Next Article in Journal
Inflammatory Proteins HMGA2 and PRTN3 as Drivers of Vulvar Squamous Cell Carcinoma Progression
Next Article in Special Issue
Daratumumab and Nanobody-Based Heavy Chain Antibodies Inhibit the ADPR Cyclase but not the NAD+ Hydrolase Activity of CD38-Expressing Multiple Myeloma Cells
Previous Article in Journal
The Association between Ranitidine Use and Gastrointestinal Cancers
Previous Article in Special Issue
Preclinical Rationale for Targeting the PD-1/PD-L1 Axis in Combination with a CD38 Antibody in Multiple Myeloma and Other CD38-Positive Malignancies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Current Understanding of Myelomatous Mesenchymal Stromal Cells Extended through Advances in Experimental Methods

Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Osaka 5650871, Japan
*
Author to whom correspondence should be addressed.
Cancers 2021, 13(1), 25; https://doi.org/10.3390/cancers13010025
Submission received: 29 November 2020 / Revised: 20 December 2020 / Accepted: 21 December 2020 / Published: 23 December 2020
(This article belongs to the Special Issue Tumor Microenvironment and Exacerbation Mechanism in Multiple Myeloma)

Abstract

:

Simple Summary

As the amount of information available has grown, now it is known that many types of non-hematopoietic cells, including mesenchymal stem/progenitor cells, mature mesenchymal cells, and endothelial cells, as well as mature hematopoietic cells such as monocytes, macrophages, T-cells, and B-cells, have roles in the pathogenesis of multiple myeloma. This review focuses on the role of mesenchymal cells in the microenvironment of multiple myeloma. We summarize the experimental strategies and current understanding of the biological roles in the pathogenesis of myeloma. Furthermore, we discuss the possible clinical applications targeting mesenchymal cells.

Abstract

Multiple myeloma is an incurable cancer formed by malignant plasma cells. For the proliferation and survival of myeloma cells, as well as the occurrence of the complications, numerous intra- and extra-cellular mechanisms are involved. The interaction of myeloma cells with the microenvironment is known to be one of the most critical mechanisms. A specific microenvironment could affect the progression and growth of tumor cells, as well as drug resistance. Among various microenvironment components, such as hematological and non-hematological cells, and soluble factors (cytokines, chemokines, and extracellular matrix (ECM) proteins), in this review, we focus on the role of mesenchymal cells. We aimed to summarize the experimental strategies used for conducting studies and current understanding of the biological roles in the pathogenesis of myeloma. Furthermore, we discuss the possible clinical applications targeting mesenchymal cells.

1. Introduction

Multiple myeloma (MM) is a malignant plasma cell (PC) disease that infiltrates the bone marrow (BM). One of the characteristics of MM is the occurrence of various symptoms and signs, such as hypercalcemia, renal dysfunction, anemia, and bone lesions, with the progression of disease [1,2]. Some of these symptoms are induced upon accumulation or hyperviscosity of monoclonal proteins produced by MM cells. Many types of soluble factors secreted by MM cells and direct and indirect crosstalk between myeloma cells and other cells affect hematopoiesis, osteoclastogenesis, angiogenesis, and osteogenesis in BM. MM is also characterized by the premalignant condition, monoclonal gammopathy of undetermined significance (MGUS), which occurs before the onset of MM. Chromosomal translocations involving 14q and hyperdiploidity are generated as an initial step of transformation to MGUS in post-germinal center B-lymphocytes or immunoglobulin-producing PCs, and then the transformed cells migrate to BM. MM develops after the several processes induced by genetic and environmental factors in BM [3,4].
In the last two decades, the development of novel agents, such as immunomodulatory drugs, proteasome inhibitors, monoclonal antibodies, and histone deacetylase inhibitors (HDACis), as well as the high-dose chemotherapy, followed by autologous stem cell transplantation, dramatically improved patients’ outcomes [5,6]. However, despite the high response rates to the initial therapy, almost all patients develop drug resistance over time, and MM is still considered to be incurable. For the migration of MM cells to BM and following the multistep transformation to malignant diseases, numerous intra- and extra- cellular mechanisms are involved. The interaction with the microenvironment is known to be one of the most important mechanisms for the progression and growth of tumor cells, as well as the development of drug resistance. In this review, we focus on the role of mesenchymal cells as a cellular component of the MM microenvironment. After summarizing the constituents of the BM microenvironment, we further discuss the experimental strategies for studies, knowledge from these experiments, and the possible clinical applications targeting mesenchymal cells.

2. Hematopoietic Microenvironment in BM

2.1. Microenvironment for Hematopoietic Stem Cells or Plasma Cells in Normal BM

Based on the hypothesis that cells surrounding hematopoietic stem cells (HSCs) determine the behavior of HSCs in BM, the effects of cellular components on HSC population have been extensively evaluated [7,8,9,10,11]. As the information available has expanded enormously, now it is known that many types of non-hematopoietic cells, including mesenchymal stem/progenitor cells, mature mesenchymal cells, endothelial cells, sympathetic neurons, non-myelinating Schwann cells, perivascular cells, and mesodermal derived cells, as well as mature hematopoietic cells such as monocytes, macrophages, regulatory T-cells, neutrophils, and megakaryocytes, exhibit crucial roles in the maintenance of the stemness of HSCs and the coordinated production of different types of hematopoietic cells [9,10,12]. These cellular components are involved in hematopoiesis through the direct interaction with HSCs, and also in the indirect ways through the secretion of soluble factors (cytokines, chemokines, and extracellular matrix (ECM) proteins) and small vesicles of exosomes packaging microRNAs (miRNAs), and the regulation of other microenvironmental cells or factors.
One of the well-studied subjects regarding the microenvironment in BM is mesenchymal lineage. Mature mesenchymal cells include bone, cartilage, fat, and muscle cells. Mesenchymal stromal cells (MSCs) with the ability to perform multi-lineage differentiation, self-renewal, and reconstitution in vivo are detected in several organs, such as BM, adipose tissue, connective tissue, umbilical cord, and placental tissue [8,11]. In BM cavity, a range of adipo- and osteo-lineage cells are found to exist at different developmental stages, including the most primitive MSCs and several distinct stages of lineage-committed progenitors, to terminally differentiated cells. In vivo experiments using transgenic mouse labeled with Nestin, leptin-receptor, or CXC chemokine ligand (CXCL), 12 have been useful to determine the critical roles of MSCs located in the perivascular area in HSC maintenance, and that the osteoprogenitors support B lymphopoiesis [11,13,14,15,16,17].
Regarding the microenvironment of plasma cells, it is reported that plasmablasts, which express high levels of CXC chemokine receptor (CXCR) 4, migrate and home to BM, where they are retained by CXCL12-expressing stromal cells, from secondary lymphoid organs [17,18,19,20,21]. Other chemokines, including CXCL9, CXCL10, and CXCL11 in mice, which are the ligands of CXCR3, and CXCL 16 in human, which is the ligand of CXCR6, are also involved in the entry of plasmablasts into BM [22,23,24]. Direct adhesion signals between PCs and stromal cells, including very late antigen 4 (VLA-4), a dimer of integrin α4 and β1, and the receptor for vascular cell adhesion molecule 1 (VCAM-1) and fibronectin, and the lymphocyte function associated antigen 1 (LFA-1), a dimer of integrin αL and β2, and the receptor for intercellular adhesion molecule 1 (ICAM-1), are essential [17,18,19,25,26]. For the survival of PCs, cytokines, including a proliferation-inducing ligand (APRIL) and B-cell activating factor (BAFF), members of tumor necrosis factor (TNF) superfamily, and IL-6, are critical [21,22,27,28]. Among cells which secret APRIL, such as monocytes, macrophages, eosinophils, osteoclasts, and fibroblasts, eosinophils have been shown to play the important role as the PC microenvironment in mice [29]. Plasma cells in BM were localized together with eosinophils, and the eosinophil-deleted mice exhibited a decrease in the number of long-lived PCs in BM with migration of PCs to spleen. For the survival of long-lived plasma cells, the retention with CXCL12 is also known to be important [14,17,22,26]. Tokoyoda et al. reported that plasma cells in murine BM were located in contact with CXCL12-abundant reticular cells, and CXCR4-deletion in B-lineage cells relocated PCs from BM to spleen, indicating an essential role of mesenchymal cells as the cellular component of PC microenvironment [17].

2.2. Microenvironment for Malignant Cells in BM

In hematological malignancies, the effects of the microenvironment are higher than previously thought [9,30,31,32]. Neoplastic cells interact with almost all of the cellular components in BM, such as endothelial, mesenchymal, and hematopoietic cells, inducing a favorable environment for their survival. Leukemic cells are involved in the secretion of various types of soluble factors and the differentiation of MSCs [33,34,35,36]. Dissemination of malignant cells changes the oxygen concentration in BM and enhances angiogenesis via increasing the levels of hypoxia-inducible factors (HIFs), angiopoietin and proangiogenic cytokines [30,32]. The tumor-specific microenvironment affects the survival and drug resistance of tumor cells [31]. For example, recent studies have reported that, in mice with myeloproliferative neoplasms, MSCs in BM are essential for tumor proliferation and creation of the fibrotic environment [37,38]. Furthermore, the tumor microenvironment suppresses the immune system through the direct activation of regulatory T-cells, inhibition of B-cell proliferation, and cytokine secretion [32,39,40]. Intriguingly, several studies proved that the abnormal BM microenvironment could lead to the development of hematological malignancies [9,38,41,42,43,44]. With the deletion of Dicer-1 in murine osteoprogenitors, normal HSCs were transformed into the myelodysplasia, which subsequently developed acute myeloid leukemia [41]. Kode et al. reported that constitutive activation of β-catenin in osteoblast induced leukemogenic transformation in mice [43,44].
Multiple myeloma cells, which reside in BM, in most cases, are highly dependent on the BM microenvironment, especially mesenchymal cells. In the following section, we focus on the studies investigating the role of mesenchymal cells in MM with different points of view.

3. The Role of Mesenchymal Cells in Myeloma Microenvironment

The crosstalk between MM cells and other cell types in BM supports the dissemination, survival, migration, transformation, drug resistance, and relapse of MM cells [32,45,46]. MM cells and the surrounding cells adapt to the microenvironmental condition to favor MM maintenance through altering the concentrations of soluble factors and oxygen, and enhancing angiogenesis and hypoxia [47,48,49]. Hematopoiesis, osteogenesis, adipogenesis, and immunity are also affected in the MM microenvironment (Figure 1).
There are various ways for studying the pathogenesis of MM microenvironment. In this section, we discuss the results obtained from using different types of approaches for the research about the role of mesenchymal cells as the MM microenvironment. Like other cancer types, MM has special properties favoring survival. Additionally, MM develops from the precursor state of MGUS, and there are many steps underlying it until the final evolution to extramedullary diseases [50]. The treatment of MM patients after the diagnosis sometimes continues over decades. In studies on MM, depending on the objective of the research, the experimental strategies and subjects are decided to clarify what is really happening in patients, and some of technical problems remain unresolved.

3.1. Studies with Myeloma Cells

The environmental influences are traceable through analyzing MM cells, and MM cells also impact MSCs. Studies using tumor cells have shown that MM cells secret the inhibitory factors for osteoblastogenesis, such as hepatocyte growth factor (HGF) and TNFα [49,51]. Inhibitors of canonical Wnt signaling, including Dickkopf (DKK)1, secreted frizzled related protein (sFRP) −2 and −3, and sclerostin, are also produced by MM cells, leading the suppression of osteoblast formation [52,53,54,55]. Activated signaling of basic fibroblast growth factor (bFGF)1 and TNFα, secreted by MM cells, stimulates IL-6 production in MSCs [49,56].
The results of several studies with MM cells indicate that they interact with the surrounding cells via the adjacent and direct contact. Chemokines, such as CC chemokine ligand (CCL) 25, produced by MM cells, and CXCR4 expressed on the surface of MM cells, lead to the migration of MM cells close to MSCs [32,48,57,58]. Cell adhesion molecules, including integrins, cadherins, and selectins, are also important for the direct interaction between MM cells and MSCs [59,60,61,62,63,64,65,66,67,68]. Overexpression of c-MAF, which is observed in half of patients with MM, increases integrin β7 at both RNA and protein levels in MM cells, and this molecule induces the adhesion of MM cells to MSCs, thereby leading the induction of vascular endothelial growth factor (VEGF) [69,70]. Another report indicated that the interaction of integrin α4β7, with its counter-receptor, mucosal addressin cell adhesion molecule (MADCAM)-1, contributed to the retention of MM cells in BM [64]. Recently, we identified the activated form of integrin β7 as a MM-specific marker through screening the monoclonal antibodies reacting with MM cell lines [65]. In our observation, MM cells derived from approximately 90% of patients (45 of 51 samples) expressed active integrin β7, while integrin β7 expressed on T- and B-cells mainly corresponded to the resting structure. Although it is known that large conformational changes are required for activating integrins, the detailed mechanisms in MM remain unknown.
Using multicolor flow-cytometry technique, Paiva et al. showed that the residual disease clones after treatment exhibited high expression of integrins, such as integrin α4, α5, and β1, as well as CXCR4, compared to MM cells isolated before the treatment [66,67]. These results indicated that residual MM cells directly interact with the special microenvironment and, thus, could be protected from chemotherapy. Loss of activated leukocyte cell adhesion molecule (ALCAM) expression on residual MM cells was involved in chemoresistance; however, the function of ALCAM in the interaction with the microenvironment is not yet fully understood.

3.2. Studies with Human MSCs

The experiments using MSCs isolated from MM patients’ BM (MM-MSCs) have provided direct evidence for the important roles of the mesenchymal cells in MM pathogenesis and the interaction between MM cells and MSCs. MM-MSCs are different from those derived from normal healthy donors (ND-MSCs), and these alternations are not caused by normal responses to osteolysis induced by activated osteoclasts [32,45,46,71,72]. Due to the difficulties in the isolating MSCs from BM, studies on MM-MSC were limited for a long time.
In 2000s, genetic analyses with MM-MSCs demonstrated that transcriptomes, such as IL-6, DKK1 and growth differentiation factor (GDF)15, which regulate angiogenesis, osteogenic differentiation, and tumor growth, were distinctively expressed in MM-MSCs, as compared to ND-MSCs [73,74]. Another study using bioinformatics analyses showed that the genes associated with cell cycle, immune response, and bone metabolism were significantly regulated [75,76,77]. In vitro experiments with a co-culture system indicated that the supportive capacity of T-cell proliferation was impaired with high expression of IL-6 by MM-MSCs [78,79]. The proliferation capacity of MSCs was inhibited by reduced expression of the receptors for platelet-derived growth factor (PDGF)α, PDGFβ, insulin-like growth factor (IGF) 1, epidermal growth factor, and bFGF on MM-MSCs, while others reported a comparable expansion between MM-MSCs and HD-MSCs [73,78,79,80,81,82]. Other studies have demonstrated the concentration of various soluble factors, such as IL-10, IL-1β, IL-3, TNFα, stem cell factor, granulocyte-macrophage colony-stimulating factor (GM-CSF), HGF, BAFF, decorin, and CCL3, in MM microenvironment are altered by MM-MSCs [75,77,83,84,85,86,87]. For the regulation of cytokine secretion, Li et al. reported that the elongation of telomere length in MM-MSCs affected the expression of IL-6 and CCL3 in MM cells [84]. McNee et al. showed that the upregulation of peptidyl arginine deaminase (PADI)2 mediated IL-6 production in MM-MSCs through the enzymatic deamination of histone H3 arginine 26 to form citrulline [88]. The upregulation of PADI2 was identified in MSCs derived from both MGUS and MM patients, indicating that the microenvironment is altered at the stage of MGUS. The activation of Notch signaling in MM-MSCs induced by MM cells has also been reported as the mechanism of IL-6 and VEGF production [89].
Increased levels of soluble factors, such as IL-6, BAFF, and GDF15, and a decreased level of decorin in BM plasma, promote the proliferation of MM cells [73,74,85,90]. In addition, recent studies showed that MM-MSCs secreted exosomes and microvesicles, which are transferred to MM cells [91,92]. The exosomes derived from MM-MSCs included oncogenic proteins, cytokines, and adhesion molecules and promoted MM proliferation, as compared to those derived from HD-MSCs [91]. Specifically, the lower levels of miRNA-15a, a tumor suppressor, and higher expression of miRNA-10a in these vesicles were identified [91,92]. While transferred miRNA-10a enhanced the proliferation of MM cells, the accumulation in MM-MSCs inhibited the proliferation of MSCs with the induced apoptosis, suggesting the therapeutic possibility of the transfer inhibition of MM-MSC-derived exosomes [92].
Using culture experiments, which are helpful for analyzing functions and interactions of specific cell subsets or molecules, the importance of interaction with CXCL12, VCAM-1, and ECM proteins has been revealed [63,93,94]. Another interesting study performed by Kikuchi et al. showed that stromal cell adhesion-dependent epigenetic alternation in MM cells contributes to drug resistance [95,96]. They demonstrated that inactivation of enhancer of zeste homolog (EZH) 2 by MM-MSCs reduced the abundance of H3K27me3 and altered the expression of several genes in MM cells. The direct interaction between MSCs and MM cells blocked H3K27 hypermethylation induced by cytotoxic anti-cancer agents, such as doxorubicin and melphalan, and maintained the expression of anti-apoptotic genes of BCL2, IGF1, and HIF1a, leading to the drug-resistant survival of MM cells. The indirect effect of MM-MSCs with the secreted soluble factors on upregulation of KDM6B gene, which removes H3K27 di- and tri-methylation in MM cells, has also been reported [97].
The abnormalities of mesenchymal cells are also involved in the bone complication in MM together with the activation of osteoclast [82,98,99]. MM-MSCs derived from patients with bone lesions showed distinct gene expression profiles, compared to those from patient without bone complications [86]. The differentiation capacity of MM-MSCs to adipocytes and osteoblasts is suppressed [78,79,100]. Circulating cytokines, such as transforming growth factor (TGF)β, TNFα, interferon (IFN)γ, IL-1β, and IL-6, as well as the direct interaction with MM cells, promote the apoptosis of osteoprogenitors [89,101,102,103]. Inhibitors of the canonical Wnt signaling pathway produced by MM cells and altered expression of miRNA, such as miRNA-138 and -223 in MM-MSCs, are also shown to impair osteoblast differentiation [52,80,89,104].
The underlying mechanisms of the transformation of HD-MSCs to MM-MSCs are partially understood. As mentioned above, soluble factors in BM influence the differentiation of MSCs. Exposure to MM cells also induces the special MSC phenotypes. Xu et al. reported that miRNA-135b, which was aberrantly expressed in MM-MSCs and inhibited the osteogenesis, was upregulated in ND-MSCs co-cultured with MM cells, while the expression recapitulated to the normal level after removing MM cells from the culture [105]. Interestingly, there is evidence indicating that MM-MSCs remain dysfunctional without the coexistence of MM cells, for example, after the achievement of complete remission in MM patients [45,78]. MM-MSCs cultured without MM cells demonstrated overproduction of cytokines, such as IL-6 and GDF15, thereby favoring growth of MM cells [73]. MM-MSCs possess genomic alternations with a specific pattern of gene region, and aberrant methylation status in MM-MSCs has also been reported [106,107,108]. Adamik et al. demonstrated that heterochromatin silencing of the promoter of Runt-related transcription factor (RUNX)2 prolongs impaired osteogenesis [109]. The transcriptional repressor of RUNX2, Gfi1, which is upregulated in MM-MSC by TNFα or IL-7 during MM progression, recruits HDAC1 and EZH2, and increases H3K27me3 on RUNX2 [109,110].
In many studies about human MSCs, adhesion methods for isolation have been commonly used, considering the difficulties in direct isolation of MSCs from BM and the proliferation capacity in vitro [8]. While macrophages and other non-mesenchymal cells are often contaminated in the adhesion methods with murine BM, mesenchymal lineage cells are efficiently cultivated in humans. The International Society for Cellular Therapy (ISCT) issued the minimal criteria for defining MSCs in BM [111]. According to the recommendation from ISCT, the cells adhering to plastic after culturing isolated marrow cells, which lack expression of CD45, CD34, CD14, CD11b, CD79a, or CD19, but express CD105, CD73, and CD90, and are capable of differentiating into osteogenic, adipogenic, and chondrogenic lineages, should be considered as MSCs. On the other hand, recent technical advances in flow cytometry have enabled the isolation of MSCs, which express several surface markers, such as CD271, CD146, SSEA4, CD49a, and PDGF receptors in human, without maintaining cell cultures. It has been reported that there are differences between freshly isolated and cultured MSCs in terms of expression of RNAs, cell surface molecules, and the reconstitution capacity in vivo [76,112,113,114,115]. Although the experimental results mentioned above were acquired from studies with MSCs derived from patients, it is possible that MSCs isolation method might affect the results. This needs to be considered especially for translating them into therapeutic applications.

3.3. Studies with Mouse Models

The concept of microenvironment explains that the neighboring subsets of cells and extracellular substrates regulate the survival and function of MM cells in the region where MM cells reside. Ex vivo studies using the cells isolated from patients have limitations on the anatomical evaluation, and the condition in culture experiments cannot recapitulates the MM microenvironment in BM. Mouse models are useful tools for understanding the interaction between MM cells and the surrounding microenvironment.
Mouse strain C57BL/KalwRij is known to spontaneously develop monoclonal disorder of immunoglobulin-producing B-cells, resembling MGUS in human [116,117,118]. Although the probability of progression to MM and Waldenstrom Macroglobulinemia is less than 1% in primary mice, the recipient mice transplanted with primary cells or the established 5T cell lines develop MM with bone lytic lesions. Using this 5T mouse model, Fowler et al. reported that adiponectin secreted from MM-MSCs decreased with the progression of disease after 5T cell transplantation, and a similar phenomenon was observed in MGUS patients who subsequently developed MM [119]. Although adiponectin was originally identified as a protein secreted from adipose tissue, it has since been recognized that adiponectin is also secreted by a range of cell types, including MSCs in BM. Adiponectin induced MM cell apoptosis, and its therapeutic effects were shown in the study. Using this model, they also demonstrated the upregulated DKK1 secretion from MM-MSCs and the critical role of BMP signaling in bone destruction induced by MM [120,121]. Intriguingly, taking advantage of mouse experiments, Lawson et al. dissected the microenvironments for two types of MM cell fractions with distinct cell-cycle status [122]. They visualized eGFP-labeled 5TGM1 MM cells in real time, using intravital two-photon microscopy, and showed that MM cells colonized in the endosteal area after transplantation. Furthermore, dormant MM cells, which highly expressed VCAM-1 and AXL, were retained in direct contact with bone-lining osteoblasts. Conversely, actively dividing MM cells were found at locations distant from the bone surface, and bone resorption induced by osteoclast activation released dormant MM cells from their own microenvironment, thereby entering into the cell cycle. They also demonstrated that the dormancy in MM cells was associated with drug resistance, using melphalan-treated mice. In addition, the distinct transcriptome profile of the dormant MM cells was induced by cocultures with osteoblastic cells [123].
Several types of transgenic (Tg) mice, in which the expression of MAF, XBP-1, or MYC genes is enforced in the B-cell compartment, develop MGUS and MM [124,125,126]. Among them, Vk*MYC Tg mice, in which the activation of MYC is under the control of Ig kappa light chain gene regulatory elements, show anemia, osteolytic bone lesion, and kidney damage, resembling human MM symptoms, and the MM cells are transplantable with the reconstitution in BM [126]. This suggests a potential application of studying the microenvironment. Using this mouse model, the detailed mechanisms of immunomodulation, anemia, and hypoxic condition in MM-specific microenvironment have been revealed [47,127,128,129].

3.4. Studies with Humanized Mice

There are species differences of MM cells and MSCs between human and mice. To evaluate the interaction of human MM cells with the microenvironment in vivo, the immunodeficient mice, such as severe combined immunodeficiency (SCID) and non-obese diabetic (NOD)/SCID mice, were first used [116,130,131]. Subsequently, the introduction of intratibial injection methods, as well as the development of NOD/SCID-based highly immunodeficient mice, such as NOG (NOD/Shi-SCID/IL2Rγ−/−), and NSG (NOD/LtSz-SCID/IL2Rγ−/−), and Rag2−/− IL2Rγ−/− mice, has led to the improvement in the ability of BM engraftment of patient MM cells [132,133,134,135]. In these humanized models, MM cells labeled with bioluminescent and fluorescent reporters can be tracked by using MRI, PET, and micro CT imaging [132,136]. MM cells in SCID mice disseminate in various organs, such as the spleen, liver, lungs, and BM, after intravenous injection, while subcutaneous injection causes palpable tumors with growth at injection site. Only a few types of human cell lines, such as KMS12-BM and U266, reconstitute dominantly in BM after injection into the immunodeficient mice [116,132]. Using these mouse models, in which the bone lytic lesion develops with the MM cell line infiltration in mice BM, the inhibitory effect of neutralizing human IL-6 receptor on MM proliferation, and the co-localization of human MM cells in endosteal area have been shown [132,137]. In some studies, to assess the role and therapeutic possibility, co-injection of MSCs producing soluble factors, such as IFN and osteoprotegerin, was conducted [131,138].
Recently, mice expressing human genes were developed to mimic human BM environment [139,140]. Humanization of IL-6, along with macrophage-colony-stimulating factor, IL-3, GM-CSF, thrombopoietin, and signal regulatory protein-α, resulted in the enhancement of patient MM cell-reconstitution in recipient BM [139]. No improvement of PC engraftment was observed in immunodeficient mice with human BAFF expression [140]. These novel techniques of mouse humanization would be useful for studying the MM microenvironment.
To reproduce human microenvironment, a distinct type of mouse model, which is known as the SCID-hu model, has been established [136,141,142]. In this model, MM cells, such as IL-6-dependent INA-6 MM cell line and primary patient MM cells, were injected into a human bone chip previously implanted in the flank of mice, and the successful engraftment in human BM was observed. This model provided the evidence of the requirement of IL-6 and human specific BM microenvironment, and a useful tool for evaluating the effects of drugs [119,136]. Using the SCID-hu model, osteoblast suppression through Ephrin B2/B4 interaction was described [143]. However, this model has several limitations on the restricted availability of human fetal bones and different nature between fetus and adult. Although rabbit bone is sometimes used for the replacement of fetal bone chip, the problems remain unresolved [135,144]. To overcome the limitations of SCID-hu models, fetal bone chip can be replaced by using an adult BM ossicle with three-dimensional scaffold, and work on technical improvements is currently in progress [145,146,147].

4. Therapeutic Application Targeting the Effects of MM-MSCs

Several studies focusing on MM-MSCs have revealed the important roles in MM pathogenesis, which include (i) the supportive effects on MM proliferation, (ii) the suppression of osteoblastogenesis from MSCs, (iii) the induction of drug resistance and dormancy in MM cells, and (iv) the immunosuppressive effects (Table 1). Some of the experimental methods, such as cultures and humanized mouse models, have been useful in predicting clinical efficacies and have shown that the inhibition of pathways in MSCs activated by MM could be promising candidates for the therapeutic target. Here, we explain the results of preclinical and clinical studies on agents with high potential for clinical application (Table 2).
MM-MSCs support the proliferation of MM cells with the production of cytokines, such as IL-6 and BAFF, transfer of exosomes into MM cells, and the direct interaction. The anti-IL-6 chimeric antibody, siltuximab has been tested in MM patients [148,149,150]. While treatment with siltuximab did not improve the outcome in studies of newly diagnosed and relapsed/refractory patients, another clinical study showed the inhibition tended to delay the progression of high-risk smoldering MM. Preclinical studies using cultures and humanized mice showed the inhibitors of JAK signaling, such as tofacitinib and ruxolitinib, were effective in suppressing MM cell proliferation [151,152]. Umezu et al., reported the inhibitory effects of the extracellular vesicle release inhibitor, FTY720, which has been approved for the treatment of multiple sclerosis in US and Europe, on MM growth in vitro and in vivo [92].
The capacity of MM-MSCs to differentiate osteoblasts is suppressed by the increased levels of cytokines, such as Wnt signaling inhibitors, TGFβ, and IL-6, as well as cell-intrinsic pathways. In clinical trials with DKK1 inhibitor, BHQ880, and TGFβ antagonists, such as sotatercept and luspatercept, significant improvements in bone complications have not been reported so far [153,154,166]. Considering the suppression of osteogenic genes with aberrant methylation status in MM-MSCs, epigenetic inhibitors, such as DNA methyltransferase inhibitors (DNMTis) and HDACis, which are used for the various types of cancer therapies, might be effective [108,109]. Knowledge about the effects of DNMTi on MSCs is limited [155]. Preclinical studies have shown that some HDACis, such as trichostain A, valproic acid, and sodium butyrate, promote osteoblastogenesis from MSCs, and the effects of vorinostat are controversial [156,157,158,159,160]. Clinical observations, as well as preclinical studies, have demonstrated that bortezomib, a proteasome inhibitor, which is commonly used for MM treatment, induces the differentiation to osteoblast [99,161].
The adhesion to mesenchymal cells within the special endosteal microenvironment induces distinct phenotypes characterized by the cell-cycle status and drug resistance of MM cells. Several studies in vitro and in MM mouse models indicate that blocking MM cell adhesion could release MM cells residing in the dormant microenvironment and improve the sensitivity to the treatment. Natalizumab, which has been used for the treatment of multiple sclerosis in clinical practices, prevents the integrin α4-mediated adhesion to their counter-receptors, including VCAM-1 and MADCAM-1. This molecule inhibited the proliferation of MM cells in preclinical studies [162]. Clinical efficacies in MM patients targeting the CXCL12–CXCR4 axis by using plerixafor, olaptesed pegol, and ulocuplumab are currently under investigation [163,164,165].
MM-MSCs exhibit immunosuppressive effects with a decreased ability to support T- and B-lymphocytes [75,78,79]. MM-MSCs regulate both innate and adaptive immune responses through the secretion of IFNγ, IL-1β, IL-6, IL-10, and TNFα [75,83,84]. Considering the recent advances in immune therapy, such as the application of immunocheckpoint inhibitors and chimeric antigen receptor (CAR)-T therapy, the regulation of immune status would be more important in the near future [167,168]. For the treatment of inflammatory diseases and tissue injuries, MSCs derived from healthy donors have already been used in clinical practices [39,169].

5. Conclusions and Perspectives

Numerous studies have shown that MM-MSCs are functionally and genetically different from ND-MSCs, leading to supportive characteristics for the migration to BM, proliferation, drug resistance of MM cells, and survival of residual tumor cells. At present, our knowledge regarding the role of MSCs in the emergence of MGUS, progression to MM, and transformation of MM clones during treatment is limited [3,50]. MSCs might affect the etiological events at HSC and B progenitor levels, which contribute to the progression of MM in some patients [170,171]. The effects of MM treatment on MSCs are also largely unknown [76]. In the near future, the improvement in humanized mouse models with advances in gene transduction methods and construction of three-dimensional tissues with scaffold could yield valuable information. Novel techniques to analyze RNA, DNA, and protein expression at the single-cell level, as well as spatial transcriptomic analyses, could help dissect the heterogeneity in MM-MSCs [35,172]. MM is still essentially incurable, and understanding the pathogenesis of MM-MSCs could contribute to developing novel therapeutic strategies and drugs.

Author Contributions

M.I. performed the literature review and drafted the manuscript. N.H. edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported in part by Japan Society for the Promotion of Science KAKENHI, grant number 19K08814 and 19K17829.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kyle, R.A.; Rajkumar, S.V. Multiple myeloma. N. Engl. J. Med. 2004, 351, 1860–1873. [Google Scholar] [CrossRef]
  2. 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]
  3. Pawlyn, C.; Morgan, G.J. Evolutionary biology of high-risk multiple myeloma. Nat. Rev. Cancer 2017, 17, 543–556. [Google Scholar] [CrossRef]
  4. Manier, S.; Salem, K.Z.; Park, J.; Landau, D.A.; Getz, G.; Ghobrial, I.M. Genomic complexity of multiple myeloma and its clinical implications. Nat. Rev. Clin. Oncol. 2017, 14, 100–113. [Google Scholar] [CrossRef] [PubMed]
  5. Kristinsson, S.Y.; Landgren, O.; Dickman, P.W.; Derolf, A.R.; Björkholm, M. Patterns of survival in multiple myeloma: A population-based study of patients diagnosed in Sweden from 1973 to 2003. J. Clin. Oncol. 2007, 25, 1993–1999. [Google Scholar] [CrossRef] [PubMed]
  6. Sonneveld, P.; Avet-Loiseau, H.; Lonial, S.; Usmani, S.; Siegel, D.; Anderson, K.C.; Chng, W.J.; Moreau, P.; Attal, M.; Kyle, R.A.; et al. Treatment of multiple myeloma with high-risk cytogenetics: A consensus of the International Myeloma Working Group. Blood 2016, 127, 2955–2962. [Google Scholar] [CrossRef] [PubMed]
  7. Schofield, R. The relationship between the spleen colony-forming cell and the haemopoietic stem cell. Blood Cells 1978, 4, 7–25. [Google Scholar]
  8. Kfoury, Y.; Scadden, D.T. Mesenchymal cell contributions to the stem cell niche. Cell Stem Cell 2015, 16, 239–253. [Google Scholar] [CrossRef] [Green Version]
  9. Pinho, S.; Frenette, P.S. Haematopoietic stem cell activity and interactions with the niche. Nat. Rev. Mol. Cell Biol. 2019, 20, 303–320. [Google Scholar] [CrossRef]
  10. Crane, G.M.; Jeffery, E.; Morrison, S.J. Adult haematopoietic stem cell niches. Nat. Rev. Immunol. 2017, 17, 573–590. [Google Scholar] [CrossRef]
  11. Kokkaliaris, K.D. Dissecting the spatial bone marrow microenvironment of hematopoietic stem cells. Curr. Opin. Oncol. 2020, 32, 154–161. [Google Scholar] [CrossRef] [PubMed]
  12. Mendez-Ferrer, S.; Scadden, D.T.; Sanchez-Aguilera, A. Bone marrow stem cells: Current and emerging concepts. Ann. N. Y. Acad. Sci. 2015, 1335, 32–44. [Google Scholar] [CrossRef] [PubMed]
  13. Yu, V.W.; Scadden, D.T. Heterogeneity of the bone marrow niche. Curr. Opin. Hematol. 2016, 23, 331–338. [Google Scholar] [CrossRef]
  14. Wei, Q.; Frenette, P.S. Niches for Hematopoietic Stem Cells and Their Progeny. Immunity 2018, 48, 632–648. [Google Scholar] [CrossRef] [Green Version]
  15. Yu, V.W.; Lymperi, S.; Oki, T.; Jones, A.; Swiatek, P.; Vasic, R.; Ferraro, F.; Scadden, D.T. Distinctive Mesenchymal-Parenchymal Cell Pairings Govern B Cell Differentiation in the Bone Marrow. Stem Cell Rep. 2016, 7, 220–235. [Google Scholar] [CrossRef] [Green Version]
  16. Cordeiro Gomes, A.; Hara, T.; Lim, V.Y.; Herndler-Brandstetter, D.; Nevius, E.; Sugiyama, T.; Tani-Ichi, S.; Schlenner, S.; Richie, E.; Rodewald, H.R.; et al. Hematopoietic Stem Cell Niches Produce Lineage-Instructive Signals to Control Multipotent Progenitor Differentiation. Immunity 2016, 45, 1219–1231. [Google Scholar] [CrossRef] [Green Version]
  17. 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]
  18. Chang, H.D.; Tokoyoda, K.; Radbruch, A. Immunological memories of the bone marrow. Immunol. Rev. 2018, 283, 86–98. [Google Scholar] [CrossRef]
  19. Cyster, J.G. Homing of antibody secreting cells. Immunol. Rev. 2003, 194, 48–60. [Google Scholar] [CrossRef]
  20. Radbruch, A.; Muehlinghaus, G.; Luger, E.O.; Inamine, A.; Smith, K.G.; Dorner, T.; Hiepe, F. Competence and competition: The challenge of becoming a long-lived plasma cell. Nat. Rev. Immunol. 2006, 6, 741–750. [Google Scholar] [CrossRef]
  21. Belnoue, E.; Tougne, C.; Rochat, A.F.; Lambert, P.H.; Pinschewer, D.D.; Siegrist, C.A. Homing and adhesion patterns determine the cellular composition of the bone marrow plasma cell niche. J. Immunol. 2012, 188, 1283–1291. [Google Scholar] [CrossRef] [PubMed]
  22. Chu, V.T.; Berek, C. The establishment of the plasma cell survival niche in the bone marrow. Immunol. Rev. 2013, 251, 177–188. [Google Scholar] [CrossRef] [PubMed]
  23. Hauser, A.E.; Debes, G.F.; Arce, S.; Cassese, G.; Hamann, A.; Radbruch, A.; Manz, R.A. Chemotactic responsiveness toward ligands for CXCR3 and CXCR4 is regulated on plasma blasts during the time course of a memory immune response. J. Immunol. 2002, 169, 1277–1282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. 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]
  25. DiLillo, D.J.; Hamaguchi, Y.; Ueda, Y.; Yang, K.; Uchida, J.; Haas, K.M.; Kelsoe, G.; Tedder, T.F. Maintenance of long-lived plasma cells and serological memory despite mature and memory B cell depletion during CD20 immunotherapy in mice. J. Immunol. 2008, 180, 361–371. [Google Scholar] [CrossRef]
  26. Zehentmeier, S.; Roth, K.; Cseresnyes, Z.; Sercan, O.; Horn, K.; Niesner, R.A.; Chang, H.D.; Radbruch, A.; Hauser, A.E. Static and dynamic components synergize to form a stable survival niche for bone marrow plasma cells. Eur. J. Immunol. 2014, 44, 2306–2317. [Google Scholar] [CrossRef] [Green Version]
  27. Belnoue, E.; Pihlgren, M.; McGaha, T.L.; Tougne, C.; Rochat, A.F.; Bossen, C.; Schneider, P.; Huard, B.; Lambert, P.H.; Siegrist, C.A. APRIL is critical for plasmablast survival in the bone marrow and poorly expressed by early-life bone marrow stromal cells. Blood 2008, 111, 2755–2764. [Google Scholar] [CrossRef]
  28. O’Connor, B.P.; Raman, V.S.; Erickson, L.D.; Cook, W.J.; Weaver, L.K.; Ahonen, C.; Lin, L.L.; Mantchev, G.T.; Bram, R.J.; Noelle, R.J. BCMA is essential for the survival of long-lived bone marrow plasma cells. J. Exp. Med. 2004, 199, 91–98. [Google Scholar] [CrossRef]
  29. Chu, V.T.; Frohlich, A.; Steinhauser, G.; Scheel, T.; Roch, T.; Fillatreau, S.; Lee, J.J.; Lohning, M.; Berek, C. Eosinophils are required for the maintenance of plasma cells in the bone marrow. Nat. Immunol. 2011, 12, 151–159. [Google Scholar] [CrossRef]
  30. Le, P.M.; Andreeff, M.; Battula, V.L. Osteogenic niche in the regulation of normal hematopoiesis and leukemogenesis. Haematologica 2018, 103, 1945–1955. [Google Scholar] [CrossRef] [Green Version]
  31. Witkowski, M.T.; Kousteni, S.; Aifantis, I. Mapping and targeting of the leukemic microenvironment. J. Exp. Med. 2020, 217. [Google Scholar] [CrossRef]
  32. Ghobrial, I.M.; Detappe, A.; Anderson, K.C.; Steensma, D.P. The bone-marrow niche in MDS and MGUS: Implications for AML and MM. Nat. Rev. Clin. Oncol. 2018, 15, 219–233. [Google Scholar] [CrossRef]
  33. Krause, D.S.; Fulzele, K.; Catic, A.; Sun, C.C.; Dombkowski, D.; Hurley, M.P.; Lezeau, S.; Attar, E.; Wu, J.Y.; Lin, H.Y.; et al. Differential regulation of myeloid leukemias by the bone marrow microenvironment. Nat. Med. 2013, 19, 1513–1517. [Google Scholar] [CrossRef] [Green Version]
  34. Battula, V.L.; Le, P.M.; Sun, J.C.; Nguyen, K.; Yuan, B.; Zhou, X.; Sonnylal, S.; McQueen, T.; Ruvolo, V.; Michel, K.A.; et al. AML-induced osteogenic differentiation in mesenchymal stromal cells supports leukemia growth. JCI Insight 2017, 2. [Google Scholar] [CrossRef]
  35. Baryawno, N.; Przybylski, D.; Kowalczyk, M.S.; Kfoury, Y.; Severe, N.; Gustafsson, K.; Kokkaliaris, K.D.; Mercier, F.; Tabaka, M.; Hofree, M.; et al. A Cellular Taxonomy of the Bone Marrow Stroma in Homeostasis and Leukemia. Cell 2019, 177, 1915–1932. [Google Scholar] [CrossRef]
  36. Duarte, D.; Hawkins, E.D.; Akinduro, O.; Ang, H.; De Filippo, K.; Kong, I.Y.; Haltalli, M.; Ruivo, N.; Straszkowski, L.; Vervoort, S.J.; et al. Inhibition of Endosteal Vascular Niche Remodeling Rescues Hematopoietic Stem Cell Loss in AML. Cell Stem Cell 2018, 22, 64–77. [Google Scholar] [CrossRef] [Green Version]
  37. Decker, M.; Martinez-Morentin, L.; Wang, G.; Lee, Y.; Liu, Q.; Leslie, J.; Ding, L. Leptin-receptor-expressing bone marrow stromal cells are myofibroblasts in primary myelofibrosis. Nat. Cell Biol. 2017, 19, 677–688. [Google Scholar] [CrossRef]
  38. Dong, L.; Yu, W.M.; Zheng, H.; Loh, M.L.; Bunting, S.T.; Pauly, M.; Huang, G.; Zhou, M.; Broxmeyer, H.E.; Scadden, D.T.; et al. Leukaemogenic effects of Ptpn11 activating mutations in the stem cell microenvironment. Nature 2016, 539, 304–308. [Google Scholar] [CrossRef] [Green Version]
  39. Uccelli, A.; de Rosbo, N.K. The immunomodulatory function of mesenchymal stem cells: Mode of action and pathways. Ann. N. Y. Acad. Sci. 2015, 1351, 114–126. [Google Scholar] [CrossRef]
  40. Ghannam, S.; Pene, J.; Moquet-Torcy, G.; Jorgensen, C.; Yssel, H. Mesenchymal stem cells inhibit human Th17 cell differentiation and function and induce a T regulatory cell phenotype. J. Immunol. 2010, 185, 302–312. [Google Scholar] [CrossRef] [Green Version]
  41. Raaijmakers, M.H.; Mukherjee, S.; Guo, S.; Zhang, S.; Kobayashi, T.; Schoonmaker, J.A.; Ebert, B.L.; Al-Shahrour, F.; Hasserjian, R.P.; Scadden, E.O.; et al. Bone progenitor dysfunction induces myelodysplasia and secondary leukaemia. Nature 2010, 464, 852–857. [Google Scholar] [CrossRef] [Green Version]
  42. Zambetti, N.A.; Ping, Z.; Chen, S.; Kenswil, K.J.G.; Mylona, M.A.; Sanders, M.A.; Hoogenboezem, R.M.; Bindels, E.M.J.; Adisty, M.N.; Van Strien, P.M.H.; et al. Mesenchymal Inflammation Drives Genotoxic Stress in Hematopoietic Stem Cells and Predicts Disease Evolution in Human Pre-leukemia. Cell Stem Cell 2016, 19, 613–627. [Google Scholar] [CrossRef] [Green Version]
  43. Kode, A.; Mosialou, I.; Manavalan, S.J.; Rathinam, C.V.; Friedman, R.A.; Teruya-Feldstein, J.; Bhagat, G.; Berman, E.; Kousteni, S. FoxO1-dependent induction of acute myeloid leukemia by osteoblasts in mice. Leukemia 2016, 30, 1–13. [Google Scholar] [CrossRef] [Green Version]
  44. Kode, A.; Manavalan, J.S.; Mosialou, I.; Bhagat, G.; Rathinam, C.V.; Luo, N.; Khiabanian, H.; Lee, A.; Murty, V.V.; Friedman, R.; et al. Leukaemogenesis induced by an activating beta-catenin mutation in osteoblasts. Nature 2014, 506, 240–244. [Google Scholar] [CrossRef] [Green Version]
  45. Reagan, M.R.; Ghobrial, I.M. Multiple myeloma mesenchymal stem cells: Characterization, origin, and tumor-promoting effects. Clin. Cancer Res. 2012, 18, 342–349. [Google Scholar] [CrossRef] [Green Version]
  46. Xu, S.; De Veirman, K.; De Becker, A.; Vanderkerken, K.; Van Riet, I. Mesenchymal stem cells in multiple myeloma: A therapeutical tool or target? Leukemia 2018, 32, 1500–1514. [Google Scholar] [CrossRef]
  47. Hu, J.; Van Valckenborgh, E.; Menu, E.; De Bruyne, E.; Vanderkerken, K. Understanding the hypoxic niche of multiple myeloma: Therapeutic implications and contributions of mouse models. Dis. Models Mech. 2012, 5, 763–771. [Google Scholar] [CrossRef] [Green Version]
  48. Azab, A.K.; Hu, J.; Quang, P.; Azab, F.; Pitsillides, C.; 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. Hideshima, T.; Chauhan, D.; Podar, K.; Schlossman, R.L.; Richardson, P.; Anderson, K.C. Novel therapies targeting the myeloma cell and its bone marrow microenvironment. Semin. Oncol. 2001, 28, 607–612. [Google Scholar] [CrossRef]
  50. Dhodapkar, M.V. MGUS to myeloma: A mysterious gammopathy of underexplored significance. Blood 2016, 128, 2599–2606. [Google Scholar] [CrossRef]
  51. Standal, T.; Abildgaard, N.; Fagerli, U.M.; Stordal, B.; Hjertner, O.; Borset, M.; Sundan, A. HGF inhibits BMP-induced osteoblastogenesis: Possible implications for the bone disease of multiple myeloma. Blood 2007, 109, 3024–3030. [Google Scholar] [CrossRef] [Green Version]
  52. Tian, E.; Zhan, F.; Walker, R.; Rasmussen, E.; Ma, Y.; Barlogie, B.; Shaughnessy, J.D., Jr. 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. [Google Scholar] [CrossRef]
  53. Giuliani, N.; Morandi, F.; Tagliaferri, S.; Lazzaretti, M.; Donofrio, G.; Bonomini, S.; Sala, R.; Mangoni, M.; Rizzoli, V. Production of Wnt inhibitors by myeloma cells: Potential effects on canonical Wnt pathway in the bone microenvironment. Cancer Res. 2007, 67, 7665–7674. [Google Scholar] [CrossRef] [Green Version]
  54. Oshima, T.; Abe, M.; Asano, J.; Hara, T.; Kitazoe, K.; Sekimoto, E.; Tanaka, Y.; Shibata, H.; Hashimoto, T.; Ozaki, S.; et al. Myeloma cells suppress bone formation by secreting a soluble Wnt inhibitor, sFRP-2. Blood 2005, 106, 3160–3165. [Google Scholar] [CrossRef] [Green Version]
  55. Brunetti, G.; Oranger, A.; Mori, G.; Specchia, G.; Rinaldi, E.; Curci, P.; Zallone, A.; Rizzi, R.; Grano, M.; Colucci, S. Sclerostin is overexpressed by plasma cells from multiple myeloma patients. Ann. N. Y. Acad. Sci. 2011, 1237, 19–23. [Google Scholar] [CrossRef]
  56. Bisping, G.; Leo, R.; Wenning, D.; Dankbar, B.; Padro, T.; Kropff, M.; Scheffold, C.; Kroger, M.; Mesters, R.M.; Berdel, W.E.; et al. Paracrine interactions of basic fibroblast growth factor and interleukin-6 in multiple myeloma. Blood 2003, 101, 2775–2783. [Google Scholar] [CrossRef] [Green Version]
  57. Feng, Y.; Wen, J.; Mike, P.; Choi, D.S.; Eshoa, C.; Shi, Z.Z.; Zu, Y.; Chang, C.C. Bone marrow stromal cells from myeloma patients support the growth of myeloma stem cells. Stem Cells Dev. 2010, 19, 1289–1296. [Google Scholar] [CrossRef]
  58. Xu, S.; Menu, E.; De Becker, A.; Van Camp, B.; Vanderkerken, K.; Van Riet, I. Bone marrow-derived mesenchymal stromal cells are attracted by multiple myeloma cell-produced chemokine CCL25 and favor myeloma cell growth in vitro and in vivo. Stem Cells 2012, 30, 266–279. [Google Scholar] [CrossRef]
  59. Sanz-Rodríguez, F.; Teixidó, J. VLA-4-dependent myeloma cell adhesion. Leuk. Lymphoma 2001, 41, 239–245. [Google Scholar] [CrossRef]
  60. Damiano, J.S.; Cress, A.E.; Hazlehurst, L.A.; Shtil, A.A.; Dalton, W.S. Cell adhesion mediated drug resistance (CAM-DR): Role of integrins and resistance to apoptosis in human myeloma cell lines. Blood 1999, 93, 1658–1667. [Google Scholar] [CrossRef] [Green Version]
  61. Noborio-Hatano, K.; Kikuchi, J.; Takatoku, M.; Shimizu, R.; Wada, T.; Ueda, M.; Nobuyoshi, M.; Oh, I.; Sato, K.; Suzuki, T.; et al. Bortezomib overcomes cell-adhesion-mediated drug resistance through downregulation of VLA-4 expression in multiple myeloma. Oncogene 2009, 28, 231–242. [Google Scholar] [CrossRef] [Green Version]
  62. Ely, S.A.; Knowles, D.M. Expression of CD56/Neural Cell Adhesion Molecule Correlates with the Presence of Lytic Bone Lesions in Multiple Myeloma and Distinguishes Myeloma from Monoclonal Gammopathy of Undetermined Significance and Lymphomas with Plasmacytoid Differentiation. Am. J. Pathol. 2002, 160, 1293–1299. [Google Scholar] [CrossRef] [Green Version]
  63. Landowski, T.H.; Olashaw, N.E.; Agrawal, D.; Dalton, W.S. Cell adhesion-mediated drug resistance (CAM-DR) is associated with activation of NF-kappa B (RelB/p50) in myeloma cells. Oncogene 2003, 22, 2417–2421. [Google Scholar] [CrossRef] [Green Version]
  64. Natoni, A.; Farrell, M.L.; Harris, S.; Falank, C.; Kirkham-McCarthy, L.; Macauley, M.S.; Reagan, M.R.; O’Dwyer, M. Sialyltransferase inhibition leads to inhibition of tumor cell interactions with E-selectin, VCAM1, and MADCAM1, and improves survival in a human multiple myeloma mouse model. Haematologica 2020, 105, 457–467. [Google Scholar] [CrossRef]
  65. Hosen, N.; Matsunaga, Y.; Hasegawa, K.; Matsuno, H.; Nakamura, Y.; Makita, M.; Watanabe, K.; Yoshida, M.; Satoh, K.; Morimoto, S.; et al. The activated conformation of integrin beta7 is a novel multiple myeloma-specific target for CAR T cell therapy. Nat. Med. 2017, 23, 1436–1443. [Google Scholar] [CrossRef]
  66. Paiva, B.; Corchete, L.A.; Vidriales, M.B.; Puig, N.; Maiso, P.; Rodriguez, I.; Alignani, D.; Burgos, L.; Sanchez, M.L.; Barcena, P.; et al. Phenotypic and genomic analysis of multiple myeloma minimal residual disease tumor cells: A new model to understand chemoresistance. Blood 2016, 127, 1896–1906. [Google Scholar] [CrossRef] [Green Version]
  67. Paino, T.; Paiva, B.; Sayagues, J.M.; Mota, I.; Carvalheiro, T.; Corchete, L.A.; Aires-Mejia, I.; Perez, J.J.; Sanchez, M.L.; Barcena, P.; et al. Phenotypic identification of subclones in multiple myeloma with different chemoresistant, cytogenetic and clonogenic potential. Leukemia 2015, 29, 1186–1194. [Google Scholar] [CrossRef] [Green Version]
  68. Furukawa, M.; Ohkawara, H.; Ogawa, K.; Ikeda, K.; Ueda, K.; Shichishima-Nakamura, A.; Ito, E.; Imai, J.I.; Yanagisawa, Y.; Honma, R.; et al. Autocrine and Paracrine Interactions between Multiple Myeloma Cells and Bone Marrow Stromal Cells by Growth Arrest-specific Gene 6 Cross-talk with Interleukin-6. J. Biol. Chem. 2017, 292, 4280–4292. [Google Scholar] [CrossRef] [Green Version]
  69. 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]
  70. Moreaux, J.; Hose, D.; Jourdan, M.; Reme, T.; Hundemer, M.; Moos, M.; Robert, N.; Moine, P.; De Vos, J.; Goldschmidt, H.; et al. TACI expression is associated with a mature bone marrow plasma cell signature and C-MAF overexpression in human myeloma cell lines. Haematologica 2007, 92, 803–811. [Google Scholar] [CrossRef]
  71. Lomas, O.C.; Tahri, S.; Ghobrial, I.M. The microenvironment in myeloma. Curr. Opin. Oncol. 2020, 32, 170–175. [Google Scholar] [CrossRef] [PubMed]
  72. Terpos, E.; Ntanasis-Stathopoulos, I.; Dimopoulos, M.A. Myeloma bone disease: From biology findings to treatment approaches. Blood 2019, 133, 1534–1539. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Corre, J.; Mahtouk, K.; Attal, M.; Gadelorge, M.; Huynh, A.; Fleury-Cappellesso, S.; Danho, C.; Laharrague, P.; Klein, B.; Reme, T.; et al. Bone marrow mesenchymal stem cells are abnormal in multiple myeloma. Leukemia 2007, 21, 1079–1088. [Google Scholar] [CrossRef] [PubMed]
  74. Corre, J.; Labat, E.; Espagnolle, N.; Hebraud, B.; Avet-Loiseau, H.; Roussel, M.; Huynh, A.; Gadelorge, M.; Cordelier, P.; Klein, B.; et al. Bioactivity and prognostic significance of growth differentiation factor GDF15 secreted by bone marrow mesenchymal stem cells in multiple myeloma. Cancer Res. 2012, 72, 1395–1406. [Google Scholar] [CrossRef] [Green Version]
  75. Fernando, R.C.; Mazzotti, D.R.; Azevedo, H.; Sandes, A.F.; Rizzatti, E.G.; de Oliveira, M.B.; Alves, V.L.F.; Eugenio, A.I.P.; de Carvalho, F.; Dalboni, M.A.; et al. Transcriptome Analysis of Mesenchymal Stem Cells from Multiple Myeloma Patients Reveals Downregulation of Genes Involved in Cell Cycle Progression, Immune Response, and Bone Metabolism. Sci. Rep. 2019, 9, 1056. [Google Scholar] [CrossRef] [Green Version]
  76. Alameda, D.; Saez, B.; Lara-Astiaso, D.; Sarvide, S.; Lasa, M.; Alignani, D.; Rodriguez, I.; Garate, S.; Vilas, A.; Paiva, B.; et al. Characterization of freshly isolated bone marrow mesenchymal stromal cells from healthy donors and patients with multiple myeloma: Transcriptional modulation of the microenvironment. Haematologica 2020, 105, e470–e473. [Google Scholar] [CrossRef] [Green Version]
  77. Garcia-Gomez, A.; De Las Rivas, J.; Ocio, E.M.; Díaz-Rodríguez, E.; Montero, J.C.; Martín, M.; Blanco, J.F.; Sanchez-Guijo, F.M.; Pandiella, A.; San Miguel, J.F.; et al. Transcriptomic profile induced in bone marrow mesenchymal stromal cells after interaction with multiple myeloma cells: Implications in myeloma progression and myeloma bone disease. Oncotarget 2014, 5, 8284–8305. [Google Scholar] [CrossRef] [Green Version]
  78. Arnulf, B.; Lecourt, S.; Soulier, J.; Ternaux, B.; Lacassagne, M.N.; Crinquette, A.; Dessoly, J.; Sciaini, A.K.; Benbunan, M.; Chomienne, C.; et al. Phenotypic and functional characterization of bone marrow mesenchymal stem cells derived from patients with multiple myeloma. Leukemia 2007, 21, 158–163. [Google Scholar] [CrossRef]
  79. Andre, T.; Meuleman, N.; Stamatopoulos, B.; De Bruyn, C.; Pieters, K.; Bron, D.; Lagneaux, L. Evidences of early senescence in multiple myeloma bone marrow mesenchymal stromal cells. PLoS ONE 2013, 8, e59756. [Google Scholar] [CrossRef]
  80. Garderet, L.; Mazurier, C.; Chapel, A.; Ernou, I.; Boutin, L.; Holy, X.; Gorin, N.C.; Lopez, M.; Doucet, C.; Lataillade, J.J. Mesenchymal stem cell abnormalities in patients with multiple myeloma. Leuk. Lymphoma 2007, 48, 2032–2041. [Google Scholar] [CrossRef]
  81. Guo, J.; Zhao, Y.; Fei, C.; Zhao, S.; Zheng, Q.; Su, J.; Wu, D.; Li, X.; Chang, C. Dicer1 downregulation by multiple myeloma cells promotes the senescence and tumor-supporting capacity and decreases the differentiation potential of mesenchymal stem cells. Cell Death Dis. 2018, 9, 512. [Google Scholar] [CrossRef] [PubMed]
  82. Kassen, D.; Moore, S.; Percy, L.; Herledan, G.; Bounds, D.; Rodriguez-Justo, M.; Croucher, P.; Yong, K. The bone marrow stromal compartment in multiple myeloma patients retains capability for osteogenic differentiation in vitro: Defining the stromal defect in myeloma. Br. J. Haematol. 2014, 167, 194–206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. 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. (Warsz) 2008, 56, 207–221. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Li, S.; Jiang, Y.; Li, A.; Liu, X.; Xing, X.; Guo, Y.; Xu, Y.; Hao, Y.; Zheng, C. Telomere length is positively associated with the expression of IL6 and MIP1alpha in bone marrow mesenchymal stem cells of multiple myeloma. Mol. Med. Rep. 2017, 16, 2497–2504. [Google Scholar] [CrossRef] [PubMed]
  85. Li, X.; Pennisi, A.; Yaccoby, S. Role of decorin in the antimyeloma effects of osteoblasts. Blood 2008, 112, 159–168. [Google Scholar] [CrossRef] [Green Version]
  86. Todoerti, K.; Lisignoli, G.; Storti, P.; Agnelli, L.; Novara, F.; Manferdini, C.; Codeluppi, K.; Colla, S.; Crugnola, M.; Abeltino, M.; et al. Distinct transcriptional profiles characterize bone microenvironment mesenchymal cells rather than osteoblasts in relationship with multiple myeloma bone disease. Exp. Hematol. 2010, 38, 141–153. [Google Scholar] [CrossRef]
  87. Wallace, S.R.; Oken, M.M.; Lunetta, K.L.; Panoskaltsis-Mortari, A.; Masellis, A.M. Abnormalities of bone marrow mesenchymal cells in multiple myeloma patients. Cancer 2001, 91, 1219–1230. [Google Scholar] [CrossRef]
  88. McNee, G.; Eales, K.L.; Wei, W.; Williams, D.S.; Barkhuizen, A.; Bartlett, D.B.; Essex, S.; Anandram, S.; Filer, A.; Moss, P.A.; et al. Citrullination of histone H3 drives IL-6 production by bone marrow mesenchymal stem cells in MGUS and multiple myeloma. Leukemia 2017, 31, 373–381. [Google Scholar] [CrossRef]
  89. Berenstein, R.; Nogai, A.; Waechter, M.; Blau, O.; Kuehnel, A.; Schmidt-Hieber, M.; Kunitz, A.; Pezzutto, A.; Dörken, B.; Blau, I.W. Multiple myeloma cells modify VEGF/IL-6 levels and osteogenic potential of bone marrow stromal cells via Notch/miR-223. Mol. Carcinog. 2016, 55, 1927–1939. [Google Scholar] [CrossRef]
  90. Kristensen, I.B.; Pedersen, L.; Ro, T.B.; Christensen, J.H.; Lyng, M.B.; Rasmussen, L.M.; Ditzel, H.J.; Borset, M.; Abildgaard, N. Decorin is down-regulated in multiple myeloma and MGUS bone marrow plasma and inhibits HGF-induced myeloma plasma cell viability and migration. Eur. J. Haematol. 2013, 91, 196–200. [Google Scholar] [CrossRef]
  91. Roccaro, A.M.; Sacco, A.; Maiso, P.; Azab, A.K.; Tai, Y.T.; Reagan, M.; Azab, F.; Flores, L.M.; Campigotto, F.; Weller, E.; et al. BM mesenchymal stromal cell-derived exosomes facilitate multiple myeloma progression. J. Clin. Investig. 2013, 123, 1542–1555. [Google Scholar] [CrossRef] [PubMed]
  92. Umezu, T.; Imanishi, S.; Yoshizawa, S.; Kawana, C.; Ohyashiki, J.H.; Ohyashiki, K. Induction of multiple myeloma bone marrow stromal cell apoptosis by inhibiting extracellular vesicle miR-10a secretion. Blood Adv. 2019, 3, 3228–3240. [Google Scholar] [CrossRef] [PubMed]
  93. Abe, M.; Hiura, K.; Ozaki, S.; Kido, S.; Matsumoto, T. Vicious cycle between myeloma cell binding to bone marrow stromal cells via VLA-4-VCAM-1 adhesion and macrophage inflammatory protein-1alpha and MIP-1beta production. J. Bone Miner. Metab. 2009, 27, 16–23. [Google Scholar] [CrossRef] [PubMed]
  94. Kirshner, J.; Thulien, K.J.; Martin, L.D.; Debes Marun, C.; Reiman, T.; Belch, A.R.; Pilarski, L.M. A unique three-dimensional model for evaluating the impact of therapy on multiple myeloma. Blood 2008, 112, 2935–2945. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Kikuchi, J.; Koyama, D.; Wada, T.; Izumi, T.; Hofgaard, P.O.; Bogen, B.; Furukawa, Y. Phosphorylation-mediated EZH2 inactivation promotes drug resistance in multiple myeloma. J. Clin. Investig. 2015, 125, 4375–4390. [Google Scholar] [CrossRef] [PubMed]
  96. Furukawa, Y.; Kikuchi, J. Epigenetic mechanisms of cell adhesion-mediated drug resistance in multiple myeloma. Int. J. Hematol. 2016, 104, 281–292. [Google Scholar] [CrossRef]
  97. Ohguchi, H.; Harada, T.; Sagawa, M.; Kikuchi, S.; Tai, Y.T.; Richardson, P.G.; Hideshima, T.; Anderson, K.C. KDM6B modulates MAPK pathway mediating multiple myeloma cell growth and survival. Leukemia 2017, 31, 2661–2669. [Google Scholar] [CrossRef] [Green Version]
  98. Matsumoto, T.; Abe, M. TGF-β-related mechanisms of bone destruction in multiple myeloma. Bone 2011, 48, 129–134. [Google Scholar] [CrossRef]
  99. Giuliani, N.; Rizzoli, V.; Roodman, G.D. Multiple myeloma bone disease: Pathophysiology of osteoblast inhibition. Blood 2006, 108, 3992–3996. [Google Scholar] [CrossRef] [Green Version]
  100. Mehdi, S.J.; Johnson, S.K.; Epstein, J.; Zangari, M.; Qu, P.; Hoering, A.; van Rhee, F.; Schinke, C.; Thanendrarajan, S.; Barlogie, B.; et al. Mesenchymal stem cells gene signature in high-risk myeloma bone marrow linked to suppression of distinct IGFBP2-expressing small adipocytes. Br. J. Haematol. 2019, 184, 578–593. [Google Scholar] [CrossRef] [Green Version]
  101. Silvestris, F.; Cafforio, P.; Calvani, N.; Dammacco, F. Impaired osteoblastogenesis in myeloma bone disease: Role of upregulated apoptosis by cytokines and malignant plasma cells. Br. J. Haematol. 2004, 126, 475–486. [Google Scholar] [CrossRef] [PubMed]
  102. Giuliani, N.; Colla, S.; Morandi, F.; Lazzaretti, M.; Sala, R.; Bonomini, S.; Grano, M.; Colucci, S.; Svaldi, M.; Rizzoli, V. Myeloma cells block RUNX2/CBFA1 activity in human bone marrow osteoblast progenitors and inhibit osteoblast formation and differentiation. Blood 2005, 106, 2472–2483. [Google Scholar] [CrossRef] [PubMed]
  103. Takeuchi, K.; Abe, M.; Hiasa, M.; Oda, A.; Amou, H.; Kido, S.; Harada, T.; Tanaka, O.; Miki, H.; Nakamura, S.; et al. Tgf-Beta inhibition restores terminal osteoblast differentiation to suppress myeloma growth. PLoS ONE 2010, 5, e9870. [Google Scholar] [CrossRef]
  104. Tsukamoto, S.; Lovendorf, M.B.; Park, J.; Salem, K.Z.; Reagan, M.R.; Manier, S.; Zavidij, O.; Rahmat, M.; Huynh, D.; Takagi, S.; et al. Inhibition of microRNA-138 enhances bone formation in multiple myeloma bone marrow niche. Leukemia 2018, 32, 1739–1750. [Google Scholar] [CrossRef]
  105. Xu, S.; Cecilia Santini, G.; De Veirman, K.; Vande Broek, I.; Leleu, X.; De Becker, A.; Van Camp, B.; Vanderkerken, K.; Van Riet, I. Upregulation of miR-135b is involved in the impaired osteogenic differentiation of mesenchymal stem cells derived from multiple myeloma patients. PLoS ONE 2013, 8, e79752. [Google Scholar] [CrossRef] [Green Version]
  106. Garayoa, M.; Garcia, J.L.; Santamaria, C.; Garcia-Gomez, A.; Blanco, J.F.; Pandiella, A.; Hernández, J.M.; Sanchez-Guijo, F.M.; del Cañizo, M.C.; Gutiérrez, N.C.; et al. Mesenchymal stem cells from multiple myeloma patients display distinct genomic profile as compared with those from normal donors. Leukemia 2009, 23, 1515–1527. [Google Scholar] [CrossRef] [PubMed]
  107. Berenstein, R.; Blau, O.; Nogai, A.; Waechter, M.; Slonova, E.; Schmidt-Hieber, M.; Kunitz, A.; Pezzutto, A.; Doerken, B.; Blau, I.W. Multiple myeloma cells alter the senescence phenotype of bone marrow mesenchymal stromal cells under participation of the DLK1-DIO3 genomic region. BMC Cancer 2015, 15, 68. [Google Scholar] [CrossRef] [Green Version]
  108. Adamik, J.; Roodman, G.D.; Galson, D.L. Epigenetic-Based Mechanisms of Osteoblast Suppression in Multiple Myeloma Bone Disease. JBMR Plus 2019, 3, e10183. [Google Scholar] [CrossRef]
  109. Adamik, J.; Jin, S.; Sun, Q.; Zhang, P.; Weiss, K.R.; Anderson, J.L.; Silbermann, R.; Roodman, G.D.; Galson, D.L. EZH2 or HDAC1 Inhibition Reverses Multiple Myeloma-Induced Epigenetic Suppression of Osteoblast Differentiation. Mol. Cancer Res. 2017, 15, 405–417. [Google Scholar] [CrossRef] [Green Version]
  110. D’Souza, S.; del Prete, D.; Jin, S.; Sun, Q.; Huston, A.J.; Kostov, F.E.; Sammut, B.; Hong, C.S.; Anderson, J.L.; Patrene, K.D.; et al. Gfi1 expressed in bone marrow stromal cells is a novel osteoblast suppressor in patients with multiple myeloma bone disease. Blood 2011, 118, 6871–6880. [Google Scholar] [CrossRef] [Green Version]
  111. Dominici, M.; Le Blanc, K.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.; Krause, D.; Deans, R.; Keating, A.; Prockop, D.; Horwitz, E. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006, 8, 315–317. [Google Scholar] [CrossRef] [PubMed]
  112. Churchman, S.M.; Ponchel, F.; Boxall, S.A.; Cuthbert, R.; Kouroupis, D.; Roshdy, T.; Giannoudis, P.V.; Emery, P.; McGonagle, D.; Jones, E.A. Transcriptional profile of native CD271+ multipotential stromal cells: Evidence for multiple fates, with prominent osteogenic and Wnt pathway signaling activity. Arthritis Rheum. 2012, 64, 2632–2643. [Google Scholar] [CrossRef] [PubMed]
  113. Harichandan, A.; Sivasubramaniyan, K.; Bühring, H.J. Prospective isolation and characterization of human bone marrow-derived MSCs. Adv. Biochem. Eng. Biotechnol. 2013, 129, 1–17. [Google Scholar] [CrossRef] [PubMed]
  114. Mabuchi, Y.; Morikawa, S.; Harada, S.; Niibe, K.; Suzuki, S.; Renault-Mihara, F.; Houlihan, D.D.; Akazawa, C.; Okano, H.; Matsuzaki, Y. LNGFR(+)THY-1(+)VCAM-1(hi+) cells reveal functionally distinct subpopulations in mesenchymal stem cells. Stem Cell Rep. 2013, 1, 152–165. [Google Scholar] [CrossRef] [Green Version]
  115. Tormin, A.; Li, O.; Brune, J.C.; Walsh, S.; Schutz, B.; Ehinger, M.; Ditzel, N.; Kassem, M.; Scheding, S. CD146 expression on primary nonhematopoietic bone marrow stem cells is correlated with in situ localization. Blood 2011, 117, 5067–5077. [Google Scholar] [CrossRef] [Green Version]
  116. Rossi, M.; Botta, C.; Arbitrio, M.; Grembiale, R.D.; Tagliaferri, P.; Tassone, P. Mouse models of multiple myeloma: Technologic platforms and perspectives. Oncotarget 2018, 9, 20119–20133. [Google Scholar] [CrossRef] [Green Version]
  117. Radl, J.; De Glopper, E.D.; Schuit, H.R.; Zurcher, C. Idiopathic paraproteinemia. II. Transplantation of the paraprotein-producing clone from old to young C57BL/KaLwRij mice. J. Immunol. 1979, 122, 609–613. [Google Scholar]
  118. Radl, J.; Croese, J.W.; Zurcher, C.; Van den Enden-Vieveen, M.H.; de Leeuw, A.M. Animal model of human disease. Multiple myeloma. Am. J. Pathol. 1988, 132, 593–597. [Google Scholar]
  119. Fowler, J.A.; Lwin, S.T.; Drake, M.T.; Edwards, J.R.; Kyle, R.A.; Mundy, G.R.; Edwards, C.M. Host-derived adiponectin is tumor-suppressive and a novel therapeutic target for multiple myeloma and the associated bone disease. Blood 2011, 118, 5872–5882. [Google Scholar] [CrossRef]
  120. Fowler, J.A.; Mundy, G.R.; Lwin, S.T.; Edwards, C.M. Bone marrow stromal cells create a permissive microenvironment for myeloma development: A new stromal role for Wnt inhibitor Dkk1. Cancer Res. 2012, 72, 2183–2189. [Google Scholar] [CrossRef] [Green Version]
  121. Gooding, S.; Olechnowicz, S.W.Z.; Morris, E.V.; Armitage, A.E.; Arezes, J.; Frost, J.; Repapi, E.; Edwards, J.R.; Ashley, N.; Waugh, C.; et al. Transcriptomic profiling of the myeloma bone-lining niche reveals BMP signalling inhibition to improve bone disease. Nat. Commun. 2019, 10, 4533. [Google Scholar] [CrossRef] [PubMed]
  122. 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]
  123. 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] [PubMed]
  124. Morito, N.; Yoh, K.; Maeda, A.; Nakano, T.; Fujita, A.; Kusakabe, M.; Hamada, M.; Kudo, T.; Yamagata, K.; Takahashi, S. A novel transgenic mouse model of the human multiple myeloma chromosomal translocation t(14;16)(q32;q23). Cancer Res. 2011, 71, 339–348. [Google Scholar] [CrossRef] [Green Version]
  125. Carrasco, D.R.; Sukhdeo, K.; Protopopova, M.; Sinha, R.; Enos, M.; Carrasco, D.E.; Zheng, M.; Mani, M.; Henderson, J.; Pinkus, G.S.; et al. The differentiation and stress response factor XBP-1 drives multiple myeloma pathogenesis. Cancer Cell 2007, 11, 349–360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Chesi, M.; Robbiani, D.F.; Sebag, M.; Chng, W.J.; Affer, M.; Tiedemann, R.; Valdez, R.; Palmer, S.E.; Haas, S.S.; Stewart, A.K.; et al. AID-dependent activation of a MYC transgene induces multiple myeloma in a conditional mouse model of post-germinal center malignancies. Cancer Cell 2008, 13, 167–180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Calcinotto, A.; Brevi, A.; Chesi, M.; Ferrarese, R.; Garcia Perez, L.; Grioni, M.; Kumar, S.; Garbitt, V.M.; Sharik, M.E.; Henderson, K.J.; et al. Microbiota-driven interleukin-17-producing cells and eosinophils synergize to accelerate multiple myeloma progression. Nat. Commun. 2018, 9, 4832. [Google Scholar] [CrossRef]
  128. Nakamura, K.; Kassem, S.; Cleynen, A.; Chretien, M.L.; Guillerey, C.; Putz, E.M.; Bald, T.; Forster, I.; Vuckovic, S.; Hill, G.R.; et al. Dysregulated IL-18 Is a Key Driver of Immunosuppression and a Possible Therapeutic Target in the Multiple Myeloma Microenvironment. Cancer Cell 2018, 33, 634–648. [Google Scholar] [CrossRef] [Green Version]
  129. Bordini, J.; Bertilaccio, M.T.; Ponzoni, M.; Fermo, I.; Chesi, M.; Bergsagel, P.L.; Camaschella, C.; Campanella, A. Erythroblast apoptosis and microenvironmental iron restriction trigger anemia in the VK*MYC model of multiple myeloma. Haematologica 2015, 100, 834–841. [Google Scholar] [CrossRef] [Green Version]
  130. Alsina, M.; Boyce, B.; Devlin, R.D.; Anderson, J.L.; Craig, F.; Mundy, G.R.; Roodman, G.D. Development of an in vivo model of human multiple myeloma bone disease. Blood 1996, 87, 1495–1501. [Google Scholar] [CrossRef]
  131. Rabin, N.; Kyriakou, C.; Coulton, L.; Gallagher, O.M.; Buckle, C.; Benjamin, R.; Singh, N.; Glassford, J.; Otsuki, T.; Nathwani, A.C.; et al. A new xenograft model of myeloma bone disease demonstrating the efficacy of human mesenchymal stem cells expressing osteoprotegerin by lentiviral gene transfer. Leukemia 2007, 21, 2181–2191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Iriuchishima, H.; Takubo, K.; Miyakawa, Y.; Nakamura-Ishizu, A.; Miyauchi, Y.; Fujita, N.; Miyamoto, K.; Miyamoto, T.; Ikeda, E.; Kizaki, M.; et al. Neovascular niche for human myeloma cells in immunodeficient mouse bone. PLoS ONE 2012, 7, e30557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Chaidos, A.; Barnes, C.P.; Cowan, G.; May, P.C.; Melo, V.; Hatjiharissi, E.; Papaioannou, M.; Harrington, H.; Doolittle, H.; Terpos, E.; et al. Clinical drug resistance linked to interconvertible phenotypic and functional states of tumor-propagating cells in multiple myeloma. Blood 2013, 121, 318–328. [Google Scholar] [CrossRef] [PubMed]
  134. Schueler, J.; Wider, D.; Klingner, K.; Siegers, G.M.; May, A.M.; Wasch, R.; Fiebig, H.H.; Engelhardt, M. Intratibial injection of human multiple myeloma cells in NOD/SCID IL-2Rgamma(null) mice mimics human myeloma and serves as a valuable tool for the development of anticancer strategies. PLoS ONE 2013, 8, e79939. [Google Scholar] [CrossRef] [Green Version]
  135. Hosen, N.; Matsuoka, Y.; Kishida, S.; Nakata, J.; Mizutani, Y.; Hasegawa, K.; Mugitani, A.; Ichihara, H.; Aoyama, Y.; Nishida, S.; et al. CD138-negative clonogenic cells are plasma cells but not B cells in some multiple myeloma patients. Leukemia 2012, 26, 2135–2141. [Google Scholar] [CrossRef] [PubMed]
  136. Tassone, P.; Neri, P.; Carrasco, D.R.; Burger, R.; Goldmacher, V.S.; Fram, R.; Munshi, V.; Shammas, M.A.; Catley, L.; Jacob, G.S.; et al. A clinically relevant SCID-hu in vivo model of human multiple myeloma. Blood 2005, 106, 713–716. [Google Scholar] [CrossRef] [Green Version]
  137. Tsunenari, T.; Koishihara, Y.; Nakamura, A.; Moriya, M.; Ohkawa, H.; Goto, H.; Shimazaki, C.; Nakagawa, M.; Ohsugi, Y.; Kishimoto, T.; et al. New xenograft model of multiple myeloma and efficacy of a humanized antibody against human interleukin-6 receptor. Blood 1997, 90, 2437–2444. [Google Scholar] [CrossRef]
  138. Sartoris, S.; Mazzocco, M.; Tinelli, M.; Martini, M.; Mosna, F.; Lisi, V.; Indraccolo, S.; Moserle, L.; Cestari, T.; Riviera, A.P.; et al. Efficacy assessment of interferon-alpha-engineered mesenchymal stromal cells in a mouse plasmacytoma model. Stem Cells Dev. 2011, 20, 709–719. [Google Scholar] [CrossRef] [Green Version]
  139. Das, R.; Strowig, T.; Verma, R.; Koduru, S.; Hafemann, A.; Hopf, S.; Kocoglu, M.H.; Borsotti, C.; Zhang, L.; Branagan, A.; et al. Microenvironment-dependent growth of preneoplastic and malignant plasma cells in humanized mice. Nat. Med. 2016, 22, 1351–1357. [Google Scholar] [CrossRef] [Green Version]
  140. Lang, J.; Zhang, B.; Kelly, M.; Peterson, J.N.; Barbee, J.; Freed, B.M.; Di Santo, J.P.; Matsuda, J.L.; Torres, R.M.; Pelanda, R. Replacing mouse BAFF with human BAFF does not improve B-cell maturation in hematopoietic humanized mice. Blood Adv. 2017, 1, 2729–2741. [Google Scholar] [CrossRef] [Green Version]
  141. Yaccoby, S.; Barlogie, B.; Epstein, J. Primary myeloma cells growing in SCID-hu mice: A model for studying the biology and treatment of myeloma and its manifestations. Blood 1998, 92, 2908–2913. [Google Scholar] [CrossRef] [PubMed]
  142. Urashima, M.; Chen, B.P.; Chen, S.; Pinkus, G.S.; Bronson, R.T.; Dedera, D.A.; Hoshi, Y.; Teoh, G.; Ogata, A.; Treon, S.P.; et al. The development of a model for the homing of multiple myeloma cells to human bone marrow. Blood 1997, 90, 754–765. [Google Scholar] [CrossRef] [PubMed]
  143. Pennisi, A.; Ling, W.; Li, X.; Khan, S.; Shaughnessy, J.D., Jr.; Barlogie, B.; Yaccoby, S. The ephrinB2/EphB4 axis is dysregulated in osteoprogenitors from myeloma patients and its activation affects myeloma bone disease and tumor growth. Blood 2009, 114, 1803–1812. [Google Scholar] [CrossRef]
  144. Yata, K.; Yaccoby, S. The SCID-rab model: A novel in vivo system for primary human myeloma demonstrating growth of CD138-expressing malignant cells. Leukemia 2004, 18, 1891–1897. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Groen, R.W.; Noort, W.A.; Raymakers, R.A.; Prins, H.J.; Aalders, L.; Hofhuis, F.M.; Moerer, P.; van Velzen, J.F.; Bloem, A.C.; van Kessel, B.; et al. Reconstructing the human hematopoietic niche in immunodeficient mice: Opportunities for studying primary multiple myeloma. Blood 2012, 120, e9–e16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Calimeri, T.; Battista, E.; Conforti, F.; Neri, P.; Di Martino, M.T.; Rossi, M.; Foresta, U.; Piro, E.; Ferrara, F.; Amorosi, A.; et al. A unique three-dimensional SCID-polymeric scaffold (SCID-synth-hu) model for in vivo expansion of human primary multiple myeloma cells. Leukemia 2011, 25, 707–711. [Google Scholar] [CrossRef] [PubMed]
  147. Reinisch, A.; Hernandez, D.C.; Schallmoser, K.; Majeti, R. Generation and use of a humanized bone-marrow-ossicle niche for hematopoietic xenotransplantation into mice. Nat. Protoc. 2017, 12, 2169–2188. [Google Scholar] [CrossRef] [PubMed]
  148. San-Miguel, J.; Blade, J.; Shpilberg, O.; Grosicki, S.; Maloisel, F.; Min, C.K.; Polo Zarzuela, M.; Robak, T.; Prasad, S.V.; Tee Goh, Y.; et al. Phase 2 randomized study of bortezomib-melphalan-prednisone with or without siltuximab (anti-IL-6) in multiple myeloma. Blood 2014, 123, 4136–4142. [Google Scholar] [CrossRef] [Green Version]
  149. Brighton, T.A.; Khot, A.; Harrison, S.J.; Ghez, D.; Weiss, B.M.; Kirsch, A.; Magen, H.; Gironella, M.; Oriol, A.; Streetly, M.; et al. Randomized, Double-Blind, Placebo-Controlled, Multicenter Study of Siltuximab in High-Risk Smoldering Multiple Myeloma. Clin. Cancer Res. 2019, 25, 3772–3775. [Google Scholar] [CrossRef] [Green Version]
  150. Orlowski, R.Z.; Gercheva, L.; Williams, C.; Sutherland, H.; Robak, T.; Masszi, T.; Goranova-Marinova, V.; Dimopoulos, M.A.; Cavenagh, J.D.; Spicka, I.; et al. A phase 2, randomized, double-blind, placebo-controlled study of siltuximab (anti-IL-6 mAb) and bortezomib versus bortezomib alone in patients with relapsed or refractory multiple myeloma. Am. J. Hematol. 2015, 90, 42–49. [Google Scholar] [CrossRef] [Green Version]
  151. Lam, C.; Ferguson, I.D.; Mariano, M.C.; Lin, Y.T.; Murnane, M.; Liu, H.; Smith, G.A.; Wong, S.W.; Taunton, J.; Liu, J.O.; et al. Repurposing tofacitinib as an anti-myeloma therapeutic to reverse growth-promoting effects of the bone marrow microenvironment. Haematologica 2018, 103, 1218–1228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. de Oliveira, M.B.; Fook-Alves, V.L.; Eugenio, A.I.P.; Fernando, R.C.; Sanson, L.F.G.; de Carvalho, M.F.; Braga, W.M.T.; Davies, F.E.; Colleoni, G.W.B. Anti-myeloma effects of ruxolitinib combined with bortezomib and lenalidomide: A rationale for JAK/STAT pathway inhibition in myeloma patients. Cancer Lett. 2017, 403, 206–215. [Google Scholar] [CrossRef] [PubMed]
  153. Iyer, S.P.; Beck, J.T.; Stewart, A.K.; Shah, J.; Kelly, K.R.; Isaacs, R.; Bilic, S.; Sen, S.; Munshi, N.C. A Phase IB multicentre dose-determination study of BHQ880 in combination with anti-myeloma therapy and zoledronic acid in patients with relapsed or refractory multiple myeloma and prior skeletal-related events. Br. J. Haematol. 2014, 167, 366–375. [Google Scholar] [CrossRef] [PubMed]
  154. Abdulkadyrov, K.M.; Salogub, G.N.; Khuazheva, N.K.; Sherman, M.L.; Laadem, A.; Barger, R.; Knight, R.; Srinivasan, S.; Terpos, E. Sotatercept in patients with osteolytic lesions of multiple myeloma. Br. J. Haematol. 2014, 165, 814–823. [Google Scholar] [CrossRef] [PubMed]
  155. Yan, X.; Ehnert, S.; Culmes, M.; Bachmann, A.; Seeliger, C.; Schyschka, L.; Wang, Z.; Rahmanian-Schwarz, A.; Stockle, U.; De Sousa, P.A.; et al. 5-azacytidine improves the osteogenic differentiation potential of aged human adipose-derived mesenchymal stem cells by DNA demethylation. PLoS ONE 2014, 9, e90846. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Schroeder, T.M.; Westendorf, J.J. Histone deacetylase inhibitors promote osteoblast maturation. J. Bone Miner. Res. 2005, 20, 2254–2263. [Google Scholar] [CrossRef]
  157. Cho, H.H.; Park, H.T.; Kim, Y.J.; Bae, Y.C.; Suh, K.T.; Jung, J.S. Induction of osteogenic differentiation of human mesenchymal stem cells by histone deacetylase inhibitors. J. Cell Biochem. 2005, 96, 533–542. [Google Scholar] [CrossRef]
  158. McGee-Lawrence, M.E.; McCleary-Wheeler, A.L.; Secreto, F.J.; Razidlo, D.F.; Zhang, M.; Stensgard, B.A.; Li, X.; Stein, G.S.; Lian, J.B.; Westendorf, J.J. Suberoylanilide hydroxamic acid (SAHA; vorinostat) causes bone loss by inhibiting immature osteoblasts. Bone 2011, 48, 1117–1126. [Google Scholar] [CrossRef] [Green Version]
  159. Pratap, J.; Akech, J.; Wixted, J.J.; Szabo, G.; Hussain, S.; McGee-Lawrence, M.E.; Li, X.; Bedard, K.; Dhillon, R.J.; van Wijnen, A.J.; et al. The histone deacetylase inhibitor, vorinostat, reduces tumor growth at the metastatic bone site and associated osteolysis, but promotes normal bone loss. Mol. Cancer Ther. 2010, 9, 3210–3220. [Google Scholar] [CrossRef] [Green Version]
  160. Xu, S.; De Veirman, K.; Evans, H.; Santini, G.C.; Vande Broek, I.; Leleu, X.; De Becker, A.; Van Camp, B.; Croucher, P.; Vanderkerken, K.; et al. Effect of the HDAC inhibitor vorinostat on the osteogenic differentiation of mesenchymal stem cells in vitro and bone formation in vivo. Acta Pharmacol. Sin. 2013, 34, 699–709. [Google Scholar] [CrossRef] [Green Version]
  161. Giuliani, N.; Morandi, F.; Tagliaferri, S.; Lazzaretti, M.; Bonomini, S.; Crugnola, M.; Mancini, C.; Martella, E.; Ferrari, L.; Tabilio, A.; et al. The proteasome inhibitor bortezomib affects osteoblast differentiation in vitro and in vivo in multiple myeloma patients. Blood 2007, 110, 334–338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Podar, K.; Zimmerhackl, A.; Fulciniti, M.; Tonon, G.; Hainz, U.; Tai, Y.T.; Vallet, S.; Halama, N.; Jager, D.; Olson, D.L.; et al. The selective adhesion molecule inhibitor Natalizumab decreases multiple myeloma cell growth in the bone marrow microenvironment: Therapeutic implications. Br. J. Haematol. 2011, 155, 438–448. [Google Scholar] [CrossRef] [PubMed]
  163. 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] [PubMed] [Green Version]
  164. 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] [PubMed]
  165. Ghobrial, I.M.; Liu, C.J.; Redd, R.A.; Perez, R.P.; Baz, R.; Zavidij, O.; Sklavenitis-Pistofidis, R.; Richardson, P.G.; Anderson, K.C.; Laubach, J.; et al. A Phase Ib/II Trial of the First-in-Class Anti-CXCR4 Antibody Ulocuplumab in Combination with Lenalidomide or Bortezomib Plus Dexamethasone in Relapsed Multiple Myeloma. Clin. Cancer Res. 2020, 26, 344–353. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Fulciniti, M.; Tassone, P.; Hideshima, T.; Vallet, S.; Nanjappa, P.; Ettenberg, S.A.; Shen, Z.; Patel, N.; Tai, Y.T.; Chauhan, D.; et al. Anti-DKK1 mAb (BHQ880) as a potential therapeutic agent for multiple myeloma. Blood 2009, 114, 371–379. [Google Scholar] [CrossRef] [Green Version]
  167. Long, M.; Beckwith, K.; Do, P.; Mundy, B.L.; Gordon, A.; Lehman, A.M.; Maddocks, K.J.; Cheney, C.; Jones, J.A.; Flynn, J.M.; et al. Ibrutinib treatment improves T cell number and function in CLL patients. J. Clin. Investig. 2017, 127, 3052–3064. [Google Scholar] [CrossRef]
  168. Gauthier, J.; Hirayama, A.V.; Purushe, J.; Hay, K.A.; Lymp, J.; Li, D.H.; Yeung, C.C.S.; Sheih, A.; Pender, B.S.; Hawkins, R.M.; et al. Feasibility and efficacy of CD19-targeted CAR T cells with concurrent ibrutinib for CLL after ibrutinib failure. Blood 2020, 135, 1650–1660. [Google Scholar] [CrossRef]
  169. Regmi, S.; Pathak, S.; Kim, J.O.; Yong, C.S.; Jeong, J.H. Mesenchymal stem cell therapy for the treatment of inflammatory diseases: Challenges, opportunities, and future perspectives. Eur. J. Cell Biol. 2019, 98, 151041. [Google Scholar] [CrossRef]
  170. Walker, B.A.; Wardell, C.P.; Johnson, D.C.; Kaiser, M.F.; Begum, D.B.; Dahir, N.B.; Ross, F.M.; Davies, F.E.; Gonzalez, D.; Morgan, G.J. Characterization of IGH locus breakpoints in multiple myeloma indicates a subset of translocations appear to occur in pregerminal center B cells. Blood 2013, 121, 3413–3419. [Google Scholar] [CrossRef]
  171. Boucher, K.; Parquet, N.; Widen, R.; Shain, K.; Baz, R.; Alsina, M.; Koomen, J.; Anasetti, C.; Dalton, W.; Perez, L.E. Stemness of B-cell progenitors in multiple myeloma bone marrow. Clin. Cancer Res. 2012, 18, 6155–6168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Al-Sabah, J.; Baccin, C.; Haas, S. Single-cell and spatial transcriptomics approaches of the bone marrow microenvironment. Curr. Opin. Oncol. 2020, 32, 146–153. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Bone marrow microenvironment of multiple myeloma. In bone marrow, myeloma cells and the surrounding cells interact with each other in direct or indirect ways, inducing a favorable environment for the survival of multiple myeloma. MM, multiple myeloma; MSC, mesenchymal stromal cell; Endothel, endothelial cell; Mono/Mac, monocyte or macrophage; NK, natural killer cell; Neu, neutrophil; Meg, Megakaryocyte; DC, dendritic cell; ●, soluble factors.
Figure 1. Bone marrow microenvironment of multiple myeloma. In bone marrow, myeloma cells and the surrounding cells interact with each other in direct or indirect ways, inducing a favorable environment for the survival of multiple myeloma. MM, multiple myeloma; MSC, mesenchymal stromal cell; Endothel, endothelial cell; Mono/Mac, monocyte or macrophage; NK, natural killer cell; Neu, neutrophil; Meg, Megakaryocyte; DC, dendritic cell; ●, soluble factors.
Cancers 13 00025 g001
Table 1. Interaction between multiple myeloma cells and mesenchymal stromal cells.
Table 1. Interaction between multiple myeloma cells and mesenchymal stromal cells.
Cell TypeEffectsEffectsEffectsRegulated FactorsReferences
MM cellsEffects on MSCsSoluble factor secretionInhibition of osteoblastogenesisWnt inhibitors (DKK1, sFRPs, sclerostin), TNFα[49,51,52,53,54,55]
IL-6 production in MSCsbFGF1, TNFα[49,56]
Localization with MSCsProliferation and drug resistance in MM cellsCCL25, CXCR4
Integrin α4, α5, β1, β7, ALCAM, VCAM-1
[32,48,57,58,59,60,61,62,63,64,65,66,67,68]
VEGF production in MSCsIntegrin β7[69]
MSCsEffects on MM cellsSoluble factor secretionMM cell proliferationIL-6, GDF15, BAFF, micro vesicles (miRNA-15a,-10a)[73,74,83,84,85,86,87,88,89,90,91]
Localization with MM cellsProliferation and drug resistance in MM cellsCXCL12, CCL3, ICAM-1, VCAM-1, MADCAM-1[63,83,84,85,86,87,88,89,90,91,92]
MSCsEffects on MM microenvironment Inhibition of osteoblastogenesisDKK1, TNFα, IL-6, TGFβ[101,102,103,104]
immunomodulationIL-6, IL-10, IL-1β, IL-3, TNFα,[75,76,77,78,79,83,84,85]
angiogenesisVEGF, HGF[69,89]
MM, multiple myeloma; MSC, mesenchymal stromal cell; DKK1, Dickkopf1; sFRP, secreted frizzled related protein; TNF, tumor necrosis factor; CCL, CC chemokine ligand; CXCR, CXC chemokine receptor; CAM, cell adhesion molecule; ALCAM, activated leukocyte cell adhesion molecule; VCAM, vascular cell adhesion molecule; VEGF, vascular endothelial growth factor; GDF, growth differentiation factor;BAFF, B-cell activating factor; ICAM, intercellular adhesion molecule; MADCAM, mucosal addressin cell adhesion molecule; TGF, transforming growth factor; HGF, hepatocyte growth factor.
Table 2. Agents for targeting the effects of MM-MSCs.
Table 2. Agents for targeting the effects of MM-MSCs.
TargetAgentAgentAgentEfficacies
MM proliferationIL-6 chimeric antibody [148,149,150]SiltuximabClinical trialsNot clear
JAK signal inhibitor [151,152]Tofacitinib, ruxolitinibPreclinical studiesYes
Microvesicle release inhibitor [92]FTY720Preclinical studiesYes
Osteoblastogenesis suppressionDKK1 inhibitor [153]BHQ880Clinical trialsNo
TGFβ antagonist [154]Sotatercept Clinical trialsNo
DNMT inhibitor [155]5-AzacytidinePreclinical studiesNot clear
HDAC inhibitor [156,157,158,159,160]VorinostatPreclinical studiesNot clear
Proteasome inhibitor [161]BortezomibClinical studiesYes
Adhesion to MM microenvironmentIntegrin α4 inhibition [162]NatalizumabPreclinical studiesYes
CXCL12-CXCR4 inhibition [162,163,164,165]Plerixafor, olaptesed pegol, ulocuplumabClinical trialsOngoing
MM, multiple myeloma; MSC, mesenchymal stromal cell; DKK1, Dickkopf1, TGF, transforming growth factor; DNMT, DNA methyltransferase; HDAC, histone deacetylase; CXCL, CXC chemokine ligand; CXCR, CXC chemokine receptor.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ichii, M.; Hosen, N. Current Understanding of Myelomatous Mesenchymal Stromal Cells Extended through Advances in Experimental Methods. Cancers 2021, 13, 25. https://doi.org/10.3390/cancers13010025

AMA Style

Ichii M, Hosen N. Current Understanding of Myelomatous Mesenchymal Stromal Cells Extended through Advances in Experimental Methods. Cancers. 2021; 13(1):25. https://doi.org/10.3390/cancers13010025

Chicago/Turabian Style

Ichii, Michiko, and Naoki Hosen. 2021. "Current Understanding of Myelomatous Mesenchymal Stromal Cells Extended through Advances in Experimental Methods" Cancers 13, no. 1: 25. https://doi.org/10.3390/cancers13010025

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