Skip to main content
Wiley Open Access Collection logoLink to Wiley Open Access Collection
. 2019 Dec 18;250(5):555–572. doi: 10.1002/path.5357

Mesenchymal stromal cells in cancer: a review of their immunomodulatory functions and dual effects on tumor progression

Sabine Galland 1, Ivan Stamenkovic 1,
PMCID: PMC7217065  PMID: 31608444

Abstract

Mesenchymal stem or stromal cells (MSCs) are pluripotent cells implicated in a broad range of physiological events, including organogenesis and maintenance of tissue homeostasis as well as tissue regeneration and repair. Because their current definition is somewhat loose – based primarily on their ability to differentiate into a variety of mesenchymal tissues, adhere to plastic, and express, or lack, a handful of cell surface markers – MSCs likely encompass several subpopulations, which may have diverse properties. Their diversity may explain, at least in part, the pleiotropic functions that they display in different physiological and pathological settings. In the context of tissue injury, MSCs can respectively promote and attenuate inflammation during the early and late phases of tissue repair. They may thereby act as sensors of the inflammatory response and secrete mediators that boost or temper the response as required by the stage of the reparatory and regenerative process. MSCs are also implicated in regulating tumor development, in which they are increasingly recognized to play a complex role. Thus, MSCs can both promote and constrain tumor progression by directly affecting tumor cells via secreted mediators and cell–cell interactions and by modulating the innate and adaptive immune response. This review summarizes our current understanding of MSC involvement in tumor development and highlights the mechanistic underpinnings of their implication in tumor growth and progression. © 2020 Authors. Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.

Keywords: mesenchymal stem/stromal cells, immune system, inflammation, cancer, Toll‐like receptors, exosomes, anti‐tumor therapy, interleukin‐6

Introduction

Few cells have attracted as much interest in the past 20 years as have mesenchymal stem or stromal cells (MSCs). The fascination with MSCs is due in part to their implication in a wide range of physiological and pathological processes, including development, tissue repair, organ transplantation, autoimmunity, and cancer, and in part to their elusive identity. There have been several excellent recent reviews on MSCs 1, 2, 3, 4, 5, 6, 7, 8, 9 and rather than discuss all of their known biological and functional properties, we will focus on their role in cancer, particularly their immunomodulatory ability.

Current experimental models suggest that MSCs may both promote and constrain tumor growth, although their net effect appears to be predominantly pro‐tumorigenic (Table 1, with references). Tumor growth, which triggers and maintains chronic inflammation, tissue remodeling, and dampened immunity, has been heralded as ‘a wound that never heals’ 44, in which MSCs actively participate. The immunomodulatory effects of MSCs on innate and adaptive immunity through secreted factors, exosomes, and cell–cell contacts constitute a major mechanism by which MSCs affect tumor initiation and progression. As MSCs may exert opposing effects on immune cells, promoting inflammation on the one hand and exhibiting immunosuppressive features, which favor tumor progression, on the other, harnessing their plasticity toward the expression of anti‐tumorigenic, anti‐inflammatory, and pro‐immunogenic properties may provide an attractive therapeutic option. However, such an endeavor requires in‐depth understanding of the functional relationship between tumor cells, MSCs, and immune cells – particularly how tumor cells subvert MSCs to function in their favor and the underpinnings of MSC plasticity that allow such subversion to occur.

Table 1.

Pro‐ and anti‐tumor effects of MSCs in the TME

References Origin of MSCs Species Tumor relevance Tumor function Mechanisms
10 Bone marrow Human Breast cancer Promoting CCL5 (RANTES)
11 Bone marrow Human Breast cancer Promoting p‐EGFR
12 Bone marrow Human Prostate cancer Promoting TGF‐β
13 Adipose tissue Human Prostate cancer Promoting TGF‐β and periostin
14 Umbilical cord and adipose tissue Human Breast cancer Suppressive Apoptosis induction (PARP cleavage)
15 Bone marrow Mouse Hepatoma Suppressive Apoptosis induction
16 Bone marrow Rat and mouse Melanoma Suppressive when administered at a 3:1 ratio with ECs Cytotoxic for endothelial cells (ROS) and anti‐angiogenic effects
17 Bone marrow Human Ovarian cancer (‘SKOV‐3’ cell line) Promoting IL‐6; transition to CAF
18 Umbilical cord blood (UCB) and adipose tissue (AT) Human Glioblastoma multiforme

UCB‐MSCs: suppressive;

AT‐MSCs: promoting

UCB‐MSCs: TRAIL (apoptosis)

AT‐MSCs:

VEGF, ANG1, PDGF, IGF, SDF‐1/CXCL12

19 Tumor tissue, bone marrow Human Gastric cancer Promoting IL‐8
20 Tumor tissue, bone marrow Human Glioma Promoting IL‐6/STAT3
21 Bone marrow Human Breast cancer ‘MDA‐MB‐231’ cells Promoting CAF differentiation
22 Bone marrow Human Breast cancer ‘MDA‐MB‐231’ cells Promoting TGF‐β/Smad pathway; TGF‐β‐dependent transition to CAF
23 Bone marrow Human Kaposi's sarcoma Suppressive MSCs target Akt activity within tumor cells
24 Dermal tissues of a human fetus Human Hepatoma Suppressive Wnt signaling pathway
25 Tumor tissue Human Gastric cancer Promoting SDF‐1 and VEGF
26 Tumor tissue Human Head and neck cancers Promoting IL‐6, IL‐8, SDF‐1α, and expression of CD54
27 Bone marrow Human Inflammatory breast cancer Promoting IL‐6
28 Bone marrow Human Pancreatic cancer Suppressive Unknown
29 Bone marrow Human Glioma Suppressive Downregulation of PDGF/PDGFR axis
30 Tumor tissue Human Ovarian cancer Promoting Promotion of Akt and XIAP phosphorylation
31 Tumor tissue Human Colon cancer Promoting IL‐6/Notch‐1/CD44 signaling axis
32 Bone marrow Mouse Melanoma Promoting Immunosuppression after priming by IFN‐γ and TNF‐α
33 Tumor tissue Human Hepatocellular carcinoma Promoting Trophic factor secretion
34 Bone marrow Human and rat Colon cancer Suppressive Immunomodulation and decrease in inflammation; increase of miR‐150 and miR‐7
35 Bone marrow Human Gastric cancer Promoting Platelet activation: TGF‐β
36 Bone marrow Mouse Breast cancer Promoting Increased stiffness (prosaposin) of the ECM induces differentiation of MSCs to CAFs, enhanced proliferation, and survival of tumor cells
37 Adipose tissue Human, rat Gastric cancer Promoting MAPK activation, decrease apoptosis
38 Adipose tissue Human Leukemia Suppressive DKK‐1‐mediated inhibition
39 Bone marrow Human Colorectal cancer Promoting CCR5
40 Tumor tissue Human Colorectal cancer Promoting Tumor cells escape from senescence via P53/P21 pathway
41 Adipose tissue Human Lung cancer Promoting IL‐6/STAT3
42 Bone marrow Human, mouse Breast cancer Promoting Chemoresistance via a CD9‐dependent mechanism
43 Bone marrow, tumor tissue Human, mouse Prostate cancer Promoting Asporin (ASPN) secreted by MSCs drives metastasis

Within the tumor, MSCs can exert both stimulatory and inhibitory effects on cancer cell growth, invasion, and metastasis through direct or indirect interactions with tumor cells. However, despite the seemingly opposing potential effects of MSCs on tumor growth, their net effect seems to be predominantly pro‐tumorigenic. This reflects the imbalance between pro‐ and anti‐tumorigenic activities that may vary depending on tumor type (and regionally within the tumors), the ecology of the host milieu, the stage of the evolution of a particular tumor, and possibly the composition of the MSC population itself. The predominantly pro‐tumorigenic effect of MSCs in vivo and the opposing effects reported may be due to differences in experimental design, models used, and MSC heterogeneity that may reflect variable responses to a given set of stimuli.

For a complete list of abbreviations see supplementary material, Table S1.

MSCs: heterogeneous cells in search of better definition

Precise definition of stromal cell populations is still lacking. Unlike hematopoietic cell subpopulations, whose identity, developmental stage, and plasticity can be predicted based on a combination of cell surface marker and transcription factor expression 45, 46, 47, stromal cells lack comparable functional and differentiation state markers. As a result, stromal cell populations are defined based on relatively loose phenotypic and functional criteria, which may be common to cells with distinct identities. Fibroblasts illustrate this notion well. Although a few cell surface receptors, including FAP (fibroblast activation protein α) and FSP (fibroblast surface protein), are commonly used to identify fibroblasts 48, 49, 50, their expression allows only approximate categorization of a subset of stromal cells. Moreover, fibroblasts are primarily defined based on their functional properties upon activation, during which they express alpha smooth muscle actin (α‐SMA) and secrete a wide range of extracellular matrix (ECM) components. These secretory products are more or less comparable in the context of wound healing (where the cells are labeled myofibroblasts) 51, 52 and cancer growth [where they are commonly referred to as cancer‐associated fibroblasts (CAFs)] 49, 50. Resting fibroblasts, which are identified largely based on morphology, remain poorly defined in terms of biological properties. Arguments have been put forth that they are multipotent cells, capable of differentiating into a spectrum of mesenchymal tissues 49, which is akin to tissue MSCs. However, adult skin fibroblasts tend not to differentiate into various mesenchymal tissues in culture and neither their origin nor their potential heterogeneity has been clearly elucidated 49, 53. Similar issues face the definition of MSCs (Figure 1).

Figure 1.

PATH-5357-FIG-0001-c

MSC definition and differentiation and comparison with fibroblasts. MSCs have been suggested to be a probable source of fibroblasts, implying that fibroblasts are one type of mesenchymal cell into which MSCs differentiate. However, as MSCs and fibroblasts share numerous functional features, it is possible that maturation or aging (although not in the sense of cell senescence) rather differentiation distinguish the two cell types. Fibroblasts may thus be a more ‘mature’ form of MSCs that have lost pluripotency and altered part of their cell surface receptor repertoire but that can respond to environmental stimuli such as injury and tumor growth in a manner akin to that of MSCs, many of whose properties they retain. MSC (left) and fibroblast (right) activation are illustrated under reversible, wound healing‐associated, and chronic tumor‐related inflammation. Some of the markers associated with each cell type in the context of wound healing and the tumor microenvironment are highlighted. (1) MSCs are a diverse and heterogeneous subset of multipotent precursors present in the stromal fraction of many adult tissues, especially bone marrow but also adipose tissue, synovial membranes, tooth pulp, and the connective tissue of most organs. Several studies show that MSCs lie adjacent to blood vessels and are localized in almost every perivascular space of the body. MSCs are the common predecessors of cells of the mesenchymal lineage, such as bone, cartilage, and fat cells. They can also differentiate into cells from unrelated germline lineages (endodermic and neuroectodermic differentiation potential), a process known as transdifferentiation. Quiescent or resting MSCs are spindle‐shaped cells (fibroblast‐like cells), but contrary to fibroblasts, which can be identified primarily based on morphology, MSCs are more heterogeneous. (2) In response to tissue injury and the associated stimuli, quiescent MSCs are activated to facilitate repair and regeneration. These MSCs may be tissue‐resident or recruited from the bone marrow or adjacent tissues and adopt a stellate morphology. The acquired synthetic properties are associated with secretory and migratory functions that amplify their activation, recruitment, and proliferation. Such activation may be reversed by reprogramming; alternatively, activated MSCs may undergo apoptosis upon completion of the repair process. (3) Chronic inflammation and/or the presence of a tumor induces prolonged activation of MSCs (tumor‐associated MSCs, T‐MSCs), which may gain further secretory properties (e.g. high secretion of IL‐6), specialized ECM remodeling ability, and robust autocrine activation and dynamic immunomodulatory signaling functions. Epigenetic regulation may help to maintain such activated states. T‐MSCs gain enhanced proliferative properties and their functional diversity adds to the dynamic complexity of the evolving tumor microenvironment. For a complete list of abbreviations see supplementary material, Table S1.

Mesenchymal stem or stromal cells are multipotent stromal cells, believed to be an important constituent of the connective tissue that forms the supportive structure in which functional cells of tissues reside. In 2006, the International Society for Cellular Therapy (ISCT) published a position paper to define MSCs and recommended renaming the cells ‘multipotent mesenchymal stromal cells’ 54, 55. However, the most commonly used terms are ‘mesenchymal stromal cells’ and ‘mesenchymal stem cells’.

Although they were initially described in the bone marrow (BM), MSCs display a broad tissue distribution, including adipose, synovial, and lung tissue as well as umbilical cord and peripheral blood 56. Their defining properties include adhesiveness, a cell surface receptor repertoire, and plasticity. Adhesiveness is determined by MSC attachment to plastic under standard culture conditions; the cell surface phenotype is defined by expression of CD105, CD73, and CD90, and lack of expression of CD45, CD34, CD14 or CD11b, CD79α, CD19, and HLA‐DR. This includes low expression levels of MHC class I molecules, and minimal or no expression of MHC class II molecules or co‐stimulatory receptors, including CD40, CD80, and CD86, precluding antigen‐presenting activity 57. Plasticity is measured by the ability of the cells to differentiate into osteocytes, adipocytes, and chondrocytes in vitro in response to appropriate growth factors 54. However, MSCs have the capacity to differentiate into both mesodermal and non‐mesodermal tissues, such that in addition to osteocytes, adipocytes, and chondrocytes, they can differentiate toward endodermal and neuroectodermal lineages (multi‐lineage plasticity 56). Furthermore, a population of MSCs that displays homogeneous expression of CD105, CD90, and CD73 may display heterogeneous differentiation properties. Exposure to differentiation factors may result in only a fraction of the cells differentiating into adipocytes, chondrocytes or osteocytes, suggesting functional heterogeneity despite common cell surface marker expression. Whether such functional heterogeneity reflects differences in adaptation to in vitro culture or the outgrowth of stromal cell subpopulations from progenitor cells bearing distinct identities remains to be resolved. It must also be noted that the above criteria have been defined using bone marrow‐derived MSCs but that there are substantial phenotypic and functional differences among MSCs from different tissues 58.

Refinement of MSC definition along with the identification of their putative subpopulations requires additional phenotypic and/or functional criteria. Candidate markers that are associated with MSCs from various tissues include Stro‐1 (BM‐MSCs) 59, CD271/NGFR (nerve growth factor receptor) 60, CD200 61, CD106/VCAM‐1 (vascular cell adhesion molecule 1) 62, CD146/MCAM‐1 (melanoma cell adhesion molecule 1) 63, 64, MSCA‐1/TNAP (mesenchymal stromal cell antigen 1/tissue‐nonspecific alkaline phosphatase) 65, and SSEA‐4 (stage‐specific embryonic antigen 4) 66. The reliability and limitations of the most commonly used markers have recently been reviewed 67. Several newly identified promising candidate markers include Meflin 68, PDPN (podoplanin) 69, and gremlin‐1 70, 71, 72.

Physiological role: tissue regeneration and wound repair

MSCs are widely believed to play a central role in tissue repair. Injury‐initiated inflammation, whose effectors include innate immune cells and their mediators, and the ensuing tissue remodeling provide signals that mobilize MSCs and drive their differentiation toward diverse stromal components, some of which replace damaged cells. Mesenchymal stromal cells may be injured‐tissue‐resident or recruited from the bone marrow. However, the mechanisms by which they are mobilized and recruited to damaged sites are not fully understood. One study, using a murine model of acute renal tubular necrosis, suggested that bone marrow‐derived MSC recruitment to sites of injury relies on the interaction between CD44 expressed on the MSC surface and hyaluronic acid produced by a variety of cells in areas of tissue remodeling 73. However, additional factors are likely implicated and the mechanisms that promote MSC survival and differentiation toward distinct cell types in vivo are still unclear 74.

The secretome and proteome of MSCs reflect their pleiotropic functions and plasticity 75. In response to soluble mediators derived from the microenvironment of injured tissues, including tumor necrosis factoralpha (TNF‐α), interleukin‐1 (IL‐1), interferon‐gamma (IFN‐γ), and toxins from infectious agents, MSCs can release a wide repertoire of soluble mediators that includes epidermal growth factor (EGF), fibroblast growth factor (FGF), platelet‐derived growth factor (PDGF), transforming growth factor‐beta (TGF‐β), vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF), insulin‐like growth factor‐1 (IGF‐1), angiopoietin‐1 (Ang‐1), keratinocyte growth factor (KGF), TNF‐stimulated G6 protein (TSG‐6), interleukin‐1 receptor antagonist (IL‐1RA), prostaglandin E2 (PGE2), indoleamine 2,3 dioxygenase (IDO), nitric oxide (NO), and stromal‐derived factor 1 (SDF‐1). These mediators promote the activation of fibroblasts, endothelial cells, and tissue progenitor cells, leading to angiogenesis, inhibition of apoptosis, ECM deposition, and damaged cell replacement 6, 76, 77, 78, 79, which in turn ensure tissue regeneration and repair 80, 81, 82. In addition to helping orchestrate regeneration and repair, MSCs can actively participate in bactericidal activity (through LL‐37) 83.

Several in vivo studies have shown the beneficial effect of allogeneic or xenogenic MSCs in a variety of disorders that require tissue regeneration and repair, including acute graft‐versus‐host disease, sepsis, acute asthma, acute renal failure, multiple sclerosis, and myocardial infarction 76, 84, 85, 86. Currently, more than 785 studies are underway or have been submitted to http://clinicaltrials.gov (https://clinicaltrials.gov/) under the terms ‘mesenchymal stem cells’ or ‘mesenchymal stromal cells’.

MSCs and cancer cell crosstalk

Accumulating evidence suggests that MSCs have the ability to migrate toward tumor sites 87 and MSC mobilization has been observed in response to a wide range of solid cancer‐derived cell types. Within the tumor microenvironment (TME), MSCs can exert both stimulatory and inhibitory effects on cancer cell growth, invasion, and metastasis through direct or indirect interactions with tumor cells (Table 1 and Figure 2). However, their net effect seems to be predominantly pro‐tumorigenic, which may reflect an imbalance between pro‐ and anti‐tumorigenic activity dictated by the tumor type, intratumoral heterogeneity, the ecology of the host milieu, and possibly the composition of the MSC population itself.

Figure 2.

PATH-5357-FIG-0002-c

Summary of MSC effects on the tumor microenvironment. MSCs have multiple effects on tumor cells, mainly promoting tumor growth due, at least in part, to their role in regulating inflammation and tissue repair (1). They affect tumor cell survival and stemness (2 and 3) and contribute to tumor vasculature by producing angiogenic factors and by differentiating into pericytes (4). MSCs promote tumor cell motility, EMT, and metastasis, and secrete chemokines, including CXCL1, CXCL2, and CXCL12, and cytokines, including IL‐6 and several matrix metalloproteinases (MMPs), which degrade the ECM and facilitate tumor cell migration (5). They exert an important immunomodulatory function, which is primarily immunosuppressive (6) and can enhance tumor cell resistance to drugs, at least in part by releasing exosomes, which harbor numerous mediators, including miRNAs that may alter tumor cell properties (7). Although MSCs are primarily pro‐tumorigenic, several studies have shown that they may display anti‐tumor effects (8) as well. For a complete list of abbreviations see supplementary material, Table S1.

MSCs interact with and may affect the function of cancer cells at multiple stages of cancer progression. Within the primary tumor, MSCs have been observed to drive tumor cells toward acquiring invasive and metastatic properties. MSCs induce expression of epithelial–mesenchymal transition (EMT)‐ and hypoxia‐related genes in primary tumor cells and promote tumor cell dissemination 70. They also deposit ECM 88; participate in the remodeling of the TME; secrete IL‐6 and TGF‐β, which induces EMT; and help to create a niche that promotes angiogenesis and tumor invasion 5, 89. These observations are consistent with the predominantly pro‐tumorigenic effect of MSCs in vivo. The tumor inhibitory effects that have been reported may be due to differences in experimental design, models used, and MSC heterogeneity, which may reflect variable responses to a given set of stimuli (explored more extensively in a review by Klopp et al) 6, 90.

Effect of the tumor microenvironment on the MSC phenotype

Tumor‐derived signals have the capacity to modulate the phenotype of tissue‐resident and tumor‐recruited MSCs (T‐MSCs), which become constituents of the tumor mass and harbor features distinct from those of normal tissue MSCs (N‐MSCs) or bone marrow MSCs (BM‐MSCs) 91. Differences between non‐tumor‐associated MSCs and T‐MSCs may arise in large part in response to cytokines and exosomes produced by the tumor microenvironment. This notion is supported by observations that MSCs treated with IFN‐γ and TNF‐α upregulate TGF‐β and VEGF expression 92, 93. TGF‐β can then promote EMT, which may facilitate invasion and metastasis 92. In addition, IFN‐γ and TNF‐α can enhance the immunosuppressive effects of MSCs 94, further helping tumor cell dissemination. Exosomes derived from breast and ovarian cancer cells can cause adipose tissue MSCs to adopt a CAF phenotype, characterized, in part, by upregulated α‐SMA expression, and can also promote MSC expression of SDF‐1, VEGF, CCL5 (RANTES), and TGF‐β 95, 96. Recently, Raz et al 97 have shown in breast tumors that resident and BM‐derived MSCs differentiate toward a subpopulation of CAF‐like cells that express distinct immune‐response‐related genes. Analysis of gene expression in these resident and BM‐derived CAF‐like cells from mammary tumors or their lung metastases revealed tissue‐specific transcriptional changes, implicating a microenvironmental influence on the reprogramming of stromal cells. Interestingly, BM‐derived CAF‐like cells were shown to be functionally important for tumor growth and were more efficient than their resident counterparts in promoting angiogenesis. Thus, MSCs recruited to neoplastic tissues can be reprogrammed in a local, tissue‐specific manner to induce tumor‐promoting inflammation and to facilitate angiogenesis and tumor growth 97.

Characteristics of tumor‐associated MSCs (T‐MSCs)

Tumor‐associated MSCs do not undergo transformation and are euploid 98, 99. Moreover, MSCs are more prevalent in tumor tissues than in adjacent normal tissues 33, 70 and exhibit a significantly greater proliferative capacity than their normal tissue‐associated counterparts 99, 100, 101, 102. In addition, T‐MSCs exhibit a stronger migratory capability than N‐MSCs and more potent immunosuppressive activity than BM‐MSCs 99, 101, 102, 103, 104, 105, 106, 107. Finally, T‐MSCs have been shown to promote tumor cell proliferation 99 and to increase the proportion of cancer stem cells 99, 108, suggesting a possible role in tumor cell reprogramming.

Pro‐inflammatory and immunosuppressive effects of MSCs in the TME: Dr Jekyll and Mr Hyde behavior?

As discussed above, MSCs affect the immune response by secreting immunomodulatory molecules as well as by cell–cell contact. Several studies have also highlighted the role of exosomes and other extracellular vesicles (EVs) on MSC‐mediated immune modulation 109, 110. Two major functional features of MSCs that are relevant to immunity include their ability to induce immunosuppression and to exert immunoprivilege. Immunosuppression has primarily been associated with BM‐MSCs, whereas studies using other sources of MSCs have shown both immunosuppressive and pro‐inflammatory effects. These seemingly contradictory observations may be due to species‐specific factors but possibly also to the tissue from which the MSCs were harvested and to priming by their microenvironment. The mechanisms underlying immunoprivilege are largely unknown but are most probably related to low expression of MHC I and MHC II molecules coupled to the immunosuppressive functions of MSCs. Furthermore, immunoprivilege is not a stable property: cellular differentiation and priming by IFN‐γ upregulate MHC‐I and, to a lesser extent, MHC‐II expression, enhancing MSC antigen‐presenting capacity and immunogenicity and reducing immunoprivilege 111.

The immunosuppressive effects of MSCs require proximity to their target cells, which include T and B lymphocytes as well as NK cells (Figure 3). Activated/primed MSCs upregulate MHC class I molecules, ICAM‐1 and VCAM‐1 adhesion receptors, and the immunosuppressive molecule PD‐L1 (programmed death‐ligand 1). The latter three molecules recognize ligands on immune cells, promote cell–cell adhesion, and facilitate immune cell exposure to secreted immunosuppressive mediators 139. Following activation, the MSC‐derived secreted immunosuppressive arsenal includes HLA‐G, TGF‐β, PGE2, TSG‐6 (tumor necrosis factor‐inducible gene 6 protein), HO‐1 (heme oxygenase 1), HGF, IL‐10, IL‐6, IDO1 (indoleamine‐pyrrole 2,3‐dioxygenase), ARG1/2 (arginase), NOS2 (nitric oxide synthase 2A), adenosine, and LIF (leukemia inhibitory factor), as well as PD‐L1/2 and Fas ligand (FasL) 140. TGF‐β and PGE2 are two key mediators of immunosuppression. TGF‐β directly inhibits the function of anti‐tumor effector cells (NK, CD8+ T cells, and γδ T cells) by downregulating the activating receptor NKG2D and generating and recruiting regulatory T cells and γδ T cells 4, 112, 143, 144, 145, 146, 147, 148, 149, 150.

Figure 3.

PATH-5357-FIG-0003-c

MSC and immune cell interactions. (1) MSCs can inhibit the proliferation, cytotoxicity, and cytokine production by NK cells by secreting several mediators, including PGE2, IDO, and sHLA‐G5. In turn, MSCs can be killed by cytokine‐activated NK cells through the engagement of NKG2D by its ligand ULBP3 or MICA expressed by MSCs, and of DNAM‐1 by MSC‐associated PVR or nectin‐2. (2) MSCs inhibit differentiation of monocytes to DCs, skew mature DCs toward an immature DC state, and inhibit TNF‐α and IL‐12 production by DCs through PGE2 secretion. (3) MSCs dampen the respiratory burst and delay spontaneous apoptosis of neutrophils by constitutively releasing IL‐6. (4) MSCs affect CD4+ T cells through PGE2, IDO, TGF‐β, HGF, iNOS, and HO1 release. MSCs increase the production of IL‐4 and IL‐10 by Th2 cells and reduce the release of IFN‐γ by Th1 and NK cells. IDO can reduce tryptophan levels and inhibit the growth of B cells, T cells, and NK cells. Defective CD4+ T‐cell activation impairs helper function for B‐cell proliferation and antibody production. CD8+ T‐cell cytotoxicity is inhibited mainly by sHLA‐G5, as well as by the increase of the regulatory T‐cell population, also induced by IL‐10. Adapted from Refs. 9, 112, 116. For a complete list of abbreviations see supplementary material, Table S1.

MSCs and innate immunity

MSCs exert their pro‐ and anti‐inflammatory effects by a variety of mechanisms. They have a functional relationship with the complement system, as BM‐MSCs express the anaphylatoxin receptors C3aR and C5aR, suggesting that C3a and C5a may be chemotactic for MSCs toward sites of injury. MSCs also express the complement inhibitors CD46, CD55, and most predominantly CD59, which protect them from complement opsonization and lysis and secrete the complement inhibitor, factor H 144, 152, 153.

MSCs play an active role in neutrophil recruitment by secreting chemotactic cytokines and chemokines, including IL‐6, IL‐8, IFN‐β, GM‐CSF, and macrophage inhibitory factor (MIF). They also promote neutrophil survival, which helps to eliminate pathogens 114 and to respond to histamine released by mast cells by inducing IL‐6 production 117.

MSCs directly and indirectly interfere with the proliferation, cytokine production, and, in some cases, cytotoxicity of NK cells. MSC–NK cell interactions are complex and largely dependent on the microenvironment and activation status of the NK cells. Bone marrow‐derived MSCs can inhibit NK cell proliferation, cytotoxicity, and cytokine production by secreting IDO1, TGF‐β, HLA‐G, and PGE2 103, 116, 117, 118, 119, 120. However, they are also vulnerable to lysis by activated NK cells, depending on their expression of activating NK receptor ligands, including the MHC class I polypeptide‐related sequence (MICA, B), UL16 binding proteins (ULBPs), CD112, and CD155 120, 121, 122. In human lung cancers, T‐MSCs have been shown to be more immunosuppressive than N‐MSCs, mainly through PGE2 and, to a lesser extent, IL‐6 secretion. They decrease IFN‐γ production and downregulate expression of the activating NK cell receptors NKp44, NKp30, NKG2D, DNAM‐I, and NKG2A. T‐MSCs also induce an inversion in the CD56bright/dim NK cell ratio in favor of the CD56dim phenotype, which is associated with degranulation rather than elevated cytokine production, in a contact‐dependent manner 103.

Dendritic cell (DC) function is affected by MSCs, which can directly inhibit both the maturation of monocytes and CD34+ precursor cells toward DCs as well as activation of DCs via PGE2, IL‐6, TSG‐6, and M‐CSF (macrophage colony‐stimulating factor) secretion, and Jagged‐2 mediated signaling 136, 137, 138. Both immature and mature DCs (iDCs and mDCs) are affected by MSCs. In the presence of MSCs, iDCs display diminished capacity to present antigen and stimulate T‐cell proliferation and naïve T‐cell differentiation, resulting in ineffective T‐cell activation. MSCs can also revert mDCs to an immature phenotype associated with downregulation of their cell surface expression of antigen‐presenting and co‐stimulatory molecules, suppression of IL‐12 secretion, and the inability to stimulate lymphocyte proliferation in vitro 138.

MSCs seem to favor the emergence of the myeloid suppressor cell (MDSC) phenotype by secreting IL‐6, HGF, and CXCL3 (C‐X‐C motif chemokine ligand 3), which stimulate MDSC production of COX2 (cyclooxygenase‐II enzyme, PTGS2), IDO, PD‐L1, and PD‐L2, and MMP9 (matrix metalloproteinase 9) 156, 157.

Mesenchymal stromal cells direct monocyte mobilization from the BM and macrophage recruitment to sites of inflammation to promote wound repair through secretion of the chemokine (C‐C motif) ligands CCL2, CCL3, and CCL12. They also participate in the differentiation of monocytes to M2 (tissue repair‐associated) macrophages via direct cell contact and secretion of PGE2, IL‐6, and IDO 124, 125. The capacity of MSCs to regulate the macrophage phenotype (M1 or M2) and to promote immunosuppression strongly depends on macrophage IL‐6 signaling 126. Finally, as discussed above, MSCs impair the maturation and differentiation of antigen‐presenting cells (APCs) 123.

Current research is exploring the effect of MSCs on γδ T cells. γδ T cells have the ability to produce the pro‐inflammatory cytokines IFN‐γ, TNF‐α, and IL‐17, as well as the anti‐inflammatory cytokines TGF‐β, IL‐4, and IL‐10, depending on the types of signals that predominate in the tissue microenvironment, and exert both anti‐ and pro‐tumoral effects 158. TGF‐β acts as a key player in the MSC‐mediated regulatory response by inducing CD4+ regulatory T cells (Treg) and γδ regulatory T cells. On the other hand, MSCs are potent suppressors of γδ‐cell proliferation, cytokine production, and cytolytic responses (anti‐tumor effect) in vitro through COX2‐dependent production of PGE2 134.

MSCs and adaptive immunity

MSCs can regulate the activation and function of T and B lymphocytes 9, 113. Many factors have been reported to be critical in MSC‐mediated suppression of T‐cell proliferation, including iNOS (inducible NO synthase), IDO, semaphorin‐3A, B7‐H4, HLA‐G, LIF, galectin(s), HO‐1, IL‐6, IL‐10, PD‐L1/2, FasL, and PGE2 127, 128, 129. MSCs exert inhibitory effects toward Th1 and Th17 (pro‐inflammatory) cells through PD‐1, PGE2, and IL‐10, and promotion of Th2 secretion of IL‐4 130. However, stimulatory effects on Th17 cells have also been observed. MSCs can promote Treg differentiation by secreting TGF‐β, IL‐6, and IL‐10, and expressing IDO 135.

The immunosuppressive function of MSCs is elicited by IFN‐γ, which induces the production of chemokines, IDO, PGE2, HGF, and TGF‐β in humans to attract and to suppress T cells 131, 159. Although soluble factors are critical to mediate the immunosuppressive functions of MSCs, cell–cell contact is involved in MSC‐based immunosuppression of T cells, including ICAM‐1–LFA‐1 and PD‐1/PD‐L1 interactions. Additional mechanisms of suppression occur through microRNA and exosome release 159, 160, 161.

B‐cell–MSC interactions are less well studied, although observations suggest an inhibitory effect on B‐cell activation, proliferation, differentiation, and chemotactic responses 132, 133.

Priming: Toll‐like receptors and the level of inflammation

MSC immunomodulatory activity is reported to be primed by cytokines from a pro‐inflammatory microenvironment (for example, inflammation, cancer or infection), particularly IFN‐γ, TNF‐α, and IL‐1β, and by Toll‐like receptor (TLR) stimulation 162, 163, 164, 165, 166, 167, 168. MSCs express TLR1–6 transcripts and TLR2–4, 7, and 9 proteins, and can be polarized toward a pro‐inflammatory or an immunosuppressive phenotype following specific TLR stimulation 169, 170, 171, 172. Thus, TLR4‐primed MSCs are polarized toward a pro‐inflammatory MSC1 phenotype (tumor‐growth inhibition), whereas TLR3‐primed MSCs are polarized toward the more classical immunosuppressive MSC2 phenotype (Figure 4) 173, 175. The degree of inflammation, as assessed by the cytokine repertoire and production level within the microenvironment, appears to be critical in MSC polarization. MSCs become immunosuppressive only when exposed to high levels of pro‐inflammatory cytokines (TNF‐α, IFN‐γ), which corresponds to late stages of inflammation 139, 176, but the MSC2 phenotype is also influenced by TLR3 stimulation 176. In the presence of low levels of TNF‐α and IFN‐γ, at the early phase of inflammation, MSCs may adopt a pro‐inflammatory phenotype (MSC1) and enhance immune responses, in part through TLR4 expressed on their surface 175. TLR4 stimulation promotes IL‐6, IL‐8, and TGF‐β secretion, whereas TLR3 stimulation increases IL‐4, IL‐1RA (interleukin‐1 receptor antagonist), IDO, and PGE2 164, 175, 177, 178. However, results are discordant among research groups and MSC activation through TLR3 and 4 is also reported to lead to the secretion of IL‐1, IL‐6, IL‐8, TRAIL (TNF‐related apoptosis‐inducing ligand), and CCL5 174, 175, 179. Some studies have shown that both TLR3 and 4 enhance immunosuppression through IDO 141, whereas others have reported an increase in pro‐inflammatory cytokines in both 142, 151, 154, 179, 180. Thus, MSC licensing to become activated depends on stimulation by pro‐inflammatory cytokines, priming signals delivered by TLRs, and the timing of MSC engagement in immune effector cell activation (Figure 5) 164.

Figure 4.

PATH-5357-FIG-0004-c

The MSC polarization paradigm. The level of inflammation and/or TLR agonists polarizes MSCs toward a pro‐inflammatory/anti‐tumor (MSC1) or an anti‐inflammatory/immunosuppressive/pro‐tumor (MSC2) phenotype. Low levels of TNF‐α, IFN‐γ (low level of inflammation), and/or TLR4 agonists (LPS) polarize MSCs toward a pro‐inflammatory phenotype, whereas the downstream consequences of high TNF‐α/IFN‐γ/TLR3 [poly(I:C)] stimulation skew MSCs toward an anti‐inflammatory MSC2 phenotype. MSC1 and MSC2 have divergent cytokine and chemokine secretion repertoires, differences in differentiation capability, extracellular matrix deposition, TGF‐β signaling pathways, and Jagged, IDO, and PGE2 expression. The most compelling outcome is the opposite effect of the two cell types on immunomodulation. As shown in the figure, existing data are conflicting regarding the repertoire of cytokines and chemokines secreted in response to the numerous stimuli from the microenvironment. MSCs are a heterogeneous cell population, and the range of the observed responses may be explained, at least in part, by their diversity itself. For a complete list of abbreviations see supplementary material, Table S1.

Figure 5.

PATH-5357-FIG-0005-c

TLR priming of MSCs. (1) The classical view of MSC polarization: different levels of inflammation and/or TLR agonist stimulation polarize MSCs toward a pro‐inflammatory/anti‐tumor (MSC1) or anti‐inflammatory/immunosuppressive/pro‐tumor (MSC2) phenotype. Low levels of TNF‐α, IFN‐γ (low level of inflammation), and/or TLR4 agonists polarize MSCs toward a pro‐inflammatory phenotype, whereas stimulation with high TNF‐α/IFN‐γ/TLR3 levels promotes induction of an anti‐inflammatory MSC2 phenotype. (2) In the tumor microenvironment, MSCs are continuously exposed to pro‐inflammatory cytokines and tend to express both TLR3 and TLR4. They acquire features that help to sustain tumor progression and that overlap with those defined by the classical MSC1 and MSC2 phenotypes. (3) Petri et al showed that the dichotomous pro‐ and anti‐inflammatory effects of TLR3‐stimulated MSCs may be time‐related 184. MSCs may initially respond by secreting pro‐inflammatory IFN‐α/β and later switch to production of the regulatory factors IL‐6 and TGF‐β. MSCs may thereby actively contribute to each phase of wound healing, progressively driving the process to completion and restoration of tissue function. For a complete list of abbreviations see supplementary material, Table S1.

Experimentally, the duration of TLR stimulation of MSCs seems to play a role in their subsequent activation profile. Brief stimulation of MSCs with poly(I:C) led to an MSC2, whereas LPS stimulation induced an inflammatory MSC1 phenotype 175. However, MSCs at an infection site are likely to be continuously exposed to TLR agonists.

In contrast to the study of Waterman et al 175, other investigators have shown that TLR3 stimulation of MSCs leads to a pro‐inflammatory response 181, 182, 183, 184. The dichotomous pro‐ and anti‐inflammatory effects of TLR3‐stimulated MSCs may be time‐related. Thus, upon arriving at an infection site, MSCs may initially respond by secreting pro‐inflammatory IFN‐α/β and later switch to production of the regulatory factors IL‐6 and TGF‐β (Figure 5). They observed that the effect of constitutively produced TGF‐β was modulated by the presence of inducible IL‐6. MSCs may thereby actively contribute to each phase of wound healing, progressively driving the process to completion and restoration of tissue function 184.

Several mediators, including COX1, COX2, LIF, HGF, Gal‐1, HO‐1, IL‐11, IL‐8, IL‐6, and TGF‐β, were observed to display variable constitutive expression in BM‐MSCs. Inflammatory priming of the cells differentially modulated expression of these mediators, strongly increasing expression of COX2, LIF, HGF, IL‐11, IL‐8, and IL‐6, while decreasing that of COX1, Gal‐1, and TGF‐β 185. According to one model 186, strong inflammation, characterized by IFN‐γ, TNF‐α, IL‐1, and IL‐17 production, induces NO/IDO production by MSCs, leading to an immunosuppressive phenotype, with secretion of TGF‐β, sHLA‐G, IL‐6, IL‐10, and IDO. In contrast, MSCs subjected to a microenvironment enriched in TGF‐β and IL‐10 acquire a pro‐inflammatory phenotype. Interestingly, MSCs themselves produce abundant TGF‐β, which can act in an autocrine manner to modulate their immunoregulatory functions 187. Thus, TGF‐β can reverse the immunosuppressive effect of MSCs induced by IFN‐γ and TNF‐α 186.

MSC‐derived exosomes

Mesenchymal stromal cell‐derived extracellular vesicles (MSC‐EVs) play a significant role in the TME 188, 189. Release of EVs is a mechanism of intercellular communication used by tumor cells and stromal cells within the TME. EVs include exosomes, the smallest EV fraction arising from intracellular endosomes, and microvesicles generated by budding from the plasma membrane. EVs can directly activate target cell surface receptors through protein and bioactive lipid ligands, and by delivering different effectors, including transcription factors, oncogenes, proteins, growth factors, and non‐coding regulatory RNAs, thus inducing functional changes in recipient cells.

MSC‐EVs are regulators of tumor cell survival and growth

Similar to their effects on immunity and the inflammatory response, MSCs have shown divergent effects on tumor cells, some of which are anti‐, whereas others are pro‐tumorigenic. Similar to MSCs, MSC‐EVs have a dual effect on tumor progression. Exosomes derived from MSCs in non‐small cell lung cancer have been shown to promote chemoresistance 190. Tumor‐EVs can also mediate drug resistance through mechanisms that include drug sequestration; delivery of specific mRNAs, miRNAs, and proteins; and crosstalk between cancer cells and MSCs 191, 192. MSC‐EVs have been found to modulate the tumor microenvironment, creating favorable conditions for cancer cell metastasis, and have been shown to mimic the effects of MSCs on tumor growth promotion 193. Kalluri 194 described tumor‐associated and circulating exosomes as heterogeneous populations that generate a unique tumor nanoenvironment (TNE).

Immunomodulation

By fulfilling the role of vehicles that deliver immunomodulatory mediators, MSC‐EVs display functions similar to those of their parent cells 195. Cytokines (including IL‐6, IDO, PGE2, IL‐10) and chemokines (including CXCL2, CCL2, CXCL8) may be packaged into EVs together with nucleic acids and post‐transcriptional modulators which can influence the inflammatory response when released 195, 196. Exosomes can inhibit B‐cell proliferation 197 and increase Treg activity 198. In physiological conditions, MSC‐EVs have been reported to modulate the immune cell response to facilitate tissue repair, through promotion of anti‐inflammatory and pro‐regenerative (M2) macrophages over pro‐inflammatory M1 macrophages and concomitant enhancement of the expression of the anti‐inflammatory cytokines IL‐10 and TGF‐β 199. In tumors, chronic inflammation promotes immunosuppression, at least in part through EV release, which contributes to tumor progression 200, 201.

Interleukin‐6

Aside from TGF‐β and PGE2, IL‐6 appears to play a major role in MSC communication with their microenvironment. IL‐6 is a pleiotropic cytokine, highly secreted by tumor stromal cells, including MSCs. The IL‐6 signaling pathway consists of IL‐6Rα (CD126) and gp130 (CD130), JAK/STAT signaling, and negative regulation by SOCS molecules. IL‐6 supports cancer cell proliferation, survival, and metastatic dissemination. Moreover, IL‐6 can act on numerous cell types within the tumor microenvironment to sustain a pro‐tumoral milieu by supporting angiogenesis and tumor evasion of immune surveillance. However, IL‐6 may also promote anti‐tumor adaptive immunity 202, 203.

MSCs, especially MSCs isolated from the tumor stroma, secrete higher levels of IL‐6 70, 103 than other non‐tumor cells, which together with PGE2 can participate in suppression of NK cell activity and facilitate tumor cell dissemination and metastasis 70. IL‐6 secretion by MSCs has been shown to be part of the late regulatory response 184, which includes TGF‐β, to induce senescent‐like NK cells. TGF‐β secreted by MSCs in osteosarcoma can increase the migratory capacity of tumor cells, which, in turn, stimulate the secretion of IL‐6 that fosters cancer cell stemness and aggressiveness 204, 205. In a model of arthritis, IL‐6‐dependent PGE2 secretion by MSCs inhibits local inflammation 206. Indeed, IL‐6 modifies the soluble mediator secretion profile of MSCs, increasing PGE2 and VEGF, among others. MSC‐derived IL‐6 and PGE2 skew monocyte differentiation toward the formation of IL‐10‐expressing macrophages 207. IL‐6 clearly plays an important role in the crosstalk between MSCs and the tumor microenvironment, and additional work is needed to elucidate the full spectrum of its effects. In different tumor models, targeted inhibition of IL‐6 may enhance the efficacy of anti‐PD‐L1 208, 209, 210.

MSCs and anti‐tumor therapy

The potential therapeutic benefit of exogenous MSCs has been under preclinical investigation since the late 1990s. Tissue regeneration‐related candidate MSC applications include bone marrow transplantation, graft‐versus‐host disease (GVHD), acute myocardial infarction, heart failure, stroke, lung diseases, acute kidney failure, liver fibrosis, juvenile diabetes, osteoarthritis and rheumatoid arthritis, inflammatory bowel disease, multiple sclerosis, Parkinson's disease, and sepsis 211, 212, some of which are approved (GVHD being one example) 213, 214, 215. There are currently also ongoing clinical trials that use MSCs to treat tumors 7. In the context of such cell therapeutic approaches, MSCs are used as gene delivery vehicles for tumor‐targeted therapies 216, 217, 218. MSCs have been engineered to deliver interleukins to improve anti‐cancer immune surveillance, as delivery agents of interferons (IFN‐α and ‐β) 159, and as carriers of prodrugs or oncolytic viruses 219, 220. MSCs have also been tested as deliverers of anti‐angiogenic agents, pro‐apoptotic proteins (TRAIL, for example) 221, and growth factor antagonists. Moreover, after exposure to high doses of chemotherapeutic drugs such as paclitaxel or gemcitabine, MSCs have been shown to accumulate and deliver the anti‐neoplastic agent without undergoing genetic modifications and to decrease tumor growth 222, 223. Their ability to preferentially migrate toward tumor sites (primary and metastatic neoplasms) in addition to their availability, non‐immunogenic nature, and relative ease of manipulation in vitro renders them attractive candidates for cell‐based therapies. However, as discussed above, increasing evidence regarding the tumor‐promoting activity of MSCs, especially when subverted by the TME, raises issues as to their safety and cautions their use in clinical trials. In addition, using engineered MSCs may be a problem after eradication of the tumor they are designed to target 224. A recent systematic review by Christodoulou et al addresses these issues in the settings of preclinical cancer cytotherapy 225.

MSC‐EVs for anti‐tumor therapy

Several studies suggest that the cell source may condition EV homing to specific sites and that their membrane can be engineered to increase tissue‐specific targeting 226, 227. These observations open new possibilities for potential future applications of MSC‐EVs as cell‐free therapeutic agents 228. MSC‐derived exosomes may thus be used as delivery vehicles to transfer genetic materials, including mRNA and non‐coding RNAs to recipient cells 160, 229, 230, 231, 232.

Targeting the pro‐tumor effects of MSCs

Approaches that could be used to target MSCs in the TME and counteract their immunosuppressive effects include direct blockade of their immunosuppressive function and reprogramming to render their immunostimulatory properties dominant over their immunosuppressive ones 5. MSC activity could be modulated in a variety of ways using, among others, drugs that inhibit one or, preferably, several of the MSC‐derived immunosuppressive molecules (e.g. IDO, TGF‐β); inhibitory antibodies (e.g. anti‐PDGF, anti‐EGFR antibodies) that block the effect of growth factors involved in MSC–tumor cell crosstalk 233, 234, 235 and elicit ADCC (antibody‐dependent cellular cytotoxicity); inhibitors of sheddases/ADAMs (a disintegrin and metalloproteinases); and tyrosine kinase inhibitors. Some of these approaches have the advantage of targeting both MSCs and tumor cells. HMG‐CoA reductase inhibitors (statins) decrease mevalonate, its metabolic product that is essential for MSC and tumor cell metabolism. However, mevalonate is also required to develop an immune response and kill tumor cells. Thus, it is important to design inhibitors that can be directly and specifically delivered to MSCs 236.

PD‐L1 expression is upregulated in MSCs by IFN‐γ 237, and PD‐L1/PD1 is involved in MSC regulation of T‐ and B‐cell proliferation 238, 239. ADAM proteins release MSC ligands for NK cells and decrease NK‐cell recognition of tumor cells. ADAMs can be released in exosomes and microvesicles.

Reprogramming MSCs from an immunosuppressive to an immunostimulatory phenotype may constitute another potentially promising approach. The effects of bisphosphonates 240, 241, as well as immunomodulatory drugs such as thalidomide and lenalidomide, that target the TME and decrease IL‐6 by regulating SOCS1 242 are being assessed by several groups.

Preclinical models

Studies on the effect of MSCs on cancer growth and their immunomodulatory properties have been based mainly on in vitro 2D co‐culture systems and on in vivo cancer models using primarily BM‐MSCs and adipose tissue‐isolated MSCs. New approaches using more complex in vitro 3D models are under development and are gaining interest, as they are more prone to mimic the in vivo features of the tumor microenvironment 204. For a more detailed review, the recent publication of Avnet et al 204 summarizes the different 3D preclinical models available, as well as their limitations.

Concluding remarks

Mesenchymal stem cells may be heralded as key preservers of tissue homeostasis. Their pleiotropic nature provides them with the unique ability to act as sensors of tissue state and as both coordinators of and participants in the effector functions required to repair and regenerate injured tissues. By sensing the degree of an inflammatory response to injury, MSCs may adapt their own regulatory and effector functions to temper or boost the response as required. Thus, they can become immunosuppressive upon exposure to elevated levels of pro‐inflammatory cytokines while providing support to tissue repair by secreting ECM components and stimulating regeneration by resident stem cells. By contrast, in the presence of low levels of IFN‐γ and TNF‐α, MSCs may adopt a pro‐inflammatory phenotype and enhance T‐cell responses as well as tissue remodeling.

Despite its highly beneficial effects in the maintenance of homeostasis, MSC plasticity is a double‐edged sword as it can be readily exploited by tumors to serve tumor cell needs. In response to tumor‐derived cytokines and signals generated by direct physical contact with tumor cells, MSCs can adopt a potent immunosuppressive phenotype that acts on both innate and adaptive immunity. As a result, they may facilitate tumor progression, which entails acquisition by tumor cells of additional genetic and epigenetic changes that may shield them from cytotoxic cells and drugs and support their formation of metastases.

Although a plethora of studies have been conducted on MSCs in recent years, several key issues remain to be resolved. Perhaps the most pressing one is the heterogeneity of MSCs, which requires the identification and definition of their putative subpopulations and the determination of their mutual relationships. For example, are the different subpopulations stable or are they transient and can one subpopulation transform itself into another in response to microenvironmental stimuli? Several other questions need to be addressed as well. What is the relationship between MSCs and ‘resting’ fibroblasts – are they distinct entities or one and the same, perhaps at different stages of differentiation? What proportion of the stromal response to injury is directed by MSCs versus other more differentiated stromal cells? These and other issues will need to be resolved if we hope to effectively disrupt the functional support that MSCs provide to tumor growth.

Author contributions statement

Both authors contributed to this review and reviewed the final manuscript.

Supporting information

Table S1. Complete list of abbreviations used

Acknowledgements

We thank Professor Christophe Galland for helpful suggestions. SG was supported by a Clinician–MD–PhD scientist research grant from the Faculty of Biology and Medicine Research Commission Fund, University of Lausanne, and by a Junior Clinical Scientist Leenaards Foundation research grant, Lausanne. This work was supported by Swiss National Science Foundation grant 310030_169563.

No conflicts of interest were declared.

References

  • 1. Bianco P, Robey PG, Simmons PJ. Mesenchymal stem cells: revisiting history, concepts, and assays. Cell Stem Cell 2008; 2: 313–319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Keating A. Mesenchymal stromal cells: new directions. Cell Stem Cell 2012; 10: 709–716. [DOI] [PubMed] [Google Scholar]
  • 3. Lazennec G, Lam PY. Recent discoveries concerning the tumor–mesenchymal stem cell interactions. Biochim Biophys Acta 2016; 1866: 290–299. [DOI] [PubMed] [Google Scholar]
  • 4. Papaccio F, Paino F, Regad T, et al Concise review: cancer cells, cancer stem cells, and mesenchymal stem cells: influence in cancer development. Stem Cells Transl Med 2017; 6: 2115–2125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Poggi A, Varesano S, Zocchi MR. How to hit mesenchymal stromal cells and make the tumor microenvironment immunostimulant rather than immunosuppressive. Front Immunol 2018; 9: 262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Ridge SM, Sullivan FJ, Glynn SA. Mesenchymal stem cells: key players in cancer progression. Mol Cancer 2017; 16: 31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Timaner M, Tsai KK, Shaked Y. The multifaceted role of mesenchymal stem cells in cancer. Semin Cancer Biol 2020; 60: 225–237. [DOI] [PubMed] [Google Scholar]
  • 8. Tolar J, Le Blanc K, Keating A, et al Concise review: hitting the right spot with mesenchymal stromal cells. Stem Cells 2010; 28: 1446–1455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Uccelli A, Moretta L, Pistoia V. Mesenchymal stem cells in health and disease. Nat Rev Immunol 2008; 8: 726–736. [DOI] [PubMed] [Google Scholar]
  • 10. Karnoub AE, Dash AB, Vo AP, et al Mesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature 2007; 449: 557–563. [DOI] [PubMed] [Google Scholar]
  • 11. Lacerda L, Debeb BG, Smith D, et al Mesenchymal stem cells mediate the clinical phenotype of inflammatory breast cancer in a preclinical model. Breast Cancer Res 2015; 17: 42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Ye H, Cheng J, Tang Y, et al Human bone marrow‐derived mesenchymal stem cells produced TGFbeta contributes to progression and metastasis of prostate cancer. Cancer Invest 2012; 30: 513–518. [DOI] [PubMed] [Google Scholar]
  • 13. Lee MJ, Heo SC, Shin SH, et al Oncostatin M promotes mesenchymal stem cell‐stimulated tumor growth through a paracrine mechanism involving periostin and TGFBI. Int J Biochem Cell Biol 2013; 45: 1869–1877. [DOI] [PubMed] [Google Scholar]
  • 14. Sun B, Roh K‐H, Park J‐R, et al Therapeutic potential of mesenchymal stromal cells in a mouse breast cancer metastasis model. Cytotherapy 2009; 11: 289–298. [DOI] [PubMed] [Google Scholar]
  • 15. Abd‐Allah SH, Shalaby SM, El‐Shal AS, et al Effect of bone marrow‐derived mesenchymal stromal cells on hepatoma. Cytotherapy 2014; 16: 1197–1206. [DOI] [PubMed] [Google Scholar]
  • 16. Otsu K, Das S, Houser SD, et al Concentration‐dependent inhibition of angiogenesis by mesenchymal stem cells. Blood 2009; 113: 4197–4205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Spaeth EL, Dembinski JL, Sasser AK, et al Mesenchymal stem cell transition to tumor‐associated fibroblasts contributes to fibrovascular network expansion and tumor progression. PLoS One 2009; 4: e4992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Akimoto K, Kimura K, Nagano M, et al Umbilical cord blood‐derived mesenchymal stem cells inhibit, but adipose tissue‐derived mesenchymal stem cells promote, glioblastoma multiforme proliferation. Stem Cells Dev 2012; 22: 1370–1386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Li W, Zhou Y, Yang J, et al Gastric cancer‐derived mesenchymal stem cells prompt gastric cancer progression through secretion of interleukin‐8. J Exp Clin Cancer Res 2015; 34: 52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Hossain A, Gumin J, Gao F, et al Mesenchymal stem cells isolated from human gliomas increase proliferation and maintain stemness of glioma stem cells through the IL‐6/gp130/STAT3 pathway. Stem Cells 2015; 33: 2400–2415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Mishra PJ, Mishra PJ, Humeniuk R, et al Carcinoma‐associated fibroblast‐like differentiation of human mesenchymal stem cells. Cancer Res 2008; 68: 4331–4339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Shangguan L, Ti X, Krause U, et al Inhibition of TGF‐β/Smad signaling by BAMBI blocks differentiation of human mesenchymal stem cells to carcinoma‐associated fibroblasts and abolishes their protumor effects. Stem Cells 2012; 30: 2810–2819. [DOI] [PubMed] [Google Scholar]
  • 23. Khakoo AY, Pati S, Anderson SA, et al Human mesenchymal stem cells exert potent antitumorigenic effects in a model of Kaposi's sarcoma. J Exp Med 2006; 203: 1235–1247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Qiao L, Xu Z, Zhao T, et al Suppression of tumorigenesis by human mesenchymal stem cells in a hepatoma model. Cell Res 2008; 18: 500–507. [DOI] [PubMed] [Google Scholar]
  • 25. Kim E‐K, Kim H‐J, Yang Y‐I, et al Endogenous gastric‐resident mesenchymal stem cells contribute to formation of cancer stroma and progression of gastric cancer. Korean J Pathol 2013; 47: 507–518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Kansy BA, Dißmann PA, Hemeda H, et al The bidirectional tumor–mesenchymal stromal cell interaction promotes the progression of head and neck cancer. Stem Cell Res Ther 2014; 5: 95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Wolfe AR, Trenton NJ, Debeb BG, et al Mesenchymal stem cells and macrophages interact through IL‐6 to promote inflammatory breast cancer in pre‐clinical models. Oncotarget 2016; 7: 82482–82492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Kidd S, Caldwell L, Dietrich M, et al Mesenchymal stromal cells alone or expressing interferon‐beta suppress pancreatic tumors in vivo, an effect countered by anti‐inflammatory treatment. Cytotherapy 2010; 12: 615–625. [DOI] [PubMed] [Google Scholar]
  • 29. Ho IAW, Toh HC, Ng WH, et al Human bone marrow‐derived mesenchymal stem cells suppress human glioma growth through inhibition of angiogenesis. Stem Cells 2013; 31: 146–155. [DOI] [PubMed] [Google Scholar]
  • 30. Castells M, Milhas D, Gandy C, et al Microenvironment mesenchymal cells protect ovarian cancer cell lines from apoptosis by inhibiting XIAP inactivation. Cell Death Dis 2013; 4: e887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Lin J‐T, Wang J‐Y, Chen M‐K, et al Colon cancer mesenchymal stem cells modulate the tumorigenicity of colon cancer through interleukin 6. Exp Cell Res 2013; 319: 2216–2229. [DOI] [PubMed] [Google Scholar]
  • 32. Han Z, Tian Z, Lv G, et al Immunosuppressive effect of bone marrow‐derived mesenchymal stem cells in inflammatory microenvironment favours the growth of B16 melanoma cells. J Cell Mol Med 2011; 15: 2343–2352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Hernanda PY, Pedroza‐Gonzalez A, van der Laan LJW, et al Tumor promotion through the mesenchymal stem cell compartment in human hepatocellular carcinoma. Carcinogenesis 2013; 34: 2330–2340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. François S, Usunier B, Forgue‐Lafitte M‐E, et al Mesenchymal stem cell administration attenuates colon cancer progression by modulating the immune component within the colorectal tumor microenvironment. Stem Cells Transl Med 2019; 8: 285–300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Wang Q, Li Z, Sun L, et al Platelets enhance the ability of bone‐marrow mesenchymal stem cells to promote cancer metastasis. OncoTargets Ther 2018; 11: 8251–8263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Ishihara S, Inman DR, Li W‐J, et al Mechano‐signal transduction in mesenchymal stem cells induces prosaposin secretion to drive the proliferation of breast cancer cells. Cancer Res 2017; 77: 6179–6189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Nomoto‐Kojima N, Aoki S, Uchihashi K, et al Interaction between adipose tissue stromal cells and gastric cancer cells in vitro . Cell Tissue Res 2011; 344: 287–298. [DOI] [PubMed] [Google Scholar]
  • 38. Zhu Y, Sun Z, Han Q, et al Human mesenchymal stem cells inhibit cancer cell proliferation by secreting DKK‐1. Leukemia 2009; 23: 925–933. [DOI] [PubMed] [Google Scholar]
  • 39. Nishikawa G, Kawada K, Nakagawa J, et al Bone marrow‐derived mesenchymal stem cells promote colorectal cancer progression via CCR5. Cell Death Dis 2019; 10: 264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Li G, Zhang R, Zhang X, et al Human colorectal cancer derived‐MSCs promote tumor cells escape from senescence via P53/P21 pathway. Clin Transl Oncol 2020; 22: 503–511. [DOI] [PubMed] [Google Scholar]
  • 41. Wang Y, Chu Y, Ren X, et al Epidural adipose tissue‐derived mesenchymal stem cell activation induced by lung cancer cells promotes malignancy and EMT of lung cancer. Stem Cell Res Ther 2019; 10: 168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Ullah M, Akbar A, Ng NN, et al Mesenchymal stem cells confer chemoresistance in breast cancer via a CD9 dependent mechanism. Oncotarget 2019; 10: 3435–3450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Hughes RM, Simons BW, Khan H, et al Asporin restricts mesenchymal stromal cell differentiation, alters the tumor microenvironment, and drives metastatic progression. Cancer Res 2019; 79: 3636–3650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Dvorak HF. Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing. N Engl J Med 1986; 315: 1650–1659. [DOI] [PubMed] [Google Scholar]
  • 45. Wood B. Multicolor immunophenotyping: human immune system hematopoiesis. Methods Cell Biol 2004; 75: 559–576. [DOI] [PubMed] [Google Scholar]
  • 46. Watt SM, Gilmore DJ, Davis JM, et al Cell‐surface markers on haemopoietic precursors. Reagents for the isolation and analysis of progenitor cell subpopulations. Mol Cell Probes 1987; 1: 297–326. [DOI] [PubMed] [Google Scholar]
  • 47. Ng AP, Alexander WS. Haematopoietic stem cells: past, present and future. Cell Death Discov 2017; 3: 17002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Kahounová Z, Kurfürstová D, Bouchal J, et al The fibroblast surface markers FAP, anti‐fibroblast, and FSP are expressed by cells of epithelial origin and may be altered during epithelial‐to‐mesenchymal transition. Cytometry A 2018; 93: 941–951. [DOI] [PubMed] [Google Scholar]
  • 49. Kalluri R. The biology and function of fibroblasts in cancer. Nat Rev Cancer 2016; 16: 582–598. [DOI] [PubMed] [Google Scholar]
  • 50. Kalluri R, Zeisberg M. Fibroblasts in cancer. Nat Rev Cancer 2006; 6: 392. [DOI] [PubMed] [Google Scholar]
  • 51. Hinz B. Formation and function of the myofibroblast during tissue repair. J Invest Dermatol 2007; 127: 526–537. [DOI] [PubMed] [Google Scholar]
  • 52. Vong S, Kalluri R. The role of stromal myofibroblast and extracellular matrix in tumor angiogenesis. Genes Cancer 2011; 2: 1139–1145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Öhlund D, Elyada E, Tuveson D. Fibroblast heterogeneity in the cancer wound. J Exp Med 2014; 211: 1503–1523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Dominici M, Le Blanc K, Mueller I, et al Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006; 8: 315–317. [DOI] [PubMed] [Google Scholar]
  • 55. Horwitz EM, Le Blanc K, Dominici M, et al Clarification of the nomenclature for MSC: the International Society for Cellular Therapy position statement. Cytotherapy 2005; 7: 393–395. [DOI] [PubMed] [Google Scholar]
  • 56. Williams AR, Hare JM. Mesenchymal stem cells: biology, pathophysiology, translational findings, and therapeutic implications for cardiac disease. Circ Res 2011; 109: 923–940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Fibbe WE, Noort WA. Mesenchymal stem cells and hematopoietic stem cell transplantation. Ann N Y Acad Sci 2003; 996: 235–244. [DOI] [PubMed] [Google Scholar]
  • 58. Phinney DG, Sensebé L. Mesenchymal stromal cells: misconceptions and evolving concepts. Cytotherapy 2013; 15: 140–145. [DOI] [PubMed] [Google Scholar]
  • 59. Kolf CM, Cho E, Tuan RS. Mesenchymal stromal cells. Biology of adult mesenchymal stem cells: regulation of niche, self‐renewal and differentiation. Arthritis Res Ther 2007; 9: 204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Quirici N, Soligo D, Bossolasco P, et al Isolation of bone marrow mesenchymal stem cells by anti‐nerve growth factor receptor antibodies. Exp Hematol 2002; 30: 783–791. [DOI] [PubMed] [Google Scholar]
  • 61. Delorme B, Ringe J, Gallay N, et al Specific plasma membrane protein phenotype of culture‐amplified and native human bone marrow mesenchymal stem cells. Blood 2008; 111: 2631–2635. [DOI] [PubMed] [Google Scholar]
  • 62. Yang ZX, Han Z‐B, Ji YR, et al CD106 identifies a subpopulation of mesenchymal stem cells with unique immunomodulatory properties. PLoS One 2013; 8: e59354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Halfon S, Abramov N, Grinblat B, et al Markers distinguishing mesenchymal stem cells from fibroblasts are downregulated with passaging. Stem Cells Dev 2011; 20: 53–66. [DOI] [PubMed] [Google Scholar]
  • 64. Sacchetti B, Funari A, Michienzi S, et al Self‐renewing osteoprogenitors in bone marrow sinusoids can organize a hematopoietic microenvironment. Cell 2007; 131: 324–336. [DOI] [PubMed] [Google Scholar]
  • 65. Wirths S, Malenke E, Kluba T, et al Shared cell surface marker expression in mesenchymal stem cells and adult sarcomas. Stem Cells Transl Med 2013; 2: 53–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Gang EJ, Bosnakovski D, Figueiredo CA, et al SSEA‐4 identifies mesenchymal stem cells from bone marrow. Blood 2007; 109: 1743–1751. [DOI] [PubMed] [Google Scholar]
  • 67. Lv F‐J, Tuan RS, Cheung KMC, et al Concise review: the surface markers and identity of human mesenchymal stem cells. Stem Cells 2014; 32: 1408–1419. [DOI] [PubMed] [Google Scholar]
  • 68. Maeda K, Enomoto A, Hara A, et al Identification of Meflin as a potential marker for mesenchymal stromal cells. Sci Rep 2016; 6: 22288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Aggoune D, Sorel N, Bonnet M‐L, et al Bone marrow mesenchymal stromal cell (MSC) gene profiling in chronic myeloid leukemia (CML) patients at diagnosis and in deep molecular response induced by tyrosine kinase inhibitors (TKIs). Leuk Res 2017; 60: 94–102. [DOI] [PubMed] [Google Scholar]
  • 70. Fregni G, Quinodoz M, Möller E, et al Reciprocal modulation of mesenchymal stem cells and tumor cells promotes lung cancer metastasis. EBioMedicine 2018; 29: 128–145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Sneddon JB, Zhen HH, Montgomery K, et al Bone morphogenetic protein antagonist gremlin 1 is widely expressed by cancer‐associated stromal cells and can promote tumor cell proliferation. Proc Natl Acad Sci U S A 2006; 103: 14842–14847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Worthley DL, Churchill M, Compton JT, et al Gremlin 1 identifies a skeletal stem cell with bone, cartilage, and reticular stromal potential. Cell 2015; 160: 269–284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Herrera MB, Bussolati B, Bruno S, et al Exogenous mesenchymal stem cells localize to the kidney by means of CD44 following acute tubular injury. Kidney Int 2007; 72: 430–441. [DOI] [PubMed] [Google Scholar]
  • 74. Shi M, Zhang Z, Xu R, et al Human mesenchymal stem cell transfusion is safe and improves liver function in acute‐on‐chronic liver failure patients. Stem Cells Transl Med 2012; 1: 725–731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Ranganath SH, Levy O, Inamdar MS, et al Harnessing the mesenchymal stem cell secretome for the treatment of cardiovascular disease. Cell Stem Cell 2012; 10: 244–258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. DiMarino AM, Caplan AI, Bonfield TL. Mesenchymal stem cells in tissue repair. Front Immunol 2013; 4: 201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Hill BS, Pelagalli A, Passaro N, et al Tumor‐educated mesenchymal stem cells promote pro‐metastatic phenotype. Oncotarget 2017; 8: 73296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Lee H, Hong I. Double‐edged sword of mesenchymal stem cells: cancer‐promoting versus therapeutic potential. Cancer Sci 2017; 108: 1939–1946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Qi K, Li N, Zhang Z, et al Tissue regeneration: the crosstalk between mesenchymal stem cells and immune response. Cell Immunol 2018; 326: 86–93. [DOI] [PubMed] [Google Scholar]
  • 80. Ma S, Xie N, Li W, et al Immunobiology of mesenchymal stem cells. Cell Death Differ 2014; 21: 216–225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Shi Y, Su J, Roberts AI, et al How mesenchymal stem cells interact with tissue immune responses. Trends Immunol 2012; 33: 136–143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Lee JW, Fang X, Krasnodembskaya A, et al Concise review: mesenchymal stem cells for acute lung injury: role of paracrine soluble factors. Stem Cells 2011; 29: 913–919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Krasnodembskaya A, Song Y, Fang X, et al Antibacterial effect of human mesenchymal stem cells is mediated in part from secretion of the antimicrobial peptide LL‐37. Stem Cells 2010; 28: 2229–2238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Abdallah BM, Kassem M. Human mesenchymal stem cells: from basic biology to clinical applications. Gene Ther 2008; 15: 109–116. [DOI] [PubMed] [Google Scholar]
  • 85. Bader P, Kuçi Z, Bakhtiar S, et al Effective treatment of steroid and therapy‐refractory acute graft‐versus‐host disease with a novel mesenchymal stromal cell product (MSC‐FFM). Bone Marrow Transplant 2018; 53: 852–862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Caplan AI. Why are MSCs therapeutic? New data: new insight. J Pathol 2009; 217: 318–324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Kidd S, Spaeth E, Dembinski JL, et al Direct evidence of mesenchymal stem cell tropism for tumor and wounding microenvironments using in vivo bioluminescent imaging. Stem Cells 2009; 27: 2614–2623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Puré E, Lo A. Can targeting stroma pave the way to enhanced antitumor immunity and immunotherapy of solid tumors? Cancer Immunol Res 2016; 4: 269–278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Hong I‐S, Lee H‐Y, Kang K‐S. Mesenchymal stem cells and cancer: friends or enemies? Mutat Res 2014; 768: 98–106. [DOI] [PubMed] [Google Scholar]
  • 90. Klopp AH, Gupta A, Spaeth E, et al Concise review: dissecting a discrepancy in the literature: do mesenchymal stem cells support or suppress tumor growth? Stem Cells 2011; 29: 11–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Sun Z, Wang S, Zhao RC. The roles of mesenchymal stem cells in tumor inflammatory microenvironment. J Hematol Oncol 2014; 7: 14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Jing Y, Han Z, Liu Y, et al Mesenchymal stem cells in inflammation microenvironment accelerates hepatocellular carcinoma metastasis by inducing epithelial–mesenchymal transition. PLoS One 2012; 7: e43272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Liu Y, Han Z, Zhang S, et al Effects of inflammatory factors on mesenchymal stem cells and their role in the promotion of tumor angiogenesis in colon cancer. J Biol Chem 2011; 286: 25007–25015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Ren G, Zhao X, Wang Y, et al CCR2‐dependent recruitment of macrophages by tumor‐educated mesenchymal stromal cells promotes tumor development and is mimicked by TNFα. Cell Stem Cell 2012; 11: 812–824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Cho JA, Park H, Lim EH, et al Exosomes from ovarian cancer cells induce adipose tissue‐derived mesenchymal stem cells to acquire the physical and functional characteristics of tumor‐supporting myofibroblasts. Gynecol Oncol 2011; 123: 379–386. [DOI] [PubMed] [Google Scholar]
  • 96. Dostert G, Mesure B, Menu P, et al How do mesenchymal stem cells influence or are influenced by microenvironment through extracellular vesicles communication? Front Cell Dev Biol 2017; 5: 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Raz Y, Cohen N, Shani O, et al Bone marrow‐derived fibroblasts are a functionally distinct stromal cell population in breast cancer. J Exp Med 2018; 215: 3075–3093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Brune JC, Tormin A, Johansson MC, et al Mesenchymal stromal cells from primary osteosarcoma are non‐malignant and strikingly similar to their bone marrow counterparts. Int J Cancer 2011; 129: 319–330. [DOI] [PubMed] [Google Scholar]
  • 99. McLean K, Gong Y, Choi Y, et al Human ovarian carcinoma‐associated mesenchymal stem cells regulate cancer stem cells and tumorigenesis via altered BMP production. J Clin Invest 2011; 121: 3206–3219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Gottschling S, Granzow M, Kuner R, et al Mesenchymal stem cells in non‐small cell lung cancer – different from others? Insights from comparative molecular and functional analyses. Lung Cancer 2013; 80: 19–29. [DOI] [PubMed] [Google Scholar]
  • 101. Johann P‐D, Vaegler M, Gieseke F, et al Tumour stromal cells derived from paediatric malignancies display MSC‐like properties and impair NK cell cytotoxicity. BMC Cancer 2010; 10: 501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Xu X, Zhang X, Wang S, et al Isolation and comparison of mesenchymal stem‐like cells from human gastric cancer and adjacent non‐cancerous tissues. J Cancer Res Clin Oncol 2011; 137: 495–504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Galland S, Vuille J, Martin P, et al Tumor‐derived mesenchymal stem cells use distinct mechanisms to block the activity of natural killer cell subsets. Cell Rep 2017; 20: 2891–2905. [DOI] [PubMed] [Google Scholar]
  • 104. Razmkhah M, Jaberipour M, Erfani N, et al Adipose derived stem cells (ASCs) isolated from breast cancer tissue express IL‐4, IL‐10 and TGF‐β1 and upregulate expression of regulatory molecules on T cells: do they protect breast cancer cells from the immune response? Cell Immunol 2011; 266: 116–122. [DOI] [PubMed] [Google Scholar]
  • 105. Razmkhah M, Mansourabadi Z, Mohtasebi MS, et al Cancer and normal adipose‐derived mesenchymal stem cells (ASCs): do they have differential effects on tumor and immune cells? Cell Biol Int 2018; 42: 334–343. [DOI] [PubMed] [Google Scholar]
  • 106. Razmkhah M, Abtahi S, Ghaderi A. Mesenchymal stem cells, immune cells and tumor cells crosstalk: a sinister triangle in the tumor microenvironment. Curr Stem Cell Res Ther 2019; 14: 43–51. [DOI] [PubMed] [Google Scholar]
  • 107. Ren G, Chen X, Dong F, et al Concise review: mesenchymal stem cells and translational medicine: emerging issues. Stem Cells Transl Med 2012; 1: 51–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Luo J, Ok Lee S, Liang L, et al Infiltrating bone marrow mesenchymal stem cells increase prostate cancer stem cell population and metastatic ability via secreting cytokines to suppress androgen receptor signaling. Oncogene 2014; 33: 2768–2778. [DOI] [PubMed] [Google Scholar]
  • 109. Burrello J, Monticone S, Gai C, et al Stem cell‐derived extracellular vesicles and immune‐modulation. Front Cell Dev Biol 2016; 4: 83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Di Trapani M, Bassi G, Midolo M, et al Differential and transferable modulatory effects of mesenchymal stromal cell‐derived extracellular vesicles on T, B and NK cell functions. Sci Rep 2016; 6: 24120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Hass R, Kasper C, Böhm S, et al Different populations and sources of human mesenchymal stem cells (MSC): a comparison of adult and neonatal tissue‐derived MSC. Cell Commun Signal 2011; 9: 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Turley SJ, Cremasco V, Astarita JL. Immunological hallmarks of stromal cells in the tumour microenvironment. Nat Rev Immunol 2015; 15: 669–682. [DOI] [PubMed] [Google Scholar]
  • 113. Tabera S, Pérez‐Simón JA, Díez‐Campelo M, et al The effect of mesenchymal stem cells on the viability, proliferation and differentiation of B‐lymphocytes. Haematologica 2008; 93: 1301–1309. [DOI] [PubMed] [Google Scholar]
  • 114. Raffaghello L, Bianchi G, Bertolotto M, et al Human mesenchymal stem cells inhibit neutrophil apoptosis: a model for neutrophil preservation in the bone marrow niche. Stem Cells 2008; 26: 151–162. [DOI] [PubMed] [Google Scholar]
  • 115. Chen W, Huang Y, Han J, et al Immunomodulatory effects of mesenchymal stromal cells‐derived exosome. Immunol Res 2016; 64: 831–840. [DOI] [PubMed] [Google Scholar]
  • 116. Casado JG, Tarazona R, Sanchez‐Margallo FM. NK and MSCs crosstalk: the sense of immunomodulation and their sensitivity. Stem Cell Rev 2013; 9: 184–189. [DOI] [PubMed] [Google Scholar]
  • 117. Hu C‐HD, Kosaka Y, Marcus P, et al Differential immunomodulatory effects of human bone marrow‐derived mesenchymal stromal cells on natural killer cells. Stem Cells Dev 2019; 28: 933–943. [DOI] [PubMed] [Google Scholar]
  • 118. Krampera M, Cosmi L, Angeli R, et al Role for interferon‐gamma in the immunomodulatory activity of human bone marrow mesenchymal stem cells. Stem Cells 2006; 24: 386–398. [DOI] [PubMed] [Google Scholar]
  • 119. Rasmusson I, Ringdén O, Sundberg B, et al Mesenchymal stem cells inhibit the formation of cytotoxic T lymphocytes, but not activated cytotoxic T lymphocytes or natural killer cells. Transplantation 2003; 76: 1208–1213. [DOI] [PubMed] [Google Scholar]
  • 120. Spaggiari GM, Capobianco A, Abdelrazik H, et al Mesenchymal stem cells inhibit natural killer‐cell proliferation, cytotoxicity, and cytokine production: role of indoleamine 2,3‐dioxygenase and prostaglandin E2. Blood 2008; 111: 1327–1333. [DOI] [PubMed] [Google Scholar]
  • 121. Poggi A, Prevosto C, Massaro A‐M, et al Interaction between human NK cells and bone marrow stromal cells induces NK cell triggering: role of NKp30 and NKG2D receptors. J Immunol 2005; 175: 6352–6360. [DOI] [PubMed] [Google Scholar]
  • 122. Spaggiari GM, Capobianco A, Becchetti S, et al Mesenchymal stem cell–natural killer cell interactions: evidence that activated NK cells are capable of killing MSCs, whereas MSCs can inhibit IL‐2‐induced NK‐cell proliferation. Blood 2006; 107: 1484–1490. [DOI] [PubMed] [Google Scholar]
  • 123. Pereira RC, Martinelli D, Cancedda R, et al Human articular chondrocytes regulate immune response by affecting directly T cell proliferation and indirectly inhibiting monocyte differentiation to professional antigen‐presenting cells. Front Immunol 2016; 7: 415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. François M, Romieu‐Mourez R, Li M, et al Human MSC suppression correlates with cytokine induction of indoleamine 2,3‐dioxygenase and bystander M2 macrophage differentiation. Mol Ther 2012; 20: 187–195. [DOI] [PubMed] [Google Scholar]
  • 125. Németh K, Leelahavanichkul A, Yuen PST, et al Bone marrow stromal cells attenuate sepsis via prostaglandin E2‐dependent reprogramming of host macrophages to increase their interleukin‐10 production. Nat Med 2009; 15: 42–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Philipp D, Suhr L, Wahlers T, et al Preconditioning of bone marrow‐derived mesenchymal stem cells highly strengthens their potential to promote IL‐6‐dependent M2b polarization. Stem Cell Res Ther 2018; 9: 286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Chabannes D, Hill M, Merieau E, et al A role for heme oxygenase‐1 in the immunosuppressive effect of adult rat and human mesenchymal stem cells. Blood 2007; 110: 3691–3694. [DOI] [PubMed] [Google Scholar]
  • 128. De Miguel MP, Fuentes‐Julián S, Blázquez‐Martínez A, et al Immunosuppressive properties of mesenchymal stem cells: advances and applications. Curr Mol Med 2012; 12: 574–591. [DOI] [PubMed] [Google Scholar]
  • 129. Nasef A, Chapel A, Mazurier C, et al Identification of IL‐10 and TGF‐β transcripts involved in the inhibition of T‐lymphocyte proliferation during cell contact with human mesenchymal stem cells. Gene Expr 2007; 13: 217–226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Najar M, Fayyad‐Kazan H, Faour WH, et al Immunological modulation following bone marrow‐derived mesenchymal stromal cells and Th17 lymphocyte co‐cultures. Inflamm Res 2019; 68: 203–213. [DOI] [PubMed] [Google Scholar]
  • 131. Zimmermann JA, Hettiaratchi MH, McDevitt TC. Enhanced immunosuppression of T cells by sustained presentation of bioactive interferon‐γ within three‐dimensional mesenchymal stem cell constructs. Stem Cells Transl Med 2017; 6: 223–237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Asari S, Itakura S, Ferreri K, et al Mesenchymal stem cells suppress B‐cell terminal differentiation. Exp Hematol 2009; 37: 604–615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Corcione A, Benvenuto F, Ferretti E, et al Human mesenchymal stem cells modulate B‐cell functions. Blood 2006; 107: 367–372. [DOI] [PubMed] [Google Scholar]
  • 134. Martinet L, Fleury‐Cappellesso S, Gadelorge M, et al A regulatory cross‐talk between Vγ9Vδ2 T lymphocytes and mesenchymal stem cells. Eur J Immunol 2009; 39: 752–762. [DOI] [PubMed] [Google Scholar]
  • 135. Aggarwal S, Pittenger MF. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood 2005; 105: 1815–1822. [DOI] [PubMed] [Google Scholar]
  • 136. Beyth S, Borovsky Z, Mevorach D, et al Human mesenchymal stem cells alter antigen‐presenting cell maturation and induce T‐cell unresponsiveness. Blood 2005; 105: 2214–2219. [DOI] [PubMed] [Google Scholar]
  • 137. Nauta AJ, Kruisselbrink AB, Lurvink E, et al Mesenchymal stem cells inhibit generation and function of both CD34+‐derived and monocyte‐derived dendritic cells. J Immunol 2006; 177: 2080–2087. [DOI] [PubMed] [Google Scholar]
  • 138. Zhang B, Liu R, Shi D, et al Mesenchymal stem cells induce mature dendritic cells into a novel Jagged‐2‐dependent regulatory dendritic cell population. Blood 2009; 113: 46–57. [DOI] [PubMed] [Google Scholar]
  • 139. Ren G, Zhao X, Zhang L, et al Inflammatory cytokine‐induced intercellular adhesion molecule‐1 and vascular cell adhesion molecule‐1 in mesenchymal stem cells are critical for immunosuppression. J Immunol 2010; 184: 2321–2328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Li N, Hua J. Interactions between mesenchymal stem cells and the immune system. Cell Mol Life Sci 2017; 74: 2345–2360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Opitz CA, Litzenburger UM, Lutz C, et al Toll‐like receptor engagement enhances the immunosuppressive properties of human bone marrow‐derived mesenchymal stem cells by inducing indoleamine‐2,3‐dioxygenase‐1 via interferon‐beta and protein kinase R. Stem Cells 2009; 27: 909–919. [DOI] [PubMed] [Google Scholar]
  • 142. Liotta F, Angeli R, Cosmi L, et al Toll‐like receptors 3 and 4 are expressed by human bone marrow‐derived mesenchymal stem cells and can inhibit their T‐cell modulatory activity by impairing Notch signaling. Stem Cells 2008; 26: 279–289. [DOI] [PubMed] [Google Scholar]
  • 143. Krampera M, Galipeau J, Shi Y, et al Immunological characterization of multipotent mesenchymal stromal cells – The International Society for Cellular Therapy (ISCT) working proposal. Cytotherapy 2013; 15: 1054–1061. [DOI] [PubMed] [Google Scholar]
  • 144. Le Blanc K, Davies LC. Mesenchymal stromal cells and the innate immune response. Immunol Lett 2015; 168: 140–146. [DOI] [PubMed] [Google Scholar]
  • 145. Poggi A, Musso A, Dapino I, et al Mechanisms of tumor escape from immune system: role of mesenchymal stromal cells. Immunol Lett 2014; 159: 55–72. [DOI] [PubMed] [Google Scholar]
  • 146. Spaggiari GM, Moretta L. Cellular and molecular interactions of mesenchymal stem cells in innate immunity. Immunol Cell Biol 2013; 91: 27–31. [DOI] [PubMed] [Google Scholar]
  • 147. Spaggiari GM, Moretta L. Interactions between mesenchymal stem cells and dendritic cells. Adv Biochem Eng Biotechnol 2013; 130: 199–208. [DOI] [PubMed] [Google Scholar]
  • 148. Stagg J, Galipeau J. Mechanisms of immune modulation by mesenchymal stromal cells and clinical translation. Curr Mol Med 2013; 13: 856–867. [DOI] [PubMed] [Google Scholar]
  • 149. Uccelli A, Moretta L, Pistoia V. Immunoregulatory function of mesenchymal stem cells. Eur J Immunol 2006; 36: 2566–2573. [DOI] [PubMed] [Google Scholar]
  • 150. Wang Y, Chen X, Cao W, et al Plasticity of mesenchymal stem cells in immunomodulation: pathological and therapeutic implications. Nat Immunol 2014; 15: 1009–1016. [DOI] [PubMed] [Google Scholar]
  • 151. Cho HH, Bae YC, Jung JS. Role of Toll‐like receptors on human adipose‐derived stromal cells. Stem Cells 2006; 24: 2744–2752. [DOI] [PubMed] [Google Scholar]
  • 152. Le Blanc K, Mougiakakos D. Multipotent mesenchymal stromal cells and the innate immune system. Nat Rev Immunol 2012; 12: 383–396. [DOI] [PubMed] [Google Scholar]
  • 153. Tu Z, Li Q, Bu H, et al Mesenchymal stem cells inhibit complement activation by secreting factor H. Stem Cells Dev 2010; 19: 1803–1809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154. Tomchuck SL, Zwezdaryk KJ, Coffelt SB, et al Toll‐like receptors on human mesenchymal stem cells drive their migration and immunomodulating responses. Stem Cells 2008; 26: 99–107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. Nemeth K, Wilson T, Rada B, et al Characterization and function of histamine receptors in human bone marrow stromal cells. Stem Cells 2012; 30: 222–231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. Chen H‐W, Chen H‐Y, Wang L‐T, et al Mesenchymal stem cells tune the development of monocyte‐derived dendritic cells toward a myeloid‐derived suppressive phenotype through growth‐regulated oncogene chemokines. J Immunol 2013; 190: 5065–5077. [DOI] [PubMed] [Google Scholar]
  • 157. Yen BL, Yen M‐L, Hsu P‐J, et al Multipotent human mesenchymal stromal cells mediate expansion of myeloid‐derived suppressor cells via hepatocyte growth factor/c‐Met and STAT3. Stem Cell Reports 2013; 1: 139–151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Paul S, Lal G. Regulatory and effector functions of gamma‐delta (γδ) T cells and their therapeutic potential in adoptive cellular therapy for cancer. Int J Cancer 2016; 139: 976–985. [DOI] [PubMed] [Google Scholar]
  • 159. Ren C, Kumar S, Chanda D, et al Cancer gene therapy using mesenchymal stem cells expressing interferon‐beta in a mouse prostate cancer lung metastasis model. Gene Ther 2008; 15: 1446–1453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160. Phinney DG, Pittenger MF. Concise review: MSC‐derived exosomes for cell‐free therapy. Stem Cells 2017; 35: 851–858. [DOI] [PubMed] [Google Scholar]
  • 161. Zhang B, Shen L, Shi H, et al Exosomes from human umbilical cord mesenchymal stem cells: identification, purification, and biological characteristics. Stem Cells Int 2016; 2016: 1929536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162. Bernardo ME, Fibbe WE. Mesenchymal stromal cells: sensors and switchers of inflammation. Cell Stem Cell 2013; 13: 392–402. [DOI] [PubMed] [Google Scholar]
  • 163. Dumitru CA, Hemeda H, Jakob M, et al Stimulation of mesenchymal stromal cells (MSCs) via TLR3 reveals a novel mechanism of autocrine priming. FASEB J 2014; 28: 3856–3866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164. Krampera M. Mesenchymal stromal cell ‘licensing’: a multistep process. Leukemia 2011; 25: 1408–1414. [DOI] [PubMed] [Google Scholar]
  • 165. Glenn JD, Whartenby KA. Mesenchymal stem cells: emerging mechanisms of immunomodulation and therapy. World J Stem Cells 2014; 6: 526–539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166. Ayala‐Cuellar AP, Kang J‐H, Jeung E‐B, et al Roles of mesenchymal stem cells in tissue regeneration and immunomodulation. Biomol Ther 2019; 27: 25–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167. Lu Y, Liu J, Liu Y, et al TLR4 plays a crucial role in MSC‐induced inhibition of NK cell function. Biochem Biophys Res Commun 2015; 464: 541–547. [DOI] [PubMed] [Google Scholar]
  • 168. Raicevic G, Rouas R, Najar M, et al Inflammation modifies the pattern and the function of Toll‐like receptors expressed by human mesenchymal stromal cells. Hum Immunol 2010; 71: 235–244. [DOI] [PubMed] [Google Scholar]
  • 169. Raicevic G, Najar M, Stamatopoulos B, et al The source of human mesenchymal stromal cells influences their TLR profile as well as their functional properties. Cell Immunol 2011; 270: 207–216. [DOI] [PubMed] [Google Scholar]
  • 170. Sallustio F, Curci C, Stasi A, et al Role of Toll‐like receptors in actuating stem/progenitor cell repair mechanisms: different functions in different cells. Stem Cells Int 2019; 2019: 6795845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171. Shirjang S, Mansoori B, Solali S, et al Toll‐like receptors as a key regulator of mesenchymal stem cell function: an up‐to‐date review. Cell Immunol 2017; 315: 1–10. [DOI] [PubMed] [Google Scholar]
  • 172. Hou R, Li J, Niu X, et al Stem cells in psoriasis. J Dermatol Sci 2017; 86: 181–186. [DOI] [PubMed] [Google Scholar]
  • 173. Waterman RS, Henkle SL, Betancourt AM. Mesenchymal stem cell 1 (MSC1)‐based therapy attenuates tumor growth whereas MSC2‐treatment promotes tumor growth and metastasis. PLoS One 2012; 7: e45590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174. Simones AA, Beisang DJ, Panoskaltsis‐Mortari A, et al Mesenchymal stem cells in the pathogenesis and treatment of bronchopulmonary dysplasia: a clinical review. Pediatr Res 2018; 83: 308–317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175. Waterman RS, Tomchuck SL, Henkle SL, et al A new mesenchymal stem cell (MSC) paradigm: polarization into a pro‐inflammatory MSC1 or an immunosuppressive MSC2 phenotype. PLoS One 2010; 5: e10088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176. Li W, Ren G, Huang Y, et al Mesenchymal stem cells: a double‐edged sword in regulating immune responses. Cell Death Differ 2012; 19: 1505–1513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Giuliani M, Bennaceur‐Griscelli A, Nanbakhsh A, et al TLR ligands stimulation protects MSC from NK killing. Stem Cells 2014; 32: 290–300. [DOI] [PubMed] [Google Scholar]
  • 178. Jiang M, Gao T, Liu Y. et al. CD14 dictates differential activation of mesenchymal stromal cells through AKT, NF‐κB and P38 signals. Biosci Rep 2019; 39: pii: BSR20190807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179. Romieu‐Mourez R, François M, Boivin M‐N, et al Cytokine modulation of TLR expression and activation in mesenchymal stromal cells leads to a proinflammatory phenotype. J Immunol 2009; 182: 7963–7973. [DOI] [PubMed] [Google Scholar]
  • 180. Rashedi I, Gómez‐Aristizábal A, Wang X‐H, et al TLR3 or TLR4 activation enhances mesenchymal stromal cell‐mediated Treg induction via Notch signaling. Stem Cells 2017; 35: 265–275. [DOI] [PubMed] [Google Scholar]
  • 181. Cassatella MA, Mosna F, Micheletti A, et al Toll‐like receptor‐3‐activated human mesenchymal stromal cells significantly prolong the survival and function of neutrophils. Stem Cells 2011; 29: 1001–1011. [DOI] [PubMed] [Google Scholar]
  • 182. Kota DJ, DiCarlo B, Hetz RA, et al Differential MSC activation leads to distinct mononuclear leukocyte binding mechanisms. Sci Rep 2014; 4: 4565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183. Zhao X, Liu D, Gong W, et al The Toll‐like receptor 3 ligand, poly(I:C), improves immunosuppressive function and therapeutic effect of mesenchymal stem cells on sepsis via inhibiting MiR‐143. Stem Cells 2014; 32: 521–533. [DOI] [PubMed] [Google Scholar]
  • 184. Petri RM, Hackel A, Hahnel K, et al Activated tissue‐resident mesenchymal stromal cells regulate natural killer cell immune and tissue‐regenerative function. Stem Cell Reports 2017; 9: 985–998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185. Najar M, Krayem M, Merimi M, et al Insights into inflammatory priming of mesenchymal stromal cells: functional biological impacts. Inflamm Res 2018; 67: 467–477. [DOI] [PubMed] [Google Scholar]
  • 186. Cao W, Cao K, Cao J, et al Mesenchymal stem cells and adaptive immune responses. Immunol Lett 2015; 168: 147–153. [DOI] [PubMed] [Google Scholar]
  • 187. Xu C, Yu P, Han X, et al TGF‐β promotes immune responses in the presence of mesenchymal stem cells. J Immunol 2014; 192: 103–109. [DOI] [PubMed] [Google Scholar]
  • 188. Adamo A, Dal Collo G, Bazzoni R, et al Role of mesenchymal stromal cell‐derived extracellular vesicles in tumour microenvironment. Biochim Biophys Acta Rev Cancer 2019; 1871: 192–198. [DOI] [PubMed] [Google Scholar]
  • 189. Whiteside TL. Exosome and mesenchymal stem cell cross‐talk in the tumor microenvironment. Semin Immunol 2018; 35: 69–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190. Lobb RJ, van Amerongen R, Wiegmans A, et al Exosomes derived from mesenchymal non‐small cell lung cancer cells promote chemoresistance. Int J Cancer 2017; 141: 614–620. [DOI] [PubMed] [Google Scholar]
  • 191. Gentile P, Garcovich S. Concise review: adipose‐derived stem cells (ASCs) and adipocyte‐secreted exosomal microRNA (A‐SE‐miR) modulate cancer growth and promote wound repair. J Clin Med 2019; 8: 855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192. Samuel P, Fabbri M, Carter DRF. Mechanisms of drug resistance in cancer: the role of extracellular vesicles. Proteomics 2017; 17: 1600375. [DOI] [PubMed] [Google Scholar]
  • 193. Zhu W, Huang L, Li Y, et al Exosomes derived from human bone marrow mesenchymal stem cells promote tumor growth in vivo . Cancer Lett 2012; 315: 28–37. [DOI] [PubMed] [Google Scholar]
  • 194. Kalluri R. The biology and function of exosomes in cancer. J Clin Invest 2016; 126: 1208–1215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195. Mardpour S, Hamidieh AA, Taleahmad S, et al Interaction between mesenchymal stromal cell‐derived extracellular vesicles and immune cells by distinct protein content. J Cell Physiol 2019; 234: 8249–8258. [DOI] [PubMed] [Google Scholar]
  • 196. Robbins PD, Morelli AE. Regulation of immune responses by extracellular vesicles. Nat Rev Immunol 2014; 14: 195–208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197. Khare D, Or R, Resnick I, et al Mesenchymal stromal cell‐derived exosomes affect mRNA expression and function of B‐lymphocytes. Front Immunol 2018; 9: 3053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198. Zhang Q, Fu L, Liang Y, et al Exosomes originating from MSCs stimulated with TGF‐β and IFN‐γ promote Treg differentiation. J Cell Physiol 2018; 233: 6832–6840. [DOI] [PubMed] [Google Scholar]
  • 199. Zhang B, Yin Y, Lai RC, et al Mesenchymal stem cells secrete immunologically active exosomes. Stem Cells Dev 2014; 23: 1233–1244. [DOI] [PubMed] [Google Scholar]
  • 200. Whiteside TL. Exosomes and tumor‐mediated immune suppression. J Clin Invest 2016; 126: 1216–1223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201. Whiteside TL. Tumor‐derived exosomes and their role in cancer progression. Adv Clin Chem 2016; 74: 103–141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202. Fisher DT, Appenheimer MM, Evans SS. The two faces of IL‐6 in the tumor microenvironment. Semin Immunol 2014; 26: 38–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203. Tanaka T, Narazaki M, Kishimoto T. IL‐6 in inflammation, immunity, and disease. Cold Spring Harb Perspect Biol 2014; 6: a016295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204. Avnet S, Lemma S, Cortini M, et al Pre‐clinical models for studying the interaction between mesenchymal stromal cells and cancer cells and the induction of stemness. Front Oncol 2019; 9: 305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205. Cortini M, Massa A, Avnet S, et al Tumor‐activated mesenchymal stromal cells promote osteosarcoma stemness and migratory potential via IL‐6 secretion. PLoS One 2016; 11: e0166500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206. Bouffi C, Bony C, Courties G, et al IL‐6‐dependent PGE2 secretion by mesenchymal stem cells inhibits local inflammation in experimental arthritis. PLoS One 2010; 5: e14247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207. Melief SM, Geutskens SB, Fibbe WE, et al Multipotent stromal cells skew monocytes towards an anti‐inflammatory interleukin‐10‐producing phenotype by production of interleukin‐6. Haematologica 2013; 98: 888–895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208. Li J, Xu J, Yan X, et al Targeting interleukin‐6 (IL‐6) sensitizes anti‐PD‐L1 treatment in a colorectal cancer preclinical model. Med Sci Monit 2018; 24: 5501–5508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209. Liu H, Shen J, Lu K. IL‐6 and PD‐L1 blockade combination inhibits hepatocellular carcinoma cancer development in mouse model. Biochem Biophys Res Commun 2017; 486: 239–244. [DOI] [PubMed] [Google Scholar]
  • 210. Mace TA, Shakya R, Pitarresi JR, et al IL‐6 and PD‐L1 antibody blockade combination therapy reduces tumour progression in murine models of pancreatic cancer. Gut 2018; 67: 320–332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211. Caplan AI, Correa D. The MSC: an injury drugstore. Cell Stem Cell 2011; 9: 11–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212. Galipeau J, Sensébé L. Mesenchymal stromal cells: clinical challenges and therapeutic opportunities. Cell Stem Cell 2018; 22: 824–833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213. Bartunek J, Davison B, Sherman W, et al Congestive heart failure cardiopoietic regenerative therapy (CHART‐1) trial design. Eur J Heart Fail 2016; 18: 160–168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214. Bianco P, Cao X, Frenette PS, et al The meaning, the sense and the significance: translating the science of mesenchymal stem cells into medicine. Nat Med 2013; 19: 35–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215. Fibbe WE, Dazzi F, LeBlanc K. MSCs: science and trials. Nat Med 2013; 19: 812–813. [DOI] [PubMed] [Google Scholar]
  • 216. Chulpanova DS, Kitaeva KV, Tazetdinova LG, et al Application of mesenchymal stem cells for therapeutic agent delivery in anti‐tumor treatment. Front Pharmacol 2018; 9: 259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217. Mohr A, Zwacka R. The future of mesenchymal stem cell‐based therapeutic approaches for cancer – from cells to ghosts. Cancer Lett 2018; 414: 239–249. [DOI] [PubMed] [Google Scholar]
  • 218. Sage EK, Thakrar RM, Janes SM. Genetically modified mesenchymal stromal cells in cancer therapy. Cytotherapy 2016; 18: 1435–1445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219. Pereboeva L, Komarova S, Mikheeva G, et al Approaches to utilize mesenchymal progenitor cells as cellular vehicles. Stem Cells 2003; 21: 389–404. [DOI] [PubMed] [Google Scholar]
  • 220. Power AT, Bell JC. Cell‐based delivery of oncolytic viruses: a new strategic alliance for a biological strike against cancer. Mol Ther 2007; 15: 660–665. [DOI] [PubMed] [Google Scholar]
  • 221. Sasportas LS, Kasmieh R, Wakimoto H, et al Assessment of therapeutic efficacy and fate of engineered human mesenchymal stem cells for cancer therapy. Proc Natl Acad Sci U S A 2009; 106: 4822–4827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222. Pessina A, Bonomi A, Coccè V, et al Mesenchymal stromal cells primed with paclitaxel provide a new approach for cancer therapy. PLoS One 2011; 6: e28321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223. Petrella F, Rimoldi I, Rizzo S, et al Mesenchymal stromal cells for antineoplastic drug loading and delivery. Medicines 2017; 4: pii: E87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224. Shah K. Mesenchymal stem cells engineered for cancer therapy. Adv Drug Deliv Rev 2012; 64: 739–748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225. Christodoulou I, Goulielmaki M, Devetzi M, et al Mesenchymal stem cells in preclinical cancer cytotherapy: a systematic review. Stem Cell Res Ther 2018; 9: 336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226. Hood JL, San RS, Wickline SA. Exosomes released by melanoma cells prepare sentinel lymph nodes for tumor metastasis. Cancer Res 2011; 71: 3792–3801. [DOI] [PubMed] [Google Scholar]
  • 227. Rani S, Ryan AE, Griffin MD, et al Mesenchymal stem cell‐derived extracellular vesicles: toward cell‐free therapeutic applications. Mol Ther 2015; 23: 812–823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228. Reis M, Mavin E, Nicholson L, et al Mesenchymal stromal cell‐derived extracellular vesicles attenuate dendritic cell maturation and function. Front Immunol 2018; 9: 2538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229. Ferguson SW, Wang J, Lee CJ, et al The microRNA regulatory landscape of MSC‐derived exosomes: a systems view. Sci Rep 2018; 8: 1419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230. Mendt M, Kamerkar S, Sugimoto H, et al Generation and testing of clinical‐grade exosomes for pancreatic cancer. JCI Insight; 3: pii: 99263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231. Valadi H, Ekström K, Bossios A, et al Exosome‐mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol 2007; 9: 654–659. [DOI] [PubMed] [Google Scholar]
  • 232. Zhou J, Tan X, Tan Y, et al Mesenchymal stem cell derived exosomes in cancer progression, metastasis and drug delivery: a comprehensive review. J Cancer 2018; 9: 3129–3137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233. Borghese C, Cattaruzza L, Pivetta E, et al Gefitinib inhibits the cross‐talk between mesenchymal stem cells and prostate cancer cells leading to tumor cell proliferation and inhibition of docetaxel activity. J Cell Biochem 2013; 114: 1135–1144. [DOI] [PubMed] [Google Scholar]
  • 234. Borriello L, Nakata R, Sheard MA, et al Cancer‐associated fibroblasts share characteristics and protumorigenic activity with mesenchymal stromal cells. Cancer Res 2017; 77: 5142–5157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235. Fierro F, Illmer T, Jing D, et al Inhibition of platelet‐derived growth factor receptor beta by imatinib mesylate suppresses proliferation and alters differentiation of human mesenchymal stem cells in vitro . Cell Prolif 2007; 40: 355–366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236. Park N, Rim YA, Jung H, et al Etanercept‐synthesising mesenchymal stem cells efficiently ameliorate collagen‐induced arthritis. Sci Rep 2017; 7: 39593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237. Davies LC, Heldring N, Kadri N, et al Mesenchymal stromal cell secretion of programmed death‐1 ligands regulates T cell mediated immunosuppression. Stem Cells 2017; 35: 766–776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238. Chinnadurai R, Copland IB, Patel SR, et al IDO‐independent suppression of T cell effector function by IFN‐γ‐licensed human mesenchymal stromal cells. J Immunol 2014; 192: 1491–1501. [DOI] [PubMed] [Google Scholar]
  • 239. Liu O, Xu J, Ding G, et al Periodontal ligament stem cells regulate B lymphocyte function via programmed cell death protein 1. Stem Cells 2013; 31: 1371–1382. [DOI] [PubMed] [Google Scholar]
  • 240. Musso A, Catellani S, Canevali P, et al Aminobisphosphonates prevent the inhibitory effects exerted by lymph node stromal cells on anti‐tumor Vδ 2 T lymphocytes in non‐Hodgkin lymphomas. Haematologica 2014; 99: 131–139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241. Zocchi MR, Costa D, Venè R, et al Zoledronate can induce colorectal cancer microenvironment expressing BTN3A1 to stimulate effector γδ T cells with antitumor activity. Oncoimmunology 2017; 6: e1278099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242. Görgün G, Calabrese E, Soydan E, et al Immunomodulatory effects of lenalidomide and pomalidomide on interaction of tumor and bone marrow accessory cells in multiple myeloma. Blood 2010; 116: 3227–3237. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Table S1. Complete list of abbreviations used


Articles from The Journal of Pathology are provided here courtesy of Wiley

RESOURCES