Next Article in Journal
Enhanced Malignant Phenotypes of Glioblastoma Cells Surviving NPe6-Mediated Photodynamic Therapy are Regulated via ERK1/2 Activation
Next Article in Special Issue
Interrupting Neuron—Tumor Interactions to Overcome Treatment Resistance
Previous Article in Journal
Evaluation of Clinicopathological and Molecular Parameters on Disease Recurrence of Papillary Thyroid Cancer Patient: A Retrospective Observational Study
Previous Article in Special Issue
Epithelial Transfer of the Tyrosine Kinase Inhibitors Erlotinib, Gefitinib, Afatinib, Crizotinib, Sorafenib, Sunitinib, and Dasatinib: Implications for Clinical Resistance
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Tumour Dissemination in Multiple Myeloma Disease Progression and Relapse: A Potential Therapeutic Target in High-Risk Myeloma

by
Mara N. Zeissig
1,2,
Andrew C. W. Zannettino
1,2,3,4 and
Kate Vandyke
1,2,*
1
Myeloma Research Laboratory, Faculty of Health and Medical Sciences, Adelaide Medical School, The University of Australia, Adelaide 5005, Australia
2
Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide 5000, Australia
3
Central Adelaide Local Health Network, Adelaide 5000, Australia
4
Centre for Cancer Biology, University of South Australia, Adelaide 5000, Australia
*
Author to whom correspondence should be addressed.
Submission received: 28 October 2020 / Revised: 2 December 2020 / Accepted: 2 December 2020 / Published: 4 December 2020

Abstract

:

Simple Summary

Like in solid cancers, the process of dissemination is a critical feature of disease progression in the blood cancer multiple myeloma. At diagnosis, myeloma patients have cancer that has spread throughout the bone marrow, with patients with more disseminatory myeloma having worse outcomes for their disease. In this review, we discuss the current understanding of the mechanisms that underpin the dissemination process in multiple myeloma. Furthermore, we discuss the potential for the use of therapies that target the dissemination process as a novel means of improving outcomes for multiple myeloma patients.

Abstract

Multiple myeloma (MM) is a plasma cell (PC) malignancy characterised by the presence of MM PCs at multiple sites throughout the bone marrow. Increased numbers of peripheral blood MM PCs are associated with rapid disease progression, shorter time to relapse and are a feature of advanced disease. In this review, the current understanding of the process of MM PC dissemination and the extrinsic and intrinsic factors potentially driving it are addressed through analysis of patient-derived MM PCs and MM cell lines as well as mouse models of homing and dissemination. In addition, we discuss how patient cytogenetic subgroups that present with highly disseminated disease, such as t(4;14), t(14;16) and t(14;20), suggest that intrinsic properties of MM PC influence their ability to disseminate. Finally, we discuss the possibility of using therapeutic targeting of tumour dissemination to slow disease progression and prevent overt relapse.

Graphical Abstract

1. The Role of Dissemination in the Progression of Multiple Myeloma

Multiple myeloma (MM) is an incurable haematological malignancy characterised by the uncontrolled proliferation of clonal malignant plasma cells (PCs) in the bone marrow (BM) [1]. Worldwide, MM accounts for 1% of all cancers [2], with approximately 140,000 people diagnosed with myeloma every year [3]. A characteristic feature of MM is that, at the time of diagnosis, MM PCs are present at multiple sites throughout the skeleton [1].
During initial disease development, malignant clonal PCs establish themselves in BM niches that support their growth [4]. Migration and population of these transformed clonal PCs throughout the BM leads to the development of the pre-myeloma disease stages monoclonal gammopathy of undetermined significance (MGUS) or smouldering MM and, ultimately, MM. Notably, the detection of multiple tumour lesions within the BM is a defining feature of MM, with elevated numbers of MRI-detectable lesions being an independent predictor of poor prognosis in asymptomatic [5,6,7] or symptomatic [8,9] myeloma. Furthermore, over two-thirds of newly diagnosed MM patients have detectable circulating PCs, as detected by flow cytometry [10], cytology [11] or slide-based immunofluorescence [12], with higher numbers being an independent predictor of shorter progression-free survival [13,14] and overall survival [10,11,12,13]. Notably, the association between poor outcomes and elevated clonal PCs in the peripheral blood (PB) is independent of BM tumour burden [13], suggesting an active role for circulating PCs in MM disease progression and relapse following therapy. Furthermore, the dissemination of MM PCs is a key feature of aggressive, advanced forms of MM, including in extramedullary disease and plasma cell leukaemia (PCL). PCL is characterised by the presence of very high (>20%) numbers of circulating PCs and is observed in 2–4% of newly diagnosed MM patients [15]. Extramedullary disease, characterised by the dissemination and growth of clonal PCs at soft-tissue sites, occurs in 3–5% of MM cases at diagnosis and increases to up to 20% at relapse (reviewed in Jagosky and Usmani [16]). Importantly, patients with either PCL or extramedullary disease have a poorer prognosis compared with other MM patients [15], highlighting the association between dissemination and aggressive disease.
MGUS and smouldering MM are largely asymptomatic precursor diseases characterised by the presence of clonal PCs in the BM in the absence of evidence of end organ damage that is characteristic of MM and, notably, in a lack of multiple MRI-detectable tumours [1]. Studies using next generation sequencing examining clonal evolution during disease progression from MGUS or smouldering MM to MM suggest that the genetic abnormalities leading to MM are already present at the pre-malignant stages in the majority of patients [17,18,19]. These studies suggest that the outgrowth of dominant PC clones, and their subsequent population of sites throughout the BM, is a key feature of progression to MM. In support of this, increased incidence of circulating PCs in MGUS and smouldering MM patients is associated with progression [20,21]. Taken together, these studies suggest that dissemination of clonal PCs is a key step for the progression to symptomatic disease.
In this review, we will discuss the current understanding of the mechanisms that underpin MM PC dissemination. Furthermore, we will discuss how this knowledge could be used to develop novel therapeutic strategies to delay disease progression and treat high-risk MM patients.

2. The Process of Dissemination in MM

Similar to the process of solid tumour metastasis, the dissemination of MM PCs is associated with a loss of their adherence to cells of the BM microenvironment that favours MM PC retention, allowing the cells to exit the niche. The tumour cells must then undergo trans-endothelial migration, mediated by chemoattractants and adhesive interactions, and intravasate (move from the BM into the bloodstream) where they are carried to a secondary site. In order to home to new BM sites, the tumour cells must arrest on the BM endothelium and extravasate (move out of the blood and into tissues), following chemotactic factors produced by BM cells. The final stage of MM PC dissemination is associated with the colonisation of new BM sites which supports tumour cell growth. The following sections will describe the molecular mechanisms that are involved in MM PC dissemination, focusing on both the intrinsic properties of the MM PCs that support their dissemination, as well as the extrinsic stimuli that drive this process.

2.1. Retention within the BM Stromal Niche

BM mesenchymal stromal cells (BMSCs) are a critical component in the BM niche that supports the growth of MM PCs. Adhesion of MM PCs to BMSCs, and extracellular matrix (ECM) components that are secreted by these cells, is a crucial mechanism by which MM PCs are maintained within the niche (Figure 1). Binding of the integrin α4β1 (also known as very late antigen 4, VLA-4), expressed by MM PCs, to vascular cell–adhesion molecule 1 (VCAM-1) and fibronectin, expressed by BMSCs, is one of the key factors mediating the strong adhesion of MM PCs to BMSCs [22]. Notably, integrin α4β1-mediated binding to fibronectin decreases the response of MM cell lines to chemotactic factors in vitro, supporting its role in MM PC retention [23]. Additionally, the integrin αLβ2 complex (also known as lymphocyte function-associated antigen 1, LFA-1) on MM PCs mediates binding to intracellular adhesion molecule 1 (ICAM-1) on BMSCs [24]. Other adhesive factors expressed by MM PCs include CD44 variants CD44v6 and CD44v9, which mediate adhesion to BMSCs [25], and syndecan-1 (also known as CD138), which is involved in adhesion to type 1 collagen [26].
The adhesion of MM PCs to BMSCs and ECM can be enhanced by exogenous secreted factors which induce rapid cytoskeletal remodelling and conformational changes in integrins, thereby increasing adhesion [22,27,28]. The C-X-C chemokine ligand 12 (CXCL12; also known as stromal-derived factor-1 [SDF-1]), abundantly produced by BMSCs [29,30,31,32], is the ligand for the C-X-C chemokine receptor 4 (CXCR4), which is expressed universally on MM PCs from MM patients [33,34,35]. Treatment of MM PCs with CXCL12 rapidly increases MM PC adhesion to fibronectin and VCAM-1 on the surface of BMSCs, through induction of conformational changes in integrin α4β1 [22,27,28,31]. Furthermore, treatment with the CXCR4 antagonist plerixafor (also known as AMD3100) rapidly mobilises PCs into the blood [36]. Additionally, other factors may increase the adhesion of MM PCs to BMSCs by increasing expression of integrins. For example, the cytokine tumour necrosis factor alpha (TNF-α), secreted by MM PCs, acts in an autocrine fashion to increase expression of integrin α4β1 and integrin αLβ2 complexes on MM PCs, thereby increasing adhesion of MM cell lines to BMSCs in vitro [24]. In addition, the CD40 ligand (CD40L), expressed on haematopoietic cells, binds to the CD40 receptor on MM PCs and stimulates the adhesion of MM cell lines to BMSCs and fibronectin in vitro [37,38], via a mechanism which may involve upregulation of integrin expression [37]. Furthermore, B-cell activating factor (BAFF), which is expressed by BMSCs and osteoclasts, has also been shown to increase the adhesion of MM PCs to BMSCs, although the mechanism involved is unclear [39,40].

2.2. Release from the BM Niche

In order to be released from the BM and enter into the circulation, MM PCs must overcome the aforementioned adhesive interactions that act as a strong BM retention signal (Figure 1). While the microenvironmental stimuli that regulate the release from the BM niche are unclear, decreased expression of key factors involved in adhesion in the stromal niche may play a role. Studies analysing the expression of cell surface adhesion factors in PB PCs compared with BM PCs from MM patients have shown that circulating MM PCs express lower levels of integrin α4β1 compared with BM-resident MM PCs [41,42]. In addition, studies show that there are decreased levels of the activated form of integrin β1 in MM PCs in the PB compared with the BM in MM patients [43], suggesting that downregulation of its active form may in part facilitate release from the BM. Syndecan-1 expression has also been shown to be decreased in PB MM PCs compared with their BM counterparts [41] and MM PC syndecan-1 expression is associated with decreased dissemination in an in vivo model of MM [44]. Notably, treatment with a syndecan-1 blocking antibody rapidly induced the mobilisation of MM PCs from the BM to the PB in a syngeneic mouse model of MM [44]. Additionally, MM cell line expression of the enzyme heparanase-1, which is responsible for cleaving proteoglycans including syndecan-1 from the cell surface, significantly increased the spontaneous dissemination of MM cells in vivo, suggesting that shedding of syndecan-1 may promote dissemination [45]. Finally, PCs from PCL patients have been reported to have decreased CD40 expression compared with MGUS PCs, supporting a potential role for loss of CD40 expression in release of MM PCs from the BM niche [46].

2.3. Microenvironmental Control of Release from the BM Niche

Release from the BM niche may also be facilitated by signals from the micro-environment (Figure 1). The BM becomes increasingly hypoxic during MM tumour growth, with highly hypoxic regions arising within the tumour mass due to rapid tumour cell growth and abnormal blood vessel formation (reviewed by Martin et al. [47]). Moreover, the role of hypoxia in tumour progression, dissemination and angiogenesis has been demonstrated in mouse models of MM [48,49,50]. In particular, Azab and colleagues demonstrated in a mouse model of MM that MM PC hypoxia, as assessed by pimonidazole staining, strongly correlated with both BM tumour burden and numbers of circulating MM PCs [48]. Furthermore, transcriptomic analyses of paired BM and PB tumour samples from MM patients have revealed a hypoxia-related gene expression signature in MM PCs in the circulation, suggesting that hypoxia may selectively induce the mobilisation of MM PCs from the BM [51]. In addition, culturing MM cell lines in hypoxic conditions significantly reduced their adherence to BMSCs or to collagen in vitro [48,52], suggesting a mechanism whereby hypoxia may induce the release of MM PCs from the BM niche. Hypoxia has been shown to increase MM PC expression of the transcription factors Snail and Twist1 that are master regulators of the epithelial to mesenchymal transition (EMT), suggesting that, like in epithelial cancers, an EMT-like process may also be occurring in MM to allow release from the niche [48,53]. Studies by our group have demonstrated that the induction of hypoxia inducible factor HIF-2α in MM cell lines can lead to decreased response to stromal cell-derived CXCL12, which may also facilitate release from the niche. HIF-2α increases CXCL12 expression by MM cell lines [54] which, in turn, reduces CXCR4 cell-surface levels on MM cells [35], forming a feedback loop that leads to a decrease in CXCR4 signalling and a desensitisation to exogenous CXCL12 [35]. In addition, our group has demonstrated that hypoxic activation of HIF-2α leads to upregulation of the C-C chemokine receptor 1 (CCR1) in MM PCs, which may also contribute to their preferential mobilisation [35]. Notably, treatment of CCR1-positive MM cell lines with the CCR1 ligand C-C chemokine ligand 3 (CCL3; also known as macrophage inflammatory protein 1 alpha [MIP-1 α]) abrogates migration towards CXCL12 in vitro, suggesting that CCL3/CCR1 signalling can desensitise cells to exogenous CXCL12 [35]. Furthermore, either CCR1 knockout or treatment with a small molecule CCR1 inhibitor strongly inhibits the spontaneous dissemination of human MM cell lines in an intratibial model of myeloma [55]. Taken together, these studies suggest that hypoxia may mediate a disruption to the CXCR4/CXCL12 retention signal to allow MM PC release from the BM.

2.4. Intravasation

In order to undergo haematogenous dissemination, following release from the BM stromal niche the MM PCs need to migrate towards the vasculature, and then invade and traverse through the basement membrane and the endothelium to enter the blood stream (Figure 1). MM PCs must migrate through and invade basement membranes comprised of type IV collagen, laminin, and other ECM components. Upregulation of MM PC secretion of specific matrix metalloprotease (MMP) enzymes that degrade ECM facilitates invasion through the ECM into the vasculature (Figure 1). MMP-2 and -9, which degrade ECM components including collagen IV and laminin, have been implicated in the invasion of MM PCs [56,57].
BM endothelial cells (BMECs) are sources of secreted factors that are known chemoattractants for MM PCs, which may encourage dissemination from the primary tumour site towards the vasculature (Table 1). Conditioned media from BMECs has been shown to contain chemotactic factors for MM cell lines, including CCL2 (also known as monocyte chemotactic protein-1 [MCP-1]; ligand for CCR2), which induce chemotaxis in MM cell lines and primary MM PCs in vitro [33,58,59]. In support of this, migration of mouse 5T MM cells towards BMEC conditioned media can be blocked using an antibody against CCL2, suggesting that BMEC-derived CCL2 may promote MM PC migration towards the vasculature [58]. The production of promigratory factors by BMECs may create a promigratory stimulus that directs the migration of MM PCs from their niche towards the vasculature, allowing subsequent entry into the blood stream.

2.5. Extravasation and Homing to the BM

Once in the blood stream, MM PCs travel in the circulation until they reach a distant site where, in response to local chemoattractant factors, they adhere to the endothelial layer and then extravasate and migrate to a new BM site (Figure 2).
MM PC adhesion to BMECs is a critical process which enables subsequent extravasation from the vasculature. In the trafficking of lymphocytes, initial reversible adhesive interactions lead to loose “tethering” of the cells to the endothelium, following which the lymphocyte rolls along the endothelium, allowing it to respond to local chemokines which provide the stimulus for extravasation and homing into the BM (Figure 2) (reviewed in Kunkel & Butcher [60]). Tethering and rolling of MM PCs is mediated by the binding of CD44 and P-selectin glycoprotein ligand-1 (PSGL-1) on MM PCs to P-selectin and hyaluronic acid, respectively, on the endothelium [61,62]. Indeed, inhibitors which prevent PSGL-1 interacting with P-selectin have been shown to result in a reduction of in vivo homing following intravenous injection of MM.1S cells [63,64]. Furthermore, treatment with an anti-CD44 antibody resulted in a decrease in 5T myeloma cell homing to the BM following intravenous injection in a mouse model [63]. Studies by our group and those of others have also implicated the homophilic adhesion molecule N-cadherin in the adhesion of MM PCs to the endothelium. shRNA knockdown of N-cadherin in human and murine MM cell lines resulted in a reduction of the adhesion of MM cell lines to BMECs under shear flow [65] without affecting transendothelial migration in vitro [65,66], suggesting a role for N-cadherin in the tethering process, but not in subsequent extravasation. In support of this, N-cadherin knockdown or an N-cadherin blocking peptide decreased the homing of MM cells to the BM following intravenous injection in mice [65,66].
Rolling along the endothelium allows the MM PCs to respond to locally produced chemokines, in particular CXCL12, which induce strong adhesive interactions through stabilising integrin-mediated adhesion to the endothelium and allowing extravasation and homing into the BM (Figure 2). Notably, studies using primary patient samples have demonstrated that CXCR4 is upregulated in MM PCs in the PB, when compared with those in the BM [28,35], potentially enabling enhanced response to local BMEC-derived CXCL12. The preferential arrest of circulating MM PCs in the BM vasculature may, at least in part, be due to the abundant expression of CXCL12 by endothelial cells in the BM (Figure 2) [29,33,67,68]. A murine MM cell line has been shown to preferentially adhere to endothelial cells from the BM, compared with those of other organs, suggesting a preference for the BM as a site of establishment [69]. Notably, Sipkins and colleagues have demonstrated that the MM cell line U266, and other B-cell lines, preferentially extravasates in the BM at specific vascular regions where endothelial expression of CXCL12 is high [68].
CXCL12 produced by BMECs has been shown to induce rapid activation of integrin α4β1 in primary MM PCs and MM cell lines, enabling arrest of the MM PCs through adhesion to BMEC VCAM-1, and subsequently enabling transendothelial migration [70]. Transendothelial migration of human myeloma cell lines in vitro [70] and in vivo [71] and primary MM PCs in vitro [70] could be completely blocked using antibodies against VCAM-1, integrin α4 or integrin β1, suggesting a role for integrin α4β1-VCAM-1 interaction in adhesion to BMECs and subsequent extravasation. In support of this, blocking of integrin α4β1 binding with an antibody delayed the in vivo homing of the human MM cell line MM.1S to the BM following intravenous injection [72]. Adhesion to the endothelium [73] and exposure to CXCL12 [74] also triggers upregulation of MM PC production of MMP9 which facilitates proteolytic degradation of the basement membrane of the endothelium [75] to further facilitate homing to the BM.

2.6. Establishment and Colonisation of MM PCs in a New BM Niche

Following extravasation, MM PCs further respond to locally produced chemokines and growth factors to direct their movement to their ultimate location in the BM (Figure 2). The migration of MM PCs towards specific niches in the BM may be driven by a range of chemoattractant molecules that are produced by BM cells, including BMSCs, osteoclasts and macrophages, which play an important role in the MM BM niche (Table 1). For both normal PCs and MM PCs, CXCL12 represents the predominant signal that is thought to drive homing from the PB into the BM [28,30,76,77] and subsequently also leads to MM PC retention in the BM (described in the retention within the niche section, above). CXCL12 expression is higher in the BM than in the PB [28,35], consistent with its abundant expression by BMSCs [29,30,31,32], establishing a gradient that enables homing to the BM. MM PCs have a strong migratory response towards CXCL12 [27,76], with CXCL12 inducing cytoskeletal remodelling that enables MM PC migration [28,35,70]. Blockade of CXCL12-CXCR4 binding slows homing of human MM cell lines from the PB and accumulation in the BM in vivo [28,78]. BMSCs also produce hepatocyte growth factor (HGF) [79,80,81], which further enhances the chemotactic effect of CXCL12 on MM PCs [82]. Furthermore, BMSC production of exosomes containing chemokines including CCL2, CCL3, and CXCL12 may also play a role in the migration of MM PCs towards BMSCs [83]. BMSC-derived exosomes increase the migration of the mouse myeloma cell line 5T33MM in vitro, which can be reversed with inhibitors of CXCR4 or CCR2 [83]. Insulin-like growth factor 1 (IGF-1) has also been shown to be a key promigratory (chemokinetic) and chemoattractant factor for murine [84] and human [82] MM cell lines in vitro which enhances the response to CXCL12 in vitro [82]. In the mouse 5T MM models, IGF-1 is a critical promoter of homing of MM PCs to the BM [84,85]. Notably, ablation of macrophages, an abundant source of IGF-1 in the BM, is sufficient to inhibit the homing of 5TGM1 cells to the BM [85]. Monocytes and macrophages also highly express the CCR1 ligand CCL3 [86,87], which is a promigratory factor for primary MM PCs and MM cell lines in vitro [76,87,88]. Osteoclasts are also a predominant source of a number of factors that induce MM PC migration in in vitro assays, including CCL3 [87,89], the CCR2 ligands CCL2, CCL7 (also known as monocyte chemotactic protein-3 [MCP-3]) and CCL8 (also known as monocyte chemotactic protein-2 [MCP-2]) [87,90] and the CXCR3 ligand CXCL10 (also known as interferon gamma-induced protein 10 [IP-10]) [76,91,92]. It is yet to be established whether these osteoclast-derived factors play a role in MM PC homing in an in vivo setting. Other cells of the BM microenvironment, including mesenchymal lineage cells such as adipocytes, may also play a role in the BM homing process. For example, adipocyte conditioned media induces the migration of MM cell lines in vitro [93,94,95] at least in part through expression of the chemoattractants CXCL12 and CCL2 [94].
Recent animal studies from our group [103] and those of others [104,105] have shed light on the fate of disseminating clones, and shown that the dissemination process is extremely inefficient: of the hundreds of MM cells that may reach the BM following intravenous injection, very few ultimately proliferate to form macroscopic tumours, with the remaining cells being maintained in a non-proliferative state [103,104,105]. This suggests that the BM microenvironment in which the MM PCs ultimately reside may determine whether an individual tumour cell is destined for dormancy or proliferation [104,106]. At this time, it remains unclear whether the homing of MM PCs to these niches is largely a stochastic process, or whether MM PCs are driven by specific microenvironment-derived factors to home to defined niches that ultimately determine their fate for dormancy or growth.

3. Intrinsic MM Characteristics that May Facilitate Dissemination

There is growing evidence that there an association between certain chromosomal translocations that are frequently observed in MM patients, including t(14;16), t(14;20) and t(4;14), and the incidence of elevated circulating PCs or PCL [14,107,108,109,110,111,112] suggesting that these chromosomal translocations may result in an increased propensity for MM PC dissemination (Figure 3; Table 2). Additionally, t(11;14) is also highly represented in PCL [107,108,109], but is not associated with increased PB MM cell numbers in MM patients [113]; the mechanism that underlies the propensity for development of PCL in these patients remains unclear. As discussed below, there is increasing evidence for functional differences arising from t(14;16), t(14;20), and t(4;14) translocations which may underlie the propensity for dissemination observed in these genetic subtypes of MM.

3.1. t(14;16) and t(14;20)

The chromosomal translocations t(14;16) and t(14;20) lead to constitutive overexpression of the transcription factors MAF and MAFB, respectively. Notably, 15% of t(14;16) MM patients present with PCL, compared with 1.5% of non-t(14;16) MM patients [111]. Furthermore, the t(14;16) translocation is seen 6.5–9% of patients with elevated PB MM cells, compared with only 2–3.7% of patients with low PB MM cells [14,113], suggesting an association with increased dissemination. The relative infrequency of t(14;20) translocations has precluded statistical analysis of the association between MM PC dissemination and t(14;20); however, t(14;20) has been reported in 3% of MM patients with elevated PB PCs [14] and 2.5% of PCL patients [108], compared with 1.0–1.5% of all MM patients [14,108], suggesting a potentially increased incidence of dissemination in these patients.
Transcriptome profiling has revealed that elevation of expression of two genes, the integrin ITGB7 and the chemokine receptor CX3CR1, are highly characteristic of MM patients and human MM cell lines with MAF or MAFB translocations [114,115,116,119]. Overexpression and siRNA knockdown studies have demonstrated that MAF regulates integrin β7 expression in human MM cell lines [116]. Integrin β7 can form a heterodimer with integrin αE, with the αEβ7 complex regulating adhesion to E-cadherin on the surface of BMSCs [116,120]. Blockade of E-cadherin on BMSCs [48,116] or of integrin β7 on human MM cell lines [116,120] decreases the adhesion of MM PCs to BMSCs in vitro, suggesting that this interaction may play a role in adhesion and retention in the niche. Additionally, the α4β7 integrin complex is involved in adhesion to VCAM-1 [121], suggesting a potential role for integrin β7 expression in adhesion to endothelial cells. shRNA knockdown of integrin β7 decreases the migration of human MM cell lines towards CXCL12 in vitro and, furthermore, delays BM homing following intravenous injection in vivo [120]. Elevated integrin β7 expression in MM PCs may, therefore, contribute to the increased dissemination seen in patients with aberrant MAF expression. Microarray data analyses have also shown that expression of the chemokine receptor CX3CR1 is a recurrent feature of patients with t(14;16) [114,115]. The ligand for CX3CR1, CX3CL1, can be released in a soluble form where it can act as a chemoattractant or, alternatively, can be presented in a membrane-bound form, where it facilitates cell-cell adhesion [122]. Notably, CX3CL1/CX3CR1 binding has been implicated in playing a key role in the transendothelial migration of lymphocytes by increasing endothelial cell adhesion and migration towards chemoattractants [123]. While the functional role for CX3CR1 in MM is unclear, the human MM cell line RPMI-8226 has been shown to bind to CX3CL1 in vitro under shear flow, suggesting that it may play a role in adhesion to endothelial cells in MM [124]. Gene expression profiling studies have also identified elevated expression of IGF1R and CCR1, the receptors for the MM PC chemoattractants IGF-1 and CCL3, respectively, are also elevated in t(14;16) human MM cell lines and primary MM PCs [99,116]. The expression of these key promigratory receptors may also, therefore, play a role in the MM PC dissemination in t(14;16) patients.

3.2. t(4;14)

The chromosomal translocation t(4;14), which leads to constitutive overexpression of the histone methyltransferase NSD2 (also known as WHSC1 and MMSET), is also associated with high risk disease and increased dissemination in MM patients. The incidence of patients with t(4;14) is 2- to 4-fold higher in patients with elevated circulating PCs, compared with those with low circulating PCs [11,14,113,125]. Additionally, there is some evidence for an increase in the incidence of t(4;14) in PCL patients [109] and t(4;14) MM is also associated with an increased incidence of extramedullary PC tumours [126].
Notably, the t(4;14) translocation is associated with an EMT-like gene expression signature in MM PCs, characterised by upregulation of mesenchymal genes, including those for N-cadherin and vimentin, and the transcription factor Twist-1 [53,66,117,118]. N-cadherin knockdown studies in MM cell lines have shown that N-cadherin is important in the homing of MM PCs from the vasculature to the BM [65,66]. In addition, studies from our group have shown that overexpression of Twist-1 increases MM PCs dissemination in an intratibial model of MM in vivo [53]. Like t(14;16) patients, tumour cells from t(4;14) patients and MM cell lines have been shown to have increased expression of IGF1R and CCR1 receptors [99,100,116] which may also contribute to the increased propensity for dissemination of t(4;14) MM PC.

3.3. Subclonal Heterogeneity and Dissemination

There is evidence to suggest that, in addition to these chromosomal translocations, other secondary chromosomal abnormalities and mutations may increase the ability of individual subclones to disseminate [41,127,128,129]. Using FISH or whole exome sequencing on paired BM and PB PC samples from MM patients, several studies have found that in some instances subclones present in the BM were not present in the PB, and vice versa [41,128,129]. However, it remains to be seen whether the subclonal differences observed between the BM and PB in MM patients are due to selective microenvironmental pressures, such as hypoxia or other as yet uncharacterised stimuli, that drive the efflux of a subclone in a particular microenvironment, or if certain secondary copy number changes or SNVs may increase the propensity for dissemination in MM, leading to an increased likelihood for dissemination of particular subclones. Furthermore, it is also possible that the observed subclonal differences may be due to the inability of single-site BM biopsy to capture the spatial heterogeneity seen in this disease [130].
There is some evidence that points to mechanisms whereby certain subclones could have an increased propensity for dissemination. Deletion of chromosome 17p13 [del(17p)] occurs in approximately 10% of newly diagnosed MM patients [109,110,113,131]. Missense mutations in TP53, on 17p, are seen in approximately 19% of patients with del(17p), leading to a complete inactivation of functional p53 [132]. There is some evidence for an increased incidence of del(17p) in PCL patients [109], although this has not been reported in MM patients with elevated PB PC [11,113]. In a study utilising whole exome sequencing of paired BM and PB tumour samples from 4 patients, Manier et al. identified one patient with 17p loss across the BM and PB sample with a TP53 missense mutation which was only detectable in the PB sample, suggesting a potential association between loss of p53 function and increased the propensity for dissemination [129]. In support of this, siRNA-mediated knockdown of p53 in NCI-H929 cells has been shown to increase invasion through Matrigel and decreased adhesion to BMSCs in vitro [133]. Although more studies are needed, these data suggest a potential role for loss of p53 in the dissemination of MM PC.

4. Future Perspectives

One of the defining features of symptomatic MM is the presence of MM tumours at multiple sites in the BM [1]. In over two-thirds of newly diagnosed MM patients, circulating MM PCs are detectable in the PB [10,11,12], with higher numbers being associated with poorer overall survival [10,11,12,13]. Furthermore, highly disseminated disease, as characterised by elevated circulating MM PCs and extramedullary tumours, is a feature of advanced disease and is associated with poorer overall survival [14,111,134,135]. There is also evidence to suggest that dissemination of therapy-resistant clones leads to shorter time to relapse, with higher numbers of circulating tumour cells at baseline being indicative of shorter progression-free survival, regardless of the therapy used [13,14,136,137].
These studies suggest that therapies which target key processes involved in MM PC dissemination may be useful to delay disease progression or as a maintenance therapy to extend progression-free survival following frontline therapy. To this end, studies suggest that therapeutic targeting adhesion molecules or chemokine receptors may be able to inhibit the ability of MM cells to egress from or to home to the BM. For example, selective blocking of adhesion molecules involved in the adhesion to endothelium, such as integrins and N-cadherin, have been shown to inhibit homing and thereby decrease subsequent tumour burden following intravenous injection of tumour cells in mouse models of MM [63,65]. Additionally, blockade of CXCL12/CXCR4 with plerixafor or a CXCL12-targeting L-RNA aptamer has been shown to decrease BM tumour establishment following intravenous injection in mouse models of MM [28,78]. However, paradoxically, CXCL12/CXCR4 blockade also mobilises MM PC from the BM niche in tumour bearing mice [36,78] and in patients [138], which could inadvertently increase tumour dissemination. Recent studies from our group have shown that treatment with a small molecule inhibitor of the chemokine receptor CCR1, CCX9588, can prevent the dissemination of MM PCs from the BM in intratibial MM models in vivo [55]. As far as we are aware, this is the first preclinical demonstration of a therapeutic intervention that can inhibit spontaneous dissemination of MM cells in vivo. However, it remains to be seen whether therapeutic targeting of dissemination will translate to a beneficial impact on subsequent disease progression and, to date, no anti-dissemination treatment strategies have reached clinical trial for MM.
An alternative approach could be to therapeutically target hypoxic MM cells before they disseminate. Evofosfamide (TH-302), a DNA alkylator designed to be selectively activated under hypoxic conditions, has been shown to cause apoptosis of MM cell lines when cultured under hypoxia in vitro and to inhibit MM tumour development in vivo [50,139]. In a Phase I/II trial in relapsed/refractory myeloma patients, treatment with evofosfamide in combination with dexamethasone, with or without bortezomib, was associated with a modest response rate, with partial or better response observed in 12% of these heavily pre-treated patients [140]. Taken together, these studies suggest that targeting of hypoxic MM PCs could be investigated as a therapeutic strategy to prevent MM PC dissemination.

5. Conclusions

In conclusion, this review has highlighted the current understanding of the processes involved in MM dissemination. Some key outstanding questions include: Are there environmental factors other than hypoxia that drive dissemination/mobilisation? Is underlying subclonal heterogeneity leading to mobilisation of specific individual subclones during dissemination? What are the features of the BM microenvironment that support the homing and subsequent growth of MM PCs at specific niches? A greater understanding the process of MM PC dissemination may allow the identification of novel therapeutic targets that could improve outcomes for MM patients, especially for the prevention of disease progression and relapse in high-risk patients.

Author Contributions

Conceptualisation, M.N.Z. and K.V.; writing—original draft preparation, M.N.Z.; writing—review and editing, K.V. and A.C.W.Z. All authors have read and agreed to the published version of the manuscript.

Funding

M.N.Z. was supported by Chun Chung Wong and So Sau Lam Memorial Postgraduate Cancer Research Top-Up Scholarship from the Florey Medical Research Foundation, a Short-Term Research Grant from the German Academic Exchange Service (DAAD) and a Hans-Jürgen and Marianne Ohff Research Grant from the University of Adelaide. K.V. was supported by an Early Career Cancer Research Fellowship from the Cancer Council SA Beat Cancer Project on behalf of its donors and the State Government of South Australia through the Department of Health.

Acknowledgments

Figures were created using BioRender.com.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Rajkumar, S.V.; Dimopoulos, M.A.; Palumbo, A.; Blade, J.; Merlini, G.; Mateos, M.V.; Kumar, S.; Hillengass, J.; Kastritis, E.; Richardson, P.; et al. International Myeloma Working Group updated criteria for the diagnosis of multiple myeloma. Lancet Oncol. 2014, 15, e538–e548. [Google Scholar] [CrossRef]
  2. Ferlay, J.; Soerjomataram, I.; Dikshit, R.; Eser, S.; Mathers, C.; Rebelo, M.; Parkin, D.M.; Forman, D.; Bray, F. Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int. J. Cancer 2015, 136, 359–386. [Google Scholar] [CrossRef] [PubMed]
  3. Cowan, A.J.; Allen, C.; Barac, A.; Basaleem, H.; Bensenor, I.; Curado, M.P.; Foreman, K.; Gupta, R.; Harvey, J.; Hosgood, H.D.; et al. Global Burden of Multiple Myeloma: A Systematic Analysis for the Global Burden of Disease Study 2016. JAMA Oncol. 2018, 4, 1221–1227. [Google Scholar] [CrossRef] [Green Version]
  4. Noll, J.E.; Williams, S.A.; Purton, L.E.; Zannettino, A.C.W. Tug of war in the haematopoietic stem cell niche: Do myeloma plasma cells compete for the HSC niche? Blood Cancer J. 2012, 2, e91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Hillengass, J.; Fechtner, K.; Weber, M.A.; Bauerle, T.; Ayyaz, S.; Heiss, C.; Hielscher, T.; Moehler, T.M.; Egerer, G.; Neben, K.; et al. Prognostic significance of focal lesions in whole-body magnetic resonance imaging in patients with asymptomatic multiple myeloma. J. Clin. Oncol. 2010, 28, 1606–1610. [Google Scholar] [CrossRef] [PubMed]
  6. Dhodapkar, M.V.; Sexton, R.; Waheed, S.; Usmani, S.; Papanikolaou, X.; Nair, B.; Petty, N.; Shaughnessy, J.D., Jr.; Hoering, A.; Crowley, J.; et al. Clinical, genomic, and imaging predictors of myeloma progression from asymptomatic monoclonal gammopathies (SWOG S0120). Blood 2014, 123, 78–85. [Google Scholar] [CrossRef] [Green Version]
  7. Kastritis, E.; Moulopoulos, L.A.; Terpos, E.; Koutoulidis, V.; Dimopoulos, M.A. The prognostic importance of the presence of more than one focal lesion in spine MRI of patients with asymptomatic (smoldering) multiple myeloma. Leukemia 2014, 28, 2402–2403. [Google Scholar] [CrossRef]
  8. Walker, R.; Barlogie, B.; Haessler, J.; Tricot, G.; Anaissie, E.; Shaughnessy, J.D., Jr.; Epstein, J.; van Hemert, R.; Erdem, E.; Hoering, A.; et al. Magnetic resonance imaging in multiple myeloma: Diagnostic and clinical implications. J. Clin. Oncol. 2007, 25, 1121–1128. [Google Scholar] [CrossRef] [Green Version]
  9. Mai, E.K.; Hielscher, T.; Kloth, J.K.; Merz, M.; Shah, S.; Raab, M.S.; Hillengass, M.; Wagner, B.; Jauch, A.; Hose, D.; et al. A magnetic resonance imaging-based prognostic scoring system to predict outcome in transplant-eligible patients with multiple myeloma. Haematologica 2015, 100, 818–825. [Google Scholar] [CrossRef] [Green Version]
  10. Nowakowski, G.S.; Witzig, T.E.; Dingli, D.; Tracz, M.J.; Gertz, M.A.; Lacy, M.Q.; Lust, J.A.; Dispenzieri, A.; Greipp, P.R.; Kyle, R.A.; et al. Circulating plasma cells detected by flow cytometry as a predictor of survival in 302 patients with newly diagnosed multiple myeloma. Blood 2005, 106, 2276–2279. [Google Scholar] [CrossRef] [Green Version]
  11. Granell, M.; Calvo, X.; Garcia-Guiñón, A.; Escoda, L.; Abella, E.; Martínez, C.M.; Teixidó, M.; Gimenez, M.T.; Senín, A.; Sanz, P.; et al. Prognostic impact of circulating plasma cells in patients with multiple myeloma: Implications for plasma cell leukemia definition. Haematologica 2017, 102, 1099. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Witzig, T.E.; Gertz, M.A.; Lust, J.A.; Kyle, R.A.; O’Fallon, W.M.; Greipp, P.R. Peripheral blood monoclonal plasma cells as a predictor of survival in patients with multiple myeloma. Blood 1996, 88, 1780–1787. [Google Scholar] [CrossRef] [PubMed]
  13. Dingli, D.; Nowakowski, G.S.; Dispenzieri, A.; Lacy, M.Q.; Hayman, S.R.; Rajkumar, S.V.; Greipp, P.R.; Litzow, M.R.; Gastineau, D.A.; Witzig, T.E.; et al. Flow cytometric detection of circulating myeloma cells before transplantation in patients with multiple myeloma: A simple risk stratification system. Blood 2006, 107, 3384–3388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Gonsalves, W.I.; Rajkumar, S.V.; Gupta, V.; Morice, W.G.; Timm, M.M.; Singh, P.P.; Dispenzieri, A.; Buadi, F.K.; Lacy, M.Q.; Kapoor, P.; et al. Quantification of clonal circulating plasma cells in newly diagnosed multiple myeloma: Implications for redefining high-risk myeloma. Leukemia 2014, 28, 2060–2065. [Google Scholar] [CrossRef] [Green Version]
  15. Fernandez de Larrea, C.; Kyle, R.A.; Durie, B.G.; Ludwig, H.; Usmani, S.; Vesole, D.H.; Hajek, R.; San Miguel, J.F.; Sezer, O.; Sonneveld, P.; et al. Plasma cell leukemia: Consensus statement on diagnostic requirements, response criteria and treatment recommendations by the International Myeloma Working Group. Leukemia 2013, 27, 780–791. [Google Scholar] [CrossRef] [Green Version]
  16. Jagosky, M.H.; Usmani, S.Z. Extramedullary Disease in Multiple Myeloma. Curr. Hematolog. Malig. Rep. 2020, 15, 62–71. [Google Scholar] [CrossRef]
  17. Dutta, A.K.; Fink, J.L.; Grady, J.P.; Morgan, G.J.; Mullighan, C.G.; To, L.B.; Hewett, D.R.; Zannettino, A.C.W. Subclonal evolution in disease progression from MGUS/SMM to multiple myeloma is characterised by clonal stability. Leukemia 2018, 33, 457–468. [Google Scholar] [CrossRef]
  18. Walker, B.A.; Wardell, C.P.; Melchor, L.; Brioli, A.; Johnson, D.C.; Kaiser, M.F.; Mirabella, F.; Lopez-Corral, L.; Humphray, S.; Murray, L.; et al. Intraclonal heterogeneity is a critical early event in the development of myeloma and precedes the development of clinical symptoms. Leukemia 2013, 28, 384. [Google Scholar] [CrossRef] [Green Version]
  19. Bustoros, M.; Sklavenitis-Pistofidis, R.; Park, J.; Redd, R.; Zhitomirsky, B.; Dunford, A.J.; Salem, K.; Tai, Y.T.; Anand, S.; Mouhieddine, T.H.; et al. Genomic Profiling of Smoldering Multiple Myeloma Identifies Patients at a High Risk of Disease Progression. J. Clin. Oncol. 2020, 38, 2380–2389. [Google Scholar] [CrossRef]
  20. Bianchi, G.; Kyle, R.A.; Larson, D.R.; Witzig, T.E.; Kumar, S.; Dispenzieri, A.; Morice, W.G.; Rajkumar, S.V. High levels of peripheral blood circulating plasma cells as a specific risk factor for progression of smoldering multiple myeloma. Leukemia 2013, 27, 680–685. [Google Scholar] [CrossRef]
  21. Gonsalves, W.I.; Rajkumar, S.V.; Dispenzieri, A.; Dingli, D.; Timm, M.M.; Morice, W.G.; Lacy, M.Q.; Buadi, F.K.; Go, R.S.; Leung, N.; et al. Quantification of circulating clonal plasma cells via multiparametric flow cytometry identifies patients with smoldering multiple myeloma at high risk of progression. Leukemia 2017, 31, 130–135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Sanz-Rodríguez, F.; Ruiz-Velasco, N.; Pascual-Salcedo, D.; Teixidó, J. Characterization of VLA-4-dependent myeloma cell adhesion to fibronectin and VCAM-1. Br. J. Haematol. 1999, 107, 825–834. [Google Scholar] [CrossRef] [PubMed]
  23. Shibayama, H.; Tagawa, S.; Hattori, H.; Inoue, R.; Katagiri, S.; Kitani, T. Laminin and fibronectin promote the chemotaxis of human malignant plasma cell lines. Blood 1995, 86, 719–725. [Google Scholar] [CrossRef] [PubMed]
  24. Hideshima, T.; Chauhan, D.; Schlossman, R.; Richardson, P.; Anderson, K.C. The role of tumor necrosis factor alpha in the pathophysiology of human multiple myeloma: Therapeutic applications. Oncogene 2001, 20, 4519–4527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Van Driel, M.; Günthert, U.; van Kessel, A.C.; Joling, P.; Stauder, R.; Lokhorst, H.M.; Bloem, A.C. CD44 variant isoforms are involved in plasma cell adhesion to bone marrow stromal cells. Leukemia 2002, 16, 135. [Google Scholar] [CrossRef] [Green Version]
  26. Ridley, R.C.; Xiao, H.; Hata, H.; Woodliff, J.; Epstein, J.; Sanderson, R.D. Expression of syndecan regulates human myeloma plasma cell adhesion to type I collagen. Blood 1993, 81, 767–774. [Google Scholar] [CrossRef] [Green Version]
  27. Sanz-Rodriguez, F.; Hidalgo, A.; Teixido, J. Chemokine stromal cell-derived factor-1alpha modulates VLA-4 integrin-mediated multiple myeloma cell adhesion to CS-1/fibronectin and VCAM-1. Blood 2001, 97, 346–351. [Google Scholar] [CrossRef] [Green Version]
  28. Alsayed, Y.; Ngo, H.; Runnels, J.; Leleu, X.; Singha, U.K.; Pitsillides, C.M.; Spencer, J.A.; Kimlinger, T.; Ghobrial, J.M.; Jia, X.; et al. Mechanisms of regulation of CXCR4/SDF-1 (CXCL12)-dependent migration and homing in multiple myeloma. Blood 2007, 109, 2708–2717. [Google Scholar] [CrossRef]
  29. Ponomaryov, T.; Peled, A.; Petit, I.; Taichman, R.S.; Habler, L.; Sandbank, J.; Arenzana-Seisdedos, F.; Magerus, A.; Caruz, A.; Fujii, N.; et al. Induction of the chemokine stromal-derived factor-1 following DNA damage improves human stem cell function. J. Clin. Investig. 2000, 106, 1331–1339. [Google Scholar] [CrossRef] [Green Version]
  30. Tokoyoda, K.; Egawa, T.; Sugiyama, T.; Choi, B.I.; Nagasawa, T. Cellular niches controlling B lymphocyte behavior within bone marrow during development. Immunity 2004, 20, 707–718. [Google Scholar] [CrossRef] [Green Version]
  31. Nakayama, T.; Hieshima, K.; Izawa, D.; Tatsumi, Y.; Kanamaru, A.; Yoshie, O. Cutting edge: Profile of chemokine receptor expression on human plasma cells accounts for their efficient recruitment to target tissues. J. Immunol. 2003, 170, 1136–1140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Honczarenko, M.; Le, Y.; Swierkowski, M.; Ghiran, I.; Glodek, A.M.; Silberstein, L.E. Human bone marrow stromal cells express a distinct set of biologically functional chemokine receptors. Stem Cells 2006, 24, 1030–1041. [Google Scholar] [CrossRef] [PubMed]
  33. Pellegrino, A.; Ria, R.; Di Pietro, G.; Cirulli, T.; Surico, G.; Pennisi, A.; Morabito, F.; Ribatti, D.; Vacca, A. Bone marrow endothelial cells in multiple myeloma secrete CXC-chemokines that mediate interactions with plasma cells. Br. J. Haematol. 2005, 129, 248–256. [Google Scholar] [CrossRef] [PubMed]
  34. Vande Broek, I.; Leleu, X.; Schots, R.; Facon, T.; Vanderkerken, K.; Van Camp, B.; Van Riet, I. Clinical significance of chemokine receptor (CCR1, CCR2 and CXCR4) expression in human myeloma cells: The association with disease activity and survival. Haematologica 2006, 91, 200–206. [Google Scholar] [PubMed]
  35. Vandyke, K.; Zeissig, M.N.; Hewett, D.R.; Martin, S.K.; Mrozik, K.M.; Cheong, C.M.; Diamond, P.; To, L.B.; Gronthos, S.; Peet, D.J.; et al. HIF-2α promotes dissemination of plasma cells in multiple myeloma by regulating CXCL12/CXCR4 and CCR1. Cancer Res. 2017, 77, 5452–5463. [Google Scholar] [CrossRef] [Green Version]
  36. Azab, A.K.; Runnels, J.M.; Pitsillides, C.; Moreau, A.-S.; Azab, F.; Leleu, X.; Jia, X.; Wright, R.; Ospina, B.; Carlson, A.L.; et al. CXCR4 inhibitor AMD3100 disrupts the interaction of multiple myeloma cells with the bone marrow microenvironment and enhances their sensitivity to therapy. Blood 2009, 113, 4341–4351. [Google Scholar] [CrossRef] [Green Version]
  37. Urashima, M.; Chauhan, D.; Uchiyama, H.; Freeman, G.J.; Anderson, K.C. CD40 ligand triggered interleukin-6 secretion in multiple myeloma. Blood 1995, 85, 1903–1912. [Google Scholar] [CrossRef]
  38. Tai, Y.T.; Podar, K.; Mitsiades, N.; Lin, B.; Mitsiades, C.; Gupta, D.; Akiyama, M.; Catley, L.; Hideshima, T.; Munshi, N.C.; et al. CD40 induces human multiple myeloma cell migration via phosphatidylinositol 3-kinase/AKT/NF-kappa B signaling. Blood 2003, 101, 2762–2769. [Google Scholar] [CrossRef]
  39. Moreaux, J.; Cremer, F.W.; Reme, T.; Raab, M.; Mahtouk, K.; Kaukel, P.; Pantesco, V.; De Vos, J.; Jourdan, E.; Jauch, A.; et al. The level of TACI gene expression in myeloma cells is associated with a signature of microenvironment dependence versus a plasmablastic signature. Blood 2005, 106, 1021–1030. [Google Scholar] [CrossRef] [Green Version]
  40. Tai, Y.T.; Li, X.F.; Breitkreutz, I.; Song, W.; Neri, P.; Catley, L.; Podar, K.; Hideshima, T.; Chauhan, D.; Raje, N.; et al. Role of B-cell-activating factor in adhesion and growth of human multiple myeloma cells in the bone marrow microenvironment. Cancer Res. 2006, 66, 6675–6682. [Google Scholar] [CrossRef] [Green Version]
  41. Paiva, B.; Paino, T.; Sayagues, J.M.; Garayoa, M.; San-Segundo, L.; Martin, M.; Mota, I.; Sanchez, M.L.; Barcena, P.; Aires-Mejia, I.; et al. Detailed characterization of multiple myeloma circulating tumor cells shows unique phenotypic, cytogenetic, functional, and circadian distribution profile. Blood 2013, 122, 3591–3598. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Paiva, B.; Perez-Andres, M.; Vidriales, M.B.; Almeida, J.; de las Heras, N.; Mateos, M.V.; Lopez-Corral, L.; Gutierrez, N.C.; Blanco, J.; Oriol, A.; et al. Competition between clonal plasma cells and normal cells for potentially overlapping bone marrow niches is associated with a progressively altered cellular distribution in MGUS vs myeloma. Leukemia 2011, 25, 697–706. [Google Scholar] [CrossRef] [Green Version]
  43. Luque, R.; Garcia-Trujillo, J.A.; Camara, C.; Moreno, A.; Eiras, P.; Roy, G.; Villar, L.M.; Lombardia, M.; Brieva, J.A.; Bootello, A.; et al. CD106 and activated-CD29 are expressed on myelomatous bone marrow plasma cells and their downregulation is associated with tumour progression. Br. J. Haematol. 2002, 119, 70–78. [Google Scholar] [CrossRef] [PubMed]
  44. Akhmetzyanova, I.; McCarron, M.J.; Parekh, S.; Chesi, M.; Bergsagel, P.L.; Fooksman, D.R. Dynamic CD138 surface expression regulates switch between myeloma growth and dissemination. Leukemia 2020, 34, 245–256. [Google Scholar] [CrossRef] [Green Version]
  45. Yang, Y.; Macleod, V.; Bendre, M.; Huang, Y.; Theus, A.M.; Miao, H.Q.; Kussie, P.; Yaccoby, S.; Epstein, J.; Suva, L.J.; et al. Heparanase promotes the spontaneous metastasis of myeloma cells to bone. Blood 2005, 105, 1303–1309. [Google Scholar] [CrossRef] [PubMed]
  46. Pérez-Andrés, M.; Almeida, J.; Martín-Ayuso, M.; Moro, M.J.; Martín-Nuñez, G.; Galende, J.; Borrego, D.; Rodríguez, M.J.; Ortega, F.; Hernandez, J.; et al. Clonal plasma cells from monoclonal gammopathy of undetermined significance, multiple myeloma and plasma cell leukemia show different expression profiles of molecules involved in the interaction with the immunological bone marrow microenvironment. Leukemia 2005, 19, 449. [Google Scholar] [CrossRef] [Green Version]
  47. Martin, S.K.; Diamond, P.; Gronthos, S.; Peet, D.J.; Zannettino, A.C. The emerging role of hypoxia, HIF-1 and HIF-2 in multiple myeloma. Leukemia 2011, 25, 1533–1542. [Google Scholar] [CrossRef] [Green Version]
  48. Azab, A.K.; Hu, J.; Quang, P.; Azab, F.; Pitsillides, C.M.; Awwad, R.; Thompson, B.; Maiso, P.; Sun, J.D.; Hart, C.P.; et al. Hypoxia promotes dissemination of multiple myeloma through acquisition of epithelial to mesenchymal transition-like features. Blood 2012, 119, 5782–5794. [Google Scholar] [CrossRef] [Green Version]
  49. Storti, P.; Bolzoni, M.; Donofrio, G.; Airoldi, I.; Guasco, D.; Toscani, D.; Martella, E.; Lazzaretti, M.; Mancini, C.; Agnelli, L.; et al. Hypoxia-inducible factor (HIF)-1alpha suppression in myeloma cells blocks tumoral growth in vivo inhibiting angiogenesis and bone destruction. Leukemia 2013, 27, 1697–1706. [Google Scholar] [CrossRef] [Green Version]
  50. Hu, J.; Handisides, D.R.; Van Valckenborgh, E.; De Raeve, H.; Menu, E.; Vande Broek, I.; Liu, Q.; Sun, J.D.; Van Camp, B.; Hart, C.P.; et al. Targeting the multiple myeloma hypoxic niche with TH-302, a hypoxia-activated prodrug. Blood 2010, 116, 1524–1527. [Google Scholar] [CrossRef] [Green Version]
  51. Garces, J.J.; Simicek, M.; Vicari, M.; Brozova, L.; Burgos, L.; Bezdekova, R.; Alignani, D.; Calasanz, M.J.; Growkova, K.; Goicoechea, I.; et al. Transcriptional profiling of circulating tumor cells in multiple myeloma: A new model to understand disease dissemination. Leukemia 2020, 34, 589–603. [Google Scholar] [CrossRef] [PubMed]
  52. Kawano, Y.; Kikukawa, Y.; Fujiwara, S.; Wada, N.; Okuno, Y.; Mitsuya, H.; Hata, H. Hypoxia reduces CD138 expression and induces an immature and stem cell-like transcriptional program in myeloma cells. Int. J. Oncol. 2013, 43, 1809–1816. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Cheong, C.M.; Mrozik, K.M.; Hewett, D.R.; Bell, E.; Panagopoulos, V.; Noll, J.E.; Licht, J.D.; Gronthos, S.; Zannettino, A.C.W.; Vandyke, K. Twist-1 is upregulated by NSD2 and contributes to tumour dissemination and an epithelial-mesenchymal transition-like gene expression signature in t(4;14)-positive multiple myeloma. Cancer Lett. 2020, 475, 99–108. [Google Scholar] [CrossRef] [PubMed]
  54. Martin, S.K.; Diamond, P.; Williams, S.A.; To, L.B.; Peet, D.J.; Fujii, N.; Gronthos, S.; Harris, A.L.; Zannettino, A.C.W. Hypoxia-inducible factor-2 is a novel regulator of aberrant CXCL12 expression in multiple myeloma plasma cells. Haematologica 2010, 95, 776–784. [Google Scholar] [CrossRef] [Green Version]
  55. Zeissig, M.N.; Hewett, D.R.; Panagopoulos, V.; Mrozik, K.M.; To, L.B.; Croucher, P.I.; Zannettino, A.C.W.; Vandyke, K. Expression of the chemokine receptor CCR1 promotes the dissemination of multiple myeloma plasma cells in vivo. Haematologica 2020. [Google Scholar] [CrossRef]
  56. Barille, S.; Akhoundi, C.; Collette, M.; Mellerin, M.P.; Rapp, M.J.; Harousseau, J.L.; Bataille, R.; Amiot, M. Metalloproteinases in multiple myeloma: Production of matrix metalloproteinase-9 (MMP-9), activation of proMMP-2, and induction of MMP-1 by myeloma cells. Blood 1997, 90, 1649–1655. [Google Scholar] [CrossRef]
  57. Asosingh, K.; Menu, E.; Van Valckenborgh, E.; Vande Broek, I.; Van Riet, I.; Van Camp, B.; Vanderkerken, K. Mechanisms involved in the differential bone marrow homing of CD45 subsets in 5T murine models of myeloma. Clin. Exp. Metastasis 2002, 19, 583. [Google Scholar] [CrossRef]
  58. Vanderkerken, K.; Vande Broek, I.; Eizirik, D.L.; Van Valckenborgh, E.; Asosingh, K.; Van Riet, I.; Van Camp, B. Monocyte chemoattractant protein-1 (MCP-1), secreted by bone marrow endothelial cells, induces chemoattraction of 5T multiple myeloma cells. Clin. Exp. Metastasis 2002, 19, 87–90. [Google Scholar] [CrossRef]
  59. Johrer, K.; Janke, K.; Krugmann, J.; Fiegl, M.; Greil, R. Transendothelial migration of myeloma cells is increased by tumor necrosis factor (TNF)-alpha via TNF receptor 2 and autocrine up-regulation of MCP-1. Clin. Cancer Res. 2004, 10, 1901–1910. [Google Scholar] [CrossRef] [Green Version]
  60. Kunkel, E.J.; Butcher, E.C. Plasma-cell homing. Nat. Rev. Immunol. 2003, 3, 822–829. [Google Scholar] [CrossRef]
  61. Okada, T.; Hawley, R.G.; Kodaka, M.; Okuno, H. Significance of VLA-4-VCAM-1 interaction and CD44 for transendothelial invasion in a bone marrow metastatic myeloma model. Clin. Exp. Metastasis 1999, 17, 623–629. [Google Scholar] [CrossRef] [PubMed]
  62. Yago, T.; Shao, B.; Miner, J.J.; Yao, L.; Klopocki, A.G.; Maeda, K.; Coggeshall, K.M.; McEver, R.P. E-selectin engages PSGL-1 and CD44 through a common signaling pathway to induce integrin αLβ2-mediated slow leukocyte rolling. Blood 2010, 116, 485–494. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Asosingh, K.; Günthert, U.; De Raeve, H.; Van Riet, I.; Van Camp, B.; Vanderkerken, K. A unique pathway in the homing of murine multiple myeloma cells: CD44v10 mediates binding to bone marrow endothelium. Cancer Res. 2001, 61, 2862. [Google Scholar] [PubMed]
  64. Azab, A.K.; Quang, P.; Azab, F.; Pitsillides, C.; Thompson, B.; Chonghaile, T.; Patton, J.T.; Maiso, P.; Monrose, V.; Sacco, A.; et al. P-selectin glycoprotein ligand regulates the interaction of multiple myeloma cells with the bone marrow microenvironment. Blood 2012, 119, 1468–1478. [Google Scholar] [CrossRef] [PubMed]
  65. Mrozik, K.M.; Cheong, C.M.; Hewett, D.; Chow, A.W.; Blaschuk, O.W.; Zannettino, A.C.; Vandyke, K. Therapeutic targeting of N-cadherin is an effective treatment for multiple myeloma. Br. J. Haematol. 2015, 171, 387–399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Groen, R.W.J.; de Rooij, M.F.M.; Kocemba, K.A.; Reijmers, R.M.; de Haan-Kramer, A.; Overdijk, M.B.; Aalders, L.; Rozemuller, H.; Martens, A.C.M.; Bergsagel, P.L.; et al. N-cadherin-mediated interaction with multiple myeloma cells inhibits osteoblast differentiation. Haematologica 2011, 96, 1653–1661. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Imai, K.; Kobayashi, M.; Wang, J.; Shinobu, N.; Yoshida, H.; Hamada, J.; Shindo, M.; Higashino, F.; Tanaka, J.; Asaka, M.; et al. Selective secretion of chemoattractants for haemopoietic progenitor cells by bone marrow endothelial cells: A possible role in homing of haemopoietic progenitor cells to bone marrow. Br. J. Haematol. 1999, 106, 905–911. [Google Scholar] [CrossRef]
  68. Sipkins, D.A.; Wei, X.; Wu, J.W.; Runnels, J.M.; Cote, D.; Means, T.K.; Luster, A.D.; Scadden, D.T.; Lin, C.P. In vivo imaging of specialized bone marrow endothelial microdomains for tumour engraftment. Nature 2005, 435, 969–973. [Google Scholar] [CrossRef]
  69. Vanderkerken, K.; Greef, C.D.; Asosingh, K.; Arteta, B.; Veerman, M.D.; Broek, I.V.; Riet, I.V.; Kobayashi, M.; Smedsrod, B.; Camp, B.V. Selective initial in vivo homing pattern of 5T2 multiple myeloma cells in the C57BL/KalwRij mouse. Br. J. Cancer 2000, 82, 953–959. [Google Scholar] [CrossRef] [Green Version]
  70. Parmo-Cabañas, M.; Bartolomé, R.A.; Wright, N.; Hidalgo, A.; Drager, A.M.; Teixidó, J. Integrin α4β1 involvement in stromal cell-derived factor-1α-promoted myeloma cell transendothelial migration and adhesion: Role of cAMP and the actin cytoskeleton in adhesion. Exp. Cell Res. 2004, 294, 571–580. [Google Scholar] [CrossRef] [Green Version]
  71. Martínez-Moreno, M.; Leiva, M.; Aguilera-Montilla, N.; Sevilla-Movilla, S.; Isern de Val, S.; Arellano-Sánchez, N.; Gutiérrez, N.C.; Maldonado, R.; Martínez-López, J.; Buño, I.; et al. In vivo adhesion of malignant B cells to bone marrow microvasculature is regulated by α4β1 cytoplasmic-binding proteins. Leukemia 2016, 30, 861–872. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Runnels, J.M.; Carlson, A.L.; Pitsillides, C.; Thompson, B.; Wu, J.; Spencer, J.A.; Kohler, J.M.; Azab, A.; Moreau, A.S.; Rodig, S.J.; et al. Optical techniques for tracking multiple myeloma engraftment, growth, and response to therapy. J. Biomed. Opt. 2011, 16, 011006. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Vande Broek, I.; Asosingh, K.; Allegaert, V.; Leleu, X.; Facon, T.; Vanderkerken, K.; Van Camp, B.; Van Riet, I. Bone marrow endothelial cells increase the invasiveness of human multiple myeloma cells through upregulation of MMP-9: Evidence for a role of hepatocyte growth factor. Leukemia 2004, 18, 976–982. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Menu, E.; Asosingh, K.; Indraccolo, S.; De Raeve, H.; Van Riet, I.; Van Valckenborgh, E.; Van de Broek, I.; Fujii, N.; Tamamura, H.; Van Camp, B.; et al. The involvement of stromal derived factor 1alpha in homing and progression of multiple myeloma in the 5TMM model. Haematologica 2006, 91, 605–612. [Google Scholar] [PubMed]
  75. Vande Broek, I.; Vanderkerken, K.; Van Camp, B.; Van Riet, I. Extravasation and homing mechanisms in multiple myeloma. Clin. Exp. Metastasis 2008, 25, 325–334. [Google Scholar] [CrossRef]
  76. Moller, C.; Stromberg, T.; Juremalm, M.; Nilsson, K.; Nilsson, G. Expression and function of chemokine receptors in human multiple myeloma. Leukemia 2003, 17, 203–210. [Google Scholar] [CrossRef] [Green Version]
  77. Roccaro, A.M.; Mishima, Y.; Sacco, A.; Moschetta, M.; Tai, Y.T.; Shi, J.; Zhang, Y.; Reagan, M.R.; Huynh, D.; Kawano, Y.; et al. CXCR4 Regulates Extra-Medullary Myeloma through Epithelial-Mesenchymal-Transition-like Transcriptional Activation. Cell Rep. 2015, 12, 622–635. [Google Scholar] [CrossRef] [Green Version]
  78. Roccaro, A.M.; Sacco, A.; Purschke, W.G.; Moschetta, M.; Buchner, K.; Maasch, C.; Zboralski, D.; Zollner, S.; Vonhoff, S.; Mishima, Y.; et al. SDF-1 inhibition targets the bone marrow niche for cancer therapy. Cell Rep. 2014, 9, 118–128. [Google Scholar] [CrossRef] [Green Version]
  79. Takai, K.; Hara, J.; Matsumoto, K.; Hosoi, G.; Osugi, Y.; Tawa, A.; Okada, S.; Nakamura, T. Hepatocyte growth factor is constitutively produced by human bone marrow stromal cells and indirectly promotes hematopoiesis. Blood 1997, 89, 1560–1565. [Google Scholar] [CrossRef] [Green Version]
  80. Zdzisinska, B.; Bojarska-Junak, A.; Dmoszynska, A.; Kandefer-Szerszen, M. Abnormal cytokine production by bone marrow stromal cells of multiple myeloma patients in response to RPMI8226 myeloma cells. Arch. Immunol. Ther. Exp. 2008, 56, 207–221. [Google Scholar] [CrossRef] [Green Version]
  81. Chen, L.; Tredget, E.E.; Wu, P.Y.; Wu, Y. Paracrine factors of mesenchymal stem cells recruit macrophages and endothelial lineage cells and enhance wound healing. PLoS ONE 2008, 3, e1886. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Ro, T.B.; Holien, T.; Fagerli, U.M.; Hov, H.; Misund, K.; Waage, A.; Sundan, A.; Holt, R.U.; Borset, M. HGF and IGF-1 synergize with SDF-1alpha in promoting migration of myeloma cells by cooperative activation of p21-activated kinase. Exp. Hematol. 2013, 41, 646–655. [Google Scholar] [CrossRef] [PubMed]
  83. Wang, J.; Hendrix, A.; Hernot, S.; Lemaire, M.; De Bruyne, E.; Van Valckenborgh, E.; Lahoutte, T.; De Wever, O.; Vanderkerken, K.; Menu, E. Bone marrow stromal cell-derived exosomes as communicators in drug resistance in multiple myeloma cells. Blood 2014, 124, 555–566. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Asosingh, K.; Gunthert, U.; Bakkus, M.H.; De Raeve, H.; Goes, E.; Van Riet, I.; Van Camp, B.; Vanderkerken, K. In vivo induction of insulin-like growth factor-I receptor and CD44v6 confers homing and adhesion to murine multiple myeloma cells. Cancer Res. 2000, 60, 3096–3104. [Google Scholar]
  85. Opperman, K.S.; Vandyke, K.; Clark, K.C.; Coulter, E.A.; Hewett, D.R.; Mrozik, K.M.; Schwarz, N.; Evdokiou, A.; Croucher, P.I.; Psaltis, P.J.; et al. Clodronate-liposome mediated macrophage depletion abrogates multiple myeloma tumor establishment in vivo. Neoplasia 2019, 21, 777–787. [Google Scholar] [CrossRef]
  86. Davatelis, G.; Tekamp-Olson, P.; Wolpe, S.D.; Hermsen, K.; Luedke, C.; Gallegos, C.; Coit, D.; Merryweather, J.; Cerami, A. Cloning and characterization of a cDNA for murine macrophage inflammatory protein (MIP), a novel monokine with inflammatory and chemokinetic properties. J. Exp. Med. 1988, 167, 1939–1944. [Google Scholar] [CrossRef] [Green Version]
  87. Moreaux, J.; Hose, D.; Kassambara, A.; Reme, T.; Moine, P.; Requirand, G.; Goldschmidt, H.; Klein, B. Osteoclast-gene expression profiling reveals osteoclast-derived CCR2 chemokines promoting myeloma cell migration. Blood 2011, 117, 1280–1290. [Google Scholar] [CrossRef] [Green Version]
  88. Lentzsch, S.; Gries, M.; Janz, M.; Bargou, R.; Dorken, B.; Mapara, M.Y. Macrophage inflammatory protein 1-alpha (MIP-1 alpha ) triggers migration and signaling cascades mediating survival and proliferation in multiple myeloma (MM) cells. Blood 2003, 101, 3568–3573. [Google Scholar] [CrossRef] [Green Version]
  89. Vallet, S.; Raje, N.; Ishitsuka, K.; Hideshima, T.; Podar, K.; Chhetri, S.; Pozzi, S.; Breitkreutz, I.; Kiziltepe, T.; Yasui, H.; et al. MLN3897, a novel CCR1 inhibitor, impairs osteoclastogenesis and inhibits the interaction of multiple myeloma cells and osteoclasts. Blood 2007, 110, 3744–3752. [Google Scholar] [CrossRef] [Green Version]
  90. Vande Broek, I.; Asosingh, K.; Vanderkerken, K.; Straetmans, N.; Van Camp, B.; Van Riet, I. Chemokine receptor CCR2 is expressed by human multiple myeloma cells and mediates migration to bone marrow stromal cell-produced monocyte chemotactic proteins MCP-1, -2 and -3. Br. J. Cancer 2003, 88, 855–862. [Google Scholar] [CrossRef] [Green Version]
  91. Pellegrino, A.; Antonaci, F.; Russo, F.; Merchionne, F.; Ribatti, D.; Vacca, A.; Dammacco, F. CXCR3-binding chemokines in multiple myeloma. Cancer Lett. 2004, 207, 221–227. [Google Scholar] [CrossRef] [PubMed]
  92. Lee, J.H.; Kim, H.N.; Kim, K.O.; Jin, W.J.; Lee, S.; Kim, H.H.; Ha, H.; Lee, Z.H. CXCL10 promotes osteolytic bone metastasis by enhancing cancer outgrowth and osteoclastogenesis. Cancer Res. 2012, 72, 3175–3186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Caers, J.; Deleu, S.; Belaid, Z.; De Raeve, H.; Van Valckenborgh, E.; De Bruyne, E.; Defresne, M.P.; Van Riet, I.; Van Camp, B.; Vanderkerken, K. Neighboring adipocytes participate in the bone marrow microenvironment of multiple myeloma cells. Leukemia 2007, 21, 1580–1584. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Trotter, T.N.; Gibson, J.T.; Sherpa, T.L.; Gowda, P.S.; Peker, D.; Yang, Y. Adipocyte-Lineage Cells Support Growth and Dissemination of Multiple Myeloma in Bone. Am. J. Pathol. 2016, 186, 3054–3063. [Google Scholar] [CrossRef] [Green Version]
  95. Morris, E.V.; Suchacki, K.J.; Hocking, J.; Cartwright, R.; Sowman, A.; Gamez, B.; Lea, R.; Drake, M.T.; Cawthorn, W.P.; Edwards, C.M. Myeloma cells down-regulate adiponectin in bone marrow adipocytes via TNF-Alpha. J. Bone Miner. Res. 2020, 35, 942–955. [Google Scholar] [CrossRef] [Green Version]
  96. Geng, S.; Wang, J.; Zhang, X.; Zhang, J.J.; Wu, F.; Pang, Y.; Zhong, Y.; Wang, J.; Wang, W.; Lyu, X.; et al. Single-cell RNA sequencing reveals chemokine self-feeding of myeloma cells promotes extramedullary metastasis. FEBS Lett. 2020, 594, 452–465. [Google Scholar] [CrossRef] [Green Version]
  97. Uneda, S.; Hata, H.; Matsuno, F.; Harada, N.; Mitsuya, Y.; Kawano, F.; Mitsuya, H. Macrophage inflammatory protein-1 alpha is produced by human multiple myeloma (MM) cells and its expression correlates with bone lesions in patients with MM. Br. J. Haematol. 2003, 120, 53–55. [Google Scholar] [CrossRef]
  98. Roussou, M.; Tasidou, A.; Dimopoulos, M.A.; Kastritis, E.; Migkou, M.; Christoulas, D.; Gavriatopoulou, M.; Zagouri, F.; Matsouka, C.; Anagnostou, D.; et al. Increased expression of macrophage inflammatory protein-1alpha on trephine biopsies correlates with extensive bone disease, increased angiogenesis and advanced stage in newly diagnosed patients with multiple myeloma. Leukemia 2009, 23, 2177–2181. [Google Scholar] [CrossRef] [Green Version]
  99. Bataille, R.; Robillard, N.; Avet-Loiseau, H.; Harousseau, J.L.; Moreau, P. CD221 (IGF-1R) is aberrantly expressed in multiple myeloma, in relation to disease severity. Haematologica 2005, 90, 706–707. [Google Scholar]
  100. Sprynski, A.C.; Hose, D.; Caillot, L.; Reme, T.; Shaughnessy, J.D., Jr.; Barlogie, B.; Seckinger, A.; Moreaux, J.; Hundemer, M.; Jourdan, M.; et al. The role of IGF-1 as a major growth factor for myeloma cell lines and the prognostic relevance of the expression of its receptor. Blood 2009, 113, 4614–4626. [Google Scholar] [CrossRef]
  101. Wynes, M.W.; Riches, D.W. Induction of macrophage insulin-like growth factor-I expression by the Th2 cytokines IL-4 and IL-13. J. Immunol. 2003, 171, 3550–3559. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Borset, M.; Hjorth-Hansen, H.; Seidel, C.; Sundan, A.; Waage, A. Hepatocyte growth factor and its receptor c-met in multiple myeloma. Blood 1996, 88, 3998–4004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Hewett, D.R.; Vandyke, K.; Lawrence, D.M.; Friend, N.; Noll, J.E.; Geoghegan, J.M.; Croucher, P.I.; Zannettino, A.C.W. DNA barcoding reveals habitual clonal dominance of myeloma plasma cells in the bone marrow microenvironment. Neoplasia 2017, 19, 972–981. [Google Scholar] [CrossRef] [PubMed]
  104. Lawson, M.A.; McDonald, M.M.; Kovacic, N.; Hua Khoo, W.; Terry, R.L.; Down, J.; Kaplan, W.; Paton-Hough, J.; Fellows, C.; Pettitt, J.A.; et al. Osteoclasts control reactivation of dormant myeloma cells by remodelling the endosteal niche. Nat. Commun. 2015, 6, 8983. [Google Scholar] [CrossRef] [Green Version]
  105. Shen, Y.J.; Mishima, Y.; Shi, J.; Sklavenitis-Pistofidis, R.; Redd, R.A.; Moschetta, M.; Manier, S.; Roccaro, A.; Sacco, A.; Tai, Y.T.; et al. Progression signature underlies clonal evolution and dissemination of multiple myeloma. Blood 2020. [Google Scholar] [CrossRef] [PubMed]
  106. Khoo, W.H.; Ledergor, G.; Weiner, A.; Roden, D.L.; Terry, R.L.; McDonald, M.M.; Chai, R.C.; De Veirman, K.; Owen, K.L.; Opperman, K.S.; et al. A niche-dependent myeloid transcriptome signature defines dormant myeloma cells. Blood 2019, 134, 30–43. [Google Scholar] [CrossRef]
  107. Avet-Loiseau, H.; Li, J.Y.; Facon, T.; Brigaudeau, C.; Morineau, N.; Maloisel, F.; Rapp, M.J.; Talmant, P.; Trimoreau, F.; Jaccard, A.; et al. High incidence of translocations t(11;14)(q13;q32) and t(4;14)(p16;q32) in patients with plasma cell malignancies. Cancer Res. 1998, 58, 5640–5645. [Google Scholar]
  108. Avet-Loiseau, H.; Daviet, A.; Brigaudeau, C.; Callet-Bauchu, E.; Terre, C.; Lafage-Pochitaloff, M.; Desangles, F.; Ramond, S.; Talmant, P.; Bataille, R. Cytogenetic, interphase, and multicolor fluorescence in situ hybridization analyses in primary plasma cell leukemia: A study of 40 patients at diagnosis, on behalf of the Intergroupe Francophone du Myelome and the Groupe Francais de Cytogenetique Hematologique. Blood 2001, 97, 822–825. [Google Scholar] [CrossRef] [Green Version]
  109. Chang, H.; Qi, X.; Yeung, J.; Reece, D.; Xu, W.; Patterson, B. Genetic aberrations including chromosome 1 abnormalities and clinical features of plasma cell leukemia. Leukemia Res. 2009, 33, 259–262. [Google Scholar] [CrossRef]
  110. Ross, F.M.; Chiecchio, L.; Dagrada, G.; Protheroe, R.K.; Stockley, D.M.; Harrison, C.J.; Cross, N.C.; Szubert, A.J.; Drayson, M.T.; Morgan, G.J.; et al. The t(14;20) is a poor prognostic factor in myeloma but is associated with long-term stable disease in monoclonal gammopathies of undetermined significance. Haematologica 2010, 95, 1221–1225. [Google Scholar] [CrossRef] [Green Version]
  111. Avet-Loiseau, H.; Malard, F.; Campion, L.; Magrangeas, F.; Sebban, C.; Lioure, B.; Decaux, O.; Lamy, T.; Legros, L.; Fuzibet, J.G.; et al. Translocation t(14;16) and multiple myeloma: Is it really an independent prognostic factor? Blood 2011, 117, 2009–2011. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. An, G.; Xu, Y.; Shi, L.; Zou, D.; Deng, S.; Sui, W.; Xie, Z.; Hao, M.; Chang, H.; Qiu, L. t(11;14) multiple myeloma: A subtype associated with distinct immunological features, immunophenotypic characteristics but divergent outcome. Leukemia Res. 2013, 37, 1251–1257. [Google Scholar] [CrossRef] [PubMed]
  113. An, G.; Qin, X.; Acharya, C.; Xu, Y.; Deng, S.; Shi, L.; Zang, M.; Sui, W.; Yi, S.; Li, Z.; et al. Multiple myeloma patients with low proportion of circulating plasma cells had similar survival with primary plasma cell leukemia patients. Ann. Hematol. 2015, 94, 257–264. [Google Scholar] [CrossRef] [PubMed]
  114. Mattioli, M.; Agnelli, L.; Fabris, S.; Baldini, L.; Morabito, F.; Bicciato, S.; Verdelli, D.; Intini, D.; Nobili, L.; Cro, L.; et al. Gene expression profiling of plasma cell dyscrasias reveals molecular patterns associated with distinct IGH translocations in multiple myeloma. Oncogene 2005, 24, 2461–2473. [Google Scholar] [CrossRef] [Green Version]
  115. Zhan, F.; Huang, Y.; Colla, S.; Stewart, J.P.; Hanamura, I.; Gupta, S.; Epstein, J.; Yaccoby, S.; Sawyer, J.; Burington, B.; et al. The molecular classification of multiple myeloma. Blood 2006, 108, 2020–2028. [Google Scholar] [CrossRef] [Green Version]
  116. Hurt, E.M.; Wiestner, A.; Rosenwald, A.; Shaffer, A.L.; Campo, E.; Grogan, T.; Bergsagel, P.L.; Kuehl, W.M.; Staudt, L.M. Overexpression of c-maf is a frequent oncogenic event in multiple myeloma that promotes proliferation and pathological interactions with bone marrow stroma. Cancer Cell 2004, 5, 191–199. [Google Scholar] [CrossRef] [Green Version]
  117. Dring, A.M.; Davies, F.E.; Fenton, J.A.; Roddam, P.L.; Scott, K.; Gonzalez, D.; Rollinson, S.; Rawstron, A.C.; Rees-Unwin, K.S.; Li, C.; et al. A global expression-based analysis of the consequences of the t(4;14) translocation in myeloma. Clin. Cancer Res. 2004, 10, 5692–5701. [Google Scholar] [CrossRef] [Green Version]
  118. Lauring, J.; Abukhdeir, A.M.; Konishi, H.; Garay, J.P.; Gustin, J.P.; Wang, Q.; Arceci, R.J.; Matsui, W.; Park, B.H. The multiple myeloma associated MMSET gene contributes to cellular adhesion, clonogenic growth, and tumorigenicity. Blood 2008, 111, 856–864. [Google Scholar] [CrossRef]
  119. Lombardi, L.; Poretti, G.; Mattioli, M.; Fabris, S.; Agnelli, L.; Bicciato, S.; Kwee, I.; Rinaldi, A.; Ronchetti, D.; Verdelli, D.; et al. Molecular characterization of human multiple myeloma cell lines by integrative genomics: Insights into the biology of the disease. Genes Chromosomes Cancer 2007, 46, 226–238. [Google Scholar] [CrossRef]
  120. Neri, P.; Ren, L.; Azab, A.K.; Brentnall, M.; Gratton, K.; Klimowicz, A.C.; Lin, C.; Duggan, P.; Tassone, P.; Mansoor, A.; et al. Integrin β7-mediated regulation of multiple myeloma cell adhesion, migration, and invasion. Blood 2011, 117, 6202. [Google Scholar] [CrossRef] [Green Version]
  121. Postigo, A.A.; Sanchez-Mateos, P.; Lazarovits, A.I.; Sanchez-Madrid, F.; de Landazuri, M.O. Alpha 4 beta 7 integrin mediates B cell binding to fibronectin and vascular cell adhesion molecule-1. Expression and function of alpha 4 integrins on human B lymphocytes. J. Immunol. 1993, 151, 2471–2483. [Google Scholar] [PubMed]
  122. Bazan, J.F.; Bacon, K.B.; Hardiman, G.; Wang, W.; Soo, K.; Rossi, D.; Greaves, D.R.; Zlotnik, A.; Schall, T.J. A new class of membrane-bound chemokine with a CX3C motif. Nature 1997, 385, 640–644. [Google Scholar] [CrossRef] [PubMed]
  123. Nishimura, M.; Umehara, H.; Nakayama, T.; Yoneda, O.; Hieshima, K.; Kakizaki, M.; Dohmae, N.; Yoshie, O.; Imai, T. Dual functions of fractalkine/CX3C ligand 1 in trafficking of perforin+/granzyme B+ cytotoxic effector lymphocytes that are defined by CX3CR1 expression. J. Immunol. 2002, 168, 6173–6180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Cederblad, L.; Rosengren, B.; Ryberg, E.; Hermansson, N.-O. AZD8797 is an allosteric non-competitive modulator of the human CX3CR1 receptor. Biochem. J. 2016, 473, 641–649. [Google Scholar] [CrossRef] [Green Version]
  125. Chakraborty, R.; Muchtar, E.; Kumar, S.K.; Jevremovic, D.; Buadi, F.K.; Dingli, D.; Dispenzieri, A.; Hayman, S.R.; Hogan, W.J.; Kapoor, P.; et al. Risk stratification in myeloma by detection of circulating plasma cells prior to autologous stem cell transplantation in the novel agent era. Blood Cancer J. 2016, 6, e512. [Google Scholar] [CrossRef] [Green Version]
  126. Katodritou, E.; Gastari, V.; Verrou, E.; Hadjiaggelidou, C.; Varthaliti, M.; Georgiadou, S.; Laschos, K.; Xirou, P.; Yiannaki, E.; Constantinou, N.; et al. Extramedullary (EMP) relapse in unusual locations in multiple myeloma: Is there an association with precedent thalidomide administration and a correlation of special biological features with treatment and outcome? Leukemia Res. 2009, 33, 1137–1140. [Google Scholar] [CrossRef]
  127. Lohr, J.G.; Kim, S.; Gould, J.; Knoechel, B.; Drier, Y.; Cotton, M.J.; Gray, D.; Birrer, N.; Wong, B.; Ha, G.; et al. Genetic interrogation of circulating multiple myeloma cells at single-cell resolution. Sci. Transl. Med. 2016, 8, 363ra147. [Google Scholar] [CrossRef] [Green Version]
  128. Mishima, Y.; Paiva, B.; Shi, J.; Park, J.; Manier, S.; Takagi, S.; Massoud, M.; Perilla-Glen, A.; Aljawai, Y.; Huynh, D.; et al. The mutational landscape of circulating tumor cells in multiple myeloma. Cell Rep. 2017, 19, 218–224. [Google Scholar] [CrossRef] [Green Version]
  129. Manier, S.; Park, J.; Capelletti, M.; Bustoros, M.; Freeman, S.S.; Ha, G.; Rhoades, J.; Liu, C.J.; Huynh, D.; Reed, S.C.; et al. Whole-exome sequencing of cell-free DNA and circulating tumor cells in multiple myeloma. Nat. Commun. 2018, 9, 1691. [Google Scholar] [CrossRef]
  130. Rasche, L.; Chavan, S.S.; Stephens, O.W.; Patel, P.H.; Tytarenko, R.; Ashby, C.; Bauer, M.; Stein, C.; Deshpande, S.; Wardell, C.; et al. Spatial genomic heterogeneity in multiple myeloma revealed by multi-region sequencing. Nat. Commun. 2017, 8, 268. [Google Scholar] [CrossRef]
  131. Avet-Loiseau, H.; Attal, M.; Moreau, P.; Charbonnel, C.; Garban, F.; Hulin, C.; Leyvraz, S.; Michallet, M.; Yakoub-Agha, I.; Garderet, L.; et al. Genetic abnormalities and survival in multiple myeloma: The experience of the Intergroupe Francophone du Myelome. Blood 2007, 109, 3489–3495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Lode, L.; Eveillard, M.; Trichet, V.; Soussi, T.; Wuilleme, S.; Richebourg, S.; Magrangeas, F.; Ifrah, N.; Campion, L.; Traulle, C.; et al. Mutations in TP53 are exclusively associated with del(17p) in multiple myeloma. Haematologica 2010, 95, 1973–1976. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Yue, Z.; Zhou, Y.; Zhao, P.; Chen, Y.; Yuan, Y.; Jing, Y.; Wang, X. p53 Deletion promotes myeloma cells invasion by upregulating miR19a/CXCR5. Leukemia Res. 2017, 60, 115–122. [Google Scholar] [CrossRef] [PubMed]
  134. Palumbo, A.; Avet-Loiseau, H.; Oliva, S.; Lokhorst, H.M.; Goldschmidt, H.; Rosinol, L.; Richardson, P.; Caltagirone, S.; Lahuerta, J.J.; Facon, T.; et al. Revised International Staging System for Multiple Myeloma: A Report From International Myeloma Working Group. J. Clin. Oncol. 2015, 33, 2863–2869. [Google Scholar] [CrossRef] [PubMed]
  135. Sher, T.; Miller, K.C.; Deeb, G.; Lee, K.; Chanan-Khan, A. Plasma cell leukaemia and other aggressive plasma cell malignancies. Br. J. Haematol. 2010, 150, 418–427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Peceliunas, V.; Janiulioniene, A.; Matuzeviciene, R.; Zvirblis, T.; Griskevicius, L. Circulating plasma cells predict the outcome of relapsed or refractory multiple myeloma. Leuk Lymphoma 2012, 53, 641–647. [Google Scholar] [CrossRef]
  137. Keats, J.J.; Chesi, M.; Egan, J.B.; Garbitt, V.M.; Palmer, S.E.; Braggio, E.; Van Wier, S.; Blackburn, P.R.; Baker, A.S.; Dispenzieri, A.; et al. Clonal competition with alternating dominance in multiple myeloma. Blood 2012, 120, 1067–1076. [Google Scholar] [CrossRef]
  138. Ludwig, H.; Weisel, K.; Petrucci, M.T.; Leleu, X.; Cafro, A.M.; Garderet, L.; Leitgeb, C.; Foa, R.; Greil, R.; Yakoub-Agha, I.; et al. Olaptesed pegol, an anti-CXCL12/SDF-1 Spiegelmer, alone and with bortezomib-dexamethasone in relapsed/refractory multiple myeloma: A Phase IIa Study. Leukemia 2017, 31, 997–1000. [Google Scholar] [CrossRef]
  139. Hu, J.; Van Valckenborgh, E.; Xu, D.; Menu, E.; De Raeve, H.; De Bruyne, E.; Xu, S.; Van Camp, B.; Handisides, D.; Hart, C.P.; et al. Synergistic induction of apoptosis in multiple myeloma cells by bortezomib and hypoxia-activated prodrug TH-302, in vivo and in vitro. Mol. Cancer Ther. 2013, 12, 1763–1773. [Google Scholar] [CrossRef] [Green Version]
  140. Laubach, J.P.; Liu, C.J.; Raje, N.S.; Yee, A.J.; Armand, P.; Schlossman, R.L.; Rosenblatt, J.; Hedlund, J.; Martin, M.; Reynolds, C.; et al. A Phase I/II study of evofosfamide, a hypoxia-activated prodrug with or without bortezomib in subjects with relapsed/refractory multiple myeloma. Clin. Cancer Res. 2019, 25, 478–486. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Multiple myeloma (MM) plasma cell (PC) intravasation is dependent on overcoming adhesive interactions within the bone marrow (BM) and invading the vasculature basement membrane. Release from the stromal niche is likely mediated by both external drivers such as hypoxia, and internal drivers in subclones with increased propensity to disseminate. This leads to a decrease in MM PC expression of key adhesion molecules such as α4β1, allowing the MM PCs to overcome the retention signal mediated by CXCL12 produced by bone marrow mesenchymal stromal cells (BMSCs). Hypoxic MM PCs upregulate epithelial to mesenchymal transition (EMT)-like genes, decrease their cell surface expression of CXCR4 and increase their expression of CCR1 to mediate mobilisation from the BM niche. Upregulation of matrix metalloproteins-2 and -9 then allows invasion into the vasculature basement membranes and intravasation into the peripheral circulation during dissemination to a new site.
Figure 1. Multiple myeloma (MM) plasma cell (PC) intravasation is dependent on overcoming adhesive interactions within the bone marrow (BM) and invading the vasculature basement membrane. Release from the stromal niche is likely mediated by both external drivers such as hypoxia, and internal drivers in subclones with increased propensity to disseminate. This leads to a decrease in MM PC expression of key adhesion molecules such as α4β1, allowing the MM PCs to overcome the retention signal mediated by CXCL12 produced by bone marrow mesenchymal stromal cells (BMSCs). Hypoxic MM PCs upregulate epithelial to mesenchymal transition (EMT)-like genes, decrease their cell surface expression of CXCR4 and increase their expression of CCR1 to mediate mobilisation from the BM niche. Upregulation of matrix metalloproteins-2 and -9 then allows invasion into the vasculature basement membranes and intravasation into the peripheral circulation during dissemination to a new site.
Cancers 12 03643 g001
Figure 2. Multiple myeloma (MM) plasma cell (PC) extravasation is dependent on expression of key adhesion and chemoattractant molecules. Tethering and rolling are mediated by MM PC CD44 and P-selectin glycoprotein ligand-1 (PSGL-1) binding to E- and P-selectin, respectively, on endothelial cells), and homophilic adhesion of N-cadherin between MM PCs and endothelial cells. CXCL12, produced by the endothelium, activates MM PC-expressed integrin complex α4β1, allowing arrest through firm adhesive interactions with VCAM-1 ligand expressed on endothelial cells. Following arrest, MM PCs undergo trans-endothelial migration following chemoattractant factors expressed within the bone marrow (BM) by BM mesenchymal stromal cells (BMSCs), macrophages and osteoclasts.
Figure 2. Multiple myeloma (MM) plasma cell (PC) extravasation is dependent on expression of key adhesion and chemoattractant molecules. Tethering and rolling are mediated by MM PC CD44 and P-selectin glycoprotein ligand-1 (PSGL-1) binding to E- and P-selectin, respectively, on endothelial cells), and homophilic adhesion of N-cadherin between MM PCs and endothelial cells. CXCL12, produced by the endothelium, activates MM PC-expressed integrin complex α4β1, allowing arrest through firm adhesive interactions with VCAM-1 ligand expressed on endothelial cells. Following arrest, MM PCs undergo trans-endothelial migration following chemoattractant factors expressed within the bone marrow (BM) by BM mesenchymal stromal cells (BMSCs), macrophages and osteoclasts.
Cancers 12 03643 g002
Figure 3. High-risk cytogenetics are associated with higher numbers of circulating multiple myeloma (MM) plasma cells (PCs) in patients. Average proportion of MM and plasma cell leukaemia (PCL) patients with common IgH translocation t(4;14), t(14;16), t(14;20) or t(11;14), or those without these translocations (other), are shown. Data is shown for patients with newly diagnosed MM or PCL (A) or for newly diagnosed MM patients with low or high circulating MM PC numbers (B). Higher incidences of high-risk cytogenetics t(4;14), t(14;16) and t(14;20) is observed in patients with PCL and in patients with high numbers of MM PCs in the peripheral blood (PB). Data is taken from [107,108,109,110,111] (A) and [11,14,113] (B).
Figure 3. High-risk cytogenetics are associated with higher numbers of circulating multiple myeloma (MM) plasma cells (PCs) in patients. Average proportion of MM and plasma cell leukaemia (PCL) patients with common IgH translocation t(4;14), t(14;16), t(14;20) or t(11;14), or those without these translocations (other), are shown. Data is shown for patients with newly diagnosed MM or PCL (A) or for newly diagnosed MM patients with low or high circulating MM PC numbers (B). Higher incidences of high-risk cytogenetics t(4;14), t(14;16) and t(14;20) is observed in patients with PCL and in patients with high numbers of MM PCs in the peripheral blood (PB). Data is taken from [107,108,109,110,111] (A) and [11,14,113] (B).
Cancers 12 03643 g003
Table 1. Receptors and secreted factors involved in the dissemination process in multiple myeloma.
Table 1. Receptors and secreted factors involved in the dissemination process in multiple myeloma.
Receptor on MM PCsPro-Disseminatory FactorPredominant Source in The
Myeloma BM
Suggested Role in Dissemination
CXCR4[33,34,35]CXCL12Hypoxic MM PCs[35,54]Mobilisation of MM PCs from the BM[35,96]
Endothelial cells[29,33,67,68]Arrest of circulating MM PCs in the BM vasculature[33,67,68,69]
BMSCs[29,30,31,32]Migration and homing to BM niches[27,28,76,77]
CCR1[34,35,55]CCL3MM PCs[35,89,97,98]Mobilisation of MM PCs from the BM[35,55]
Osteoclasts, macrophages[86,87,89]Migration and homing to BM niches[87,88,89]
CCR2[33,34,87]CCL2Endothelial cells[33,58]Migration towards BMECs[33,58,59]
BMSCs, osteoclasts, adipocytes[32,81,87,90,94]Migration and homing to BM niches[33,58]
CCL7, CCL8Osteoclasts[87]Migration and homing to BM niches[90]
IGF-1R[99,100]IGF-1Macrophages[85,87,101]Migration and homing to BM niches[84,85]
Met[100,102]HGFBMSCs[79,80,81]Synergises with CXCL12 to increase MM PC migration [82]
Table 2. Chromosomal translocations associated with increased dissemination in multiple myeloma (MM) and plasma cell leukaemia (PCL).
Table 2. Chromosomal translocations associated with increased dissemination in multiple myeloma (MM) and plasma cell leukaemia (PCL).
Chromosomal TranslocationDriver GeneAssociation with Increased Dissemination 1Putative Pro-Disseminatory Factors
t(14;16) and t(14;20)MAF/MAFBIncreased incidence in PCL and high-PB PC MMIntegrin β7[102,103,104,105]
CX3CR1[114,115]
IGF-1R[99,116]
CCR1[99,116]
t(4;14)NSD2Increased incidence in PCL and high-PB PC MMEMT-like gene signature
(including CDH2, VIM and TWIST1)
[53,66,117,118]
IGF-1R[99,100,116]
CCR1[99,100,116]
t(11;14)CCND1Increased incidence in PCLUnclear-
1 See Figure 3.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Zeissig, M.N.; Zannettino, A.C.W.; Vandyke, K. Tumour Dissemination in Multiple Myeloma Disease Progression and Relapse: A Potential Therapeutic Target in High-Risk Myeloma. Cancers 2020, 12, 3643. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers12123643

AMA Style

Zeissig MN, Zannettino ACW, Vandyke K. Tumour Dissemination in Multiple Myeloma Disease Progression and Relapse: A Potential Therapeutic Target in High-Risk Myeloma. Cancers. 2020; 12(12):3643. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers12123643

Chicago/Turabian Style

Zeissig, Mara N., Andrew C. W. Zannettino, and Kate Vandyke. 2020. "Tumour Dissemination in Multiple Myeloma Disease Progression and Relapse: A Potential Therapeutic Target in High-Risk Myeloma" Cancers 12, no. 12: 3643. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers12123643

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop