Next Article in Journal
External Quality Assessment 2.0: The Importance of a Standardized Implementation of TILs for Daily and Trial Practices
Next Article in Special Issue
Mesenchymal Stromal Cells: From Therapeutic Option to Therapeutic Target
Previous Article in Journal
Knockdown of CKAP2 Inhibits Proliferation, Migration, and Aggregate Formation in Aggressive Breast Cancer
Previous Article in Special Issue
Fight the Cancer, Hit the CAF!
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mesenchymal Stem/Stromal Cells May Decrease Success of Cancer Treatment by Inducing Resistance to Chemotherapy in Cancer Cells

by
Taja Železnik Ramuta
and
Mateja Erdani Kreft
*
Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
*
Author to whom correspondence should be addressed.
Submission received: 31 May 2022 / Revised: 27 July 2022 / Accepted: 29 July 2022 / Published: 2 August 2022

Abstract

:

Simple Summary

Tumours consist of different cell types and an extracellular matrix, all of which together form a complex microenvironment. The tumour microenvironment plays a critical role in various aspects of tumour development and progression. Mesenchymal stem/stromal cells (MSCs) are multipotent stem cells that have a tri-lineage differentiation capacity and are one of the key stromal cells in the tumour microenvironment. Following the interaction with cancer cells, they are transformed from naïve MSCs to tumour-associated MSCs, which substantially affect tumour growth and progression as well as the development of chemoresistance in cancer cells. The aim of this review article is to provide an overview of studies that have investigated how MSCs affect the susceptibility of cancer cells to chemotherapeutics. Their results show that MSCs protect cancer cells from chemotherapeutics by influencing several signalling pathways. This knowledge is crucial for the development of new treatment approaches that will lead to improved treatment outcomes.

Abstract

The tumour microenvironment, which is comprised of various cell types and the extracellular matrix, substantially impacts tumour initiation, progression, and metastasis. Mesenchymal stem/stromal cells (MSCs) are one of the key stromal cells in the tumour microenvironment, and their interaction with cancer cells results in the transformation of naïve MSCs to tumour-associated MSCs. The latter has an important impact on tumour growth and progression. Recently, it has been shown that they can also contribute to the development of chemoresistance in cancer cells. This review provides an overview of 42 studies published between 1 January 2001 and 1 January 2022 that examined the effect of MSCs on the susceptibility of cancer cells to chemotherapeutics. The studies showed that MSCs affect various signalling pathways in cancer cells, leading to protection against chemotherapy-induced damage. Promising results emerged from the use of inhibitors of various signalling pathways that are affected in cancer cells due to interactions with MSCs in the tumour microenvironment. These studies present a good starting point for the investigation of novel treatment approaches and demonstrate the importance of targeting the stroma in the tumour microenvironment to improve treatment outcomes.

1. Introduction

Mesenchymal stem/stromal cells (MSCs) are multipotent stem cells that have atri-lineage differentiation capacity, namely, into osteoblasts, chondrocytes, and adipocytes [1,2,3]. They express the surface markers CD73, CD105, and CD90 [1,4]. MSCs can be isolated from various sources, such as the bone marrow [5], adipose tissue [5], dental pulp [6], umbilical cord [7], placenta [8], endometrium [9], amniotic fluid [10], skeletal muscle tissue [11], liver tissue [5], and other tissues [1,2,12]. MSCs are capable of homing to various injury (inflammation) sites, where they support tissue repair, stem cell homeostasis, and immunomodulation [12,13,14]. As the tumour microenvironment is similar to the microenvironment of chronic wounds, it attracts MSCs, especially those derived from the bone marrow, adipose tissue, and umbilical cord [14]. The homing and migration of MSCs are mediated by tumour-cell-specific receptors, the extracellular matrix, and various inflammatory cytokines, chemokines, and growth factors (e.g., MCP-1, SDF-1, IL-8, TGF-β, and VEGF) [12,15,16,17]. Moreover, a pro-inflammatory toll-like receptor ligand LL-37 has been linked to tumour-homing, as various tumour types (e.g., ovarian, breast, and lung cancer) demonstrated the overexpression of LL-37, which acted as a proliferative signal and pro-angiogenic factor and promoted the migration of MSCs into the tumour microenvironment, while its inhibition resulted in reduced tumour growth [18].
Tumours are comprised of various cell types and extracellular matrix, which together form a complex microenvironment. It has been shown that the tumour stroma, which is comprised of MSCs, tumour vasculature, infiltrating inflammatory and immune cells, extracellular matrix, and fibroblasts, plays a crucial role in tumour initiation, progression, and metastasis [19,20]. Furthermore, MSCs are one of the crucial stromal cells in the tumour microenvironment, and their interactions with tumour cells and various other components of the tumour microenvironment lead to the transformation from naïve MSCs to tumour-associated MSCs [21,22]. The latter are implicated in tumour growth and progression as well as the development of chemoresistance in cancer cells [23,24,25]. Since the distinction between “naïve” and “tumour-associated” MSCs is not consistently listed in most studies, we will use the general term MSCs in this review unless otherwise stated. However, more attention needs to be paid to these specific stages of MSCs in the future.
In many patients, the initial response to anticancer agents is favourable and improves disease-free survival. However, many tumours eventually become resistant to therapy, leading to relapse and the recurrence of cancer. Hence, it is of the utmost importance to elucidate the mechanisms that contribute to chemoresistance, which will lead to the development of novel strategies to overcome this issue.

2. The Ambivalent Nature of MSCs in the Tumour Microenvironment

In recent years, numerous studies have investigated the properties of MSCs in the tumour microenvironment, which have been reported in detail elsewhere [21,23,25,26,27]. Hence, herein we will briefly describe the properties of MSCs and then focus on the effects of MSCs on chemotherapeutic agents in the tumour microenvironment.
MSCs present an integral part of the tumour microenvironment and largely [28] contribute to tumour progression and metastasis [29]. First, MSCs are recruited to the tumour microenvironment, where the direct interactions between the tumour cells, MSCs, and tumour-secreted factors lead to the development of a pro-tumourigenic subpopulation of MSCs, i.e., tumour-associated MSCs [21,30,31]. These contribute to tumour progression by (a) differentiating into other pro-tumourigenic cells in the tumour microenvironment (e.g., cancer-associated fibroblasts) [14,32,33], (b) suppressing the immune response, (c) promoting the epithelial-to-mesenchymal transition, (d) promoting angiogenesis, (e) enriching the cancer stem cell population [29,30,34,35,36], and (f) promoting tumour cell survival and metastasis [14,21,24,33,37].
When using naïve MSCs in in vitro and in vivo studies, researchers have shown that the cells possess tumour-promoting and tumour-suppressing effects. Several studies demonstrated the capability of MSCs to promote tumour survival and angiogenesis, enhance invasion and metastatic potential, and inhibit the apoptosis of cancer cells, thus supporting tumour growth [24,38,39,40,41,42]. MSCs have been shown to release factors such as VEGF, FGF-2, PDGF, HGF, BDNF, SDF-1α, IGF-1, IGF-2, TGF-β, TNFα IGFBP-2, LIF, M-CSF, MIP-2, IL-8, IL-6, and IFNγ that promote tumour survival, proliferation, migration, invasion [21,43,44,45,46,47], and angiogenesis [21,24,25,33,48,49,50,51,52]. Furthermore, it has been shown that cancer cells induce epigenomic mesenchymal-to-epithelial transition in MSCs, resulting in pro-tumorigenic functions of MSCs, which then serve as a metastatic driver/chaperone [28]. Interestingly, other cell types in the tumour microenvironment may also transform into MSC-like cells and drive tumour resistance to cytotoxic agents, which was demonstrated in the case of glioblastoma, as the Wnt-mediated endothelial cell transformation resulted in MSC-like cells [53].
In addition, MSCs promote the epithelial-to-mesenchymal transformation by secreting CCL5, which promotes the secretion of matrix metalloproteinases that loosen the extracellular matrix in the tumour microenvironment and consequently contribute to cancer cell motility [21,54]. MSCs also have immunosuppressive properties that facilitate cancer cells to escape immune surveillance. They secrete several mediators, e.g., TGFβ, TNFα, IFNγ, indoleamine 2,3-dioxygenase (IDO), IL-1β, IL-1α, IL-4, and IL-6, and interact with various immune cells, all leading to immunosuppression [25,55,56,57]. For example, MSCs promote CD8+ T-cell tumour immune exclusion and decrease the response to anti-PD-L1 immune checkpoint inhibitor due to the secretion of the chemokines CCL2, CX3CL1, and TGFβ1 [58].
On the other hand, in some cases, MSCs are also capable of inhibiting tumour growth, which has been shown, for example, in the in vitro and in vivo models of breast cancer, Kaposi’s sarcoma, liver cancer, and melanoma [1,59,60,61,62]. The MSCs suppress tumour growth by inhibiting the signalling pathways associated with proliferation (e.g., STAT3, AKT, PI3K, and Wnt signalling pathways), cell cycle progression [13,52,60,63,64,65,66], the inhibition of apoptosis, and the suppression of angiogenesis [13,25,41,52,59,60,61,62,63,67,68,69,70,71,72]. The anticancer properties of MSCs have been reported in more detail elsewhere [19,73,74].
Another important point to consider is the variation in the structure of the tumour microenvironment among different cancers, as it has been shown that tumour microenvironments differ in terms of cell types and the composition of the extracellular matrix [75,76,77]. Moreover, crosstalk between MSCs and other cell types in the tumour microenvironment and certain properties of the microenvironment (e.g., pH, O2 concentration, and ion gradients) importantly affect MSC development and characteristics and the heterogeneity of the population [73,78]. To conclude, the ambivalent nature of MSCs was well-demonstrated by Ramasamy et al. (2007). They showed that naïve MSCs inhibit the proliferation of cancer cells in vitro, but the injection of MSCs into nude mice promotes tumour growth [38]. While the discrepancies in the effect of MSCs are partly due to the heterogeneity of the MSC populations and the different experimental conditions [13,19], these findings also underline the importance of considering the contribution of non-cancer cells in the tumour microenvironment to disease development and the response to treatment.

3. MSCs Promote Resistance of Cancer Cells to Chemotherapeutic Agents

Drug resistance may result from genetic changes in cancer cells (intrinsic resistance) or from the tumour microenvironment (extrinsic resistance) [29,79]. The latter promotes drug resistance by (1) preventing drug penetration into the tumour due to changes in the composition of the extracellular matrix, (2) by secreting protective cytokines, or (3) by changing gene expression in tumour cells, which leads to decreased cytotoxicity of chemotherapeutics [79,80,81,82,83] (Figure 1). As chemotherapeutics induce damage in the tumour microenvironment, this results in a stress response in MSCs and cancer-associated fibroblasts, which secrete various growth and anti-apoptotic factors (e.g., IL-6, IL-7, IL-8, CXCL12, and VEGF) that promote cell survival, proliferation, invasion, and the metastasis of cancer cells [84,85,86,87,88,89,90,91]. Furthermore, the latest studies show that MSCs also play an important role in the development of drug resistance, which is the main focus of this review article.
To investigate this topic in detail, we performed a comprehensive literature search using PubMed to identify articles investigating the effects of MSCs on chemotherapy resistance. We used the following search terms: ((“mesenchymal stem”) OR (“mesenchymal stromal”) OR (“MSCs”) AND (“chemotherapy”) AND (“resistance”)) and included only articles in which these terms appeared in the title and/or the abstract. Only full-text articles that were published in the English language between 1 January 2001 and 1 January 2022 were included. We identified 224 articles through database research, and after the removal of duplicates and review articles, editorials, and conference proceedings, there were 155 articles that were assessed in detail. Next, we excluded the articles that were irrelevant for the topic of this review article. The remaining 42 articles were included in the analysis, and their conclusions are presented in the following chapters and Table 1.

3.1. Blood Cancers

3.1.1. Acute Lymphoblastic Leukaemia

To investigate the mechanism of resistance to chemotherapy-induced apoptosis in B-cell acute lymphoblastic leukaemia (B-ALL), Yu et al. (2020) focused on the protein tyrosine phosphatase Shp2, which is highly expressed in bone marrow MSCs of patients. The plasmid-induced Shp2 activation in MSCs led to an increase in chemotherapy-resistant cancer cells in the in vitro and in vivo settings. Moreover, they demonstrated that Shp2 activation in MSCs upregulates vascular cell adhesion molecule 1 (VCAM-1) expression by increasing the PI3K/AKT phosphorylation level. Moreover, the inhibition of very late antigen-4 (VLA-4) reversed the chemotherapy resistance, which indicates that targeting VCAM-1/VLA-4 signalling may have clinically relevant value [92]. Its importance was also demonstrated by Jacamo et al. (2014), who showed that VCAM-1/VLA-4 signalling plays an integral role in the activation of NF-κB in leukaemia and stromal cells, which is also crucial for promoting the chemoresistance of cancer cells [93].
The CXCR/CXCL12 axis plays a crucial role in the homing of hematopoietic stem cells and ALL cells to the bone marrow microenvironment, and the overexpression of CXCR4 in ALL patients is associated with disease progression and poor prognosis [94,95]. Wang et al. (2020) cultured cells from patients with relapsed/refractory ALL with MSCs, which resulted in a higher expression of CXCR4 and CXCL12 in the presence of the chemotherapeutic agent vincristine. However, when the CXCR4 antagonist AMD3100 was added, the expression of CXCR4 and CXCL12 was reduced, which showed that blocking the CXCR4/CXCL12 axis results in sensitizing the relapsed/refractory ALL cells to chemotherapeutic agents [96]. Moreover, Pillozzi et al. (2019) identified the novel peptide 4-1-17 and the small molecule 8673 for targeting the CXCR4/CXCL12 axis, the use of which, in an in vitro setting, induced a pro-apoptotic effect that was not reduced by the presence of MSCs. The treatment using both novel tools was capable of overcoming the MSC-induced resistance to the chemotherapeutic agent cytarabine in ALL-derived cell lines [97].
The Wnt signalling pathway regulates cell proliferation, survival, differentiation, and migration and is also involved in MSC-mediated drug resistance [98]. Yang et al. (2013) grew co-cultures of ALL cells and MSCs and found that MSCs protect cancer cells from spontaneous cytarabine-induced apoptosis, which results from the decreased cleavage of apoptotic proteins and the modified expression of various cell-cycle-related proteins. Moreover, they also demonstrated that MSCs activate the Wnt signalling pathway in ALL cells. Importantly, by using a Wnt signalling inhibitor, the susceptibility of ALL cells to chemotherapy was improved [99].
In the case of T-cell ALL, MSCs contribute to the development of chemoresistance by accepting mitochondria from cancer cells that underwent chemotherapy-induced intracellular oxidative stress. The transfer is mediated by tunnelling nanotubes, and by inhibiting the mitochondria transfer using cytochalasin D, the capacity of MSCs to protect cancer cells from a chemotherapeutic agent decreased. Interestingly, in the co-culture setting, most of the cancer cells adhered to MSCs, and researchers showed that the adhesion is ICAM-1-mediated and contributes to MSC-induced chemoresistance [100].

3.1.2. Acute Myeloid Leukaemia

Various studies have demonstrated that MSCs protect cancer cells from chemotherapy-induced genotoxicity. For example, Gynn et al. (2021) showed that primary MSCs in co-culture with leukaemic HL-60 cells are sensitized to cytarabine-induced genotoxicity, while leukaemic cells remain protected from the agent. Moreover, primary MSCs from patients who had undergone chemotherapy show reduced survival and increased genotoxicity in comparison to patients’ MSCs at diagnosis, and MSCs also retain the long-term genotoxic and functional damage following exposure to cytarabine [101]. Moreover, patient-derived data show that the poor therapy response in patients with AML correlates with increased quiescence. The phenomenon was also confirmed in an in vitro study in which co-cultures of MSCs and AML cells (in direct contact and in the transwell setting) showed that MSCs protect cancer cells from the apoptosis induced by the chemotherapeutic agent cytarabine by enriching the population of quiescent cells [102].
MSCs secrete TGF-β1 and CXCL12, which are the key factors in the bone marrow niche that govern hematopoietic cell proliferation and survival; TGF-β1 maintains quiescent hematopoietic stem cells, while CXCL12 promotes their homing and anchoring in the niche [103,104]. The co-culture of bone marrow MSCs and AML cells led to a higher proliferation capacity of AML cells in comparison to a monoculture. Moreover, the blockade of TGF-β1 increased the proliferation and chemosensitivity of AML cells, while the addition of a CXCR4 antagonist resulted in anti-proliferative effects, but it did not affect the susceptibility of AML cells to the chemotherapeutic cytarabine [105].
Increased aldehyde dehydrogenase (ALDH) activity in AML cells is associated with greater tumourigenicity and chemotherapy resistance. Yuan et al. (2020) demonstrated that the ratio of ALDH-positive cells increased when cancer cells were incubated with MSCs, and further analysis revealed that the TGF-β1-regulated gene signature was activated in cancer cells when they were in co-culture with MSCs. Further data demonstrated that the knockdown of TGF-β1 in MSCs inhibited stroma-induced ALDH activity in cancer cells, while on the other hand, the treatment with recombinant TGF-β1 induced ALDH activity in cancer cells. Moreover, the TGF-β1-induced ALDH2 expression in AML cells was regulated by the non-canonical pathway through the activation of p38. Importantly, the use of two inhibitors of ALDH2 led to the sensitization of AML cells to chemotherapy, even in the presence of MSCs [106].
Dong-Feng et al. (2014) investigated the role of the connection between the leukaemic stem cells and the hematopoietic microenvironment, mediated by the thrombopoietin/c-MPL pathway, in developing chemoresistance. They showed that when in co-culture with osteoblasts differentiated from bone-marrow-derived MSCs from acute myeloid leukaemia (AML) patients, cancer cells secreted more thrombopoietin and were more resistant to the chemotherapeutic daunorubicin than when co-cultured with osteoblasts derived from normal control MSCs. Hence, their results indicate that a high level of thrombopoietin/c-MPL signalling may contribute to increased chemoresistance in AML [107].
In 2014, Xia et al. demonstrated that bone-marrow-derived MSCs protect AML cells from chemotherapy-induced apoptosis via the c-Myc-dependent pathway [108]. Next, in 2017, the same research group showed that in the co-culture with MSCs, AML cells decreased the expression of miRNA-494, which is one of the c-Myc regulators, and its downregulation is associated with poor prognosis in AML patients. Moreover, the activation of miR-494 in AML cells in combination with mitoxantrone in vitro led to suppressed proliferation and induced apoptosis and in vivo resulted in suppressed tumour growth and prolonged survival in mice. Therefore, their results indicated that miR-494 suppresses drug resistance in AML cells by downregulating c-Myc [109].
O’Reilly et al. (2018) showed that bone-marrow-derived MSCs promote the resistance of AML cells to the chemotherapeutic agents cytarabine and daunorubicin and to BH3 mimetics. One of the most studied mechanisms of chemoresistance is the activation of pro-survival signal transduction and the induction of anti-apoptotic Bcl-2 proteins [110,111]. Moreover, another anti-apoptotic protein, myeloid cell leukemia-1 (Mcl-1) protein, acts as a master regulator of apoptosis [112]. The results of O’Reilly et al. show that chemotherapy resistance is mediated by MSC-induced Mcl-1 expression over Bcl-2 and/or Bcl-XL in AML cells, and the inhibition of Mcl-1 restores sensitivity to BH3 mimetics, while the combined inhibition of Bcl-2/Bcl-XL and Mcl-1 restores susceptibility to cytarabine and daunorubicin [110].
Prostaglandin E2 (PGE2) suppresses apoptosis in multiple cell types, and it has been shown that malignant cells release IL1β, which induces PGE2 synthesis in MSCs, which is followed by the activation of β-catenin signalling and the induction of the cancer stem cell phenotype. PGE2 is an enzymatic product of Cox-2, and both proteins are involved in inflammation and cancer, although the role and regulation of PGE2 in the case of AML are not well-documented [113,114]. Carter et al. (2016) showed that apoptosis repressor with caspase recruitment domain (ARC) protein induces the expression of IL-1β in cancer cells when they are in co-culture with MSCs and increases the expression of CCL2, CCL4, and CXCL12 in MSCs. Hence, ARC confers chemoresistance by controlling the interactions between cancer cells and their microenvironment through an NFκB/IL1β signalling network. Importantly, the inhibition of IL1β suppressed cancer cell migration and increased their susceptibility to chemotherapeutics [115]. Next, the same research group showed that co-cultures of AML cells and MSCs promoted COX-2 expression in MSCs and PGE-2 production. The latter induces the expression of β-catenin, which regulates the protein ARC and promotes chemoresistance in AML cells. Furthermore, the study showed that ARC is an integral component of the IL1β/COX-2/PGE2/β-catenin circuit and that ARC, which is regulated by β-catenin, mediates AML-stromal interactions. Importantly, the inhibition of β-catenin led to a decrease in ARC and sensitized AML cells to chemotherapy [114].

3.1.3. Chronic Lymphocytic Leukaemia

In 2012, Nwabo Kamdje et al. investigated the role of stromal-cell-mediated signalling in chronic lymphocytic leukaemia resistance to chemotherapy. They showed that when cancer cells were in co-culture with autologous or allogeneic bone-marrow-derived MSCs, the latter protected cancer cells from spontaneous or chemotherapy-induced apoptosis. Moreover, following treatment with a combination of anti-Notch-1, anti-Notch-2, and anti-Notch-4 antibodies or γ-secretase inhibitor XII, cancer cells had increased susceptibility to chemotherapeutics [116]. Similarly, Kurtova et al. (2009) and Severin et al. (2019) demonstrated that human and murine MSCs are effective in protecting chronic lymphocytic leukaemia cells from fludarabine-, dexamethasone-, cyclophosphamide-, and ibrutinib-induced apoptosis. Interestingly, Kurtova et al. showed that the MSCs triggered this effect only when in cell–cell contact with cancer cells but not when cultivated in a transwell setting [117,118].

3.1.4. Multiple Myeloma

Yang et al. (2017) investigated the effect of bone-marrow-derived MSCs from patients with multiple myeloma on cell lines of multiple myeloma. They showed that MSCs decreased melphalan- or doxorubicin-induced cell cycle arrest and apoptosis. Moreover, MSCs promoted the expression of autophagy-related genes in cancer cells, leading to the activation of autophagy and subsequently NF-κB signalling [119], which promotes the proliferation, survival, and drug resistance of cancer cells [120]. When NF-κB signalling was inhibited, MSCs no longer had a protective effect on cancer cells. Using autophagy inhibitors (chloroquine and 3-methyladenine), it was shown that treatment suppressed the phosphorylation and consequent degradation of NF-κB inhibitor (I-κBα), which prevented the resistance of cancer cells to chemotherapeutic agents [119]. Furthermore, it has been shown that bone-marrow-derived MSCs, together with the non-cellular components, promote protective endoplasmic reticulum stress in multiple myeloma cells, thus bolstering their resistance to melphalan and bortezomib [121].

3.2. Glioma

The conditioned medium of glioma-associated MSCs has been shown to induce the overexpression of FOXS1 in glioma cells, which leads to the resistance of cancer cells to the chemotherapeutic agent temozolomide. Interestingly, the researchers identified two subpopulations of glioma-derived MSCs that play distinct roles in tumour progression. The CD90high population of MSCs increases glioma cell proliferation, migration, and adhesion, while the CD90low population of MSCs is involved in the development of glioma cell resistance to chemotherapy [122,123].
On the other hand, de Castro et al. (2017) showed that a conditioned medium from Wharton’s jelly-derived MSCs promoted the viability, proliferation, and migration of glioblastoma cell lines, but it did not significantly affect the susceptibility of cancer cells to temozolomide [124].

3.3. Neuroblastoma

Lifshitz et al. (2017) obtained the conditioned medium from bone-marrow-derived MSCs from patients with neuroblastoma and applied it to the neuroblastoma cell line CHLA-255. They showed that MSCs promote the expression of sphingosine-1-phosphate receptor 1 (S1PR1) expression in cancer cells as well as the phosphorylation of its downstream molecules JAK2 and STAT3 [125]. This leads to increased cell proliferation, the suppression of apoptosis, improved cell survival, and chemoresistance [126,127]. Next, they prepared an S1PR1 knockdown in neuroblastoma cells by shRNA, which enhanced chemotherapeutic agent etoposide-induced apoptosis in neuroblastoma cell lines. Similarly, the treatment of neuroblastoma cell lines with an antagonist of S1PR1 significantly increased the susceptibility of cancer cells to etoposide. On the other hand, the overexpression of S1PR1 protected the cancer cells from chemotherapy-induced apoptosis by activating JAK-STAT3 signalling, supporting the claim that S1PR1 plays an important role in chemoresistance [125].

3.4. Oral Squamous Cell Carcinoma

Receptor tyrosine kinases, such as the platelet-derived growth factor receptor (PDGFR), play a crucial role in promoting cell growth and proliferation [128]. In the case of oral squamous cell carcinoma (OSCC), it was shown that in the co-culture of OSCC cells and MSCs the latter activated a PDGF-AA/PDGFR-α autocrine loop, which led to reduced cancer cell sensitivity to cisplatin due to alterations in apoptosis. The authors concluded that the crosstalk between cancer cells and MSCs is at least in part mediated by the activation of PDGFR-α/AKT signalling pathways [129].

3.5. Head and Neck Cancers

The resistance of cancer cells to chemotherapeutics as a result of an interaction with MSCs is also induced in an epigenetic manner. This was demonstrated by Liu et al. (2021), who showed that the presence of MSCs supported the resistance of head and neck cancer lines (SCC-25, HSC-2) to paclitaxel by affecting the expression of genes that have been implicated in drug resistance (e.g., GDF15, CXCL11, TFP1-2, POSTN, IGFBP5, and DAPK1). The effect of MSCs on the drug resistance of cancer cells lasted as long as 30 days after the exposure of cancer cells to MSCs, while the largest effect was recorded at 3 days after exposure to MSCs [130].
Moreover, MSCs had a protective effect on head and neck cancer cell lines in the presence of paclitaxel, as the co-culture led to the decreased expression of cleaved-caspase 3 and cleaved-PARP-1 in cancer cells, which is associated a with lower level of drug-induced apoptosis [130].

3.6. Breast Cancer

Ullah et al. (2019) injected bone-marrow-derived MSCs and breast cancer cells into NOD/SCID mice and observed that xenograft tumours grew smaller in comparison to tumours induced by breast cancer cells alone but exhibited resistance to chemotherapeutic agents. Moreover, they showed that the chemoresistance was associated with an increased expression of tetraspanins (CD9 and CD81) and the drug resistance proteins BCRP and MDR1. Their findings showed that crosstalk between MSCs and cancer cells can be attributed to CD9, the overexpression of which is associated with a higher risk of invasion and metastasis as well as the promotion of the expression of BCRP and MDR1, which is mediated via CCL5, CCR5 and CXCR12 [131]. Next, Yeh et al. (2017) treated the MDA-MB-231 breast cancer cell line with the conditioned medium from adipose-derived MSCs isolated from breast cancer patients. They demonstrated that MSCs secreted CXCL1, which promoted doxorubicin resistance. MSCs downregulated miR-106a expression, which enhanced the expression of ATP-binding cassette transporters (ABCG2) through which doxorubicin is removed from the cell [132].
Luo et al. (2020) demonstrated that exosomes secreted by MSCs promote chemoresistance in breast cancer by enhancing the miR-21-5p-mediated expression of S100A6, a chemoresistance gene. Moreover, doxorubicin induced the expression of miR-21-5p, which was necessary for the expression of the S100A6 gene in cancer cells. However, silencing miR-21-5p expression in cancer cells resulted in cancer cells being susceptible to doxorubicin [133].
Lu et al. (2017) co-cultured breast cancer cells (MCF-7/ADR cell line) with adipose-derived MSCs and investigated the effect of MSCs on the expression of C-terminal Src kinase-binding protein (Cbp), which is a transmembrane adaptor protein that regulates Src family kinase activities. The presence of MSCs induced the increased expression of Cbp and increased cell proliferation and chemoresistance. Using RNA interference, the presence of Cbp in breast cancer cells was depleted, which led to the inhibition of cell proliferation and invasion. Moreover, in these conditions, the use of doxorubicin induced the suppression of tumour growth. The silencing of Cbp was followed by the inhibition of the expression of phosphoryl Src, AKT, and mTOR signals. Therefore, these results indicate that the PAG1/Cbp pathway is involved in breast cancer tumour progression and acquired chemoresistance and that the PAG1/Cbp pathway modulates MSC-promoted tumourigenesis via the Src and AKT/mTOR pathways [134].
Barneh et al. (2018) demonstrated that valproic acid, which is already being used in the clinic for the treatment of seizures and bipolar disorder [135], inhibits the protective effects of MSCs against chemotherapy in breast cancer. Using the proteomic dataset, the research group showed that secreted cytokines from MSCs activated various signalling pathways in cancer cells, including the JAK-STAT, PI3K, MAPK, and TGFβ signalling pathways, which lead to the activation of NF-kB transcription factor. Next, they showed that valproic acid affects the expression of 34 genes within the identified pathways, and in vitro experiments confirmed that valproic acid inhibits NF-kB activation in cancer cells. Moreover, the patients taking oral valproic acid demonstrated decreased expression of antioxidant enzymes, which led to increased oxidative stress in cancer cells. Hence, valproic acid hinders the protective role of MSCs by inhibiting the mechanisms allowing the adaptation of cancer cells to oxidative stress [136].

3.7. Lung Adenocarcinoma

Wang et al. (2020) showed that, in the tumour microenvironment of lung adenocarcinoma, bone-marrow-derived MSCs produced molecules associated with adipocytes, which promoted resistance to the chemotherapeutic agent erlotinib in cancer cells by activating the insulin-like growth factor 1 receptor (IGF-1R). The MSCs in the hypoxic tumour microenvironment produced leptin, which is capable of inducing erlotinib resistance in lung adenocarcinoma cells through the activation of IGF-R1 signalling in the absence of IGF-1 in hypoxia. On the other hand, IGF binding protein 2 (IGFBP2) counteracted the activation of IGF-1 and induced erlotinib resistance by activating IGF-1R signalling in an IGF-independent manner. Moreover, the inhibition of IGF-1R with the inhibitor BMS754807 reversed the erlotinib resistance [137].

3.8. Gastric Cancer

Gastric-cancer-associated MSCs promote the resistance of gastric cancer cells to chemotherapy through the secretion of IL-8 and the consequent induction of PD-L1 [138]. The overexpression of PD-L1 affects the therapeutic efficacy of chemotherapy and is associated with a shorter survival period for patients [139]. Importantly, the sensitivity of gastric cancer cells to chemotherapy was enhanced when PD-L1 was blocked [138]. Another study showed that the co-culture of gastric cancer cells with MSCs induced the long non-coding RNA (lncRNA) HCP5 (histocompatibility leukocyte antigen complex P5), which drove fatty acid oxidation through the miR-3619-5p-AMPK-PGC1α/CEBPB axis and resulted in the promotion of stemness and the induction of chemoresistance [140]. Moreover, He et al. (2019) demonstrated that MSCs promote the stemness and chemoresistance of gastric cancer cells through fatty acid oxidation in vitro and in vivo. The MSCs secrete TGF-β1, which activates SMAD2/3 and induces long non-coding RNA (lncRNA) MACC1-AS1 expression in cancer cells. MACC-AS1 promotes chemoresistance by antagonizing miR-145-5p. Importantly, the inhibition of fatty acid oxidation by the inhibitor etomoxir attenuated the MSC-induced chemoresistance in vivo and therefore underlined the importance of targeting fatty acid oxidation [141].
In 2015, Ji et al. showed that MSC-derived exosomes significantly increased the chemoresistance of gastric cancer cells to 5-fluorouracil, resulting in decreased apoptosis and the increased expression of MDR, MRP, and LRP proteins, which are associated with multidrug resistance, leading to an enhanced capability of cancer cells to expel chemotherapeutics. In cancer cells, the exosomes of MSCs triggered the activation of calcium/calmodulin-dependent protein kinases (CaM-Ks) and the Raf/MEK/ERK cascade. Importantly, the tumour-promoting role of MSC-derived exosomes in chemoresistance was inhibited by blocking the CaM-KS/Raf/MEK/ERK pathway [142].
In 2016, Ji et al. showed that bone-marrow-derived MSCs increase drug resistance in CD133-expressing gastric cancer cells, which have been shown to possess cancer stem cell properties. Moreover, MSCs triggered the activation of the PI3K/AKT pathway in CD133-positive cells, resulting in decreased apoptosis and increased chemoresistance of CD133-positive cells due to the upregulation of Bcl-2 and the downregulation of BAX. Importantly, blocking the PI3K/AKT pathway prevented the promotion of chemoresistance, and they also showed that MSCs had no effect on drug resistance in CD133-knockdown cells, highlighting the involvement of CD133 in this process [143].

3.9. Pancreatic Adenocarcinoma

Timaner et al. (2018) showed that MSCs, which homed to pancreatic adenocarcinoma and were located in close proximity to tumour-initiating cells, promoted the enrichment of tumour-initiating cells in vitro and enhanced tumour growth in vivo. These effects were due to the secretion of CXCL10 by gemcitabine-treated MSCs, which led to activation of the CXCL10-CXCR3 axis and subsequently to the activation of the STAT3 signalling pathway in tumour-initiating cells. Importantly, when the tumour-initiating cells were treated with gemcitabine and, at the same time, the CXCL10-CXCR3 axis was disrupted by the treatment with nanovesicles that contained CXCR3 antagonist, the treatment delayed tumour regrowth. Interestingly, they also showed that the MSC-dependent accumulation of tumour-initiating cells depends on the cell type, as this effect occurred in pancreatic adenocarcinomas but not in lung cancer, although both tumours express CXCR3 [144].

3.10. Genitourinary Cancers

Yu et al. (2021) showed that MSCs reduce the sensitivity of castration-resistant prostate cancer to docetaxel. Interestingly, the co-culture of cancer cells with MSCs in the presence of docetaxel led to an increase in autophagy activation, but the application of the autophagy inhibitor effectively reversed the resistance to docetaxel. Moreover, the increase in autophagy activation in the presence of docetaxel depended on TGF-β1 secretion, and the inhibition of TGF-β1 secretion increased the sensitivity of cancer cells to docetaxel [145].
Maj et al. (2016) investigated the effect of amniotic-fluid-derived MSCs and adipose-derived MSCs on the susceptibility of the 786-O human renal carcinoma and T24 bladder carcinoma cell lines to ciprofloxacin. They demonstrated that the MSC-derived conditioned medium reduced the sensitivity of cancer cells to ciprofloxacin and that the viability of cancer cells was 2.4–2.6-times higher when cells were incubated with ciprofloxacin in the presence of MSC-derived conditioned medium than in the presence of standard culture medium [146]. Similarly, conditioned medium from adipose MSCs reduced cisplatin-induced apoptosis in epithelial ovarian cancer cells, which resulted in cisplatin resistance and the increased viability of cancer cells [147].
Furthermore, Coffman et al. (2016) isolated cancer-associated MSCs from patient-derived tissue from patients with ovarian cancer. They demonstrated that Hedgehog secreted by ovarian cancer cells induces BMP4 expression in MSCs, which leads to the increased expression of Hedgehog in ovarian cancer cells, indicating a positive feedback loop [148]. Hedgehog signalling is crucial in ovarian cancer, as it is associated with increased chemoresistance and decreased survival [149,150]. Next, using a Hedgehog inhibitor or a BMP4-blocking antibody resulted in a decreased level of BMP4 derived from MSCs, which reversed resistance to cisplatin [148].

3.11. Liver Cancer

Han et al. (2014) treated hepatocellular carcinoma cells (SMMC-7721, Hep-G2 cell lines) with the conditioned medium derived from MSCs stimulated with IFN-γ and/or TNF-α. They demonstrated that in the presence of the MSC-derived conditioned medium the cancer cells underwent autophagy, which protected them against the cell toxicity of chemotherapeutics. However, when treated with an autophagy inhibitor, the cancer cells became susceptible to the chemotherapeutic agent 3-methyladenine. Additionally, they also showed that the stimulation of MSCs with IFN-γ and TNF-α promoted the expression of TGF-β in MSCs and that the upregulation of TGF-β correlates with chemoresistance in cancer cells. Moreover, the inhibition of TGF-β expression in MSCs diminished the capability of MSCs to induce autophagy, which resulted in diminished chemoresistance in cancer cells [151].

3.12. Bone Cancers

Tu et al. (2016) obtained the conditioned medium from bone-marrow-derived MSCs and used it for the treatment of the human osteosarcoma cell lines Saos-2 and U2-OS. They showed that MSCs had a protective role, as the presence of MSC-derived conditioned medium protected cancer cells from apoptosis when cultured with the chemotherapeutic agents doxorubicin or cisplatin. STAT3 activation by IL-6 has been shown to regulate MSC-induced chemoresistance, and the blockage of STAT3 activation (with AG490, a JAK2 inhibitor) improved the susceptibility of cancer cells to chemotherapeutic agents. Furthermore, when osteosarcoma mice were injected with MSCs and Saos-2 cells, the inhibition of STAT3 improved survival by suppressing tumour growth and increasing their susceptibility to doxorubicin. The role of STAT3 in chemoresistance was further supported by data obtained from osteosarcoma samples from patients, namely, increased expression of p-STAT3, MRP (multidrug resistance protein), and MDR-1 (P-glycoprotein) correlated with chemoresistance [152].
Table 1. Overview of studies investigating the effects of MSCs on chemotherapeutic-agent-treated cancer cells. Bold text indicates the crucial effects of MSCs on chemotherapeutic-agent-treated cancer cells.
Table 1. Overview of studies investigating the effects of MSCs on chemotherapeutic-agent-treated cancer cells. Bold text indicates the crucial effects of MSCs on chemotherapeutic-agent-treated cancer cells.
Type of CancerSource of MSCsIn Vitro ModelIn Vivo ModelChemotherapeutic AgentEffects of MSCs
on Chemotherapeutic-Agent-Treated Cancer Cells
References
ALLHuman umbilical-cord-derived MSCsHuman patient-derived ALL cells N/AVincristineMSCs mediated vincristine resistance by blocking the CXCR4/CXCL12 axis.[96]
Human bone-marrow-derived MSCs from ALL patients and healthy volunteersHuman CCRF-SB B lymphoma cell lineNOD/SCID mice VincristineProtein tyrosine phosphatase 2 (Shp2) activation in the BMSC upregulates VCAM-1 expression through activating the PI3K/AKT signalling pathway, which thereby interacts with VLA-4 in B-ALL cells to resist vincristine.[92]
Human bone-marrow-derived MSCsHuman B-cell precursor-ALL cells and AML cells; human normal Epstein–Barr-virus-infected B lymphocytes N/APeptide 4-1-17, small molecule 8673, cytarabine, doxorubicinPeptide 4-1-17 and small molecule 8673 inhibit leukaemia cell proliferation and induce a pro-apoptotic effect that is not reduced by the presence of MSCs. The combined treatment with 4-1-17 and 8673 has a stronger pro-apoptotic effect, particularly on cells cultured on MSCs in normoxic and hypoxic conditions, and can overcome MSC-induced resistance to cytarabine.[97]
Human bone-marrow-derived MSCsHuman T-cell ALL cell line Jurkat; human primary T-cell ALL cellsN/ACytarabineThe chemotherapeutic drug causes intracellular oxidative stress in Jurkat cells. Jurkat cells transfer mitochondria to MSCs via tunnelling membrane nanotubes but receive few mitochondria from MSCs, resulting in chemoresistance.[100]
Human bone-marrow-derived MSCs from healthy volunteersHuman leukaemia pre-B ALL cells; human primary ALL mononuclear cells N/AEverolimusBM stromal cells enhance HIF-1α expression under hypoxia, leading to an HIF-1α-dependent upregulation of glucose transport and a switch to glycolytic metabolism in leukaemic cells and primary ALL blasts. Downregulation of HIF-1α expression or blockade of mTOR signalling with everolimus promoted chemosensitivity.[153]
Human bone-marrow-derived MSCs, human MSC line HS-5, murine MSC line M2-10B4The human leukaemia cell lines Reh, RS4;11, and SEMK2 NOD/SCID mice CytarabineMSCs induce activation of the Wnt pathway in ALL cells, and this activation contributes to the survival of ALL cells. Blocking the Wnt pathway with the β-catenin inhibitor XAV939 partially overcame the MSC-mediated cytarabine resistance of ALL cells both in vitro and in vivo.[99]
Human bone-marrow-derived MSCs from patients with ALL or AMLThe human leukaemia cell lines OCI-AML3, Reh, NALM6-luciferaseCopGFP, and RS4;11N/AVincristine, doxorubicine, cytarabidine The blockade of NF-κB activation via chemical agents or the overexpression of the mutant form of the inhibitor κB-α (IκBα) in BM-MSCs markedly reduced the stromal-mediated chemotherapeutic drug resistance in leukaemia cells in vitro and in vivo. An in vivo model of the human leukaemia BM microenvironment also illustrated a direct link between NF-κB activation and stromal-associated chemoprotection.[93]
AMLHuman bone-marrow-derived MSCs from patientsLeukaemic cell line HL-60 N/ACytarabinePrimary mesenchymal stromal cells in co-culture with leukaemic HL-60 cells are sensitized to cytarabine-induced genotoxicity, while leukaemic cells are protected. Malignant HSCs and MSCs bidirectionally modulate genotoxicity.[101]
Human bone-marrow-derived MSCs from healthy volunteersLeukaemic cell line HL-60 NOD/SCID miceCytarabineBM stromal cells induce ALDH activity in AML cells through increased expression of the ALDH2 isoform. BM-MSCs secrete TGF-β1, which exerts its effect through a noncanonical/p38-dependent signalling mechanism, leading to a stem-like phenotype in AML cells. Inhibition of downstream targets of this pathway, such as p38 MAPK, inhibits ALDH activity in AML cells. ALDH2 inhibition sensitizes AML cells to standard cytarabine chemotherapy in vitro. However, these findings were not validated in vivo.[106]
Human bone-marrow-derived MSCs from healthy volunteersHuman leukaemia cell line OCI-AML3; human primary cells from AML patients or healthy volunteersNSG miceCytarabineCox-2 expression and PGE2 generation are ARC/IL1β-dependent. The apoptosis repressor with caspase recruitment domain (ARC) protein, regulated by β-catenin, is an integral component of an IL1β/PGE2/β-catenin circuit. Cox-2/PGE2, regulated by ARC and induced by AML-MSC co-culture contributes to MSC-mediated chemoprotection in AML.[114]
Human bone-marrow-derived MSCsHuman leukaemia cell lines OCI-AML2, OCI-AML3, HL-60, ML-1, and Molm-13N/ABH3 mimetics, cytarabine, and daunorubicinBcl-2/Bcl-XL and Mcl-1 act redundantly as effectors of BMM-mediated AML drug resistance, highlighting the potential of Mcl-1 suppression to reverse BMM-mediated drug resistance in the leukaemic stem cell population, thus preventing disease relapse and ultimately improving patient survival. Suppression of Mcl-1 expression by the CDC7/CDK9 inhibitor PHA-767491 overcomes bone marrow stroma-mediated drug resistance in AML.[110]
Human bone-marrow-derived MSCs from ALL patientsMurine cell line MS-5; human bone-marrow-derived cells from patients with AMLN/ACytarabineHuman MSCs are potent feeder cells that are able to maintain AML cells in long-term culture. Co-culture of AML cells on MSCs results in a significantly higher proliferation capacity than on MS-5 or liquid culture. This favourable co-existence seems to be due, in part, to molecules important for communication within the niche. Blockade of TGF-β1 increases AML cell proliferation and chemosensitivity, while the CXCR4 antagonist plerixafor shows anti-proliferative effects and does not change cytarabine-induced cell death compared to control.[105]
Human MSCs from healthy volunteersHuman bone-marrow-derived and peripheral blood cells from AML patientsN/ACytarabineHuman MSCs contribute to quiescence and therapy resistance of persistent AML cells. Co-culture studies demonstrate that hMSCs protect leukaemic cells from the effect of AraC (cytarabine) treatment by enriching quiescent cells, mimicking the effects observed in patients. This effect is even detectable when no direct stromal contact is established.[102]
Human bone-marrow-derived MSCs from ALL patientsLeukaemia cell lines U937, HL60, and KG1aNOD/SCID mice MitoxantroneMSC protects AML cells from apoptosis through the c-Myc-dependent pathway. The expression of microRNA-494 (miR-494) in AML cells after
co-culture with MSCs is downregulated. In the co-culture system, activation of miR-494 in AML cells suppresses proliferation and induces apoptosis of AML cells in vitro. After the addition of mitoxantrone to the co-culture system, the proliferation of AML cells with miR-494 activation is suppressed more than that of control cells. After subcutaneous injection of AML cell lines in combination with MSC, tumour growth is suppressed in mice injected with miR-494-overexpressing AML cells. The rate of tumour formation is even lower after mitoxantrone treatment in the miR-494-overexpressing group. miR-494 suppresses drug resistance in AML cells by downregulating c-Myc through interaction with MSCs.
[109]
Human bone-marrow-derived MSCs from healthy volunteersLeukaemia cell lines OCI-AML3, KG-1 cells, and Molm13 NSG mice CytarabineARC enhances the migration and adhesion of leukaemia cells to MSCs both in vitro and in a novel human extramedullary bone/bone marrow mouse model. ARC induces IL1β expression in AML cells and increases CCL2, CCL4, and CXCL12 expression in MSCs, both through ARC-mediated activation of NFκB. Cells from AML patients express the receptors for and migrate toward CCL2, CCL4, and CXCL12. Inhibition of IL1β suppresses AML cell migration and sensitizes the cells co-cultured with MSCs to chemotherapy.[115]
Human bone-marrow-derived MSCs from AML patientsAML cell line HEL; ossification cell line hFOB1.19N/ADaunorubicinA strong positive correlation between the thrombopoietin (TPO) level and c-MPL expression is found in the bone marrow mononuclear cells of relapsed AML patients. A high level of TPO/c-MPL signalling may protect MSCs from daunorubicin chemotherapy in AML. The effects of inhibition of the TPO/c-MPL pathway on enhancing the chemotherapy sensitivity of AML cells and on their downstream effector molecules that direct the interactions between patient-derived blasts and leukaemia-repopulating cells need to be further studied.[107]
CLLHuman bone-marrow-derived MSCs from CLL patients and healthy volunteersHuman peripheral-blood-derived cells from patients with CLLN/AFludarabine, Cyclophosphamide, Bendamustine, Prednisone, and HydrocortisoneThe presence of BM-MSCs rescues chronic lymphocytic leukaemia (CLL) cells from apoptosis both spontaneously and following induction with various drugs, including fludarabine, cyclophosphamide, bendamustine, prednisone, and hydrocortisone. The treatment with a combination of anti-Notch-1, Notch-2, and Notch-4 antibodies or γ-secretase inhibitor XII
reverts this protective effect by day 3, even in the presence of the above-mentioned drugs.
[116]
Multiple myelomaHuman bone-marrow-derived MSCs from patients with multiple myeloma and healthy volunteersHuman multiple myeloma cell lines U266 and RPMI-8226 N/AMelphalan or doxorubicinBM-MSCs derived from patients with multiple myeloma (MM-MSCs), but not from healthy subjects (NM-MSCs), protect MM cells against the cytotoxicity of two chemotherapeutic agents (melphalan and doxorubicin). The pyrrolidine dithiocarbamate (PDTC), a potent and specific inhibitor of NF-κB, reverses the protective effects of MM-MSCs on MM cells in response to chemotherapy-induced apoptosis. Moreover, blocking autophagy by CQ or 3MA induces I-κBα phosphorylation and consequently prevents MM-MSC-mediated NF-κB signalling activation in MM cells. The results highlight the significance of MM-MSC-activated autophagy in MM chemotherapy resistance.[119]
GliomaHuman glioma-associated MSCs from patientsHuman cell line U87MG; human primary glioblastoma cellsBALB/c-nu miceTemozolomide Conditioned media of glioma-associated mesenchymal stromal/stem cells (gaMSCs) promotes the proliferation, migration, and chemotherapy resistance of glioma cells. The increased expression of FOXS1 and the activation of the EMT process in glioma cells under gaMSC-conditioned media was detected. The relationship of FOXS1, EMT, and temozolomide resistance in glioma cells was demonstrated through the regulation of FOXS1 expression in vitro and in vivo.[122]
GlioblastomaHuman umbilical cord perivascular cells Human glioblastoma cell lines SNB-19 and U251 Chicken chorioallantoic membraneTemozolomide Human umbilical cord perivascular cells (HUCPVCs, an MSC population present in the Wharton’s jelly of the umbilical cord) secrete molecules that contribute to glioblastoma aggressiveness by increasing cell proliferation, migration, and viability in vitro and by stimulating higher tumour growth in vivo. In contrast, the resistance of glioblastoma cells to temozolomide chemotherapy is not significantly affected by HUCPVC-conditioned medium.[124]
NeuroblastomaHuman bone-marrow-derived MSCs from patients with neuroblastomaHuman neuroblastoma cell lines CHLA-171 and CHLA-255N/AEtoposide, JAK inhibitor (AZD1480)Human bone marrow mesenchymal stromal cells induce tumour expression of sphingosine-1-phosphate receptor-1 (S1PR1), leading to their resistance to chemotherapy. Targeting S1PR1 by shRNA markedly enhances etoposide-induced apoptosis in neuroblastoma (NB) cells, while overexpression of S1PR1 significantly protects NB cells from multidrug-induced apoptosis via activating JAK-STAT3 signalling. Treatment with FTY720, an FDA-approved drug and antagonist of S1PR1, dramatically sensitizes drug-resistant NB cells to etoposide in in vitro and human neuroblastoma xenograft models.[125]
Oral squamous cell carcinomaHuman bone-marrow-derived MSCsHuman oral squamous cell carcinoma cell lines JHU-012, JHU-019, and OKF-TERT1N/ACisplatinThe crosstalk between human oral squamous carcinoma cells and MSCs is mediated, at least in part, by the activation of the autocrine PDGF-AA/PDGFR-α loop driving AKT-mediated signalling pathways, resulting in reduced cancer cell sensitivity to cisplatin through alterations in apoptosis.[129]
Head and neck cancerHuman bone-marrow-derived MSCsHuman head and neck cancer cell line SCC-25BALB/c nude micePaclitaxel MSC-exposed head and neck cancer cells develop paclitaxel resistance that can be maintained up to 30 d after the initial co-incubation period. The secretory profile of the MSCs suggested IL-6 to be a potential mediator of epigenetic imprinting on the head and neck cancer cells. When the MSC-imprinted cancer cells are exposed to the demethylation agent 5-aza-2′deoxycytidine, it restores the expression of the drug resistance genes to that of parental cells.[130]
Breast cancerHuman bone-marrow-derived MSCs from healthy volunteers; Murine bone-marrow- or blood-derived MSCsHuman breast cancer cell line MDA-MB-231, human colon carcinoma cell line C26, human lung adenocarcinoma cell line LLCBALB/c, C57Bl/6, athymic nude mice. Cisplatin, irinotecanMSCs become activated during treatment with platinum analogues and secrete factors that protect cancer cells against a range of chemotherapeutics. A metabolomics approach reveals two distinct platinum-induced polyunsaturated fatty acids (PIFAs), 12-oxo-5,8,10-heptadecatrienoic acid (KHT) and hexadeca-4,7,10,13-tetraenoic acid (16:4(n-3)), that in minute quantities, induce resistance to a broad spectrum of chemotherapeutic agents. Blocking central enzymes involved in the production of these PIFAs (cyclooxygenase-1 and thromboxane synthase) prevents MSC-induced resistance.[154]
Human bone-marrow-derived MSCsHuman breast cancer cell lines MDA-MB-231 and MCF-7 BALB/c female nude miceDoxorubicinDoxorubicin treatment induces the expression of miR-21-5p in MSCs and in mesenchymal stem-cell-derived exosomes, leading to the induction of S100A6 in the breast cancer cells (BCs). Silencing of miR-21-5p expression in MSCs and MSC exosomes abolished the resistance of BCs to doxorubicin, indicating an exosomal miR-21-5p regulated the role of S100A6 in chemoresistance both in vitro and in vivo.[133]
Human bone-marrow-derived MSCsHuman breast cancer cell line HCC1806NOD/SCID mice Mithramycin A, Doxycycline, 5-fluorouracilWhen GFP-labelled BMMSCs and RFP-labelled HCC1806 cells are injected together in vivo, they create tumours that contain a new hybrid cell that has characteristics of both BMMSCs and HCC1806 cells. When hybrid cells are injected into the mammary fat pad of NOD/SCID mice, they produce xenograft tumours that are smaller in size and exhibit resistance to chemotherapy drugs (i.e., doxorubicin and 5-fluorouracil) compared tumours from HCC1806 cells alone. This chemoresistance is shown to be associated with an increased expression of tetraspanins (CD9 and CD81) and drug resistance proteins (BCRP and MDR1). Subsequent siRNA-mediated knockdown of BMMSC-CD9 in DP-HCC1806:BMMSCs results in an attenuation of doxorubicin and 5-fluorouracil chemoresistance associated with decreased BCRP and serum cytokine expression (CCL5, CCR5, and CXCR12). It is suggested that within the tumour microenvironment CD9 is responsible for the crosstalk between BMMSCs and HCC1806 breast cancer cells (via CCL5, CCR5, and CXCR12), which contributes to chemoresistance.[131]
Bone-marrow-derived MSCsHuman breast cancer cell line MDA-MB-231N/AValproic acidIn vitro experiments confirm that VA inhibits NF-kB activation in cancer cells. In addition, analysing gene expression data in patients taking oral valproic acid showed that this drug decreased the expression of antioxidant enzymes, culminating in increased oxidative stress in tumour cells. Analysis of publicly available genome-wide drug-induced effects reveals that valproic acid, as a histone deacetylase inhibitor (HDACI), is the most effective drug in disturbing the signalling pathways activated by tumour–stromal interaction.[136]
Human adipose-derived MSCsHuman breast cancer cell line MCF-7/ADRN/AAdriamycin
hydrochloride
Conditioned medium derived from adipose mesenchymal stem cells induces increased expression of C-terminal Src kinase (Csk)-binding protein (Cbp), accompanied by enhanced cell proliferation and chemotherapy resistance in MCF-7/ADR breast cancer cells. Depletion of Cbp in breast cancer cells by RNA interference leads to remarkable inhibition of cell proliferation and invasion as well as synergy with adriamycin hydrochloride to suppress tumour growth.[134]
Human adipose-derived MSCsHuman breast cancer cell line MDA-MB-231N/ADoxorubicinConditioned medium collected from hAdSCs elicits doxorubicin resistance and enhances the expression of ABCG2, which is a transporter responsible for the efflux of doxorubicin. CXCL1 secreted by hAdSCs downregulates miR-106a expression in triple-negative breast cancer and thus upregulates the ABCG2 and doxorubicin resistance.[132]
Lung adenocarcinomaHuman bone-marrow-derived MSCs from patients with non-haematological malignant tumoursHuman lung adenocarcinoma cell lines H358, A549, and H460 and murine LLC cell lineC57BL/6 miceErlotinib Bone-marrow-derived MSCs residing in the hypoxic solid cancer microenvironment produce high levels of molecules associated with adipocytes, including adipokine leptin and IGFBPs. It is suggested that leptin induces the resistance of lung cancer cells to erlotinib through activating IGF-1R signalling. IGFBP2 induces erlotinib resistance by activating IGF-1R signalling in an IGF-1-independent manner. IGFBP2 had a synergistic effect with leptin to induce erlotinib resistance in vitro and in vivo.[137]
Gastric cancerHuman bone-marrow-derived MSCsHuman gastric cancer cell lines AGS and MKN45BALB/C nude miceFOLFOX regiment, composed of 5-FU, oxaliplatin, and calcium folinateMSCs promote stemness and chemoresistance in gastric cancer (GC) cells through fatty acid oxidation (FAO). TGF-β1 secreted by MSCs activates SMAD2/3 through TGF-β receptors, which then induce lncRNA MACC1-AS1 expression in GC cells and promote FAO-dependent stemness and chemoresistance through antagonizing miR145-5p. Pharmacologic inhibition of FAO with etomoxir (ETX) attenuates MSC-induced FOLFOX regiment resistance in vivo.[141]
Human gastric-cancer-associated MSCs from patientsThe human GC cell lines SGC-7901, MGC-803, HGC-27, and AGS BALB/c nude mice5-fluorouracilGastric cancer MSCs upregulate the levels of PD-L1 bound to the transcription factor CCCTC binding factor (CTCF), enhance the CSC-like
properties of GC cells, and lead to tumorigenesis. In vivo, PD-L1-positive GC cells have greater stemness potential and tumorigenicity than PD-L1-negative GC cells. GC cells are heterogeneous, and PD-L1s in GC cells have different reactivities to GCMSCs.
[138]
Human bone-marrow-derived MSCsHuman gastric cancer cell lines AGS and MKN45, human renal epithelial cell line HEK293TBALB/C nude mice FOLFOX regiment, composed of 5-FU, oxaliplatin, and calcium folinateMSC co-culture improves stemness and drug-resistance of gastric cancer (GC) cells. LncRNA histocompatibility leukocyte antigen complex P5 (HCP5) is induced in GC cells by MSC co-culture, contributing to stemness and drug resistance. MSC-induced lncRNA HCP5 drives FAO through
miR-3619-5p/AMPK/PGC1α/CEBPB axis to promote stemness and chemo-resistance of GC.
[140]
Human bone-marrow-derived MSCsHuman gastric cancer cell lines SGC7901, KATO-III, MKN45, and AGS N/ACisplatinBone marrow MSCs increase the antiapoptotic abilities and chemoresistance of CD133+ cells via upregulation of Bcl-2 and downregulation of BAX. BM-MSCs triggered activation of the PI3K/AKT signalling cascade in CD133+ cells. Blocking the PI3K/AKT pathway inhibited the promotion of chemoresistance. BM-MSCs enhance the drug resistance of CD133-overexpressing cells in vitro and in vivo but not that of CD133-knockdown cells, which demonstrates the contribution of CD133 to this process.[143]
Human umbilical-cord-derived MSCsHuman gastric cancer cell lines HGC-27, MGC-803, and SGC-7901 and human foetal lung fibroblast cell line HFL1BALB/c nu/nu mice5-fluorouracil, cisplatin MSC exosomes induce the resistance of gastric cancer cells to 5-fluorouracil both in vivo and ex vivo. MSC exosomes antagonise 5-fluorouracil-induced apoptosis and enhance the expression of multi-drug-resistance-associated proteins, including MDR, MRP, and LRP. MSC exosomes trigger the activation of calcium/calmodulin-dependent protein kinases (CaMKs) and Raf/MEK/ERK kinase cascade in gastric cancer cells. Blocking the CaM-Ks/Raf/MEK/ERK pathway inhibits the promoting role of MSC exosomes in chemoresistance.[142]
Pancreatic cancerHuman bone-marrow-derived MSCsHuman pancreatic adenocarcinoma cell lines PANC1, MIA PaCa-2, and BxPC3, human glioblastoma cell line U-87MG, human colon carcinoma cell line HT29, human non-small-cell lung carcinoma cell line A549, human breast carcinoma cell line MCF7SCID miceGemcitabine The inhibiting of crosstalk between MSCs and tumour-initiating cells (TIC) disrupts the CXCL10–CXCR3 axis and sensitizes tumour cells to chemotherapy, mainly by targeting TICs residing in the treated tumour. Based on the ability of MSCs to specifically home to tumours and target the TIC population, the use of MSC-derived nanovesicles as “Trojan horses” is presented as a strategy to overcome resistance, especially in desmoplastic cancers such as pancreatic adenocarcinomas.[144]
Genitourinary cancersHuman bone-marrow-derived MSCsHuman prostatic carcinoma cell lines PC3 and DU145Nude miceDocetaxel MSCs reduce the sensitivity of castration-resistant prostate cancer (CRPC) cells to docetaxel-induced proliferation inhibition and apoptosis promotion in vivo and in vitro. CRPC cells co-cultured with MSCs under docetaxel administration have an increased autophagy activation through TGF-β1 signalling. The autophagy inhibitor could effectively reverse MSC-induced resistance to docetaxel, e.g., inhibition of TGF-β1 secretion in MSCs increases the sensitivity of CRPC cells to docetaxel.[145]
Human adipose-derived MSCsHuman epithelial ovarian carcinoma cell lines ES2 and SKOV3N/ACisplatinPrimary omental adipose-derived mesenchymal stem cells (ADSCs) are a contributor to cisplatin resistance, exhibiting an ability to reduce
caspase-3-dependent apoptosis and intracellular platinum accumulation in epithelial ovarian carcinoma EOC.
[147]
Human amniotic-fluid-derived MSCs and adipose-derived MSCs from healthy volunteersHuman renal carcinoma cell line 786-0, human bladder carcinoma line T24N/ACiprofloxacinMSC-conditioned medium reduces bladder and renal cancer cell viability in vitro, induces cell cycle perturbations in bladder cancer T24 cells without significant influence on apoptosis rate in both studied cancer cell lines, and reduces cell sensitivity to ciprofloxacin after incubation in vitro with conditioned media.[146]
Human ovarian-cancer-associated MSCs from patients; human adipose-derived MSCs from healthy volunteers, human ovary-derived MSCs from healthy volunteersHuman ovarian cancer cell lines SKOV3, CAOV-3, COV318, Hey1, and PEO1 NOD/SCID mice Cisplatin Ovarian tumour-cell-secreted Hedgehog (HH) induces CA-MSC BMP4 expression. CA-MSC-derived BMP4 reciprocally increases ovarian tumour cell HH expression, indicating a positive feedback loop. Interruption of this loop with an HH pathway inhibitor or BMP4-blocking antibody decreases CA-MSC-derived BMP4 and tumour-derived HH, preventing enrichment of cancer stem-like cells (CSCs) and reversing chemotherapy resistance. The impact of HH inhibition is only seen in CA-MSC-containing tumours, indicating the importance of a humanized stroma. The results are reciprocal to findings in pancreatic and bladder cancer, suggesting HH signalling effects are tumour-tissue-specific, warranting careful investigation in each tumour type.[148]
Liver cancerHuman MSCsHuman hepatocarcinoma cell lines SMMC-7721 and HepG2 N/A3-methyladenineTumour inflammatory microenvironment is a key player in activating MSCs to induce chemoresistance of hepatocarcinoma cells. Inflammation is a fundamental feature during the development of hepatocellular carcinoma, which exists not only within the tumour tissue but also in the tissues surrounding the tumour. MSCs in the inflammatory microenvironment may persistently promote the development of chemoresistance in HCC cells during tumour growth. One mechanism underlying the MSC-promoted development of chemoresistance in HCC cells is their overexpression of TGF-β in response to inflammatory stimuli in the tumour microenvironment. Treatment of HCC cells with autophagy inhibitor effectively reverses the MSC-induced resistance to chemotherapy, and knockdown of TGF-β expression by MSCs with siRNA attenuates MSC-induced chemoresistance in HCC cells.[151]
Bone cancerHuman bone-marrow-derived MSCsHuman osteosarcoma cell lines Saos-2 and U2-OS; primary cells from patients with osteosarcomaBALB/c nude miceDoxorubicin MSCs promote osteosarcoma cell survival and drug resistance through activation of STAT3. Inhibition of STAT3 prolongs the survival time of tumour-bearing mice by suppressing tumour growth and increasing the sensitivity of tumour cells to doxorubicin. The increased expression of p-STAT3, multidrug resistance protein (MRP), and P-glycoprotein (MDR-1) is associated with high chemotherapy resistance in clinical osteosarcoma samples.[152]

4. For a Successful Treatment, the Whole Tumour Microenvironment Must Be Considered: Crucial Findings

Based on the results of the studies presented in Chapter 3, we have identified crucial findings about the effect of MSCs on cancer cells, which are presented in the following subchapters.

4.1. MSCs Affect Various Signalling Pathways in Cancer Cells to Protect Them from Chemotherapy-Induced Damage

Studies have shown that in the presence of chemotherapeutic agents MSCs in the tumour microenvironment play a protective role that prevents or at least decreases chemotherapy-induced damage in cancer cells. MSCs affect a large number of signalling pathways in cancer cells, most commonly those that protect cancer cells from apoptosis (e.g., increased expression of Bcl-2, Mcl-1, PGE2, ARC, and drug resistance proteins [110,114,115]) and promote their proliferation (e.g., altered expression of the Wnt, c-Myc, PI3K/AKT, and JAK2/STAT3 signalling pathways [99,106,108,125,155]). Moreover, these studies demonstrate the multifactorial ways in which the MSCs protect cancer cells and underline the reason for the difficulty in successfully preventing chemotherapy resistance and disease recurrence.

4.2. MSCs Affect the Susceptibility of Cancer Cells to Chemotherapy When within the Tumour Microenvironment and Distant from the Tumour (Systemic Use)

While most of the studies included in this review article focused on the effects of MSCs or their conditioned medium in the tumour microenvironment, another study demonstrated that MSCs are not only capable of initiating a response within the tumour microenvironment but may also induce drug resistance when placed distant from the tumour. This was demonstrated by Roodhart et al. (2011), who first developed two mice tumour models using C26 colon carcinoma cells in BALB/c mice and Lewis lung carcinoma (LLC) cells in C57BL/6 mice. Next, they applied an intravenous injection of bone marrow MSCs at a distant site from the tumour at the time of cisplatin treatment, which led to complete resistance to the treatment in mice, which was mediated by the release of two platinum-induced fatty acids [154]. These were able to induce resistance against a broad spectrum of chemotherapeutic agents, especially platinum-containing chemotherapeutics. Interestingly, even the platinum-induced fatty acids alone (in the absence of MSCs) were able to induce resistance to chemotherapy, but the addition of COX-1 and thromboxane synthase to the MSC culture blocked the formation of chemotherapy resistance, as the production of the platinum-induced fatty acids depends on these enzymes [79].

4.3. How to Improve Treatment Outcomes

Successful cancer treatment without eventual disease relapse is extremely difficult to achieve due to the high complexity of the tumour microenvironment and the immense adaptability of cancer cells. However, several studies [30,96,99,106,116,119] demonstrated encouraging results when using inhibitors of various signalling pathways that are affected in cancer cells due to the presence and actions of MSCs in the tumour microenvironment. These results offer a good starting point for the investigation of novel treatment approaches and a much-needed shift from a cancer-cell-oriented treatment toward including the targeting of the stroma in the tumour microenvironment.
In the last years, many studies investigated the use of MSCs as a drug delivery tool for cancer treatment due to their ability to home to the tumour microenvironment. While there are still considerable challenges limiting effective translation, such as a limited tumour tropism and broad biodistribution when using conventional (non-altered) MSCs, the field is quickly developing [156,157,158]. However, one must consider the effect of the newly arrived MSCs on the tumour microenvironment after the delivery of chemotherapeutic agents. It is not clear whether the MSCs integrate into the tumour microenvironment and what effect they have on it, which raises crucial safety concerns. Hence, the studies included in this review article expose the hazards of bringing additional MSCs into the tumour microenvironment and underline the importance of thoroughly investigating the long-term effects of MSCs used as delivery tools.

5. Conclusions

The role of MSCs in the tumour microenvironment and the development of chemotherapy resistance has been quite well elucidated in the case of ALL, AML, and breast cancer. However, for most cancers, only few or even no studies investigating the role of MSCs in the development of resistance to chemotherapy have been published. Therefore, to ensure better treatment outcomes, future research should focus on the effect of non-cancerous cells, especially MSCs, in the tumour microenvironment on chemotherapeutic agents in all cancers and plan treatments in compliance with these results.
Furthermore, discrepancies may arise when comparing results from the in vitro and in vivo settings, as MSCs can reduce cancer cell proliferation in the in vitro setting while promoting tumour growth in mice [38]. These results highlight the impact of the complex tumour microenvironment on cancer cells, which should be especially considered when using in vitro models that must contain multiple cell types in biologically relevant ratios. Furthermore, to obtain clear and solid data on the effects of non-cancerous cells in the tumour microenvironment, studies should include a combination of investigations in the in vitro and in vivo settings.

Author Contributions

Conceptualization, T.Ž.R. and M.E.K.; methodology, T.Ž.R. and M.E.K.; writing—original draft preparation, T.Ž.R.; writing—review and editing, T.Ž.R. and M.E.K.; funding acquisition, M.E.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Slovenian Research Agency (J7-2594, J3-2526, P3-0108).

Acknowledgments

This work was supported by: COST Action CA17116 International Network for Translating Research on Perinatal Derivatives into Therapeutic Approaches (SPRINT), supported by COST (European Cooperation in Science and Technology).

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ALDHAldehyde dehydrogenase
ALLAcute lymphocytic leukaemia
AMLAcute myeloid leukaemia
ARCApoptosis repressor with caspase recruitment domain
BDNFBrain-derived neurotrophic factor
CXCLChemokine (C-X-C motif) ligand
CXCRChemokine (C-X-C motif) receptor
FGF-2Fibroblast growth factor 2
HGFHepatocyte growth factor
ICAM-1Intercellular adhesion molecule 1
IDOIndoleamine 2,3-dioxygenase
IFNγInterferon γ
IGF-1Insulin-like growth factor 1
IGFBP-2Insulin-like growth factor-binding protein-2
IL-8Interleukin-8
IGF-1RInsulin-like growth factor 1 receptor
LIFLeukaemia inhibitory factor
MAPKMitogen-activated protein kinase
Mcl-1Myeloid cell leukaemia -1
MCP-1Monocyte chemotactic protein-1
M-CSFMacrophage colony-stimulating factor
MIP-2Macrophage inflammatory protein 2
MSCsMesenchymal stem/stromal cells
PGDFPlatelet-derived growth factor
PGE2Prostaglandin E2
PI3KPhosphoinositide 3-kinase
S1PR1Sphingosine-1-phosphate receptor 1
SDF-1Stromal cell-derived factor 1
STAT3Signal transducer and activator of transcription 3
TGF-βTransforming growth factor β
TNFαTumour necrosis factor α
VCAM-1Vascular cell adhesion protein 1
VEGFVascular endothelial growth factor
VLA-4Very late antigen-4

References

  1. Ridge, S.M.; Sullivan, F.J.; Glynn, S.A. Mesenchymal stem cells: Key players in cancer progression. Mol. Cancer 2017, 16, 31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Pittenger, M.F.; Mackay, A.M.; Beck, S.C.; Jaiswal, R.K.; Douglas, R.; Mosca, J.D.; Moorman, M.A.; Simonetti, D.W.; Craig, S.; Marshak, D.R. Multilineage potential of adult human mesenchymal stem cells. Science 1999, 284, 143–147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Fan, M.; Zhang, J.; Xin, H.; He, X.; Zhang, X. Current Perspectives on Role of MSC in Renal Pathophysiology. Front. Physiol. 2018, 9, 1323. [Google Scholar] [CrossRef] [Green Version]
  4. Dominici, M.; Le Blanc, K.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.; Krause, D.; Deans, R.; Keating, A.; Prockop, D.; Horwitz, E. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006, 8, 315–317. [Google Scholar] [CrossRef]
  5. Heidari, B.; Shirazi, A.; Akhondi, M.M.; Hassanpour, H.; Behzadi, B.; Naderi, M.M.; Sarvari, A.; Borjian, S. Comparison of proliferative and multilineage differentiation potential of sheep mesenchymal stem cells derived from bone marrow, liver, and adipose tissue. Avicenna J. Med. Biotechnol. 2013, 5, 104–117. [Google Scholar] [PubMed]
  6. Gronthos, S.; Mankani, M.; Brahim, J.; Robey, P.G.; Shi, S. Postnatal human dental pulp stem cells (DPSCs) in vitro and in vivo. Proc. Natl. Acad. Sci. USA 2000, 97, 13625–13630. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Erices, A.; Conget, P.; Minguell, J.J. Mesenchymal progenitor cells in human umbilical cord blood. Br. J. Haematol. 2000, 109, 235–242. [Google Scholar] [CrossRef] [PubMed]
  8. Fukuchi, Y.; Nakajima, H.; Sugiyama, D.; Hirose, I.; Kitamura, T.; Tsuji, K. Human placenta-derived cells have mesenchymal stem/progenitor cell potential. Stem Cells 2004, 22, 649–658. [Google Scholar] [CrossRef] [Green Version]
  9. Schwab, K.E.; Gargett, C.E. Co-expression of two perivascular cell markers isolates mesenchymal stem-like cells from human endometrium. Hum. Reprod. 2007, 22, 2903–2911. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. In ’t Anker, P.S.; Scherjon, S.A.; Kleijburg-van der Keur, C.; Noort, W.A.; Claas, F.H.; Willemze, R.; Fibbe, W.E.; Kanhai, H.H. Amniotic fluid as a novel source of mesenchymal stem cells for therapeutic transplantation. Blood 2003, 102, 1548–1549. [Google Scholar] [CrossRef] [PubMed]
  11. Young, H.E.; Steele, T.A.; Bray, R.A.; Hudson, J.; Floyd, J.A.; Hawkins, K.; Thomas, K.; Austin, T.; Edwards, C.; Cuzzourt, J.; et al. Human reserve pluripotent mesenchymal stem cells are present in the connective tissues of skeletal muscle and dermis derived from fetal, adult, and geriatric donors. Anat. Rec. 2001, 264, 51–62. [Google Scholar] [CrossRef] [PubMed]
  12. Zhuang, W.Z.; Lin, Y.H.; Su, L.J.; Wu, M.S.; Jeng, H.Y.; Chang, H.C.; Huang, Y.H.; Ling, T.Y. Mesenchymal stem/stromal cell-based therapy: Mechanism, systemic safety and biodistribution for precision clinical applications. J. Biomed. Sci. 2021, 28, 28. [Google Scholar] [CrossRef] [PubMed]
  13. Sun, Z.; Wang, S.; Zhao, R.C. The roles of mesenchymal stem cells in tumor inflammatory microenvironment. J. Hematol. Oncol. 2014, 7, 14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Hill, B.S.; Pelagalli, A.; Passaro, N.; Zannetti, A. Tumor-educated mesenchymal stem cells promote pro-metastatic phenotype. Oncotarget 2017, 8, 73296–73311. [Google Scholar] [CrossRef] [Green Version]
  15. Kouris, N.A.; Schaefer, J.A.; Hatta, M.; Freeman, B.T.; Kamp, T.J.; Kawaoka, Y.; Ogle, B.M. Directed Fusion of Mesenchymal Stem Cells with Cardiomyocytes via VSV-G Facilitates Stem Cell Programming. Stem Cells Int. 2012, 2012, 414038. [Google Scholar] [CrossRef] [Green Version]
  16. Caplan, A.I.; Bruder, S.P. Mesenchymal stem cells: Building blocks for molecular medicine in the 21st century. Trends Mol. Med. 2001, 7, 259–264. [Google Scholar] [CrossRef]
  17. Thompson, M.; Mei, S.H.J.; Wolfe, D.; Champagne, J.; Fergusson, D.; Stewart, D.J.; Sullivan, K.J.; Doxtator, E.; Lalu, M.; English, S.W.; et al. Cell therapy with intravascular administration of mesenchymal stromal cells continues to appear safe: An updated systematic review and meta-analysis. EClinicalMedicine 2020, 19, 100249. [Google Scholar] [CrossRef] [Green Version]
  18. Coffelt, S.B.; Marini, F.C.; Watson, K.; Zwezdaryk, K.J.; Dembinski, J.L.; LaMarca, H.L.; Tomchuck, S.L.; Honer zu Bentrup, K.; Danka, E.S.; Henkle, S.L.; et al. The pro-inflammatory peptide LL-37 promotes ovarian tumor progression through recruitment of multipotent mesenchymal stromal cells. Proc. Natl. Acad. Sci. USA 2009, 106, 3806–3811. [Google Scholar] [CrossRef] [Green Version]
  19. Christodoulou, I.; Goulielmaki, M.; Devetzi, M.; Panagiotidis, M.; Koliakos, G.; Zoumpourlis, V. Mesenchymal stem cells in preclinical cancer cytotherapy: A systematic review. Stem Cell Res. Ther. 2018, 9, 336. [Google Scholar] [CrossRef]
  20. Balkwill, F.R.; Capasso, M.; Hagemann, T. The tumor microenvironment at a glance. J. Cell Sci. 2012, 125, 5591–5596. [Google Scholar] [CrossRef] [Green Version]
  21. Atiya, H.; Frisbie, L.; Pressimone, C.; Coffman, L. Mesenchymal Stem Cells in the Tumor Microenvironment. Adv. Exp. Med. Biol. 2020, 1234, 31–42. [Google Scholar] [CrossRef] [PubMed]
  22. Li, W.; Yang, J.; Zheng, P.; Li, H.; Zhao, S. The Origins and Generation of Cancer-Associated Mesenchymal Stromal Cells: An Innovative Therapeutic Target for Solid Tumors. Front. Oncol. 2021, 11, 723707. [Google Scholar] [CrossRef] [PubMed]
  23. Ahn, S.Y. The Role of MSCs in the Tumor Microenvironment and Tumor Progression. Anticancer. Res. 2020, 40, 3039–3047. [Google Scholar] [CrossRef] [PubMed]
  24. Suzuki, K.; Sun, R.; Origuchi, M.; Kanehira, M.; Takahata, T.; Itoh, J.; Umezawa, A.; Kijima, H.; Fukuda, S.; Saijo, Y. Mesenchymal stromal cells promote tumor growth through the enhancement of neovascularization. Mol. Med. 2011, 17, 579–587. [Google Scholar] [CrossRef]
  25. Liang, W.; Chen, X.; Zhang, S.; Fang, J.; Chen, M.; Xu, Y. Mesenchymal stem cells as a double-edged sword in tumor growth: Focusing on MSC-derived cytokines. Cell Mol. Biol. Lett. 2021, 26, 3. [Google Scholar] [CrossRef] [PubMed]
  26. Galland, S.; Stamenkovic, I. Mesenchymal stromal cells in cancer: A review of their immunomodulatory functions and dual effects on tumor progression. J. Pathol. 2020, 250, 555–572. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Papait, A.; Stefani, F.R.; Cargnoni, A.; Magatti, M.; Parolini, O.; Silini, A.R. The Multifaceted Roles of MSCs in the Tumor Microenvironment: Interactions With Immune Cells and Exploitation for Therapy. Front. Cell Dev. Biol. 2020, 8, 447. [Google Scholar] [CrossRef]
  28. Fan, H.; Atiya, H.I.; Wang, Y.; Pisanic, T.R.; Wang, T.H.; Shih, I.M.; Foy, K.K.; Frisbie, L.; Buckanovich, R.J.; Chomiak, A.A.; et al. Epigenomic Reprogramming toward Mesenchymal-Epithelial Transition in Ovarian-Cancer-Associated Mesenchymal Stem Cells Drives Metastasis. Cell Rep. 2020, 33, 108473. [Google Scholar] [CrossRef]
  29. Li, C.; Zhao, H.; Wang, B. Mesenchymal stem/stromal cells: Developmental origin, tumorigenesis and translational cancer therapeutics. Transl. Oncol. 2021, 14, 100948. [Google Scholar] [CrossRef]
  30. Coffman, L.G.; Pearson, A.T.; Frisbie, L.G.; Freeman, Z.; Christie, E.; Bowtell, D.D.; Buckanovich, R.J. Ovarian Carcinoma-Associated Mesenchymal Stem Cells Arise from Tissue-Specific Normal Stroma. Stem Cells 2019, 37, 257–269. [Google Scholar] [CrossRef] [Green Version]
  31. McLean, K.; Gong, Y.; Choi, Y.; Deng, N.; Yang, K.; Bai, S.; Cabrera, L.; Keller, E.; McCauley, L.; Cho, K.R.; et al. Human ovarian carcinoma–associated mesenchymal stem cells regulate cancer stem cells and tumorigenesis via altered BMP production. J. Clin. Investig. 2011, 121, 3206–3219. [Google Scholar] [CrossRef] [Green Version]
  32. Mishra, P.J.; Humeniuk, R.; Medina, D.J.; Alexe, G.; Mesirov, J.P.; Ganesan, S.; Glod, J.W.; Banerjee, D. Carcinoma-associated fibroblast-like differentiation of human mesenchymal stem cells. Cancer Res. 2008, 68, 4331–4339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Spaeth, E.L.; Dembinski, J.L.; Sasser, A.K.; Watson, K.; Klopp, A.; Hall, B.; Andreeff, M.; Marini, F. Mesenchymal stem cell transition to tumor-associated fibroblasts contributes to fibrovascular network expansion and tumor progression. PLoS ONE 2009, 4, e4992. [Google Scholar] [CrossRef] [Green Version]
  34. Tsai, K.S.; Yang, S.H.; Lei, Y.P.; Tsai, C.C.; Chen, H.W.; Hsu, C.Y.; Chen, L.L.; Wang, H.W.; Miller, S.A.; Chiou, S.H.; et al. Mesenchymal stem cells promote formation of colorectal tumors in mice. Gastroenterology 2011, 141, 1046–1056. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Luo, J.; Ok Lee, S.; Liang, L.; Huang, C.K.; Li, L.; Wen, S.; Chang, C. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Hossain, A.; Gumin, J.; Gao, F.; Figueroa, J.; Shinojima, N.; Takezaki, T.; Priebe, W.; Villarreal, D.; Kang, S.G.; Joyce, C.; 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Guan, J.; Chen, J. Mesenchymal stem cells in the tumor microenvironment. Biomed. Rep. 2013, 1, 517–521. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Ramasamy, R.; Lam, E.W.; Soeiro, I.; Tisato, V.; Bonnet, D.; Dazzi, F. Mesenchymal stem cells inhibit proliferation and apoptosis of tumor cells: Impact on in vivo tumor growth. Leukemia 2007, 21, 304–310. [Google Scholar] [CrossRef] [PubMed]
  39. Huang, W.H.; Chang, M.C.; Tsai, K.S.; Hung, M.C.; Chen, H.L.; Hung, S.C. Mesenchymal stem cells promote growth and angiogenesis of tumors in mice. Oncogene 2013, 32, 4343–4354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Lin, G.; Yang, R.; Banie, L.; Wang, G.; Ning, H.; Li, L.C.; Lue, T.F.; Lin, C.S. Effects of transplantation of adipose tissue-derived stem cells on prostate tumor. Prostate 2010, 70, 1066–1073. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Lee, J.K.; Park, S.R.; Jung, B.K.; Jeon, Y.K.; Lee, Y.S.; Kim, M.K.; Kim, Y.G.; Jang, J.Y.; Kim, C.W. Exosomes derived from mesenchymal stem cells suppress angiogenesis by down-regulating VEGF expression in breast cancer cells. PLoS ONE 2013, 8, e84256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Waghray, M.; Yalamanchili, M.; Dziubinski, M.; Zeinali, M.; Erkkinen, M.; Yang, H.; Schradle, K.A.; Urs, S.; Pasca Di Magliano, M.; Welling, T.H.; et al. GM-CSF Mediates Mesenchymal-Epithelial Cross-talk in Pancreatic Cancer. Cancer Discov. 2016, 6, 886–899. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Berniakovich, I.; Giorgio, M. Low oxygen tension maintains multipotency, whereas normoxia increases differentiation of mouse bone marrow stromal cells. Int. J. Mol. Sci. 2013, 14, 2119–2134. [Google Scholar] [CrossRef] [PubMed]
  44. Efimenko, A.; Starostina, E.; Kalinina, N.; Stolzing, A. Angiogenic properties of aged adipose derived mesenchymal stem cells after hypoxic conditioning. J. Transl. Med. 2011, 9, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Hung, S.C.; Pochampally, R.R.; Chen, S.C.; Hsu, S.C.; Prockop, D.J. Angiogenic effects of human multipotent stromal cell conditioned medium activate the PI3K-Akt pathway in hypoxic endothelial cells to inhibit apoptosis, increase survival, and stimulate angiogenesis. Stem. Cells 2007, 25, 2363–2370. [Google Scholar] [CrossRef] [PubMed]
  46. Crisostomo, P.R.; Wang, Y.; Markel, T.A.; Wang, M.; Lahm, T.; Meldrum, D.R. Human mesenchymal stem cells stimulated by TNF-alpha, LPS, or hypoxia produce growth factors by an NF kappa B- but not JNK-dependent mechanism. Am. J. Physiol. Cell Physiol. 2008, 294, C675–C682. [Google Scholar] [CrossRef]
  47. Rodini, C.O.; Gonçalves da Silva, P.B.; Assoni, A.F.; Carvalho, V.M.; Okamoto, O.K. Mesenchymal stem cells enhance tumorigenic properties of human glioblastoma through independent cell-cell communication mechanisms. Oncotarget 2018, 9, 24766–24777. [Google Scholar] [CrossRef] [Green Version]
  48. Zhu, W.; Huang, L.; Li, Y.; Zhang, X.; Gu, J.; Yan, Y.; Xu, X.; Wang, M.; Qian, H.; Xu, W. Exosomes derived from human bone marrow mesenchymal stem cells promote tumor growth in vivo. Cancer Lett. 2012, 315, 28–37. [Google Scholar] [CrossRef]
  49. Li, G.C.; Zhang, H.W.; Zhao, Q.C.; Sun, L.I.; Yang, J.J.; Hong, L.; Feng, F.; Cai, L. Mesenchymal stem cells promote tumor angiogenesis via the action of transforming growth factor β1. Oncol. Lett. 2016, 11, 1089–1094. [Google Scholar] [CrossRef] [Green Version]
  50. Feng, B.; Chen, L. Review of mesenchymal stem cells and tumors: Executioner or coconspirator? Cancer Biother. Radiopharm. 2009, 24, 717–721. [Google Scholar] [CrossRef] [PubMed]
  51. Du, W.J.; Chi, Y.; Yang, Z.X.; Li, Z.J.; Cui, J.J.; Song, B.Q.; Li, X.; Yang, S.G.; Han, Z.B.; Han, Z.C. Heterogeneity of proangiogenic features in mesenchymal stem cells derived from bone marrow, adipose tissue, umbilical cord, and placenta. Stem Cell Res. Ther. 2016, 7, 163. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Zhu, Y.; Sun, Z.; Han, Q.; Liao, L.; Wang, J.; Bian, C.; Li, J.; Yan, X.; Liu, Y.; Shao, C.; et al. Human mesenchymal stem cells inhibit cancer cell proliferation by secreting DKK-1. Leukemia 2009, 23, 925–933. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Huang, M.; Zhang, D.; Wu, J.Y.; Xing, K.; Yeo, E.; Li, C.; Zhang, L.; Holland, E.; Yao, L.; Qin, L.; et al. Wnt-mediated endothelial transformation into mesenchymal stem cell-like cells induces chemoresistance in glioblastoma. Sci. Transl. Med. 2020, 12, aay7522. [Google Scholar] [CrossRef] [PubMed]
  54. Karnoub, A.E.; Dash, A.B.; Vo, A.P.; Sullivan, A.; Brooks, M.W.; Bell, G.W.; Richardson, A.L.; Polyak, K.; Tubo, R.; Weinberg, R.A. Mesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature 2007, 449, 557–563. [Google Scholar] [CrossRef] [PubMed]
  55. Rivera-Cruz, C.M.; Shearer, J.J.; Figueiredo Neto, M.; Figueiredo, M.L. The Immunomodulatory Effects of Mesenchymal Stem Cell Polarization within the Tumor Microenvironment Niche. Stem Cells Int. 2017, 2017, 4015039. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Poggi, A.; Varesano, S.; Zocchi, M.R. How to Hit Mesenchymal Stromal Cells and Make the Tumor Microenvironment Immunostimulant Rather Than Immunosuppressive. Front. Immunol. 2018, 9, 262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Torisu, H.; Ono, M.; Kiryu, H.; Furue, M.; Ohmoto, Y.; Nakayama, J.; Nishioka, Y.; Sone, S.; Kuwano, M. Macrophage infiltration correlates with tumor stage and angiogenesis in human malignant melanoma: Possible involvement of TNFalpha and IL-1alpha. Int. J. Cancer 2000, 85, 182–188. [Google Scholar] [CrossRef]
  58. Cascio, S.; Chandler, C.; Zhang, L.; Sinno, S.; Gao, B.; Onkar, S.; Bruno, T.C.; Vignali, D.A.A.; Mahdi, H.; Osmanbeyoglu, H.U.; et al. Cancer-associated MSC drive tumor immune exclusion and resistance to immunotherapy, which can be overcome by Hedgehog inhibition. Sci. Adv. 2021, 7, eabi5790. [Google Scholar] [CrossRef] [PubMed]
  59. Otsu, K.; Das, S.; Houser, S.D.; Quadri, S.K.; Bhattacharya, S.; Bhattacharya, J. Concentration-dependent inhibition of angiogenesis by mesenchymal stem cells. Blood 2009, 113, 4197–4205. [Google Scholar] [CrossRef] [Green Version]
  60. Qiao, L.; Xu, Z.; Zhao, T.; Zhao, Z.; Shi, M.; Zhao, R.C.; Ye, L.; Zhang, X. Suppression of tumorigenesis by human mesenchymal stem cells in a hepatoma model. Cell Res. 2008, 18, 500–507. [Google Scholar] [CrossRef] [Green Version]
  61. Khakoo, A.Y.; Pati, S.; Anderson, S.A.; Reid, W.; Elshal, M.F.; Rovira, I.I.; Nguyen, A.T.; Malide, D.; Combs, C.A.; Hall, G.; et al. Human mesenchymal stem cells exert potent antitumorigenic effects in a model of Kaposi’s sarcoma. J. Exp. Med. 2006, 203, 1235–1247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Sun, B.; Roh, K.H.; Park, J.R.; Lee, S.R.; Park, S.B.; Jung, J.W.; Kang, S.K.; Lee, Y.S.; Kang, K.S. Therapeutic potential of mesenchymal stromal cells in a mouse breast cancer metastasis model. Cytotherapy 2009, 11, 289–298. [Google Scholar] [CrossRef] [PubMed]
  63. Lu, Y.R.; Yuan, Y.; Wang, X.J.; Wei, L.L.; Chen, Y.N.; Cong, C.; Li, S.F.; Long, D.; Tan, W.D.; Mao, Y.Q.; et al. The growth inhibitory effect of mesenchymal stem cells on tumor cells in vitro and in vivo. Cancer Biol. Ther. 2008, 7, 245–251. [Google Scholar] [CrossRef] [Green Version]
  64. Khalil, C.; Moussa, M.; Azar, A.; Tawk, J.; Habbouche, J.; Salameh, R.; Ibrahim, A.; Alaaeddine, N. Anti-proliferative effects of mesenchymal stem cells (MSCs) derived from multiple sources on ovarian cancer cell lines: An in-vitro experimental study. J. Ovarian. Res. 2019, 12, 70. [Google Scholar] [CrossRef]
  65. Wu, X.; Tang, Y.; Yue, X.; Chen, P.; Hu, Z.; Shen, Y. Inhibition of cervical cancer cells by co-culturing with mesenchymal stem cells. Int. J. Clin. Exp. Pathol. 2018, 11, 2506–2513. [Google Scholar] [PubMed]
  66. He, N.; Kong, Y.; Lei, X.; Liu, Y.; Wang, J.; Xu, C.; Wang, Y.; Du, L.; Ji, K.; Wang, Q.; et al. MSCs inhibit tumor progression and enhance radiosensitivity of breast cancer cells by down-regulating Stat3 signaling pathway. Cell Death Dis. 2018, 9, 1026. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Rhee, K.J.; Lee, J.I.; Eom, Y.W. Mesenchymal Stem Cell-Mediated Effects of Tumor Support or Suppression. Int. J. Mol. Sci. 2015, 16, 30015–30033. [Google Scholar] [CrossRef] [Green Version]
  68. Qiao, L.; Xu, Z.L.; Zhao, T.J.; Ye, L.H.; Zhang, X.D. Dkk-1 secreted by mesenchymal stem cells inhibits growth of breast cancer cells via depression of Wnt signalling. Cancer Lett. 2008, 269, 67–77. [Google Scholar] [CrossRef] [PubMed]
  69. Cousin, B.; Ravet, E.; Poglio, S.; De Toni, F.; Bertuzzi, M.; Lulka, H.; Touil, I.; André, M.; Grolleau, J.L.; Péron, J.M.; et al. Adult stromal cells derived from human adipose tissue provoke pancreatic cancer cell death both in vitro and in vivo. PLoS ONE 2009, 4, e6278. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Dasari, V.R.; Velpula, K.K.; Kaur, K.; Fassett, D.; Klopfenstein, J.D.; Dinh, D.H.; Gujrati, M.; Rao, J.S. Cord blood stem cell-mediated induction of apoptosis in glioma downregulates X-linked inhibitor of apoptosis protein (XIAP). PLoS ONE 2010, 5, e11813. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Alcayaga-Miranda, F.; González, P.L.; Lopez-Verrilli, A.; Varas-Godoy, M.; Aguila-Díaz, C.; Contreras, L.; Khoury, M. Prostate tumor-induced angiogenesis is blocked by exosomes derived from menstrual stem cells through the inhibition of reactive oxygen species. Oncotarget 2016, 7, 44462–44477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Rosenberger, L.; Ezquer, M.; Lillo-Vera, F.; Pedraza, P.L.; Ortúzar, M.I.; González, P.L.; Figueroa-Valdés, A.I.; Cuenca, J.; Ezquer, F.; Khoury, M.; et al. Stem cell exosomes inhibit angiogenesis and tumor growth of oral squamous cell carcinoma. Sci. Rep. 2019, 9, 663. [Google Scholar] [CrossRef]
  73. Hass, R. Role of MSC in the Tumor Microenvironment. Cancers 2020, 12, 2107. [Google Scholar] [CrossRef] [PubMed]
  74. Hmadcha, A.; Martin-Montalvo, A.; Gauthier, B.R.; Soria, B.; Capilla-Gonzalez, V. Therapeutic Potential of Mesenchymal Stem Cells for Cancer Therapy. Front. Bioeng. Biotechnol. 2020, 8, 43. [Google Scholar] [CrossRef] [PubMed]
  75. Buenrostro, D.; Mulcrone, P.L.; Owens, P.; Sterling, J.A. The Bone Microenvironment: A Fertile Soil for Tumor Growth. Curr. Osteoporos. Rep. 2016, 14, 151–158. [Google Scholar] [CrossRef] [PubMed]
  76. Peddareddigari, V.G.; Wang, D.; Dubois, R.N. The tumor microenvironment in colorectal carcinogenesis. Cancer Microenviron. 2010, 3, 149–166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Li, J.J.; Tsang, J.Y.; Tse, G.M. Tumor Microenvironment in Breast Cancer-Updates on Therapeutic Implications and Pathologic Assessment. Cancers 2021, 13, 4233. [Google Scholar] [CrossRef]
  78. Selich, A.; Daudert, J.; Hass, R.; Philipp, F.; von Kaisenberg, C.; Paul, G.; Cornils, K.; Fehse, B.; Rittinghausen, S.; Schambach, A.; et al. Massive Clonal Selection and Transiently Contributing Clones During Expansion of Mesenchymal Stem Cell Cultures Revealed by Lentiviral RGB-Barcode Technology. Stem Cells Transl. Med. 2016, 5, 591–601. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Houthuijzen, J.M.; Daenen, L.G.; Roodhart, J.M.; Voest, E.E. The role of mesenchymal stem cells in anti-cancer drug resistance and tumour progression. Br. J. Cancer 2012, 106, 1901–1906. [Google Scholar] [CrossRef] [PubMed]
  80. Meads, M.B.; Hazlehurst, L.A.; Dalton, W.S. The bone marrow microenvironment as a tumor sanctuary and contributor to drug resistance. Clin. Cancer Res. 2008, 14, 2519–2526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Huang, J.; Zhang, L.; Wan, D.; Zhou, L.; Zheng, S.; Lin, S.; Qiao, Y. Extracellular matrix and its therapeutic potential for cancer treatment. Signal Transduct. Target. Ther. 2021, 6, 153. [Google Scholar] [CrossRef] [PubMed]
  82. Valkenburg, K.C.; de Groot, A.E.; Pienta, K.J. Targeting the tumour stroma to improve cancer therapy. Nat. Rev. Clin. Oncol. 2018, 15, 366–381. [Google Scholar] [CrossRef] [PubMed]
  83. Henke, E.; Nandigama, R.; Ergün, S. Extracellular Matrix in the Tumor Microenvironment and Its Impact on Cancer Therapy. Front. Mol. Biosci. 2019, 6, 160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Gilbert, L.A.; Hemann, M.T. DNA damage-mediated induction of a chemoresistant niche. Cell 2010, 143, 355–366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Huber, R.M.; Lucas, J.M.; Gomez-Sarosi, L.A.; Coleman, I.; Zhao, S.; Coleman, R.; Nelson, P.S. DNA damage induces GDNF secretion in the tumor microenvironment with paracrine effects promoting prostate cancer treatment resistance. Oncotarget 2015, 6, 2134–2147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Gomez-Sarosi, L.; Sun, Y.; Coleman, I.; Bianchi-Frias, D.; Nelson, P.S. DNA Damage Induces a Secretory Program in the Quiescent TME that Fosters Adverse Cancer Phenotypes. Mol. Cancer Res. 2017, 15, 842–851. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Sun, Y.; Campisi, J.; Higano, C.; Beer, T.M.; Porter, P.; Coleman, I.; True, L.; Nelson, P.S. Treatment-induced damage to the tumor microenvironment promotes prostate cancer therapy resistance through WNT16B. Nat. Med. 2012, 18, 1359–1368. [Google Scholar] [CrossRef]
  88. Ni, Y.; Zhou, X.; Yang, J.; Shi, H.; Li, H.; Zhao, X.; Ma, X. The Role of Tumor-Stroma Interactions in Drug Resistance Within Tumor Microenvironment. Front. Cell Dev. Biol. 2021, 9, 637675. [Google Scholar] [CrossRef]
  89. Vianello, F.; Villanova, F.; Tisato, V.; Lymperi, S.; Ho, K.K.; Gomes, A.R.; Marin, D.; Bonnet, D.; Apperley, J.; Lam, E.W.; et al. Bone marrow mesenchymal stromal cells non-selectively protect chronic myeloid leukemia cells from imatinib-induced apoptosis via the CXCR4/CXCL12 axis. Haematologica 2010, 95, 1081–1089. [Google Scholar] [CrossRef]
  90. Scherzed, A.; Hackenberg, S.; Froelich, K.; Kessler, M.; Koehler, C.; Hagen, R.; Radeloff, A.; Friehs, G.; Kleinsasser, N. BMSC enhance the survival of paclitaxel treated squamous cell carcinoma cells in vitro. Cancer Biol. Ther. 2011, 11, 349–357. [Google Scholar] [CrossRef] [Green Version]
  91. Agarwal, P.; Isringhausen, S.; Li, H.; Paterson, A.J.; He, J.; Gomariz, Á.; Nagasawa, T.; Nombela-Arrieta, C.; Bhatia, R. Mesenchymal Niche-Specific Expression of Cxcl12 Controls Quiescence of Treatment-Resistant Leukemia Stem Cells. Cell Stem Cell 2020, 26, 123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Yu, K.; Yin, Y.; Ma, D.; Lu, T.; Wei, D.; Xiong, J.; Zhou, Z.; Zhang, T.; Zhang, S.; Fang, Q.; et al. Shp2 activation in bone marrow microenvironment mediates the drug resistance of B-cell acute lymphoblastic leukemia through enhancing the role of VCAM-1/VLA-4. Int. Immunopharmacol. 2020, 80, 106008. [Google Scholar] [CrossRef] [PubMed]
  93. Jacamo, R.; Chen, Y.; Wang, Z.; Ma, W.; Zhang, M.; Spaeth, E.L.; Wang, Y.; Battula, V.L.; Mak, P.Y.; Schallmoser, K.; et al. Reciprocal leukemia-stroma VCAM-1/VLA-4-dependent activation of NF-κB mediates chemoresistance. Blood 2014, 123, 2691–2702. [Google Scholar] [CrossRef] [PubMed]
  94. Peled, A.; Klein, S.; Beider, K.; Burger, J.A.; Abraham, M. Role of CXCL12 and CXCR4 in the pathogenesis of hematological malignancies. Cytokine 2018, 109, 11–16. [Google Scholar] [CrossRef]
  95. Möhle, R.; Schittenhelm, M.; Failenschmid, C.; Bautz, F.; Kratz-Albers, K.; Serve, H.; Brugger, W.; Kanz, L. Functional response of leukaemic blasts to stromal cell-derived factor-1 correlates with preferential expression of the chemokine receptor CXCR4 in acute myelomonocytic and lymphoblastic leukaemia. Br. J. Haematol. 2000, 110, 563–572. [Google Scholar] [CrossRef]
  96. Wang, S.; Wang, X.; Liu, S.; Zhang, S.; Wei, X.; Song, Y.; Yin, Q. The CXCR4 Antagonist, AMD3100, Reverses Mesenchymal Stem Cell-Mediated Drug Resistance in Relapsed/Refractory Acute Lymphoblastic Leukemia. Onco Targets Ther. 2020, 13, 6583–6591. [Google Scholar] [CrossRef] [PubMed]
  97. Pillozzi, S.; Bernini, A.; Spiga, O.; Lelli, B.; Petroni, G.; Bracci, L.; Niccolai, N.; Arcangeli, A. Peptides and small molecules blocking the CXCR4/CXCL12 axis overcome bone marrow-induced chemoresistance in acute leukemias. Oncol. Rep. 2019, 41, 312–324. [Google Scholar] [CrossRef] [Green Version]
  98. Clevers, H.; Loh, K.M.; Nusse, R. Stem cell signaling. An integral program for tissue renewal and regeneration: Wnt signaling and stem cell control. Science 2014, 346, 1248012. [Google Scholar] [CrossRef]
  99. Yang, Y.; Mallampati, S.; Sun, B.; Zhang, J.; Kim, S.B.; Lee, J.S.; Gong, Y.; Cai, Z.; Sun, X. Wnt pathway contributes to the protection by bone marrow stromal cells of acute lymphoblastic leukemia cells and is a potential therapeutic target. Cancer Lett. 2013, 333, 9–17. [Google Scholar] [CrossRef] [Green Version]
  100. Wang, J.; Liu, X.; Qiu, Y.; Shi, Y.; Cai, J.; Wang, B.; Wei, X.; Ke, Q.; Sui, X.; Wang, Y.; et al. Cell adhesion-mediated mitochondria transfer contributes to mesenchymal stem cell-induced chemoresistance on T cell acute lymphoblastic leukemia cells. J. Hematol. Oncol. 2018, 11, 11. [Google Scholar] [CrossRef]
  101. Gynn, L.E.; Anderson, E.; Robinson, G.; Wexler, S.A.; Upstill-Goddard, G.; Cox, C.; May, J.E. Primary mesenchymal stromal cells in co-culture with leukaemic HL-60 cells are sensitised to cytarabine-induced genotoxicity, whilst leukaemic cells are protected. Mutagenesis 2021, 36, 419–428. [Google Scholar] [CrossRef] [PubMed]
  102. Wang, W.; Bochtler, T.; Wuchter, P.; Manta, L.; He, H.; Eckstein, V.; Ho, A.D.; Lutz, C. Mesenchymal stromal cells contribute to quiescence of therapy-resistant leukemic cells in acute myeloid leukemia. Eur. J. Haematol. 2017, 99, 392–398. [Google Scholar] [CrossRef]
  103. Ruscetti, F.W.; Akel, S.; Bartelmez, S.H. Autocrine transforming growth factor-beta regulation of hematopoiesis: Many outcomes that depend on the context. Oncogene 2005, 24, 5751–5763. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Fortunel, N.O.; Hatzfeld, A.; Hatzfeld, J.A. Transforming growth factor-beta: Pleiotropic role in the regulation of hematopoiesis. Blood 2000, 96, 2022–2036. [Google Scholar] [CrossRef] [PubMed]
  105. Schelker, R.C.; Iberl, S.; Müller, G.; Hart, C.; Herr, W.; Grassinger, J. TGF-β1 and CXCL12 modulate proliferation and chemotherapy sensitivity of acute myeloid leukemia cells co-cultured with multipotent mesenchymal stromal cells. Hematology 2018, 23, 337–345. [Google Scholar] [CrossRef] [Green Version]
  106. Yuan, B.; El Dana, F.; Ly, S.; Yan, Y.; Ruvolo, V.; Shpall, E.J.; Konopleva, M.; Andreeff, M.; Battula, V.L. Bone marrow stromal cells induce an ALDH+ stem cell-like phenotype and enhance therapy resistance in AML through a TGF-β-p38-ALDH2 pathway. PLoS ONE 2020, 15, e0242809. [Google Scholar] [CrossRef] [PubMed]
  107. Dong-Feng, Z.; Ting, L.; Yong, Z.; Cheng, C.; Xi, Z.; Pei-Yan, K. The TPO/c-MPL pathway in the bone marrow may protect leukemia cells from chemotherapy in AML Patients. Pathol. Oncol. Res. 2014, 20, 309–317. [Google Scholar] [CrossRef]
  108. Xia, B.; Tian, C.; Guo, S.; Zhang, L.; Zhao, D.; Qu, F.; Zhao, W.; Wang, Y.; Wu, X.; Da, W.; et al. c-Myc plays part in drug resistance mediated by bone marrow stromal cells in acute myeloid leukemia. Leuk. Res. 2015, 39, 92–99. [Google Scholar] [CrossRef] [PubMed]
  109. Tian, C.; Zheng, G.; Zhuang, H.; Li, X.; Hu, D.; Zhu, L.; Wang, T.; You, M.J.; Zhang, Y. MicroRNA-494 Activation Suppresses Bone Marrow Stromal Cell-Mediated Drug Resistance in Acute Myeloid Leukemia Cells. J. Cell Physiol. 2017, 232, 1387–1395. [Google Scholar] [CrossRef] [PubMed]
  110. O’ Reilly, E.; Dhami, S.P.S.; Baev, D.V.; Ortutay, C.; Halpin-McCormick, A.; Morrell, R.; Santocanale, C.; Samali, A.; Quinn, J.; O’Dwyer, M.E.; et al. Repression of Mcl-1 expression by the CDC7/CDK9 inhibitor PHA-767491 overcomes bone marrow stroma-mediated drug resistance in AML. Sci. Rep. 2018, 8, 15752. [Google Scholar] [CrossRef] [PubMed]
  111. Konopleva, M.; Konoplev, S.; Hu, W.; Zaritskey, A.Y.; Afanasiev, B.V.; Andreeff, M. Stromal cells prevent apoptosis of AML cells by up-regulation of anti-apoptotic proteins. Leukemia 2002, 16, 1713–1724. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Bolomsky, A.; Vogler, M.; Köse, M.C.; Heckman, C.A.; Ehx, G.; Ludwig, H.; Caers, J. MCL-1 inhibitors, fast-lane development of a new class of anti-cancer agents. J. Hematol. Oncol. 2020, 13, 173. [Google Scholar] [CrossRef] [PubMed]
  113. Nakanishi, M.; Rosenberg, D.W. Multifaceted roles of PGE2 in inflammation and cancer. Semin. Immunopathol. 2013, 35, 123–137. [Google Scholar] [CrossRef] [PubMed]
  114. Carter, B.Z.; Mak, P.Y.; Wang, X.; Tao, W.; Ruvolo, V.; Mak, D.; Mu, H.; Burks, J.K.; Andreeff, M. An ARC-Regulated IL1β/Cox-2/PGE2/β-Catenin/ARC Circuit Controls Leukemia-Microenvironment Interactions and Confers Drug Resistance in AML. Cancer Res. 2019, 79, 1165–1177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Carter, B.Z.; Mak, P.Y.; Chen, Y.; Mak, D.H.; Mu, H.; Jacamo, R.; Ruvolo, V.; Arold, S.T.; Ladbury, J.E.; Burks, J.K.; et al. Anti-apoptotic ARC protein confers chemoresistance by controlling leukemia-microenvironment interactions through a NFκB/IL1β signaling network. Oncotarget 2016, 7, 20054–20067. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Nwabo Kamdje, A.H.; Bassi, G.; Pacelli, L.; Malpeli, G.; Amati, E.; Nichele, I.; Pizzolo, G.; Krampera, M. Role of stromal cell-mediated Notch signaling in CLL resistance to chemotherapy. Blood Cancer J. 2012, 2, e73. [Google Scholar] [CrossRef]
  117. Severin, F.; Frezzato, F.; Visentin, A.; Martini, V.; Trimarco, V.; Carraro, S.; Tibaldi, E.; Brunati, A.M.; Piazza, F.; Semenzato, G.; et al. In Chronic Lymphocytic Leukemia the JAK2/STAT3 Pathway Is Constitutively Activated and Its Inhibition Leads to CLL Cell Death Unaffected by the Protective Bone Marrow Microenvironment. Cancers 2019, 11, 1939. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Kurtova, A.V.; Balakrishnan, K.; Chen, R.; Ding, W.; Schnabl, S.; Quiroga, M.P.; Sivina, M.; Wierda, W.G.; Estrov, Z.; Keating, M.J.; et al. Diverse marrow stromal cells protect CLL cells from spontaneous and drug-induced apoptosis: Development of a reliable and reproducible system to assess stromal cell adhesion-mediated drug resistance. Blood 2009, 114, 4441–4450. [Google Scholar] [CrossRef]
  119. Yang, H.; Zheng, Y.; Zhang, Y.; Cao, Z.; Jiang, Y. Mesenchymal stem cells derived from multiple myeloma patients protect against chemotherapy through autophagy-dependent activation of NF-κB signaling. Leuk. Res. 2017, 60, 82–88. [Google Scholar] [CrossRef] [PubMed]
  120. Baud, V.; Karin, M. Is NF-kappaB a good target for cancer therapy? Hopes and pitfalls. Nat. Rev. Drug Discov. 2009, 8, 33–40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Manni, S.; Carrino, M.; Semenzato, G.; Piazza, F. Old and Young Actors Playing Novel Roles in the Drama of Multiple Myeloma Bone Marrow Microenvironment Dependent Drug Resistance. Int. J. Mol. Sci. 2018, 19, 1512. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Xue, B.Z.; Xiang, W.; Zhang, Q.; Wang, H.F.; Zhou, Y.J.; Tian, H.; Abdelmaksou, A.; Xue, J.; Sun, M.X.; Yi, D.Y.; et al. CD90 glioma—Associated mesenchymal stromal/stem cells promote temozolomide resistance by activating FOXS1-mediated epithelial-mesenchymal transition in glioma cells. Stem Cell Res. Ther. 2021, 12, 394. [Google Scholar] [CrossRef]
  123. Zhang, Q.; Yi, D.Y.; Xue, B.Z.; Wen, W.W.; Lu, Y.P.; Abdelmaksou, A.; Sun, M.X.; Yuan, D.T.; Zhao, H.Y.; Xiong, N.X.; et al. CD90 determined two subpopulations of glioma-associated mesenchymal stem cells with different roles in tumour progression. Cell Death Dis. 2018, 9, 1101. [Google Scholar] [CrossRef]
  124. Vieira de Castro, J.; Gomes, E.D.; Granja, S.; Anjo, S.I.; Baltazar, F.; Manadas, B.; Salgado, A.J.; Costa, B.M. Impact of mesenchymal stem cells’ secretome on glioblastoma pathophysiology. J. Transl. Med. 2017, 15, 200. [Google Scholar] [CrossRef]
  125. Lifshitz, V.; Priceman, S.J.; Li, W.; Cherryholmes, G.; Lee, H.; Makovski-Silverstein, A.; Borriello, L.; DeClerck, Y.A.; Yu, H. Sphingosine-1-Phosphate Receptor-1 Promotes Environment-Mediated and Acquired Chemoresistance. Mol. Cancer Ther. 2017, 16, 2516–2527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Maceyka, M.; Harikumar, K.B.; Milstien, S.; Spiegel, S. Sphingosine-1-phosphate signaling and its role in disease. Trends Cell Biol. 2012, 22, 50–60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Spiegel, S.; Milstien, S. The outs and the ins of sphingosine-1-phosphate in immunity. Nat. Rev. Immunol. 2011, 11, 403–415. [Google Scholar] [CrossRef]
  128. Zhang, H.; Bajraszewski, N.; Wu, E.; Wang, H.; Moseman, A.P.; Dabora, S.L.; Griffin, J.D.; Kwiatkowski, D.J. PDGFRs are critical for PI3K/Akt activation and negatively regulated by mTOR. J. Clin. Investig. 2007, 117, 730–738. [Google Scholar] [CrossRef] [Green Version]
  129. Wang, J.; Cui, R.; Clement, C.G.; Nawgiri, R.; Powell, D.W.; Pinchuk, I.V.; Watts, T.L. Activation PDGFR-α/AKT Mediated Signaling Pathways in Oral Squamous Cell Carcinoma by Mesenchymal Stem/Stromal Cells Promotes Anti-apoptosis and Decreased Sensitivity to Cisplatin. Front. Oncol. 2020, 10, 552. [Google Scholar] [CrossRef]
  130. Liu, C.; Billet, S.; Choudhury, D.; Cheng, R.; Haldar, S.; Fernandez, A.; Biondi, S.; Liu, Z.; Zhou, H.; Bhowmick, N.A. Bone marrow mesenchymal stem cells interact with head and neck squamous cell carcinoma cells to promote cancer progression and drug resistance. Neoplasia 2021, 23, 118–128. [Google Scholar] [CrossRef] [PubMed]
  131. Ullah, M.; Akbar, A.; Ng, N.N.; Concepcion, W.; Thakor, A.S. Mesenchymal stem cells confer chemoresistance in breast cancer via a CD9 dependent mechanism. Oncotarget 2019, 10, 3435–3450. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Yeh, W.L.; Tsai, C.F.; Chen, D.R. Peri-foci adipose-derived stem cells promote chemoresistance in breast cancer. Stem Cell Res. Ther. 2017, 8, 177. [Google Scholar] [CrossRef] [PubMed]
  133. Luo, T.; Liu, Q.; Tan, A.; Duan, L.; Jia, Y.; Nong, L.; Tang, J.; Zhou, W.; Xie, W.; Lu, Y.; et al. Mesenchymal Stem Cell-Secreted Exosome Promotes Chemoresistance in Breast Cancer via Enhancing miR-21-5p-Mediated. Mol. Ther. Oncolytics 2020, 19, 283–293. [Google Scholar] [CrossRef] [PubMed]
  134. Lu, Y.; Yang, Y.; Liu, Y.; Hao, Y.; Zhang, Y.; Hu, Y.; Jiang, L.; Gong, Y.; Wu, K. Upregulation of PAG1/Cbp contributes to adipose-derived mesenchymal stem cells promoted tumor progression and chemoresistance in breast cancer. Biochem. Biophys. Res. Commun. 2017, 494, 719–727. [Google Scholar] [CrossRef]
  135. Romoli, M.; Mazzocchetti, P.; D’Alonzo, R.; Siliquini, S.; Rinaldi, V.E.; Verrotti, A.; Calabresi, P.; Costa, C. Valproic Acid and Epilepsy: From Molecular Mechanisms to Clinical Evidences. Curr. Neuropharmacol. 2019, 17, 926–946. [Google Scholar] [CrossRef] [PubMed]
  136. Barneh, F.; Salimi, M.; Goshadrou, F.; Ashtiani, M.; Mirzaie, M.; Zali, H.; Jafari, M. Valproic acid inhibits the protective effects of stromal cells against chemotherapy in breast cancer: Insights from proteomics and systems biology. J. Cell Biochem. 2018, 119, 9270–9283. [Google Scholar] [CrossRef] [PubMed]
  137. Wang, F.; Zhang, L.; Sai, B.; Wang, L.; Zhang, X.; Zheng, L.; Tang, J.; Li, G.; Xiang, J. BMSC-derived leptin and IGFBP2 promote erlotinib resistance in lung adenocarcinoma cells through IGF-1R activation in hypoxic environment. Cancer Biol. Ther. 2020, 21, 61–71. [Google Scholar] [CrossRef]
  138. Sun, L.; Huang, C.; Zhu, M.; Guo, S.; Gao, Q.; Wang, Q.; Chen, B.; Li, R.; Zhao, Y.; Wang, M.; et al. Gastric cancer mesenchymal stem cells regulate PD-L1-CTCF enhancing cancer stem cell-like properties and tumorigenesis. Theranostics 2020, 10, 11950–11962. [Google Scholar] [CrossRef]
  139. Vrankar, M.; Zwitter, M.; Kern, I.; Stanic, K. PD-L1 expression can be regarded as prognostic factor for survival of non-small cell lung cancer patients after chemoradiotherapy. Neoplasma 2018, 65, 140–146. [Google Scholar] [CrossRef] [PubMed]
  140. Wu, H.; Liu, B.; Chen, Z.; Li, G.; Zhang, Z. MSC-induced lncRNA HCP5 drove fatty acid oxidation through miR-3619-5p/AMPK/PGC1α/CEBPB axis to promote stemness and chemo-resistance of gastric cancer. Cell Death Dis. 2020, 11, 233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. He, W.; Liang, B.; Wang, C.; Li, S.; Zhao, Y.; Huang, Q.; Liu, Z.; Yao, Z.; Wu, Q.; Liao, W.; et al. MSC-regulated lncRNA MACC1-AS1 promotes stemness and chemoresistance through fatty acid oxidation in gastric cancer. Oncogene 2019, 38, 4637–4654. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Ji, R.; Zhang, B.; Zhang, X.; Xue, J.; Yuan, X.; Yan, Y.; Wang, M.; Zhu, W.; Qian, H.; Xu, W. Exosomes derived from human mesenchymal stem cells confer drug resistance in gastric cancer. Cell Cycle 2015, 14, 2473–2483. [Google Scholar] [CrossRef] [Green Version]
  143. Ji, N.; Yu, J.W.; Ni, X.C.; Wu, J.G.; Wang, S.L.; Jiang, B.J. Bone marrow-derived mesenchymal stem cells increase drug resistance in CD133-expressing gastric cancer cells by regulating the PI3K/AKT pathway. Tumour. Biol. 2016, 37, 14637–14651. [Google Scholar] [CrossRef]
  144. Timaner, M.; Letko-Khait, N.; Kotsofruk, R.; Benguigui, M.; Beyar-Katz, O.; Rachman-Tzemah, C.; Raviv, Z.; Bronshtein, T.; Machluf, M.; Shaked, Y. Therapy-Educated Mesenchymal Stem Cells Enrich for Tumor-Initiating Cells. Cancer Res. 2018, 78, 1253–1265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Yu, Y.; Yang, F.H.; Zhang, W.T.; Guo, Y.D.; Ye, L.; Yao, X.D. Mesenchymal stem cells desensitize castration-resistant prostate cancer to docetaxel chemotherapy via inducing TGF-β1-mediated cell autophagy. Cell Biosci. 2021, 11, 7. [Google Scholar] [CrossRef] [PubMed]
  146. Maj, M.; Bajek, A.; Nalejska, E.; Porowinska, D.; Kloskowski, T.; Gackowska, L.; Drewa, T. Influence of Mesenchymal Stem Cells Conditioned Media on Proliferation of Urinary Tract Cancer Cell Lines and Their Sensitivity to Ciprofloxacin. J. Cell Biochem. 2017, 118, 1361–1368. [Google Scholar] [CrossRef] [PubMed]
  147. Wen, Y.; Guo, Y.; Huang, Z.; Cai, J.; Wang, Z. Adipose-derived mesenchymal stem cells attenuate cisplatin-induced apoptosis in epithelial ovarian cancer cells. Mol. Med. Rep. 2017, 16, 9587–9592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Coffman, L.G.; Choi, Y.J.; McLean, K.; Allen, B.L.; di Magliano, M.P.; Buckanovich, R.J. Human carcinoma-associated mesenchymal stem cells promote ovarian cancer chemotherapy resistance via a BMP4/HH signaling loop. Oncotarget 2016, 7, 6916–6932. [Google Scholar] [CrossRef] [Green Version]
  149. Liao, X.; Siu, M.K.; Au, C.W.; Wong, E.S.; Chan, H.Y.; Ip, P.P.; Ngan, H.Y.; Cheung, A.N. Aberrant activation of hedgehog signaling pathway in ovarian cancers: Effect on prognosis, cell invasion and differentiation. Carcinogenesis 2009, 30, 131–140. [Google Scholar] [CrossRef] [PubMed]
  150. McCann, C.K.; Growdon, W.B.; Kulkarni-Datar, K.; Curley, M.D.; Friel, A.M.; Proctor, J.L.; Sheikh, H.; Deyneko, I.; Ferguson, J.A.; Vathipadiekal, V.; et al. Inhibition of Hedgehog signaling antagonizes serous ovarian cancer growth in a primary xenograft model. PLoS ONE 2011, 6, e28077. [Google Scholar] [CrossRef] [Green Version]
  151. Han, Z.; Jing, Y.; Xia, Y.; Zhang, S.; Hou, J.; Meng, Y.; Yu, F.; Liu, X.; Wu, M.; Zhang, P.; et al. Mesenchymal stem cells contribute to the chemoresistance of hepatocellular carcinoma cells in inflammatory environment by inducing autophagy. Cell Biosci. 2014, 4, 22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Tu, B.; Zhu, J.; Liu, S.; Wang, L.; Fan, Q.; Hao, Y.; Fan, C.; Tang, T.T. Mesenchymal stem cells promote osteosarcoma cell survival and drug resistance through activation of STAT3. Oncotarget 2016, 7, 48296–48308. [Google Scholar] [CrossRef] [Green Version]
  153. Frolova, O.; Samudio, I.; Benito, J.M.; Jacamo, R.; Kornblau, S.M.; Markovic, A.; Schober, W.; Lu, H.; Qiu, Y.H.; Buglio, D.; et al. Regulation of HIF-1α signaling and chemoresistance in acute lymphocytic leukemia under hypoxic conditions of the bone marrow microenvironment. Cancer Biol. Ther. 2012, 13, 858–870. [Google Scholar] [CrossRef]
  154. Roodhart, J.M.; Daenen, L.G.; Stigter, E.C.; Prins, H.J.; Gerrits, J.; Houthuijzen, J.M.; Gerritsen, M.G.; Schipper, H.S.; Backer, M.J.; van Amersfoort, M.; et al. Mesenchymal stem cells induce resistance to chemotherapy through the release of platinum-induced fatty acids. Cancer Cell 2011, 20, 370–383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Bajetto, A.; Pattarozzi, A.; Corsaro, A.; Barbieri, F.; Daga, A.; Bosio, A.; Gatti, M.; Pisaturo, V.; Sirito, R.; Florio, T. Different Effects of Human Umbilical Cord Mesenchymal Stem Cells on Glioblastoma Stem Cells by Direct Cell Interaction or Via Released Soluble Factors. Front. Cell Neurosci. 2017, 11, 312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Krueger, T.E.G.; Thorek, D.L.J.; Denmeade, S.R.; Isaacs, J.T.; Brennen, W.N. Concise Review: Mesenchymal Stem Cell-Based Drug Delivery: The Good, the Bad, the Ugly, and the Promise. Stem Cells Transl. Med. 2018, 7, 651–663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Babajani, A.; Soltani, P.; Jamshidi, E.; Farjoo, M.H.; Niknejad, H. Recent Advances on Drug-Loaded Mesenchymal Stem Cells With Anti-neoplastic Agents for Targeted Treatment of Cancer. Front. Bioeng. Biotechnol. 2020, 8, 748. [Google Scholar] [CrossRef]
  158. Takayama, Y.; Kusamori, K.; Nishikawa, M. Mesenchymal stem/stromal cells as next-generation drug delivery vehicles for cancer therapeutics. Expert Opin. Drug Deliv. 2021, 18, 1627–1642. [Google Scholar] [CrossRef] [PubMed]
Figure 1. MSCs in the tumour microenvironment affect the success of anticancer therapy. There are two potential outcomes: (A) chemotherapeutic agents induce cell death in cancer cells or (B) the interaction of cancer cells, treated with a chemotherapeutic agent, with MSCs and the extracellular matrix in the tumour microenvironment may result in an increased risk of cancer cells developing drug resistance.
Figure 1. MSCs in the tumour microenvironment affect the success of anticancer therapy. There are two potential outcomes: (A) chemotherapeutic agents induce cell death in cancer cells or (B) the interaction of cancer cells, treated with a chemotherapeutic agent, with MSCs and the extracellular matrix in the tumour microenvironment may result in an increased risk of cancer cells developing drug resistance.
Cancers 14 03761 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ramuta, T.Ž.; Kreft, M.E. Mesenchymal Stem/Stromal Cells May Decrease Success of Cancer Treatment by Inducing Resistance to Chemotherapy in Cancer Cells. Cancers 2022, 14, 3761. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers14153761

AMA Style

Ramuta TŽ, Kreft ME. Mesenchymal Stem/Stromal Cells May Decrease Success of Cancer Treatment by Inducing Resistance to Chemotherapy in Cancer Cells. Cancers. 2022; 14(15):3761. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers14153761

Chicago/Turabian Style

Ramuta, Taja Železnik, and Mateja Erdani Kreft. 2022. "Mesenchymal Stem/Stromal Cells May Decrease Success of Cancer Treatment by Inducing Resistance to Chemotherapy in Cancer Cells" Cancers 14, no. 15: 3761. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers14153761

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

Article Metrics

Back to TopTop