Open Access

Exosome as a crucial communicator between tumor microenvironment and gastric cancer (Review)

  • Authors:
    • Menghui Wang
    • Hongxin Shu
    • Xifu Cheng
    • Hong Xiao
    • Zhenhua Jin
    • Nan Yao
    • Shengxun Mao
    • Zhen Zong
  • View Affiliations

  • Published online on: January 19, 2024     https://doi.org/10.3892/ijo.2024.5616
  • Article Number: 28
  • Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Gastric cancer is one of the most common malignancies and has relatively high morbidity and mortality rates. Exosomes are nanoscale extracellular vesicles that originate from a diverse array of cells and may be found throughout various bodily fluids. These vesicles are endogenous nanocarriers in their natural state with the unique ability to transport lipids, proteins, DNA and RNA. Exosomes contain DNA, RNA, proteins, lipids and other bioactive components that have crucial roles in the transmission of information and regulation of cell activities in gastric cancer. This paper begins with an exploration of the composition, formation and release mechanisms of exosomes. Subsequently, the role of exosomes in the tumor microenvironment is reviewed in terms of the immune cell population, nonimmune cell population and other factors. Finally, the current status and challenges of exosome‑based research on the progression, diagnosis and therapeutic methods of gastric cancer are summarized. This holistic review offers insight that may guide future research directions for exosomes and potentially pave the way for novel therapeutic interventions in the management of gastric cancer.

1. Introduction

Gastric cancer poses a significant threat to human health and well-being and results in extensive mortality worldwide (1). Characterized by its malignant properties, gastric cancer is the fifth most common malignancy and fourth leading cause of cancer-related death globally (2). Prevalence rates of gastric cancer are higher in East Asian and Eastern European countries compared to North American and Northern European regions, which is possibly due to variations in dietary patterns and lifestyle choices among local populations (3,4). The primary risk factor associated with gastric cancer is Helicobacter pylori (H. pylori) infection, which is widely recognized as the main causative agent (5). H. pylori infection may accelerate the progression of intestinal metaplasia in the stomach and results in gastric inflammation, and eventually, the development of gastric cancer (6). However, other environmental factors, such as tobacco smoking, alcohol consumption and high consumption of grilled foods, may increase the risk of developing this disease (7). Given the seriousness of this clinical condition, additional research into the aetiology and pathogenesis of gastric cancer is urgently needed. Furthermore, efforts must be made to develop appropriate diagnostic approaches and therapeutic measures to combat this condition.

Exosomes, which were discovered in sheep reticulocytes by Trams et al (8) in the previous century, have demonstrated great therapeutic promise in a variety of clinical conditions (9). These subcellular, membrane-bound nanovesicles are characterized by their small size, which ranges from 30 to 200 nm in diameter, and their distinctive single-layered membrane structure (10). Exosomes contain a wide range of biologically active components, including proteins, lipids and nucleic acids (11). These vesicles are increasingly recognized as critical mediators of cell-cell communication and influence a wide range of biological processes, such as cancer growth, immune system responses and inflammatory reactions (12). Of note, exosomes have a pivotal role in the interface between cancer cells and the tumor microenvironment (TME), which is a critical interplay that is central to cancer genesis and progression (13). Exosome-secreted compounds have also been shown to have diagnostic value, particularly as biomarkers for early-stage gastric cancer (14,15). Understanding mechanisms that drive the initiation and progression of gastric cancer and the identification of additional biological markers for early detection is crucial to the advancement of exosome-based therapeutic methods for gastric cancer. In this comprehensive review, a detailed and systematic examination of the components of exosomes and their intricate interactions with gastric cancer within the TME setting is provided. This includes discussions about gastric cancer progression, diagnosis and therapeutic methods.

2. Composition, formation and release of exosomes

Exosomes are nanosized membrane vesicles that are released into the extracellular environment and contain a diverse mixture of essential biomolecules including proteins, lipids, functional RNA species and other bioactive substances that perform crucial cellular functions (16,17). Exosomes exhibit significant heterogeneity and their molecular composition varies based on the cell type and the fluidic microenvironment (18). Proteins are a crucial component of exosomes and their profile differs based on the cell type and extracellular fluid conditions (19). Exosomal proteins regulate cellular processes in tumor cells and thus promote the metastatic cascade (20,21). Lipids, which consist of cholesterol, phosphatidylserine and sphingolipids, are essential constituents of exosomes and have a critical role in their release. These proteins serve as anchor sites for membrane proteins and protect proteins from protease-related damage (22). Exosomes are membrane-bound vesicles that originate from endocytic pathways and have a crucial role in intercellular communication. These extracellular vesicles encapsulate various RNA molecules, including microRNAs (miRNAs), mRNAs and long noncoding RNAs (lncRNAs), which may be transferred between cells to influence cellular behaviour and signalling pathways (23). These RNAs modulate gene expression and thereby impact various biological processes, such as cellular proliferation, differentiation and apoptosis (24). Furthermore, the multifunctional nucleic acid molecules that are miRNAs indubitably have a fundamental role in the pathogenesis of cancer, particularly in its progression and metastatic spread across diverse cancer types (25,26). In summary, exosomes exhibit significant heterogeneity, which suggests that their compositional profiles are viable biomarkers for both the identification and amelioration of various malignancies. Therefore, exosomes and their molecular components, including proteins, lipids and RNA species, are highly important in cancer research and may be the key to developing novel therapeutic approaches for cancer management.

The generation of exosomes has been extensively studied (27). This process begins with the invagination of the cell membrane, which leads to the formation of early endosomes that are enriched with various bioactive components derived from the extracellular environment. These endosomes then undergo a maturation process during which they transform into late endosomes (28). These organelles may undergo a fate determination process in which they can either be destined for lysosomal degradation or progress towards the formation of multivesicular bodies (MVBs) through the inwards budding of their limiting membranes. This bifurcation outcome is tightly controlled and represents a fundamental cellular mechanism for membrane trafficking and protein sorting (29,30). In different cellular contexts, MVBs can have distinct fates. Specifically, MVBs may undergo exocytosis via fusion with the plasma membrane, where they release intraluminal vesicles as exosomes, or they can be targeted for degradation by lysosomes or recycled through the trans-Golgi network (Fig. 1). Of note, mechanisms underlying exosome biogenesis can vary across different cellular and organismal contexts (31). A deeper understanding of mechanisms driving exosome biogenesis may lead to critical insight into the biological functions of these extracellular vesicles and help guide the development of effective therapeutic strategies. By unravelling the complexities of this process, a more nuanced appreciation of the role that exosomes have in health and disease and how their potential may be harnessed for therapeutic gain may be acquired.

3. Role of exosomes in the TME

The TME has a pivotal role in tumor metabolism, development, progression and therapeutic response. The tumorigenic potential of cancer cells is significantly influenced by alterations in the TME (32). This dynamic interplay may be likened to the correlation between the quality of soil and growth of seeds, where enriched soil fertility facilitates rapid and robust seedling development (33). The composition of the TME varies depending on the type of tumor but hallmark features include immune cells, nonimmune cells and the extracellular matrix (34). Both immune and nonimmune cell populations and other factors influence the role of exosomes within the TME. As shown in Fig. 2, exosomes released by immune cells and nonimmune cells are present in the TME and have an important role in regulating intercellular communication by transmitting signals. In the chapters below, the impacts of immune and nonimmune cell populations and other factors were summarized to shed light on the crucial role of exosomes within the TME. This provides novel insight and perspectives for further understanding the significant contributions of these cells to the treatment of tumors.

From the immune cell population perspective

The role of the immune cell population in the occurrence and progression of cancer is paramount, and this population holds promise as a novel approach for cancer therapeutics (35,36). The discovery of this immune cell population has propelled the development of immunotherapy and opened avenues for treating a subset of cancers (37). For instance, strategies aimed at reversing the suppressive immune milieu and augmenting effector T-cell function may potentially increase the success rate of immunotherapy for cervical cancer (38,39). However, immunotherapy regimens do not uniformly yield positive outcomes in all cancer treatments (40). Consequently, unravelling the elusive molecular mechanisms underlying cancer-related immune responses has emerged as an imperative objective.

Exosomes have been shown to have a significant role in the polarization of macrophages towards the M1 and M2 phenotypes (41). M1 macrophages are proinflammatory and may promote inflammation and swallow extraneous substances to provide relative protection to tissues and organs (41,42). M2 macrophages are anti-inflammatory agents that may suppress the activity of M1 macrophages through the secretion of anti-inflammatory factors and have essential effects on processes such as wound healing and tissue repair (41-43). Pritchard et al (44) demonstrated that lung tumor-derived exosomes promote M2 macrophage polarization and are a novel therapeutic target for immunotherapy in lung cancer. Tumor cells can recruit, aggregate and induce macrophages to transform into tumor-associated macrophages and facilitate tumor development within the TME (45-47). Previous studies revealed that exosome-borne Epstein-Barr virus-encoded small RNA-1 induces indoleamine 2,3-dioxygenase expression in tumor-infiltrating macrophages of oral squamous-cell carcinomas and suppresses T-cell activity by activating the retinoic acid-inducible gene I/IL-6/TNF-α pathway (48). In addition, the intracellular signalling mechanisms involved in exosome uptake and the regulatory payload of the TME in macrophages have yet to be fully elucidated. Dendritic cells constitute a vital component of the TME. Previous studies have shown that tumor-derived exosomes may impinge on dendritic cells through the heat shock protein (HSP)72/HSP105-Toll-like receptor (TLR)2/TLR4 pathway and thereby promote tumor metastasis and development (49). Exosomes can stimulate both the innate and adaptive immune systems by activating dendritic cells, natural killer (NK) cells and T cells. This activation enables these immune cells to exert antitumor effects, particularly in ovarian cancer (50). Tumor-associated neutrophils mediate protumor immunosuppressive activity and their infiltration into tumors is associated with adverse outcomes in a wide array of malignant diseases (51). Of note, tumor-derived exosomes can, to a certain extent, induce N2 polarization of neutrophils and promote the migration of gastric cancer cells, leading to further deterioration of gastric cancer (52). Multiple myeloma-derived exosomes have been shown to promote apoptosis and inhibit T-lymphocyte proliferation, which highlights their significant role in regulating immune cell function (53). The innate ability of NK cells to recognize and eliminate target cells renders them the first line of defence in identifying and eliminating infected or transformed cells (54). Furthermore, there is growing evidence demonstrating crosstalk between exosomal biogenesis and autophagy pathways with autophagy-mediated exosomes acting as immunomodulators of NK cells in the pancreatic cancer microenvironment. This crosstalk pathway holds promising potential in advancing the development of pancreatic cancer immunotherapy (55). In addition, NK cell-derived exosomes have been shown to enhance antitumor effects in ovarian cancer by delivering cisplatin and reactivating NK-cell function, which underscores their therapeutic relevance (56). Of note, polymorphonuclear myelopoietic suppressor cells are prevalent in the peripheral blood of cancer patients and these cells can inhibit the antitumor activity of NK cells, highlighting the complex interplay between different immune cell populations in cancer progression (57). The interaction between exosomes derived from immune cells and cancer cells is illustrated in Fig. 3.

From the perspective of nonimmune cell populations

In addition to immune cell populations, the TME is populated by diverse and significant groups of nonimmune cells. These nonimmune cells in the TME include cancer-associated fibroblasts (CAFs) and endothelial cells, among others, which have pivotal roles in regulating tumor progression, metastasis and sensitivity to anticancer drugs. CAFs influence the occurrence and development of gastric cancer by controlling individual cytokine profiles and direct cell-cell interactions (58). Previous studies demonstrated that hepatocyte growth factor (HGF), which is derived from CAFs, promotes angiogenesis in gastric cancer through the PI3K/AKT and ERK1/2 signalling pathways (59). CAFs can foster chemotherapy resistance in gastric cancer by secreting IL-11 to target the JAK/STAT3/Bcl2 signalling pathway (60). A study conducted by Liu et al (61) confirmed that immunosuppressive microfibril associated protein 2 CAFs can lead to adverse outcomes and therapeutic resistance in gastric cancer. Furthermore, recent studies have revealed that CAF-derived slit homolog 2 protein can promote metastasis of gastric cancer cells by activating NIMA-related kinase 9, which leads to the exacerbation of gastric cancer (62). The exosome glucose-regulated protein 78 derived from gastric cancer cells stimulates the secretion of cytokines by vascular endothelial cells to induce angiogenesis (63). In gastric cancer exosome metastasis, Y-box binding protein 1 can also promote angiogenesis through increased expression of angiogenic factors in vascular endothelial cells (64). Exosomes carrying miR-155 can promote angiogenesis in gastric cancer by targeting the forkhead box O3 protein in endothelial cells (65). Endothelial cells not only supply essential nutrients to tumor cells but also engage in cross-talk with other cells through the secretion of cytokines. Deciphering these complex interactions within the TME may pave the way for the identification of novel therapeutic targets in the treatment of gastric cancer. In general, nonimmune cell populations have an equally critical and irreplaceable role in the TME.

The other factors' perspective

In addition to the pivotal role of exosomes in cell populations, other factors within the TME, such as metabolic processes, inflammatory responses and gene expression, also influence the function of exosomes (66). Tumor cells exhibit metabolic profiles distinct from those of normal cells and require an abundance of energy to support their rapid growth and dissemination. This distinct metabolic pattern can influence the function of exosomes. Previous studies demonstrated that certain tumor cells release substantial amounts of lactic acid, which results in a correspondingly acidic environment that impacts the stability of exosomes and the activity of the bioactive molecules they carry (67). An emerging study revealed that microbial proteins encoded by exosomal IncRNA aldo-keto reductase family 1 member C2 can facilitate lymph node metastasis in gastric cancer by modulating fatty acid metabolism (68). Exosomes have been implicated in lipid metabolism and the regulation of multiple processes in gastric cancer, including communication between gastric cancer cells, the establishment of the TME and the biological behaviour of gastric cancer, such as metastasis, invasion and chemotherapy resistance. Inflammation is a pivotal initiating step in the formation of the TME and is intricately linked to the recruitment of various immune cells and activation of epithelial stem cells or direct progenitors, which serve as the primary origin of gastrointestinal cancers (69). Therefore, inflammation has an important role in the complex interplay of events within the TME and warrants further exploration in the context of gastric cancer development and progression. Furthermore, gene expression patterns are frequently altered, which can influence the function of exosomes in tumors (70). The incorrect expression of certain key genes can affect the communication between exosomes and tumor cells, which contributes to the development and progression of tumor cells.

4. Progression, diagnosis and therapeutic methods for gastric cancer treatment based on exosomes

The relationship between gastric cancer cells and the TME can be likened to that between a seed and the soil. In this context, exosomes have emerged as important factors that facilitate the development of metastatic gastric cancer by serving as crucial molecular agents of intercellular communication (71). Through their ability to transfer proteins and other related substances between cells via tissue fluid, exosomes are capable of regulating various phenotypic changes within gastric cancer cells while also inducing metastasis of distant tumor cells (72,73). Of note, exosomes are also capable of modulating the TME by regulating diverse physiological processes, such as immunity, angiogenesis and metastasis, and thereby promote the progression of gastric cancer (74,75). Based on the above research, it was found that exosomes have critical roles in the development of novel diagnostic and therapeutic strategies for gastric cancer (Fig. 4).

The progression of gastric cancer based on exosomes

Exosomes have emerged as crucial players in the carcinogenesis and pathogenesis of gastric cancer and are intricately involved in multiple tumorigenic events (76,77). Exosomes are involved in the antiapoptotic, proliferative, metastatic, autophagic and angiogenic processes of gastric cancer cells and are an important means of intercellular communication between tumor cells and the surrounding microenvironment (78). Previous studies demonstrated that exosomes derived from gastric cancer cells can impede the processes of senescence and apoptosis in primary normal gastric epithelial cells. This suggests that gastric cancer exosomes influence the development of field cancer within surrounding gastric epithelial cells (79). Exosomes originating from gastric cancer cells were found to elicit neutrophil-mediated autophagy and facilitate activation of the same cancer cell type, predominantly via the high mobility group box 1 protein/TLR4/NF-κB signalling pathway (52). Liang et al (80) reported that the circ670 molecule, which is present in exosomes, has an influential role in the pathogenesis of tobacco-induced gastric cancer. Chang et al (81) demonstrated that the ectopic expression of miR-1228 promoted the emergence of exosomes, which in turn led to a discernible attenuation of recombinant matrix metalloproteinase (MMP)-14 expression. As a consequence, the incidence and progression of gastric cancer is thwarted. The present study revealed that decreased expression of miR-3184-5p within exosomes from individuals diagnosed with gastric cancer may potentially augment the overall proliferative capacity of these malignant cells through the modulation of distinct molecular signalling pathways, namely, the AKT, STAT3 and inositol-requiring enzyme 1 pathways (82). Exosomes have emerged as a promising modality for promoting the development of gastric cancer cells by serving as transport vehicles. Specifically, exosomes ferry the lncRNA prostate cancer gene expression marker 1 to support gastric cancer invasion and metastasis via stabilization of SNAI1. This fascinating mechanism underscores the crucial role that exosomes play as functional vectors in orchestrating pathogenic progression in gastric cancer (83). Exosomes derived from gastric cancer cells can deliver miR-130a to vascular cells. This transfer is mediated by targeting the c-MYB gene and ultimately promoting angiogenesis and enhancing tumor growth (84). Extracellular vesicles, specifically exosomes originating from mesenchymal stem cells (MSCs), promote the proliferation and metastasis of gastric carcinoma cells through the promotion of AKT pathway activation (85). These findings align with the current research and demonstrate that exosomes derived from gastric cancer cells stimulate programmed cell death 1 ligand 1 expression in MSCs via the AKT/C-Myc signalling axis and promote the metastasis of gastric cancer cells (86). Exosomes have a pivotal role in the pathogenesis of gastric cancer. Despite numerous strides in elucidating the functional relevance of these genes, several aspects of these genes remain poorly understood and necessitate further investigation.

Diagnosis of gastric cancer based on exosomes

Numerous studies on exosomes have shown their potential as biomarkers for various types of cancer. In particular, the concentration of exosomes has emerged as a promising diagnostic indicator for gastric cancer. Xia et al (15) demonstrated that patients with gastric cancer exhibit greater specificity and sensitivity in terms of exosome expression and reached 65.2 and 73.1%, respectively, compared to healthy individuals. This was achieved through characterization of exosome morphology via transmission electron microscopy and the detection of exosome size and concentration through nanoparticle tracking analysis (15). Furthermore, exosomal tripartite motif-containing 3 protein has potential as a diagnostic biomarker for gastric cancer and has paved the way for a novel approach to the management of this malignancy (87). In another study conducted by Sun et al (88) confirmed that inter-alpha-trypsin inhibitor heavy chain 4 was identified as a crucial serum biomarker for the detection of early gastric cancer (EGC) in patients. Specifically, mass spectrometry has validated its value as a highly valuable diagnostic tool for EGC detection compared to that of a healthy control group (88,89). Another investigation of patients with gastric cancer suggests that the serum exosomal membrane type 1-MMP mRNA and the serum exosomal lncRNA pcsk2-2:1 has pivotal roles in the progression of this disease. These findings underscore the potential of targeting serum exosomes in a pancancer liquid biopsy to uncover specific diagnostic biomarkers for gastric cancer (90,91). Although these studies have unveiled the potential significance of exosomes in gastric cancer diagnosis, further elucidation is required regarding the exact threshold and practical scope of application of these markers. Given the inherent heterogeneity of gastric cancer, it may be imperative to employ a combination of multiple markers for enhanced diagnostic accuracy and reliability. It is worth mentioning that the precise detection of exosomes was found to be highly important for the early identification of gastric cancer. However, achieving this precision in exosome detection remains a challenging task and demands extensive investigative efforts. Research indicates that exosomes derived from MSCs possess immense potential for mitigating disease pathologies and enhancing cognitive function in patients with conditions such as Alzheimer's disease, Parkinson's disease and vascular dementia (92). Prospective studies must further explore the concentration of exosomes, establish a specific diagnostic threshold and define the applicable range of pertinent markers to facilitate a more precise foundation for clinical diagnosis. Concurrently, a surge in clinical trials is imperative to validate these findings and propel the practical use of exosomes in gastric cancer diagnosis. It is envisaged that continued research and scientific scrutiny will pave the way for the effective utilization of exosomes as diagnostic biomarkers and revolutionize the management of gastric cancer and other malignancies.

Therapeutic methods for gastric cancer based on exosomes

Exosomes are increasingly assumed to play a critical role in public health medicine and exhibit potential applications for treating a wide range of diseases including cancer and central nervous system disorders (93,94). Due to their low immunogenicity and efficient ability to cross the blood-brain barrier, exosomes have the potential to become crucial players in the management of ischaemic stroke (95). However, this investigation is limited by a lack of convincing evidence regarding the safety of exosome therapy. Of note, exosomal polyphosphates can be stimulated through the mediation of exosomes derived from cancer cells, which elicit the activation of factor XII (96). Growing evidence suggests that exosomes may hold significant potential in the development of effective therapeutics for gastric cancer. Specifically, exosomes have been demonstrated to hinder tumor cell growth and angiogenesis through the transport of HGF small inhibitory RNA (97). Furthermore, exosomes serve as ideal nanoparticles for transporting miR-214 and effectively reversing chemical resistance in gastric cancer and thus represent a potentially selective treatment option for cisplatin-resistant gastric cancer (98). However, despite recent progress, numerous unknowns surround the use of exosomes in clinical practice. Significant improvements and challenges must be addressed before exosomes can become a viable therapeutic tool. The field of exosome-based medicine is in its infancy and further research is needed to determine its full potential.

5. Conclusions

Exosomes serve as crucial transporters in promoting the interaction between gastric cancer cells and the TME. These nanoscale extracellular vesicles possess several potential advantages, including biocompatibility, stability and intrinsic targeting capabilities, which render them promising therapeutic options for gastric cancer. However, the application of exosomes as drug delivery systems is still in its infancy and numerous challenges need to be addressed. Research geared towards clinical translation should strive to improve the yield, purity, targeting efficiency and biological activity of exosomes. Overall, it may take time for clinical success to be achieved regarding exosomes, but with the wealth of research and clinical trials underway, it is expected that exosome-related applications will soon benefit a majority of gastric cancer patients.

Availability of data and materials

Not applicable.

Authors' contributions

ZZ, SM, SH and MW conceived and designed the approach. MW, SH, XC and HX wrote and edited the manuscript. MW, SH, XC, ZJ and NY generated the figures. All authors have read and approved the final manuscript. Data authentication is not applicable.

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

All figures were created with Figdraw.

Funding

This research was funded by the National Natural Science Foundation of China (grant nos. 82060453 and 82103645 and 82260596); the Training Plan for Academic and Technical Young Leaders of Major Disciplines in Jiangxi Province (grant no. 20204BCJ23021), the Science and Technology Program of Jiangxi Provincial Health and Family Planning Commission (grant no. 202410246) and the China Postdoctoral Science Foundation (grant no. 2023M741523).

References

1 

Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011.

2 

Siegel RL, Miller KD, Fuchs HE and Jemal A: Cancer Statistics, 2021. CA Cancer J Clin. 71:7–33. 2021.

3 

Estevez-Ordonez D, Montalvan-Sanchez EE, Wong RE, Montalvan-Sanchez DM, Rodriguez-Murillo AA, Dominguez RL and Morgan DR: Health barriers and patterns of gastric cancer care in rural central American resource-limited settings. JAMA Oncol. 4:1131–1133. 2018.

4 

Yeoh KG and Tan P: Mapping the genomic diaspora of gastric cancer. Nat Rev Cancer. 22:71–84. 2022.

5 

Walker MM, Teare L and McNulty C: Gastric cancer and Helicobacter pylori: The bug, the host or the environment? Postgrad Med J. 84:169–170. 2008.

6 

Zeyaullah M, AlShahrani AM and Ahmad I: Association of helicobacter pylori infection and host cytokine gene polymorphism with gastric cancer. Can J Gastroenterol Hepatol. 2021:88106202021.

7 

Zeng Y and Jin RU: Molecular pathogenesis, targeted therapies, and future perspectives for gastric cancer. Semin Cancer Biol. 86:566–582. 2021.

8 

Trams EG, Lauter CJ, Salem N and Heine U: Exfoliation of membrane ecto-enzymes in the form of micro-vesicles. Biochim Biophys Acta. 645:63–70. 1981.

9 

Li Y, Tian L, Zhao T and Zhang J: A nanotherapeutic system for gastric cancer suppression by synergistic chemotherapy and immunotherapy based on iPSCs and DCs exosomes. Cancer Immunol Immunother. 72:1673–1683. 2023.

10 

Chen BY, Sung CWH, Chen C, Cheng CM, Lin DP, Huang CT and Hsu MY: Advances in exosomes technology. Clin Chim Acta. 493:14–19. 2019.

11 

Beit-Yannai E, Tabak S and Stamer WD: Exosomes. Nat Biotechnol. 38:11502020.

12 

Ayyar KK and Moss AC: Exosomes in intestinal inflammation. Front Pharmacol. 12:6585052021.

13 

Beit-Yannai E, Tabak S and Stamer WD: Physical exosome: Exosome interactions. J Cell Mol Med. 22:2001–2006. 2018.

14 

Wang J, Liu Y, Sun W, Zhang Q, Gu T and Li G: Plasma exosomes as novel biomarker for the early diagnosis of gastric cancer. Cancer Biomark. 21:805–812. 2018.

15 

Xia Y, Hu X, Di K, Liu C, Tan T, Lin Y, Xu H, Xie H, Wang S, Yang Z, et al: Combined detection of exosome concentration and tumor markers in gastric cancer. J Biomed Nanotechnol. 16:252–258. 2020.

16 

Exosomes. Nat Biotechnol. 38:11502020.

17 

Li J, Zhang Y and Luo B: Effects of exosomal viral components on the tumor microenvironment. Cancers (Basel). 14:35522022.

18 

Pegtel DM and Gould SJ: Exosomes. Annu Rev Biochem. 88:487–514. 2019.

19 

Wang X, Huang J, Chen W, Li G, Li Z and Lei J: The updated role of exosomal proteins in the diagnosis, prognosis, and treatment of cancer. Exp Mol Med. 54:1390–1400. 2022.

20 

Wang M, Zhao X, Huang F, Wang L, Huang J, Gong Z and Yu W: Exosomal proteins: Key players mediating pre-metastatic niche formation and clinical implications (Review). Int J Oncol. 58:42021.

21 

Huda MN and Nurunnabi M: Potential application of exosomes in vaccine development and delivery. Pharm Res. 39:2635–2671. 2022.

22 

Wang W, Zhu N, Yan T, Shi YN, Chen J, Zhang CJ, Xie XJ, Liao DF and Qin L: The crosstalk: Exosomes and lipid metabolism. Cell Commun Signal. 18:1192020.

23 

Puno MR, Weick E-M, Das M and Lima CD: SnapShot: The RNA exosome. Cell. 179:282–282.e1. 2019.

24 

Kilchert C, Wittmann S and Vasiljeva L: The regulation and functions of the nuclear RNA exosome complex. Nat Rev Mol Cell Biol. 17:227–239. 2016.

25 

Bach DH, Hong JY, Park HJ and Lee SK: The role of exosomes and miRNAs in drug-resistance of cancer cells. Int J Cancer. 141:220–230. 2017.

26 

Wu Z, Fang ZX, Hou YY, Wu BX, Deng Y, Wu HT and Liu J: Exosomes in metastasis of colorectal cancers: Friends or foes? World J Gastrointest Oncol. 15:731–756. 2023.

27 

Patil AA and Rhee WJ: Exosomes: Biogenesis, composition, functions, and their role in pre-metastatic niche formation. Biotechnol Bioprocess Engineering. 24:689–701. 2019.

28 

Kanemoto S, Nitani R, Murakami T, Kaneko M, Asada R, Matsuhisa K, Saito A and Imaizumi K: Multivesicular body formation enhancement and exosome release during endoplasmic reticulum stress. Biochem Biophys Res Commun. 480:166–172. 2016.

29 

Xia Z, Qing B, Wang W, Gu L, Chen H and Yuan Y: Formation, contents, functions of exosomes and their potential in lung cancer diagnostics and therapeutics. Thorac Cancer. 12:3088–3100. 2021.

30 

Wang Z, Zhao Z, Gao B and Zhang L: Exosome mediated biological functions within skeletal microenvironment. Front Bioeng Biotechnol. 10:9539162022.

31 

Zhang Y, Liu Y, Liu H and Tang WH: Exosomes: Biogenesis, biologic function and clinical potential. Cell Biosci. 9:192019.

32 

Kuang G, Wang Z, Luo C, Luo J and Wang J: Mechanism of exosomes in the tumor microenvironment in the abscopal effect (Review). Int J Oncol. 62:22023.

33 

Han L, Lam EWF and Sun Y: Extracellular vesicles in the tumor microenvironment: Old stories, but new tales. Mol Cancer. 18:592019.

34 

Anderson NM and Simon MC: The tumor microenvironment. Curr Biol. 30:R921–R925. 2020.

35 

Guerra E, Di Pietro R, Basile M, Trerotola M and Alberti S: Cancer-Homing CAR-T cells and endogenous immune population dynamics. Int J Mol Sci. 23:4052021.

36 

Kong H and Kim SB: Exosomal communication between the tumor microenvironment and innate immunity and its therapeutic application. Immune Netw. 22:e382022.

37 

Wang Z, Chen JQ, Liu JL and Tian L: Exosomes in tumor microenvironment: Novel transporters and biomarkers. J Transl Med. 14:2972016.

38 

Louise F, Ken YL, Richard BSR, Chien-Fu H and Wu TC: Cervical cancer immunotherapy: Facts and hopes. Clin Cancer Res. 27:4953–4973. 2021.

39 

Li I and Nabet BY: Exosomes in the tumor microenvironment as mediators of cancer therapy resistance. Mol Cancer. 18:322019.

40 

Hu X, Qiu Y, Zeng X and Wang H: Exosomes reveal the dual nature of radiotherapy in tumor immunology. Cancer Sci. 113:1105–1112. 2022.

41 

Baig MS, Roy A, Rajpoot S, Liu D, Savai R, Banerjee S, Kawada M, Faisal SM, Saluja R, Saqib U, et al: Tumor-derived exosomes in the regulation of macrophage polarization. Inflamm Res. 69:435–451. 2020.

42 

Yunna C, Mengru H, Lei W and Weidong C: Macrophage M1/M2 polarization. Eur J Pharmacol. 877:1730902020.

43 

Xia Y, Rao L, Yao H, Wang Z, Ning P and Chen X: Engineering macrophages for cancer immunotherapy and drug delivery. Adv Mater. 32:e20020542020.

44 

Pritchard A, Tousif S, Wang Y, Hough K, Khan S, Strenkowski J, Chacko BK, Darley-Usmar VM and Deshane JS: Lung tumor Cell-Derived exosomes promote M2 macrophage polarization. Cells. 9:13032020.

45 

Khan FH, Reza MJ, Shao YF, Perwez A, Zahra H, Dowlati A and Abbas A: Role of exosomes in lung cancer: A comprehensive insight from immunomodulation to theragnostic applications. Biochim Biophys Acta Rev Cancer. 1877:1887762022.

46 

Guo W, Li Y, Pang W and Shen H: Exosomes: A potential therapeutic tool targeting communications between tumor cells and macrophages. Mol Ther. 28:1953–1964. 2020.

47 

Bożyk A, Wojas-Krawczyk K, Krawczyk P and Milanowski J: Tumor Microenvironment-A short review of cellular and interaction diversity. Biology (Basel). 11:9292022.

48 

Burassakarn A, Srisathaporn S, Pientong C, Wongjampa W, Vatanasapt P, Patarapadungkit N and Ekalaksananan T: Exosomes-carrying Epstein-Barr virus-encoded small RNA-1 induces indoleamine 2, 3-dioxygenase expression in tumor-infiltrating macrophages of oral squamous-cell carcinomas and suppresses T-cell activity by activating RIG-I/IL-6/TNF-α pathway. Oral Oncol. 117:1052792021.

49 

Shen Y, Guo D, Weng L, Wang S, Ma Z, Yang Y, Wang P, Wang J and Cai Z: Tumor-derived exosomes educate dendritic cells to promote tumor metastasis via HSP72/HSP105-TLR2/TLR4 pathway. Oncoimmunology. 6:e13625272017.

50 

Li X, Liu Y, Zheng S, Zhang T, Wu J, Sun Y, Zhang J and Liu G: Role of exosomes in the immune microenvironment of ovarian cancer. Oncol Lett. 21:3772021.

51 

Rubenich DS, Omizzollo N, Szczepański MJ, Reichert TE, Whiteside TL, Ludwig N and Braganhol E: Small extracellular vesicle-mediated bidirectional crosstalk between neutrophils and tumor cells. Cytokine Growth Factor Rev. 61:16–26. 2021.

52 

Zhang X, Shi H, Yuan X, Jiang P, Qian H and Xu W: Tumor-derived exosomes induce N2 polarization of neutrophils to promote gastric cancer cell migration. Mol Cancer. 17:1462018.

53 

Shao Q, Deng L, Liu H, Liu Z, Chen J, Jiang F, Yan S and Fu R: Involvement of MM cell-derived exosomes in T lymphocytes immune responses. Oncol Lett. 20:312020.

54 

Ferguson Bennit HR, Gonda A, Kabagwira J, Oppegard L, Chi D, Licero Campbell J, De Leon M and Wall NR: Natural killer cell phenotype and functionality affected by exposure to extracellular survivin and Lymphoma-Derived exosomes. Int J Mol Sci. 22:12552021.

55 

Papademetrio DL, Garcia MN, Grasso D and Alvarez É: Autophagy-Mediated exosomes as immunomodulators of natural killer cells in pancreatic cancer microenvironment. Front Oncol. 10:6229562021.

56 

Luo H, Zhou Y, Zhang J, Zhang Y, Long S, Lin X, Yang A, Duan J, Yang N, Yang Z, et al: NK cell-derived exosomes enhance the anti-tumor effects against ovarian cancer by delivering cisplatin and reactivating NK cell functions. Front Immunol. 13:10876892023.

57 

Tumino N, Besi F, Martini S, Di Pace AL, Munari E, Quatrini L, Pelosi A, Fiore PF, Fiscon G, Paci P, et al: Polymorphonuclear Myeloid-Derived suppressor cells are abundant in peripheral blood of cancer patients and suppress natural killer cell Anti-tumor activity. Front Immunol. 12:8030142022.

58 

Yan Y, Wang R, Guan W, Qiao M and Wang L: Roles of microRNAs in cancer associated fibroblasts of gastric cancer. Pathol Res Pract. 213:730–736. 2017.

59 

Ding X, Xi W, Ji J, Cai Q, Jiang J, Shi M, Yu Y, Zhu Z and Zhang J: HGF derived from cancer-associated fibroblasts promotes vascularization in gastric cancer via PI3K/AKT and ERK1/2 signaling. Oncol Rep. 40:1185–1195. 2018.

60 

Ma J, Song X, Xu X and Mou Y: Cancer-Associated-Fibroblasts promote the chemo-resistance in gastric cancer through secreting IL-11 targeting JAK/STAT3/Bcl2 pathway. Cancer Res Treat. 51:194–210. 2019.

61 

Wei R, Song J and Liu X, Huo S, Liu C and Liu X: Immunosuppressive MFAP2+ cancer associated fibroblasts conferred unfavorable prognosis and therapeutic resistance in gastric cancer. Cell Oncol (Dordr). Aug 4–2023. View Article : Google Scholar : Epub ahead of print.

62 

Lu G, Du R, Dong J, Sun Y, Zhou F, Feng F, Feng B, Han Y and Shang Y: Cancer associated fibroblast derived SLIT2 drives gastric cancer cell metastasis by activating NEK9. Cell Death Dis. 14:4212023.

63 

Iha K, Sato A, Tsai HY, Sonoda H, Watabe S, Yoshimura T, Lin MW and Ito E: Gastric cancer cell-derived exosomal GRP78 enhances angiogenesis upon stimulation of vascular endothelial cells. Curr Issues Mol Biol. 44:6145–6157. 2022.

64 

Xue X, Huang J, Yu K, Chen X, He Y, Qi D and Wu Y: YB-1 transferred by gastric cancer exosomes promotes angiogenesis via enhancing the expression of angiogenic factors in vascular endothelial cells. BMC Cancer. 20:9962020.

65 

Zhou Z, Zhang H, Deng T, Ning T, Liu R, Liu D, Bai M, Ying G and Ba Y: Exosomes Carrying MicroRNA-155 target forkhead Box O3 of endothelial cells and promote angiogenesis in gastric cancer. Mol Ther Oncolytics. 25:2622022.

66 

Oya Y, Hayakawa Y and Koike K: Tumor microenvironment in gastric cancers. Cancer Sci. 111:2696–2707. 2020.

67 

Hao Y, Zheng C, Wang L, Hu Y, Guo H, Song Q, Zhang H, Zhang Z and Zhang Y: Covalent self-assembled nanoparticles with pH-dependent enhanced tumor retention and drug release for improving tumor therapeutic efficiency. J Mater Chem B. 5:2133–2144. 2017.

68 

Zhu KG, Yang J, Zhu Y, Zhu Q, Pan W, Deng S, He Y, Zuo D, Wang P, Han Y and Zhang HY: The microprotein encoded by exosomal lncAKR1C2 promotes gastric cancer lymph node metastasis by regulating fatty acid metabolism. Cell Death Dis. 14:7082023.

69 

Wang R, Liang L, Matsumoto M, Iwata K, Umemura A and He F: Reactive Oxygen species and NRF2 signaling, friends or foes in cancer? Biomolecules. 13:3532023.

70 

Chen H, Sun Q, Zhang C, She J, Cao S, Cao M, Zhang N, Adiila AV, Zhong J, Yao C, et al: Identification and Validation of CYBB, CD86, and C3AR1 as the key genes related to macrophage infiltration of gastric cancer. Front Mol Biosci. 8:7560852021.

71 

Zhao LX, Zhang K, Shen BB and Li JN: Mesenchymal stem cell-derived exosomes for gastrointestinal cancer. World J Gastrointest Oncol. 13:1981–1996. 2021.

72 

Tian X, Shen H, Li Z, Wang T and Wang S: Tumor-derived exosomes, myeloid-derived suppressor cells, and tumor microenvironment. J Hematol Oncol. 12:842019.

73 

Liu K, Gao X, Kang B, Liu Y, Wang D and Wang Y: The role of tumor stem cell exosomes in cancer invasion and metastasis. Front Oncol. 12:8365482022.

74 

da Costa VR, Araldi RP, Vigerelli H, D'Ámelio F, Mendes TB, Gonzaga V, Policíquio B, Colozza-Gama GA, Valverde CW and Kerkis I: Exosomes in the tumor microenvironment: From biology to clinical applications. Cells. 10:26172021.

75 

Wang T, Nasser MI, Shen J, Qu S, He Q and Zhao M: Functions of exosomes in the triangular relationship between the tumor, inflammation, and immunity in the tumor microenvironment. J Immunol Res. 2019:41978292019.

76 

Lopez K, Lai SWT, Lopez Gonzalez EDJ, Dávila RG and Shuck SC: Extracellular vesicles: A dive into their role in the tumor microenvironment and cancer progression. Front Cell Dev Biol. 11:11545762023.

77 

Thakur A, Johnson A, Jacobs E, Zhang K, Chen J, Wei Z, Lian Q and Chen HJ: Energy sources for exosome communication in a cancer microenvironment. Cancers (Basel). 14:16982022.

78 

Yan Y, Fu G, Ye Y and Ming L: Exosomes participate in the carcinogenesis and the malignant behavior of gastric cancer. Scand J Gastroenterol. 52:499–504. 2017.

79 

Yoon JH, Choi BJ, Nam SW and Park WS: Gastric cancer exosomes contribute to the field cancerization of gastric epithelial cells surrounding gastric cancer. Gastric Cancer. 25:490–502. 2022.

80 

Liang ZF, Zhang Y, Guo W, Chen B, Fang S and Qian H: Gastric cancer stem cell-derived exosomes promoted tobacco smoke-triggered development of gastric cancer by inducing the expression of circ670. Med Oncol. 40:242022.

81 

Chang L, Gao H, Wang L, Wang N, Zhang S, Zhou X and Yang H: Exosomes derived from miR-1228 overexpressing bone marrow-mesenchymal stem cells promote growth of gastric cancer cells. Aging (Albany NY). 13:11808–11821. 2021.

82 

Lin S, Que Y, Que C, Li F, Deng M and Xu D: Exosome miR-3184-5p inhibits gastric cancer growth by targeting XBP1 to regulate the AKT, STAT3, and IRE1 signalling pathways. Asia Pac J Clin Oncol. 19:e27–e38. 2023.

83 

Piao HY, Guo S, Wang Y and Zhang J: Exosome-transmitted lncRNA PCGEM1 promotes invasive and metastasis in gastric cancer by maintaining the stability of SNAI1. Clin Transl Oncol. 23:246–256. 2021.

84 

Yang H, Zhang H, Ge S, Ning T, Bai M, Li J, Li S, Sun W, Deng T, Zhang L, et al: Exosome-Derived miR-130a activates angiogenesis in gastric cancer by targeting C-MYB in vascular endothelial cells. Mol Ther. 26:2466–2475. 2018.

85 

Gu H, Ji R, Zhang X, Wang M, Zhu W, Qian H, Chen Y, Jiang P and Xu W: Exosomes derived from human mesenchymal stem cells promote gastric cancer cell growth and migration via the activation of the Akt pathway. Mol Med Rep. 14:3452–3458. 2016.

86 

Gao Q, Cui L, Huang C, Zhou C, Chen B, Wang Q, Wang M, Shene B, Xu W and Zhu W: Gastric cancer-derived exosomes induce PD-L1 expression on human bone marrow mesenchymal stem cells through the AKT-c-Myc signal axis. Front Life Sci. 442–451. 2022.

87 

Fu H, Yang H, Zhang X, Wang B, Mao J, Li X, Wang M, Zhang B, Sun Z, Qian H and Xu W: Exosomal TRIM3 is a novel marker and therapy target for gastric cancer. J Exp Clin Cancer Res. 37:1622018.

88 

Sun Y, Jin J, Jing H, Lu Y, Zhu Q, Shu C, Zhang Q and Jing D: ITIH4 is a novel serum biomarker for early gastric cancer diagnosis. Clin Chim Acta. 523:365–373. 2021.

89 

Wang S, He Y, Lu J, Wang Y, Wu X, Yan G, Fang X and Liu B: All-in-One strategy for downstream molecular profiling of tumor-derived exosomes. ACS Appl Mater Interfaces. 14:36341–36352. 2022.

90 

Dong Z, Sun X, Xu J, Han X, Xing Z, Wang D, Ge J, Meng L and Xu X: Serum Membrane Type 1-Matrix Metalloproteinase (MT1-MMP) mRNA Protected by exosomes as a potential biomarker for gastric cancer. Med Sci Monit. 25:7770–7783. 2019.

91 

Cai C, Zhang H, Zhu Y, Zheng P, Xu Y, Sun J, Zhang M, Lan T, Gu B, Li S and Ma P: Serum exosomal long noncoding RNA pcsk2-2:1 as a potential novel diagnostic biomarker for gastric cancer. OncoTargets Ther. 12:10035–10041. 2019.

92 

Joo HS, Jeon HY, Hong EB, Kim HY and Lee JM: Exosomes for the diagnosis and treatment of dementia. Curr Opin Psychiatry. 36:119–125. 2023.

93 

Aheget H, Mazini L, Martin F, Belqat B, Marchal JA and Benabdellah K: Exosomes: Their role in pathogenesis, diagnosis and treatment of diseases. Cancers (Basel). 13:842020.

94 

Wang J, Sun X, Zhao J, Yang Y, Cai X, Xu J and Cao P: Exosomes: A novel strategy for treatment and prevention of diseases. Front Pharmacol. 8:3002017.

95 

Jiang L, Chen W, Ye J and Wang Y: Potential role of exosomes in ischemic stroke treatment. Biomolecules. 115:122022.

96 

You D, Kundu S, Schmaier AH, Khorana AA and McCrae KR: Exosome polyphosphate mediates the activation of FXII by cancer cell-derived exosomes. Blood. 132:38002018.

97 

Zhang H, Wang Y, Bai M, Wang J, Zhu K, Liu R, Ge S, Li J, Ning T, Deng T, et al: Exosomes serve as nanoparticles to suppress tumor growth and angiogenesis in gastric cancer by delivering hepatocyte growth factor siRNA. Cancer Sci. 109:629–641. 2018.

98 

Wang X, Zhang H, Bai M, Ning T, Ge S, Deng T, Liu R, Zhang L, Ying G and Ba Y: Exosomes serve as nanoparticles to deliver Anti-miR-214 to reverse chemoresistance to cisplatin in gastric cancer. Mol Ther. 26:774–783. 2018.

Related Articles

Journal Cover

March-2024
Volume 64 Issue 3

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang M, Shu H, Cheng X, Xiao H, Jin Z, Yao N, Mao S and Zong Z: Exosome as a crucial communicator between tumor microenvironment and gastric cancer (Review). Int J Oncol 64: 28, 2024
APA
Wang, M., Shu, H., Cheng, X., Xiao, H., Jin, Z., Yao, N. ... Zong, Z. (2024). Exosome as a crucial communicator between tumor microenvironment and gastric cancer (Review). International Journal of Oncology, 64, 28. https://doi.org/10.3892/ijo.2024.5616
MLA
Wang, M., Shu, H., Cheng, X., Xiao, H., Jin, Z., Yao, N., Mao, S., Zong, Z."Exosome as a crucial communicator between tumor microenvironment and gastric cancer (Review)". International Journal of Oncology 64.3 (2024): 28.
Chicago
Wang, M., Shu, H., Cheng, X., Xiao, H., Jin, Z., Yao, N., Mao, S., Zong, Z."Exosome as a crucial communicator between tumor microenvironment and gastric cancer (Review)". International Journal of Oncology 64, no. 3 (2024): 28. https://doi.org/10.3892/ijo.2024.5616