Research advances in the role of gastric cancer‑derived mesenchymal stem cells in tumor progression (Review)

  • Authors:
    • Jiaqi Shen
    • Wei Zhu
  • View Affiliations

  • Published online on: December 4, 2020     https://doi.org/10.3892/ijmm.2020.4810
  • Pages: 455-462
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Gastric cancer (GC) is one of the most common malignancies and the second leading cause of cancer‑associated death in the world. The carcinogenesis and development of GC involves complicated steps and various factors, in which the tumor microenvironment serves a vital role. Mesenchymal stem cells (MSCs), also known as mesenchymal stromal cells, are multipotent stromal cells, and have gained increasing attention due to their wound‑healing ability, as well as their tumor‑promoting potential. MSCs are essential components of the tumor microenvironment and serve important roles in tumor initiation, progression and metastasis. The present review focuses on GC and discusses recent advances in understanding the effect of GC‑derived MSC‑like cells (GC‑MSCs) on tumor progression, chemoresistance and immune escape. Additionally, the mechanism underlying the tumor tropism of bone marrow‑derived MSCs and the malignant transition of these cells to GC‑MSCs are addressed. The potential of GC‑MSCs in the treatment of GC, such as for predicting prognosis and as therapeutic targets, is also discussed in association with their critical role in tumor progression. The information on the characteristics and functions of GC‑MSCs provided in the present review may promote the development of novel therapeutic strategies against GC.

1. Introduction

According to global cancer statistics, gastric cancer (GC) ranked fifth in incidence (5.7% of the total cases) and third in mortality (8.2% of the total cancer deaths) among malignancies worldwide in 2018 (1). When detected early, GC is usually curable and has a 5-year survival rate of >90%; however, the prognosis of patients with advanced GC remains poor (2). The lack of early clinical symptoms often delays the diagnosis of GC, resulting in the development of an incurable disease in a number of patients (3). Another reason for the high mortality is that surgical resection is the only curative treatment for GC (4). Despite the availability of a number of novel therapies, including targeted therapy and immunotherapy (3,5), the treatment of GC remains unsatisfactory. Therefore, elucidating the underlying mechanisms underlying tumor progression may help develop more effective treatments.

The tumor microenvironment (TME) is a complex and dynamic cellular community composed of cancer cells, endothelial cells, fibroblasts, immune cells, and mesenchymal stem cells (MSCs) (6). The TME is formed via the recruitment of tumor-supporting MSCs and extensive remodeling of adjacent tissues; thus, the TME differs from normal tissues in numerous aspects, including the extracellular matrix, blood vessels and phenotypes of cells (7). The interaction between tumor cells and the TME serves an important role in tumor initiation, progression, chemoresistance and immune escape, and certain molecules present in the TME are prognosis predictors in various types of cancer, such as pancreatic cancer, GC and urothelial carcinoma (8-11). MSCs, which are important components of the TME and serve critical roles in tumor progression, have been extensively studied.

MSCs, also known as mesenchymal stromal cells, are multipotent stromal cells with the ability to differentiate into osteoblasts, adipocytes, chondrocytes and other types of cells under different conditions (12). Despite extensive research efforts, the multi-differentiation potential of MSCs has only been demonstrated in vitro, and there are few studies describing their characteristics in vivo (13,14). MSCs can accelerate wound healing by regulating inflammation (15). Additionally, MSCs can suppress the function of both innate and adaptive immune cells, including macrophages and lymphocytes (15). In cases of weak inflammatory responses, MSCs can act as antigen-presenting cells and increase the immune response (15). In cancer, MSCs are critical components of the TME and promote the progression of several types of tumor, such as hepatocarcinoma and colorectal cancer (16,17). Tumors are considered to be 'wounds that never heal', and MSCs can migrate to injured tissues, supporting the tumor tropism of MSCs and their ability to sense wound-associated signals (18). Upon recruitment to tumors, MSCs are converted into tumor-associated MSCs (TA-MSCs), which can promote tumor progression more potently (19). TA-MSCs interact with tumor cells and are involved in the remodeling of the TME in response to signals from tumor cells and other stromal cells (19,20). The crosstalk between TA-MSCs and tumor cells can be mediated by several mechanisms, including paracrine signals, exosomes and direct contact (21,22). Furthermore, TA-MSCs can differentiate into cancer-associated fibroblasts, which promote tumor progression (23).

In the present review, the effect of GC-derived MSC-like cells (GC-MSCs) on GC progression, metastasis, chemo-resistance and immune escape are described. Additionally, the mechanisms by which GC cells, immune cells and other stromal cells educate MSCs and skew MSCs towards the GC-MSC fate are discussed. Finally, the therapeutic potential of GC-MSCs as both targets and biomarkers are summarized.

2. Biological characteristics of GC-MSCs

Friedenstein et al (24) discovered the presence of mouse bone marrow (BM)-derived fibroblast-like cells in the early 1970s, which are currently known as MSCs. MSCs are present in numerous tissues, including the stomach (25). GC-MSCs have been successfully isolated and characterized from patients with GC (26,27). Similarly to other stem cells, GC-MSCs have the potential to differentiate into adipocytes and osteocytes (28). Since no specific surface markers of GC-MSCs have been identified, they can only be characterized by in vitro experiments; GC-MSCs express high levels of CD29, CD90 and CD105 and barely detectable levels of CD34, CD45 and CD19 (28). Although BM-MSCs and GC-adjacent non-cancerous tissue-derived MSCs (GCN-MSCs) can transform into GC-MSCs in the TME, these three types of cells exhibit both similarities and differences (23). The three types of cells have similar morphology when isolated from patients (Fig. 1A), as well as surface markers and differentiation potential. They are all slender and look like a shuttle under the microscope. The three types of cells are positive for CD29, CD44 and CD105, and negative for CD14 and CD34 (26). BM-MSCs and GCN-MSCs express CD13, whereas GC-MSCs do not (26). They all have a pluripotent differentiation potential and can differentiate into osteocytes and adipocytes (26). However, GC-MSCs can proliferate twice as fast as GCN-MSCs and BM-MSCs (26). GC-MSCs differ from BM-MSCs in the number of cytoplasmic organelles, tumor-promoting capacity and secretion levels of numerous inflammatory cytokines, such as IL-6, monocyte chemoattractant protein 1 and VEGF (26,29). GC-MSCs can improve the proliferative, migratory and proangiogenic abilities of GC cells more potently than GCN-MSCs and BM-MSCs, partly by secreting IL-8 (30). These findings suggest that GC-MSCs maintain the mesenchymal lineage and stem cell capabilities, and differ from MSCs from non-GC tissues in several aspects, especially their tumor-promoting capabilities.

3. Formation of GC-MSCs

MSCs migrate to the tumor

Studies have revealed that BM-MSCs have tropism to several types of tumor, such as glioma and breast cancer, by intravenous or intraperitoneal injection (31,32). CXC-chemokine ligand 16 (CXCL16) secreted by breast cancer cells binds to its receptor CXC-chemokine receptor 6 (CXCR6) on BM-MSCs, which in turn produce CXCL10, thereby promoting the recruitment of BM-MSCs to cancer cells (33). The combination of CXCL12 and CXCR6 facilitates the recruitment of BM-MSCs to prostate tumors (34). Other chemokines and growth factors also participate in the migration of MSCs from non-cancer to cancer tissues, such as transforming growth factor-β (TGF-β), platelet-derived growth factor, monocyte chemoattractant protein-1 and stromal cell-derived factor-1 (35-37). Hypoxia in the TME induces BM-MSCs tropism to breast cancer (33). Thus, there are various mechanisms involved in the migration of MSCs from non-cancer to tumor tissues. In GC, Berger et al (38) performed a 'plug assay', in which GC and lung carcinoma cell-derived microvesicles (MVs) were collected and used as a Matrigel plug, which was implanted into teratoma tissues; 6-10 days after the injection, the plug was harvested and subjected to histological analysis or dissociated into a single cell suspension. The results revealed that MSCs were present in the plug containing GC cell-derived MVs, whereas the control group did not exhibit MSCs, suggesting that GC cell-derived MVs are responsible for the migration of MSCs, although the underlying mechanism remains unknown (38). To the best of our knowledge, there are no studies investigating the tropism of MSCs from non-GC to GC tissues, and additional studies are required to understand this migration and to identify key cytokines or chemokines.

MSCs transform into GC-MSCs

After their recruitment to the tumor site, MSCs from non-cancer tissues are converted into TA-MSCs under the influence of tumor cells, immune cells, local TA-MSCs and other stromal cells (34). In GC, tumor cells are involved in the malignant transition of MSCs through several mechanisms (Table I). For example, Shamai et al (39) revealed that GC cells have the capacity to increase R-spondin expression in GC-MSCs, and GC-MSCs can in turn upregulate Lgr5 expression in GC cells. Therefore, tumor cells can alter gene expression in GC-MSCs and GC-MSCs can induce tumor cell stemness. Exosomes from GC cells regulate the immunomodulatory properties of adipose-derived MSCs by activating the NF-κB signaling pathway (40). GC-MSCs exposed to GC cell-derived exosomes can activate both macrophages and T lymphocytes, thus maintaining the inflammatory TME and promoting tumor progressio (40). MicroRNAs (miRNAs/miRs) are involved in the malignant transition of MSCs from non-GC tissues. The downregulation of miR-155-5p expression caused by the activation of NF-κB signaling in GC-MSCs promotes this process, as demonstrated by the effect of miR-155-5p inhibition on conferring BM-MSCs a GC-MSC-like phenotype and function (41). Unlike miR-155-5p, miR-374 expression is upregulated in GC-MSCs, and overexpression of miR-374 promotes the proliferation and migration of MSCs from normal gastric tissues by regulating the Wnt5a/β-catenin signaling pathway (42,43). Additionally, immune cells can participate in the malignant transition of MSCs. Macrophages activate umbilical cord-derived MSCs and confer these MSCs a pro-inflammatory phenotype, which can promote GC progression partly through the NF-κB signaling pathway, as demonstrated by in vitro and in vivo experiments (44). CD4+ T lymphocytes may also be involved in this process; although the effect of CD4+ T cells on the malignant transition of MSCs has not been investigated, these cells upregulate the expression levels of PD-L1 in GC-MSCs through the p-STAT3 signaling pathway, and GC-MSCs with positive PDL1 expression protect tumor cells from immune cells (45). Patients with GC infected with Helicobacter pylori have a high mortality rate because this bacterium confers MSCs a pro-inflammatory phenotype through the activation of the NF-κB signaling pathway (46). Overall, these findings suggest that GC cells, as well as immune cells and bacteria, can convert BM-MSCs into GC-MSCs (Fig. 1B).

Table I

Mechanisms of the malignant transition of MSCs to GC-MSCs.

Table I

Mechanisms of the malignant transition of MSCs to GC-MSCs.

First author, yearRegulation mechanismEffect on malignant transition(Refs.)
Shamai et al, 2019GC cells increased R-spondin expression in GC-MSCsPromote(39)
Shen et al, 2019GC cells secreted exosomes to activate NF-κB signaling pathwayPromote(40)
Zhu et al, 2016miR-155-5p inhibited NF-κB signaling pathwayInhibit(41)
Ji et al, 2017; Sun et al, 2018miR-374 activated Wnt5a/β-catenin signaling pathwayPromote(42,43)
Yang et al, 2014Macrophages activated NF-κB signaling pathwayPromote(44)
Xu et al, 2018CD4+ T cells upregulated PD-L1 expression in GC-MSCs through phosphorylated-STAT3Enhance GC-MSCs tumor-promoting effect(45)
Zhang et al, 2016Helicobacter pylori infection activated NF-κB signaling pathwayPromote(46)

[i] miR, microRNA; GC-MSCs, gastric cancer-derived MSC-like cells; PD-L1, programmed cell death-ligand 1.

4. Effects of GC-MSCs on tumor progression and the tumor microenvironment

GC-MSCs promote tumor progression

TA-MSCs promote tumor progression by secreting cytokines, which act directly on tumor cells or other stromal cells in the TME (47-49). GC-MSCs can promote GC progression through various mechanisms (Fig. 2). For example, IL-15 secreted by GC-MSCs induces GC cell epithelial-mesenchymal transition and promotes GC cell migration, which are associated with tumor growth and metastasis, respectively (28). Consistently, IL-15 levels are higher in patients with GC than in healthy donors in both serum and tissues in association with lymph node metastasis (28). In addition, GC-MSCs produce high levels of IL-8, which serves a role in tumor cell proliferation and migration, and in angiogenesis (30). However, the tumor-promoting ability of IL-8 has only been demonstrated in vitro and the underlying mechanisms remain unknown. In addition to cytokines, other molecules serve roles in the tumor-promoting effect of GC-MSCs. The interaction between GC cells and GC-MSCs maintains GC cell stemness through the activation of the R-spondin/Lgr5 axis and WNT/β-catenin signaling pathway (39). Hepatocyte growth factor (HGF) exclusively secreted by GC-MSCs promotes the proliferation and migration of GC cells through the activation of the HGF/c-MET signaling pathway (38). Furthermore, platelet-derived growth factor-DD (PDGF-DD) secreted by GC-MSCs increases the phosphorylation of PDGF receptor-β in GC cells, thus promoting GC cell proliferation and migration; recombinant PDGF-DD can mimic the tumor-promoting effect of GC-MSCs conditioned medium (CM) on GC cell proliferation and migration (50). GC-MSCs induce VEGF expression in GC cells in vitro and in vivo, and contribute to GC-MSC-mediated angiogenesis by activating the NF-κB/VEGF signaling pathway (51). Since tumor neovascularization is indispensable for continuous tumor growth, this pathway may be a potential target to inhibit tumor growth. Notably, lung carcinoma cell proliferation is independent of lung carcinoma MSCs, whereas GC cell proliferation is critically dependent on the presence of their counterparts GC-MSCs (38). This phenomenon suggests that tumor growth does not always depend on the counterpart TA-MSCs. The interaction between tumor cells and TA-MSCs is specific not only for the requirements of the tumor cells, but also their capacity to recruit MSCs and educate them to further promote tumor progression (38). Overall, these results demonstrate that GC-MSCs serve essential roles in GC progression.

GC-MSCs induce tumor chemoresistance

Chemotherapy is the first-line treatment for patients with advanced GC; pre- or post-operative chemotherapy with 5-fluorouracil (5-FU) and cisplatin has improved the survival rates of patients with GC (52-54). However, the response rate for this therapy is limited by the development of chemoresistance (55,56). Therefore, it is urgent to investigate the mechanisms underlying GC cell chemoresistance. The TME can protect tumors from chemo-therapy through physical barriers and metabolites, exosomes and other substances secreted by tumor stromal cells (57). One of the key components of the TME, TA-MSCs, induces tumor cell chemoresistance through various mechanism, such as elevating tumor cell stemness and secreting certain molecules, such as interleukins and chemokines (58-60). Cancer stem cells (CSCs), also known as cancer initiation cells, were identified based on the observation of histological heterogeneity in tumors and on the fact that a single mouse tumor cell can form a new tumor (61). Although the definition of CSC is contentious, these cells have self-renewal and differentiation capacities and sustain the growth of tumors (62,63), which is associated with tumor cell chemoresistance. In GC, He et al (64) suggested that MSCs promote GC cell stemness and chemoresistance through fatty acid oxidation (FAO) based on in vitro and in vivo experiments. Mechanistically, TGF-β1 secreted by MSCs activates SMAD2/3 through TGF-β recep-tors, which then induces lncRNA MACC1-AS1 expression in GC cells and promotes FAO-dependent stemness and chemoresistance by antagonizing miR-145-5p (64). GC-MSCs express high levels of the ATP-binding cassette subfamily B member 1 transporter, which results in decreased drug accumulation in chemoresistant cells (39). Exosomes secreted by GC-MSCs induce GC cell resistance to 5-FU by activating calcium/calmodulin-dependent protein kinases and the Raf/MEK/ERK kinase cascade, thus upregulating the expression levels of multi-drug resistance-associated proteins (65). The aforementioned studies indicate that GC-MSCs can induce GC cell chemoresistance by secreting cytokines and exosomes, although there may be other undiscovered mechanisms that may help develop strategies to overcome chemoresistance.

GC-MSCs suppress antitumor immunity

The number and types of inflammatory factors in the TME can alter the immune responses to tumors, although the underlying mechanisms remain obscure. MSCs modulate immune cells and can suppress the immune response; however, they can also promote immune responses when inflammatory conditions are not enough (66). Additionally, GC-MSCs can modulate antitumor immunity by interacting with immune cells, such as macro-phages, neutrophils and T lymphocytes. Macrophages are associated with a poor prognosis of GC and are used as prognostic indicators (67). Li et al (68) suggested that GC-MSCs may trigger the polarization and generation of M2-like macro-phages by activating the JAK2/STAT3 signaling pathway via high secretion of IL-6/IL-8. M2-like macrophages can facilitate the metastasis and progression of GC by enhancing epithelial-mesenchymal transition in GC cells (68). Exosomes extracted from the GC AGS cell line can induce macrophage phagocytosis and promote the secretion of pro-inflammatory factors, thereby activating CD69 and CD25 on the surface of T cells through the NF-κB signaling pathway in MSCs (40). Regarding neutrophils, there is a reciprocal interaction between GC-MSCs and neutrophils. GC-MSCs can induce chemotaxis and neutrophil activation, as well as suppress neutrophil spontaneous apoptosis through the activation of the STAT3 and ERK1/2 signaling pathways (29). Neutrophils incubated with GC-MSCs or GC-MSCs-CM can promote the migration of tumor cells and induce the formation of tube-like structures in endothelial cells (29). Furthermore, GC-MSC-treated neutrophils can in turn convert normal MSCs into tumor-associated fibroblasts (30). GC-MSCs-CM pretreatment reverses the inhibitory effect of peripheral blood mononuclear cells and promotes GC liver metastases by disrupting the balance of regulatory T cells and T helper 17 cells (69). Both innate and adaptive immune cells can be affected by GC-MSCs, and they can gain tumor-promoting abilities or be inhibited.

GC-MSCs upregulate PD-L1 expression in GC cells

In past years, immune checkpoints, including programmed cell death protein 1 (PD-1)/programmed cell death 1 ligand 1 (PD-L1), cytotoxic T-lymphocyte-associated protein 4 and T-cell immunoglobulin domain and mucin domain-3, have attracted increasing attention for their ability to weaken the function of T lymphocytes and induce tumor immune escape. Blocking the interaction between PD1 and PD-L1 has exhibited promising results in the treatment of several types of cancer, including breast cancer and squamous cell carcinoma of the head and neck (70,71). In GC, pembrolizumab, a PD-1 inhibitor, has shown good results in phase I/II trials, with objective response rates of 11.6-25.8% and low toxicity (72,73), and the combination of nivolumab and regorafenib, which respectively inhibit PD-1 or VEFGR tyrosine kinase, also exhibits antitumor activity (74). However, the response to this therapy cannot be predicted due to a lack of effective biomarkers (72). GC-MSCs upregulate PD-L1 expression in GC cells by secreting IL-8, which can regulate the STAT3 and mTOR signaling pathways (75). The role of GC-MSCs in regulating PD-L1 in GC cells has not been investigated extensively. Further investigation of the involvement of GC-MSCs in the regulation of PD-L1 expression and elucidation of the underlying mechanism may help the development of anti-PD-L1 therapy and may provide novel biomarkers.

5. Therapeutic opportunities for GC-MSCs

Since MSCs can be isolated from bone marrow and adipose tissues, they have been used in numerous studies in past years (76,77). MSCs from non-cancer tissues can suppress immune responses and have immune evasion properties; they are therefore stable in an allogeneic environment and display promise for cell therapy (78). For instance, exosomes from MSCs from non-GC tissues and MSCs themselves can serve as vehicles to transport miRNAs or drugs and cytokines to tumors, thus suppressing tumor progression, according to laboratory tests and clinic trials (79-81).

Unlike MSCs from non-GC tissues, GC-MSCs cannot be used as drug carriers due to their critical role in tumor progression. Instead, they should be specifically targeted to eliminate their effect on tumor cell proliferation, drug-resistance and migration. However, GC-MSCs have no specific surface markers, and therefore they cannot be targeted without affecting other cells. Only the substances released by GC-MSCs can serve as targets to suppress their tumor-promoting capability. For example, GC-MSCs can induce chemoresistance in tumor cells through FAO, and inhibition of FAO attenuates this phenomenon, suggesting that FAO may be a potential target to reduce chemoresistance in GC (64). Similarly, GC-MSCs-CM treatment can promote tumor cell proliferation and migration and increase pro-angiogenic abilities through the secretion of IL-8; therefore, the use of an IL-8 neutralizing antibody may suppress the effects of GC-MSCs (30). In addition, IL-8 produced by GC-MSCs upregulates PD-L1 expression in tumor cells and can thus induce tumor cell resistance to CD8+ T cells, and inhibition of IL-8 can eliminate resistance (75). The afore-mentioned studies indicate that target key modulators in the tumor-promoting process of GC-MSCs can suppress tumor progression and chemoresistance. Furthermore, this strategy may be effective in combination with other therapies, such as chemotherapy and anti-PD-L1 therapy.

Furthermore, miRNAs and cytokines secreted by GC-MSCs can predict prognosis. For instance, GC-MSCs have higher expression levels of miR-214, miR-221 and miR-222 than GCN-MSCs, and high expression levels of miR-221 and miR-222, or miR-214 and miR-222 in the tissues of patients with GC are positively associated with lymph node metastasis and serosal invasion, respectively (82). IL-15 in the GC microenvironment is mostly derived from GC-MSCs and is associated with lymph node metastasis (28). GC-MSCs can secrete high levels of IL-8, which predicts a poor prognosis in patients with GC (30). In summary, GC-MSCs can potentially be developed into novel therapies and prognostic biomarkers.

6. Conclusions

In past years, the interaction between MSCs and tumors has gained increasing attention. The present review focused on the transformation of MSCs from non-GC tissues into GC-MSCs and the role of GC-MSCs in tumor progression, chemoresistance and immune escape. In addition to GC cells, immune cells and bacteria can be involved in the malignant transformation of MSCs into GC-MSCs. GC-MSCs can in turn promote tumor progression, induce chemoresistance and confer immune cells a tumor-promoting phenotype. Regarding their therapeutic potential, the upstream or downstream modulators of GC-MSCs can serve as targets to weaken their effect on tumor progression. However, the interaction between other stromal cells and GC-MSCs, and the underlying mechanisms require further investigation, including the roles of natural killer cells and endothelial cells. In addition, specific surface markers of GC-MSCs remain to be identified, which may facilitate the specific targeting of GC-MSCs without affecting other cells. The association between GC-MSCs and PD-L1 should be investigated further, as it may provide new insight into PD-L1 co-therapy. Despite numerous advances in the understanding of the effect of GC-MSCs on tumor progression, elucidating the function and underlying mechanisms of GC-MSCs may provide valuable information to improve the treatment of GC.

Funding

The present study was supported by the National Science Foundation of China (grant nos. 81972313, 81972822 and 81772641), the Wu Jieping Medical Foundation (grant no. 320.6750.19060) and the Bethune Charitable Foundation (grant no. G-X-2019-0101-12).

Availability of data and materials

Not applicable.

Authors' contributions

JS and WZ reviewed the literature on the role of GC-MSCs in tumor progression and the tumor microenvironment, and wrote most of the review. WZ discussed the role of GC-MSCs in GC treatment. Both authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

References

1 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Suzuki H, Oda I, Abe S, Sekiguchi M, Mori G, Nonaka S, Yoshinaga S and Saito Y: High rate of 5-year survival among patients with early gastric cancer undergoing curative endoscopic submucosal dissection. Gastric Cancer. 19:198–205. 2016. View Article : Google Scholar

3 

Hsu A, Chudasama R, Almhanna K and Raufi A: Targeted therapies for gastroesophageal cancers. Ann Transl Med. 8:11042020. View Article : Google Scholar : PubMed/NCBI

4 

Hunt RH, Camilleri M, Crowe SE, El-Omar EM, Fox JG, Kuipers EJ, Malfertheiner P, McColl KE, Pritchard DM, Rugge M, et al: The stomach in health and disease. Gut. 64:1650–1668. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Dolcetti R, De Re V and Canzonieri V: Immunotherapy for Gastric Cancer: Time for a personalized approach? Int J Mol Sci. 19:16022018. View Article : Google Scholar :

6 

Cheng HS, Lee JXT, Wahli W and Tan NS: Exploiting vulnerabilities of cancer by targeting nuclear receptors of stromal cells in tumor microenvironment. Mol Cancer. 18:512019. View Article : Google Scholar : PubMed/NCBI

7 

Zhou ZX and Lu ZR: Molecular imaging of the tumor microen-vironment. Adv Drug Deliver Rev. 113:24–48. 2017. View Article : Google Scholar

8 

Denton AE, Roberts EW and Fearon DT: Stromal Cells in the Tumor Microenvironment. Adv Exp Med Biol. 1060:99–114. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Zhan HX, Zhou B, Cheng YG, Xu JW, Wang L, Zhang GY and Hu SY: Crosstalk between stromal cells and cancer cells in pancreatic cancer: New insights into stromal biology. Cancer Lett. 392:83–93. 2017. View Article : Google Scholar : PubMed/NCBI

10 

Chen DX, Chen G, Jiang W, Fu M, Liu W, Sui J, Xu S, Liu Z, Zheng X, Chi L, et al: Association of the collagen signature in the tumor microenvironment with lymph node metastasis in early gastric cancer. JAMA Surg. 154:e1852492019. View Article : Google Scholar : PubMed/NCBI

11 

Hodgson A, Xu B, Satkunasivam R and Downes MR: Tumour front inflammation and necrosis are independent prognostic predictors in high-grade urothelial carcinoma of the bladder. J Clin Pathol. 71:154–160. 2018. View Article : Google Scholar

12 

Patil S and Singh N: Spatially controlled functional group grafting of silk films to induce osteogenic and chondrogenic differentiation of human mesenchymal stem cells. Mater Sci Eng C Mater Biol Appl. 91:796–805. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Ghaneialvar H, Soltani L, Rahmani HR, Lotfi AS and Soleimani M: Characterization and classification of mesenchymal stem cells in several species using surface markers for cell therapy purposes. Indian J Clin Biochem. 33:46–52. 2018. View Article : Google Scholar : PubMed/NCBI

14 

Schafer R and Bieback K: Characterization of mesenchymal stem or stromal cells: Tissue sources, heterogeneity, and function. Transfusion. 56:S2–S5. 2016. View Article : Google Scholar

15 

Wang Y, Chen X, Cao W and Shi Y: Plasticity of mesenchymal stem cells in immunomodulation: Pathological and therapeutic implications. Nat Immunol. 15:1009–1016. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Zong C, Zhang HJ, Yang X, Gao L, Hou J, Ye F, Jiang J, Yang Y, Li R, Han Z and Wei L: The distinct roles of mesenchymal stem cells in the initial and progressive stage of hepatocarcinoma. Cell Death Dis. 9:3452018. View Article : Google Scholar : PubMed/NCBI

17 

Zhang X, Hu FY, Li G, Li G, Yang X, Liu L, Zhang R, Zhang B and Feng Y: Human colorectal cancer-derived mesenchymal stem cells promote colorectal cancer progression through IL-6/JAK2/STAT3 signaling. Cell Death Dis. 9:252018. View Article : Google Scholar : PubMed/NCBI

18 

Dvorak HF: Tumors: Wounds that do not heal. Similarities between tumor stroma generation and wound healing. N Engl J Med. 315:1650–1659. 1986. View Article : Google Scholar : PubMed/NCBI

19 

Chen J, Ma L, Zhang N, Zhu Y, Zhang K, Xu Z and Wang Q: Mesenchymal stem cells promote tumor progression via inducing stroma remodeling on rabbit VX2 bladder tumor model. Int J Biol Sci. 14:1012–1021. 2018. View Article : Google Scholar : PubMed/NCBI

20 

Gao T, Yu Y, Cong Q, Wang Y, Sun M, Yao L, Xu C and Jiang W: Human mesenchymal stem cells in the tumour micro-environment promote ovarian cancer progression: The role of platelet-activating factor. BMC Cancer. 18:9992018. View Article : Google Scholar

21 

Shojaei S, Hashemi SM, Ghanbarian H, Salehi M and Mohammadi-Yeganeh S: Effect of mesenchymal stem cells-derived exosomes on tumor microenvironment: Tumor progression versus tumor suppression. J Cell Physiol. 234:3394–3409. 2019. View Article : Google Scholar

22 

Worner PM, Schachtele DJ, Barabadi Z, Srivastav S, Chandrasekar B, Izadpanah R and Alt EU: Breast tumor micro-environment can transform naive mesenchymal stem cells into tumor-forming cells in nude mice. Stem Cells Dev. 28:341–352. 2019. View Article : Google Scholar

23 

Shi Y, Du L, Lin L and Wang Y: Tumour-associated mesenchymal stem/stromal cells: Emerging therapeutic targets. Nat Rev Drug Discov. 16:35–52. 2017. View Article : Google Scholar

24 

Friedenstein AJ, Chailakhjan RK and Lalykina KS: The development of fibroblast colonies in monolayer cultures of guinea-pig bone marrow and spleen cells. Cell Tissue Kine. 3:393–403. 1970.

25 

Mushahary D, Spittler A, Kasper C, Weber V and Charwat V: Isolation, cultivation, and characterization of human mesen-chymal stem cells. Cytometry A. 93:19–31. 2018. View Article : Google Scholar

26 

Xu X, Zhang X, Wang S, Qian H, Zhu W, Cao H, Wang M, Chen Y and Xu W: Isolation and comparison of mesenchymal stem-like cells from human gastric cancer and adjacent non-cancerous tissues. J Cancer Res Clin. 137:495–504. 2011. View Article : Google Scholar

27 

Cao H, Xu W, Qian H, Zhu W, Yan Y, Zhou H, Zhang X and Xu X, Li J, Chen Z and Xu X: Mesenchymal stem cell-like cells derived from human gastric cancer tissues. Cancer Lett. 274:61–71. 2009. View Article : Google Scholar

28 

Sun L, Wang Q, Chen B, Zhao Y, Shen B, Wang X, Zhu M, Li Z, Zhao X, Xu C, et al: Human gastric cancer mesenchymal stem cell-derived IL15 contributes to tumor cell epithelial-mesenchymal transition via upregulation tregs ratio and PD-1 expression in CD4 + T cell. Stem Cells Dev. 27:1203–1214. 2018. View Article : Google Scholar : PubMed/NCBI

29 

Zhu Q, Zhang X, Zhang L, Li W, Wu H, Yuan X, Mao F, Wang M, Zhu W, Qian H and Xu W: The IL-6-STAT3 axis mediates a reciprocal crosstalk between cancer-derived mesenchymal stem cells and neutrophils to synergistically prompt gastric cancer progression. Cell Death Dis. 5:e12952014. View Article : Google Scholar : PubMed/NCBI

30 

Li W, Zhou Y, Yang J, Zhang X, Zhang H, Zhang T, Zhao S, Zheng P, Huo J and Wu H: Gastric cancer-derived mesenchymal stem cells prompt gastric cancer progression through secretion of interleukin-8. J Exp Clin Cancer Res. 34:522015. View Article : Google Scholar : PubMed/NCBI

31 

Cao M, Mao J, Duan X, Lu L, Zhang F, Lin B, Chen M, Zheng C, Zhang X and Shen J: In vivo tracking of the tropism of mesenchymal stem cells to malignant gliomas using reporter gene-based MR imaging. Int J Cancer. 142:1033–1046. 2018. View Article : Google Scholar

32 

Chen Y, He Y, Wang X, Lu F and Gao J: Adiposederived mesenchymal stem cells exhibit tumor tropism and promote tumorsphere formation of breast cancer cells. Oncol Rep. 41:2126–2136. 2019.PubMed/NCBI

33 

Chaturvedi P, Gilkes DM, Takano N and Semenza GL: Hypoxia-inducible factor-dependent signaling between triple-negative breast cancer cells and mesenchymal stem cells promotes macrophage recruitment. Proc Natl Acad Sci USA. 111:E2120–E2129. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Jung YH, Kim JK, Shiozawa Y, Wang J, Mishra A, Joseph J, Berry JE, McGee S, Lee E, Sun H, et al: Recruitment of mesenchymal stem cells into prostate tumours promotes metastasis. Nat Commun. 4:17952013. View Article : Google Scholar : PubMed/NCBI

35 

Camorani S, Hill BS, Fontanella R, Greco A, Gramanzini M, Auletta L, Gargiulo S, Albanese S, Lucarelli E, Cerchia L and Zannetti A: Inhibition of bone marrow-derived mesenchymal stem cells homing towards triple-negative breast cancer micro-environment using an Anti-PDGFRβ Aptamer. Theranostics. 7:3595–3607. 2017. View Article : Google Scholar :

36 

Barcellos-de-Souza P, Comito G, Pons-Segura C, Taddei ML, Gori V, Becherucci V, Bambi F, Margheri F, Laurenzana A, Del Rosso M and Chiarugi P: Mesenchymal stem cells are recruited and activated into carcinoma-associated fibroblasts by prostate cancer microenvironment-derived TGF-β1. Stem Cells. 34:2536–2547. 2016. View Article : Google Scholar : PubMed/NCBI

37 

Pavon LF, Sibov TT, de Souza AV, da Cruz EF, Malheiros SMF, Cabral FR, de Souza JG, Boufleur P, de Oliveira DM, de Toledo SRC, et al: Tropism of mesenchymal stem cell toward CD133+ stem cell of glioblastoma in vitro and promote tumor proliferation in vivo. Stem Cell Res Ther. 9:3102018. View Article : Google Scholar

38 

Berger L, Shamai Y, Skorecki KL and Tzukerman M: Tumor specific recruitment and reprogramming of mesenchymal stem cells in tumorigenesis. Stem Cells. 34:1011–1026. 2016. View Article : Google Scholar

39 

Shamai Y, Alperovich DC, Yakhini Z, Skorecki K and Tzukerman M: Reciprocal reprogramming of cancer cells and associated mesenchymal stem cells in gastric cancer. Stem Cells. 37:176–189. 2019. View Article : Google Scholar

40 

Shen Y, Xue C, Li X, Ba L, Gu J, Sun Z, Han Q and Zhao RC: Effects of gastric cancer cell-derived exosomes on the immune regulation of mesenchymal stem cells by the NF-κB signaling pathway. Stem Cells Dev. 28:464–476. 2019. View Article : Google Scholar : PubMed/NCBI

41 

Zhu M, Wang M, Yang F, Tian Y, Cai J, Yang H, Fu H, Mao F, Zhu W, Qian H and Xu W: miR-155-5p inhibition promotes the transition of bone marrow mesenchymal stem cells to gastric cancer tissue derived MSC-like cells via NF-κ B p65 activation. Oncotarget. 7:16567–16580. 2016. View Article : Google Scholar : PubMed/NCBI

42 

Ji RB, Zhang X, Qian H, Gu H, Sun Z, Mao F, Yan Y, Chen J, Liang Z and Xu W: miR-374 mediates the malignant transformation of gastric cancer-associated mesenchymal stem cells in an experimental rat model. Oncol Rep. 38:1473–1481. 2017. View Article : Google Scholar : PubMed/NCBI

43 

Sun Z, Chen J, Zhang J, Ji R, Xu W, Zhang X and Qian H: The role and mechanism of miR-374 regulating the malignant transformation of mesenchymal stem cells. Am J Transl Res. 10:3224–3232. 2018.PubMed/NCBI

44 

Yang T, Zhang X, Wang M, Zhang J, Huang F, Cai J, Zhang Q, Mao F, Zhu W, Qian H and Xu W: Activation of mesenchymal stem cells by macrophages prompts human gastric cancer growth through NF-κ B pathway. PLoS One. 9:e975692014. View Article : Google Scholar

45 

Xu R, Zhao X, Zhao Y, Chen B, Sun L, Xu C, Shen B, Wang M, Xu W and Zhu W: Enhanced gastric cancer growth potential of mesenchymal stem cells derived from gastric cancer tissues educated by CD4+ T cells. Cell Prolif. 51:e123992018. View Article : Google Scholar

46 

Zhang Q, Ding J, Liu J, Wang W, Zhang F, Wang J and Li Y: Helicobacter pylori-infected MSCs acquire a pro-inflammatory phenotype and induce human gastric cancer migration by promoting EMT in gastric cancer cells. Oncol Lett. 11:449–457. 2016. View Article : Google Scholar : PubMed/NCBI

47 

Vafaei R, Nassiri SM and Siavashi V: 3-Adrenergic regulation of EPC features through manipulation of the bone marrow MSC niche. J Cell Biochem. 118:4753–4761. 2017. View Article : Google Scholar : PubMed/NCBI

48 

Yang Y, Bucan V, Baehre H, Von der Ohe J, Otte A and Hass R: Acquisition of new tumor cell properties by MSC-derived exosomes. Int J Oncol. 47:244–252. 2015. View Article : Google Scholar : PubMed/NCBI

49 

Whiteside TL: Exosome and mesenchymal stem cell cross-talk in the tumor microenvironment. Semin Immunol. 35:69–79. 2018. View Article : Google Scholar : PubMed/NCBI

50 

Huang F, Wang M, Yang T, Cai J, Zhang Q, Sun Z, Wu X, Zhang X, Zhu W, Qian H and Xu W: Gastric cancer-derived MSC-secreted PDGF-DD promotes gastric cancer progression. J Cancer Res Clin. 140:1835–1848. 2014. View Article : Google Scholar

51 

Huang F, Yao Y, Wu J, Liu Q, Zhang J, Pu X, Zhang Q and Xia L: Curcumin inhibits gastric cancer-derived mesenchymal stem cells mediated angiogenesis by regulating NF-κB/VEGF signaling. Am J Transl Res. 9:5538–5547. 2017.

52 

Sun J, Song Y, Wang Z, Chen X, Gao P, Xu Y, Zhou B and Xu H: Clinical significance of palliative gastrectomy on the survival of patients with incurable advanced gastric cancer: A systematic review and meta-analysis. BMC Cancer. 13:5772013. View Article : Google Scholar : PubMed/NCBI

53 

Orditura M, Galizia G, Sforza V, Gambardella V, Fabozzi A, Laterza MM, Andreozzi F, Ventriglia J, Savastano B, Mabilia A, et al: Treatment of gastric cancer. World J Gastroenterol. 20:1635–1649. 2014. View Article : Google Scholar : PubMed/NCBI

54 

Kwee RM and Kwee TC: Role of imaging in predicting response to neoadjuvant chemotherapy in gastric cancer. World J Gastroenterol. 20:1650–1656. 2014. View Article : Google Scholar : PubMed/NCBI

55 

Shitara K: Chemotherapy for advanced gastric cancer: Future perspective in Japan. Gastric Cancer. 20:102–110. 2017. View Article : Google Scholar

56 

Roberto M, Romiti A, Onesti CE, Zullo A, Falcone R and Marchetti P: Evolving treatments for advanced gastric cancer: Appraisal of the survival trend. Expert Rev Anticancer Ther. 16:717–729. 2016. View Article : Google Scholar : PubMed/NCBI

57 

McMillin DW, Negri JM and Mitsiades CS: The role of tumour-stromal interactions in modifying drug response: Challenges and opportunities. Nat Rev Drug Discov. 12:217–228. 2013. View Article : Google Scholar : PubMed/NCBI

58 

Zeng J, Chen S, Li C, Ye Z, Lin B, Liang Y, Wang B, Ma Y, Chai X, Zhang X, et al: Mesenchymal stem/stromal cells-derived IL-6 promotes nasopharyngeal carcinoma growth and resistance to cisplatin via upregulating CD73 expression. J Cancer. 11:2068–2079. 2020. View Article : Google Scholar : PubMed/NCBI

59 

Le Naour A, Prat M, Thibault B, Mével R, Lemaitre L, Leray H, Joubert MV, Coulson K, Golzio M, Lefevre L, et al: Tumor cells educate mesenchymal stromal cells to release chemoprotective and immunomodulatory factors. J Mol Cell Biol. 12:202–215. 2020. View Article : Google Scholar :

60 

Pasquier J, Gosset M, Geyl C, Hoarau-Véchot J, Chevrot A, Pocard M, Mirshahi M, Lis R, Rafii A and Touboul C: CCL2/CCL5 secreted by the stroma induce IL-6/PYK2 depen-dent chemoresistance in ovarian cancer. Mol Cancer. 17:472018. View Article : Google Scholar

61 

Wang X: Stem cells in tissues, organoids, and cancers. Cell Mol Life Sci. 76:4043–4070. 2019. View Article : Google Scholar : PubMed/NCBI

62 

Polejaeva IA, Chen SH, Vaught TD, Page RL, Mullins J, Ball S, Dai Y, Boone J, Walker S, Ayares DL, et al: Cloned pigs produced by nuclear transfer from adult somatic cells. Nature. 407:86–90. 2000. View Article : Google Scholar : PubMed/NCBI

63 

Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K and Yamanaka S: Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 131:861–872. 2007. View Article : Google Scholar : PubMed/NCBI

64 

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. 38:4637–4654. 2019. View Article : Google Scholar : PubMed/NCBI

65 

Ji R, Zhang B, Zhang X, Xue J, Yuan X, Yan Y, Wang M, Zhu W, Qian H and Xu W: Exosomes derived from human mesenchymal stem cells confer drug resistance in gastric cancer. Cell Cycle. 14:2473–2483. 2015. View Article : Google Scholar : PubMed/NCBI

66 

Chaudhary D, Trivedi RN, Kathuria A, Goswami TK, Khandia R and Munjal A: In vitro and in vivo immunomodulating properties of mesenchymal stem cells. Recent Pat Inflamm Allergy Drug Discov. 12:59–68. 2018. View Article : Google Scholar : PubMed/NCBI

67 

Su CY, Fu XL, Duan W, Yu PW and Zhao YL: High density of CD68+ tumor-associated macrophages predicts a poor prognosis in gastric cancer mediated by IL-6 expression. Oncol Lett. 15:6217–6224. 2018.PubMed/NCBI

68 

Li W, Zhang X, Wu F, Zhou Y, Bao Z, Li H, Zheng P and Zhao S: Gastric cancer-derived mesenchymal stromal cells trigger M2 macrophage polarization that promotes metastasis and EMT in gastric cancer. Cell Death Dis. 10:9182019. View Article : Google Scholar : PubMed/NCBI

69 

Wang M, Chen B, Sun XX, Zhao XD, Zhao YY, Sun L, Xu CG, Shen B, Su ZL, Xu WR and Zhu W: Gastric cancer tissue-derived mesenchymal stem cells impact peripheral blood mononuclear cells via disruption of Treg/Th17 balance to promote gastric cancer progression. Exp Cell Res. 361:19–29. 2017. View Article : Google Scholar : PubMed/NCBI

70 

Schmid P, Cortes J, Pusztai L, McArthur H, Kümmel S, Bergh J, Denkert C, Park YH, Hui R, Harbeck N, et al: Pembrolizumab for early triple-negative breast cancer. N Engl J Med. 382:810–821. 2020. View Article : Google Scholar : PubMed/NCBI

71 

Burtness B, Harrington KJ, Greil R, Soulières D, Tahara M, de Castro G Jr, Psyrri A, Basté N, Neupane P, Bratland Å, et al: Pembrolizumab alone or with chemotherapy versus cetuximab with chemotherapy for recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-048): A randomised, open-label, phase 3 study. Lancet. 394:1915–1928. 2019. View Article : Google Scholar : PubMed/NCBI

72 

Kamath SD, Kalyan A and Benson A: Pembrolizumab for the treatment of gastric cancer. Expert Rev Anticancer Ther. 18:1177–1187. 2018. View Article : Google Scholar : PubMed/NCBI

73 

Bang YJ, Kang YK, Catenacci DV, Muro K, Fuchs CS, Geva R, Hara H, Golan T, Garrido M, Jalal SI, et al: Pembrolizumab alone or in combination with chemotherapy as first-line therapy for patients with advanced gastric or gastroesophageal junction adenocarcinoma: Results from the phase II nonrandomized KEYNOTE-059 study. Gastric Cancer. 22:828–837. 2019. View Article : Google Scholar : PubMed/NCBI

74 

Fukuoka S, Hara H, Takahashi N, Kojima T, Kawazoe A, Asayama M, Yoshii T, Kotani D, Tamura H, Mikamoto Y, et al: Regorafenib plus nivolumab in patients with advanced gastric or colorectal cancer: An open-label, dose-escalation, and dose-expansion phase Ib trial (REGONIVO, EPOC1603). J Clin Oncol. 38:2053–2061. 2020. View Article : Google Scholar : PubMed/NCBI

75 

Sun L, Wang Q, Chen B, Zhao Y, Shen B, Wang H, Xu J, Zhu M, Zhao X, Xu C, et al: Gastric cancer mesenchymal stem cells derived IL-8 induces PD-L1 expression in gastric cancer cells via STAT3/mTOR-c-Myc signal axis. Cell Death Dis. 9:9282018. View Article : Google Scholar : PubMed/NCBI

76 

Song Y, Du H, Dai C, Zhang L, Li S, Hunter DJ, Lu L and Bao C: Human adipose-derived mesenchymal stem cells for osteoarthritis: A pilot study with long-term follow-up and repeated injections. Regen Med. 13:295–307. 2018. View Article : Google Scholar : PubMed/NCBI

77 

Ahn SY, Chang YS, Sung SI and Park WS: Mesenchymal stem cells for severe intraventricular hemorrhage in preterm infants: Phase I dose-escalation clinical trial. Stem Cells Transl Med. 7:847–856. 2018. View Article : Google Scholar : PubMed/NCBI

78 

Lin W, Huang L, Li Y, Fang B, Li G, Chen L and Xu L: Mesenchymal stem cells and cancer: Clinical challenges and opportunities. Biomed Res Int. 2019:28208532019. View Article : Google Scholar : PubMed/NCBI

79 

Pakravan K, Babashah S, Sadeghizadeh M, Mowla SJ, Mossahebi-Mohammadi M, Ataei F, Dana N and Javan M: MicroRNA-100 shuttled by mesenchymal stem cell-derived exosomes suppresses in vitro angiogenesis through modulating the mTOR/HIF-1α/VEGF signaling axis in breast cancer cells. Cell Oncol (Dordr). 40:457–470. 2017. View Article : Google Scholar

80 

Zhang J, Hou L, Wu X, Zhao D, Wang Z, Hu H, Fu Y and He J: Inhibitory effect of genetically engineered mesenchymal stem cells with Apoptin on hepatoma cells in vitro and in vivo. Mol Cell Biochem. 416:193–203. 2016. View Article : Google Scholar : PubMed/NCBI

81 

Shojaati G, Khandaker I, Funderburgh ML, Mann MM, Basu R, Stolz DB, Geary ML, Dos Santos A, Deng SX and Funderburgh JL: Mesenchymal stem cells reduce corneal fibrosis and inflammation via extracellular vesicle-mediated delivery of miRNA. Stem Cells Transl Med. 8:1192–1201. 2019. View Article : Google Scholar : PubMed/NCBI

82 

Wang M, Zhao C, Shi H, Zhang B, Zhang L, Zhang X, Wang S, Wu X, Yang T, Huang F, et al: Deregulated microRNAs in gastric cancer tissue-derived mesenchymal stem cells: Novel biomarkers and a mechanism for gastric cancer. Br J Cancer. 110:1199–1210. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2021
Volume 47 Issue 2

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Shen J and Shen J: Research advances in the role of gastric cancer‑derived mesenchymal stem cells in tumor progression (Review). Int J Mol Med 47: 455-462, 2021
APA
Shen, J., & Shen, J. (2021). Research advances in the role of gastric cancer‑derived mesenchymal stem cells in tumor progression (Review). International Journal of Molecular Medicine, 47, 455-462. https://doi.org/10.3892/ijmm.2020.4810
MLA
Shen, J., Zhu, W."Research advances in the role of gastric cancer‑derived mesenchymal stem cells in tumor progression (Review)". International Journal of Molecular Medicine 47.2 (2021): 455-462.
Chicago
Shen, J., Zhu, W."Research advances in the role of gastric cancer‑derived mesenchymal stem cells in tumor progression (Review)". International Journal of Molecular Medicine 47, no. 2 (2021): 455-462. https://doi.org/10.3892/ijmm.2020.4810