Skip to main content
MedComm logoLink to MedComm
. 2021 Jul 1;2(3):297–314. doi: 10.1002/mco2.49

The multifaceted involvement of exosomes in tumor progression: Induction and inhibition

Wen‐Jie Gu 1,#, Yi‐Wen Shen 1,#, Li‐Jun Zhang 1, Hong Zhang 1, Dale G Nagle 1,2, Xin Luan 1,, San‐Hong Liu 1,
PMCID: PMC8554660  PMID: 34766148

Abstract

As key performers in intercellular communication, exosomes released by tumor cells play an important role in cancer development, including angiogenesis, cancer‐associated fibroblasts activation, epithelial‐mesenchymal transformation (EMT), immune escape, and pre‐metastatic niche formation. Meanwhile, other cells in tumor microenvironment (TME) can secrete exosomes and facilitate tumor progression. Elucidating mechanisms regarding these processes may offer perspectives for exosome‐based antitumor strategies. In this review, we mainly introduce the versatile roles of tumor or stromal cell derived exosomes in cancer development, with a particular focus on the biological capabilities and functionalities of their diverse contents, such as miRNAs, lncRNAs, and circRNAs. The potential clinical application of exosomes as biomarkers in cancer diagnosis and prognosis is also discussed. Finally, the current antitumor strategies based on exosomes in immunotherapy and targeted delivery for chemotherapeutic or biological agents are summarized.

Keywords: biomarker, cancer, drug delivery, exosome, microRNAs


Exosomes released by tumor cells and tumor microenvironment‐associated cells play a pivotal role in cancer initiation and progression. Exosomes facilitate cancer development in multiple aspects, including angiogenesis, cancer‐associated fibroblasts activation, immune escape, epithelial‐mesenchymal transformation, pre‐metastasis niche formation, and drug resistance.

graphic file with name MCO2-2-297-g003.jpg

1. INTRODUCTION

Exosomes, the nanosized (40‐160 nm) subgroups of extracellular vesicles (EVs), were initially believed to be the platelet dust in plasma. 1 , 2 Exosomes are generated in the maturation process of endosomes to multivesicular bodies (MVBs). In early endosomes, molecular materials such as proteins, RNA, and DNA, are collected and further processed. Then the late endosomes mature into MVBs, and intraluminal vesicles (ILVs) are formed. Eventually, MVBs can fuse with the plasma membrane, and ILVs are released as exosomes into extracellular space. 3 In the past decades, studies have unraveled that exosomes are important mediators of intercellular communication and are involved in a diverse range of biological processes with the varying cargos inside, including lipids, proteins, nucleic acids, and metabolites. 2 When exosomes are released into the extracellular space, they can reach recipient cells and deliver cargos, exerting functional effects and inducing related phenotypic changes. 4

Exosomes are transferred between cells and exhibit multi‐faceted physiological and pathological function. Exosomes can mediate the cell‐to‐cell communication and maintain the normal bioactivities of recipient cells through the transportation of cargos within extracellular space. 5 On the other hand, exosomes also play a critical role in the progress of some diseases, especially as the transmitters in tumor microenvironment (TME). 6 , 7 It has been demonstrated that cancer cells exhibited the significantly increased exosomes secretion compared with normal cells with profound influences on tumor progression and metastasis. 6 , 7 For example, exosomes derived from tumor cells can alter the properties of normal cells by carrying oncogenic materials and initiate phenotypic changes in TME, thus progressing cancer development. 8 , 9 Emerging evidences have also proved that exosomes are responsible for the tumor cell expansion, metabolic activity remodeling, tumor angiogenesis, immunosuppressive TME, and acquired drug resistance (Figures 1 and 2). 10 These studies urge the need of corresponding regarding these revealed mechanisms.

FIGURE 1.

FIGURE 1

The functions of tumor cell‐derived exosomes in cancer development. Tumor cell‐derived exosomes carrying biological molecules, such as miRNAs, proteins, and nucleic acids, are involved in various aspects of tumor progression, including angiogenesis, cancer‐associated fibroblasts activation, immune escape, epithelial‐mesenchymal transformation, pre‐metastasis niche formation, and drug resistance

FIGURE 2.

FIGURE 2

The functions of exosomes from other cells in TME in cancer development. Exosomes from other cells in TME, such as mesenchymal stem cells, cancer‐associated fibroblasts and macrophages can promote cell proliferation, drug resistance, and tumor metastasis, thus facilitating tumor progression

Research have shown that exosomes are recognized as important transporters in cancer and exhibit promising prospects for cancer diagnosis (Figure 3). 11 Recently, the applications of exosomes have been focused on biomarkers in diagnosis, underlying functions and mechanisms, and drug delivery systems owing to its unique natural features. 12 , 13 In this review, we focus on the present studies and the underlying mechanisms of exosomes, especially regarding the involvement in TME. Additionally, we conclude the application exosomes in tumor diagnosis and therapy.

FIGURE 3.

FIGURE 3

Application of exosomes in cancer diagnosis and therapy. A, Exosomes containing specific RNAs and proteins can be utilized as noninvasive biomarkers for cancer diagnosis and prognosis. B, Exosomes can serve as nanocarriers for drug delivery and can be applied in immunotherapy as cancer therapeutic vaccine

2. ROLE OF TUMOR CELL‐DERIVED EXOSOMES FOR TUMOR PROGRESSION

Exosomes function variably according to the origin of cells. Tumor cells‐derived exosomes (TCDEs) contribute to the tumor progression through the induction of tumor cell proliferation, invasion, and migration, and are also proved to be the key attributes of tumorigenicity and TME maintenance. Targeting TME is widely accepted as a promising strategy to effectively suppress tumor progression, and six important aspects of involvement in tumor progression regarding tumor cell‐released exosomes have been summarized as follows (Figure 1 and Table 1).

TABLE 1.

Role of tumor cell‐derived exosomes for tumor progression

Cell type Exosomal cargo Function Mechanism Reference
Angiogenesis
Papillary thyroid cancer cells miR‐21‐5p Increase angiogenesis Target and suppress TGFBI and COL4A1 17
Ovarian cancer cells miR‐205 Induce angiogenesis and promote metastasis Via the PTEN‐AKT pathway 18
Lung cancer cells miR‐23a Increase angiogenesis and vascular permeability Target prolyl hydroxylase and tight junction protein ZO‐1 20
Glioblastoma cells miR‐182‐5p Promote angiogenesis Target kruppel‐like factor 2 and 4 21
Glioblastoma cells miR‐26a Promote angiogenesis Activation of the PI3K/Akt signaling pathway by targeting PTEN. 22
Cervical squamous cells miR‐221‐3p Promote angiogenesis Target THBS2 23
Gastric cancer cells miR‐130a Activate angiogenesis Target C‐MYB 16
Gastric cancer cells miR‐155 Promote angiogenesis Target forkhead box O3 24
Gastric cancer cells miR‐135b Promote angiogenesis Inhibit FOXO1 expression 25
Gastric cancer cells miR‐23a Promote angiogenesis Target PTEN 26
Gastric carcinoma cells miR‐155 Promote angiogenesis Target the c‐MYB/ VEGF axis 27
Colorectal cancer cells miR‐183‐5p Promote angiogenesis Regulation of FOXO1 28
Colorectal cancer cells miR‐1229 Promote angiogenesis Target HIPK2 29
Hepatocellular carcinoma cells Angiopoietin‐2 Induce angiogenesis Activate the AKT/eNOS and AKT/β‐catenin pathways in HUVECs 30
Hepatocellular carcinoma cells LOXL4 Promote angiogenesis and cell migration Paracrine transfer mechanism; activate FAK/Src pathway 31
Colorectal cancer cells miR‐590‐5p Inhibit angiogenesis and metastasis Regulate NF 90/VEGFA Axis 32
Menstrual mesenchymal stem cells / Inhibit angiogenesis and tumor growth Induce endothelial cell death, modulate VEGF secretion 33
Liver stem cells

miR‐15a

miR181b

miR320c

miR‐874

Inhibit angiogenesis Downregulation of FGF1 and PLAU 34
Breast cancer cells

miR‐23b

miR‐27b

miR‐320b

Inhibit angiogenesis Decrease the expression of PLAU, AMOTL1, NRP1, ETS2 35
Cancer‐associated fibroblast
Hepatocellular carcinoma cells miR‐21 Convert hepatocyte stellate cells to cancer‐associated fibroblasts, contribute to tumor progression miRNA‐21 targets PTEN, leading to activation of PDK1/AKT signaling; CAF secrete VEGF, MMP2, MMP9, bFGF, and TGF‐β 39
Melanoma cells miR‐155‐5p

Induce reprogramming of fibroblasts into CAFs

miR‐155 trigger the proangiogenic switch of CAFs

promote the expression of VEGFa, FGF2, and MMP9

Via the SOCS1/JAK2/STAT3 signaling pathway 40
Oral cancer cells miR‐34a‐5p

Confer aggressiveness

Induce EMT and promote cancer cells metastasis

Via the AKT/GSK‐3β/β‐catenin/Snail signaling cascade 41
Colorectal cancer cells miR‐93‐5p Higher miR‐93‐5p leads to radioresistance and increase the tumor growth Downregulation of FOXA1 and upregulation of TGF‐β3 42
Immune escape
Melanoma cells PD‐L1 Increased PD‐L1 suppresses CD8+T cells and facilitates tumor growth Through stimulation with interferon‐γ (IFN‐γ) 48
Non‐small cell lung cancer cells PD‐L1 Reduce cytokine production and induce apoptosis in CD8+ T cells Inhibit IFN‐γ secretion and impair immune functions 50
Hepatocellular carcinoma cells HMGB1 Foster immune evasion and promote the advanced disease stage Activate B cells and promote TIM‐1+ Breg expansion via TLR 2/4 and MAPK signaling pathways 52
Melanoma cells

miR‐3187‐3p

miR‐498

miR‐122

miR149

miR‐181a/b

Downregulate T‐cell responses Through decreased TCR signaling, diminished cytokine and granzyme B and TNFα secretion 53
Chronic lymphocytic leukemia monocytes HY4 Release CCL2, CCL4, and IL‐6, and induce PD‐L1 expression, contribute to cancer‐related inflammation and concurrent immune escape Through TLR7 signaling 54
Epithelial‐mesenchymal transformation
Hypoxic bone marrow‐derived mesenchymal stem cells

miR‐193a‐3p

miR‐210‐3p

miR‐5100

Induce EMT and promote tumor cell invasion Activation of STAT3 signaling 55
Clear cell renal cell carcinoma cells miR‐19b‐3p Induce EMT and confer organotropism Decreased PTEN, CD103+ 56
Melanoma cells Decreased let‐7

Promote phenotype switching;

Initiate EMT and promote metastasis

Through paracrine/autocrine signaling;

Activation of MAPK pathway

57
Hepatocellular carcinoma cells Rab27a blockade Elicit EMT and facilitate the tumor progression Through MAPK/ERK pathway 58
Hepatocellular carcinoma cells / Inhibit EMT Vps4A mediates the plasma membrane localization and decreases exosome‐released β‐catenin 60
Mesenchymal stem cells / Reduce CAFs and suppress EMT, induce angiogenesis and maintain vascular homeostasis / 62
Colorectal cancer cells miR‐92a‐3p

Lead to EMT,

promote metastasis and chemoresistance

Activation of Wnt/β‐catenin pathway and inhibition of mitochondrial apoptosis 63
Breast cancer cells miR‐181d‐5p Promote EMT Downregulation of HOXA5 and CDX2 64
Pre‐metastatic niche
Pancreatic ductal adenocarcinoma cells Migration inhibitory factor (MIF) Induce pre‐metastatic niche formation and metastasis Driven by TGFβ‐signaling, FN deposition, and recruitment of BM‐derived macrophages 67
Colorectal cancer cells miR‐25‐3p Promote angiogenesis and vascular permeability, induce vascular leakiness and enhance metastasis Regulate the expression of VEGFR2, ZO‐1, occludin, and Claudin5 by targeting KLF2 and KLF4 68
Lung epithelial cells RNA Initiate neutrophil recruitment and lung metastatic niche formation Activate toll‐like receptor 3 69
Melanoma cells Pigment epithelium‐derived factor (PEDF) Loss of PEDF enables immunosuppression and abrogates the immune clearance of cancer cells metastasis PEDF alerts host immune system, innate immune responses 71
Drug resistance
Gastric cancer cells miR‐21 Reduce cisplatin chemosensitivity and suppress apoptosis MiR‐21 transfer enhances activation of PI3K/AKT signaling pathway by downregulation of PTEN 76
Advanced colorectal cancer cells miR‐128‐3p Suppress EMT and increase intracellular oxaliplatin accumulation Negative regulation of Bmi1 and MRP5 77
Glioblastoma cells miR‐15a Enhance chemosensitivity to temozolomide Target XRCC4 78
Glioblastoma cells miR‐1238 Confer temozolomide chemoresistance Target the CAV1/EGFR pathway 79
Multidrug‐resistant hepatocellular carcinoma cells miR‐32‐5p Induce multidrug resistance Target PTEN via the PI3K/Akt pathway 80
Pancreatic cancer cells EphA2 Transmit chemoresistance / 81
Ovarian cancer cells / Increase cisplatin efflux, augmenting metastasis and chemotherapy resistance Increased exosome secretion through regulation of Rab proteins by STAT3 82
Ovarian cancer cells miR‐1246 Acquire chemoresistance Modulate Cav1and p‐gp interaction 83

Symbol “/” represents the unknown cargo.

2.1. TCDE and angiogenesis

Angiogenesis is a crucial part which shows a profound influence on promoting tumor progression, consequently leading to poor prognosis. In the preexisting vascular network, new blood vessels are formed by either endothelial cells mediated‐sprouting angiogenesis or interstitial tissues‐expanded intussusceptive angiogenesis. 14 Besides, the secretion of pro‐angiogenic factors by tumor cells triggers the creation of this disorganized, immature, and permeable vascular network, promoting cancer cell invasion and impeding the tumor‐killing action of immune cells. 15

Surprisingly, exosomes show great potential in inducing either pro‐ or anti‐angiogenic signaling through the information delivery to endothelial cells. Additionally, exosomes adjust their cargo composition to fine tune the process of blood vessel formation in response to changes in TME. 14 Contents of exosomes, especially miRNAs potentiate this process, and multiple studies have validated the role of exosomal miRNA in the activation of angiogenesis. 16 TCDEs are identified to trigger angiogenesis in papillary thyroid cancer, 17 ovarian cancer, 18 breast cancer, 19 and lung cancer 20 through exosome‐mediated miRNA signaling. Under hypoxic conditions, glioblastoma cells derived‐exosomes carrying miR‐182‐5p promote tumor angiogenesis and increase vascular permeability, which are supportive for microenvironment. 21 Consistently, glioma stem cells‐derived exosomal miR‐26a enhances angiogenesis of microvessel endothelial cells in glioma. 22 In cervical squamous cell carcinoma (CSCC), CSCC cell‐secreted exosomal miR‐221‐3p was found to enhance the angiogenesis in the transfer to vessel endothelial cells targeting THBS2. 23 In gastric cancer (GC), TCDEs carrying miR‐130a, 16 miR‐155, 24 miR‐135b, 25 miR‐23a, 26 and miR‐155 27 were investigated to exert identical effect on promotion of angiogenesis through varying mechanisms. In colorectal cancer (CRC), exosomal miR‐183‐5p induces angiogenesis by regulation of FOXO1, 28 and miR‐1229 promotes angiogenesis by targeting HIPK2. 29 In hepatocellular carcinoma (HCC), exosome‐mediated secretion of Angiopoietin‐2‐induced angiogenesis 30 and exosomal lysyl oxidase like 1 (LOXL) promotes cell invasion and metastasis. 31 Most of TCDE‐related miRNAs exhibit angiogenic promotion effect, however, some exosomes are the opposite. miR‐590‐5p acted as an anti‐onco‐miR which was found to inhibit angiogenesis and metastasis through the regulation of nuclear factor (NF) 90/VEGFA axis. 32 Several stem cells, including menstrual mesenchymal stem cells (MSCs) and liver stem cells, have been identified as inhibitors of angiogenesis, and secreted exosomes loading miRNA are detected to suppress angiogenesis and tumor growth. 33 , 34 In breast cancer, researchers discovered that an antiangiogenic agent, docosahexaenoic acid (DHA), triggered the secretion of exosomes and the expression level of miRNAs including miR‐23b, miR‐27b, and miR‐320b, which inhibited tube formation of endothelial cells by decreasing the expression of their respective pro‐angiogenic target genes, including PLAU, AMOTL1, NRP1, and ETS2. 35

2.2. TCDE and cancer‐associated fibroblasts

Cancer‐associated fibroblasts (CAFs), a kind of stromal cells, form a part of TME and play an important role in the regulation of the tumor progression. 36 Based on the demonstrated presence of CAFs in malignancy, Paggetti et al unveiled the presence of CAFs in chronic lymphocytic leukemia (CLL) lymph node. What's more, AKT serine/threonine kinase 1 (AKT) and NF‐κB in stromal cell were demonstrated to be essential for induction of the inflammatory phenotype. It is worth noting that, the uptake of cancer exosomes by stromal cells elicits the inflammatory effects which contribute to the CAFs transition. 37

Ringuette et al determined the fibroblast differentiation into CAFs through transforming growth factor beta 1 (TGF‐β) via bladder cancer (BC) cells‐derived exosomes, suggesting exosomal TGF‐β can be considered as a novel molecular mechanism involved in CAF activation in tumor. 38 Previous studies also have shown that exosomes act as a messenger between the communication/crosstalk of tumor cells and CAFs. Therefore, a deep understanding of this interaction may make contribution to the cancer therapy.

Exosome‐secreted miRNAs exert the inducing progression effects on tumors. For example, HCC‐derived exosomal miRNA‐21 induces tumor progression by activation of hepatic stellate cells into CAFs, demonstrating the role of exosome mediator. 39 Another case in melanoma shows that exosomal miR‐155‐5p induces CAF proangiogenic function via suppressor of cytokine signaling 1 (SOCS1)/Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling pathway, 40 and exosome‐mediated paracrine miR‐34a‐5p CAF triggers oral cancer cells proliferation and metastasis. 41 Chen et al revealed that exosome‐mediated transfer of miR‐93‐5p from CAFs led to radioresistance in CRC cells through the downregulation of FOXA1 and upregulation of TGF‐β3. 42

2.3. TCDE and immune escape

Immune system displays a critical role in maintaining tumor development and progression. Exosomes from both non‐immune and immune cells participate in immune regulation, such as mediating T cell apoptosis, reducing NK‐ and T cells, and triggering the differentiation of myeloid cells to myeloid derived suppressor cells. 43 To destroy the anti‐tumor immunity, tumor‐derived‐exosomes deliver the suppressive proteins and nucleic acids to immune cells including monocytes, macrophages, dendritic cells (DCs), natural killer cells, T cells, B cells and thus evading immune surveillance. 44 , 45 programmed death‐ligand 1 (PD‐L1) is one of these suppressive proteins that exist on the surface of exosomes. 46 Exosomal PD‐L1 systemically tends to suppress the anti‐tumor immune response, and its genetic blockage promotes T cell activity in the draining lymph node resulting in the induction of systemic anti‐tumor immunity and memory. 47 On this basis, investigations revealed that exosomal PD‐L1 can be upregulated by interferon‐γ, thereby suppressing CD8+T cells and inducing tumor growth. 48 Besides, it has been revealed that PD‐L1‐induced immunosuppression is correlated with anti‐PD‐1 response, implying exosomal PD‐L1 can be applied as a biomarker for anti‐PD‐1 therapy. 48 In addition, compared to soluble PD‐L1 which increases in healthy individuals with age, exosomal PD‐L1 provides alternative options for cancer diagnosis and prognosis prediction. 49 In parallel, non‐small cell lung cancer (NSCLC) cell‐derived exosomal PD‐L1 fosters tumor growth through immune escape of identical mechanisms. 50 Another study in HCC reported that tumor‐derived exosomes could promote the immune‐suppressive regulatory B cells (Bregs), a kind of B cells subsets. 51

High mobility group box 1 (HMGB1), a kind of DNA‐binding nuclear protein, is expressed on exosome membranes. Interestingly, studies illustrated that Bregs infiltrated tumors, and T‐cell immunoglobulin mucin domain‐1 (TIM‐1) (a marker of Bregs)+Bregs exhibited enhanced suppressive activity against CD8+T cells. Researchers detected that the expansion of Bregs promoted the advanced disease stage through the exosomal‐derived HMGB1 activation. 52 In melanoma, tumor exosomes harboring enriched hsa‐miR‐3187‐3p, hsa‐miR‐498, hsa‐miR‐122, hsa‐miR149, and hsa‐miR‐181a/b elicit the immune escape by regulating TCR signaling and TNFα secretion. 53 Furthermore, Haderk et al discovered the protumorigenic skewing mechanisms related to PD‐L1 expression in CLL and concluded that CLL‐derived exosomes mediated the transfer of non‐coding RNAs to monocytes, leading to the immune escape. 54

2.4. TCDE and epithelial‐mesenchymal transformation

Epithelial‐mesenchymal transformation (EMT) is a conserved process characterized as the loss of epithelial features and the possession of mesenchymal phenotype. 8 Prior research generally demonstrates that EMT is a tumor‐associated process that may elicit tumor cell invasion, metastasis, and drug resistance. Zhang et al proved that hypoxic bone marrow‐derived MSCs (BMSCs)‐derived exosomes promoted tumor cell invasion and EMT. 55 Additionally, using miR‐193a‐3p, miR‐210‐3p, and miR‐5100‐loading exosomes together exerts better diagnostic accuracy in patients with lung cancer metastasis compared with respective treatment. 55 Exosomes secreted from cancer cells are implicated in inducing EMT in cancer development. Wang et al conducted the study on the correlation between cancer stem cells (CSC) from clear cell renal cell carcinoma and EMT. The outcome revealed that CSC exosomes initiated EMT through the transmission of MIR‐19b‐3p to CCSCC cells, which was related to the repressed expression of phosphatase and tensin homolog (PTEN). What's more, metastasis exosomes display stronger EMT effect. 56 EMT occurs in melanoma cell‐derived exosomes as a consequence of the activation of MAPK pathway, thus leading to metastasis. Decreased let‐7, a miRNA modulator of EMT is detected, which may account for this phenomenon. 57 Similar examples are shown in HCC‐derived exosomes. Highly metastatic HCC‐derived exosomes improve migration, chemotaxis, and invasion of low metastatic cells by transferring pro‐metastatic molecules. Abrogating the release of these exosomal factors through inhibition of Rab27a, a modulator of exosome secretion, promotes invasion of parental cells. 58 Vps4A, functioning as a tumor suppressor through regulation of exosomal miRNAs in HCC, is demonstrated to mediate the plasma membrane localization and decrease exosome‐released β‐catenin, thus inhibiting EMT when it is overexpressed. 59 , 60

As mentioned above, CAFs act as abettors in tumor progression, including the induction of EMT. 61 Yeon et al observed that MSC‐derived exosomes reduced CAFs and suppressed EMT, thereby inducing angiogenesis and maintaining vascular homeostasis, while cancer‐derived exosomes that transdifferentiate CAFs promoted EMT. 62 Hu et al examined the elevated level of miR‐92a‐3p as a consequence of CAFs transferring exosomes to CRC cells. Additionally, increased miR‐92a‐3p activated Wnt/β‐catenin pathway and inhibited mitochondrial apoptosis, leading to EMT and thus promoting metastasis and chemoresistance. 63 Another similar study shows the promotion of EMT in breast cancer caused by CAFs secreted exosomal microRNA‐181d‐5p and the underlying mechanisms is related to the downregulation of homeobox A5 (HOXA5) and caudal‐related homeobox 2 (CDX2). 64 Studies regarding the correlation between CAFs and EMT have been performed, providing understandings on repressing tumor progression.

2.5. TCDE and pre‐metastatic niche

Pre‐metastatic niche in cancers is a prevalent precondition of metastasis. Liu et al elucidated it with six pro‐metastasis features including immunosuppression, inflammation, angiogenesis/ vascular permeability, lymphangiogenesis, organotropism, and reprogramming. 65 There is an urgent need to seek novel and effective approaches targeting pre‐metastasis niche to make amends for the delayed detection and prevent further metastasis.

The consensus has been that exosomes participate in pre‐metastatic niche formation of cancers. 66 Costa‐Silva et al found that pancreatic ductal adenocarcinomas (PDAC)‐derived exosomes were integrated with Kupffer cells, a kind of macrophage, to stimulate the fibronectin production and induce the formation of pre‐metastasis niche. Furthermore, macrophage migration inhibitory factors (MIF), which were released by macrophages in fibrotic microenvironment and enriched in PDAC‐exosomes, could prevent the niche formation when it was blocked. 67 Zeng et al manifested the involvement of CRC‐derived exosomal miR‐25‐3p in promoting pre‐metastatic niche formation through mechanisms related to the angiogenesis and vascular permeability. 68 Toll‐like receptor 3 (TLR3) are able to recognize inflammatory signals and promote tumor progression. Liu et al uncovered that deficiency of TLR3 in lung epithelial cells prevented the niche formation, while exosomal RNA reversed this process by activating TLR3, consequently recruiting Neutrophils and favoring the niche formation. 69 Similarly, melanosomes foster tumor niche by reprogramming dermal fibroblast into CAFs through transferring miR‐211. 70 Contrary to the aforementioned research, Plebanek et al discovered that non‐metastasic exosomes resulted in the inhibition of metastasis. It is identified that pre‐metastatic tumors release exosomes, simultaneously eliciting immune surveillance, resulting in tumor cell clearance at the pre‐metastatic niche. 71

It has also been reported that melanoma‐derived exosomes facilitate the formation of pre‐metastatic niche via induction of vascular leakiness and pro‐vasculogenic phenotype of bone marrow progenitors. 72 Another notable investigation based on previous exosome‐mediated metastasis theory revealed the crucial role of exosome integrin in promoting metastasis. Hoshino et al provided a rational of exosome integrin predicting organotropism of metastatic cells, which indicated the promising prospect of the application in diagnosis and treatment. 73

2.6. TCDE and drug resistance

Acquired drug resistance is a troublesome obstacle that causes limited therapeutic response in cancer patients. 74 Previous studies have documented that exosomes derived from tumor cells or related stromal cells mediate the drug resistance through the exosomal transfer of miRNAs. 75 The miR‐21 transferred from macrophages to GC cells was detected to reduce cisplatin chemosensitivity and suppress apoptosis. 76 However, exosome‐transmitted miR‐128‐3p overexpression displays enhanced chemosensitivity in oxaliplatin‐resistant advanced CRC through altering target gene expression, while lower expression of miR‐128‐3p leads to poor drug response. 77 In glioblastoma, exosomal transfer of miR‐15a and miR‐1238 was substantiated to contribute to chemotherapeutic‐resistance. 78 , 79 In multidrug‐resistant HCC cells, elevated exosomal miRNA‐32‐5p and reduced PTEN are associated with poor prognosis. Additionally, miRNA‐32‐5p targeting PTEN induces multidrug resistance via the PI3K/Akt pathway. 80 EphA2 is overexpressed in tumors and has been recognized as a pivotal mediator of chemoresistance. In pancreatic cancer, exosome‐mediated EphA2 transfer is revealed to perform chemoresistance transmission between variable gemcitabine resistance cell lines. 81 Dorayappan et al demonstrated that exosomes were greatly secreted under hypoxic condition, and these hypoxia‐induced exosomes increased cisplatin efflux, augmenting metastasis and chemotherapy resistance in ovarian cancer. 82 Another investigation in ovarian cancer has shown that exosomal miR‐1246 is potent effector in acquiring chemoresistance via modulating Cav1and p‐gp interaction. 83

In addition, the expression of drug targets on exosomes, including PD‐L1, CD20, and CTLA‐4, may be a novel mechanism for drug resistance. These molecules carried on exosomes can deplete the specific drugs or antibodies to waken the therapeutic effects. 46 Yang et al have proved that the accumulation of exosomal PD‐L1 in TME induced therapeutic resistance, and the successful inhibition of these exosomes’ secretion could recover the efficacy of anti‐PD‐1 therapy. 84

3. ROLE OF EXOSOMES FROM OTHER CELLS IN TME FOR TUMOR PROGRESSION

3.1. Mesenchymal stem cell‐derived exosomes

MSCs are considered as crucial regulators of TME and control multiple aspects of cancer progression, including tumorigenesis, angiogenesis, and metastasis. 85 It has been reported that the interactions between MSCs and tumor cells, predominately mediated by exosomes, is the key performer of MSCs functions. On the one hand, exosomes derived from tumor cells transfer biologically active proteins and nuclear acids to MSCs, co‐opting MSCs and transforming them into tumor supportive types. On the other hand, MSCs reprogrammed by tumor exosomes become avid producers of their own exosomes, secreting various factors accumulating cancer progression (Table 2). 2 , 86

TABLE 2.

Role of exosomes from other cells in TME for tumor progression

Cancer type Cargo Function Mechanism Reference
Mesenchymal stem cells
Lung cancer

miR‐193a‐3p

miR‐210‐3p

miR‐5100

Promote cancer cell invasion and EMT Activate STAT3 signaling pathway 55
Glioblastoma miR‐1587

Increase the proliferation and clonogenicity of tumor‐initiating stem‐like cells,

lead to greater tumor burden and decreased survival immune regulation and inhibit B‐lymphocytes functions

Downregulation of NCOR1 87
Breast cancer / Promote cell growth and protect tumor cells from chemotherapeutic drug‐induced apoptosis in vitro Activation of Hippo signaling pathway 89
Prostate cancer miR‐205 Repress cell proliferation, invasion and migration, impede cancer progression Inhibit RHPN2 90
Pancreatic Cancer miR‐126‐3p Inhibit the proliferation, invasion, and metastasis of pancreatic cancer cells, and promote apoptosis; suppress cancer development Target ADAM9 91
Glioblastoma miR‐133b Suppress cancer development Inhibit the Wnt/β‐catenin signaling pathway 92
Fibroblast
Pancreatic cancer / Promote proliferation and drug resistance Increased snail 94
Colorectal cancer miR‐92a‐3p overexpression Promote metastasis and chemotherapy resistance Activate Wnt/β‐catenin pathway, inhibit mitochondrial apoptosis 63
Gastric cancer miR‐522 Decrease chemo‐sensitivity and inhibit ferroptosis Suppress ALOX15 expression and lipid‐ROS accumulation 95
Colorectal cancer lncRNAs H19 Promote the stemness and chemoresistance Activate the β‐catenin pathway 96
Macrophage
Colorectal cancer

miR‐21‐5p

miR‐155‐5p

Induce cancer cell migration and invasion Downregulate BRG1 expression 98
Pancreatic ductal adenocarcinoma miR‐501‐3p Promote tumor progression Target TGFBR3 and activate TGF‐β signaling pathway 99
Gastric cancer miR‐21 Contribute to cisplatin resistance Activate PI3K/AKT signaling pathway and inhibit cell apoptosis 76
Gastric cancer Functional apolipoprotein E Remodel the cytoskeleton‐supporting migration Activate PI3K‐Akt signaling pathway 100

Symbol “/” represents the unknown cargo.

Zhang et al found that exosomes released by bone marrow‐derived MSCs (BMSCs) could promote invasion of lung cancer cells by activating STAT3 signaling pathway. The microRNAs, including miR‐193a‐3p, miR‐210‐3p, and miR‐5100, transferred by exosomes from BMSCs to epithelial cancer cells exhibited diagnostic accuracy that discriminate cancer patients from non‐cancerous controls. 55 Additionally, MSC‐derived exosomes from glioma tissues increase the proliferation and clonogenicity of tumor‐initiating stem‐like cells, leading to greater tumor burden and decreased survival. 87 Moreover, MSC‐derived exosomes are implicated in immune regulation and inhibit the functions of B‐lymphocytes by affecting the expression of specific mRNAs. 88 In a recent study, Wang et al demonstrated that exosomes secreted by MSC‐differentiated adipocytes promote breast cancer cell growth and protect tumor cells from chemotherapeutic drug‐induced apoptosis in vitro. The inhibition of exosomes attenuates the tumor‐promoting effects of adipocytes. 89

While MSC‐derived exosomes primarily exert pro‐tumor effects in cancer development, studies also reported the tumor suppressive activities of exosomes secreted by MSCs. Jiang and colleagues showed that MSC‐derived exosomes carrying miR‐205 could repress prostate cancer cell proliferation, invasion, and migration, thus impeding cancer progression through inhibiting rhophilin Rho GTPase binding protein 2 (RHPN2). 90 Similarly, MSC‐derived exosomal miR‐126‐3p suppresses the development of pancreatic cancer by targeting ADAM9. 91 In another study, Xu et al found that exosomes containing miR‐133b released by MSCs attenuated glioma development via inhibiting the Wnt/β‐catenin signaling pathway. 92 Collectively, the diverse cargos transferred by MSC‐derived exosomes are the predominant determinant of their effects in tumor progression, and MSCs also varies dependent on local TME. 85

3.2. Fibroblast‐derived exosomes

Exosomes released by tumor cells promote tumorigenesis through activation of CAFs, which actively remold TME and sustain a cancer permissive state. Conversely, CAFs‐derived exosomes also significantly affect tumor cells and facilitate tumor progression. 93 A number of studies have demonstrated that CAFs‐derived exosomes are involved in tumor cell proliferation, migration, metastasis, and drug resistance. 63 , 94 , 95 , 96 For example, fibroblast exosomes have been shown to enhance the survival and proliferation of pancreatic cancer cells, while inhibition of exosome release by GW4869 significantly reduces survival of cancer epithelial cells. 94 Hu et al found that exosomes secreted by CAFs are transferred to CRC cells and lead to overexpression of miR‐92a‐3p, activating Wnt/β‐catenin pathway and inhibiting mitochondrial apoptosis, thus promoting metastasis and chemotherapy resistance. 63 In another study, exosomal miR‐522 derived from CAFs suppresses ALOX15 expression and lipid‐ROS accumulation in cancer cells, decreasing chemo‐sensitivity and inhibiting ferroptosis, a novel mode of non‐apoptotic cell death. 95 CAFs‐derived exosomes have also been shown to contain long non‐coding RNAs (lncRNAs) that promote the stemness and chemoresistance of CRC. 96

3.3. Macrophage‐derived exosomes

Macrophages within the TME play a critical role in cancer development, which promote cancer initiation and progression by facilitating cancer cell proliferation, migration, angiogenesis, and immunosuppression. 97 Macrophage‐derived exosomes orchestrate their communication with cancer cells, significantly affecting the malignant progression.

It has been shown that M2 macrophage‐derived exosomes transfer miR‐21‐5p and miR‐155‐5p to CRC cells, inducing cancer cell migration and invasion. 98 In addition, exosomes released by macrophages promote progression of PDAC by targeting transforming growth factor beta receptor 3 (TGFBR3), a tumor suppressor gene, and activating TGF‐β signaling pathway. 99 It has also been found that M2 macrophage‐derived exosomes contribute to cisplatin resistance in GC cells through delivery of miR‐21, which activates PI3K/AKT signaling pathway and inhibits cell apoptosis. 76 In another study, the authors demonstrated that macrophage‐derived exosomes transfer functional apolipoprotein E to GC cells and promote the migration of cancer cells. 100

4. APPLICATION OF EXOSOMES IN DIAGNOSIS

In most cases, the poor therapeutic response and prognosis in cancer patients are due to the late detection of diseases, which is largely attributed to the lack of appropriate biomarkers that serve as efficient diagnostic tools. 101 Therefore, considerable attention has been drawn to identifying and validating disease‐specific molecules that could be utilized for cancer diagnosis and prediction (Figure 3). Exosomes containing various proteins, lipids, nucleic acids, and metabolites are considered as the rich source of potential biomarkers with unparalleled advantages. 102 First, almost all cells secrete exosomes, and the multiple cargos carried by exosomes reflect the physiological status of their parental cells. 103 Second, apart from being secreted by different cells in vivo, exosomes also widely exist in various body fluids, such as blood, urine, and saliva. 103 Thus, exosomes are ideal noninvasive biomarkers and easily accessible for clinical detection. Third, due to the unique structure of exosomes, proteins and nucleic acids could be well protected by the lipid bilayer, and important information about disease characteristics is preserved. 101

miRNAs are one of the biological cargos in exosomes that participate in multiple aspects of cancer progression. Emerging evidence suggests that miRNAs are potential biomarkers for cancer diagnosis and prediction due to their stable properties and extreme abundance in exosomes (Table 3). 19 Importantly, with the development of specific and sensitive quantitative techniques, exosomal miRNAs can be detected in small volume samples with high accuracy. 103 Clinical studies have shown that miR‐126, a tumor suppressor and metastasis inhibitor, is downregulated in NSCLC patients. Additionally, miR‐126 is equally distributed between exosomes and exosome‐free serum fractions in healthy people while mainly present in exosomes of NSCLC patients, suggesting the potential of miR‐126 as a reliable disease biomarker. 104 Furthermore, Jin et al have identified AC‐ and SCC‐specific miRNAs as diagnostic biomarkers for early stage NSCLC by using next‐generation sequencing. The validated miRNAs with diagnostic accuracy such as AC‐specific miR‐181‐5p, miR‐30a‐3p, miR‐30e‐3p, and miR‐361‐5p, and SCC‐specific miR‐10b‐5p, miR‐15b‐5p, and miR‐320b could be promising indicators in the diagnosis of NSCLC. 105 It has also been reported that the expression level of exosomal miR‐25‐3p is significantly elevated in CRC patients with metastasis than those without metastasis. Mechanism analysis has shown that miR‐25‐3p regulates the expression of VEGFR2, ZO‐1, occludin, and Claudin5 in endothelial cells by targeting KLF2 and KLF4, promoting angiogenesis and pre‐metastatic niche formation. 68 Interestingly, Cheng et al have demonstrated that the secretion of colorectal CSC exosomes carrying biomarker miR‐146a‐5p is dependent on the expression of RAB27B by activation of β‐catenin/Tcf‐4. 106 In addition, Xue et al identified the differentially expressed exosomal miRNAs in cholangiocarcinoma and gallbladder carcinoma patients with high‐throughput small RNA sequencing. After a larger‐scale validation, they found that miR‐96‐5p, miR‐151a‐5p, miR‐191‐5p, and miR‐4732‐3p were significantly increased in the exosomes of cholangiocarcinoma patients, while miR‐151a‐5p was increased in the exosomes of gallbladder carcinoma patients. 107 In summary, although a multitude of miRNAs have been identified and validated as potential diagnostic markers for cancer in the basic research studies, much clinical data are also required to confirm their diagnostic effects. 19

TABLE 3.

Exosomal diagnostic biomarkers for detection of tumor progression

Cancer type Biomarker Source of exosomes Potential mechanisms Isolation methods Reference
Non‐small cell lung cancer (NSCLC) miR‐126↓ Serum Suppress tumorigenic function by targeting EGFL7 Ultracentrifugation 104
Non‐small cell lung cancer‐ adenocarcinoma (NSCLC‐AC)

miR‐181‐5p↑

miR‐30a‐3p↓

miR‐30e‐3p↓

miR‐361‐5p↑

Plasma Not mentioned Ultracentrifugation and immunoaffinity magnetic beads 105
Non‐small cell lung cancer‐ squamous cell carcinoma (NSCLC‐SCC)

miR‐10b‐5p↓

miR‐15b‐5p↓

miR‐320b↑

Plasma Not mentioned Ultracentrifugation and immunoaffinity magnetic beads 105
Colorectal cancer (CRC) miR‐25‐3p↑ CRC cells Regulate VEGFR2, ZO‐1, occludin and Claudin5 by targeting KLF2 and KLF4 Ultracentrifugation 68
Colorectal cancer (CRC) miR‐146a‐5p↑ CRC stem cells Target Numb in recipient CRC cells Ultracentrifugation 106
Cholangiocarcinoma (CCA)

miR‐96‐5p↑

miR‐151a‐5p↑

miR‐191‐5p↑

miR‐4732‐3p↑

Plasma Not mentioned exoEasy maxi kit (QIAGEN) 107
Bladder cancer (BC) lncRNA‐UCA1↑ 5637 cells and serum Promote tumor growth and progression through EMT Ultracentrifugation 108
Bladder cancer (BC) lncRNA PTENP1↓ Plasma Mediate PTEN expression by competitively binding to microRNA‐17, inhibit cell migration, invasion and induce apoptosis Exoquick exosome precipitation solution (System Biosciences) 109
Bladder cancer (BC)

MALAT1↑,

PCAT‐1↑

SPRY4‐IT1↑

Urine Not mentioned Not mentioned 110
Gastric cancer (GC)

lncUEGC1↑

lncUEGC2↑

Plasma, epithelial cells; GC cells Not mentioned Ultracentrifugation and discontinuous iodixanol gradient methods. 111
Gastric cancer (GC) lncRNA HOTTIP↑ Serum Positively associate with invasion depth and TNM stage Ultracentrifugation 112
Glioblastoma (GBM) lncSBF2‐AS1↑ GBM cells Confer temozolomide chemoresistance Ultracentrifugation 113
Pancreatic ductal adenocarcinomas (PDAC) circ‐PDE8A↑ PDAC cells Promote PDAC cells invasion by upregulating MET TRIzol LS (Thermo, Carlsbad, CA) 116
Pancreatic ductal adenocarcinomas (PDAC) circ‐IARS↑ PDAC cells Regulate endothelial monolayer permeability Total Exosome Isolation Kit (Thermo, Carlsbad, CA) 117
Gastric cancer (GC) circ‐KIAA1244↓ GC tissues, plasmas, and cells Not mentioned Hieff Quick exosome isolation kit (for serum/plasma, Shanghai, 41202ES30) 118
Gastric cancer (GC) circNRIP1↑ GC cells Via the AKT1/mTOR pathway, function as a microRNA‐149‐5p sponge Exosome isolation reagents (4,478,359, Invitrogen) 119
Head and neck squamous cell carcinomas (HNSCCs) PD‐L1↑ Plasma Induce immune escape and lead to immunotherapy resistance Ultracentrifugation 120
Pancreatic cancer (PC) ZIP4↑ PC cells Enhance cell proliferation, migration and invasion abilities SBI ExoQuick‐TC Kit (System Biosciences, Mountain View, CA) 121

Arrow ↑/↓ represents the increased/decreased expression of biomarker.

In addition to miRNAs, lncRNAs are also attractive candidates in the development of sensitive and specific biomarkers for cancer diagnosis and prediction. These lncRNAs are enriched in exosomes and stable in blood, reflecting the cell status that they origin. Additionally, the different expression level of lncRNAs in tumor tissues and healthy organs suggests their potential clinical application as cancer‐specific biomarkers. For instance, the expression level of lncRNA‐UCA1 is significantly higher in exosomes of BC patients than in healthy donors and could be used for the clinical detection of BC. 108 While in another study, Zheng et al found that lncRNA PTENP1 was significantly reduced in BC tissues, and normal cell derived‐exosomal PTENP1 could be transferred to cancer cells, impeding cancer progression by inhibiting cell migration, invasion, and inducing apoptosis. 109 Moreover, reports have established that a urinary exosome‐derived lncRNA panel (MALAT1, PCAT‐1, and SPRY4‐IT1) could be used as noninvasive diagnostic and prognostic biomarker of BC with considerable clinical value. The upregulation of PCAT‐1 and MALAT1 indicates poor recurrence‐free survival. 110 Tumor‐originated exosomal lncUEGC1 is expressed at a high level in stage I GC patients and GC cells. Notably, it exhibits higher diagnostic accuracy than carcinoembryonic antigen in discriminating early GC patients from healthy individuals and late stage patients. 111 In addition, Zhao el al demonstrated that the expression level of lncRNA HOTTIP is significantly correlated with invasion depth, tumor‐node‐metastasis (TNM) stage, and overall survival of GC patients, functioning as an independent prognostic factor. 112 Recent studies have reported that exosomal lncSBF2‐AS1 confers chemoresistance to glioblastoma cells, and high levels of lncSBF2‐AS1 are associated with poor response to temozolomide treatment in glioblastoma patients. 113

In recent years, circular RNAs (circRNAs) are emerged as novel biomarkers for cancer diagnosis due to their universal existence in eukaryotic cells and tissue‐ and developmental stage‐specific expression patterns. 114 Accumulating evidence indicates that circRNAs are regulators of parental gene transcription and exhibit diverse biological functions in the progression of cancer. 115 Exosomal circ‐PDE8A derived from plasma of PDAC patients has been reported to promote tumor invasion by upregulating MET (MET proto‐oncogene, receptor tyrosine kinase). The expression level of circ‐PDE8A is positively correlated with lymphatic invasion, TNM stage and negatively correlated with survival rate, acting as a promising indicator for PDAC diagnosis and prognosis. 116 In addition, Li et al found that circ‐IARS secreted by PDAC tumor cells could be transferred to human microvascular vein endothelial (HUVECs) cells and facilitate tumor invasion and metastasis. 117 By screening circRNA expression profiles for GC, Tang and colleagues identified a novel circulating biomarker circ‐KIAA1244 in the detection of GC. Clinical data analysis demonstrated that low expression level of circ‐KIAA1244 indicated lymphatic metastasis and a shorter overall survival time. 118 A recent study revealed that circNRIP1 delivered by exosomes promoted GC progression via the AKT1/mTOR pathway, functioning as a microRNA‐149‐5p sponge. 119

Finally, proteins within exosomes also possess clinical potential as diagnostic markers. It has been reported that PD‐L1 levels in exosomes are correlated with progression of head and neck SCCs (HNSCCs), and exosomal PD‐L1 is involved in inducing immune escape that leads to resistance of immunotherapy. 120 Studies have demonstrated that ZIP4, a membrane‐located zinc ion transporter, could be carried by exosomes to tumor cells and promotes pancreatic cancer growth. The expression level of ZIP4 varies in different stages of pancreatic cancer, and high ZIP4 expression is correlated with worse survival rates. 121 Although numerous basic studies have shown strong relationship between exosome‐associated proteins and cancer progression, the high heterogeneity of exosome‐associated proteins and the limited isolation methods significantly hinder their clinical practice. In addition, further clinical research are also required to verify their application in the diagnosis of cancer or monitoring during treatment. 122

Although exosomes serving as biomarkers hold great promise in cancer diagnosis and prognosis prediction, most of research is currently in the early stage of exploration and lacks of clinical practice. 103 In addition, given the predominance of non‐tumor derived exosomes in human body fluids such as platelet‐derived exosomes in blood plasma, the accurate separation, identification, and purification of exosomes are still facing considerable challenges. Further research on exosomal proteomics and novel detection methods will undoubtedly accelerate the clinical application of exosomes in cancer diagnosis. 123

5. APPLICATION OF EXOSOMES IN THERAPY

5.1. Exosome‐based immunotherapy

In recent years, immunotherapy is emerged as critically important foci in the field of cancer research, and exosomes have attracted enormous attention in the development of novel immunotherapeutic modalities. 124 Exosomes are proved to be vital mediators of immune regulation in TME. 125 It has been showed that exosomes play significant roles in transfer of antigen and signals to antigen‐presenting cells directly or indirectly, thereby promoting the activation of immunity. 43 What's more, the intrinsic immunomodulatory capability of exosomes endows them with high clinical application serving as cancer therapeutic vaccines that are able to induce adaptive and innate immunosurveillance. 124 An impressive study by Lu et al revealed that exosomes derived from α‐fetoprotein expressing dendritic cells (DEXAFP) could trigger potent antigen‐specific antitumor immune responses and result in significant tumor growth retardation. Moreover, DEXAFP improved the TME and prolonged the survival rates in three different HCC mouse models with antigenic and pathological heterogeneity. 126 Importantly, the loading of exosomes to DCs can augment the vaccine efficacy, which is even superior to lysate‐loaded DCs. 127 In addition, the combination of exosome‐based therapeutic cancer vaccines with other therapeutic modalities, such as chemotherapy and photothermal therapy, may enhance the efficacy of immunotherapy and exert stronger antitumor effect. Morishita and colleagues designed an exosome‐based tumor antigens‐adjuvant co‐delivery system that exosomes were modified with CpG DNA (CpG‐SAV‐exo). CpG‐SAV‐exo induced tumor antigen‐specific immune response more effectively than simple coadministration of exosomes and CpG DNA and significantly inhibited tumor growth. 128

5.2. Exosome‐based drug delivery

With momentous progress in elucidating the biological and functional properties of exosomes, these natural nano‐vehicles with good biocompatibility, stability, and safety are recognized as compelling candidate platforms for drug delivery. 125 Meanwhile, exosomes display exquisite target‐homing specificity and can be preferentially transferred to tumor cells with reduced side effects. 122 Exosome‐based nanoplatforms have been employed to encapsulate and transfer multiple functional molecules in cancer treatment, such as chemotherapeutic drugs, proteins, and nucleic acids, and yielded encouraging preclinical results. 124 For instance, doxorubicin has been successfully encapsulated in exosomes to ameliorate their biodistribution. 129 Exosomes can also be deployed in delivering natural phytochemical compounds that with poor solubility, such as curcumin, to improve their bioavailability and accelerate their clinical translation. 130 Recently, with the rapid development of gene therapy, exosomes hold great promise in the delivery of nucleic acid macromolecules. Kamerkar et al developed an exosome­based nanoplatform carrying short interfering RNA or short hairpin RNA for specific targeting of oncogenic KRAS. The exosomal formulation displayed enhanced circulating stability due to CD47‐mediated protection from phagocytosis. 131

6. CONCLUSION

The past two decades have witnessed the admirable progress of exosomal science and therapeutics. These natural cell‐derived vesicles possessing unique structural characteristics are regarded as key mediators in intercellular communications and play an important role in cancer progression. The thorough understanding of these specific mechanisms by which exosome‐based communication regulates the malignant behavior of cancer cells and stromal cells in TME can make substantial contributions to the development of next‐generation exosomal therapies. In addition, as one of the most distinguishing hallmarks of exosomes, the variable cargo contents within exosomes carrying abundant, specific, and sensitive biological information are emerged as valid biomarkers in cancer diagnosis and prognosis. The enticing results of exosome based‐therapeutics and diagnosis in experimental settings have granted them a fast‐tracking into clinic.

With the rapid development of nanotechnology, exosomes are heralded as attractive candidates in the establishment of novel nanoplatforms for cancer treatment. Compared with conventional synthetic nanocarriers, these exosomal nanoformulations display fascinating advantages in drug delivery and are being rampantly developed. However, some obstacles still lay ahead for the optimal translation of these promising results into clinic, such as the technical challenges of exosome isolation, purification, and identification; and tremendous efforts are required to overcome the existing limitations to achieve clinical benefit.

CONFLICT OF INTEREST

The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of the research reported.

AUTHOR CONTRIBUTIONS

Wen‐Jie Gu and Yi‐Wen Shen searched the literature and wrote the manuscript. Li‐Jun Zhang and Hong Zhang gave some supplements of current literature. Dale G. Nagle checked the grammar and polished the language. Xin Luan and San‐Hong Liu made conceptual revision and checked the final paper to be published.

ETHICS APPROVAL STATEMENT

Not applicable.

ACKNOWLEDGMENTS

This work was supported by National Natural Science Foundation of China (grant numbers: 81772798 and 81903654), Program for Professor of Special Appointment (Young Eastern Scholar) at Shanghai Institutions of Higher Learning (QD2018035), Shanghai “Chenguang Program” of Education Commission of Shanghai Municipality (grant number: 18CG46), Shanghai Sailing Program (grant number: 19YF1449400), Ruijin Youth NSFC Cultivation Fund (KY20194297), and Shanghai “Super Postdoctoral Fellow” incentive program (grant number: 2019334).

Gu W‐J, Shen Y‐W, Zhang L‐J, et al. The multifaceted involvement of exosomes in tumor progression: Induction and inhibition. MedComm. 2021;2:297–314. 10.1002/mco2.49

Contributor Information

Xin Luan, Email: luanxin@shutcm.edu.cn.

San‐Hong Liu, Email: liush@shutcm.edu.cn.

DATA AVAILABILITY STATEMENT

Not applicable.

REFERENCES

  • 1. Hessvik NP, Llorente A. Current knowledge on exosome biogenesis and release. Cell Mol Life Sci. 2018;75(2):193‐208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Meng W, Hao Y, He C, et al. Exosome‐orchestrated hypoxic tumor microenvironment. Mol Cancer. 2019;18(1):57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Théry C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and function. Nat Rev Immunol. 2002;2(8):569‐579. [DOI] [PubMed] [Google Scholar]
  • 4. van Niel G, D'Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol. 2018;19(4):213‐228. [DOI] [PubMed] [Google Scholar]
  • 5. Fan Q, Yang L, Zhang X, et al. The emerging role of exosome‐derived non‐coding RNAs in cancer biology. Cancer Lett. 2018;414:107‐115. [DOI] [PubMed] [Google Scholar]
  • 6. Li K, Chen Y, Li A, et al. Exosomes play roles in sequential processes of tumor metastasis. Int J Cancer. 2019;144(7):1486‐1495. [DOI] [PubMed] [Google Scholar]
  • 7. Mashouri L, Yousefi H, Aref AR, et al. Exosomes: composition, biogenesis, and mechanisms in cancer metastasis and drug resistance. Mol Cancer. 2019;18(1):75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Greening DW, Gopal SK, Mathias RA, et al. Emerging roles of exosomes during epithelial‐mesenchymal transition and cancer progression. Semin Cell Dev Biol. 2015;40:60‐71. [DOI] [PubMed] [Google Scholar]
  • 9. Xu R, Rai A, Chen M, et al. Extracellular vesicles in cancer ‐ implications for future improvements in cancer care. Nat Rev Clin Oncol. 2018;15(10):617‐638. [DOI] [PubMed] [Google Scholar]
  • 10. Han L, Lam EWF, Sun Y. Extracellular vesicles in the tumor microenvironment: old stories, but new tales. Mol Cancer. 2019;18(1):59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Li X, Wang Y, Wang Q, et al. Exosomes in cancer: small transporters with big functions. Cancer Lett. 2018;435:55‐65. [DOI] [PubMed] [Google Scholar]
  • 12. Wu K, Xing F, Wu SY, et al. Extracellular vesicles as emerging targets in cancer: recent development from bench to bedside. Biochim Biophys Acta Rev Cancer. 2017;1868(2):538‐563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Luan X, Sansanaphongpricha K, Myers I, et al. Engineering exosomes as refined biological nanoplatforms for drug delivery. Acta pharmacologica Sinica. 2017;38(6):754‐763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Todorova D, Simoncini S, Lacroix R, et al. Extracellular vesicles in angiogenesis. Circ Res. 2017;120(10):1658‐1673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Viallard C, Larrivée B. Tumor angiogenesis and vascular normalization: alternative therapeutic targets. Angiogenesis. 2017;20(4):409‐426. [DOI] [PubMed] [Google Scholar]
  • 16. Yang H, Zhang H, Ge S, et al. Exosome‐derived miR‐130a activates angiogenesis in gastric cancer by targeting C‐MYB in vascular endothelial cells. Mol Ther. 2018;26(10):2466‐2475. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 17. Wu F, Li F, Lin X, et al. Exosomes increased angiogenesis in papillary thyroid cancer microenvironment. Endocr Relat Cancer. 2019;26(5):525‐538. [DOI] [PubMed] [Google Scholar]
  • 18. He L, Zhu W, Chen Q, et al. Ovarian cancer cell‐secreted exosomal miR‐205 promotes metastasis by inducing angiogenesis. Theranostics. 2019;9(26):8206‐8220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Maji S, Chaudhary P, Akopova I, et al. Exosomal annexin II promotes angiogenesis and breast cancer metastasis. Mol Cancer Res. 2017;15(1):93‐105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Hsu YL, Hung JY, Chang WA, et al. Hypoxic lung cancer‐secreted exosomal miR‐23a increased angiogenesis and vascular permeability by targeting prolyl hydroxylase and tight junction protein ZO‐1. Oncogene. 2017;36(34):4929‐4942. [DOI] [PubMed] [Google Scholar]
  • 21. Li J, Yuan H, Xu H, et al. Hypoxic cancer‐secreted exosomal miR‐182‐5p promotes glioblastoma angiogenesis by targeting kruppel‐like factor 2 and 4. Mol Cancer Res. 2020;18(8):1218‐1231. [DOI] [PubMed] [Google Scholar]
  • 22. Wang Z, Liao F, Wu H, et al. Glioma stem cells‐derived exosomal miR‐26a promotes angiogenesis of microvessel endothelial cells in glioma. J Exp Clin Cancer Res. 2019;38(1):201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Wu XG, Zhou CF, Zhang YM, et al. Cancer‐derived exosomal miR‐221‐3p promotes angiogenesis by targeting THBS2 in cervical squamous cell carcinoma. Angiogenesis. 2019;22(3):397‐410. [DOI] [PubMed] [Google Scholar]
  • 24. Zhou Z, Zhang H, Deng T, et al. Exosomes carrying microRNA‐155 target forkhead box O3 of endothelial cells and promote angiogenesis in gastric cancer. Mol Ther Oncolytics. 2019;15:223‐233. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 25. Bai M, Li J, Yang H, et al. miR‐135b delivered by gastric tumor exosomes inhibits FOXO1 expression in endothelial cells and promotes angiogenesis. Mol Ther. 2019;27(10):1772‐1783. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 26. Du J, Liang Y, Li J, et al. Gastric cancer cell‐derived exosomal microRNA‐23a promotes angiogenesis by targeting PTEN. Front Oncol. 2020;10:326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Deng T, Zhang H, Yang H, et al. Exosome miR‐155 derived from gastric carcinoma promotes angiogenesis by targeting the c‐MYB/VEGF axis of endothelial cells. Mol Ther Nucleic Acids. 2020;19:1449‐1459. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 28. Shang A, Wang X, Gu C, et al. Exosomal miR‐183‐5p promotes angiogenesis in colorectal cancer by regulation of FOXO1. Aging (Albany NY). 2020;12(9):8352‐8371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Hu HY, Yu CH, Zhang HH, et al. Exosomal miR‐1229 derived from colorectal cancer cells promotes angiogenesis by targeting HIPK2. Int J Biol Macromol. 2019;132:470‐477. [DOI] [PubMed] [Google Scholar]
  • 30. Xie JY, Wei JX, Lv LH, et al. Angiopoietin‐2 induces angiogenesis via exosomes in human hepatocellular carcinoma. Cell Commun Signal. 2020;18(1):46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Li R, Wang Y, Zhang X, et al. Exosome‐mediated secretion of LOXL4 promotes hepatocellular carcinoma cell invasion and metastasis. Mol Cancer. 2019;18(1):18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Zhou Q, Zhu Y, Wei X, et al. MiR‐590‐5p inhibits colorectal cancer angiogenesis and metastasis by regulating nuclear factor 90/vascular endothelial growth factor A axis. Cell Death Dis. 2016;7(10):e2413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Rosenberger L, Ezquer M, Lillo‐Vera F, et al. Stem cell exosomes inhibit angiogenesis and tumor growth of oral squamous cell carcinoma. Sci Rep. 2019;9(1):663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Lopatina T, Grange C, Fonsato V, et al. Extracellular vesicles from human liver stem cells inhibit tumor angiogenesis. Int J Cancer. 2019;144(2):322‐333. [DOI] [PubMed] [Google Scholar]
  • 35. Hannafon BN, Carpenter KJ, Berry WL, et al. Exosome‐mediated microRNA signaling from breast cancer cells is altered by the anti‐angiogenesis agent docosahexaenoic acid (DHA). Mol Cancer. 2015;14:133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Ishii G, Ochiai A, Neri S. Phenotypic and functional heterogeneity of cancer‐associated fibroblast within the tumor microenvironment. Adv Drug Deliver Rev. 2016;99(Pt B):186‐196. [DOI] [PubMed] [Google Scholar]
  • 37. Paggetti J, Haderk F, Seiffert M, et al. Exosomes released by chronic lymphocytic leukemia cells induce the transition of stromal cells into cancer‐associated fibroblasts. Blood. 2015;126(9):1106‐1117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Ringuette Goulet C, Bernard G, Tremblay S, et al. Exosomes induce fibroblast differentiation into cancer‐associated fibroblasts through TGFβ signaling. Mol Cancer Res. 2018;16(7):1196‐1204. [DOI] [PubMed] [Google Scholar]
  • 39. Zhou Y, Ren H, Dai B, et al. Hepatocellular carcinoma‐derived exosomal miRNA‐21 contributes to tumor progression by converting hepatocyte stellate cells to cancer‐associated fibroblasts. J Exp Clin Cancer Res. 2018;37(1):324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Zhou X, Yan T, Huang C, et al. Melanoma cell‐secreted exosomal miR‐155‐5p induce proangiogenic switch of cancer‐associated fibroblasts via SOCS1/JAK2/STAT3 signaling pathway. J Exp Clin Cancer Res. 2018;37(1):242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Li YY, Tao YW, Gao S, et al. Cancer‐associated fibroblasts contribute to oral cancer cells proliferation and metastasis via exosome‐mediated paracrine miR‐34a‐5p. EBioMedicine. 2018;36:209‐220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Chen X, Liu J, Zhang Q, et al. Exosome‐mediated transfer of miR‐93‐5p from cancer‐associated fibroblasts confer radioresistance in colorectal cancer cells by downregulating FOXA1 and upregulating TGFB3. J Exp Clin Cancer Res. 2020;39(1):65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Robbins PD, Morelli AE. Regulation of immune responses by extracellular vesicles. Nat Rev Immunol. 2014;14(3):195‐208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Whiteside TL. Tumor‐derived exosomes and their role in tumor‐induced immune suppression. Vaccines (Basel). 2016;4(4):35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Whiteside TL. Exosomes and tumor‐mediated immune suppression. J Clin Invest. 2016;126(4):1216‐1223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Xie F, Xu M, Lu J, et al. The role of exosomal PD‐L1 in tumor progression and immunotherapy. Mol Cancer. 2019;18(1):146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Poggio M, Hu T, Pai CC, et al. Suppression of exosomal PD‐L1 induces systemic anti‐tumor immunity and memory. Cell. 2019;177(2):414‐427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Chen G, Huang AC, Zhang W, et al. Exosomal PD‐L1 contributes to immunosuppression and is associated with anti‐PD‐1 response. Nature. 2018;560(7718):382‐386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Daassi D, Mahoney KM, Freeman GJ. The importance of exosomal PDL1 in tumour immune evasion. Nat Rev Immunol. 2020;20(4):209‐215. [DOI] [PubMed] [Google Scholar]
  • 50. Kim DH, Kim H, Choi YJ, et al. Exosomal PD‐L1 promotes tumor growth through immune escape in non‐small cell lung cancer. Exp Mol Med. 2019;51(8):1‐13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Han Q, Zhao H, Jiang Y, et al. HCC‐derived exosomes: critical player and target for cancer immune escape. Cells‐Basel. 2019;8(6):588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Ye L, Zhang Q, Cheng Y, et al. Tumor‐derived exosomal HMGB1 fosters hepatocellular carcinoma immune evasion by promoting TIM‐1(+) regulatory B cell expansion. J Immunother Cancer. 2018;6(1):145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Vignard V, Labbé M, Marec N, et al. MicroRNAs in tumor exosomes drive immune escape in melanoma. Cancer Immunol Res. 2020;8(2):255‐267. [DOI] [PubMed] [Google Scholar]
  • 54. Haderk F, Schulz R, Iskar M, et al. Tumor‐derived exosomes modulate PD‐L1 expression in monocytes. Sci Immunol. 2017;2(13):eaah5509. [DOI] [PubMed] [Google Scholar]
  • 55. Zhang X, Sai B, Wang F, et al. Hypoxic BMSC‐derived exosomal miRNAs promote metastasis of lung cancer cells via STAT3‐induced EMT. Mol Cancer. 2019;18(1):40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Wang L, Yang G, Zhao D, et al. CD103‐positive CSC exosome promotes EMT of clear cell renal cell carcinoma: role of remote MiR‐19b‐3p. Mol Cancer. 2019;18(1):86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Xiao D, Barry S, Kmetz D, et al. Melanoma cell‐derived exosomes promote epithelial‐mesenchymal transition in primary melanocytes through paracrine/autocrine signaling in the tumor microenvironment. Cancer Lett. 2016;376(2):318‐327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Chen L, Guo P, He Y, et al. HCC‐derived exosomes elicit HCC progression and recurrence by epithelial‐mesenchymal transition through MAPK/ERK signalling pathway. Cell Death Dis. 2018;9(5):513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Wei JX, Lv LH, Wan YL, et al. Vps4A functions as a tumor suppressor by regulating the secretion and uptake of exosomal microRNAs in human hepatoma cells. Hepatology. 2015;61(4):1284‐1294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Han Q, Lv L, Wei J, et al. Vps4A mediates the localization and exosome release of β‐catenin to inhibit epithelial‐mesenchymal transition in hepatocellular carcinoma. Cancer Lett. 2019;457:47‐59. [DOI] [PubMed] [Google Scholar]
  • 61. Fiori ME, Di Franco S, Villanova L, et al. Cancer‐associated fibroblasts as abettors of tumor progression at the crossroads of EMT and therapy resistance. Mol Cancer. 2019;18(1):70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Yeon JH, Jeong HE, Seo H, et al. Cancer‐derived exosomes trigger endothelial to mesenchymal transition followed by the induction of cancer‐associated fibroblasts. Acta Biomater. 2018;76:146‐153. [DOI] [PubMed] [Google Scholar]
  • 63. Hu JL, Wang W, Lan XL, et al. CAFs secreted exosomes promote metastasis and chemotherapy resistance by enhancing cell stemness and epithelial‐mesenchymal transition in colorectal cancer. Mol Cancer. 2019;18(1):91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Wang H, Wei H, Wang J, et al. MicroRNA‐181d‐5p‐containing exosomes derived from CAFs promote EMT by regulating CDX2/HOXA5 in breast cancer. Mol Ther Nucleic Acids. 2020;19:654‐667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Liu Y, Cao X. Characteristics and significance of the pre‐metastatic niche. Cancer Cell. 2016;30(5):668‐681. [DOI] [PubMed] [Google Scholar]
  • 66. Lobb RJ, Lima LG, Möller A. Exosomes: key mediators of metastasis and pre‐metastatic niche formation. Semin Cell Dev Biol. 2017;67:3‐10. [DOI] [PubMed] [Google Scholar]
  • 67. Costa‐Silva B, Aiello NM, Ocean AJ, et al. Pancreatic cancer exosomes initiate pre‐metastatic niche formation in the liver. Nat Cell Biol. 2015;17(6):816‐826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Zeng Z, Li Y, Pan Y, et al. Cancer‐derived exosomal miR‐25‐3p promotes pre‐metastatic niche formation by inducing vascular permeability and angiogenesis. Nat Commun. 2018;9(1):5395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Liu Y, Gu Y, Han Y, et al. Tumor exosomal RNAs promote lung pre‐metastatic niche formation by activating alveolar epithelial TLR3 to recruit neutrophils. Cancer Cell. 2016;30(2):243‐256. [DOI] [PubMed] [Google Scholar]
  • 70. García‐Silva S, Peinado H. Melanosomes foster a tumour niche by activating CAFs. Nat Cell Biol. 2016;18(9):911‐913. [DOI] [PubMed] [Google Scholar]
  • 71. Plebanek MP, Angeloni NL, Vinokour E, et al. Pre‐metastatic cancer exosomes induce immune surveillance by patrolling monocytes at the metastatic niche. Nat Commun. 2017;8(1):1319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Peinado H, Alečković M, Lavotshkin S, et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro‐metastatic phenotype through MET. Nat Med. 2012;18(6):883‐891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Hoshino A, Costa‐Silva B, Shen TL, et al. Tumour exosome integrins determine organotropic metastasis. Nature. 2015;527(7578):329‐335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Namee NM, O'Driscoll L. Extracellular vesicles and anti‐cancer drug resistance. Biochim Biophys Acta Rev Cancer. 2018;1870(2):123‐136. [DOI] [PubMed] [Google Scholar]
  • 75. Steinbichler TB, Dudás J, Skvortsov S, et al. Therapy resistance mediated by exosomes. Mol Cancer. 2019;18(1):58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Zheng P, Chen L, Yuan X, et al. Exosomal transfer of tumor‐associated macrophage‐derived miR‐21 confers cisplatin resistance in gastric cancer cells. J Exp Clin Cancer Res. 2017;36(1):53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Liu T, Zhang X, Du L, et al. Exosome‐transmitted miR‐128‐3p increase chemosensitivity of oxaliplatin‐resistant colorectal cancer. Mol Cancer. 2019;18(1):43. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 78. Zeng A, Wei Z, Yan W, et al. Exosomal transfer of miR‐151a enhances chemosensitivity to temozolomide in drug‐resistant glioblastoma. Cancer Lett. 2018;436:10‐21. [DOI] [PubMed] [Google Scholar]
  • 79. Yin J, Zeng A, Zhang Z, et al. Exosomal transfer of miR‐1238 contributes to temozolomide‐resistance in glioblastoma. EBioMedicine. 2019;42:238‐251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Fu X, Liu M, Qu S, et al. Exosomal microRNA‐32‐5p induces multidrug resistance in hepatocellular carcinoma via the PI3K/Akt pathway. J Exp Clin Cancer Res. 2018;37(1):52. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 81. Fan J, Wei Q, Koay EJ, et al. Chemoresistance transmission via exosome‐mediated ephA2 transfer in pancreatic cancer. Theranostics. 2018;8(21):5986‐5994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Dorayappan KDP, Wanner R, Wallbillich JJ, et al. Hypoxia‐induced exosomes contribute to a more aggressive and chemoresistant ovarian cancer phenotype: a novel mechanism linking STAT3/Rab proteins. Oncogene. 2018;37(28):3806‐3821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Kanlikilicer P, Bayraktar R, Denizli M, et al. Exosomal miRNA confers chemo resistance via targeting Cav1/p‐gp/M2‐type macrophage axis in ovarian cancer. EBioMedicine. 2018;38:100‐112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Yang Y, Li CW, Chan LC, et al. Exosomal PD‐L1 harbors active defense function to suppress T cell killing of breast cancer cells and promote tumor growth. Cell Res. 2018;28(8):862‐864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Whiteside TL. Exosome and mesenchymal stem cell cross‐talk in the tumor microenvironment. Semin Immunol. 2018;35:69‐79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Vakhshiteh F, Atyabi F, Ostad SN. Mesenchymal stem cell exosomes: a two‐edged sword in cancer therapy. Int J Nanomed. 2019;14:2847‐2859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Figueroa J, Phillips LM, Shahar T, et al. Exosomes from glioma‐associated mesenchymal stem cells increase the tumorigenicity of glioma stem‐like cells via transfer of miR‐1587. Cancer Res. 2017;77(21):5808‐5819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Khare D, Or R, Resnick I, et al. Mesenchymal stromal cell‐derived exosomes affect mRNA expression and function of B‐Lymphocytes. Front Immunol. 2018;9:3053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Wang S, Su X, Xu M, et al. Exosomes secreted by mesenchymal stromal/stem cell‐derived adipocytes promote breast cancer cell growth via activation of Hippo signaling pathway. Stem Cell Res Ther. 2019;10(1):117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Jiang S, Mo C, Guo S, et al. Human bone marrow mesenchymal stem cells‐derived microRNA‐205‐containing exosomes impede the progression of prostate cancer through suppression of RHPN2. J Exp Clin Cancer Res. 2019;38(1):495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Wu DM, Wen X, Han XR, et al. Bone marrow mesenchymal stem cell‐derived exosomal microRNA‐126‐3p inhibits pancreatic cancer development by targeting ADAM9. Mol Ther Nucleic Acids. 2019;16:229‐245. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 92. Xu H, Zhao G, Zhang Y, et al. Mesenchymal stem cell‐derived exosomal microRNA‐133b suppresses glioma progression via Wnt/β‐catenin signaling pathway by targeting EZH2. Stem Cell Res Ther. 2019;10(1):381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Li I, Nabet BY. Exosomes in the tumor microenvironment as mediators of cancer therapy resistance. Mol Cancer. 2019;18(1):32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Richards KE, Zeleniak AE, Fishel ML, et al. Cancer‐associated fibroblast exosomes regulate survival and proliferation of pancreatic cancer cells. Oncogene. 2017;36(13):1770‐1778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Zhang H, Deng T, Liu R, et al. CAF secreted miR‐522 suppresses ferroptosis and promotes acquired chemo‐resistance in gastric cancer. Mol Cancer. 2020;19(1):43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Ren J, Ding L, Zhang D, et al. Carcinoma‐associated fibroblasts promote the stemness and chemoresistance of colorectal cancer by transferring exosomal lncRNA H19. Theranostics. 2018;8(14):3932‐3948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Ngambenjawong C, Gustafson HH, Pun SH. Progress in tumor‐associated macrophage (TAM)‐targeted therapeutics. Adv Drug Deliver Rev. 2017;114:206‐221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Lan J, Sun L, Xu F, et al. M2 macrophage‐derived exosomes promote cell migration and invasion in colon cancer. Cancer Res. 2019;79(1):146‐158. [DOI] [PubMed] [Google Scholar]
  • 99. Yin Z, Ma T, Huang B, et al. Macrophage‐derived exosomal microRNA‐501‐3p promotes progression of pancreatic ductal adenocarcinoma through the TGFBR3‐mediated TGF‐β signaling pathway. J Exp Clin Cancer Res. 2019;38(1):310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Zheng P, Luo Q, Wang W, et al. Tumor‐associated macrophages‐derived exosomes promote the migration of gastric cancer cells by transfer of functional apolipoprotein E. Cell Death Dis. 2018;9(4):434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Milane L, Singh A, Mattheolabakis G, et al. Exosome mediated communication within the tumor microenvironment. J Control Release. 2015;219:278‐294. [DOI] [PubMed] [Google Scholar]
  • 102. He C, Zheng S, Luo Y, Wang B. Exosome theranostics: biology and translational medicine. Theranostics. 2018;8(1):237‐255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Kosaka N, Yoshioka Y, Fujita Y, et al. Versatile roles of extracellular vesicles in cancer. J Clin Invest. 2016;126(4):1163‐1172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Grimolizzi F, Monaco F, Leoni F, et al. Exosomal miR‐126 as a circulating biomarker in non‐small‐cell lung cancer regulating cancer progression. Sci Rep. 2017;7(1):15277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Jin X, Chen Y, Chen H, et al. Evaluation of tumor‐derived exosomal miRNA as potential diagnostic biomarkers for early‐stage non‐small cell lung cancer using next‐generation sequencing. Clin Cancer Res. 2017;23(17):5311‐5319. [DOI] [PubMed] [Google Scholar]
  • 106. Cheng WC, Liao TT, Lin CC, et al. RAB27B‐activated secretion of stem‐like tumor exosomes delivers the biomarker microRNA‐146a‐5p, which promotes tumorigenesis and associates with an immunosuppressive tumor microenvironment in colorectal cancer. Int J Cancer. 2019;145(8):2209‐2224. [DOI] [PubMed] [Google Scholar]
  • 107. Xue XY, Liu YX, Wang C, et al. Identification of exosomal miRNAs as diagnostic biomarkers for cholangiocarcinoma and gallbladder carcinoma. Signal Transduct Target Ther. 2020;5(1):77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Xue M, Chen W, Xiang A, et al. Hypoxic exosomes facilitate bladder tumor growth and development through transferring long non‐coding RNA‐UCA1. Mol Cancer. 2017;16(1):143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Zheng R, Du M, Wang X, et al. Exosome‐transmitted long non‐coding RNA PTENP1 suppresses bladder cancer progression. Mol Cancer. 2018;17(1):143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Zhan Y, Du L, Wang L, et al. Expression signatures of exosomal long non‐coding RNAs in urine serve as novel non‐invasive biomarkers for diagnosis and recurrence prediction of bladder cancer. Mol Cancer. 2018;17(1):142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Lin LY, Yang L, Zeng Q, et al. Tumor‐originated exosomal lncUEGC1 as a circulating biomarker for early‐stage gastric cancer. Mol Cancer. 2018;17(1):84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Zhao R, Zhang Y, Zhang X, et al. Exosomal long noncoding RNA HOTTIP as potential novel diagnostic and prognostic biomarker test for gastric cancer. Mol Cancer. 2018;17(1):68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Zhang Z, Yin J, Lu C, et al. Exosomal transfer of long non‐coding RNA SBF2‐AS1 enhances chemoresistance to temozolomide in glioblastoma. J Exp Clin Cancer Res. 2019;38(1):166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Bai H, Lei K, Huang F, et al. Exo‐circRNAs: a new paradigm for anticancer therapy. Mol Cancer. 2019;18(1):56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Li R, Jiang J, Shi H, et al. CircRNA: a rising star in gastric cancer. Cell Mol Life Sci. 2020;77(9):1661‐1680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Li Z, Yanfang W, Li J, et al. Tumor‐released exosomal circular RNA PDE8A promotes invasive growth via the miR‐338/MACC1/MET pathway in pancreatic cancer. Cancer Lett. 2018;432:237‐250. [DOI] [PubMed] [Google Scholar]
  • 117. Li J, Li Z, Jiang P, et al. Circular RNA IARS (circ‐IARS) secreted by pancreatic cancer cells and located within exosomes regulates endothelial monolayer permeability to promote tumor metastasis. J Exp Clin Cancer Res. 2018;37(1):177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Tang W, Fu K, Sun H, et al. CircRNA microarray profiling identifies a novel circulating biomarker for detection of gastric cancer. Mol Cancer. 2018;17(1):137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Zhang X, Wang S, Wang H, et al. Circular RNA circNRIP1 acts as a microRNA‐149‐5p sponge to promote gastric cancer progression via the AKT1/mTOR pathway. Mol Cancer. 2019;18(1):20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Theodoraki MN, Yerneni SS, Hoffmann TK, et al. Clinical significance of PD‐L1(+) exosomes in plasma of head and neck cancer patients. Clin Cancer Res. 2018;24(4):896‐905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Jin H, Liu P, Wu Y, et al. Exosomal zinc transporter ZIP4 promotes cancer growth and is a novel diagnostic biomarker for pancreatic cancer. Cancer Sci. 2018;109(9):2946‐2956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Kosaka N, Yoshioka Y, Fujita Y, et al. Versatile roles of extracellular vesicles in cancer. J Clin Invest. 2016;126(4):1163‐1172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Li W, Li C, Zhou T, et al. Role of exosomal proteins in cancer diagnosis. Mol Cancer. 2017;16(1):145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Yang B, Chen Y, Shi J. Exosome biochemistry and advanced nanotechnology for next‐generation theranostic platforms. Adv Mater. 2019;31(2):e1802896. [DOI] [PubMed] [Google Scholar]
  • 125. Wu Q, Zhou L, Lv D, et al. Exosome‐mediated communication in the tumor microenvironment contributes to hepatocellular carcinoma development and progression. J Hematol Oncol. 2019;12(1):53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Lu Z, Zuo B, Jing R, et al. Dendritic cell‐derived exosomes elicit tumor regression in autochthonous hepatocellular carcinoma mouse models. J Hepatol. 2017;67(4):739‐748. [DOI] [PubMed] [Google Scholar]
  • 127. Gu X, Erb U, Buchler MW, et al. Improved vaccine efficacy of tumor exosome compared to tumor lysate loaded dendritic cells in mice. Int J Cancer. 2015;136(4):E74‐E84. [DOI] [PubMed] [Google Scholar]
  • 128. Morishita M, Takahashi Y, Matsumoto A, et al. Exosome‐based tumor antigens‐adjuvant co‐delivery utilizing genetically engineered tumor cell‐derived exosomes with immunostimulatory CpG DNA. Biomaterials. 2016;111:55‐65. [DOI] [PubMed] [Google Scholar]
  • 129. Yong T, Zhang X, Bie N, et al. Tumor exosome‐based nanoparticles are efficient drug carriers for chemotherapy. Nat Commun. 2019;10(1):3838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Oskouie MN, Aghili Moghaddam NS, Butler AE, et al. Therapeutic use of curcumin‐encapsulated and curcumin‐primed exosomes. J Cell Physiol. 2019;234(6):8182‐8191. [DOI] [PubMed] [Google Scholar]
  • 131. Kamerkar S, Lebleu VS, Sugimoto H, et al. Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer. Nature. 2017;546(7659):498‐503. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

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

Data Availability Statement

Not applicable.


Articles from MedComm are provided here courtesy of Wiley

RESOURCES