Abstract
Exosomes are a type of small endosomal-derived vesicles ranging from 30 to 150 nm, which can serve as functional mediators in cell-to-cell communication and various physiological and pathological processes. In recent years, exosomes have emerged as crucial mediators of intracellular communication among tumor cells, immune cells, and stromal cells, which can shuttle bioactive molecules, such as proteins, lipids, RNA, and DNA. Exosomes exhibit the high bioavailability, biological stability, targeting specificity, low toxicity, and immune characteristics, suggesting their potentials in the diagnosis and treatment of cancers. They can be applied as an effective tool in the diagnostics, therapeutics, and drug delivery in cancers. This review summarizes the regulation and functions of exosomes in various cancers to augment our understanding of exosomes, which paves the way for parallel advancements in the therapeutic approach of cancers. In this review, we also discuss the challenges and prospects for clinical application of exosome-based diagnostics and therapeutics for cancers.
1. Introduction
Extracellular vesicles (EVs) are a collective term containing various types of cell-released membranous structures, including exosomes, microvesicles, microparticles, ectosomes, oncosomes, and apoptotic bodies [1, 2]. Among these EVs, exosomes are able to escape from the phagocyte system because of their small size and exert their superiority in intercellular communications. Exosomes are a type of endocytic and heterogeneous membrane-derived vesicles, which could be secreted by multiple cell types to induce or inhibit different cellular and molecular pathways, including immune cells, stem cells, and tumor cells [3, 4]. Exosomes used to be regarded as garbage cans for abandoning redundant cell materials. Recently, the roles of exosomes are gradually suggested, including immune regulation, intercellular communications, and biological events.
Cancer is the second leading cause of death all over the world [5]. Continuous improvements in the treatment of cancers, including surgery, chemotherapy, and radiotherapy, have significantly increased the survival, but these strategies cannot effectively control the recurrence and metastasis of cancers [6]. Thus, novel therapeutic methods are urgently needed. Since exosomes can play key roles in various physiological and pathological processes, it seems likely that they can also affect a variety of pathophysiological processes of cancers, including cancer development, migration, drug resistance, and metastasis. In this review, we comprehensively searched the PubMed database (https://pubmed.ncbi.nlm.nih.gov/) with the combined keywords “exosomes” and “cancers,” and focus on the biogenesis, regulation, and function of exosomes in cancers, which are capable of being used as a powerful weapon for the treatment of cancers.
2. Exosomes
2.1. Characteristics and Biogenesis
In 1981, Trams and his colleagues firstly described exosomes as microvesicles, which contained 5′-nucleotidase activity, released by neoplastic cell lines [7]. However, after that, the concept had been corrected and the endocytic origins of exosomes were proven, which had been regarded as natural nanocarriers. Exosomes are characterized by having a size between 30 and 150 nm, a cup-shaped structure, a density of 1.13–1.19 g/mL, and unique double lipid layer [8–10]. In addition, exosomes exhibit high biocompatibility and low toxicity. Exosomes are considered as a crucial player in the intercellular communication by transferring cellular contents to recipient cells [11]. They could transfer the bioactive molecules from cancer cells to cells in tumor microenvironment, thereby facilitating the development and progression of cancers [12]. The bioactive molecules in exosomes are transferred to nearby cells or recipient cells faraway, where they can modulate the signaling pathways involved in the cell proliferation, differentiation, and apoptosis. Exosomes regulate the biological behaviors of cancer cells, tumor microenvironmental cells, and distant recipient cells in this way and participate in the growth, invasion, metastasis, angiogenesis, and drug resistance of cancer cells [13].
Exosomes are commonly obtained from cell culture supernatants and various body fluids, including blood, cerebrospinal fluid, saliva, breast milk, bile, and urine. Exosomes contain a multitude of biomolecules, such as DNA, RNA, mRNA, lipids, metabolites, cytosolic, and proteins, and play essential roles in intercellular communications [14]. They can be secreted after fusion of multivesicular endosomes with the cell surface or directly sprout from plasma membranes. The biogenesis of exosomes mainly includes four stages, including initiation (membrane formation), endocytosis, formation of multivesicular bodies (MVBs), and sorting (secretion/degradation/recycling) [11]. At first, the cell membrane with ubiquitinated surface receptors is internalized endocytosis of the plasma membrane. Then, endosomal sorting complexes, which are necessary for the transport-0 (ESCRT-0), can bind to the ubiquitinated surface receptors via a FYVE domain to be changed into the early endosomes. And MVBs are generated in the form of trans-budding when ESCRT-I and ESCRT-II are assembled into the endosome membranes. At last, ESCRT-III induces the release of exosomes by fusing with the plasma membrane or the degradation by fusing with lysosomes.
2.2. Isolation and Purification
Differential ultracentrifugation method is usually used for the isolation of exosomes. The exosomes were extracted and isolated through multiple centrifugation steps, containing increasing centrifugal strength to sequentially pellet cells, microvesicles, and exosomes [15]. The method is fast and simple, while soluble proteins and nonexosomal particles in exosomes are hardly removed. Based on the centrifugation, ExoQuick precipitation method is also applied to isolate exosomes. After chemically precipitated with ExoPrep solution, the ultracentrifugation is performed. The advantages and disadvantages are similar to the differential ultracentrifugation method. Besides, there are also other techniques, such as ultrafiltration, antibody-coated magnetic beads, commercial precipitation kits, microfiltration techniques, and the state-of-the-art microfluidic technology, used for exosome separation.
Until now, the purest exosomes are isolated through density gradient isolation method by using sucrose or iodixanol. This method can obtain the purest exosomes, but are more time-consuming and more costly. Besides, microfluidic immunoisolation is applied for purification of exosomes by using antibodies against exosomal markers, such as EpCAM and CD63. This method is centered on exosomes expressing the target surface markers rather than other subpopulations of exosomes (27440105). There are also other novel detection modalities, such as biosensing and basic proteomics methods. Despite their unique characteristics, there is still a long way in the research of exosome-based assays.
2.3. Identification
Accurately detecting exosomes are important in the biology of vesicles. Currently, optical techniques and nonoptical methods are applied to characterize the vesicles. The optical methods contain nanoparticle tracking analysis (NTA), dynamic light scattering (DLS), fluorescence signals, and flow cytometry. The nonoptical methods include resistive pulse sensing (RPS), micronuclear magnetic resonance (μNMR), nanoholographic imaging, atomic force microscopy (AFM), enzyme-linked immunosorbent assay (ELISA), transmission electron microscopy (TEM), field-effect transistors (FET), lateral flow immunoassay (LFIA), and Raman spectroscopy. The above and emerging exosome-based technologies are helpful and urgently needed for the application of exosomes in the diagnosis and treatment of cancers. With gradually better understanding of exosomes, the corresponding technologies are also improving, which is helpful for developing the exosome-based application.
3. Exosomal RNAs in Cancers
Exosomes comprise various cellular elements, which are reflected by parental cells and affect the development of cancers. Increasing evidence shows that the functions of exosomes in cancers depend on their cargos and the identification of the components is one of the major challenges in the fields [16]. The uniqueness and enrichment of exosomal nucleic acids (RNAs) have been recently reported to be produced during the process of endosomes endocytose, collecting a large number of noncoding sequences. There are mainly three types of RNAs, including microRNAs (miRNAs), long noncoding RNAs (lncRNAs), circular RNAs (circRNAs), and other noncoding RNAs (ncRNAs) [17, 18].
3.1. miRNAs
It is widely accepted that miRNAs are a class of evolutionarily endogenous small noncoding RNAs that are about 20 to 25 nucleotide long sequences at length, which can bind to the target mRNAs and negatively regulate the gene expression via post-transcriptional inhibition or target mRNA degradation [19, 20]. As reported that, miRNAs are involved in the development and progression of cancers, which have brought increasing attraction [21]. For example, prostate cancer (PCa) cell-derived exosomes promote the angiogenesis, while the exosomes obtained from noncancer individuals are regarded as suppressors in the local environment [22]. Owing to the specific sorting mechanisms, exosomes contain greater concentrations of miRNAs, which are more stable than circulating miRNAs. The stability of miRNAs in exosomes is significantly correlated with the expression levels, which are able to transfer the information to various cellular processes [23]. According to the results of a large cohort including 195 non-small-cell lung cancer (NSCLC) patients and 30 healthy controls, in the research by Dejima et al., the plasma levels of exosomal miR-21 and miR-4257 derived from NSCLC were markedly higher in patients with recurrence than that in patients without recurrence or healthy individuals [24]. Besides, this study also indicated that the higher levels of exosomal miR-21 or miR-4257 are correlated with a worse prognosis with a shorter disease-free survival (DFS) [24]. It has been demonstrated that hepatocellular carcinoma (HCC) cells (Huh7)-derived exosomes restrain the growth and promote the aging of HepG2 cells by transferring miR-122 [25]. Previous studies indicated that breast cancer cell-derived exosomes promoted the growth, metastasis, and autophagy of breast cancer cells by transferring miR-1910-3p [26]. It has been shown that exosome-derived miR-200b promotes the proliferation of colorectal cancer, which is transferred to the recipient cells and suppresses the expression of p27 in the target cells [27]. Growing body of evidence indicated that exosomal RNAs play important roles in cancers (Table 1). With the in-depth research on the transportation functions of exosomes, exosomal miRNAs obtain a focus of attention. Thus, miRNAs play crucial roles in the development and progression of cancers.
Table 1.
Cancer type | Exosomal miRNAs | Functions of exosomal miRNAs | References |
---|---|---|---|
BCa | miR-10b | Associated with the acquisition of malignant characteristics | [28] |
miR-223 | Promote the invasion of breast cancer cells | [29] | |
miR-19a | Represent a biomarker | [30] | |
miR-105 | A potent regulator of migration | [31] | |
miR-379 | Tumor suppressor | [32] | |
miR-222 | Upregulated in BCa patients with lymphatic metastasis | [33] | |
miR-770 | Inhibit the chemoresistance and metastasis | [34] | |
miR-181d-5p | Promote epithelial-mesenchymal transition | [35] | |
Bladder cancer | miR-23b | Acquire metastatic potentials | [36] |
Colon cancer | miR-193a | Inhibit cell proliferation and cause cell cycle G1 arrest | [37] |
| |||
CRC | Let-7a, miR-1229, miR-1246, miR-150, miR-21, miR-223, and miR-23a | Upregulated in serum exosomes of primary CRC patients | [38] |
miR-301a and miR-23a | Upregulated in serum samples of CRC patients | [39] | |
miR-17-5p and miR-92a-3p | Prognostic biomarker | [40] | |
miR-150-5p | Diagnosis and prognosis | [41] | |
miR-27a and miR-130a | Diagnosis and prognosis | [42] | |
miR-196b-5p | Diagnosis and prognosis | [43] | |
miR-25-3p | Promote premetastatic niche formation | [44] | |
ESCC | miR-21 | Upregulated in serum from ESCC patients | [45] |
GBM | miR-21 | Upregulated in GBM patients | [46] |
Glioma | miR-146b | Inhibit glioma growth | [47] |
| |||
HCC | miR-1247-3p | Correlated with lung metastasis in HCC patients | [48] |
miR-122 | Increased the antitumor efficacy of sorafenib on HCC | [25] | |
miR-101, miR-106b, miR-122, and miR-195 | Downregulated in the serum exosome of HCC patients | [49] | |
miR-718 | Inhibit cell proliferation of HCC cells | [50] | |
miR-335-5p | Inhibit HCC cell proliferation and invasion, induce HCC tumor shrinkage | [51] | |
Leukemia | miR-210 | Upregulated in exosomes and enhances endothelial migration and tube formation | [52] |
| |||
Lung cancer | miR-29a | Tumor growth and metastasis | [53] |
miR-17-3p, miR-21, miR-106a, miR-146, miR-155, miR-191, miR-192, miR-203, miR-205, miR-210, miR-212, and miR-214 | Diagnostic markers | [54] | |
miR-155 and low let-7a-2 | Diagnosis and prognosis | [55] | |
Lymphoma | miR-155, miR-210, and miR-21 | Diagnostic markers | [56] |
MM | miR-135b | Upregulated in exosomes from HR-MM and enhances endothelial tube formation | [57] |
| |||
Ovarian cancer | miR-30a-5p | Promote malignant phenotypes of ovarian cancer | [58] |
miR-21, miR-141, miR-200a, miR-200c, miR-200b, miR-203, miR-205, and miR-214 | Surrogate diagnostic markers for biopsy profiling | [59] | |
miR-21 | Promote oncogenic transformation in target cells | [60] | |
PaCa | miR-4306, miR-4644, miR-3976, and miR-1246 | Upregulated in serum exosomes of PaCa patients | [61] |
| |||
PC | miR-125b, miR-130b, and miR-155 | Promote neoplastic transformation in adipose derived stem cells | [62] |
miR-196a-5p and miR-501-3p | Downregulated in exosomes from PC patients | [63] | |
miR-141, miR-375, miR-107 and miR-574-3p | Biomarkers for the diagnosis, staging, and prediction of PC | [64] | |
miR-1290 and miR-375 | Prognostic biomarkers for castration-resistant PC patients | [65] | |
PDAC | miR-10b | Upregulated in plasma-derived exosomes from PDAC patients | [66] |
miR-181a, miR-10 b, miR-21, and miR-30c | Upregulated in exosomes from PDAC patients | [67] | |
miR-145-5p | Inhibit the growth of xenograft tumors | [68] |
BCa, breast cancer; CRC, colorectal cancer; ESCC, esophageal squamous cell cancer; GBM, glioblastoma; HCC, hepatocellular carcinoma; MM, multiple myeloma; PaCa, pancreatic cancer; PC, prostate cancer; PDAC, pancreatic ductal adenocarcinoma.
3.2. lncRNAs
Other molecules, such as lncRNAs, have also been found in exosomes. LncRNAs contain 200 nucleotide sequences at length, which can regulate the expression of genes or proteins via coordinating epigenetic, transcriptional, and post-transcriptional steps [69]. Numerous studies showed that lncRNAs exert their functions via multiple molecular mechanisms, including binding with DNA to regulate the gene transcription, acting as the competing endogenous RNA (ceRNA) to control the gene expression at post-transcriptional level, combining with proteins, and encoding functional small peptides [70]. Interestingly, lncRNAs could be selectively sorted from exosomes and involved in the intercellular communication in the tumor microenvironment [71].
Exosomal lncRNAs can play important roles in the occurrence and development of cancers, including proliferation, invasion, angiogenesis, and drug resistance, which might act as attractive therapeutic targets and diagnostic biomarkers [72] (Table 2). A recent study showed that in patients with NSCLC, the exosomal lncRNA MALAT-1 was much higher than healthy controls. Besides, the upregulation of exosomal lncRNA MALAT-1 is closely related to an advanced tumor stage and lymphatic metastasis [84]. Although a growing number of evidence reveals that found that lncRNAs could be transferred between various cells, which is related to exosomes, many questions are still doubtful. For example, it is unclear whether nuclear and cytoplasmic lncRNAs exert the similar function that can be loaded into exosomes, and how their sequences, structures, and protein-binding partners affect the sorting. How many copies of lncRNAs are involved in exosomes are needed for phenotypically affecting the recipient cells? There is a dramatical difference between physiological exosome communication and in vitro treatment with purified vesicle preparations in these aspects, including time and intensity, which require further investigation and careful evaluation. In addition, while lncRNAs are upregulated in specific cancers in comparison with normal tissues and could be detected in exosomes in the circulation of patients with cancers, it is unclear whether the incorporation into exosomes depends on the specific sorting mechanisms in cancer cells [86].
Table 2.
Cancer type | Exosomal lncRNAs | Functions of exosomal lncRNAs | References |
---|---|---|---|
BCa | lncRNA-SNHG1 | Promote trastuzumab chemoresistance | [73] |
lncRNA UCA1 | Induce drug resistance | [74] | |
lncRNA AGAP2-AS1 | Induce drug resistance | [75] | |
lncRNA AFAP1-AS1 | Induce drug resistance | [76] | |
lncRNA H19 | Induce drug resistance | [77] | |
| |||
CRC | LNCV6_116109, LNCV6_98390, LNCV6_38772, LNCV_108266, LNCV6_84003, and LNCV6_98602 | Upregulated in CRC patients | [78] |
lncRNA LINC02418 | Diagnosis and prognosis | [79] | |
lncRNA RPPH1 | Diagnostic marker | [80] | |
lncRNA CRNDE-h | Diagnosis and prognosis | [81] | |
GC | LncRNA HOTTIP | Upregulated in serum exosomes from GC patients | [82] |
HCC | LncRNA-H19 | Promote angiogenesis and adhesion of CD90+Huh7 cells to endothelial cell monolayer | [83] |
NSCLC | lncRNA MALAT-1 | Promote cell proliferation and migration | [84] |
RCC | LncRNA-ARSR | Transmit to sensitive cells and disseminate sunitinib resistance | [85] |
BCa, breast cancer; CRC, colorectal cancer; GC, gastric cancer; HCC, hepatocellular carcinoma; NSCLC, non-small-cell lung cancer; RCC, renal cell carcinoma.
3.3. circRNAs
As a type of exosome-derived noncoding RNAs, exosomal circRNAs have been found to play key roles in the development and progression of cancers and act as novel diagnostic and prognostic biomarkers in cancers. CircRNAs are a class of tissue specific and covalently closed circular noncoding RNA, which is widely present in eukaryote. Exosomal circRNAs are novel frontiers in cancer research, and the important exosomal circRNAs are summarized in Table 3. These studies have suggested that exosomal circRNAs might exhibit an important influence on the development and occurrence of cancers, which suggested the potential diagnostic and therapeutic values of exosomal circRNAs. To explore the mysterious connections of exosomes and circRNAs, it might provide a key hint to probe into the biological functions of exosomal circRNAs. In addition, studying the pathogenesis mechanisms of cancers and identifying novel promising diagnostic biomarkers and therapeutic targets are a popular topic in the future.
Table 3.
Cancer type | Exosomal circRNAs | Functions of exosomal circRNAs in cancers | References |
---|---|---|---|
CRC | circ-ABCC1 | Promote CRC progression | [87] |
circ-KLDHC10 | Upregulated in serum of CRC patients | [88] | |
circ-0004771 | Diagnosis and prognosis | [89] | |
GC | ciRS-133 | Promote cancer cachexia, oxygen consumption, and heat production | [90] |
circ-SHKBP1 | Promote GC progression | [91] | |
circ-RanGAP1 | Promote invasion and metastasis | [92] | |
circ-0032821 | Promote tumor growth | [93] | |
Glioma | circ-MMP1 | Promote the progression of glioma | [94] |
HCC | circ-ZNF652 | Promote cell proliferation, migration, invasion, and glycolysis | [95] |
circ-100284 | Promote the cell cycle | [96] | |
circ-DB | Promote tumor cell proliferation | [97] | |
circ-100338 | Promote HCC metastasis | [98] | |
circ- Cdr1as | Promote HCC proliferation and migration | [99] | |
circ-0051443 | Downregulated in plasma exosomes and tissues from HCC patients | [100] | |
circ-PTGR1 | Upregulated in serum exosomes from HCC patients | [101] | |
circ-UHRF1 | Upregulated in human HCC tissues | [102] | |
circ-AKT3 | Upregulated in exosome from HCC patients | [103] | |
LSCC | circRASSF2 | Promote cancer cell proliferation and migration | [104] |
LUAD | circ-002178 | Upregulated in the LUAD tissues and LUAD cancer cells | [105] |
MM | circ-MYC | Upregulated in exosome from MM patients | [106] |
OSCC | circGDI2 | Inhibit OSCC cell proliferation, migration, invasion, and glycolysis | [107] |
PC | circ-0044516 | Promote cell proliferation and metastasis | [108] |
PDAC | circ-PDE8A | Promote the invasive growth of PDAC cell | [109] |
circ-IRAS | Promote tumor invasion and metastasis | [110] | |
UCB | circ-PRMT5 | Upregulated in serum and urine exosomes from UCB patients | [111] |
CRC, colorectal cancer; HCC, hepatocellular carcinoma; LSCC, laryngeal squamous cell carcinoma; LUAD, lung adenocarcinoma; MM, multiple myeloma; OSCC, oral squamous cell carcinoma; PC, prostate cancer; PDAC, pancreatic ductal adenocarcinoma; UCB, urothelial carcinoma of the bladder.
4. Exosomal Proteins in Cancers
Exosomes can carry a broad variety of molecules depending on the origins and in vitro culture conditions, such as proteins. It has been shown that exosomes can express a wide range of proteins involved in membrane-related functions, including Rab GTPase, Annexin, cellular adhesion proteins (integrins and tetraspanins), cytoskeletal proteins (actin and myosin), and heat shock proteins (Hsp70) [112]. Recent studies suggest that membrane transport and fusion proteins in exosomes play vital roles in the occurrence and development of cancers, including annexins, RAB5/RAB7, and TSG101. For example, dendritic cells (DCs)-derived exosomes are capable of carrying MHC-I, which can bind to the tumor-derived peptides, and then, the complex activates the immune cells to play crucial antitumor roles [113]. An interesting study has revealed that membrane surface protein TRAIL from exosomes can transfer the apoptosis-related signals to the tumor cells and induce the apoptosis of tumor cells [114]. Moreover, exosomes with SIRPα are capable of binding to CD47 on tumor cells, and promoting the phagocytosis of macrophages and finally inhibiting the cancer progression [115].
In addition to the RNAs and DNA present in exosomes, exosomal proteins could also be used as promising biomarkers (Table 4). It has been reported that exosomal EGFR protein can be regarded as a potential biomarker for the characterization of lung cancer [141, 142]. As demonstrated, the diagnostic potentials of 49 exosomal membrane-attached proteins in 336 patients with lung cancer and 127 controls are confirmed. Among these proteins, CD151, CD171, and tetraspanin 8 are reported to be the strongest biomarkers for patients with lung cancer [143].
Table 4.
Cancer type | Exosomal proteins | Functions of exosomal proteins | References |
---|---|---|---|
BC | L-plastin | Promote osteolysis | [116] |
| |||
CRC | CD147 | Diagnosis and prognosis | [117] |
IRF-2 | Remodel the lymphatic network | [118] | |
| |||
GBM | IL-6, IL-8, and angiogenin | Diagnosis and prognosis | [119] |
EGFR, EGFRvIII, and TGF-β | Diagnosis | [120] | |
EGFR, EGFRvIII, PDPN, and IDH1 R132H | Diagnosis and prognosis | [121] | |
VEGF-A | Diagnosis | [122] | |
CD97 | Promote metastasis | [123] | |
HMGB1 | Diagnosis | [124] | |
Glioma | IL13QD | Diagnosis and prognosis | [125] |
| |||
Lung cancer | IQGAP, MUC5B, BPIFA1, and CRNN | Diagnosis | [126] |
LRG1 | Diagnosis | [127] | |
| |||
Melanoma | HSP70, HSP90, TYRP2, VLA-4, and MET | Diagnosis and prognosis | [128] |
CD63 and Caveolin-1 | Diagnosis | [129] | |
MHC-I/peptide complexes | Diagnosis | [130] | |
Podoplanin | Diagnosis | [131] | |
TYRP2, VLA-4, HSP70, and HSP90 | Diagnosis and prognosis | [128] | |
Myeloma | HSP70 | Diagnosis | [132] |
NSCLC | Amphiregulin | Promote osteoclastogenesis and metastasis | [133] |
| |||
Ovarian cancer | HNRHPU, U2AF2 TGM2, and U2AF1 | Diagnosis | [134] |
EpCAM and CD24 | Diagnosis | [135] | |
PaCa | Glypican-1 | Diagnosis | [136] |
| |||
PC | PDCD6IP, FASN, XPO1, and ENO1 | Diagnosis | [137] |
αvβ6 integrin | Regulate monocyte M2 polarization | [138] | |
| |||
PDAC | MIF | Promote fibronectin secretion and metastasis | [139] |
CD151 and TSPAN8 | Promote EMT gene expression | [140] |
BC, breast cancer; CRC, colorectal cancer; GBM, glioblastoma; GC, gastric cancer; NSCLC, non-small-cell lung cancer; PaCa, pancreatic cancer; PC, prostate cancer; PDAC, pancreatic ductal adenocarcinoma.
5. Exosome-Based Diagnostics and Therapeutics in Cancers
5.1. Diagnosis
With the aging and growth of the population, the incidence and mortality of cancers are rapidly growing and cancer is one of the most leading causes of death. Cancers seriously endanger human health, and early diagnosis of cancers is required for increasing the survival rates of patients and reducing the mortality. Recently, one of the latest concepts regarding early diagnosis is based on the extracellular vesicles released by cells where the exosomes are at the frontiers. The biomarker potentials of exosomes in liquid biopsies hold huge promise and might revolutionize the diagnostics strategies of cancers. For example, exosome-derived miRNAs can act as novel potential biomarkers in the diagnostics and prediction of colorectal cancer [144, 145]. It has been found that miR-423-5p is increased in the serum exosomes of patients with gastric cancer (GC), and the levels of miR-423-5p are related to lymph node metastasis and poor outcome [146]. And the levels of miR-126 and let-7a are much higher in exosomes collected from the bronchoalveolar fluid of patients with lung adenocarcinoma than healthy controls [147]. Moreover, patients with pancreatic cancer (PC) exhibit higher level of exosomal miR-191, miR-21, and miR-451a than healthy subjects [148]. Exosomal lncRNA SAP30L-AS1 has been discovered to upregulated in benign prostatic hyperplasia (BPH) and lncRNA SChLAP1 is increased in prostate cancer than in BPH and healthy controls, which is confirmed to possess diagnostic values in distinguishing prostate cancer by the receiver operating characteristic curve [149]. A study containing 246 subjects (126 patients and 120 healthy controls) showed that the expression levels of exosomal lncRNA HOTTIP were much higher in gastric cancer patients than normal controls, which was in close correlation with invasion depth [82].
In addition, the protein levels in exosomes might also act as biomarkers in the diagnosis of cancers. As previously described, the patients with acute myeloid leukemia (AML) exhibit higher levels of plasma exosomes with a specific phenotype containing transforming growth factor beta 1 (TGFβ-1), which has a suppressive effect in the cytotoxic activity of NK cells [150]. According to the results of proteomics analysis by mass spectrometry of urinary exosomes obtained from 16 patients with prostate cancer (PC) and 15 controls, the transmembrane protein 256 is the highest sensitivity of 94% and distinctly enriched with high specificity in patients with PC [151]. As previous detailed, an inhibitor of apoptosis protein (Survivin-2B) has been found to be differentially expressed in exosomes from patients with breast cancer, suggesting its values in the diagnosis and prognosis of breast cancer [152]. The results of gene microarray from blood exosomes of mice with glioblastoma multiforme (GBM) revealed that the levels of DNM3, p65, and CD117 are significantly increased, whereas PTEN and p53 are decreased, which might be served as novel diagnostic markers for GBM [153].
Some studies also focus on the roles of circRNAs in cancer diagnosis. For example, the level of circ-PDE8A has been detected in the plasma exosomes from patients with pancreatic ductal adenocarcinoma (PDAC) and exosomal circ-PDE8A has been found to be related to the progression and prognosis in PDAC patients [109]. Many studies have stressed the roles of exosomes in the diagnosis and prediction of cancers owing to the exosomal molecules, including miRNAs, proteins, and lncRNAs. We believe that in-depth understanding of exosomes in the diagnosis of cancers could help to design novel cancer-diagnostic and cancer-prognostic tools. Effective diagnostic strategies for cancers based on exosomes are expected in the near future. Despite these advances in exosomes-based diagnostics and prediction of cancers, there are still many challenges needed to be overcome until clinical application. Much research needs to be done about the roles of exosomes in the diagnosis and prediction of cancers in the future.
5.2. Treatment
Regarded as the best-studied natural nanomaterials in the past decades in cancer therapy, exosomes are able to pass through the biological barriers, such as the blood-brain barrier (BBB) and blood-tumor barrier, indicating their potentials in the diagnosis and treatment of brain cancers. Mesenchymal stem cells (MSCs)-derived exosomes have been reported to be applied in the treatment of pancreatic cancer (PC) in animal experiments by delivering KRAS, G12D, and siRNAs [154, 155]. Besides, exosomes could be applied as novel antitumor vaccines. For example, exosomes obtained from dendritic cells can deliver the melanoma antigen identified by T cells 1 (MART-1) into patients with NSCLC at IIIB/IV stage and then inhibit the progression of NSCLC and improve the survival through activation of immune response, which is revealed by phase II clinical trials [113].
Many targets of anticancer chemical agents are intracellular, which need to cross the cell membranes and then exert anticancer functions. Because of their poor water solubility and easy degradation, the chemical agents are difficult to produce the desired effects or avoid adverse reactions. Exosomes have the advantages of low immune prototype and weak side effects, whose lipid bilayer can play protective roles in the contents in vivo [156]. Besides, exosomes are easy to enter the cells via the interaction between their membrane proteins and target cells. Thus, exosomes are regarded as the best natural carriers for chemical agents [115]. Hence, a better understanding of how to engineer and deliver exosomes to specific cells is crucial to improve the cancer therapy potential of exosomes. For example, plant-derived exosomes are easily to be absorbed by the intestine and are used for inhibiting the development of colon cancer by delivering curcumin, confirmed by clinical trials (ClinicalTrials.gov identifier: NCT01294072). It has been confirmed that the treatment with DCs-derived exosomes carrying doxorubicin significantly inhibits the growth of breast cancer cells and exerts no toxicity effects on mouse models [157]. In the study of K. Tang et al., exosomes loaded with cisplatin exert significant anticancer effects on ovarian cancer and cause no side effects on other tissues in mice, including liver and kidney. And this is an advantage in comparison with cisplatin treated alone [158]. Moreover, exosomes from plants can also be used in the adjuvant therapy for cancers. It has been shown that during the treatment of head and neck cancer, the patients suffer from oral mucositis. And grape-derived exosomes are able to ease the oral mucositis and pain, revealed by clinical trials (ClinicalTrials.gov identifier: NCT01668849). Exosomes can extend the circulation half-life and block the drug accumulation in nontarget organs by targeting specific cells. Thus, the release of exosomes in biofluids could have major advantages for exploration of the complex mechanisms of tumor progression and treatment response.
To successfully develop novel and efficient therapeutic strategies in the treatment of cancers, exosomes should not be disregarded. And the application of exosomes in clinical treatment of cancers is a challenging but intriguing endeavor that needs further investigation and exploration.
6. Conclusion and Future Prospective
Increasing evidence concerning exosomes has stressed their important roles in cancer development and clinical application recently. Exosome-derived cargos are involved in the pathophysiological processes of cancers, including cancer development, migration, drug resistance, and metastasis. In this review, we have elaborated upon the characteristics, biogenesis, isolation, purification, and identification of exosomes and summarized the roles of exosomal RNAs (miRNAs, lncRNAs, and circRNAs) and proteins in cancers. Investigation of the molecular mechanisms underlying the biogenesis and biological functions of exosomes will help to design more novel therapeutic methods targeting exosome-mediated pathophysiological processes in cancers.
It is noteworthy that various exosomal molecules are applied for developing emerging diagnostic strategies and clinical treatments of cancers. Obviously, the specific RNA in exosomes is one of the most robust surrogates of cancers and affects the current states of cancers. Thus, the corresponding targeted agents for these RNAs and molecules can be designed, which can be further personalized medicine. In addition to the research progress in mechanism, there are also further challenges in clinical application. The current knowledge on the roles of exosomes in cancers is limited; therefore, their potentials in the diagnosis and treatment of cancers require extensively studied.
Although exosome-based cancer treatment exerts great potentials, there are still many problems need to be solved until clinical application. The technologies of production and quality control are flawed. Although some preclinical studies evaluate the roles of exosomes as therapeutic drug delivery carriers in the cancer therapy and suggest their clinical application potentials, few exosomes-based clinical trials have been conducted, which might be the focus in the future research. Besides, several technical limitations, including high quality and bulk exosome preparation based on standard protocol, and exosome target specificity, are the biggest barriers presently impeded exosome-based diagnostic and therapeutic applications. Currently, there is still a long way to overcome the difficulty in the exosome-based therapeutic strategies in the treatment of cancers. Exosomes-based research might need to put more energy into in vivo models and clinical application in the future so that it is helpful to elucidate these questions.
Acknowledgments
The authors thank Prof. Yuemao Shen (Shandong University) for critical reading and discussions of the paper. This work was supported by grants from the Taishan Scholars Program (TSQN201812015), the Rongxiang Regenerative Medicine Foundation (2019SDRX-04), and the program for Multidisciplinary Research and Innovation Team of Young Scholars of Shandong University (2020QNQT007). This work was also supported by the Postdoctoral Science Foundation of China (2021M691956), the Natural Science Foundation of Shandong Province (ZR2021QH087), the Medicine and Health Science and Technology Development Plan Project of Shandong Province (202002040581), the Natural Science Foundation of Jiangsu Province (BK20210110), and the Science and Technology Development Project of Suzhou (SYG202119).
Contributor Information
Yuan Li, Email: yuanli@sdu.edu.cn.
Baobing Zhao, Email: baobingzh@sdu.edu.cn.
Conflicts of Interest
The authors declare that they have no conflicts of interest.
Authors' Contributions
Yuan Li conceived of the review paper with the guidance of Baobing Zhao; Yuan Li wrote the original draft; Yuan Li, Li Meng, Bo Li, Yanxia Li, Tao Shen, and Baobing Zhao reviewed and edited the paper. All authors have read and agreed to the published version of the manuscript.
References
- 1.Jalalian S. H., Ramezani M., Jalalian S. A., Abnous K., Taghdisi S. M. Exosomes, new biomarkers in early cancer detection. Analytical Biochemistry . 2019;571:1–13. doi: 10.1016/j.ab.2019.02.013. [DOI] [PubMed] [Google Scholar]
- 2.Théry C., Witwer K. W., Aikawa E., et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the international society for extracellular vesicles and update of the MISEV2014 guidelines. Journal of Extracellular Vesicles . 2018;7(1) doi: 10.1080/20013078.2018.1535750.1535750 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.He C., Zheng S., Luo Y., Wang B. Exosome theranostics: biology and translational medicine. Theranostics . 2018;8(1):237–255. doi: 10.7150/thno.21945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Yang B., Chen Y., Shi J. Exosome biochemistry and advanced nanotechnology for next-generation theranostic platforms. Advanced Materials . 2019;31(2) doi: 10.1002/adma.201802896.1802896 [DOI] [PubMed] [Google Scholar]
- 5.Siegel R. L., Miller K. D., Jemal A. Cancer statistics, 2020. CA: A Cancer Journal for Clinicians . 2020;70(1):7–30. doi: 10.3322/caac.21590. [DOI] [PubMed] [Google Scholar]
- 6.Xu Z., Zeng S., Gong Z., Yan Y. Exosome-based immunotherapy: a promising approach for cancer treatment. Molecular Cancer . 2020;19(1):p. 160. doi: 10.1186/s12943-020-01278-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Trams E. G., Lauter C. J., Salem J. N., Heine U. Exfoliation of membrane ecto-enzymes in the form of micro-vesicles. Biochimica et Biophysica Acta (BBA) - Biomembranes . 1981;645(1):63–70. doi: 10.1016/0005-2736(81)90512-5. [DOI] [PubMed] [Google Scholar]
- 8.Yu W., Hurley J., Roberts D., et al. Exosome-based liquid biopsies in cancer: opportunities and challenges. Annals of Oncology . 2021;32(4):466–477. doi: 10.1016/j.annonc.2021.01.074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Zhang L., Yu D. Exosomes in cancer development, metastasis, and immunity. Biochimica et Biophysica Acta (BBA) - Reviews on Cancer . 2019;1871(2):455–468. doi: 10.1016/j.bbcan.2019.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Kalluri R., LeBleu V. S. The biology , function , and biomedical applications of exosomes. Science . 2020;367 doi: 10.1126/science.aau6977. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Mashouri L., Yousefi H., Aref A. R., Ahadi A. M., Molaei F., Alahari S. K. Exosomes: composition, biogenesis, and mechanisms in cancer metastasis and drug resistance. Molecular Cancer . 2019;18(1):p. 75. doi: 10.1186/s12943-019-0991-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Fu M., Gu J., Jiang P., Qian H., Xu W., Zhang X. Exosomes in gastric cancer: roles, mechanisms, and applications. Molecular Cancer . 2019;18(1):p. 41. doi: 10.1186/s12943-019-1001-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Dai J., Su Y., Zhong S., et al. Exosomes: key players in cancer and potential therapeutic strategy. Signal Transduction and Targeted Therapy . 2020;5(1):p. 145. doi: 10.1038/s41392-020-00261-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Pi Y. N., Xia B. R., Jin M. Z., Jin W. L., Lou G. Exosomes: Powerful weapon for cancer nano-immunoengineering. Biochemical Pharmacology . 2021;186 doi: 10.1016/j.bcp.2021.114487.114487 [DOI] [PubMed] [Google Scholar]
- 15.Zhang J., Li S., Li L., et al. Exosome and exosomal microRNA: trafficking, sorting, and function. Genomics, Proteomics & Bioinformatics . 2015;13(1):17–24. doi: 10.1016/j.gpb.2015.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Bebelman M. P., Smit M. J., Pegtel D. M., Baglio S. R. Biogenesis and function of extracellular vesicles in cancer. Pharmacology & Therapeutics . 2018;188:1–11. doi: 10.1016/j.pharmthera.2018.02.013. [DOI] [PubMed] [Google Scholar]
- 17.Yu T., Wang Y., Fan Y., et al. CircRNAs in cancer metabolism: a review. Journal of Hematology & Oncology . 2019;12(1):p. 90. doi: 10.1186/s13045-019-0776-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Poulet C., Njock M. S., Moermans C., et al. Exosomal long non-coding rnas in lung diseases. International Journal of Molecular Sciences . 2020;21(10):p. 3580. doi: 10.3390/ijms21103580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Li Y., Yang F., Gao M., et al. miR-149-3p regulates the switch between adipogenic and osteogenic differentiation of BMSCs by targeting FTO. Molecular Therapy - Nucleic Acids . 2019;17:590–600. doi: 10.1016/j.omtn.2019.06.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Li Y., Feng C., Gao M., et al. MicroRNA‐92b‐5p modulates melatonin‐mediated osteogenic differentiation of bone marrow mesenchymal stem cells by targeting ICAM‐1. Journal of Cellular and Molecular Medicine . 2019;23(9):6140–6153. doi: 10.1111/jcmm.14490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Yang F., Ning Z., Ma L., et al. Exosomal miRNAs and miRNA dysregulation in cancer-associated fibroblasts. Molecular Cancer . 2017;16(1):p. 148. doi: 10.1186/s12943-017-0718-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Kharmate G., Hosseini-Beheshti E., Caradec J., Chin M. Y., Tomlinson Guns E. S. Epidermal growth factor receptor in prostate cancer derived exosomes. PLoS One . 2016;11(5) doi: 10.1371/journal.pone.0154967.e0154967 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Malla B., Zaugg K., Vassella E., Aebersold D. M., Dal Pra A. Exosomes and exosomal micrornas in prostate cancer radiation therapy. International Journal of Radiation Oncology, Biology, Physics . 2017;98(5):982–995. doi: 10.1016/j.ijrobp.2017.03.031. [DOI] [PubMed] [Google Scholar]
- 24.Dejima H., Iinuma H., Kanaoka R., Matsutani N., Kawamura M. Exosomal microRNA in plasma as a non-invasive biomarker for the recurrence of non-small cell lung cancer. Oncology Letters . 2017;13(3):1256–1263. doi: 10.3892/ol.2017.5569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Lou G., Song X., Yang F., et al. Exosomes derived from miR-122-modified adipose tissue-derived MSCs increase chemosensitivity of hepatocellular carcinoma. Journal of Hematology & Oncology . 2015;8(1):p. 122. doi: 10.1186/s13045-015-0220-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Wang B., Mao J. H., Wang B. Y., et al. Exosomal miR-1910-3p promotes proliferation, metastasis, and autophagy of breast cancer cells by targeting MTMR3 and activating the NF-κB signaling pathway. Cancer Letters . 2020;489:87–99. doi: 10.1016/j.canlet.2020.05.038. [DOI] [PubMed] [Google Scholar]
- 27.Zhang Z., Xing T., Chen Y., Xiao J. Exosome-mediated miR-200b promotes colorectal cancer proliferation upon TGF-β1 exposure. Biomedicine & Pharmacotherapy . 2018;106:1135–1143. doi: 10.1016/j.biopha.2018.07.042. [DOI] [PubMed] [Google Scholar]
- 28.Singh R., Pochampally R., Watabe K., Lu Z., Mo Y. Y. Exosome-mediated transfer of miR-10b promotes cell invasion in breast cancer. Molecular Cancer . 2014;13(1):p. 256. doi: 10.1186/1476-4598-13-256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Yang M., Chen J., Su F., et al. Microvesicles secreted by macrophages shuttle invasion-potentiating microRNAs into breast cancer cells. Molecular Cancer . 2011;10(1):p. 117. doi: 10.1186/1476-4598-10-117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Anfossi S., Giordano A., Gao H., et al. High serum miR-19a levels are associated with inflammatory breast cancer and are predictive of favorable clinical outcome in patients with metastatic HER2+ inflammatory breast cancer. PLoS One . 2014;9(1) doi: 10.1371/journal.pone.0083113.e83113 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Zhou W., Fong M., Min Y., et al. Cancer-secreted miR-105 destroys vascular endothelial barriers to promote metastasis. Cancer Cell . 2014;25(4):501–515. doi: 10.1016/j.ccr.2014.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.O’Brien K. P., Khan S., Gilligan K. E., et al. Employing mesenchymal stem cells to support tumor-targeted delivery of extracellular vesicle (EV)-encapsulated microRNA-379. Oncogene . 2018;37(16):2137–2149. doi: 10.1038/s41388-017-0116-9. [DOI] [PubMed] [Google Scholar]
- 33.Ding J., Xu Z., Zhang Y., et al. Exosome-mediated miR-222 transferring: An insight into NF-κB-mediated breast cancer metastasis. Experimental Cell Research . 2018;369(1):129–138. doi: 10.1016/j.yexcr.2018.05.014. [DOI] [PubMed] [Google Scholar]
- 34.Li Y., Liang Y., Sang Y., et al. MiR-770 suppresses the chemo-resistance and metastasis of triple negative breast cancer via direct targeting of STMN1. Cell Death & Disease . 2018;9(1):p. 14. doi: 10.1038/s41419-017-0030-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Wang H., Wei H., Wang J., Li L., Chen A., Li Z. MicroRNA-181d-5p-Containing Exosomes Derived from CAFs Promote EMT by Regulating CDX2/HOXA5 in Breast Cancer. Molecular Therapy - Nucleic Acids . 2020;19:654–667. doi: 10.1016/j.omtn.2019.11.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Ostenfeld M. S., Jeppesen D. K., Laurberg J. R., et al. Cellular disposal of miR23b by RAB27-dependent exosome release is linked to acquisition of metastatic properties. Cancer Research . 2014;74(20):5758–5771. doi: 10.1158/0008-5472.can-13-3512. [DOI] [PubMed] [Google Scholar]
- 37.Teng Y., Ren Y., Hu X., et al. MVP-mediated exosomal sorting of miR-193a promotes colon cancer progression. Nature Communications . 2017;8(1) doi: 10.1038/ncomms14448.14448 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Ogata-Kawata H., Izumiya M., Kurioka D., et al. Circulating exosomal microRNAs as biomarkers of colon cancer. PLoS One . 2014;9(4) doi: 10.1371/journal.pone.0092921.e92921 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Karimi N., Feizi M. A. H., Safaralizadeh R., et al. Serum overexpression of miR-301a and miR-23a in patients with colorectal cancer. Journal of the Chinese Medical Association . 2019;82(3):215–220. doi: 10.1097/jcma.0000000000000031. [DOI] [PubMed] [Google Scholar]
- 40.Fu F., Jiang W., Zhou L., Chen Z. Circulating exosomal mir-17-5p and mir-92a-3p predict pathologic stage and grade of colorectal cancer. Translational Oncology . 2018;11(2):221–232. doi: 10.1016/j.tranon.2017.12.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Zou S. L., Chen Y. L., Ge Z. Z., Qu Y. Y., Cao Y., Kang Z. X. Downregulation of serum exosomal miR-150-5p is associated with poor prognosis in patients with colorectal cancer. Cancer Biomarkers . 2019;26(1):69–77. doi: 10.3233/cbm-190156. [DOI] [PubMed] [Google Scholar]
- 42.Liu X., Pan B., Sun L., et al. Circulating exosomal mir-27a and mir-130a act as novel diagnostic and prognostic biomarkers of colorectal cancer. Cancer Epidemiology, Biomarkers & Prevention . 2018;27(7):746–754. doi: 10.1158/1055-9965.epi-18-0067. [DOI] [PubMed] [Google Scholar]
- 43.Ren D., Lin B., Zhang X., et al. Maintenance of cancer stemness by miR-196b-5p contributes to chemoresistance of colorectal cancer cells via activating STAT3 signaling pathway. Oncotarget . 2017;8(30):49807–49823. doi: 10.18632/oncotarget.17971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.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. Nature Communications . 2018;9(1):p. 5395. doi: 10.1038/s41467-018-07810-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Tanaka Y., Kamohara H., Kinoshita K., et al. Clinical impact of serum exosomal microRNA-21 as a clinical biomarker in human esophageal squamous cell carcinoma. Cancer . 2013;119(6):1159–1167. doi: 10.1002/cncr.27895. [DOI] [PubMed] [Google Scholar]
- 46.Akers J. C., Ramakrishnan V., Kim R., et al. MiR-21 in the extracellular vesicles (EVs) of cerebrospinal fluid (CSF): a platform for glioblastoma biomarker development. PLoS One . 2013;8(10) doi: 10.1371/journal.pone.0078115.e78115 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Katakowski M., Buller B., Zheng X., et al. Exosomes from marrow stromal cells expressing miR-146b inhibit glioma growth. Cancer Letters . 2013;335(1):201–204. doi: 10.1016/j.canlet.2013.02.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Fang T., Lv H., Lv G., et al. Tumor-derived exosomal miR-1247-3p induces cancer-associated fibroblast activation to foster lung metastasis of liver cancer. Nature Communications . 2018;9(1):p. 191. doi: 10.1038/s41467-017-02583-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Sohn W., Kim J., Kang S. H., et al. Serum exosomal microRNAs as novel biomarkers for hepatocellular carcinoma. Experimental & Molecular Medicine . 2015;47(9) doi: 10.1038/emm.2015.68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Sugimachi K., Matsumura T., Hirata H., et al. Identification of a bona fide microRNA biomarker in serum exosomes that predicts hepatocellular carcinoma recurrence after liver transplantation. British Journal of Cancer . 2015;112(3):532–538. doi: 10.1038/bjc.2014.621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Wang F., Li L., Piontek K., Sakaguchi M., Selaru F. M. Exosome miR-335 as a novel therapeutic strategy in hepatocellular carcinoma. Hepatology . 2018;67(3):940–954. doi: 10.1002/hep.29586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Tadokoro H., Umezu T., Ohyashiki K., Hirano T., Ohyashiki J. H. Exosomes derived from hypoxic leukemia cells enhance tube formation in endothelial cells. Journal of Biological Chemistry . 2013;288(48):34343–34351. doi: 10.1074/jbc.m113.480822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Fabbri M., Paone F., Calore R., et al. Micrornas bind to toll-like receptors to induce prometastatic inflammatory response. Proceedings of the National Academy of Sciences . 2012;109(31):6. doi: 10.1073/pnas.1209414109.E2110 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Rabinowits G., Gerçel-Taylor C., Day J. M., Taylor D. D., Kloecker G. H. Exosomal microRNA: a diagnostic marker for lung cancer. Clinical Lung Cancer . 2009;10(1):42–46. doi: 10.3816/clc.2009.n.006. [DOI] [PubMed] [Google Scholar]
- 55.Yanaihara N., Caplen N., Bowman E., et al. Unique microRNA molecular profiles in lung cancer diagnosis and prognosis. Cancer Cell . 2006;9(3):189–198. doi: 10.1016/j.ccr.2006.01.025. [DOI] [PubMed] [Google Scholar]
- 56.Lawrie C. H., Gal S., Dunlop H. M., et al. Detection of elevated levels of tumour-associated microRNAs in serum of patients with diffuse large B-cell lymphoma. British Journal of Haematology . 2008;141(5):672–675. doi: 10.1111/j.1365-2141.2008.07077.x. [DOI] [PubMed] [Google Scholar]
- 57.Umezu T., Tadokoro H., Azuma K., Yoshizawa S., Ohyashiki K., Ohyashiki J. H. Exosomal miR-135b shed from hypoxic multiple myeloma cells enhances angiogenesis by targeting factor-inhibiting HIF-1. Blood . 2014;124(25):3748–3757. doi: 10.1182/blood-2014-05-576116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Zhou J., Gong G., Tan H., et al. Urinary microRNA-30a-5p is a potential biomarker for ovarian serous adenocarcinoma. Oncology Reports . 2015;33(6):2915–2923. doi: 10.3892/or.2015.3937. [DOI] [PubMed] [Google Scholar]
- 59.Taylor D. D., Gercel-Taylor C. MicroRNA signatures of tumor-derived exosomes as diagnostic biomarkers of ovarian cancer. Gynecologic Oncology . 2008;110(1):13–21. doi: 10.1016/j.ygyno.2008.04.033. [DOI] [PubMed] [Google Scholar]
- 60.Cappellesso R., Tinazzi A., Giurici T., et al. Programmed cell death 4 and microRNA 21 inverse expression is maintained in cells and exosomes from ovarian serous carcinoma effusions. Cancer Cytopathology . 2014;122(9):685–693. doi: 10.1002/cncy.21442. [DOI] [PubMed] [Google Scholar]
- 61.Madhavan B., Yue S., Galli U., et al. Combined evaluation of a panel of protein and miRNA serum-exosome biomarkers for pancreatic cancer diagnosis increases sensitivity and specificity. International Journal of Cancer . 2015;136(11):2616–2627. doi: 10.1002/ijc.29324. [DOI] [PubMed] [Google Scholar]
- 62.Elmageed Z. Y. A., Yang Y., Thomas R., et al. Neoplastic reprogramming of patient-derived adipose stem cells by prostate cancer cell-associated exosomes. Stem Cells . 2014;32(4):983–997. doi: 10.1002/stem.1619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Rodríguez M., Bajo-Santos C., Hessvik N. P., et al. Identification of non-invasive miRNAs biomarkers for prostate cancer by deep sequencing analysis of urinary exosomes. Molecular Cancer . 2017;16(1):p. 156. doi: 10.1186/s12943-017-0726-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Bryant R. J., Pawlowski T., Catto J. W. F., et al. Changes in circulating microRNA levels associated with prostate cancer. British Journal of Cancer . 2012;106(4):768–774. doi: 10.1038/bjc.2011.595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Huang X., Yuan T., Liang M., et al. Exosomal miR-1290 and miR-375 as prognostic markers in castration-resistant prostate cancer. European Urology . 2015;67(1):33–41. doi: 10.1016/j.eururo.2014.07.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Joshi G. K., Deitz-McElyea S., Liyanage T., et al. Label-free nanoplasmonic-based short noncoding RNA sensing at attomolar concentrations allows for quantitative and highly specific assay of microrna-10b in biological fluids and circulating exosomes. ACS Nano . 2015;9(11):11075–11089. doi: 10.1021/acsnano.5b04527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Lai X., Wang M., McElyea S. D., Sherman S., House M., Korc M. A microRNA signature in circulating exosomes is superior to exosomal glypican-1 levels for diagnosing pancreatic cancer. Cancer Letters . 2017;393:86–93. doi: 10.1016/j.canlet.2017.02.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Ding Y., Cao F., Sun H., et al. Exosomes derived from human umbilical cord mesenchymal stromal cells deliver exogenous miR-145-5p to inhibit pancreatic ductal adenocarcinoma progression. Cancer Letters . 2019;442:351–361. doi: 10.1016/j.canlet.2018.10.039. [DOI] [PubMed] [Google Scholar]
- 69.Yang L., Li Y., Gong R., et al. The long non-coding RNA-ORLNC1 regulates bone mass by directing mesenchymal stem cell fate. Molecular Therapy . 2019;27(2):394–410. doi: 10.1016/j.ymthe.2018.11.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Cheng J., Meng J., Zhu L., Peng Y. Exosomal noncoding RNAs in glioma: biological functions and potential clinical applications. Molecular Cancer . 2020;19(1):p. 66. doi: 10.1186/s12943-020-01189-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Dai X., Liao K., Zhuang Z., et al. AHIF promotes glioblastoma progression and radioresistance via exosomes. International Journal of Oncology . 2019;54(1):261–270. doi: 10.3892/ijo.2018.4621. [DOI] [PubMed] [Google Scholar]
- 72.Wang M., Zhou L., Yu F., Zhang Y., Li P., Wang K. The functional roles of exosomal long non-coding RNAs in cancer. Cellular and Molecular Life Sciences . 2019;76(11):2059–2076. doi: 10.1007/s00018-019-03018-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Dong H., Wang R., Chen Y., et al. Exosome-mediated transfer of lncRNA‑SNHG14 promotes trastuzumab chemoresistance in breast cancer. International Journal of Oncology . 2018;53(3):1013–1026. doi: 10.3892/ijo.2018.4467. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 74.Xu C. G., Yang M. F., Ren Y. Q., Wu C. H., Wang L. Q. Exosomes mediated transfer of lncRNA UCA1 results in increased tamoxifen resistance in breast cancer cells. European review for medical and pharmacological sciences . 2016;20(20):4362–4368. [PubMed] [Google Scholar]
- 75.Zheng Z., Chen M., Xing P., Yan X., Xie B. Increased expression of exosomal AGAP2-AS1 (AGAP2 antisense RNA 1) in breast cancer cells inhibits trastuzumab-induced cell cytotoxicity. Medical Science Monitor . 2019;25:2211–2220. doi: 10.12659/msm.915419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Han M., Gu Y., Lu P., et al. Exosome-mediated lncRNA AFAP1-AS1 promotes trastuzumab resistance through binding with AUF1 and activating ERBB2 translation. Molecular Cancer . 2020;19(1):p. 26. doi: 10.1186/s12943-020-1145-5. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 77.Wang X., Pei X., Guo G., et al. Exosome‐mediated transfer of long noncoding RNA H19 induces doxorubicin resistance in breast cancer. Journal of Cellular Physiology . 2020;235(10):6896–6904. doi: 10.1002/jcp.29585. [DOI] [PubMed] [Google Scholar]
- 78.Hu D., Zhan Y., Zhu K., et al. Plasma exosomal long non-coding RNAs serve as biomarkers for early detection of colorectal cancer. Cellular Physiology and Biochemistry . 2018;51(6):2704–2715. doi: 10.1159/000495961. [DOI] [PubMed] [Google Scholar]
- 79.Zhao Y., Du T., Du L., et al. Long noncoding RNA LINC02418 regulates MELK expression by acting as a ceRNA and may serve as a diagnostic marker for colorectal cancer. Cell Death & Disease . 2019;10(8):p. 568. doi: 10.1038/s41419-019-1804-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Liang Z. X., Liu H. S., Wang F. W., et al. LncRNA RPPH1 promotes colorectal cancer metastasis by interacting with TUBB3 and by promoting exosomes-mediated macrophage M2 polarization. Cell Death & Disease . 2019;10(11):p. 829. doi: 10.1038/s41419-019-2077-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Liu T., Zhang X., Gao S., et al. Exosomal long noncoding RNA CRNDE-h as a novel serum-based biomarker for diagnosis and prognosis of colorectal cancer. Oncotarget . 2016;7(51):85551–85563. doi: 10.18632/oncotarget.13465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Zhao R., Zhang Y., Zhang X., et al. Exosomal long noncoding RNA HOTTIP as potential novel diagnostic and prognostic biomarker test for gastric cancer. Molecular Cancer . 2018;17(1):p. 68. doi: 10.1186/s12943-018-0817-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Conigliaro A., Costa V., Lo Dico A., et al. CD90+ liver cancer cells modulate endothelial cell phenotype through the release of exosomes containing H19 lncRNA. Molecular Cancer . 2015;14(1):p. 155. doi: 10.1186/s12943-015-0426-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Zhang R., Xia Y., Wang Z., et al. Serum long non coding RNA MALAT-1 protected by exosomes is up-regulated and promotes cell proliferation and migration in non-small cell lung cancer. Biochemical and Biophysical Research Communications . 2017;490(2):406–414. doi: 10.1016/j.bbrc.2017.06.055. [DOI] [PubMed] [Google Scholar]
- 85.Qu L., Ding J., Chen C., et al. Exosome-transmitted lncarsr promotes sunitinib resistance in renal cancer by acting as a competing endogenous RNA. Cancer Cell . 2016;29(5):653–668. doi: 10.1016/j.ccell.2016.03.004. [DOI] [PubMed] [Google Scholar]
- 86.Léveillé N., Baglio S. R. Exosome-transferred lncRNAs at the core of cancer bone lesions. Critical Reviews in Oncology/Hematology . 2019;139:125–127. doi: 10.1016/j.critrevonc.2019.03.002. [DOI] [PubMed] [Google Scholar]
- 87.Zhao H., Chen S., Fu Q. Exosomes from CD133 + cells carrying circ‐ABCC1 mediate cell stemness and metastasis in colorectal cancer. Journal of Cellular Biochemistry . 2020;121(5-6):3286–3297. doi: 10.1002/jcb.29600. [DOI] [PubMed] [Google Scholar]
- 88.Li Y., Zheng Q., Bao C., et al. Circular RNA is enriched and stable in exosomes: a promising biomarker for cancer diagnosis. Cell Research . 2015;25(8):981–984. doi: 10.1038/cr.2015.82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Pan B., Qin X., Liu B., et al. Identification of serum exosomal hsa-circ-0004771 as a novel diagnostic biomarker of colorectal cancer. Frontiers in Genetics . 2019;10:p. 1096. doi: 10.3389/fgene.2019.01096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Zhang H., Zhu L., Bai M., et al. Exosomal circRNA derived from gastric tumor promotes white adipose browning by targeting the miR‐133/PRDM16 pathway. International Journal of Cancer . 2019;144(10):2501–2515. doi: 10.1002/ijc.31977. [DOI] [PubMed] [Google Scholar]
- 91.Xie M., Yu T., Jing X., et al. Exosomal circSHKBP1 promotes gastric cancer progression via regulating the miR-582-3p/HUR/VEGF axis and suppressing HSP90 degradation. Molecular Cancer . 2020;19(1):p. 112. doi: 10.1186/s12943-020-01208-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Lu J., Wang Y. H., Yoon C., et al. Circular RNA circ-RanGAP1 regulates VEGFA expression by targeting miR-877-3p to facilitate gastric cancer invasion and metastasis. Cancer Letters . 2020;471:38–48. doi: 10.1016/j.canlet.2019.11.038. [DOI] [PubMed] [Google Scholar]
- 93.Zhong Y., Wang D., Ding Y., Tian G., Jiang B. Circular RNA circ_0032821 contributes to oxaliplatin (OXA) resistance of gastric cancer cells by regulating SOX9 via miR-515-5p. Biotechnology Letters . 2021;43(2):339–351. doi: 10.1007/s10529-020-03036-3. [DOI] [PubMed] [Google Scholar]
- 94.Yin K., Liu X. CircMMP1 promotes the progression of glioma through miR ‐433/ HMGB3 axis in vitro and in vivo. IUBMB Life . 2020;72(11):2508–2524. doi: 10.1002/iub.2383. [DOI] [PubMed] [Google Scholar]
- 95.Li Y., Zang H., Zhang X., Huang G. Exosomal circ-ZNF652 promotes cell proliferation, migration, invasion and glycolysis in hepatocellular carcinoma via miR-29a-3p/GUCD1 axis. Cancer Management and Research . 2020;Volume 12:7739–7751. doi: 10.2147/cmar.s259424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Dai X., Chen C., Yang Q., et al. Exosomal circRNA_100284 from arsenite-transformed cells, via microRNA-217 regulation of EZH2, is involved in the malignant transformation of human hepatic cells by accelerating the cell cycle and promoting cell proliferation. Cell Death & Disease . 2018;9(5):p. 454. doi: 10.1038/s41419-018-0485-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Zhang H., Deng T., Ge S., et al. Exosome circRNA secreted from adipocytes promotes the growth of hepatocellular carcinoma by targeting deubiquitination-related USP7. Oncogene . 2019;38(15):2844–2859. doi: 10.1038/s41388-018-0619-z. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 98.Huang X. Y., Huang Z. L., Huang J., et al. Exosomal circRNA-100338 promotes hepatocellular carcinoma metastasis via enhancing invasiveness and angiogenesis. Journal of Experimental & Clinical Cancer Research . 2020;39(1):p. 20. doi: 10.1186/s13046-020-1529-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Su Y., Lv X., Yin W., et al. CircRNA Cdr1as functions as a competitive endogenous RNA to promote hepatocellular carcinoma progression. Aging . 2019;11(19):8183–8203. doi: 10.18632/aging.102312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Chen W., Quan Y., Fan S., et al. Exosome-transmitted circular RNA hsa_circ_0051443 suppresses hepatocellular carcinoma progression. Cancer Letters . 2020;475:119–128. doi: 10.1016/j.canlet.2020.01.022. [DOI] [PubMed] [Google Scholar]
- 101.Wang G., Liu W., Zou Y., et al. Three isoforms of exosomal circPTGR1 promote hepatocellular carcinoma metastasis via the miR449a-MET pathway. EBioMedicine . 2019;40:432–445. doi: 10.1016/j.ebiom.2018.12.062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Zhang P. F., Gao C., Huang X. Y., et al. Cancer cell-derived exosomal circUHRF1 induces natural killer cell exhaustion and may cause resistance to anti-PD1 therapy in hepatocellular carcinoma. Molecular Cancer . 2020;19(1):p. 110. doi: 10.1186/s12943-020-01222-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Luo Y., Liu F., Gui R. High expression of circulating exosomal circAKT3 is associated with higher recurrence in HCC patients undergoing surgical treatment. Surgical Oncology . 2020;33:276–281. doi: 10.1016/j.suronc.2020.04.021. [DOI] [PubMed] [Google Scholar]
- 104.Tian L., Cao J., Jiao H., et al. CircRASSF2 promotes laryngeal squamous cell carcinoma progression by regulating the miR-302b-3p/IGF-1R axis. Clinical Science . 2019;133(9):1053–1066. doi: 10.1042/cs20190110. [DOI] [PubMed] [Google Scholar]
- 105.Wang J., Zhao X., Wang Y., et al. circRNA-002178 act as a ceRNA to promote PDL1/PD1 expression in lung adenocarcinoma. Cell Death & Disease . 2020;11(1):p. 32. doi: 10.1038/s41419-020-2230-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Luo Y., Gui R. Circulating exosomal circMYC is associated with recurrence and bortezomib resistance in patients with multiple myeloma. Turkish Journal of Hematology . 2020;37(4):248–255. doi: 10.4274/tjh.galenos.2020.2020.0243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Zhang Y., Tang K., Chen L., Du M., Qu Z. Exosomal circGDI2 suppresses oral squamous cell carcinoma progression through the regulation of miR-424-5p/SCAI axis. Cancer Management and Research . 2020;12:7501–7514. doi: 10.2147/cmar.s255687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Li T., Sun X., Chen L. Exosome circ_0044516 promotes prostate cancer cell proliferation and metastasis as a potential biomarker. Journal of Cellular Biochemistry . 2020;121(3):2118–2126. doi: 10.1002/jcb.28239. [DOI] [PubMed] [Google Scholar]
- 109.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 Letters . 2018;432:237–250. doi: 10.1016/j.canlet.2018.04.035. [DOI] [PubMed] [Google Scholar]
- 110.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. Journal of Experimental & Clinical Cancer Research . 2018;37(1):p. 177. doi: 10.1186/s13046-018-0822-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Chen X., Chen R. X., Wei W. S., et al. PRMT5 circular rna promotes metastasis of urothelial carcinoma of the bladder through sponging mir-30c to induce epithelial-mesenchymal transition. Clinical Cancer Research . 2018;24(24):6319–6330. doi: 10.1158/1078-0432.ccr-18-1270. [DOI] [PubMed] [Google Scholar]
- 112.Milane L., Singh A., Mattheolabakis G., Suresh M., Amiji M. M. Exosome mediated communication within the tumor microenvironment. Journal of Controlled Release . 2015;219:278–294. doi: 10.1016/j.jconrel.2015.06.029. [DOI] [PubMed] [Google Scholar]
- 113.Zhao X., Wu D., Ma X., Wang J., Hou W., Zhang W. Exosomes as drug carriers for cancer therapy and challenges regarding exosome uptake. Biomedicine & Pharmacotherapy . 2020;128 doi: 10.1016/j.biopha.2020.110237.110237 [DOI] [PubMed] [Google Scholar]
- 114.Rivoltini L., Chiodoni C., Squarcina P., et al. TNF-related apoptosis-inducing ligand (TRAIL)-armed exosomes deliver proapoptotic signals to tumor site. Clinical Cancer Research . 2016;22(14):3499–3512. doi: 10.1158/1078-0432.ccr-15-2170. [DOI] [PubMed] [Google Scholar]
- 115.Koh E., Lee E. J., Nam G. H., et al. Exosome-SIRPα, a CD47 blockade increases cancer cell phagocytosis. Biomaterials . 2017;121:121–129. doi: 10.1016/j.biomaterials.2017.01.004. [DOI] [PubMed] [Google Scholar]
- 116.Tiedemann K., Sadvakassova G., Mikolajewicz N., et al. Exosomal release of l-plastin by breast cancer cells facilitates metastatic bone osteolysis. Translational Oncology . 2019;12(3):462–474. doi: 10.1016/j.tranon.2018.11.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Tian Y., Ma L., Gong M., et al. Protein profiling and sizing of extracellular vesicles from colorectal cancer patients via flow cytometry. ACS Nano . 2018;12(1):671–680. doi: 10.1021/acsnano.7b07782. [DOI] [PubMed] [Google Scholar]
- 118.Sun B., Zhou Y., Fang Y., Li Z., Gu X., Xiang J. Colorectal cancer exosomes induce lymphatic network remodeling in lymph nodes. International Journal of Cancer . 2019;145(6):1648–1659. doi: 10.1002/ijc.32196. [DOI] [PubMed] [Google Scholar]
- 119.Skog J., Würdinger T., van Rijn S., et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nature Cell Biology . 2008;10(12):1470–1476. doi: 10.1038/ncb1800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Graner M. W., Alzate O., Dechkovskaia A. M., et al. Proteomic and immunologic analyses of brain tumor exosomes. The FASEB Journal . 2009;23(5):1541–1557. doi: 10.1096/fj.08-122184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Shao H., Chung J., Balaj L., et al. Protein typing of circulating microvesicles allows real-time monitoring of glioblastoma therapy. Nature Medicine . 2012;18(12):1835–1840. doi: 10.1038/nm.2994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Treps L., Perret R., Edmond S., Ricard D., Gavard J. Glioblastoma stem-like cells secrete the pro-angiogenic VEGF-A factor in extracellular vesicles. Journal of Extracellular Vesicles . 2017;6(1) doi: 10.1080/20013078.2017.1359479.1359479 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Liu D., Li C., Trojanowicz B., et al. CD97 promotion of gastric carcinoma lymphatic metastasis is exosome dependent. Gastric Cancer . 2016;19(3):754–766. doi: 10.1007/s10120-015-0523-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Zhang X., Shi H., Yuan X., Jiang P., Qian H., Xu W. Tumor-derived exosomes induce N2 polarization of neutrophils to promote gastric cancer cell migration. Molecular Cancer . 2018;17(1):p. 146. doi: 10.1186/s12943-018-0898-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Madhankumar A. B., Mrowczynski O. D., Patel S. R., et al. Interleukin-13 conjugated quantum dots for identification of glioma initiating cells and their extracellular vesicles. Acta Biomaterialia . 2017;58:205–213. doi: 10.1016/j.actbio.2017.06.002. [DOI] [PubMed] [Google Scholar]
- 126.Sun Y., Huo C., Qiao Z., et al. Comparative proteomic analysis of exosomes and microvesicles in human saliva for lung cancer. Journal of Proteome Research . 2018;17(3):1101–1107. doi: 10.1021/acs.jproteome.7b00770. [DOI] [PubMed] [Google Scholar]
- 127.Li Y., Zhang Y., Qiu F., Qiu Z. Proteomic identification of exosomal LRG1: a potential urinary biomarker for detecting NSCLC. Electrophoresis . 2011;32(15):1976–1983. doi: 10.1002/elps.201000598. [DOI] [PubMed] [Google Scholar]
- 128.Peinado H., Alečković M., Lavotshkin S., et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nature Medicine . 2012;18(6):883–891. doi: 10.1038/nm.2753. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Logozzi M., De Milito A., Lugini L., et al. High levels of exosomes expressing CD63 and caveolin-1 in plasma of melanoma patients. PLoS One . 2009;4(4) doi: 10.1371/journal.pone.0005219.e5219 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.André F., Chaput N., Schartz N. E. C., et al. Exosomes as potent cell-free peptide-based vaccine. I. Dendritic cell-derived exosomes transfer functional MHC class I/peptide complexes to dendritic cells. The Journal of Immunology . 2004;172(4):2126–2136. doi: 10.4049/jimmunol.172.4.2126. [DOI] [PubMed] [Google Scholar]
- 131.Carrasco-Ramírez P., Greening D. W., Andrés G., et al. Podoplanin is a component of extracellular vesicles that reprograms cell-derived exosomal proteins and modulates lymphatic vessel formation. Oncotarget . 2016;7(13):16070–16089. doi: 10.18632/oncotarget.7445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Xie Y., Bai O., Zhang H., et al. Membrane-bound HSP70-engineered myeloma cell-derived exosomes stimulate more efficient CD8+ CTL- and NK-mediated antitumour immunity than exosomes released from heat-shocked tumour cells expressing cytoplasmic HSP70. Journal of Cellular and Molecular Medicine . 2010;14(11):2655–2666. doi: 10.1111/j.1582-4934.2009.00851.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Taverna S., Pucci M., Giallombardo M., et al. Amphiregulin contained in NSCLC-exosomes induces osteoclast differentiation through the activation of EGFR pathway. Scientific Reports . 2017;7(1):p. 3170. doi: 10.1038/s41598-017-03460-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Shender V. O., Pavlyukov M. S, Ziganshin R. H., et al. Proteome-metabolome profiling of ovarian cancer ascites reveals novel components involved in intercellular communication. Molecular & Cellular Proteomics . 2014;13(12):3558–3571. doi: 10.1074/mcp.m114.041194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Im H., Shao H., Park Y. I., et al. Label-free detection and molecular profiling of exosomes with a nano-plasmonic sensor. Nature Biotechnology . 2014;32(5):490–495. doi: 10.1038/nbt.2886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Melo S. A., Luecke L. B., Kahlert C., et al. Glypican-1 identifies cancer exosomes and detects early pancreatic cancer. Nature . 2015;523(7559):177–182. doi: 10.1038/nature14581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Duijvesz D., Burnum-Johnson K. E., Gritsenko M. A., et al. Proteomic profiling of exosomes leads to the identification of novel biomarkers for prostate cancer. PLoS One . 2013;8(12) doi: 10.1371/journal.pone.0082589.e82589 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Lu H., Bowler N., Harshyne L. A., et al. Exosomal αvβ6 integrin is required for monocyte M2 polarization in prostate cancer. Matrix Biology . 2018;70:20–35. doi: 10.1016/j.matbio.2018.03.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Costa-Silva B., Aiello N. M., Ocean A. J., et al. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nature Cell Biology . 2015;17(6):816–826. doi: 10.1038/ncb3169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Yue S., Mu W., Erb U., Zöller M. The tetraspanins CD151 and Tspan8 are essential exosome components for the crosstalk between cancer initiating cells and their surrounding. Oncotarget . 2015;6(4):2366–2384. doi: 10.18632/oncotarget.2958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Yamashita T., Kamada H., Kanasaki S., et al. Epidermal growth factor receptor localized to exosome membranes as a possible biomarker for lung cancer diagnosis. Die Pharmazie . 2013;68(12):969–73. [PubMed] [Google Scholar]
- 142.Huang S. H., Li Y., Zhang J., Rong J., Ye S. Epidermal growth factor receptor-containing exosomes induce tumor-specific regulatory T cells. Cancer Investigation . 2013;31(5):330–335. doi: 10.3109/07357907.2013.789905. [DOI] [PubMed] [Google Scholar]
- 143.Sandfeld-Paulsen B., Jakobsen K. R., Bæk R., et al. Exosomal proteins as diagnostic biomarkers in lung cancer. Journal of Thoracic Oncology . 2016;11(10):1701–1710. doi: 10.1016/j.jtho.2016.05.034. [DOI] [PubMed] [Google Scholar]
- 144.Hosseini M., Khatamianfar S., Hassanian S. M., et al. Exosome-encapsulated microRNAs as potential circulating biomarkers in colon cancer. Current Pharmaceutical Design . 2017;23(11):1705–1709. doi: 10.2174/1381612822666161201144634. [DOI] [PubMed] [Google Scholar]
- 145.Tsukamoto M., Iinuma H., Yagi T., Matsuda K., Hashiguchi Y. Circulating Exosomal MicroRNA-21 as a Biomarker in Each Tumor Stage of Colorectal Cancer. Oncology . 2017;92(6):360–370. doi: 10.1159/000463387. [DOI] [PubMed] [Google Scholar]
- 146.Yang H., Fu H., Wang B., et al. Exosomal miR-423-5p targets SUFU to promote cancer growth and metastasis and serves as a novel marker for gastric cancer. Molecular Carcinogenesis . 2018;57(9):1223–1236. doi: 10.1002/mc.22838. [DOI] [PubMed] [Google Scholar]
- 147.Kim J. E., Eom J. S., Kim W. Y., et al. Diagnostic value of microRNAs derived from exosomes in bronchoalveolar lavage fluid of early-stage lung adenocarcinoma: A pilot study. Thoracic Cancer . 2018;9(8):911–915. doi: 10.1111/1759-7714.12756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Bhat E. A., Sajjad N., Thokar F. M. Current advancement of exosomes as biomarkers for cancer diagnosis and forecasting. Cancer Treatment and Research Communications . 2021;28 doi: 10.1016/j.ctarc.2021.100417.100417 [DOI] [PubMed] [Google Scholar]
- 149.Wang Y. H., Ji J., Wang B. C., et al. Tumor-derived exosomal long noncoding rnas as promising diagnostic biomarkers for prostate cancer. Cellular Physiology and Biochemistry . 2018;46(2):532–545. doi: 10.1159/000488620. [DOI] [PubMed] [Google Scholar]
- 150.Williams C., Royo F., Aizpurua-Olaizola O., et al. Glycosylation of extracellular vesicles: current knowledge, tools and clinical perspectives. Journal of Extracellular Vesicles . 2018;7(1) doi: 10.1080/20013078.2018.1442985.1442985 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Øverbye A., Skotland T., Koehler C. J., et al. Identification of prostate cancer biomarkers in urinary exosomes. Oncotarget . 2015;6(30):30357–30376. doi: 10.18632/oncotarget.4851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Khan S., Bennit H. F., Turay D., et al. Early diagnostic value of survivin and its alternative splice variants in breast cancer. BMC Cancer . 2014;14(1):p. 176. doi: 10.1186/1471-2407-14-176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Yang J. K., Song J., Huo H. R, et al. DNM3, p65 and p53 from exosomes represent potential clinical diagnosis markers for glioblastoma multiforme. Therapeutic Advances in Medical Oncology . 2017;9(12):741–754. doi: 10.1177/1758834017737471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Kamerkar S., LeBleu V. S., Sugimoto H., et al. Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer. Nature . 2017;546(7659):498–503. doi: 10.1038/nature22341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Mendt M., Kamerkar H., Sugimoto K. M., et al. Generation and testing of clinical-grade exosomes for pancreatic cancer. JCI Insight . 2018;3(8) doi: 10.1172/jci.insight.99263.99263 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Kim M. S., Haney M. J., Zhao Y., et al. Engineering macrophage-derived exosomes for targeted paclitaxel delivery to pulmonary metastases: in vitro and in vivo evaluations. Nanomedicine: Nanotechnology, Biology and Medicine . 2018;14(1):195–204. doi: 10.1016/j.nano.2017.09.011. [DOI] [PubMed] [Google Scholar]
- 157.Tian Y., Li S., Song J., et al. A doxorubicin delivery platform using engineered natural membrane vesicle exosomes for targeted tumor therapy. Biomaterials . 2014;35(7):2383–2390. doi: 10.1016/j.biomaterials.2013.11.083. [DOI] [PubMed] [Google Scholar]
- 158.Tang K., Zhang Y., Zhang H., et al. Delivery of chemotherapeutic drugs in tumour cell-derived microparticles. Nature Communications . 2012;3(1):p. 1282. doi: 10.1038/ncomms2282. [DOI] [PubMed] [Google Scholar]