiating event in leukaemogenesis in mice,53 demonstrating the complexities of understanding oncogenesis in the BMM. Additionally, other BM inflammatory components such as SDF1, a chemoattractant for CXCR4, expressed on MM cells, are also known to promote MM tumor cell BM homing and engraftment;23 increased expression of CXCR4 in clonal cells supported epithelial-to-mesenchymal transition (EMT)-like transcriptional patterns.23,54 A hypoxic microenvironment can further enhance the genomic instability for cells within the BM, select for dormant clones, and also contribute to MM cell drug resistance.23,55,56 Still, the effects of hypoxia on MM remain under investigation as novel model systems to measure or induce hypoxia and quantify cellular responses in the BMM are being engineered and optimized.

The tumor microenvironment is ever evolving in response to changes in the molecular biochemistry, genetic profile, and diversity in type and number of cell populations. Tumors and the surrounding microenvironment are thought to communicate in a highly bi-directional, parasitic manner, each with the potential to alter inherent characteristics of the other.57 A typical MM tumor microenvironment contains several cellular mediators/cell subtypes, including MM mesenchymal stromal cells, osteoblast (OB) and osteoclast (OC) bone cells, BM adipocytes, and a variety of immunomodulatory cell types (e.g., macrophages, NK cells, regulatory T cells etc.). This type of diverse microenvironment contributes to the dynamic characteristics of healthy BMM as well as to the growth and survival of a tumor.6 The roles of these cell types in MM progression are outlined below.5

Mesenchymal stem cells

Mesenchymal stem cells (MSCs) are of key importance in the BM as they serve as the common progenitor to a variety of BMM cells including osteoblasts (OBs), osteocytes (OCs), and adipocytes. MSCs found in BM are a dynamic cell type with many capabilities including self-renewal, differentiation, cell signaling, injury- and tumor-homing, and immunomodulation.7,58 MSC function is highly influenced by the surrounding BMM, and is thus altered in many disease states.6 MSCs from MM patients (MM-MSCs)7 interact with MM tumor cell clusters,6,59 and are altered by this interaction but are not genetically compromised. MM-MSCs have also been shown to produce exosomes that specifically contain proteins and microRNAs that promote tumor growth. The role of pharmacological treatments in targeting MM-MSCs are under current investigation, as previously reviewed.60

Twenty years ago, it was first shown that MM-MSCs were altered and “hijacked” by myeloma cells to change their expression of certain cytokines (TGF-β1, MIP-1β, and IL-7);61 this led to a feedback loop enhancing tumor growth by inhibiting pre-B cells to create a more immunosuppressed environment for MM cells to grow. Since then, many abnormalities in MM-MSCs in terms of gene and microRNA expression have been found, some of which enhance drug-resistance of MM cells in the BMM or contribute to decreases in osteoblastic activity.62 MM-MSCs have been shown to induce bortezomib resistance in MM cells through secretion of IL-8 and other factors that increase MM NF-κB signaling.63 IL-6, IGF-1, VEGF, TNF-α, SDF-1 and BAFF constitute other stromal elements that promote the survival, migration, and drug resistance of MM cells.64 IL-6 is of particular importance, as it acts both in paracrine and autocrine manners65 and targeting MSC-derived IL-6 using siRNAs has recently been proven effective pre-clinically to inhibit MM cell growth.66

Osteoblasts

Osteoblasts (OBs) line the bone surface in basic multicellular units (BMUs), separated from the BM by a layer of canopy cells in what has been termed a “bone remodeling compartment (BRC)” (in the trabeculae) or behind a cutting zone led by osteoclasts (in the cortex).67 Within this compartment, osteoblasts produce and mineralize bone osteoid as osteoclasts resorb bone.68 In healthy conditions, once activated by coupling-factors released during osteoclast maturation and activity, osteoblasts mineralize and rebuild healthy bone matrices.69 In MM, osteoblast growth is significantly repressed, resulting in unbalanced osteoclastic bone resorption and abnormal bone turnover.4,6 The role of osteoblasts in MM and MM drug resistance is still unclear, but osteoblasts appear to suppress MM growth or induce dormancy via paracrine signaling.7073 Decorin is one proteoglycan produced by osteoblasts that has been demonstrated to directly inhibit growth and survival of MM cells in vitro by inducing apoptosis and subsequent arrest of MM cell cycle.6,74,75 By contrast, osteoblasts may also promote growth and survival of MM cells through the production of various growth factors or providing a niche to harbor quiescent MM cells, inducing chemotherapy resistance. Factors such as osteocalcin, osteopontin, FGF and TGFβ, which are synthesized and secreted by osteoblasts, have been identified as factors which modulate the growth and survival of MM cells.6 Direct osteoblast support, inhibition, or effects on drug resistance of MM cells may be dependent on the characteristics of the surrounding BMM. More remains to be examined in this realm, and three-dimensional models of osteoblasts and MM cells may help us explore these interactions in a more physiologically relevant model than possible in two-dimensional co-cultures.6,70,76

Osteoclasts

Osteoclasts (OCs), the major target of bisphosphonate therapies, are the primary mediators of bone resorption in both healthy and pathological bone turnover.4,76 Clinically, the presence of lytic bone lesions in MM manifests with severe bone pain, pathological fractures, increased bone turnover serum markers, and hypercalcemia.6 In MM, OC’s are increasingly prominent in the BMM due to their activation by various cytokines and signaling pathways.7779 With excessive bone resorption overpowering the protective “rebuilding” effects of OBs, the integrity of bone structure becomes compromised by the presence of lytic bone lesions. Pathological lysis of bone tissue activates a vicious cycle, resulting in the ongoing release of calcium, growth factors, and ECM proteins, all of which enhance tumor growth and survival.80 Osteoclasts may also reactivate dormant MM cells by remodeling the endosteal niche, releasing MM cells from their physical and temporal hibernation.73 Moreover, osteoclasts, as well as stromal cells, have been shown to protect MM cells from dexamethasone-induced apoptosis.81 Osteoclasts also support angiogenesis through secretion of proangiogenic factors,82 hence, targeting osteoclasts through bisphosphonates is efficacious through numerous avenues.

The RANK/RANKL/OPG signaling pathway, deregulated in MM, is one of the important regulatory pathways involved in maintaining the balance between bone remodeling and resorption.8385 Receptor activator of NF-κB ligand (RANKL) is produced by osteoblast-progenitor cells as well as MM cells, and signals by binding to its receptor on the surface of the osteoclast-progenitors to stimulate osteoclast differentiation. RANKL also binds osteoprotegerin (OPG) which inhibits RANK/RANKL signaling and, in turn, osteoclastogenesis.76 In metastatic bone disease, such as MM, this system is dysregulated, leading to increased osteolysis and bone resorption. MM cells directly stimulate osteoclastogenesis through their own production of RANKL but also indirectly manipulate this system by driving increased RANKL expression and decreased OPG in both BM stromal cells and osteoblast.76 Denosumab, a RANKL neutralizing antibody, was found to be non-inferior and trending towards superior versus zoledronic acid (a bisphosphonate) in one randomized, double-blind study in delaying time to first on-study skeletal related event (SRE) in the treatment of bone metastases in patients with advanced cancer (excluding breast and prostate cancer) or multiple myeloma.86 However, overall survival and disease progression were similar between groups and hypocalcemia occurred more frequently with denosumab. Another clinical study, in the recruiting stage, will compare denosumab to zoledronic acid in the treatment of bone disease in subjects with MM.

Osteocytes

A subset of osteoblasts that slow in their building bone activities become buried by their more active osteoblast neighbors until they are fully embedded in bone matrix and differentiate into osteocytes. Osteocytes sit within lacunae, tiny crevices within bone, and make up ~95% of bone cells.87 These cells regulate the response of bone to mechanical stress through the secretion of factors including sclerostin and RANKL into the BMM to modulate osteoblast and osteoclast activity.8890 Sclerostin, a Wnt inhibitor,91 decreases OB differentiation from MSCs,92,93 and is also secreted from some MM cells to suppress OB differentiation in vitro.94 Interestingly, osteocyte number is decreased and osteocyte apoptosis is increased in MM,95 while circulating sclerostin levels are positively correlated with bone disease severity in MM patients.96 Sclerostin is also upregulated in the BMM of MM patients versus healthy people and sclerostin is overexpressed by plasma cells from MM patients.97,98 These findings implicate sclerostin as a novel target corrupting the BMM, skewing the balance away from osteoblastogenesis and toward osteolysis.

Adipocytes

In addition to bone cells, adipocytes in the BMM also interact with MM cells. In the classical model of lineage commitment, adipocytes arise from MSCs and are the major cell type found in adipose tissue. Lipid-laden adipocytes serve as an energy reserve in the BM and also serve an endocrine function through the expression and secretion of hormones known as adipokines (e.g., leptin and adiponectin), and growth factors (e.g., IL-6, TNF-α, MCP-1, and insulin), which may promote myelomagenesis or disease progression.18 Bone marrow adipose tissue (BMAT) has been characterized as yellow adipose tissue, which has gene expression patterns that overlap with both white and brown fat.99 This suggests that BMAT can serve as an important regulator of energy maintenance in the BM niche during times of stress.99 Clinical studies have suggested an association between BMAT and an increased likelihood of MM100,101 but the direct relationship and potential mechanisms remain largely unexplained. Adipocytes isolated from MM patient femoral biopsies can support myeloma growth in vitro,102 and a recent study suggests that they may protect MM cells from chemotherapy-induced apoptosis.103

Obesity has been established as a physiological risk factor for MM development, and obese individuals have shown a 1.5–2 fold elevated risk for developing MM when compared to individuals of normal weight.18,22 Circulating adiponectin, which is inversely proportional to fat mass,104 has been shown to inhibit proliferation of MM and reduce tumorigenic angiogenesis.105,106 Inhibition via adiponectin occurs through several signaling pathways, including cyclic AMP-dependent protein kinase A (PKA), signal transducer and signal activator 3 (STAT3), mitogen-activated protein kinase (MAPK), β-catenin, and phosphatidylinositol 3-kinase (PI3K/AKT).106 This results in the anti-proliferative and anti-tumorigenic effects seen in MM, breast, prostate, colon, and liver epithelial cell cancers.18,106 From a mechanistic perspective, deficiency in adiponectin (as seen in obesity) limits the biological actions of several signaling pathways that are essential to preventing growth, proliferation, migration, and drug resistance in MM cells.22 Additionally, leptin may play a role in the transition from MGUS to MM through the regulation of the osteoprotegrin/RANKL signaling pathway. Elevated serum leptin levels have been documented in MM patient populations when compared to healthy individuals.107,108 Together these results suggest that adipocytes may support advancement of MM in the BM niche; however, additional studies are required to evaluate this hypothesis.

Immunomodulatory factors

In addition to osteoclasts, other HSC-derived cells populate the BM niche, including tissue-specific macrophages known as osteomacs and other immune cells.48 Osteomacs are responsible for the regulation of HSCs in the BM niche by directing their homing and colonization, as well as their transition between active stem cell status and dormancy.109 Other immune cells that populate the BM include additional macrophages, neutrophils, and myeloid-derived suppressor cells, all of which may respond to stress, affect tumor growth, and affect bone turnover and other BM cells.110,111 Regulatory T cells in the BM niche create an immune-privileged112 or immunosuppressive environment that can be corrupted by the colonization of foreign entities including cancer cell types.48,111 Importantly, immune cells in the microenvironment can be modulated by the tumor to provide favorable conditions for MM tumors. Specifically, myeloid-lineage progenitor cells have been demonstrated to support tumor cells both in vitro and in vivo.113,114 Immune cell populations are often skewed by MM cells, with increased regulatory T cells (immune-inhibiting) and decreased activity and/or number of effector T cells (tumor killing) present in the BM.115 Upon tumor invasion, mononuclear cells begin to produce and secrete VEGF, which stimulates mobilization of endothelial cells and subsequent tumor vascularization.116

Endothelial cells

The BMM is conducive to MM cell homing and proliferation in part due to unique BM endothelial cells that express adhesion proteins that specifically enable the rolling and intravasation of MM cells through fenestrated BM capillaries.57 Angiogenic cytokines and growth factors are primarily modulated through BM stromal cells.6,54 Endothelial cells and endothelial progenitor cells, which are associated with BM stromal cells, mobilize in response to microenvironment stress, such as MM invasion, to stimulate vasculogenesis of the BM niche.117 In addition to stimulating VEGF secretion from mononuclear cells, tumor cells also secrete extremely high amounts of VEGF to stimulate the production and release of IL-6 from endothelial cells and BM stromal cells.116 This creates a feedback stimulus of IL-6 signals from the endogenous endothelial cells to increase VEGF, indirectly promoting vascularization of MM-infiltrated bone. Studies suggest that vasculogenesis as mediated by endothelial and endothelial progenitor cells may represent an early disease event that takes place during the transition from either MGUS or smoldering MM to overt MM.118

Treatments

While MM remains incurable, new therapies have substantially improved patient quality of life and survival rates. Treatment of MM has substantially progressed in the last decade, with the development of novel pharmaceutical agents and the use of bisphosphonate drugs in conjunction with autologous stem cell transplantation (ASCT). New approaches have focused on the co-treatment of specific proteasome inhibitors (PIs) with immunomodulatory drugs (IMiDs), the use of monoclonal antibodies, and histone deacetylase inhibitors. Nonetheless, chemotherapy and steroids remain effective treatments for most MM patients. Highlighted below and in Table 1 is a compilation of the most common treatments currently used for MM.

Table 1.

Current treatments used to treat multiple myeloma

Drug Type of drug Clinical trial phase References and/or trial
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.227
CARF PI FDA approved
CARF/LEN/DEX PI/IMiD/steroid III Stewart et al.228; 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 Pre-clinical Hurchla et al.229
Ixazomib/LEN/DEX PI/IMiD/steroid III NCI-2012-02752,
2011-005468-10, C16011-CTIL,
NL41603.028.12, U1111-1164-7621,
NCT01659658
Delazomib PI I Gallerani et al.230
Melphalan/THAL/PRED/DEF Chemo/IMiD/steroid/antic I/II Palumbo et al.231
Vincristine Chemo III Kyle et al.232
CYCLO/THAL/DEX Chemo/IMiD/steroid Garcia-Sanz et al.127
DOX/LEN/BORT Chemo/IMiD/PI I/II Jakubowiak et al.168
Liposomal DOX/DEX/LEN Chemo II Baz et al.233
Bendamustine Chemo I Michael et al.234
Etoposide Chemo Pre-clinical Wood et al.235
LEN IMiD FDA approved
POM IMiD FDA approved
POM/DEX IMiD/steroid III San Miguel et al.205
ELO IMiD FDA approved Starr236
LEN/BORT/DEX/ELO IMiD/PI/steroid/Ab I/II Usmani et al.237
Daratumumab Ab FDA approved
BI-505 Ab I/II Hansson et al.238
Rituximab Ab II Baz et al.239; Bergua et al.240; Korte et al.241
SAHA HDACi I Mitsidades et al.242; Richardson et al.243
LBH589 HDACi III San-Miguel et al.207
AR-42 HDACi Pre-clinical Canella et al.200
Vorinostat HDACi I Vesole et al.244
Ricolinostat (ACY-1215) HDACi Ib/II Yee et al.245

BORT, bortezomib; LEN, lenalidomide; DEX, dexamethasone; CARF, carfilzomib; POM, pomalidomide; ELO, elotuzumab; Ab, antibody; PI, proteasome inhibitor; IMiD, immunomodulatory drug; HDACi, histone deacetylase inhibitor; Chemo, chemotherapy; CYCLO, cyclophosphamide; DOX, doxorubicin; PRED, prednisone; DEF, defibrotide; antic, anticoagulant

Chemotherapy

MM is often treated with classical chemotherapies to target and destroy cancer cells.95 Chemotherapeutics commonly used for MM include: melphalan, vincristine, doxorubicin and liposomal doxorubicin.119 These are used primarily in the treatment of overt MM, as patients with asymptomatic, smoldering MM do not benefit from chemotherapy.16,120 Chemotherapeutics are often used in conjunction with ASCT, which is able to both replenish stem cells and potentially induce BM remodeling.121 This treatment combination has been shown to increase progression-free survival and overall survival in MM patients, and may be used in the treatment of relapsed, refractory MM.122,123 Additional combination treatments that utilize the cancer targeting aspects of chemotherapy drugs as well as agents that target and modulate bone remodeling such as proteasome inhibitors and bisphosphonates are now used to treat MM. Specifically, doxorubicin has been combined with the proteasome inhibitor, bortezomib, to treat patients with refractory MM. This dual treatment resulted in a greater myeloma-suppressive effect than bortezomib alone.124,125 Combination therapies utilizing chemotherapy and bisphosphonates have response rates as high as 80% in newly diagnosed patients126, and up to 75% in patients with relapsed disease.127

Proteasome inhibitors

Proteasome inhibitors such as bortezomib or the second-generation carfilzomib block the proteasome, leading to the accumulation of proteins in MM cells, causing their destruction.128,129 Specifically, bortezomib has been shown to promote apoptosis in MM cells as well as osteoclasts (attenuating bone loss) and induce osteoblast differentiation80,130,131 Bortezomib has been shown to enhance the activity of certain concurrent therapies (e.g., lenalidomide and dexamethasone) and is often prescribed in combination with other anti-MM drugs due to its chemosensitizing effects.132138 However, there are clinical challenges associated with bortezomib therapy including drug resistance and off-target effects including peripheral neuropathy.139,140 Carfilzomib has been demonstrated to also be effective when used with lenalidomide and dexamethasone by significantly reducing the minimal residual disease in patients that are newly diagnosed with MM.141143 Due to the effectiveness of bortezomib, next-generation proteasome inhibitors are currently being developed (e.g., oprozomib, ixazomib, marizomib, and delanzomib). Many of these new proteasome inhibitors are being used after treatment with bortezomib, where MM progresses to bortezomib resistance or becomes relapsed and refractory MM.144

Bisphosphonates

Bisphosphonates are a class of drugs that inhibit resorption of bone by blocking osteoclasts and decrease osteolytic bone pain.145,146 It is well known that MM is able to increase the number of OCs within the BMM,147,148 leading to bone pain, hypercalcemia, and increased bone fractures in patients.149151 Therefore treatment with bisphosphonates is commonly used in conjunction with proteasome inhibitor drugs152154 or chemotherapy to kill tumor cells and inhibit bone destruction.155,156 Several studies suggest that bisphosphonates may not have direct anti-tumor effects, despite their bone-protective effects.157159 Specifically, nitrogen-containing bisphosphonates alendronate, pamidronate and zoledronate are all FDA approved and are often administered as a crucial aspect of MM patient pain and bone disease management.160

Immunomodulatory drugs and monoclonal antibodies

Endogenous immunomodulatory actions and/or immunodeficiency can significantly impact the dynamics of the BMM to either promote or suppress MM tumor cell growth.23,115 Recent drug therapies for many cancer types, including MM, continue to support a paradigm shift in using an immunomodulatory approach for the management and treatment of malignant disease. As recently reviewed by Kawano et al., MM tumor cells can induce pronounced immunosuppression both systemically as well as locally at the BMM.115 Immunodeficiency in the BMM is characterized by an increase in immunosuppressive cell populations, including regulatory T cells and myeloid derived suppressor cells (MDSCs).161 In the pathogenesis of MM, the ability of MDSCs to differentiate into immune-protective macrophages and granulocytes is inhibited.115,162 Thus, restoration of tumor-suppressor immunomodulation is a crucial component in the development of anti-MM drug therapies.

Lenalidomide and pomalidomide, second generation immunomodulatory drugs (IMiDs), enhance the sensitivity of MM cells to both bortezomib and dexamethasone. They also promote the tumor suppressor actions of host immune defense mechanisms.163 For this reason, multiple combination therapies are often utilized in clinical practice to improve treatment response by delaying progression or recurrence and improving symptom management.164168 A Phase 3 trial found pomalidomide therapy combined with low-dose dexamethasone to be safe and effective in patients with relapsed MM disease, who had previously been treated with bortezomib or thalidomide.169 Richardson and colleagues have recently published Phase 2 clinical trial outcomes exploring the maximum tolerated dose (MTD) of lenalidomide in patients with relapsed or refractory MM disease.165 Findings from this investigation revealed that the MTD of lenalidomide (15 mg/day) administered alongside low-dose dexamethasone (40 mg/week) on the first 21 of 28 total days of treatment was tolerable and resulted in favorable results in response and 2-year survival rates.165 Thalidomide, the first generation IMiD, is still commonly used for managing MM by inhibiting angiogenesis, inducing apoptosis of MM cells, and inhibiting the secretion of IL-6 and VEGF by MM cells during adhesion to BM stromal cells.170 Like other IMiDs, thalidomide is currently being used with PIs (bortezomib or carfilzomib) and corticosteroids (e.g., dexamethasone, prednisone, prednisolone, and methylprednisone).171175 Developments in MM drug therapies will likely continue to maximize selectivity through antibody-mediated mechanisms combined with proteasome inhibitors, IMiDs, bisphosphonates and traditional chemotherapy.117,161

Monoclonal antibodies are increasingly being utilized to treat myeloma patients, both to target the myeloma cells directly and to modulate the immune response and microenvironment.176 Recent work with daratumumab, a human monoclonal antibody that binds CD38,177 has shown target-cell killing of tumor cells.178 CD38 is a transmembrane glycoprotein that is highly expressed by myeloma cells, making this a desirable, myeloma-specific target.179 Importantly, single agent daratumumab treatment, which has recently been approved by the FDA, was effective in patients with refractory disease, including those who had previously been treated with lenalidomide and bortezomib.178 Radioimmunotherapy (RIT), which combines radiation with immunotherapy, is in preclinical stages as a treatment for MM. Perhaps the most promising RIT in respect to MM treatments is the use of indatuximab ravtansine, an antibody targeting CD138 antigen, which is expressed in greater than 95% of MM cells.180,181 Recent work has shown that coupling this antibody to radioactive isotopes significantly increased survival in animal models of MM.181 The targeting of antigens and other cell-surface proteins in the treatment of cancers is rapidly expanding through the use of antibody-drug conjugates, which combine the specificity of a monoclonal antibody with a small anti-cancer drug molecule. Pre-clinical in vitro and in vivo work with these conjugates has demonstrated promise in the treatment of MM and other hematological cancers.182

Another promising new approach to MM treatment is the addition of an immunostimulatory agent, elotuzumab, to the now standard treatment of lenalidomide and dexamethasone therapy.1,30 Elotuzumab, a FDA approved humanized immunoglobulin G1 monoclonal antibody, targets signaling lymphocytic activation molecule F7 (SLAMF7).183 SLAMF7, expressed by both natural killer (NK) and MM cells, plays an integral role in the activation of NK cells through binding with the EAT-2 adapter protein.161,184 In MM cells, EAT-2 is significantly under-expressed, resulting in poor SLAMF7 signaling. Elotuzumab is able to reduce tumor burden through the activation of NK cells that induce antibody-mediated apoptosis of MM cells, with little effect on healthy cells.161,184

In addition to bisphosphonates, other bone anabolic and anti-resorption agents such as anti-DKK1, anti-sclerostin, and anti-activin A antibodies are being explored for stimulating bone healing and regrowth in MM patients, and may also have anti-tumor effects.94,9698,185192 Specifically, antibodies used to target and suppress DKK1 in pre-clinical trials have been shown to prevent the development of MM-induced osteolysis and increased bone formation in animal models.193 SOST-neutralizing antibodies (anti-SOST), which have been shown to increase bone formation and bone volume in both experimental models of and patients with osteoporosis, are also under development as a potential treatment for MM.194,195 Two recent studies have indicated the efficacy of anti-SOST antibody to prevent MM induced bone loss and inhibit MM growth in mouse myeloma models.192,196

The use of monoclonal antibodies and their effectiveness to treat MM both in early experimental models and in clinical trials has demonstrated the importance of understanding the molecular mechanisms of MM progression, including MM interaction with other cell types within the BM niche. For more information on the potential of monoclonal antibodies in the treatment of myeloma, please refer to the review by Sondergeld et al.197

Histone deacetylase inhibitors

Epigenetic modifications have been shown to play a major role in cancer development resulting in aberrant histone modifications.198 Histone acetylation is a major regulatory mechanism of gene transcription, where histone deacetylases remove acetyl groups from the histone lysine residues, resulting in repressed transcription.199 Histone deacetylase inhibitors (HDACi) have been shown to inhibit cell growth and induce apoptosis in MM cells as both a single treatment or in conjunction with bortezomib treatment.200,201 Furthermore, HDACi are able to overcome the bortezomib resistance that is commonly seen in MM cells.202 There are several HDACi that are currently under evaluation.203 The first FDA-approved HDAC inhibitor was suberoylanilide hydroxamic acid (SAHA), which is commonly used in combination with bortezomib or lenalidomide.204206 MM treatment with the HDACi vorinostat or panobinostat, with bortezomib or bortezomib/dexamethasone, has been shown to prolong MM patient survival for 0.8 months and 3.9 months, respectively.204,207 Yet, further studies are needed in order to fully elucidate the epigenetic modifications and interactions in MM and the role of epigenetics in MM progression and pathogenesis.

Future directions and conclusions

The pathogenesis of multiple myeloma is influenced by alterations, aberrations, and/or dysregulation in endocrine, vascular, genetic, and metabolic factors. Thus, identifying and targeting the corresponding molecular pathways is crucial in working toward the optimal delivery of individualized pharmacologic therapies in MM. Increases or excess in adiposity may contribute to MM risk via systemic pro-inflammatory effects or BM specific adiposity; this requires further investigation in patient populations as well as in both in vitro and in vivo models. Multiple myeloma is characterized by the presence of osteolytic bone lesions, resulting from prolonged declines in osteoblast-mediated bone mineralization, increases in bone resorption by osteoclasts, and propagation of the vicious cycle.208 While this review has focused mainly on the effects of the BM niche on MM pathogenesis, neuro-endocrine regulation of bone metabolism could also play a role in MM progression. For example, sympathetic nervous system (SNS) fibers, known to innervate cortical bone, modulate a number of outcomes, including regulating homeostatic hormonal control of bone turnover.208 Please refer to the elegant review by Elefteriou et al.209 for more information on the effects of the SNS on bone remodeling in response to many factors including oncogenesis. The complex regulation of MM from multiple systems (the BMM and beyond) makes understanding and combatting disease initiation and progression difficult. Integration of these effects is necessary to provide better therapeutics and work toward a cure.

Myeloma is now being treated by therapies aimed at either directly killing tumor cells or normalizing the aberrant tumor microenvironment. Treatments such as bisphosphonates, which inhibit osteolysis, and IMiDs that stimulate immune cell activation have been implemented with success. As we gain better understanding of the bone, BM, and blood biology disrupted by MM, we are developing safer, more efficacious treatment strategies. Exciting, novel strategies to inhibit the vicious cycle and osteolysis and heal osteolytic lesions based on a better understanding of the basic biology of the disease and the BMM are in development.6,210212 These microenvironment-targeting treatments, in conjunction with ASCT will induce better patient outcomes, and are leading the way in the treatment of relapsed, refractory MM.123 Interesting new directions may target osteocytes and osteoblasts, as we learn more about the roles of these and other cells in the BM.

Novel strategies are also being explored in the preclinical setting targeting NF-κB, JAK, MEK, Pim, PYK2, Src, mTor, PDPK1/RSK2, PERK, and other kinases and pathways.213223 Targeting hypoxia (e.g., HIF1α) and double-strand break repairs are other avenues being explored. Moreover, many novel strategies are being employed using chimeric antigen receptor T cells224226 and other engineered immune responses against MM in preclinical models that may soon make their way to the clinic. Combinatorial strategies, as with all cancers, are inevitably required for the most efficacious treatments. More targetable and prognostically significant genomic alterations are continually arising as our bioinformatics, sequencing, and preclinical validation capacities grow, and high-throughput drug screens are enabling more rapid, accurate identification of novel natural and synthetic anti-MM therapies.

A crucial aspect in the fight against MM and other cancers is the prevention of the disease, an issue arguably underrepresented in cancer research due to challenges in proving effectiveness. The integration of multi-system regulatory factors through the use of better disease models is crucial for continued understanding of MM pathogenesis for better prevention, as well as treatment approaches. In the meantime, more thorough characterization of patient tumors through genetic and epigenetic sequencing, and other profiling techniques, as well as increased sharing of patient data among centers, will provide critical information about potential causations linking systemic health, genetics, epigenetics, and environment, to cancers including myeloma. Lastly, we suggest one of the greatest strategies in accelerating effective myeloma research may be a collaborative, transparent “pre-competitive” space for researchers and pharmaceutical companies alike. We envision that transparent sharing of data and results will push the myeloma research field forward in a novel, patient-centric manner towards a cure, or, more realistically, patient-specific cures. Such a space and cooperative agreement would optimize our global scientific journey towards a greater understanding of this and other diseases, and accelerate research and discoveries relevant for those who rely on rapid development of effective therapies.

FIGURE 1.

FIGURE 1

The bone marrow microenvironment: osteoclastic and vascular niches. Schematic of the bone marrow microenvironment and its role in multiple myeloma. Pathogenesis and progression of 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.

Acknowledgments

The authors thank Dr. Michael Erard, Scientific Editor and Writing consultant at Maine Medical Center Research Institute (MMCRI) for editorial assistance. The authors’ work is supported by Start-up funds, a Pilot Project Grant from NIH/NIGMS (P30GM106391), and the NIH/NIDDK (R24 DK092759-01) at Maine Medical Center Research Institute. The authors’ work is also supported by the Department of Defense (DoD) Visionary Postdoctoral Fellowship Award, through the Peer Reviewed Cancer Research Program, under Award FY14 DoD Congressionally Directed Medical Research Programs 30 No. (W81XWH-13-1-0390); opinions, interpretations, conclusions and recommendations are those of the author and are not necessarily endorsed by the DoD.

Footnotes

Competing interest’s statement

The authors declare no competing interests.

References

  • 1.Alexander DD, Mink PJ, Adami HO, et al. Multiple myeloma: A review of the epidemiologic literature. Int. J. Cancer. 2007;120:40–61. doi: 10.1002/ijc.22718. [DOI] [PubMed] [Google Scholar]
  • 2.Morgan GJ, Walker Ba, Davies FE. The genetic architecture of multiple myeloma. Nat. Rev. Cancer. 2012;12:335–348. doi: 10.1038/nrc3257. [DOI] [PubMed] [Google Scholar]
  • 3.Kingsley LA, Fournier PGJ, Chirgwin JM, Guise TA. Molecular biology of bone metastasis. Mol. Cancer Ther. 2007;6:2609–2617. doi: 10.1158/1535-7163.MCT-07-0234. [DOI] [PubMed] [Google Scholar]
  • 4.Fowler JA, Edwards CM, Croucher PI. Tumor-host cell interactions in the bone disease of myeloma. Bone. 2011;48:121–128. doi: 10.1016/j.bone.2010.06.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Roccaro AM, Sacco A, Maiso P, et al. BM mesenchymal stromal cell-derived exosomes facilitate multiple myeloma progression. J. Clin. Invest. 2013;123:1542–1555. doi: 10.1172/JCI66517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Reagan MR, Liaw L, Rosen CJ, Ghobrial IM. Dynamic interplay between bone and multiple myeloma: emerging roles of the osteoblast. Bone. 2015;75:161–169. doi: 10.1016/j.bone.2015.02.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Reagan MR, Ghobrial IM. Multiple myeloma mesenchymal stem cells: Characterization, origin, and tumor-promoting effects. Clin. Cancer Res. 2012;18:342–349. doi: 10.1158/1078-0432.CCR-11-2212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Alegre A, Gironella M, Bailén A, Giraldo P. Zoledronic acid in the management of bone disease as a consequence of multiple myeloma: A review. Eur. J. Haematol. 2014;92:181–188. doi: 10.1111/ejh.12239. [DOI] [PubMed] [Google Scholar]
  • 9.Pozzi S, Raje N. The role of bisphosphonates in multiple myeloma: mechanisms, side effects, and the future. Oncologist. 2011;16:651–662. doi: 10.1634/theoncologist.2010-0225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Guise Ta, Mohammad KS, Clines G, et al. Basic mechanisms responsible for osteolytic and osteoblastic bone metastases. Clin. Cancer Res. 2006;12:6213–6217. doi: 10.1158/1078-0432.CCR-06-1007. [DOI] [PubMed] [Google Scholar]
  • 11.Furukawa Y, Kikuchi J. Molecular pathogenesis of multiple myeloma. Int. J. Clin. Oncol. 2015;20:413–422. doi: 10.1007/s10147-015-0837-0. [DOI] [PubMed] [Google Scholar]
  • 12.Bergsagel DE. Total body irradiation for myelomatosis. Br. Med. J. 1971;2:325. doi: 10.1136/bmj.2.5757.325-a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Swami A, Reagan MR, Basto P, et al. Engineered nanomedicine for myeloma and bone microenvironment targeting. Proc. Natl. Acad. Sci. U. S. A. 2014;111:10287–10292. doi: 10.1073/pnas.1401337111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Schmiedel BJ, Scheible CA, Nuebling T, et al. RANKL expression, function, and therapeutic targeting in multiple myeloma and chronic lymphocytic leukemia. Cancer Res. 2013;73:683–694. doi: 10.1158/0008-5472.CAN-12-2280. [DOI] [PubMed] [Google Scholar]
  • 15.Siegel R, Naishadham D, Jemal A. Cancer statistics, 2013. CA. Cancer J. Clin. 2013;63:11–30. doi: 10.3322/caac.21166. [DOI] [PubMed] [Google Scholar]
  • 16.Berenson JR, Anderson KC, Audell Ra, et al. Monoclonal gammopathy of undetermined significance: A consensus statement: Guideline. Br. J. Haematol. 2010;150:28–38. doi: 10.1111/j.1365-2141.2010.08207.x. [DOI] [PubMed] [Google Scholar]
  • 17.Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ. Cancer Statistics 2009. CA Cancer J Clin. 2009;59:1–25. doi: 10.3322/caac.20006. [DOI] [PubMed] [Google Scholar]
  • 18.Medina EA, Oberheu K, Polusani SR, Ortega V, Velagaleti GVN, Oyajobi BO. PKA/AMPK signaling in relation to adiponectin’s antiproliferative effect on multiple myeloma cells. Leukemia. 2014;28:2080–2089. doi: 10.1038/leu.2014.112. [DOI] [PubMed] [Google Scholar]
  • 19.Blair CK, Cerhan JR, Folsom AR, Ross JA. Anthropometric characteristics and risk of multiple myeloma. Epidemiology. 2005;16:691–694. doi: 10.1097/01.ede.0000172135.61188.2d. [DOI] [PubMed] [Google Scholar]
  • 20.Renehan AG, Tyson M, Egger M, Heller RF, Zwahlen M. Body-mass index and incidence of cancer: a systematic review and meta-analysis of prospective observational studies. Lancet. 2008;371:569–578. doi: 10.1016/S0140-6736(08)60269-X. [DOI] [PubMed] [Google Scholar]
  • 21.De Pergola G, Silvestris F. Obesity as a major risk factor for cancer. J Obes. 2013;2013:291546. doi: 10.1155/2013/291546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Greenfield DM, Boland E, Ezaydi Y, Ross RJM, Ahmedzai SH, Snowden Ja. Endocrine, metabolic, nutritional and body composition abnormalities are common in advanced intensively-treated (transplanted) multiple myeloma. Bone Marrow Transplant. 2014;49:907–912. doi: 10.1038/bmt.2014.63. [DOI] [PubMed] [Google Scholar]
  • 23.Grivennikov SI, Greten FR, Karin M. Immunity, Inflammation, and Cancer. Cell. 2010;140:883–899. doi: 10.1016/j.cell.2010.01.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Azab AK, Hu J, Quang P, et al. Hypoxia promotes dissemination of multiple myeloma through acquisition of epithelial to mesenchymal transition-like features. Blood. 2012;119:5782–5794. doi: 10.1182/blood-2011-09-380410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Lwin ST, Olechnowicz SWZ, Fowler JA, Edwards CM. Diet-induced obesity promotes a myeloma-like condition in vivo. Leukemia. 2015;29:507–510. doi: 10.1038/leu.2014.295. [DOI] [PubMed] [Google Scholar]
  • 26.Lohr JG, Stojanov P, Carter SL, et al. Widespread genetic heterogeneity in multiple myeloma: implications for targeted therapy. Cancer Cell. 2014;25:91–101. doi: 10.1016/j.ccr.2013.12.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Chng WJ, Santana-Dávila R, Van Wier SA, et al. Prognostic factors for hyperdiploid-myeloma: effects of chromosome 13 deletions and IgH translocations. Leukemia. 2006;20:807–813. doi: 10.1038/sj.leu.2404172. [DOI] [PubMed] [Google Scholar]
  • 28.Chubb D, Weinhold N, Broderick P, et al. Common variation at 3q26.2, 6p21.33, 17p11.2 and 22q13.1 influences multiple myeloma risk. Nat. Genet. 2013;45:1221–1225. doi: 10.1038/ng.2733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Walker Ba, Leone PE, Chiecchio L, et al. A compendium of myeloma associated chromosomal copy number abnormalities and their prognostic value. Blood. 2010;116:56–65. doi: 10.1182/blood-2010-04-279596. [DOI] [PubMed] [Google Scholar]
  • 30.Bergsagel PL, Kuehl WM, Ferrarini M, et al. Decoding the pathophysiology and the genetics of multiple myeloma to identify new therapeutic targets. J. Clin. Oncol. 2013;23:2997. [Google Scholar]
  • 31.Pawlyn C, Melchor L, Murison A, et al. Coexistent hyperdiploidy does not abrogate poor prognosis in myeloma with adverse cytogenetics and may precede IGH translocations. 2015;125:831–841. doi: 10.1182/blood-2014-07-584268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Amend SR, Wilson WC, Chu L, et al. Whole Genome Sequence of Multiple Myeloma-Prone C57BL/KaLwRij Mouse Strain Suggests the Origin of Disease Involves Multiple Cell Types. PLoS One. 2015;10:e0127828. doi: 10.1371/journal.pone.0127828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Rajkumar SV, Kyle RA, Therneau TM, et al. Serum free light chain ratio is an independent risk factor for progression in monoclonal gammopathy of undetermined significance. Blood. 2005;106:812–817. doi: 10.1182/blood-2005-03-1038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Pérez-Persona E, Vidriales MB, Mateo G, et al. New criteria to identify risk of progression in monoclonal gammopathy of uncertain significance and smoldering multiple myeloma based on multiparameter flow cytometry analysis of bone marrow plasma cells. Blood. 2007;110:2586–2592. doi: 10.1182/blood-2007-05-088443. [DOI] [PubMed] [Google Scholar]
  • 35.Kumar S, Rajkumar SV, Kyle Ra, et al. Prognostic value of circulating plasma cells in monoclonal gammopathy of undetermined significance. J. Clin. Oncol. 2005;23:5668–5674. doi: 10.1200/JCO.2005.03.159. [DOI] [PubMed] [Google Scholar]
  • 36.Drake MT. Unveiling Skeletal Fragility in Patients Diagnosed With MGUS: No Longer a Condition of Undetermined Significance? J. Bone Miner. Res. 2014;29:2529–2533. doi: 10.1002/jbmr.2387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Barlogie B, Shaughnessy J, Tricot G, et al. Treatment of multiple myeloma. Blood. 2004;103:20–32. doi: 10.1182/blood-2003-04-1045. [DOI] [PubMed] [Google Scholar]
  • 38.Röllig C, Knop S, Bornhäuser M. Multiple myeloma. Lancet. 2014:385. doi: 10.1016/S0140-6736(14)60493-1. [DOI] [PubMed] [Google Scholar]
  • 39.Lorsbach RB, Hsi ED, Dogan A, Fend F. Plasma cell myeloma and related neoplasms. Am. J. Clin. Pathol. 2011;136:168–182. doi: 10.1309/AJCPENJ68FFBRIYB. [DOI] [PubMed] [Google Scholar]
  • 40.Glavey SV, Ghobrial IM. American Society of Hematology Annual Meeting 2014: highlights in multiple myeloma. Expert Rev. Hematol. 2015;8:273–275. doi: 10.1586/17474086.2015.1029449. [DOI] [PubMed] [Google Scholar]
  • 41.Rajkumar SV, Dimopoulos MA, Palumbo A, et al. International Myeloma Working Group updated criteria for the diagnosis of multiple myeloma. Lancet Oncol. 2014;15:e538–e548. doi: 10.1016/S1470-2045(14)70442-5. [DOI] [PubMed] [Google Scholar]
  • 42.Simeon V, Todoerti K, La Rocca F, et al. Molecular Classification and Pharmacogenetics of Primary Plasma Cell Leukemia: An Initial Approach toward Precision Medicine. Int. J. Mol. Sci. 2015;16:17514–17534. doi: 10.3390/ijms160817514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Podar K, Chauhan D, Anderson KC. Bone marrow microenvironment and the identification of new targets for myeloma therapy. Leukemia. 2009;23:10–24. doi: 10.1038/leu.2008.259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Ecklund K, Vajapeyam S, Feldman Ha, et al. Bone marrow changes in adolescent girls with anorexia nervosa. J. Bone Miner. Res. 2010;25:298–304. doi: 10.1359/jbmr.090805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Rosen CJ, Ackert-Bicknell C, Rodriguez JP, Pino AM. Marrow fat and the bone microenvironment: developmental, functional, and pathological implications. Crit. Rev. Eukaryot. Gene Expr. 2009;19:109–124. doi: 10.1615/critreveukargeneexpr.v19.i2.20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Kawai M, de Paula FJ, a Rosen CJ. New insights into osteoporosis: The bone-fat connection. J. Intern. Med. 2012;272:317–329. doi: 10.1111/j.1365-2796.2012.02564.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Hardaway AL, Herroon MK, Rajagurubandara E, Podgorski I. Bone marrow fat: linking adipocyte-induced inflammation with skeletal metastases. Cancer Metastasis Rev. 2014;33:527–543. doi: 10.1007/s10555-013-9484-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Reagan MR, Rosen CJ. Navigating the bone marrow niche: translational insights and cancer-driven dysfunction. Nat. Rev. Rheumatol. 2015 doi: 10.1038/nrrheum.2015.160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Ibrahim M, Jang M, Park M, et al. Capsaicin inhibits the adipogenic differentiation of bone marrow mesenchymal stem cells by regulating cell proliferation, apoptosis, oxidative and nitrosative stress. Food Funct. 2015;6:2165–2178. doi: 10.1039/c4fo01069h. [DOI] [PubMed] [Google Scholar]
  • 50.Yagi H, Tan J, Tuan RS. Polyphenols suppress hydrogen peroxide-induced oxidative stress in human bone-marrow derived mesenchymal stem cells. J. Cell. Biochem. 2013;114:1163–1173. doi: 10.1002/jcb.24459. [DOI] [PubMed] [Google Scholar]
  • 51.Hole PS, Darley RL, Tonks A. Do reactive oxygen species play a role in myeloid leukemias? Blood. 2011;117:5816–5826. doi: 10.1182/blood-2011-01-326025. [DOI] [PubMed] [Google Scholar]
  • 52.Gilbert LC, Chen H, Lu X, Nanes MS. Chronic low dose tumor necrosis factor-alpha (TNF) suppresses early bone accrual in young mice by inhibiting osteoblasts without affecting osteoclasts. Bone. 2013;56:174–183. doi: 10.1016/j.bone.2013.06.002. [DOI] [PubMed] [Google Scholar]
  • 53.Raaijmakers MHGP, Mukherjee S, Guo S, et al. Bone progenitor dysfunction induces myelodysplasia and secondary leukaemia. Nature. 2010;464:852–857. doi: 10.1038/nature08851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Roccaro AMM, Mishima Y, Sacco A, et al. CXCR4 Regulates Extra-Medullary Myeloma through Epithelial-Mesenchymal-Transition-like Transcriptional Activation. Cell Rep. 2015;12:622–635. doi: 10.1016/j.celrep.2015.06.059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Asosingh K, De Raeve H, de Ridder M, et al. Role of the hypoxic bone marrow microenvironment in 5T2MM murine myeloma tumor progression. Haematologica. 2005;90:810–817. [PubMed] [Google Scholar]
  • 56.Martin SK, Diamond P, Williams SA, et al. Hypoxia-inducible factor-2 is a novel regulator of aberrant CXCL12 expression in multiple myeloma plasma cells. Haematologica. 2010;95:776–784. doi: 10.3324/haematol.2009.015628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.De Raeve HR, Vanderkerken K. The role of the bone marrow microenvironment in multiple myeloma. Histol. Histopathol. 2005;20:1227–1250. doi: 10.14670/HH-20.1227. [DOI] [PubMed] [Google Scholar]
  • 58.Reagan MR, Kaplan DL. Concise review: Mesenchymal stem cell tumor-homing: Detection methods in disease model systems. Stem Cells. 2011;29:920–927. doi: 10.1002/stem.645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Abe M. Targeting the interplay between myeloma cells and the bone marrow microenvironment in myeloma. Int. J. Hematol. 2011;94:334–343. doi: 10.1007/s12185-011-0949-x. [DOI] [PubMed] [Google Scholar]
  • 60.Garcia-Gomez A. Multiple myeloma mesenchymal stromal cells: Contribution to myeloma bone disease and therapeutics. World J. Stem Cells. 2014;6:322. doi: 10.4252/wjsc.v6.i3.322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Tsujimoto T, Lisukov IA, Huang N, Mahmoud MS, Kawano MM. Plasma cells induce apoptosis of pre-B cells by interacting with bone marrow stromal cells. Blood. 1996;87:3375–3383. [PubMed] [Google Scholar]
  • 62.Reagan MR, Mishima Y, Glavey SV, et al. Investigating osteogenic differentiation in multiple myeloma using a novel 3D bone marrow niche model. Blood. 2014;124:3250–3259. doi: 10.1182/blood-2014-02-558007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Markovina S, Callander NS, O’Connor SL, et al. Bone marrow stromal cells from multiple myeloma patients uniquely induce bortezomib resistant NF-kappaB activity in myeloma cells. Mol. Cancer. 2010;9:176. doi: 10.1186/1476-4598-9-176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Kastritis E, Palumbo A, Dimopoulos MA. Treatment of relapsed/refractory multiple myeloma. Semin. Hematol. 2009;46:143–157. doi: 10.1053/j.seminhematol.2009.01.004. [DOI] [PubMed] [Google Scholar]
  • 65.Kawano M, Hirano T, Matsuda T, et al. Autocrine generation and requirement of BSF-2/IL-6 for human multiple myelomas. Nature. 1988;332:83–85. doi: 10.1038/332083a0. [DOI] [PubMed] [Google Scholar]
  • 66.Teoh HK, Chong PP, Abdullah M, et al. Small interfering RNA silencing of interleukin-6 in mesenchymal stromal cells inhibits multiple myeloma cell growth. Leuk. Res. 2016;40:44–53. doi: 10.1016/j.leukres.2015.10.004. [DOI] [PubMed] [Google Scholar]
  • 67.Sims NA, Martin TJ. Coupling the activities of bone formation and resorption: a multitude of signals within the basic multicellular unit. Bonekey Rep. 2014;3:481. doi: 10.1038/bonekey.2013.215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Feng X, McDonald JM. Disorders of bone remodeling. Annu. Rev. Pathol. 2011;6:121–145. doi: 10.1146/annurev-pathol-011110-130203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Andersen TL, Søe K, Sondergaard TE, Plesner T, Delaisse JM. Myeloma cell-induced disruption of bone remodelling compartments leads to osteolytic lesions and generation of osteoclast-myeloma hybrid cells: Research paper. Br. J. Haematol. 2010;148:551–561. doi: 10.1111/j.1365-2141.2009.07980.x. [DOI] [PubMed] [Google Scholar]
  • 70.Chen Z, Orlowski RZ, Wang M, Kwak L, McCarty N. Osteoblastic niche supports the growth of quiescent multiple myeloma cells. Blood. 2014;123:2204–2208. doi: 10.1182/blood-2013-07-517136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Yaccoby S, Wezeman MJ, Zangari M, et al. Inhibitory effects of osteoblasts and increased bone formation on myeloma in novel culture systems and a myelomatous mouse model. Haematologica. 2006;91:192–199. [PMC free article] [PubMed] [Google Scholar]
  • 72.Li X, Pennisi A, Yaccoby S. Role of decorin in the antimyeloma effects of osteoblasts. Blood. 2008;112:159–168. doi: 10.1182/blood-2007-11-124164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Lawson MA, McDonald MM, Kovacic N, et al. Osteoclasts control reactivation of dormant myeloma cells by remodelling the endosteal niche. Nat. Commun. 2015;6:8983. doi: 10.1038/ncomms9983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Yaccoby S, Wezeman MJ, Zangari M, et al. Inhibitory effects of osteoblasts and increased bone formation on myeloma in novel culture systems and a myelomatous mouse model. Haematologica. 2006;91:192–199. [PMC free article] [PubMed] [Google Scholar]
  • 75.Li X, Pennisi A, Yaccoby S. Role of decorin in the antimyeloma effects of osteoblasts. Blood. 2008;112:159–168. doi: 10.1182/blood-2007-11-124164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Kovacic N, Croucher PI, McDonald MM. Signaling Between Tumor Cells and the Host Bone Marrow Microenvironment. Calcif. Tissue Int. 2013;94:125–139. doi: 10.1007/s00223-013-9794-7. [DOI] [PubMed] [Google Scholar]
  • 77.Tian E, Zhan F, Walker R, et al. The role of the Wnt-signaling antagonist DKK1 in the development of osteolytic lesions in multiple myeloma. N. Engl. J. Med. 2003;349:2483–2494. doi: 10.1056/NEJMoa030847. [DOI] [PubMed] [Google Scholar]
  • 78.Zannettino ACW, Farrugia AN, Kortesidis A, et al. Elevated serum levels of stromal-derived factor-1alpha are associated with increased osteoclast activity and osteolytic bone disease in multiple myeloma patients. Cancer Res. 2005;65:1700–1709. doi: 10.1158/0008-5472.CAN-04-1687. [DOI] [PubMed] [Google Scholar]
  • 79.Choi SJ, Cruz JC, Craig F, et al. Macrophage inflammatory protein 1-alpha is a potential osteoclast stimulatory factor in multiple myeloma. Blood. 2000;96:671–675. [PubMed] [Google Scholar]
  • 80.Obeng Ea, Obeng Ea, Carlson LM, et al. Proteasome inhibitors induce a terminal unfolded protein response in multiple myeloma cells. Cell. 2006;107:4907–4916. doi: 10.1182/blood-2005-08-3531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Ramasamy K, Khatun H, Macpherson L, et al. Fluorescence-based experimental model to evaluate the concomitant effect of drugs on the tumour microenvironment and cancer cells. Br. J. Haematol. 2012;157:564–579. doi: 10.1111/j.1365-2141.2012.09103.x. [DOI] [PubMed] [Google Scholar]
  • 82.Tanaka Y, Abe M, Hiasa M, et al. Myeloma cell-osteoclast interaction enhances angiogenesis together with bone resorption: a role for vascular endothelial cell growth factor and osteopontin. Clin. Cancer Res. 2007;13:816–823. doi: 10.1158/1078-0432.CCR-06-2258. [DOI] [PubMed] [Google Scholar]
  • 83.Lacey DL, Timms E, Tan HL, et al. Osteoprotegerin ligand is a cytokine that regulates osteoclast differentiation and activation. Cell. 1998;93:165–176. doi: 10.1016/s0092-8674(00)81569-x. [DOI] [PubMed] [Google Scholar]
  • 84.Kong YY, Yoshida H, Sarosi I, et al. OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesis. Nature. 1999;397:315–323. doi: 10.1038/16852. [DOI] [PubMed] [Google Scholar]
  • 85.Roodman GD, Dougall WC. RANK ligand as a therapeutic target for bone metastases and multiple myeloma. Cancer Treat. Rev. 2008;34:92–101. doi: 10.1016/j.ctrv.2007.09.002. [DOI] [PubMed] [Google Scholar]
  • 86.Henry DH, Costa L, Goldwasser F, et al. Randomized, double-blind study of denosumab versus zoledronic acid in the treatment of bone metastases in patients with advanced cancer (excluding breast and prostate cancer) or multiple myeloma. J. Clin. Oncol. 2011;29:1125–1132. doi: 10.1200/JCO.2010.31.3304. [DOI] [PubMed] [Google Scholar]
  • 87.Dallas SL, Prideaux M, Bonewald LF. The osteocyte: an endocrine cell … and more. Endocr. Rev. 2013;34:658–690. doi: 10.1210/er.2012-1026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.van Bezooijen RL. Sclerostin Is an Osteocyte-expressed Negative Regulator of Bone Formation, But Not a Classical BMP Antagonist. J. Exp. Med. 2004;199:805–814. doi: 10.1084/jem.20031454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Kogianni G, Noble BS. The biology of osteocytes. Curr. Osteoporos. Rep. 2007;5:81–86. doi: 10.1007/s11914-007-0007-z. [DOI] [PubMed] [Google Scholar]
  • 90.Nakashima T, Hayashi M, Fukunaga T, et al. Evidence for osteocyte regulation of bone homeostasis through RANKL expression. Nat. Med. 2011;17:1231–1234. doi: 10.1038/nm.2452. [DOI] [PubMed] [Google Scholar]
  • 91.Baron R, Kneissel M. WNT signaling in bone homeostasis and disease: from human mutations to treatments. Nat. Med. 2013;19:179–192. doi: 10.1038/nm.3074. [DOI] [PubMed] [Google Scholar]
  • 92.Semenov M, Tamai K, He X. SOST Is a Ligand for LRP5/LRP6 and a Wnt Signaling Inhibitor. J. Biol. Chem. 2005;280:26770–26775. doi: 10.1074/jbc.M504308200. [DOI] [PubMed] [Google Scholar]
  • 93.Niedźwiedzki T, Filipowska J. Bone remodeling in the context of cellular and systemic regulation: the role of osteocytes and the nervous system. J. Mol. Endocrinol. 2015;55:R23–R36. doi: 10.1530/JME-15-0067. [DOI] [PubMed] [Google Scholar]
  • 94.Colucci S, Brunetti G, Oranger A, et al. Myeloma cells suppress osteoblasts through sclerostin secretion. Blood Cancer J. 2011;1:e27. doi: 10.1038/bcj.2011.22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Giuliani N, Ferretti M, Bolzoni M, et al. Increased osteocyte death in multiple myeloma patients: role in myeloma-induced osteoclast formation. Leukemia. 2012;26:1391–1401. doi: 10.1038/leu.2011.381. [DOI] [PubMed] [Google Scholar]
  • 96.Terpos E, Christoulas D, Katodritou E, et al. Elevated circulating sclerostin correlates with advanced disease features and abnormal bone remodeling in symptomatic myeloma: reduction post-bortezomib monotherapy. Int. J. Cancer. 2012;131:1466–1471. doi: 10.1002/ijc.27342. [DOI] [PubMed] [Google Scholar]
  • 97.Brunetti G, Oranger A, Mori G, et al. Sclerostin is overexpressed by plasma cells from multiple myeloma patients. Ann. N. Y. Acad. Sci. 2011;1237:19–23. doi: 10.1111/j.1749-6632.2011.06196.x. [DOI] [PubMed] [Google Scholar]
  • 98.Wang X-T, He Y-C, Zhou S-Y, et al. Bone marrow plasma macrophage inflammatory protein protein-1 alpha(MIP-1 alpha) and sclerostin in multiple myeloma: relationship with bone disease and clinical characteristics. Leuk. Res. 2014;38:525–531. doi: 10.1016/j.leukres.2014.02.010. [DOI] [PubMed] [Google Scholar]
  • 99.Lecka-Czernik B. Marrow fat metabolism is linked to the systemic energy metabolism. Bone. 2012;50:534–539. doi: 10.1016/j.bone.2011.06.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Vogl DT, Wang T, Pérez WS, et al. Effect of obesity on outcomes after autologous hematopoietic stem cell transplantation for multiple myeloma. Biol. Blood Marrow Transplant. 2011;17:1765–1774. doi: 10.1016/j.bbmt.2011.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Islam R, Altundag K, Kurt M, Altundag O, Turen S. Association between obesity and multiple myeloma in postmenopausal women may be attributed to increased aromatization of androgen in adipose tissue. Med. Hypotheses. 2005;65:1001–1002. doi: 10.1016/j.mehy.2005.05.014. [DOI] [PubMed] [Google Scholar]
  • 102.Caers J, Deleu S, Belaid Z, et al. Neighboring adipocytes participate in the bone marrow microenvironment of multiple myeloma cells. Leukemia. 2007;21:1580–1884. doi: 10.1038/sj.leu.2404658. [DOI] [PubMed] [Google Scholar]
  • 103.Liu Z, Xu J, He J, et al. Mature adipocytes in bone marrow protect myeloma cells against chemotherapy through autophagy activation. Oncotarget. 2015;6:34329–34241. doi: 10.18632/oncotarget.6020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Arita Y, Kihara S, Ouchi N, et al. Paradoxical Decrease of an Adipose-Specific Protein, Adiponectin, in Obesity. Biochem. Biophys. Res. Commun. 1999;257:79–83. doi: 10.1006/bbrc.1999.0255. [DOI] [PubMed] [Google Scholar]
  • 105.Bråkenhielm E, Veitonmäki N, Cao R, et al. Adiponectin-induced antiangiogenesis and antitumor activity involve caspase-mediated endothelial cell apoptosis. Proc. Natl. Acad. Sci. U. S. A. 2004;101:2476–2481. doi: 10.1073/pnas.0308671100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Dalamaga M, Diakopoulos KN, Mantzoros CS. The role of adiponectin in cancer: A review of current evidence. Endocr. Rev. 2012;33:547–594. doi: 10.1210/er.2011-1015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Esheba NE, Shahba A, El Shora O, Shora OEl. Assessment of leptin and resistin levels in non-obese multiple myeloma patients and their relation with Ig level and disease stage. J. Egypt. Natl. Canc. Inst. 2014;26:61–66. doi: 10.1016/j.jnci.2014.02.002. [DOI] [PubMed] [Google Scholar]
  • 108.Hofmann JN, Liao LM, Pollak MN, et al. A prospective study of circulating adipokine levels and risk of multiple myeloma. Blood. 2012;120:4418–4421. doi: 10.1182/blood-2012-06-438606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Winkler IG, Sims NA, Pettit AR, et al. Bone marrow macrophages maintain hematopoietic stem cell (HSC) niches and their depletion mobilizes HSCs. Blood. 2010;116:4815–4828. doi: 10.1182/blood-2009-11-253534. [DOI] [PubMed] [Google Scholar]
  • 110.Casanova-Acebes M, A-González N, Weiss LA, Hidalgo A. Innate immune cells as homeostatic regulators of the hematopoietic niche. Int. J. Hematol. 2014;99:685–694. doi: 10.1007/s12185-014-1561-7. [DOI] [PubMed] [Google Scholar]
  • 111.Baschuk N, Rautela J, Parker BS. Bone specific immunity and its impact on metastasis. Bonekey Rep. 2015;4:665. doi: 10.1038/bonekey.2015.32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Reya T. Illuminating immune privilege--a role for regulatory T cells in preventing rejection. N. Engl. J. Med. 2011;365:956–957. doi: 10.1056/NEJMcibr1107781. [DOI] [PubMed] [Google Scholar]
  • 113.Dawson MR, Chae S-S, Jain RK, Duda DG. Direct evidence for lineage-dependent effects of bone marrow stromal cells on tumor progression. Am. J. Cancer Res. 2011;1:144–154. [PMC free article] [PubMed] [Google Scholar]
  • 114.Asimakopoulos F, Kim J, Denu RA, et al. Macrophages in multiple myeloma: emerging concepts and therapeutic implications. Leuk. Lymphoma. 2013;54:2112–2121. doi: 10.3109/10428194.2013.778409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Kawano Y, Moschetta M, Manier S, et al. Targeting the bone marrow microenvironment in multiple myeloma. Immunol. Rev. 2015;263:160–172. doi: 10.1111/imr.12233. [DOI] [PubMed] [Google Scholar]
  • 116.Dankbar B, Padró T, Leo R, et al. Vascular endothelial growth factor and interleukin-6 in paracrine tumor-stromal cell interactions in multiple myeloma. Blood. 2000;95:2630–2636. [PubMed] [Google Scholar]
  • 117.Moschetta M, Mishima Y, Sahin I, et al. Role of endothelial progenitor cells in cancer progression. Biochim. Biophys. Acta. 2014;1846:26–39. doi: 10.1016/j.bbcan.2014.03.005. [DOI] [PubMed] [Google Scholar]
  • 118.Mishima Y, Aljawai Y, Sahin I, et al. Bone Marrow Mobilization Of Endothelial Progenitor Cells Represents An Early Pathogenic Event During Multiple Myeloma Progression. Blood. 2013;122:680. [Google Scholar]
  • 119.Kyle RA, Rajkumar SV. Multiple myeloma. N. Engl. J. Med. 2004;351:1860–1873. doi: 10.1056/NEJMra041875. [DOI] [PubMed] [Google Scholar]
  • 120.Kyle RA, Remstein ED, Therneau TM, et al. Clinical course and prognosis of smoldering (asymptomatic) multiple myeloma. N. Engl. J. Med. 2007;356:2582–2590. doi: 10.1056/NEJMoa070389. [DOI] [PubMed] [Google Scholar]
  • 121.Hussein K, Stucki-Koch A, Alchalby H, Triviai I, Kröger N, Kreipe H. Cytokine Expression Pattern in Bone Marrow Microenvironment after Allogeneic Stem Cell Transplantation in Primary Myelofibrosis. Biol. Blood Marrow Transplant. 2015 doi: 10.1016/j.bbmt.2015.12.006. [DOI] [PubMed] [Google Scholar]
  • 122.Gay F, Oliva S, Petrucci MT, et al. Chemotherapy plus lenalidomide versus autologous transplantation, followed by lenalidomide plus prednisone versus lenalidomide maintenance, in patients with multiple myeloma: a randomised, multicentre, phase 3 trial. Lancet. Oncol. 2015;16:1617–1629. doi: 10.1016/S1470-2045(15)00389-7. [DOI] [PubMed] [Google Scholar]
  • 123.Laubach J, Garderet L, Mahindra A, et al. Management of relapsed multiple myeloma: Recommendations of the international myeloma working group. Leukemia. 2015 doi: 10.1038/leu.2015.356. [DOI] [PubMed] [Google Scholar]
  • 124.Orlowski RZ, Nagler A, Sonneveld P, et al. Randomized Phase III Study of Pegylated Liposomal Doxorubicin Plus Bortezomib Compared With Bortezomib Alone in Relapsed or Refractory Multiple Myeloma: Combination Therapy Improves Time to Progression. J. Clin. Oncol. 2007;25:3892–3901. doi: 10.1200/JCO.2006.10.5460. [DOI] [PubMed] [Google Scholar]
  • 125.Sonneveld P, Hajek R, Nagler A, et al. Combined pegylated liposomal doxorubicin and bortezomib is highly effective in patients with recurrent or refractory multiple myeloma who received prior thalidomide/lenalidomide therapy. Cancer. 2008;112:1529–1537. doi: 10.1002/cncr.23326. [DOI] [PubMed] [Google Scholar]
  • 126.Palumbo A, Bertola A, Musto P, et al. Oral melphalan, prednisone, and thalidomide for newly diagnosed patients with myeloma. Cancer. 2005;104:1428–1433. doi: 10.1002/cncr.21342. [DOI] [PubMed] [Google Scholar]
  • 127.García-Sanz R, González-Fraile MI, Sierra M, López C, González M, San Miguel JF. The combination of thalidomide, cyclophosphamide and dexamethasone (ThaCyDex) is feasible and can be an option for relapsed/refractory multiple myeloma. Hematol. J. 2002;3:43–48. doi: 10.1038/sj.thj.6200150. [DOI] [PubMed] [Google Scholar]
  • 128.Kiziltepe T, Hideshima T, Catley L, et al. 5-Azacytidine, a DNA methyltransferase inhibitor, induces ATR-mediated DNA double-strand break responses, apoptosis, and synergistic cytotoxicity with doxorubicin and bortezomib against multiple myeloma cells. Mol. Cancer Ther. 2007;6:1718–1727. doi: 10.1158/1535-7163.MCT-07-0010. [DOI] [PubMed] [Google Scholar]
  • 129.Ciechanover A, Orian A, Schwartz AL. The ubiquitin-mediated proteolytic pathway: Mode of action and clinical implications. J. Cell. Biochem. 2000;77:40–51. doi: 10.1002/(sici)1097-4644(2000)77:34+<40::aid-jcb9>3.0.co;2-6. [DOI] [PubMed] [Google Scholar]
  • 130.Giuliani N, Morandi F, Tagliaferri S, et al. The proteasome inhibitor bortezomib affects osteoblast differentiation in vitro and in vivo in multiple myeloma patients. Blood. 2007;110:334–338. doi: 10.1182/blood-2006-11-059188. [DOI] [PubMed] [Google Scholar]
  • 131.von Metzler I, Krebbel H, Hecht M, et al. Bortezomib inhibits human osteoclastogenesis. Leukemia. 2007;21:2025–2034. doi: 10.1038/sj.leu.2404806. [DOI] [PubMed] [Google Scholar]
  • 132.Popat R, Oakervee H, Williams C, et al. Bortezomib, low-dose intravenous melphalan, and dexamethasone for patients with relapsed multiple myeloma. Br. J. Haematol. 2009;144:887–894. doi: 10.1111/j.1365-2141.2008.07572.x. [DOI] [PubMed] [Google Scholar]
  • 133.Kropff M, Bisping G, Schuck E, et al. Bortezomib in combination with intermediate-dose dexamethasone and continuous low-dose oral cyclophosphamide for relapsed multiple myeloma. Br. J. Haematol. 2007;138:330–337. doi: 10.1111/j.1365-2141.2007.06656.x. [DOI] [PubMed] [Google Scholar]
  • 134.Ludwig H, Kasparu H, Leitgeb C, et al. Bendamustine-bortezomib-dexamethasone is an active and well-tolerated regimen in patients with relapsed or refractory multiple myeloma. Blood. 2014;123:985–991. doi: 10.1182/blood-2013-08-521468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Offidani M, Corvatta L, Maracci L, et al. Efficacy and tolerability of bendamustine, bortezomib and dexamethasone in patients with relapsed-refractory multiple myeloma: a phase II study. Blood Cancer J. 2013;3:e162. doi: 10.1038/bcj.2013.58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Harousseau J-L, Dimopoulos MA, Wang M, et al. Better quality of response to lenalidomide plus dexamethasone is associated with improved clinical outcomes in patients with relapsed or refractory multiple myeloma. Haematologica. 2010;95:1738–1744. doi: 10.3324/haematol.2009.015917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Mitsiades N, Mitsiades CS, Richardson PG, et al. The proteasome inhibitor PS-341 potentiates sensitivity of multiple myeloma cells to conventional chemotherapeutic agents: therapeutic applications. Blood. 2003;101:2377–2380. doi: 10.1182/blood-2002-06-1768. [DOI] [PubMed] [Google Scholar]
  • 138.Ma MH, Yang HH, Parker K, et al. The proteasome inhibitor PS-341 markedly enhances sensitivity of multiple myeloma tumor cells to chemotherapeutic agents. Clin. Cancer Res. 2003;9:1136–1144. [PubMed] [Google Scholar]
  • 139.Richardson PG, Barlogie B, Berenson J, et al. A phase 2 study of bortezomib in relapsed, refractory myeloma. N. Engl. J. Med. 2003;348:2609–2617. doi: 10.1056/NEJMoa030288. [DOI] [PubMed] [Google Scholar]
  • 140.Petrucci MT, Giraldo P, Corradini P, et al. A prospective, international phase 2 study of bortezomib retreatment in patients with relapsed multiple myeloma. Br. J. Haematol. 2013;160:649–659. doi: 10.1111/bjh.12198. [DOI] [PubMed] [Google Scholar]
  • 141.Jakubowiak AJ, Dytfeld D, Griffith KA, et al. A phase 1/2 study of carfilzomib in combination with lenalidomide and low-dose dexamethasone as a frontline treatment for multiple myeloma. Blood. 2012;120:1801–1809. doi: 10.1182/blood-2012-04-422683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Wang M, Cheng J. Overview and management of cardiac and pulmonary adverse events in patients with relapsed and/or refractory multiple myeloma treated with single-agent carfilzomib. Oncology (Williston Park) 2013;27(Suppl 3):24–30. [PubMed] [Google Scholar]
  • 143.Siegel DS, Martin T, Wang M, et al. A phase 2 study of single-agent carfilzomib (PX-171-003-A1) in patients with relapsed and refractory multiple myeloma. Blood. 2012;120:2817–2825. doi: 10.1182/blood-2012-05-425934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Shah JJ, Stadtmauer EA, Abonour R, et al. Carfilzomib, pomalidomide, and dexamethasone for relapsed or refractory myeloma. Blood. 2015;126:2284–2290. doi: 10.1182/blood-2015-05-643320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Averbuch SD. New bisphosphonates in the treatment of bone metastases. Cancer. 1993;72:3443–3452. doi: 10.1002/1097-0142(19931201)72:11+<3443::aid-cncr2820721611>3.0.co;2-3. [DOI] [PubMed] [Google Scholar]
  • 146.Baron R, Ferrari S, Russell RGG. Denosumab and bisphosphonates: different mechanisms of action and effects. Bone. 2011;48:677–692. doi: 10.1016/j.bone.2010.11.020. [DOI] [PubMed] [Google Scholar]
  • 147.Mundy GR, Ibbotson KJ, D’Souza SM, Simpson EL, Jacobs JW, Martin TJ. The hypercalcemia of cancer. Clinical implications and pathogenic mechanisms. N. Engl. J. Med. 1984;310:1718–1727. doi: 10.1056/NEJM198406283102607. [DOI] [PubMed] [Google Scholar]
  • 148.Croucher PI, Apperley JF. Bone disease in multiple myeloma. Br. J. Haematol. 1998;103:902–910. doi: 10.1046/j.1365-2141.1998.01082.x. [DOI] [PubMed] [Google Scholar]
  • 149.Bataille R, Delmas PD, Chappard D, Sany J. Abnormal serum bone Gla protein levels in multiple myeloma. Crucial role of bone formation and prognostic implications. Cancer. 1990;66:167–172. doi: 10.1002/1097-0142(19900701)66:1<167::aid-cncr2820660130>3.0.co;2-9. [DOI] [PubMed] [Google Scholar]
  • 150.Bataille R, Klein B. Serum beta-2-microglobulin (beta 2m) in myeloma: toward a simple prognostic stratification using beta 2M and acute-phase proteins? Blood. 1991;77:1616–1617. [PubMed] [Google Scholar]
  • 151.Abildgaard N, Rungby J, Glerup H, et al. Long-term oral pamidronate treatment inhibits osteoclastic bone resorption and bone turnover without affecting osteoblastic function in multiple myeloma. Eur. J. Haematol. 1998;61:128–134. doi: 10.1111/j.1600-0609.1998.tb01073.x. [DOI] [PubMed] [Google Scholar]
  • 152.Jagannath S, Durie BGM, Wolf J, et al. Bortezomib therapy alone and in combination with dexamethasone for previously untreated symptomatic multiple myeloma. Br. J. Haematol. 2005;129:776–783. doi: 10.1111/j.1365-2141.2005.05540.x. [DOI] [PubMed] [Google Scholar]
  • 153.Levy J, Roodman GD. The role of bisphosphonates in multiple myeloma. Curr. Hematol. Malig. Rep. 2009;4:108–112. doi: 10.1007/s11899-009-0015-4. [DOI] [PubMed] [Google Scholar]
  • 154.Merin NM, Kelly KR. Clinical use of proteasome inhibitors in the treatment of multiple myeloma. Pharmaceuticals (Basel) 2014;8:1–20. doi: 10.3390/ph8010001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Berenson JR, Lichtenstein A, Porter L, et al. Efficacy of pamidronate in reducing skeletal events in patients with advanced multiple myeloma. Myeloma Aredia Study Group. N. Engl. J. Med. 1996;334:488–493. doi: 10.1056/NEJM199602223340802. [DOI] [PubMed] [Google Scholar]
  • 156.Berenson JR, Lichtenstein A, Porter L, et al. Long-term pamidronate treatment of advanced multiple myeloma patients reduces skeletal events. Myeloma Aredia Study Group. J. Clin. Oncol. 1998;16:593–602. doi: 10.1200/JCO.1998.16.2.593. [DOI] [PubMed] [Google Scholar]
  • 157.Chlebowski RT, Chen Z, Cauley JA, et al. Oral bisphosphonate use and breast cancer incidence in postmenopausal women. J. Clin. Oncol. 2010;28:3582–3590. doi: 10.1200/JCO.2010.28.2095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Holen I, Coleman RE. Bisphosphonates as treatment of bone metastases. Curr. Pharm. Des. 2010;16:1262–1271. doi: 10.2174/138161210791034003. [DOI] [PubMed] [Google Scholar]
  • 159.Gnant M, Eidtmann H. The anti-tumor effect of bisphosphonates ABCSG-12, ZO-FAST and more. Crit. Rev. Oncol. Hematol. 2010;74(Suppl 1):S2–S6. doi: 10.1016/S1040-8428(10)70003-2. [DOI] [PubMed] [Google Scholar]
  • 160.Van Acker HH, Anguille S, Willemen Y, Smits EL, Van Tendeloo VF. Bisphosphonates for cancer treatment: Mechanisms of action and lessons from clinical trials. Pharmacol. Ther. 2016;158:24–40. doi: 10.1016/j.pharmthera.2015.11.008. [DOI] [PubMed] [Google Scholar]
  • 161.Glavey S, Reagan M, Manier S, et al. Dissecting the Mechanisms of Activity of SLAMF7 and the Targeting Antibody Elotuzumab in Multiple Myeloma. Blood. 2014;124:3431. [Google Scholar]
  • 162.Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat. Rev. Immunol. 2009;9:162–174. doi: 10.1038/nri2506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Zhu YX, Kortuem KM, Stewart a. K. Molecular mechanism of action of immune-modulatory drugs thalidomide, lenalidomide and pomalidomide in multiple myeloma. Leuk. Lymphoma. 2012;54:1–5. doi: 10.3109/10428194.2012.728597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Dimopoulos MA, Swern AS, Li JS, et al. Efficacy and safety of long-term treatment with lenalidomide and dexamethasone in patients with relapsed/refractory multiple myeloma. Blood Cancer J. 2014;4:e257. doi: 10.1038/bcj.2014.77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Richardson PG, Xie W, Jagannath S, et al. A phase 2 trial of lenalidomide, bortezomib, and dexamethasone in patients with relapsed and relapsed/refractory myeloma. Blood. 2014;123:1461–1469. doi: 10.1182/blood-2013-07-517276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Santo L, Hideshima T, Cirstea D, et al. Antimyeloma activity of a multitargeted kinase inhibitor, AT9283, via potent Aurora kinase and STAT3 inhibition either alone or in combination with lenalidomide. Clin. Cancer Res. 2011;17:3259–3271. doi: 10.1158/1078-0432.CCR-10-3012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.van der Veer MS, de Weers M, van Kessel B, et al. Towards effective immunotherapy of myeloma: enhanced elimination of myeloma cells by combination of lenalidomide with the human CD38 monoclonal antibody daratumumab. Haematologica. 2011;96:284–290. doi: 10.3324/haematol.2010.030759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Jakubowiak AJ, Griffith KA, Reece DE, et al. Lenalidomide, bortezomib, pegylated liposomal doxorubicin, and dexamethasone in newly diagnosed multiple myeloma: a phase 1/2 Multiple Myeloma Research Consortium trial. Blood. 2011;118:535–543. doi: 10.1182/blood-2011-02-334755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Weisel K, Dimopoulos M, Song KW, et al. Pomalidomide and Low-Dose Dexamethasone Improves Health-Related Quality of Life and Prolongs Time to Worsening in Relapsed/Refractory Patients With Multiple Myeloma Enrolled in the MM-003 Randomized Phase III Trial. Clin. Lymphoma. Myeloma Leuk. 2015;15:519–530. doi: 10.1016/j.clml.2015.05.007. [DOI] [PubMed] [Google Scholar]
  • 170.Mitsiades N, Mitsiades CS, Poulaki V, et al. Apoptotic signaling induced by immunomodulatory thalidomide analogs in human multiple myeloma cells: therapeutic implications. Blood. 2002;99:4525–4530. doi: 10.1182/blood.v99.12.4525. [DOI] [PubMed] [Google Scholar]
  • 171.Prince HM, Adena M, Smith DK, Hertel J. Efficacy of single-agent bortezomib vs. single-agent thalidomide in patients with relapsed or refractory multiple myeloma: a systematic comparison. Eur. J. Haematol. 2007;79:93–99. doi: 10.1111/j.1600-0609.2007.00886.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Pineda-Roman M, Zangari M, van Rhee F, et al. VTD combination therapy with bortezomib-thalidomide-dexamethasone is highly effective in advanced and refractory multiple myeloma. Leukemia. 2008;22:1419–1427. doi: 10.1038/leu.2008.99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Garrison LP, Wang S-T, Huang H, et al. The cost-effectiveness of initial treatment of multiple myeloma in the U.S. with bortezomib plus melphalan and prednisone versus thalidomide plus melphalan and prednisone or lenalidomide plus melphalan and prednisone with continuous lenalidomide maintenance treatment. Oncologist. 2013;18:27–36. doi: 10.1634/theoncologist.2011-0380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Garderet L, Iacobelli S, Moreau P, et al. Superiority of the triple combination of bortezomib-thalidomide-dexamethasone over the dual combination of thalidomide-dexamethasone in patients with multiple myeloma progressing or relapsing after autologous transplantation: the MMVAR/IFM 2005-04 Randomi. J. Clin. Oncol. 2012;30:2475–2482. doi: 10.1200/JCO.2011.37.4918. [DOI] [PubMed] [Google Scholar]
  • 175.Sonneveld P, Schmidt-Wolf IGH, van der Holt B, et al. Bortezomib induction and maintenance treatment in patients with newly diagnosed multiple myeloma: results of the randomized phase III HOVON-65/ GMMG-HD4 trial. J. Clin. Oncol. 2012;30:2946–2955. doi: 10.1200/JCO.2011.39.6820. [DOI] [PubMed] [Google Scholar]
  • 176.Laubach JP, Voorhees PM, Hassoun H, Jakubowiak A, Lonial S, Richardson PG. Current strategies for treatment of relapsed/refractory multiple myeloma. Expert Rev. Hematol. 2014;7:97–111. doi: 10.1586/17474086.2014.882764. [DOI] [PubMed] [Google Scholar]
  • 177.de Weers M, Tai Y-T, van der Veer MS, et al. Daratumumab, a novel therapeutic human CD38 monoclonal antibody, induces killing of multiple myeloma and other hematological tumors. J. Immunol. 2011;186:1840–1848. doi: 10.4049/jimmunol.1003032. [DOI] [PubMed] [Google Scholar]
  • 178.Lokhorst HM, Plesner T, Laubach JP, et al. Targeting CD38 with Daratumumab Monotherapy in Multiple Myeloma. N. Engl. J. Med. 2015;373:1207–1219. doi: 10.1056/NEJMoa1506348. [DOI] [PubMed] [Google Scholar]
  • 179.Santonocito AM, Consoli U, Bagnato S, et al. Flow cytometric detection of aneuploid CD38(++) plasmacells and CD19(+) B-lymphocytes in bone marrow, peripheral blood and PBSC harvest in multiple myeloma patients. Leuk. Res. 2004;28:469–477. doi: 10.1016/j.leukres.2003.09.015. [DOI] [PubMed] [Google Scholar]
  • 180.Bayer-Garner IB, Sanderson RD, Dhodapkar MV, Owens RB, Wilson CS. Syndecan-1 (CD138) immunoreactivity in bone marrow biopsies of multiple myeloma: shed syndecan-1 accumulates in fibrotic regions. Mod. Pathol. 2001;14:1052–1058. doi: 10.1038/modpathol.3880435. [DOI] [PubMed] [Google Scholar]
  • 181.Fichou N, Gouard S, Maurel C, et al. Single-Dose Anti-CD138 Radioimmunotherapy: Bismuth-213 is More Efficient than Lutetium-177 for Treatment of Multiple Myeloma in a Preclinical Model. Front. Med. 2015;2:76. doi: 10.3389/fmed.2015.00076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Kim SY, Theunissen J-W, Balibalos J, et al. A novel antibody-drug conjugate targeting SAIL for the treatment of hematologic malignancies. Blood Cancer J. 2015;5:e316. doi: 10.1038/bcj.2015.39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Glavey SV, Manier S, Natoni A, et al. The sialyltransferase ST3GAL6 influences homing and survival in multiple myeloma. Blood. 2014;124:1765–1777. doi: 10.1182/blood-2014-03-560862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Lonial S, Dimopoulos M, Palumbo A, et al. Elotuzumab Therapy for Relapsed or Refractory Multiple Myeloma. N. Engl. J. Med. 2015;373:621–631. doi: 10.1056/NEJMoa1505654. [DOI] [PubMed] [Google Scholar]
  • 185.Jin H, Wang B, Li J, et al. Anti-DKK1 antibody promotes bone fracture healing through activation of β-catenin signaling. Bone. 2015;71:63–75. doi: 10.1016/j.bone.2014.07.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Feng X, Deng S, An G, et al. [Detection of serum DKK1 in multiple myeloma and myeloma bone disease] Zhonghua Xue Ye Xue Za Zhi. 2015;36:682–685. doi: 10.3760/cma.j.issn.0253-2727.2015.08.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Ščudla V, Petrová P, Pika T, et al. [Analysis of serum levels of Dickkopf-1 (DKK-1) in monoclonal gammopathy of undetermined significance and multiple myeloma] Casopís lékarŭ ceských. 2015;154:181–188. [PubMed] [Google Scholar]
  • 188.Pozzi S, Fulciniti M, Yan H, et al. In vivo and in vitro effects of a novel anti-Dkk1 neutralizing antibody in multiple myeloma. Bone. 2013;53:487–496. doi: 10.1016/j.bone.2013.01.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Scullen T, Santo L, Vallet S, et al. Lenalidomide in combination with an activin A-neutralizing antibody: preclinical rationale for a novel anti-myeloma strategy. Leukemia. 2013;27:1715–1721. doi: 10.1038/leu.2013.50. [DOI] [PubMed] [Google Scholar]
  • 190.Vallet S, Mukherjee S, Vaghela N, et al. Activin A promotes multiple myeloma-induced osteolysis and is a promising target for myeloma bone disease. Proc. Natl. Acad. Sci. U. S. A. 2010;107:5124–5129. doi: 10.1073/pnas.0911929107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Kristensen IB, Christensen JH, Lyng MB, et al. Expression of osteoblast and osteoclast regulatory genes in the bone marrow microenvironment in multiple myeloma: only up-regulation of Wnt inhibitors SFRP3 and DKK1 is associated with lytic bone disease. Leuk. Lymphoma. 2014;55:911–919. doi: 10.3109/10428194.2013.820288. [DOI] [PubMed] [Google Scholar]
  • 192.Reagan MR, McDonald MM, Terry R, et al. Anti-Sclerostin Treatment Prevents Multiple Myeloma Induced Bone Loss and Reduces Tumor Burden [Abstract] 57th Annu. Am. Soc. Hematol. Meet. 2015:119. [Google Scholar]
  • 193.Heath DJ, Chantry AD, Buckle CH, et al. Inhibiting Dickkopf-1 (Dkk1) removes suppression of bone formation and prevents the development of osteolytic bone disease in multiple myeloma. J. Bone Miner. Res. 2009;24:425–436. doi: 10.1359/jbmr.081104. [DOI] [PubMed] [Google Scholar]
  • 194.van Dinther M, Zhang J, Weidauer SE, et al. Anti-Sclerostin antibody inhibits internalization of Sclerostin and Sclerostin-mediated antagonism of Wnt/LRP6 signaling. PLoS One. 2013;8:e62295. doi: 10.1371/journal.pone.0062295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Mazziotti G, Bilezikian J, Canalis E, Cocchi D, Giustina A. New understanding and treatments for osteoporosis. Endocrine. 2012;41:58–69. doi: 10.1007/s12020-011-9570-2. [DOI] [PubMed] [Google Scholar]
  • 196.Eda H, Santo L, Wein MN, et al. Regulation of Sclerostin Expression in Multiple Myeloma by Dkk-1; A Potential Therapeutic Strategy for Myeloma Bone Disease. J. Bone Miner. Res. 2016 doi: 10.1002/jbmr.2789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Sondergeld P, van de Donk NWCJ, Richardson PG, Plesner T. Monoclonal antibodies in myeloma. Clin. Adv. Hematol. Oncol. 2015;13:599–609. [PubMed] [Google Scholar]
  • 198.Bolden JE, Peart MJ, Johnstone RW. Anticancer activities of histone deacetylase inhibitors. Nat. Rev. Drug Discov. 2006;5:769–784. doi: 10.1038/nrd2133. [DOI] [PubMed] [Google Scholar]
  • 199.Grunstein M. Histone acetylation in chromatin structure and transcription. Nature. 1997;389:349–352. doi: 10.1038/38664. [DOI] [PubMed] [Google Scholar]
  • 200.Canella A, Nieves HC, Sborov DW, et al. HDAC inhibitor AR-42 decreases CD44 expression and sensitizes myeloma cells to lenalidomide. Oncotarget. 2015;6:31134–31150. doi: 10.18632/oncotarget.5290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Dimopoulos K, Gimsing P, Grønbæk K. The role of epigenetics in the biology of multiple myeloma. Blood Cancer J. 2014;4:e207. doi: 10.1038/bcj.2014.29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Chauhan D, Tian Z, Nicholson B, et al. A small molecule inhibitor of ubiquitin-specific protease-7 induces apoptosis in multiple myeloma cells and overcomes bortezomib resistance. Cancer Cell. 2012;22:345–358. doi: 10.1016/j.ccr.2012.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Qian DZ, Kato Y, Shabbeer S, et al. Targeting tumor angiogenesis with histone deacetylase inhibitors: the hydroxamic acid derivative LBH589. Clin. Cancer Res. 2006;12:634–642. doi: 10.1158/1078-0432.CCR-05-1132. [DOI] [PubMed] [Google Scholar]
  • 204.Dimopoulos M, Siegel DS, Lonial S, et al. Vorinostat or placebo in combination with bortezomib in patients with multiple myeloma (VANTAGE 088): a multicentre, randomised, double-blind study. Lancet. Oncol. 2013;14:1129–1140. doi: 10.1016/S1470-2045(13)70398-X. [DOI] [PubMed] [Google Scholar]
  • 205.San Miguel J, Weisel K, Moreau P, et al. Pomalidomide plus low-dose dexamethasone versus high-dose dexamethasone alone for patients with relapsed and refractory multiple myeloma (MM-003): a randomised, open-label, phase 3 trial. Lancet. Oncol. 2013;14:1055–1066. doi: 10.1016/S1470-2045(13)70380-2. [DOI] [PubMed] [Google Scholar]
  • 206.Biran N, Shahnaz S, Jagannath S, et al. A Phase II, Single-Center, Open-Label Study Of Oral Panobinostat In Combination With Lenalidomide and Weekly Dexamethasone In Patients With Multiple Myeloma. Blood. 2013;122:5392. [Google Scholar]
  • 207.San-Miguel JF, Hungria VTM, Yoon S-S, et al. Panobinostat plus bortezomib and dexamethasone versus placebo plus bortezomib and dexamethasone in patients with relapsed or relapsed and refractory multiple myeloma: a multicentre, randomised, double-blind phase 3 trial. Lancet. Oncol. 2014;15:1195–1206. doi: 10.1016/S1470-2045(14)70440-1. [DOI] [PubMed] [Google Scholar]
  • 208.Olechnowicz SWZ, Edwards CM. Contributions of the host microenvironment to cancer-induced bone disease. Cancer Res. 2014;74:1625–1631. doi: 10.1158/0008-5472.CAN-13-2645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Elefteriou F, Campbell P, Ma Y. Control of bone remodeling by the peripheral sympathetic nervous system. Calcif. Tissue Int. 2014;94:140–151. doi: 10.1007/s00223-013-9752-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Ashjian E, Redic K. Multiple myeloma: Updates for pharmacists in the treatment of relapsed and refractory disease. J. Oncol. Pharm. Pract. 2015 doi: 10.1177/1078155215572036. [DOI] [PubMed] [Google Scholar]
  • 211.Auner HW, Cenci S. Recent advances and future directions in targeting the secretory apparatus in multiple myeloma. Br. J. Haematol. 2015;168:14–25. doi: 10.1111/bjh.13172. [DOI] [PubMed] [Google Scholar]
  • 212.Ayed AO, Chang L-J, Moreb JS. Immunotherapy for multiple myeloma: Current status and future directions. Crit. Rev. Oncol. Hematol. 2015;96:399–412. doi: 10.1016/j.critrevonc.2015.06.006. [DOI] [PubMed] [Google Scholar]
  • 213.Lorenz J, Waldschmidt J, Wider D, et al. From CLL to Multiple Myeloma - Spleen Tyrosine Kinase (SYK) influences multiple myeloma cell survival and migration. Br. J. Haematol. 2015 doi: 10.1111/bjh.13825. [DOI] [PubMed] [Google Scholar]
  • 214.Brown R, Yang S, Weatherburn C, et al. Phospho-flow detection of constitutive and cytokine-induced pSTAT3/5, pAKT and pERK expression highlights novel prognostic biomarkers for patients with multiple myeloma. Leukemia. 2015;29:483–490. doi: 10.1038/leu.2014.204. [DOI] [PubMed] [Google Scholar]
  • 215.Pemmaraju N, Kantarjian H, Kadia T, et al. A phase I/II study of the Janus kinase (JAK)1 and 2 inhibitor ruxolitinib in patients with relapsed or refractory acute myeloid leukemia. Clin. Lymphoma. Myeloma Leuk. 2015;15:171–176. doi: 10.1016/j.clml.2014.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Nur H, Rao L, Frassanito MA, et al. Stimulation of invariant natural killer T cells by α-Galactosylceramide activates the JAK-STAT pathway in endothelial cells and reduces angiogenesis in the 5T33 multiple myeloma model. Br. J. Haematol. 2014;167:651–663. doi: 10.1111/bjh.13092. [DOI] [PubMed] [Google Scholar]
  • 217.Duan W, Chen S, Zhang Y, et al. Protein C-terminal enzymatic labeling identifies novel caspase cleavages during the apoptosis of multiple myeloma cells induced by kinase inhibition. Proteomics. 2015;16:60–69. doi: 10.1002/pmic.201500356. [DOI] [PubMed] [Google Scholar]
  • 218.Suzuki R, Kikuchi S, Harada T, et al. Combination of a Selective HSP90α/β Inhibitor and a RAS-RAF-MEK-ERK Signaling Pathway Inhibitor Triggers Synergistic Cytotoxicity in Multiple Myeloma Cells. PLoS One. 2015;10:e0143847. doi: 10.1371/journal.pone.0143847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Burger MT, Nishiguchi G, Han W, et al. Identification of N-(4-((1R,3S,5S)-3-Amino-5-methylcyclohexyl)pyridin-3-yl)-6-(2,6-difluorophenyl)-5-fluoropicolinamide (PIM447), a Potent and Selective Proviral Insertion Site of Moloney Murine Leukemia (PIM) 1, 2, and 3 Kinase Inhibitor in Clinical Tria. J. Med. Chem. 2015;58:8373–8386. doi: 10.1021/acs.jmedchem.5b01275. [DOI] [PubMed] [Google Scholar]
  • 220.Meads MB, Fang B, Mathews L, et al. Targeting PYK2 mediates microenvironment-specific cell death in multiple myeloma. Oncogene. 2015 doi: 10.1038/onc.2015.334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Zhang Y, Moschetta M, Huynh D, et al. Pyk2 promotes tumor progression in multiple myeloma. Blood. 2014;124:2675–2686. doi: 10.1182/blood-2014-03-563981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Han K, Xu X, Xu Z, et al. SC06, a novel small molecule compound, displays preclinical activity against multiple myeloma by disrupting the mTOR signaling pathway. Sci. Rep. 2015;5:12809. doi: 10.1038/srep12809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Chinen Y, Kuroda J, Shimura Y, et al. Phosphoinositide protein kinase PDPK1 is a crucial cell signaling mediator in multiple myeloma. Cancer Res. 2014;74:7418–7429. doi: 10.1158/0008-5472.CAN-14-1420. [DOI] [PubMed] [Google Scholar]
  • 224.Garfall AL, Fraietta JA, Maus MV. Immunotherapy with Chimeric Antigen Receptors for Multiple Myeloma. Discov. Med. 2014;17:37–46. [PubMed] [Google Scholar]
  • 225.Maus MV, June CH. CARTs on the road for myeloma. Clin. Cancer Res. 2014;20:3899–3901. doi: 10.1158/1078-0432.CCR-14-0721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Guo B, Chen M, Han Q, et al. CD138-directed adoptive immunotherapy of chimeric antigen receptor (CAR)-modified T cells for multiple myeloma. J. Cell. Immunother. 2015 [Google Scholar]
  • 227.Roussel M, Lauwers-Cances V, Robillard N, et al. Front-line transplantation program with lenalidomide, bortezomib, and dexamethasone combination as induction and consolidation followed by lenalidomide maintenance in patients with multiple myeloma: a phase II study by the Intergroupe Francophone du Myélo. J. Clin. Oncol. 2014;32:2712–2717. doi: 10.1200/JCO.2013.54.8164. [DOI] [PubMed] [Google Scholar]
  • 228.Stewart AK, Rajkumar SV, Dimopoulos MA, et al. Carfilzomib, lenalidomide, and dexamethasone for relapsed multiple myeloma. N. Engl. J. Med. 2015;372:142–152. doi: 10.1056/NEJMoa1411321. [DOI] [PubMed] [Google Scholar]
  • 229.Hurchla MA, Garcia-Gomez A, Hornick MC, et al. The epoxyketone-based proteasome inhibitors carfilzomib and orally bioavailable oprozomib have anti-resorptive and bone-anabolic activity in addition to anti-myeloma effects. Leukemia. 2013;27:430–440. doi: 10.1038/leu.2012.183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Gallerani E, Zucchetti M, Brunelli D, et al. A first in human phase I study of the proteasome inhibitor CEP-18770 in patients with advanced solid tumours and multiple myeloma. Eur. J. Cancer. 2013;49:290–296. doi: 10.1016/j.ejca.2012.09.009. [DOI] [PubMed] [Google Scholar]
  • 231.Palumbo A, Larocca A, Genuardi M, et al. Melphalan, prednisone, thalidomide and defibrotide in relapsed/refractory multiple myeloma: results of a multicenter phase I/II trial. Haematologica. 2010;95:1144–1149. doi: 10.3324/haematol.2009.017913. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Kyle RA, Jacobus S, Friedenberg WR, Slabber CF, Rajkumar SV, Greipp PR. The treatment of multiple myeloma using vincristine, carmustine, melphalan, cyclophosphamide, and prednisone (VBMCP) alternating with high-dose cyclophosphamide and alpha(2)beta interferon versus VBMCP: results of a phase III Eastern Cooperative Oncology. Cancer. 2009;115:2155–2164. doi: 10.1002/cncr.24221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Baz RC, Shain KH, Hussein MA, et al. Phase II study of pegylated liposomal doxorubicin, low-dose dexamethasone, and lenalidomide in patients with newly diagnosed multiple myeloma. Am. J. Hematol. 2014;89:62–67. doi: 10.1002/ajh.23587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Michael M, Bruns I, Bölke E, et al. Bendamustine in patients with relapsed or refractory multiple myeloma. Eur. J. Med. Res. 2010;15:13–19. doi: 10.1186/2047-783X-15-1-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Wood WA, Whitley J, Moore D, et al. Chemomobilization with Etoposide is Highly Effective in Patients with Multiple Myeloma and Overcomes the Effects of Age and Prior Therapy. Biol. Blood Marrow Transplant. 2011;17:141–146. doi: 10.1016/j.bbmt.2010.06.021. [DOI] [PubMed] [Google Scholar]
  • 236.Starr P. Elotuzumab, First-in-Class Monoclonal Antibody Immunotherapy, Improves Outcomes in Patients with Multiple Myeloma. Am. Heal. drug benefits. 2015;8:17. [PMC free article] [PubMed] [Google Scholar]
  • 237.Usmani SZ, Sexton R, Ailawadhi S, et al. Phase I safety data of lenalidomide, bortezomib, dexamethasone, and elotuzumab as induction therapy for newly diagnosed symptomatic multiple myeloma: SWOG S1211. Blood Cancer J. 2015;5:e334. doi: 10.1038/bcj.2015.62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Hansson M, Gimsing P, Badros A, et al. A Phase I Dose-Escalation Study of Antibody BI-505 in Relapsed/Refractory Multiple Myeloma. Clin. Cancer Res. 2015;21:2730–2736. doi: 10.1158/1078-0432.CCR-14-3090. [DOI] [PubMed] [Google Scholar]
  • 239.Baz R, Fanning S, Kunkel L, et al. Combination of rituximab and oral melphalan and prednisone in newly diagnosed multiple myeloma. Leuk. Lymphoma. 2007;48:2338–2344. doi: 10.1080/10428190701644330. [DOI] [PubMed] [Google Scholar]
  • 240.Bergua JM, Cabrera C, Arteta EG, Prieto J. Rituximab in CD20 positive multiple myeloma. Leukemia. 2008;22:1082–1083. doi: 10.1038/sj.leu.2405010. [DOI] [PubMed] [Google Scholar]
  • 241.Korte W, Jost C, Cogliatti S, Hess U, Cerny T. Accelerated progression of multiple myeloma during anti-CD20 (Rituximab) therapy. Ann. Oncol. 1999;10:1249–1250. doi: 10.1023/a:1008310819049. [DOI] [PubMed] [Google Scholar]
  • 242.Mitsiades CS, Mitsiades NS, McMullan CJ, et al. Transcriptional signature of histone deacetylase inhibition in multiple myeloma: biological and clinical implications. Proc. Natl. Acad. Sci. U. S. A. 2004;101:540–545. doi: 10.1073/pnas.2536759100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Richardson P, Mitsiades C, Colson K, et al. Phase I trial of oral vorinostat (suberoylanilide hydroxamic acid, SAHA) in patients with advanced multiple myeloma. Leuk. Lymphoma. 2008;49:502–507. doi: 10.1080/10428190701817258. [DOI] [PubMed] [Google Scholar]
  • 244.Vesole DH, Bilotti E, Richter JR, et al. Phase I study of carfilzomib, lenalidomide, vorinostat, and dexamethasone in patients with relapsed and/or refractory multiple myeloma. Br. J. Haematol. 2015;171:52–59. doi: 10.1111/bjh.13517. [DOI] [PubMed] [Google Scholar]
  • 245.Yee AJ, Voorhees PM, Bensinger W, et al. Ricolinostat (ACY-1215), a Selective HDAC6 Inhibitor, in Combination with Lenalidomide and Dexamethasone: Results of a Phase 1b Trial in Relapsed and Relapsed Refractory Multiple Myeloma. Blood. 2014;124:4772. [Google Scholar]

RESOURCES