You are on page 1of 12

Trends in Pharmacological Sciences

Review

Mesenchymal Stem Cell Immunomodulation:


Mechanisms and Therapeutic Potential
Na Song,1,2,3 Martijn Scholtemeijer,1,2 and Khalid Shah1,2,4,*

Mesenchymal stem/stromal cells (MSCs) are multipotent cells that are emerging Highlights
as the most promising means of allogeneic cell therapy. MSCs have inherent MSCs are multipotent cells that are
immunomodulatory characteristics, trophic activity, high in vitro self-renewal emerging as the most promising means
of allogeneic cell therapy.
ability, and can be readily engineered to enhance their immunomodulatory func-
tions. MSCs affect the functions of most immune effector cells via direct contact MSCs participate in both innate and
with immune cells and local microenvironmental factors. Previous studies have adaptive immunity, and their immuno-
confirmed that the immunomodulatory effects of MSCs are mainly communi- modulatory functions are exerted mainly
via interactions with immune cells
cated via MSC-secreted cytokines; however, apoptotic and metabolically through cell-to-cell contact and para-
inactivated MSCs have more recently been shown to possess immunomodula- crine activity.
tory potential, in which regulatory T cells and monocytes play a key role. We
review the immunomodulatory aspects of naïve and engineered MSCs, and Engineering MSCs to express specific
immunomodulatory agents contributes
discuss strategies for increasing the potential of successfully using MSCs in to MSC capacity and pluripotency, and
clinical settings. also enables them to deliver large
doses of cancer-targeting biologics with
a single dose.
Mesenchymal Stem Cells
MSCs are pluripotent T cells that have self-renewing, differentiation, and immunomodulatory MSC administration has shown potential
properties. Their two most attractive features are plasticity (see Glossary) and tropism. They efficacy in the treatment of several dis-
are distinguished from other cell types by the expression of cell-surface markers including eases that resist standard treatment.
However, there are some challenges in
CD73, CD90, and CD105, and by the lack of expression of CD45, CD34, CD14, CD19, efficiently translating MSC-based thera-
CD11b, and human leukocyte antigen DR isotype (HLA-DR) [1], and play a central role in tissue peutics into the clinic.
repair in addition to their antitumorigenic, antifibrotic, antiapoptotic, anti-inflammatory,
proangiogenic, neuroprotective, antibacterial, and chemoattractive effects [2,3]. This unique set Efficient homing and migration of MSCs
to the target tissue will be essential for
of characteristics makes MSCs attractive for their therapeutic potential in the fields of regenerative the future development of MSC-based
medicine [4], inflammatory disorders [2], and, increasingly, cancer therapy [5,6]. therapies.

Initially, MSCs were mainly used for tissue repair and regeneration [3]. However, they have been
increasingly used for diseases including graft-versus-host disease (GVHD) and autoimmune
1
Center for Stem Cell Therapeutics and
diseases such as lupus and Crohn’s disease [2]. In addition, the clinical potential of MSCs has
Imaging (CSTI), Harvard Medical School,
been extended to the treatment of myocardial infarction, stroke, multiple sclerosis, liver cirrhosis, Boston, MA 02115, USA
2
diabetes, lung injuries, and cancer [2,3]. MSCs have been harvested and expanded from a variety Department of Neurosurgery, Brigham
and Women’s Hospital, Harvard Medical
of adult and perinatal tissues such as bone marrow [7], adipose tissue [7,8], peripheral blood, fetal
School, Boston, MA 02115, USA
tissues [9], dental pulp [8], umbilical cord tissue [7,8], and placental tissues [7]. Most preclinical 3
Department of Medical Oncology, the
studies have been performed with bone marrow-derived MSCs (BM-MSCs); however, adipose First Hospital of China Medical
University, Shenyang 110001, China
tissue-derived MSCs (A-MSCs) and umbilical cord blood-derived MSCs (UC-MSCs) have also 4
Harvard Stem Cell Institute, Harvard
received considerable attention in recent years [9]. University, Cambridge, MA 02138, USA

Immunomodulation by MSCs
MSCs have recently been shown to possess immunomodulatory potential in which interactions
with regulatory T cells (Tregs) and monocytes play a key role [2,10]. Several studies have sug-
gested that A-MSCs exert more potent immunomodulatory effects than BM-MSCs, implying that *Correspondence:
A-MSCs are a better alternative for immunomodulatory therapy [11]. UC-MSCs, on the other kshah@bwh.harvard.edu (K. Shah).

Trends in Pharmacological Sciences, September 2020, Vol. 41, No. 9 https://doi.org/10.1016/j.tips.2020.06.009 653
© 2020 Elsevier Ltd. All rights reserved.
Trends in Pharmacological Sciences

hand, have been suggested to show minimal risk of initiating an allogeneic immune response Glossary
when administered in vivo. This, as well as their ease of collection, makes UC-MSCs suitable ther- Adaptive immunity: a subset of the
apeutic candidates [8]. In the following we focus on immunomodulatory aspects of naïve and immune system that is activated by
engineered MSCs, and synthesize our current understanding of the effects of MSC modulation exposure to pathogens and acts on the
threat using an immunological memory
on immune cells.
to enhance its effect. Cells of the
adaptive immune system include B and
Immunomodulatory Actions through Cell-to-Cell Contacts T cells.
MSCs participate in both innate immunity and adaptive immunity, and their immunomodula- Allogeneic: tissues or cells from
individuals of the same species, but
tory functions are exerted mainly via interactions with immune cells through cell-to-cell contact which are genetically dissimilar and
and paracrine activity involving T cells, B cells, natural killer (NK) cells, macrophages, hence immunologically incompatible.
monocytes, dendritic cells (DCs), and neutrophils [12] (Figure 1). Dendritic cells (DCs): antigen-
presenting cells in the mammalian
immune system. DCs act as
Cell-to-Cell Contact with Immune Cells of Adaptive Immunity
messengers between the innate and the
In vitro, MSCs have been shown to inhibit naïve T cell and memory T cell responses to adaptive immune systems.
communicate with antigen-presenting cells [13] by upregulating intercellular adhesion molecule 1 Eczema area and severity index
(EASI): a clinical scoring system that is
used to measure the extent and severity
of actopic eczema.
Granule polarization: a prelude to the
release of cytotoxic contents in
response to target-cell binding.
Innate immunity: a subset of the
immune system that is activated by the
presence of antigens and their chemical
properties, using nonspecific defense
mechanisms, meaning that anything that
is identified as foreign or non-self is a
target. Cells of the innate immune
system include natural killer (NK) cells,
macrophages, mast cells, neutrophils,
and DCs.
Lymphocyte: a subtype of white blood
cells that include NK cells, T cells, and
B cells.
M1 macrophages: polarized
macrophages that encourage
inflammation.
M2 macrophages: polarized
macrophages that decrease
inflammation and encourage tissue
repair.
Macrophage polarization: refers to
how macrophages have been activated
at a given point in space and time in
response to signals from their
microenvironment.
Membrane particles (MPs): particles
of the membrane of a cell ranging from
63 to 700 nm in diameter.
Trends in Pharmacological Sciences Memory T cells: a subset of infection-
and cancer-fighting T cells. They can
Figure 1. Multifaceted Immunomodulatory Interactions between Mesenchymal Stem Cells (MSCs) and
reproduce to mount a faster and
Immune Cells. MSCs exert immunomodulatory functions mainly via interactions with immune cells, such as T cells, stronger immune response, and
B cells, natural killer (NK) cells, macrophages, monocytes, dendritic cells (DCs), and neutrophils, as well as through cell-to-
recognize foreign invaders, such as
cell contacts (blue arrows) and paracrine activity (shown by secretome). The MSC secretome includes several cytokines,
bacteria or viruses, as well as cancer
growth factors, and chemokines, and their immunomodulatory functions vary depending on the source of the MSCs, the
cells.
target cells, and the microenvironment. Abbreviations: ICAM-1, intercellular adhesion molecule-1; IDO, indoleamine-pyrrole
Monocytes: the largest type of white
2,3-dioxygenase; IFN, interferon; IL, interleukin; PD-L1, programmed death ligand 1; PD-L2, programmed death ligand 2;
blood cells that can differentiate into
PGE2, prostaglandin E2; TGF-β, transforming growth factor-β; TNF-α, tumor necrosis factor-α; VEGF, vascular
macrophages and myeloid-lineage DCs.
endothelial growth factor. Figure drawn with BioRender (https://biorender.com/).

654 Trends in Pharmacological Sciences, September 2020, Vol. 41, No. 9


Trends in Pharmacological Sciences

(ICAM-1) and vascular cell adhesion molecule 1 (VCAM-1) that are crucial for T cell activation and Naïve T cells: primary cellular effectors
in the adaptive immune system that play
leukocyte (white blood cell, WBC) recruitment to the site of inflammation [14]. Furthermore,
crucial roles in antigen specificity.
coculture of BM-MSCs with activated T cells has been shown to induce lymphocytes that ex- Natural killer (NK) cells: cytotoxic
press interleukin-17A (IL-17A) [15]. MSCs cocultured with CD4+ T cells activate the Notch1/ lymphocytes that are crucial for the
Forkhead box P3 (FOXP3) pathway and increase the percentage of CD4+CD25 FOXP3+ cells innate immune system and provide rapid
responses to virus-infected cells and
[16]. Furthermore, knockdown of galectin 1, a protein that is abundantly expressed intracellularly tumor cells.
and on the cell surface of MSCs, and that has effects on T lymphocytes and cytokine secretion Neutrophils: the most abundant type
[17], in MSCs results in loss of immunomodulatory properties and restores the proliferation of of white blood cells and an essential part
CD4+ and CD8+ T cells [17]. In addition, BM-MSCs express high levels of Toll-like receptors of the innate immune system.
Plasticity: the capacity to differentiate
(TLRs) 3 and 4, which are responsible for nuclear factor κB (NF-κB) activity and cytokine produc- into other cell types in response to
tion. Expression of TLR3 and TLR4 in MSCs has been shown to restore an efficient T cell response different stimuli.
to infection [18]. Human placenta MSCs (PMSCs) have been shown to express high levels of the Regulatory T cells (Tregs): formerly
known as suppressor T cells, Tregs are
cell adhesion molecules programmed death ligand 1 (PD-L1) and PD-L2, which can inhibit T cell
a subpopulation of T cells that modulate
proliferation by arresting the cell cycle [19]. the immune system to downregulate the
induction and proliferation of effector
In vivo mouse models have also shown interactive immune regulation between MSCs and T cells. T cells.
Secretome: the set of proteins that are
In a syngeneic orthotopic mouse model of ovarian cancer, compact bone-derived MSCs
expressed by an organism and secreted
(CB-MSCs) have antitumor effects in combination with a fusion protein consisting of an anti- into the extracellular space, including
mesothelin single-chain antibody variable region (scFv) genetically fused to Mycobacterium cytokines, growth factors, extracellular
tuberculosis heat-shock protein 70 (Hsp70) (designated VIC-008), which is involved in activating matrix proteins, regulators, and shed
receptors.
CD4+ and CD8+ T cells and inhibiting Tregs in the tumor microenvironment (TME) [20]. In fetal
T helper 17 (Th17) cells: a subset of
abortion models, MSCs have been shown to enhance the suppressive regulation of T cells and proinflammatory T helper cells that are
macrophages [21]. Conversely, MSCs primed by activated T cells derived from IFN-γ null mice defined by the production of interleukin
exhibited a dramatically reduced ability to suppress T cell proliferation, and this supports the cell- 17 (IL-17).
Toll-like receptors (TLRs): receptors
to-cell contact mechanism for the immunosuppressive function of MSCs [22]. that sense invading pathogens or
endogenous damage signals, and
In addition to T cells, MSCs also affect B cells through cell-to-cell contact in vitro. A-MSCs have initiate the innate and adaptive immune
been shown to increase the survival of quiescent B cells via contact-dependent mechanisms and response. TLR3 and TLR4 are highly
expressed in BM-MSCs, and their
to facilitate B cell differentiation independently of T cells [23]. In addition, A-MSCs not only inhibit activation induces nuclear factor κB
caspase 3-mediated apoptosis of B cells by upregulating vascular endothelial growth (NF-κB) activity and cytokine production.
factor (VEGF) [24] but also inhibit cell proliferation by blocking the cell cycle of B lymphocytes in Tropism: the ability to migrate to
damaged or diseased tissues or cells.
G0/G1 phase by activating p38 mitogen-activated protein kinase (MAPK) pathways [25].

Cell-to-Cell Contacts with Immune Cells of Innate Immunity


In addition to the adaptive immune system, MSCs also affect the innate immune system through
cell-to-cell contacts. Tracking studies reveal that infused UC-MSCs briefly reside in the lungs
and are rapidly phagocytosed by monocytes, which subsequently migrate to other body sites.
UC-MSC phagocytosis induces phenotypic and functional changes in monocytes, which in turn
modulate cells of the adaptive immune system and thus play a crucial role in mediating, distributing,
and transferring the immunomodulatory effects of MSCs [26]. Coculture studies of MSCs with
different types of NK cell lines (KHYG-1 and NK-92) have shown that granule polarization is either
suppressed or induced, indicating differential crosstalk between MSCs and cytotoxic NK cells [27].
In addition, A-MSCs are known to switch activated inflammatory M1 macrophages to an M2
macrophage-like phenotype via prostaglandin E2 (PGE2) [28]. MSCs can also prevent neutrophil
death via an ICAM-1-dependent mechanism to further exert tissue-protective effects [29].

Immunomodulatory Actions through Paracrine Activity


Importantly, MSCs also exert their immunomodulatory properties by secreting multifunctional
molecules via paracrine mechanisms [12] (Figure 1). This secretome is a diverse repertoire of
multifaceted cytokines, growth factors, and chemokines, which combine to modulate the

Trends in Pharmacological Sciences, September 2020, Vol. 41, No. 9 655


Trends in Pharmacological Sciences

function of immune and cancer cells. These include transforming growth factor-β1 (TGF-β1),
tumor necrosis factor-α (TNF-α), PGE2, IFN-γ, hepatocyte growth factor (HGF), fibroblast
growth factor (FGF), indoleamine-pyrrole 2,3-dioxygenase (IDO), and nitric oxide, among many
others [30,31]. These paracrine factors are encapsulated in cell-secreted extracellular vesicles
(EVs), which in turn are usually divided into exosomes, microvesicles (MVs), and apoptotic bodies
according to their size and cell of origin [30,32]. Although MSC-derived EVs (MSC-EVs) display
immunoregulatory functions similar to the parent MSCs [33], their paracrine actions vary
according to the source of the MSCs, the target cells, and the microenvironment surrounding
the cells [34].

Paracrine Activity on the Adaptive Immune System


MSCs have been shown to act on the adaptive immune system, particularly T cells, via paracrine
secretion. MSCs inhibit T helper 17 (Th17) cell differentiation by inducing the production of IL-10
and PGE2, and by inhibiting IL-17, IL-22, and IFN-γ [35]. Coculture studies of BM-MSCs with
T cells have shown that both priming with cytokines and the cell ratio of BM-MSCs influence their
cytokine profiles, and suggest that BM-MSCs modulate the Th17 lymphocyte pathway in a complex
manner [15]. However, the mechanisms underlying interactions between MSCs and Th17 lympho-
cytes are not fully understood. Previous studies utilizing knockdown of IL-25 in vitro and in vivo have
shown that MSCs suppress Th17 responses via regulation of the IL-25/STAT3/PD-L1 axis [36].
MSC-secreted IDO induces Tregs that are responsible for kidney allograft tolerance [37]. Moreover,
cocultured exosomes derived from BM-MSCs transfected with a plasmid encoding sh RNA
targeting Fas cell death receptor (shFas) and anti-miR-375 and peripheral blood mononuclear
cells (PBMCs) suppress the immune response in an immunodeficient mouse model by inhibiting
PBMC proliferation and enhancing Treg function [38]. In addition, MSCs secrete PD-1 ligands
(including PD-L1 and PD-L2) and exert immunosuppressive effects directly on T cell behavior by
suppressing the activation of CD4+ T cells [39].

Paracrine Activity on the Innate Immune System


In the innate immune system MSCs are known to interact with NK cells by inhibiting IL-2-induced NK
cell proliferation [40] and by inducing cytotoxic activity or cytokine production via secretion of IDO and
PGE2 [41]. MSCs have also been shown to enhance the ability of IL-12/IL-18-stimulated NK cells to
secrete IFN-γ, which has the potential to improve defense against infections at the site of injury and
also influence tissue regeneration [42]. Moreover, MSC-derived IL-6 has been shown to protect neu-
trophils from apoptosis, thus preserving them in the bone marrow niche [43]. MSC-derived exosomes
have been shown to augment neutrophil viability, whereas MSC-conditioned medium (CM) increases
neutrophil function, demonstrating that both MSC-derived exosomes and CM are useful for increas-
ing immunity by modulating neutrophils [44]. Lipopolysaccharide (LPS)-stimulated MSCs augment
the antimicrobial functions of neutrophils via the release of IL-8 and macrophage migration inhibi-
tory factor (MIF), thus contributing to the resolution of infection and inflammation [45].

MSCs-secreted IL-6 can prevent the differentiation of monocytes towards an anti-inflammatory


IL-10 producing phenotype [46], whereas MSCs-derived PGE2 empower MSCs to suppress
the differentiation of monocytes to mature DCs [47]. MSC-derived EVs have been shown to pre-
vent antigen uptake by immature DCs and attenuate DC maturation, accompanied by downreg-
ulation of markers of mature DCs (CD83, CD38, and CD80) as well as of proinflammatory
cytokines (IL-6 and IL-12p70), and upregulation of the anti-inflammatory cytokine TGF-β [48].
In addition, miR-21-5p, a microRNA that is enriched in MSC-derived EVs, has been shown to
influence DC maturation and function [48]. MSC-EVs also play a crucial role in triggering
macrophage polarization [49] by enhancing the formation of anti-inflammatory M2 macro-
phages over M1-like inflammatory macrophages via downregulation of IL-23 and IL-22 [50].

656 Trends in Pharmacological Sciences, September 2020, Vol. 41, No. 9


Trends in Pharmacological Sciences

BM-MSCs activated by LPS or TNF-α reprogram macrophages by releasing PGE2 that acts on
the macrophages through the prostaglandin EP2 and EP4 receptors [51]. Human PMSCs have
also been shown to transform macrophages from an inflammatory M1 into an anti-
inflammatory M2 phenotype via soluble IL-10, IL-1β, IL-12, and macrophage inflammatory
protein 1α (MIP-1α), as well as via the glucocorticoid and progesterone receptors [52].

Preconditioning MSCs To Increase Their Immunomodulatory Functions and


Therapeutic Efficacy
Preconditioning of MSCs is known to influence their therapeutic efficacy, and modification
with diverse factors and a variety of conditions have therefore been explored to manipulate
the secretory profiles and enhance the therapeutic efficacy of MSCs [53]. Preconditioning
MSCs with hypoxia, oxidative stress, heat shock, starvation, or inflammatory biological
agents has potential to increase their survival and potency [53]. The main methods of pre-
conditioning are hypoxia and priming with immunomodulatory factors [32]. We discuss
these briefly next.

Preconditioning with Hypoxia


Hypoxia-preconditioning has been shown to have a positive influence on the MSC immune phe-
notype. Specifically, it increases paracrine and antioxidative effects, particularly the secretion of
angiogenic factors, as observed in examples of acute kidney injury [54] and bleomycin-induced
pulmonary fibrosis [55]. Hypoxia-preconditioning of dental MSCs overexpressing hypoxia-
inducible factor 1α (HIF-1α) can impair DC differentiation, attract more monocytes, and increase
resistance to NK cell-mediated lysis [56]. Hypoxia-preconditioning of CB-MSCs coupled to pre-
treatment with proinflammatory cytokines IFN-γ, TNF-α, and IL-17A improves suppression of
CD8+ T cells [57]. Hypoxia-conditioning of MSCs results in higher infiltration of endothelial cells
into MSC-containing scaffolds for dermal regeneration [58], positively affecting cell fitness and
the transcriptome, and potentially improving cellular therapies targeting orthopedic disorders
[59] as well as promoting maintenance of stem-like characteristics with IDO upregulation [60].
Furthermore, MSCs primed with hypoxia and calcium ions (HC-MSCs) have been shown to be
resistant to passage-dependent senescence mediated via the monocyte chemoattractant pro-
tein 1 and p53/p21 cascade, and secrete large amounts of proangiogenic and immunomodula-
tory factors, resulting in suppression of T cell proliferation in vitro. Administration of HC-MSCs
significantly attenuated the symptoms of GVHD in a humanized mouse model, resulting in signif-
icantly improved survival [61].

Preconditioning by Priming with Immunomodulatory Factors


Another important method of preconditioning is priming with immunomodulatory factors. IFN-γ is
a primary factor that can activate the transcription and synthesis of IDO-1, HGF, and TGF-β in
MSCs [62]. Single IFN-γ priming induces IDO expression and leads to sustained improvement
in the MSC T cell-suppressive phenotype [63]. MSCs primed by IFN-γ have been shown to
have enhanced immunosuppressive function via the IFN-γ–JAK–STAT1 pathway [64,65]. In
addition, membrane particles (MPs) of MSCs stimulated with IFN-γ can increase mRNA
expression of PD-L1 in monocytes as well as the percentage of anti-inflammatory PDL-1- and
CD90-positive monocytes, thus demonstrating the potential of MSC derived MPs as a novel
cell-free therapy for the treatment of immunological disorders [66]. Priming of MSCs with TNF-α
and IFN-γ has been shown to reverse the proinflammatory effects of palmitate, a potent modulator
of MSC immunosuppressive function that is enriched in type 2 diabetes, and can maintain the abil-
ity to block T cell proliferation and cytokine production [62]. More recently, it has been shown that
modification of BM-MSCs with small heat-shock proteins 27 and 20 (sHsp27 and sHsp20) and E7
oncoprotein significantly enhances E7-specific T cell responses and suppresses tumor growth in

Trends in Pharmacological Sciences, September 2020, Vol. 41, No. 9 657


Trends in Pharmacological Sciences

mice, indicating that MSC-based vaccination is a promising approach for immunotherapy and pro-
tection against HPV-associated cancers [67].

Engineering MSCs To Increase Their Immunomodulatory Functions and


Therapeutic Efficacy
MSCs have an intrinsic ability to evade the immune response temporarily. In the context of using
MSCs for targeted therapy, they are mainly considered as gene delivery vehicles for immunomod-
ulatory molecules such as IFNs and ILs, or are engineered to deliver oncolytic viruses (OVs)
(Figure 2A). Engineering MSCs to express specific immunomodulatory agents contribute to
their immunomodulatory capacity and pluripotency, and also enables them to deliver large
doses of cancer-targeting biologics with a single dose. Some examples of engineered MSCs
expressing diverse immunomodulatory molecules are given in Table 1 and are discussed in the
following section.

We previously showed that encapsulated MSCs engineered to express the cytokine IFN-β can
increase the survival of mice with a highly malignant brain tumor, glioblastoma, by promoting se-
lective postsurgical infiltration of CD8 T cells and inducing cell-cycle arrest in tumor cells (Table 1)
[68]. Likewise, recently, engineered MSCs expressing IFN-γ have also been shown to elicit tumor
growth reduction and increase survival via polarization of murine macrophages to the proinflam-
matory M1 phenotype in vitro and in vivo [69]. Similarly, MSCs engineered to express IL-4 have

Trends in Pharmacological Sciences

Figure 2. Three Strategies To Enhance the Therapeutic Efficacy of Mesenchymal Stem Cells (MSCs). (A) MSCs can be engineered with immunomodulatory
molecules such as interferons (IFNs), interleukins (ILs), and miRNA, or can be engineered to deliver oncolytic viruses. (B) MSCs can also be primed, which can contribute to
their efficient homing and migration to the target tissue. (C) Different methods have been modified and standardized to improve the cryopreservation of MSCs.
Abbreviations: CSF-2, colony-stimulating factor 2; CXCR4, CXC chemokine receptor 4; IFN, interferon; IL, interleukin; IGFBP3, insulin-like growth factor binding protein
3; miRNA, microRNA, SDF-1, stromal cell-derived factor-1. Figure drawn with BioRender (https://biorender.com/).

658 Trends in Pharmacological Sciences, September 2020, Vol. 41, No. 9


Trends in Pharmacological Sciences

Table 1. Engineered MSCs Expressing Diverse Immunomodulatory Moleculesa


Engineered agent Model Mechanism Refs
IFN-β Syngeneic orthotopic glioblastoma mouse model Infiltration of CD8 T cells, cell-cycle arrest [68]
IFN-γ Neuroblastoma murine model Macrophage polarization [69]
IL-4 Osteoarthritis rat model Chondroprotective and anti-inflammatory effects [70]
IL-12 Renal cell carcinoma, melanoma, breast tumor, and Reduction of lymphatic sprouts [71,72]
HCC mouse models Apoptosis induction
IL-15 Pancreatic tumor mouse model Tumor-specific T cell immune memory response [73]
IL-21 B cell lymphoma mouse model Induction of effector T and NK cells [74]
SDF-1 Streptozotocin-induced diabetic rats Increasing nNOS, VEGF, and bFGF [75]
miR-199a HCC mouse model mTOR pathway inhibition [76]
Oncolytic virus Brain metastatic melanoma mouse model Increasing IFN-γ-producing CD8+ tumor-infiltrating T lymphocytes [78]
with PD-L1 blockade
HCC mouse model Inhibiting HCC cell proliferation [79]
Lung adenocarcinoma mouse model TLR9 overexpression and activation of the NF-κB pathway [80]

a
Abbreviations: bFGF, basic fibroblast growth factor; IFN, interferon; GVHD, graft-versus-host disease; IL, interleukin; HCC, hepatocellular carcinoma; mTOR, mammalian
target of rapamycin; NK cell, natural killer cell; nNOS, neuronal nitric oxide synthase; SDF-1, stromal cell-derived factor-1; VEGF, vascular endothelial growth factor.

increased chondroprotective and anti-inflammatory effects in a rat model of osteoarthritis [70],


whereas IL-12-engineered MSCs reduce tumor growth and prolong survival in mouse models
of renal cell carcinoma, melanoma, breast tumor, and hepatocellular carcinoma (HCC) via reduc-
tion of lymphatic sprouts and induction of apoptosis [71,72]. Engineered MSCs expressing mu-
rine IL-15 significantly inhibit pancreatic cancer growth and prolong the survival of tumor-bearing
mice, as well as promoting resistance to pancreatic tumor rechallenge, via the induction of tumor-
specific T cell immune memory response [73]. Moreover, MSCs engineered to express IL-21 in-
duce effector T and NK cells, and can prevent the formation of tumor nodules in a mouse model of
B cell lymphoma [74]. In addition, MSCs engineered to express stromal cell-derived factor 1
(SDF-1) have been shown to alleviate erectile dysfunction in streptozotocin-induced diabetic
rats (Table 1) mediated by increasing the levels of neuronal nitric oxide synthase (nNOS), VEGF,
and basic fibroblast growth factor (bFGF,) and by lowering the levels of the apoptosis factors Bcl2-
associated x (Bax) and caspase 3 [75]. Furthermore, exosomes derived from A-MSCs expressing
miR-199 can promote miR-199a delivery to HCC cells and effectively sensitize cancer cells to che-
motherapeutic agents by targeting the mammalian target of rapamycin (mTOR) pathway [76].

MSCs have also been explored as a potential means of OV delivery because they can shield the
virus from host antiviral immunity and transport viral particles systemically or intratumorally [77].
We discuss some examples where the immunomodulatory function of MSCs has been shown
to be enhanced (Table 1). MSCs loaded with oncolytic herpes simplex virus (oHSV) or other
OVs can successfully deliver viral progeny to established glioblastoma tumors and brain-
metastatic melanoma, eventually decreasing tumor growth and prolonging mouse survival
[77,78]. Another study reported that MSCs loaded with HCC-targeted OVs effectively lyze
HCC cells in vitro, ultimately leading to potent tumor growth inhibition [79]. When OV-loaded
MSCs were cocultured with allogeneic PBMCs, they induced TLR9 overexpression and activa-
tion of the NF-κB pathway, creating a proinflammatory environment and enhancing antitumor ef-
ficacy both in vitro and in vivo in lung adenocarcinoma xenograft tumors [80]. In addition,
combined blockade of MSC-oHSV and PD-L1 increases the number of IFN-γ-producing CD8+
tumor-infiltrating T cells, and further increases the median survival of mice in melanoma brain
metastasis models [78,81].

Trends in Pharmacological Sciences, September 2020, Vol. 41, No. 9 659


Trends in Pharmacological Sciences

MSC-Mediated Immunomodulation in Clinical Studies and Trials


MSC infusion has shown potential efficacy in the treatment of several diseases that resist stan-
dard treatment. For example, in a Phase I/II clinical trial (EuDRA CTi: 2012-003741-15) involving
autologous tumor-specific herpes simplex virus thymidine kinase (HSV-TK), MSCs demonstrated fa-
vorable safety and tolerability in patients with gastrointestinal tumors: the median time to progression
was 1.8 months, and median overall survival was 15.6 months (Table 2) [82,83]. However, the study
was terminated early because of an insufficient number of patients. Another recent Phase I/IIa trial
(clinical trialii NCT02351011) using BM-MSCs in patients with osteoarthritis demonstrated that
autologous BM-MSCs are safe and elicit significant reductions in pain and synovial inflammation
[84]. Similarly, an expanded Phase II/III study (clinical trial UMIN-CTR numberiii UMIN000006719)
demonstrated that BM-MSC infusion improved the overall survival rate in patients with steroid-
resistant GVHD (Table 1) [85]. In a Phase IIa study of secondary progressive multiple sclerosis,
autologous BM-MSCs elicited safe and effective neuroprotection (clinical trial NCT00395200) [86].
For patients with moderate-to-severe atopic dermatitis, subcutaneous administration of human
UC-MSCs brought about marked improvement of disease features without serious adverse events,
and 55% of patients showed a 50% reduction in the eczema area and severity index (EASI)
(Table 1) [87]. In refractory fistulizing Crohn's disease, a pilot study therapy with autologous BM-
MSCs proved feasible, safe, and beneficial, and sustained complete closure was observed in 7/12
patients [88]. Another important Phase III study (clinical trial NCT01541579) has assessed the efficacy
and safety of expanded A-MSCs (Cx601) for the treatment of complex perianal fistulas in patients with
Crohn's disease. Cx601 treatment significantly achieved remission with fewer treatment-related ad-
verse events, suggesting that MSCs may be an effective strategy for clinical applications [89].

Nevertheless, it should be noted that MSC-based treatment might produce some potential unex-
pected side effects. For example, recent preclinical studies suggest that alteration of the brain mi-
croenvironment by acute hypothermia modulates MSC function, resulting in a proinflammatory
environment and increasing long-lasting motor-cognitive deficits in the neonatal hypoxic/ischemic
brain [90].

Promising Preclinical Studies That Have Potential To Increase the Efficacy of


Future MSC Trials
Apart from improving immunomodulatory capacity of MSCs (Figure 2A), there are several ap-
proaches to improve MSC migration or homing mechanisms (Figure 2B). Recent studies have

Table 2. Examples of Clinical Trials Currently Using MSCsa


Disease Origin of MSCs Treatment route Clinical trial number Phase Therapeutic efficacy Refs
Gastrointestinal Autologous BM-MSCs Intravenous infusion EudraCT numberi I/II Favorable safety and tolerability, median [82,83]
adenocarcinoma 2012-003741-15 overall survival is 15.6 months
Osteoarthritis Autologous BM-MSCs Intralesional injection Clinical trial I/II Safe, reduction in pain and symptoms, [84]
numberii and reduced synovial inflammation
NCT02351011
GVHD Allogeneic BM-MSCs Infusion UMIN-CTR II/III Improve the overall survival rate [85]
numberiii
UMIN000006719
Multiple sclerosis Autologous BM-MSCs Intravenous infusion NCT00395200 IIa Safe, effective neuroprotection [86]
Atopic dermatitis Human UC-MSCs Subcutaneous NCT01927705 I/II Marked improvement of features without [87]
serious adverse events
Crohn's disease Allogeneic A-MSCs Intralesional injection NCT01541579 III Increased remission with less [89]
treatment-related adverse events

a
Abbreviations: A-MSCs, adipose tissue-derived MSCs; BM-MSCs, bone marrow-derived MSCs; GVHD, graft-versus-host disease; UC-MSCs, umbilical cord blood-derived
MSCs.

660 Trends in Pharmacological Sciences, September 2020, Vol. 41, No. 9


Trends in Pharmacological Sciences

shown that MSC homing is driven by SDF-1-stimulated endothelial cell production of platelet- Outstanding Questions
derived growth factor (PDGF) via activation of PDGF receptor A (PDGFRA)/PI3K/Akt, PDGFRA/ What external factors influence
MAPK/Grb2, and PDGFRA/JAK2/STAT signaling [91]. In addition, FGF21 also influences the immunomodulation mediated by
MSCs?
homing ability of MSCs to injury sites [92], and colony-stimulating factor 2 (CSF-2) has been
shown to significantly enhance their differentiation and migratory capacity [93]. IL-1β induces ma- How can we minimize variation in MSC
trix metalloproteinase-1 (MMP-1), which subsequently activates the protease-activated receptor paracrine effectiveness and therapeu-
1 (PAR1), and G protein-coupled signaling pathways have been recently shown to promote MSC tic efficacy for clinical translation?
migration [94]. Interestingly, intranasally administered MSC-derived exosomes specifically target
Which new preconditioning techniques
and accumulate in the brain in pathological murine models for up to 96 h, and this long homing can lead to better MSC therapy out-
response is driven by neuroinflammatory signals [95]. Hypoxic preconditioning also promotes comes and further enhance therapeu-
the migration and survival of MSCs through increased expression of lincRNA-p21 via the tic efficacy?

HIF-1α/CXCR4 and CXCR7 pathway [96]. Can MSCs be engineered with multiple
immunomodulators that mediate key
Another way to increase the efficacy of MSCs would be to improve methods for their cryopreser- biological signaling pathways?
vation. Recent studies have indicated that cryopreservation modulates the efficacy and clinical
How can we efficiently translate
application of MSCs. Specifically, cryopreserved and thawed MSCs have impaired functional promising preclinical studies into the
properties and stunted immunosuppressive capabilities [97,98], which have significant implica- clinic?
tions for therapeutic outcomes [98]. Therefore, MSCs need to be standardized for reliability and
robust quality-control purposes, especially in large-scale production of frozen products. A recent
study revealed the feasibility of two freezing steps after a cell culture phase of at least one pas-
sage, without affecting basic cell manufacturing parameters or quality attributes of the final frozen
and thawed product [98]. A successful Phase III clinical trial (NCT01541579) using expanded A-
MSCs (Cx601) to treat patients with Crohn's disease revealed that a recovery period of at least 24
h after cell thawing improved MSC product quality [89]. These MSCs maintained their multipotent
differentiation, immunomodulatory, and anti-inflammatory properties [97]. Therefore, a 24 h accli-
mation period to reactivate thawed cells is crucial to restore their diminished MSC function
(Figure 2C).

Concluding Remarks and Future Perspectives


MSCs have emerged as a promising therapeutic strategy because of their tropism for other cell
types as well as their immunomodulatory functions. The immunomodulatory ability of MSCs is
regulated by different inflammatory cytokines, and the interaction between immune cells and
MSCs could contribute to regeneration as well as to the progression of different inflammatory dis-
eases. The main mechanisms involved in MSC immunomodulation are cell-to-cell contact and
paracrine activity primed by cytokines, chemokines, extracellular vesicles, inflammatory stimuli,
or coculture with other cells. Therefore, priming or licensing of MSCs has made cell-free therapy
a controllable, manageable, and feasible method. However, several issues remain to be solved
(see Outstanding Questions). MSCs are very heterogeneous and change significantly with inflam-
matory or anti-inflammatory stimuli. Therefore, it is difficult to determine how MSC variability influ-
ences their induced immunomodulatory effects. It is possible that viable MSCs provoke more
complex immunomodulatory mechanisms because of their intact secretome. Future work should
explore the influence of other factors and/or chronic inflammation on immunomodulation medi-
ated by MSCs. This has the potential to identify new methods of preconditioning to enhance
MSC efficacy and minimize variation in MSC paracrine effectiveness and therapeutic efficacy, es-
pecially with regards to clinical translatability.

In the context of using MSCs for targeted therapy in the clinic, they are mainly considered as gene
delivery vehicles for immunomodulatory molecules or OVs. Because engineered MSCs releasing
targeted therapeutics are already being used in multiple clinical trials for immune and inflamma-
tory diseases, it is essential to define the targets in the diseased cells/microenvironment to

Trends in Pharmacological Sciences, September 2020, Vol. 41, No. 9 661


Trends in Pharmacological Sciences

develop effective MSC therapies. For example, previous studies have shown that MSCs
engineered to coexpress an antigen-specific domain (VHH) targeting epidermal growth factor re-
ceptor (EGFR) and death receptor (DR4/5) ligand (DRL) target both cell proliferation and death
pathways in tumors [99]. MSCs engineered to express TandAb, a tetravalent bispecific tandem
diabody for CD3 and CD19, were recently found to have enhanced therapeutic efficacy in a
mouse model of B cell lymphoma as compared with targeting CD3 or CD19 alone [100].

A challenge in the field is the discrepancy between in vitro and in vivo findings in MSC research. For
example, in a New Zealand white rabbit osteogenesis model, osteogenic cells differentiated from
MSCs (DOC cells) retained their immunoprivileged and immunomodulatory properties in vitro,
but their immunomodulatory properties were lost following transplantation [101]. In a rhesus ma-
caque model, allogeneic versus autologous MSCs transplanted intracranially revealed that alloge-
neic MSCs, but not autologous MSCs, were weakly immunogenic post-transplantation, thus
negatively affecting the levels of long-term engraftment [102]. These discrepancies underline the
importance of careful monitoring of MSC immunogenicity when moving from an in vitro to an
in vivo model, as well as in further clinical applications, so as to ensure a lasting effect. Ultimately,
by enhancing their immunomodulatory potential and homing mechanisms, and by improving cryo-
preservation techniques, in conjunction with rigorous preclinical and clinical studies, MSCs have
enormous potential for wider application in clinical settings in the future.

Acknowledgments
This work was supported by National Institutes of Health grant R01-NS107857 and Department of Defence grant
CA180698 to K.S.

Disclaimer Statement
K.S. owns equity in and is a member of the Board of D irectors of AMASA Therapeutics, a company developing stem cell-based
therapies for cancer; his interests were reviewed and are managed by Brigham and Women’s Hospital and Partners HealthCare
in accordance with their conflict of interest policies. The other authors declare that they have no competing interests.

Resources
i
https://eudract.ema.europa.eu/
ii
https://clinicaltrials.gov/
iii
https://www.umin.ac.jp/

References
1. Dominici, M. et al. (2006) Minimal criteria for defining 9. Shah, K. (2016) Stem cell-based therapies for tumors in the
multipotent mesenchymal stromal cells. The International brain: are we there yet? Neuro-oncology 18, 1066–1078
Society for Cellular Therapy position statement. Cytotherapy 10. Weiss, A.R.R. and Dahlke, M.H. (2019) Immunomodulation by
8, 315–317 mesenchymal stem cells (MSCs): mechanisms of action of
2. Galipeau, J. and Sensébé, L. (2018) Mesenchymal stromal living, apoptotic, and dead MSCs. Front. Immunol. 10, 1191
cells: clinical challenges and therapeutic opportunities. Cell 11. Melief, S.M. et al. (2013) Multipotent stromal cells skew monocytes
Stem Cell 22, 824–833 towards an anti-inflammatory interleukin-10-producing phenotype
3. Timaner, M. et al. (2019) The multifaceted role of mesenchymal by production of interleukin-6. Haematologica 98, 888–892
stem cells in cancer. Semin. Cancer Biol. 60, 225–237 12. Zhou, Y. et al. (2019) The immunomodulatory functions of
4. Chen, Y. et al. (2008) Mesenchymal stem cells: a promising mesenchymal stromal/stem cells mediated via paracrine
candidate in regenerative medicine. Intl. J. Biochem. Cell activity. J. Clin. Med. 8, 1022
Biol. 40, 815–820 13. Krampera, M. et al. (2003) Bone marrow mesenchymal stem
5. Dai, L.-J. et al. (2011) Potential implications of mesenchymal cells inhibit the response of naive and memory antigen-specific
stem cells in cancer therapy. Cancer Lett. 305, 8–20 T cells to their cognate peptide. Blood 101, 3722
6. Shah, K. (2012) Mesenchymal stem cells engineered for 14. Ren, G. et al. (2010) Inflammatory cytokine-induced intercellu-
cancer therapy. Adv. Drug Deliv. Rev. 64, 739–748 lar adhesion molecule-1 and vascular cell adhesion molecule-1
7. Heo, J.S. et al. (2016) Comparison of molecular profiles of in mesenchymal stem cells are critical for immunosuppression.
human mesenchymal stem cells derived from bone marrow, J. Immunol. 184, 2321–2322
umbilical cord blood, placenta and adipose tissue. Intl. 15. Najar, M. et al. (2019) Immunological modulation following
J. Molec. Med. 37, 115–125 bone marrow-derived mesenchymal stromal cells and Th17
8. Kim, J.-H. et al. (2018) Comparison of immunological lymphocyte co-cultures. Inflamm. Res. 68, 203–212
characteristics of mesenchymal stem cells from the periodon- 16. Del Papa, B. et al. (2013) Notch1 modulates mesenchymal stem
tal ligament, umbilical cord, and adipose tissue. Stem Cells Int. cells mediated regulatory T-cell induction. Eur. J. Immunol. 43,
2018, 8429042 182–187

662 Trends in Pharmacological Sciences, September 2020, Vol. 41, No. 9


Trends in Pharmacological Sciences

17. Gieseke, F. et al. (2010) Human multipotent mesenchymal 40. Spaggiari, G.M. et al. (2006) Mesenchymal stem cell-natural
stromal cells use galectin-1 to inhibit immune effector cells. killer cell interactions: evidence that activated NK cells are
Blood 116, 3770–3772 capable of killing MSCs, whereas MSCs can inhibit IL-2-
18. Liotta, F. et al. (2008) Toll-like receptors 3 and 4 are expressed induced NK-cell proliferation. Blood 107, 1484–1492
by human bone marrow-derived mesenchymal stem cells and 41. Spaggiari, G.M. et al. (2008) Mesenchymal stem cells inhibit
can inhibit their T-cell modulatory activity by impairing Notch natural killer-cell proliferation, cytotoxicity, and cytokine
signaling. Stem Cells 26, 279–289 production: role of indoleamine 2,3-dioxygenase and
19. Wang, G. et al. (2013) Expression and biological function of prostaglandin E2. Blood 111, 1327–1332
programmed death ligands in human placenta mesenchymal 42. Thomas, H. et al. (2014) Interaction with mesenchymal stem
stem cells. Cell Biol. Int. 37, 137–142 cells provokes natural killer cells for enhanced IL-12/IL-18-
20. Zeng, Y. et al. (2019) CD90low MSCs modulate intratumoral induced interferon-gamma secretion. Mediat. Inflamm. 2014,
immunity to confer antitumor activity in a mouse model of 143463
ovarian cancer. Oncotarget 10, 4472 43. Raffaghello, L. et al. (2008) Human mesenchymal stem cells in-
21. Li, Y. et al. (2019) Cell-cell contact with proinflammatory hibit neutrophil apoptosis: a model for neutrophil preservation
macrophages enhances the immunotherapeutic effect of in the bone marrow niche. Stem Cells 26, 151–162
mesenchymal stem cells in two abortion models. Cell. Mol. 44. Mahmoudi, M. et al. (2019) Comparison of the effects of adi-
Immunol. 16, 908–920 pose tissue mesenchymal stromal cell-derived exosomes
22. Sheng, H. et al. (2008) A critical role of IFNγ in priming MSC- with conditioned media on neutrophil function and apoptosis.
mediated suppression of T cell proliferation through up- Int. Immunopharmacol. 74, 105682
regulation of B7-H1. Cell Res. 18, 846–852 45. Brandau, S. et al. (2014) Mesenchymal stem cells augment the
23. Franquesa, M. et al. (2015) Human adipose tissue-derived anti-bacterial activity of neutrophil granulocytes. PLoS One 9,
mesenchymal stem cells abrogate plasmablast formation and e106903
induce regulatory B cells independently of T helper cells. 46. Melief, S.M. et al. (2013) Adipose tissue-derived multipotent
Stem Cells 33, 880–892 stromal cells have a higher immunomodulatory capacity than
24. Healy, M.E. et al. (2015) Mesenchymal stromal cells protect their bone marrow-derived counterparts. Stem Cells Transl.
against caspase 3-mediated apoptosis of CD19+ peripheral Med. 2, 455–463
B cells through contact-dependent upregulation of VEGF. 47. Spaggiari, G.M. et al. (2009) MSCs inhibit monocyte-derived
Stem Cells Dev. 24, 2391–2402 DC maturation and function by selectively interfering with the
25. Tabera, S. et al. (2008) The effect of mesenchymal stem cells on generation of immature DCs: central role of MSC-derived
the viability, proliferation and differentiation of B-lymphocytes. prostaglandin E2. Blood 113, 6576–6583
Haematologica 93, 1301–1309 48. Reis, M. et al. (2018) Mesenchymal stromal cell-derived
26. de Witte, S.F. et al. (2018) Immunomodulation by therapeutic extracellular vesicles attenuate dendritic cell maturation and
mesenchymal stromal cells (MSC) is triggered through phago- function. Front. Immunol. 9, 2538
cytosis of MSC by monocytic cells. Stem Cells 36, 602–615 49. Sicco, C.L. et al. (2017) Mesenchymal stem cell-derived
27. Hu, C-H.D. et al. (2019) Differential immunomodulatory effects extracellular vesicles as mediators of anti-inflammatory effects:
of human bone marrow-derived mesenchymal stromal cells on endorsement of macrophage polarization. Stem Cells Transl.
natural killer cells. Stem Cells Dev. 28, 933–942 Med. 6, 1018–1028
28. Manferdini, C. et al. (2017) Adipose stromal cells mediated 50. Hyvärinen, K. et al. (2018) Mesenchymal stromal cells and
switching of the pro-inflammatory profile of M1-like macro- their extracellular vesicles enhance the anti-inflammatory
phages is facilitated by PGE2: in vitro evaluation. Osteoarthr. phenotype of regulatory macrophages by downregulating
Cartil. 25, 1161–1172 the production of interleukin (IL)-23 and IL-22. Front.
29. Jiang, D. et al. (2016) Suppression of neutrophil-mediated tis- Immunol. 9, 772
sue damage – a novel skill of mesenchymal stem cells. Stem 51. Németh, K. et al. (2009) Bone marrow stromal cells attenuate
Cells 34, 2393–2402 sepsis via prostaglandin E 2-dependent reprogramming of
30. Li, N. and Hua, J. (2017) Interactions between mesenchymal stem host macrophages to increase their interleukin-10 production.
cells and the immune system. Cell. Mol. Life Sci. 74, 2345–2362 Nat. Med. 15, 42
31. Salgado, A.J. et al. (2010) Adipose tissue derived stem cells 52. Abumaree, M. et al. (2013) Human placental mesenchymal
secretome: soluble factors and their roles in regenerative stem cells (pMSCs) play a role as immune suppressive cells
medicine. Curr. Stem Cell Res. Ther. 5, 103–112 by shifting macrophage differentiation from inflammatory M1
32. Ferreira, J.R. et al. (2018) Mesenchymal stromal cell secretome: to anti-inflammatory M2 macrophages. Stem Cell Rev. Rep.
influencing therapeutic potential by cellular pre-conditioning. 9, 620–642
Front. Immunol. 9, 2832 53. Silva, L.H. et al. (2018) Strategies to improve the therapeutic ef-
33. Mardpour, S. et al. (2019) Interaction between mesenchymal fects of mesenchymal stromal cells in respiratory diseases.
stromal cell-derived extracellular vesicles and immune cells by Stem Cell Res Ther 9, 42
distinct protein content. J. Cell. Physiol. 234, 8249–8252 54. Zhang, W. et al. (2014) Hypoxia-pretreated human MSCs at-
34. Madrigal, M. et al. (2014) A review of therapeutic effects of mes- tenuate acute kidney injury through enhanced angiogenic
enchymal stem cell secretions and induction of secretory modifi- and antioxidative capacities. Biomed. Res. Int. 2014,
cation by different culture methods. J. Transl. Med. 12, 262 462–472
35. Ghannam, S. et al. (2010) Mesenchymal stem cells inhibit 55. Lan, Y.-W. et al. (2015) Hypoxia-preconditioned mesenchymal
human Th17 cell differentiation and function and induce a T stem cells attenuate bleomycin-induced pulmonary fibrosis.
regulatory cell phenotype. J. Immunol. 185, 302–312 Stem Cell Res Ther 6, 92
36. Wang, W.-B. et al. (2015) Interleukin-25 mediates transcrip- 56. Martinez, V.G. et al. (2017) Overexpression of hypoxia-
tional control of PD-L1 via STAT3 in multipotent human inducible factor 1 alpha improves immunomodulation by dental
mesenchymal stromal cells (hMSCs) to suppress Th17 mesenchymal stem cells. Stem Cell Res Ther 8, 202
responses. Stem Cell Reports 5, 392–402 57. Sivanathan, K.N. et al. (2017) Immunodepletion and hypoxia
37. Ge, W. et al. (2010) Regulatory T-cell generation and kidney preconditioning of mouse compact bone cells as a novel pro-
allograft tolerance induced by mesenchymal stem cells associ- tocol to isolate highly immunosuppressive mesenchymal stem
ated with indoleamine 2,3-dioxygenase expression. Transplan- cells. Stem Cells Dev. 26, 512–522
tation 90, 1312–1322 58. Fierro, F.A. et al. (2015) Hypoxic pre-conditioning increases the
38. Wen, D. et al. (2016) Mesenchymal stem cell and derived infiltration of endothelial cells into scaffolds for dermal regener-
exosome as small RNA carrier and Immunomodulator to im- ation pre-seeded with mesenchymal stem cells. Front. Cell
prove islet transplantation. J. Control. Release 238, 166–172 Dev. Biol. 3, 62
39. Davies, L.C. et al. (2017) Mesenchymal stromal cell secretion of 59. Elabd, C. et al. (2018) Comparing atmospheric and hypoxic cul-
programmed death-1 ligands regulates T cell mediated tured mesenchymal stem cell transcriptome: implication for stem
immunosuppression. Stem Cells 35, 766–772 cell therapies targeting intervertebral discs. J. Transl. Med. 16, 222

Trends in Pharmacological Sciences, September 2020, Vol. 41, No. 9 663


Trends in Pharmacological Sciences

60. Kadle, R.L. et al. (2018) Microenvironmental cues enhance 82. Niess, H. et al. (2015) Treatment of advanced gastrointestinal
mesenchymal stem cell-mediated immunomodulation and reg- tumors with genetically modified autologous mesenchymal
ulatory T-cell expansion. PLoS One 13, e0193178 stromal cells (TREAT-ME1): study protocol of a phase I/II clini-
61. Kim, Y. et al. (2018) Small hypoxia-primed mesenchymal stem cells cal trial. BMC Cancer 15, 232
attenuate graft-versus-host disease. Leukemia 32, 2672–2682 83. von Einem, J.C. et al. (2019) Treatment of advanced gastroin-
62. Boland, L. et al. (2018) IFN-γ and TNF-α pre-licensing protects testinal cancer with genetically modified autologous mesen-
mesenchymal stromal cells from the pro-inflammatory effects chymal stem cells: results from the phase 1/2 TREAT-ME-1
of palmitate. Mol. Ther. 26, 860–872 trial. Int. J. Cancer 145, 1538–1542
63. Zimmermann, J.A. et al. (2017) Enhanced immunosuppression 84. Chahal, J. et al. (2019) Bone marrow mesenchymal stromal
of T cells by sustained presentation of bioactive interferon-γ cell treatment in patients with osteoarthritis results in overall im-
within three-dimensional mesenchymal stem cell constructs. provement in pain and symptoms and reduces synovial
Stem Cells Transl. Med. 6, 223–232 inflammation. Stem Cells Transl. Med. 8, 746–752
64. Klinker, M.W. et al. (2017) Morphological features of IFN-γ- 85. Muroi, K. et al. (2016) Bone marrow-derived mesenchymal
stimulated mesenchymal stromal cells predict overall stem cells (JR-031) for steroid-refractory grade III or IV acute
immunosuppressive capacity. Proc. Natl. Acad. Sci. 114, graft-versus-host disease: a phase II/III study. Int. J. Hematol.
E2598–E2602 103, 243–252
65. Kim, D.S. et al. (2018) Enhanced immunosuppressive properties 86. Connick, P. et al. (2012) Autologous mesenchymal stem cells
of human mesenchymal stem cells primed by interferon-γ. for the treatment of secondary progressive multiple sclerosis:
EBioMedicine 28, 261–272 an open-label phase 2a proof-of-concept study. Lancet
66. da Costa Gonçalves, F. et al. (2017) Membrane particles Neurol. 11, 150–152
generated from mesenchymal stromal cells modulate immune 87. Kim, H.S. et al. (2017) Clinical trial of human umbilical cord blood-
responses by selective targeting of pro-inflammatory derived stem cells for the treatment of moderate-to-severe atopic
monocytes. Sci. Rep. 7, 12100 dermatitis: Phase I/IIa studies. Stem Cells 35, 248–252
67. Bolhassani, A. et al. (2019) Modified DCs and MSCs with HPV 88. Ciccocioppo, R. et al. (2011) Autologous bone marrow-derived
E7 antigen and small Hsps: which one is the most potent strat- mesenchymal stromal cells in the treatment of fistulising
egy for eradication of tumors? Mol. Immunol. 108, 102–112 Crohn's disease. Gut 60, 788–792
68. Choi, S.H. et al. (2017) Tumor resection recruits effector T cells and 89. Panés, J. et al. (2016) Expanded allogeneic adipose-derived
boosts therapeutic efficacy of encapsulated stem cells expressing mesenchymal stem cells (Cx601) for complex perianal fistulas
IFNβ in glioblastomas. Clin. Cancer Res. 23, 7047–7052 in Crohn's disease: a phase 3 randomised, double-blind con-
69. Relation, T. et al. (2018) Intratumoral delivery of interferonγ- trolled trial. Lancet 388, 1281–1292
secreting mesenchymal stromal cells repolarizes tumor- 90. Herz, J. et al. (2018) Interaction between hypothermia and de-
associated macrophages and suppresses neuroblastoma layed mesenchymal stem cell therapy in neonatal hypoxic-
proliferation in vivo. Stem Cells 36, 915–922 ischemic brain injury. Brain Behav. Immun. 70, 118–132
70. Song, S.Y. et al. (2020) Interleukin-4 gene transfection and 91. Popielarczyk, T.L. et al. (2019) Human bone marrow-derived
spheroid formation potentiate therapeutic efficacy of mesen- mesenchymal stem cells home via the PI3K-Akt, MAPK, and
chymal stem cells for osteoarthritis. Adv. Healthc. Mater. 9, Jak/Stat signaling pathways in response to platelet-derived
e1901612 growth factor. Stem Cells Dev. 28, 1191–1202
71. Gao, P. et al. (2010) Therapeutic potential of human mesen- 92. Shahror, R.A. et al. (2019) Enhanced homing of mesenchymal stem
chymal stem cells producing IL-12 in a mouse xenograft cells overexpressing fibroblast growth factor 21 to injury site in a
model of renal cell carcinoma. Cancer Lett. 290, 157–162 mouse model of traumatic brain injury. Int. J. Mol. Sci. 20, 2622
72. Chen, X. et al. (2008) A tumor-selective biotherapy with 93. Park, S.-R. et al. (2019) A novel endogenous damage signal,
prolonged impact on established metastases based on cyto- CSF-2, activates multiple beneficial functions of adipose tissue-
kine gene-engineered MSCs. Mol. Ther. 16, 749–752 derived mesenchymal stem cells. Mol. Ther. 27, 1087–1102
73. Jing, W. et al. (2014) Human umbilical cord blood-derived mesen- 94. Chen, M.-S. et al. (2018) IL-1β-Induced matrix metalloprotease-1
chymal stem cells producing IL15 eradicate established pancreatic promotes mesenchymal stem cell migration via PAR1 and
tumor in syngeneic mice. Mol. Cancer Ther. 13, 2127–2132 G-protein-coupled signaling pathway. Stem Cells Int.
74. Kim, N. et al. (2015) IL-21-expressing mesenchymal stem 2018, 3524759
cells prevent lethal B-cell lymphoma through efficient delivery 95. Perets, N. et al. (2019) Golden exosomes selectively target brain
of IL-21, which redirects the immune system to target the pathologies in neurodegenerative and neurodevelopmental dis-
tumor. Stem Cells Dev. 24, 2808–2822 orders. Nano Lett. 19, 3422–3432
75. Jeon, S.H. et al. (2018) Engineered mesenchymal stem cells ex- 96. Meng, S.-S. et al. (2018) LincRNA-p21 promotes mesenchy-
pressing stromal cell-derived factor-1 improve erectile dysfunction mal stem cell migration capacity and survival through hypoxic
in streptozotocin-induced diabetic rats. Int. J. Mol. Sci. 19, 3732 preconditioning. Stem Cell Res. Ther. 9, 280
76. Lou, G. et al. (2020) MiR-199a-modified exosomes from adi- 97. Antebi, B. et al. (2019) Cryopreserved mesenchymal stem cells
pose tissue-derived mesenchymal stem cells improve hepato- regain functional potency following a 24-h acclimation period.
cellular carcinoma chemosensitivity through mTOR pathway. J. Transl. Med. 17, 297
J. Exp. Clin. Cancer Res. 39, 4 98. Oja, S. et al. (2019) The utilization of freezing steps in mesenchymal
77. Kavari, S.L. and Shah, K. (2020) Engineered stem cells targeting stromal cell (MSC) manufacturing: potential impact on quality and
multiple cell surface receptors in tumors. Stem Cells 38, 34–44 cell functionality attributes. Front. Immunol. 10, 1622
78. Du, W. et al. (2017) Stem cell-released oncolytic herpes 99. Zhu, Y. et al. (2017) Bi-specific molecule against EGFR and
simplex virus has therapeutic efficacy in brain metastatic death receptors simultaneously targets proliferation and
melanomas. Proc. Natl. Acad. Sci. 114, E6157–E6162 death pathways in tumors. Sci. Rep. 7, 2602
79. Yoon, A.-R. et al. (2019) Mesenchymal stem cell-mediated de- 100. Zhang, X. et al. (2017) Mesenchymal stromal cells as vehicles
livery of an oncolytic adenovirus enhances antitumor efficacy in of tetravalent bispecific Tandab (CD3/CD19) for the treatment
hepatocellular carcinoma. Cancer Res. 79, 4503–4512 of B cell lymphoma combined with IDO pathway inhibitor
80. Moreno, R. et al. (2019) Enhanced antitumor efficacy of D-1-methyl-tryptophan. J. Hematol. Oncol. 10, 52
oncolytic adenovirus-loaded menstrual blood-derived 101. Liu, H. et al. (2006) The immunogenicity and immunomodula-
mesenchymal stem cells in combination with peripheral blood tory function of osteogenic cells differentiated from mesenchy-
mononuclear cells. Mol. Cancer Ther. 18, 127–132 mal stem cells. J. Immunol. 176, 2864–2872
81. Kaczorowski, A. et al. (2016) Delivery of improved oncolytic 102. Isakova, I.A. et al. (2014) Allo-reactivity of mesenchymal stem
adenoviruses by mesenchymal stromal cells for elimination of cells in rhesus macaques is dose and haplotype dependent
tumorigenic pancreatic cancer cells. Oncotarget 7, 9042 and limits durable cell engraftment in vivo. PLoS One 9, e87232

664 Trends in Pharmacological Sciences, September 2020, Vol. 41, No. 9

You might also like