Open Access

Clinical significance of exosomal miRNAs and proteins in three human cancers with high mortality in China (Review)

  • Authors:
    • Li‑Man Li
    • Huan Liu
    • Xing‑Hui Liu
    • Hong‑Bin Hu
    • Song‑Mei Liu
  • View Affiliations

  • Published online on: October 29, 2018     https://doi.org/10.3892/ol.2018.9631
  • Pages: 11-22
  • Copyright: © Li et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

Cancer is the second leading cause of mortality worldwide. More importantly, the mortality rates for cancer are increasing. In China, lung cancer, liver cancer and gastric cancer are the top three leading causes of mortality in males, whereas lung cancer, gastric cancer and liver cancer are ranked the top three causes of mortality in females. Exosomes are extracellular vesicles that are produced and released by many different cells; these vesicles have a size range between 30 and 100 nm in diameter, and contain a lipid bilayer. Exosomes exist in various bodily fluids, contain plentiful amounts of nucleic acids and proteins, and shuttle these materials between cells to mediate the development of cancers. The present review summarizes the composition of exosomes and methods for their isolation and then intensively highlights the latest findings on the contributions of exosomal microRNAs (miRNAs) and proteins to lung cancer, liver cancer and gastric cancer. Taken together, exosomal miRNAs and proteins may be used as noninvasive, novel biomarkers for cancer diagnosis, prognosis or precision treatment owing to their ability to promote tumor progression and metastasis, and their ability to regulate the immune response and tumor cell sensitivity to chemotherapy drugs.

Introduction

Cancer is the second leading cause of death globally (1). In China, the numbers of newly diagnosed cases and deaths were approximately 3.0 million and 1.9 million, respectively, in 2010 (2). According to 2013 data, lung cancer, liver cancer and gastric cancer are the top three leading causes of mortality in males in China, whereas lung cancer, gastric cancer and liver cancer are the top three leading causes of mortality in females (3) (Fig. 1).

A growing number of studies have focused on the biology, function and clinical implications of exosomes in cancers (4,5), and it has been demonstrated that exosomal miRNAs and proteins can act as tumor biomarkers for clinical diagnosis or prognosis and that exosomes shuttle between cells to exchange genetic material, which promotes tumor progression, metastasis and prognosis; regulates the immune response; and affects the sensitivity of tumor cells to chemotherapy drugs (68). Therefore, exosomal miRNAs and proteins potentially play critical roles in cancers with high mortality rates.

Exosome composition

Exosomes are extracellular vesicles (EVs) that are produced and released by many different cells; and these vesicles range in size from 30 to 100 nm in diameter and contain a lipid bilayer (9,10). Proteins, DNA, mRNAs, miRNAs and lipids are enriched in exosomes (11). Exosomes transfer nucleic acids and proteins between different cells, leading to both the transportation of materials and cell-cell communication (6,12,13).

A set of distinct proteins are contained in exosomes (14), including heat-shock proteins (Hsp70, Hsp90), tetraspanins (CD9, CD81), ESCRT-related proteins (Alix, Tsg101), cytoskeletal proteins (actin, Tubulin) and GTPases (EEF1A1, EEF2) (15,16). These proteins are known to be involved in biogenesis, the sorting and secretion of exosomes (17), antigen presentation, the organization of membrane microdomains, the cytoskeleton, and the endosomal system (18,19). Typically, exosomes contain both cell-type specific proteins and proteins that are expressed in various cell types (20).

In addition to proteins, exosomes contain a significant amount of nucleic acids, including DNA, mRNAs, miRNAs, circular RNAs (circRNAs) and long noncoding RNAs (lncRNAs) (21). Of these, miRNAs are a class of well-known regulatory molecules that control posttranscriptional gene regulation (22). Increasing evidence has shown that exosomal miRNAs are associated with many diseases, such as cancers, diabetes and obesity (2326). Interestingly, the miRNA content of exosomes is similar to that of the original tumor; thus, a series of studies has focused on exosomal miRNA profiles for cancer diagnostics (16). In particular, the shuttling of miRNAs may act as a tumor promoter or a tumor suppressor during tumorigenesis (27). Previous studies have uncovered exosomal miRNAs that are closely associated with tumorigenesis, metastasis and drug resistance in various kinds of cancers (28,29). All of these findings suggest that exosomal miRNAs play a pivotal role in the diagnosis, treatment and prognosis of cancers (30,31).

Additionally, cholesterols, diglycerides, phospholipids, glycerophospholipids, sphingomyelins and ceramides are enriched in exosomes (32). These lipids participate in exosome biogenesis, function and release. For example, the cellular trafficking of the tetraspanin CD82 to endosomes is regulated by the cholesterol content of the membrane, and ceramides can protect miRNAs from degradation by circulating RNases and govern the cellular distribution of the tetraspanin CD81. In addition, bioactive lipids such as prostaglandins, leukotrienes, fatty acids and lipid-related enzymes such as phospholipases A2 have been detected in exosomes (33).

Exosome isolation

Exosomes secreted by various types of living cells have been detected in a diverse range of bodily fluids, including peripheral blood, saliva, cerebrospinal fluid, ascites fluid, amniotic fluid, urine, breast milk and semen (31,34) (Fig. 2). It is clear that the utility of exosomes goes beyond basic research and extends to clinical practice. For this reason, an efficient and accurate method for exosome isolation is crucial.

Here, we compare the common methods for exosome isolation (Table I), including ultracentrifugation (UC), ultrafiltration (UF), immunomagnetic beads, size exclusion chromatography (SEC) and ExoQuick (35,36). UC is a common and simple method (37); however, recent studies indicated that more contaminants were found in exosomes isolated by UC compared to other methods mainly due to the presence of albumins. Furthermore, the high-velocity ultracentrifugation process could cause some exosomes to rupture, resulting in exosome loss (38). Recently, the challenges of UC approach have been again discussed, the conventional biophysical UC cannot distinguish exosomes from lipoproteins and oncosomes, other types of small EVs with sedimentation velocities and gradient densities similar to those of exosomes (39). UF does not require special equipment, although it leads to a reduction in the membranes' lifespan and a low isolation efficiency (35,40). The use of immunomagnetic beads is an alternative method with high specificity and purity, but it is limited to exosomes with a known antigen and has a high reagent cost (35). Although SEC does not lead to significant albumin contamination, the efficiency is low (35,37,41). ExoQuick produces excellent reproducibility and sensitivity. However, the proprietary reagents exhibit contamination from unknown sources, and the polymer leads to protein aggregation (35,36,42,43). Moreover, the ExoQuick kit does not specifically precipitate exosomes, which means that other types of nanovesicles with similar sizes (30–100 nm) might also be coprecipitated (39). Recently, a new technique developed by the microfluidics community has been used to approach some of the problems with exosome isolation mentioned above. The most important feature of this method is exosome enrichment during isolation, which is beneficial for the detection of early-stage cancers. This microfluidics approach showed a superior recovery of 60–80% compared to the conventional techniques of UC (6%) and ExoQuick (30%) based on nanoparticle tracking analysis (NTA) (43).

Table I.

Comparison of exosome isolation methods.

Table I.

Comparison of exosome isolation methods.

Author, yearMethodPrincipleAdvantagesDisadvantages(Refs.)
Baranyai et al, 2015; Peterson et al, 2015UCSeparating the exosomes through differential mass, density and shape• Available technology• The high velocity ultracentrifugation process could cause some exosomes rupture that results in some exosomes loss(37,38)
• Simple operation
• Contaminated with albumin and IgG
• Time consuming (16–20 h)
Li et al, 2017; Zeringer et al, 2015UFDepending on exosomal size or molecular weight• No need of special equipment• Clogging and vesicle trapping lead to reduce the membranes' lifetime and low isolation efficiency(35,40)
• Good portability
Li et al, 2017Immunom-agnetic beadsSpecific exosomal antigens (receptors) can be captured by magnetic beads (ligands)• High specificity and purity• High reagent cost(35)
• Low yield
• No damage on the integrity of the exosomes' morphology and structure
Li et al, 2017; Baranyai et al, 2015; Taylor and Shah, 2015SECA porous stationary phase is utilized to sort exosomes out according to the size• Obtaining high-purity exosomes without significant albumin contamination• Require dedicated equipment(35,37,41)
• Low efficiency
• Excellent reproducibility and sensitivity
Li et al, 2017; Caradec et al, 2014; Ban et al, 2015ExoQuick™By the precipitation approach• Efficient (around 100%) and reproducible• Isolation procedure should be under acidic conditions (pH=4)(35,36,42)
• Decreasing albumin contamination• Polymer precipitates protein aggregation
• Fast (within 30 min)

[i] UC, ultracentrifugation; UF, ultrafiltration; SEC, size exclusion chromatography.

Indeed, the high quantity and purity of exosomes are extremely important for exosomal biology studies. Thus, western blotting should be used to determine whether exosomal protein markers (Alix, Tsg101, Hsp70 or others) are present in exosome isolations (44). Simultaneously, transmission electron microscopy (TEM) is often utilized to observe exosome morphology, NTA is used to measure particle size, and the bicinchoninic acid assay (BCA) is performed to examine the protein concentration of exosomes (45). Additionally, to ensure the sensitivity of isolations and achieve a robust result, pre-analytical factors should be taken into consideration (Table II) (46,47).

Table II.

Pre-analytical considerations.

Table II.

Pre-analytical considerations.

Author, yearPre-analytical considerations(Refs.)
Muller et al, 2014; Witwer et al, 2013Venous blood from patients or healthy volunteers is collected into tubes without heparin-based anticoagulants, EDTA may be more appropriate.(46,47)
Witwer et al, 2013Blood should be processed quickly at room temperature.(47)
Witwer et al, 2013Collected blood should be handled gently and tubes should be vertically positioned prior to centrifugation.(47)
Witwer et al, 2013Both plasma and serum can be used, but most studies indicate the isolation of exosomes prefers to plasma.(47)
Muller et al, 2014Harvested plasma or serum should be immediately used or stored at −80°C.(46)

Exosomal miRNAs and proteins in lung cancer

The latest report showed that lung cancer caused approximately 597,000 deaths in China in 2013 (3). Of lung cancer cases, approximately 95% are non-small-cell lung cancer (NSCLC) and small-cell lung cancer (SCLC) (48), which together represent the most common cause of cancer-related death globally (49,50).

Serving as biomarkers

Exosomes and exosomal miRNAs differed between patients with lung cancer and controls (51). By comparing 12 specific tumor- and exosome-derived miRNAs (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) in lung cancer, previous studies revealed that there was no significant difference between circulating miRNAs and tumor miRNAs, demonstrating that exosome-derived miRNAs can serve as diagnostic biomarkers for lung cancer (51). In a nude mouse model of subcutaneous primary and recurrent lung cancer xenografts in vivo, miR-21 and miR-155 were found to be up-regulated in serum exosomes derived from recurrent tumor-bearing nude mice compared to nontumor- or primary tumor-bearing nude mice (52), suggesting that these two miRNAs might be potential prognostic biomarkers for noninvasive diagnosis of recurrent lung cancer. In addition, Liu et al (53) first reported that elevation of plasma exosomal miR-23b-3p, miR-10b-5p and miR-21-5p predicted a significantly poor survival, implying that these three exosomal miRNAs could serve as independent prognostic biomarkers for NSCLC.

Exosomal membrane-bound proteins, for example, the epidermal growth factor receptor (EGFR), NY-ESO-1 and CD91, are also promising diagnostic or prognostic biomarker candidates for lung cancer. Yamashita et al (54) demonstrated that the measurement of plasma exosomal proteins might be helpful for in vitro diagnosis, and exosomal EGFR was a potential diagnostic biomarker for the characterization of lung cancer. In NSCLC patients, exosomal NY-ESO-1 was a strong prognostic biomarker of poorer survival (55). CD91 expression was significantly increased in serum exosomes derived from patients with lung adenocarcinoma (ADC), and its detection power for early-stage patients was higher than that of carcinoembryonic antigen (CEA) (56).

Stimulating angiogenesis and inducing metastasis

Angiogenesis is essential for tumor growth, progression and metastasis (57). Liu et al (58) found that exosomal miR-21 derived from cigarette smoke extract (CSE)-transformed human bronchial epithelial (HBE) cells was elevated, and this increased exosomal miR-21 led to STAT3 activation and altered the vascular endothelial growth factor (VEGF) expression of recipient cells, promoting CSE-induced angiogenesis and the malignant transformation of HBE cells. These results provided a novel intervention strategy to prevent carcinogenesis of lung cancer. In addition, hypoxic lung cancer cell (hypoxic CL1-5)-derived exosomal miR-23a enhanced neovascularization and tumor growth, and serum exosomal miR-23a was also elevated in patients with lung cancer. These findings provided strong evidence that an increase in exosomal miR-23a contributes to angiogenesis, intravasation and extravasation in lung cancer (59).

Exosomes play a fundamental role in the premetastatic niche and metastasis (4). Results from Fabbri et al (60) indicated that miRNAs (miR-21/29a) derived from lung cancer cell line (A549 and SK-MES) exosomes activate members of the Toll-like receptor (TLR) family (murine TLR7 and human TLR8) in immune cells, leading to a TLR-mediated prometastatic inflammatory response that might ultimately trigger tumor growth and metastasis.

Mediating cisplatin (DDP) resistance

Lung cancer cell-derived exosomes could confer DDP resistance to other cancer cells. Qin et al (61) established A549 cells that were resistant to DDP (A549/DDP). Compared with A549 exosomes, miR-100-5p was downregulated by 75% in A549/DDP cell exosomes. Lower expression of miR-100-5p induced DDP resistance in recipient cells (other lung cancer cell lines). miR-100-5p negatively regulated mTOR, the mammalian target of rapamycin, to alter the recipient lung cancer cells' resistance to DDP. Additionally, the chemosensitivity of NSCLC to DDP could be regulated by serum exosomal miR-146a-5p. The overexpression of miR-146a-5p reversed the resistance of A549/DDP cells by targeting Atg12 to inhibit autophagy (62). Furthermore, in a human bronchial epithelial cell (HBEC) model, exosomes derived from chemoresistant mesenchymal NSCLC cells were able to transfer chemoresistance and mesenchymal phenotypes to recipient cells, thereby enhancing resistance to gemcitabine and cisplatin/gemcitabine combination therapy (63).

Exosomal miRNAs and proteins in liver cancer

Liver cancer is a common malignancy with a high mortality rate both in China and around the world (64,65). Liver cancer includes primary liver cancer (PLC) and secondary liver cancer. Hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC) are two different histologic types of PLC, which is the second most common cause of cancer-related deaths worldwide (66).

Serving as biomarkers

Differential expression of exosomal miRNAs in serum could serve as a diagnostic biomarker for HCC. Sohn et al (67) reported that the levels of serum exosomal miR-18a, miR-221, miR-222 and miR-224 were remarkably higher in HCC patients compared with patients with liver cirrhosis (LC) or chronic hepatitis B (CHB); however, the levels of serum exosomal miR-101, miR-106b, miR-122 and miR-195 were lower in HCC patients than in CHB patients. In addition, other studies have shown that expression of exosomal miR-21 and miR-125b was upregulated in HCC patients compared with CHB patients or healthy controls. More importantly, the levels of miR-21 and miR-125b were higher in exosomes than in serum samples (68,69).

Promoting proliferation, invasion and metastasis

Exosomal miRNAs could affect cellular gene expression and cellular behaviors in target cells (70). Wei et al (71) showed that exosomes derived from HCC cells (SMMC-7721, Hep3B, and Huh-7) could functionally deliver miRNAs to target cells and that Vps4A regulated the secretion and uptake of these miRNAs in hepatoma cells by utilizing exosomes as mediators. Vps4A-associated miRNAs are believed to regulate the PI3K/AKT signaling pathway and promote the proliferation, invasion and metastasis of HCC cells. It has been suggested that a large number of protumorigenic RNAs and proteins, such as the MET proto-oncogene, caveolins (CAV1, CAV2) and an S100 family member (S100A4), are enriched in metastatic HCC-derived exosomes (7274). Moreover, He et al (75) showed that uptake of these shuttling molecules in exosomes derived from motile HCC cell lines (HKCI-C3, HKCI-8 and MHCC97 L) markedly enhanced the invasive and migratory abilities of nonmotile immortalized hepatocyte (MIHA) cell lines by activating the PI3K/AKT and MAPK signaling pathways and increasing the secretion of matrix metalloproteinases (MMP)-2 and MMP-9, which induced cell invasion.

Mediating sensitivity to sorafenib

Sorafenib is predominantly used for the treatment of liver cancer and can improve the overall survival of patients with advanced HCC (76). Exosomes may mediate sorafenib resistance in HCC cells. Guo et al (77) revealed that miR-122 contained in adipose tissue mesenchymal stem cell (AMSC) exosomes enhanced HCC cell sensitivity to chemotherapeutic agents. Compared with the control groups, the inhibitory effect of 5-fluorouracil (5-FU) or sorafenib on HCC cells (HepG2 and Huh7) treated with AMSC-derived exosomes (122-Exo) was significantly enhanced, thereby providing a new strategy for HCC therapy. An important mechanism of sorafenib resistance is the overexpression of c-Met, a proto-oncogene that serves as a receptor for hepatocyte growth factor (HGF) in tumor cells (78). Further investigations confirmed that HGF upregulation and c-Met/AKT pathway activation triggered sorafenib resistance induced by exosomes derived from HCC cells (MHCC-97L and MHCC-97H), indicating that HGF/c-Met might be a possible target for decreasing sorafenib resistance of HCC cells (79).

Exosomal miRNAs and proteins in gastric cancer

Gastric cancer (GC), a malignant tumor of the digestive system, is the second leading cause of cancer-related death and the fourth most common cancer worldwide (80). Although its incidence and mortality have appreciably decreased globally over recent decades, the mortality of GC is still relatively high in Asia (81).

Serving as biomarkers

Recent research suggested that serum exosomal miR-19b-3p and miR-106a-5 could be potential biomarkers for the early diagnosis of GC (82). Additionally, Tokuhisa et al (83) assessed exosomal miRNA profiles in peritoneal fluid and found that miR-21 and miR-1225-5p might be prognostic biomarkers for peritoneal recurrence after curative GC resection. miR-10b-5p, miR-195-5p, miR-20a-3p and miR-296-5p were significantly upregulated in serum exosomes derived from patients with GC and were able to discriminate GC patients from healthy controls (84).

Promoting metastasis

miR-214, miR-221 and miR-222 are commonly upregulated in gastric cancer tissue-derived mesenchymal stem cells (GC-MSCs) and tumor tissues; moreover, GC-MSC-derived exosomes deliver miR-221 to HGC-27 cells and promote the proliferation and migration (85). The serum exosomes of GC patients transport EGFR to liver cells, and EGFR activates HGF by suppressing miR-26a/b, stimulating the development of a liver-like microenvironment that promotes gastric cancer liver metastasis (86). In later studies, proliferation and Matrigel invasion of gastric cancer cells in the presence of exosomes derived from gastric cancer cells (SGC-7901) with either high (SGC/wt) or low (SGC/kd) CD97 expression were investigated, and the results indicated that CD97 promoted gastric cancer cell proliferation and invasion through exosome-mediated activation of the MAPK signaling pathway (87,88).

Regulating the immune response

Compared with exosomes derived from the untreated malignant ascites of GC patients, exosomes derived from heat-treated malignant ascites contained higher concentrations of the heat shock proteins Hsp70 and Hsp60, which might play an important role in inducing a tumor-specific cytotoxic T lymphocyte (CTL) response in vitro and are involved in the promotion of dendritic cell (DC) maturation (89). Additionally, HSPs have been identified as damage-associated molecular patterns (DAMPs), a class of self-danger signals released by stressed cells that elicited immune responses. Mechanistically, HSPs respond to the innate immune system both directly with inflammation and indirectly by recruiting reinforcements (90). However, there is some evidence showing that HSPs have a dampening effect on the immune system under physiological conditions, indicating that HSPs are actually DAMPERs, a class of molecules that reduces the activity of the innate immune system (91).

Mediating DDP resistance

The level of miR-21 in exosomes derived from tumor-associated macrophages (M2 macrophages) has been shown to be increased, and exosomal miR-21 can be directly transferred from tumor-associated macrophages to gastric cancer cells, conferring DDP resistance to gastric cancer cells by downregulating PTEN and activating signaling through the PI3K/AKT pathway (92). However, exosome-delivered anti-miR-214 was able to reverse the resistance of gastric cancer cells to DDP, leading to suppressed migration in vitro, inhibited tumor growth in vivo, and increased cellular apoptosis (93). Additionally, MSC-derived exosomes significantly induced gastric cancer cell resistance to 5-FU both in vivo and ex vivo by activating the calmodulin-dependent protein kinase (CaM-K)/Raf/MEK/ERK pathway (94).

Conclusion and future studies

Exosomes have established a role in cancer biology, immunology, drug sensitivity and clinical diagnosis. In particular, exosomal miRNAs and proteins play important roles in cancers with high mortality rates (lung cancer, liver cancer and gastric cancer) (Tables III and IV).

Table III.

Exosomal miRNAs in the top three mortality cancer types.

Table III.

Exosomal miRNAs in the top three mortality cancer types.

A, Lung cancer

Author, yearmiRNAsStudy designSampleClinical significanceApproachPerformance(Refs.)
Rabinowits et al, 2009 miR-17-p/21/106a/146/155/191/192/203/205/210/212/214Case-controlHuman plasmaDiagnostic biomarkers for NSCLCMicroarrayIncrease(51)
Munagala et al, 2016miR-21/155Animal model Cell modelAthymic nude mice H1299, Beas-2bPossible prognostic markers for lung cancer recurrenceMicroarray, qPCRIncrease(52)
Liu et al, 2017 miR-23b-3p/10b-5p/21-5pCase-controlHuman plasmaIndependent non-invasive prognostic markers for NSCLCqPCRIncrease(53)
Liu et al, 2016miR-21Patients Cell modelHuman serum CSE-transformed HBE cellsPromoting CSE-induced angiogenesis and malignant transformation of HBE cellsqPCRIncrease(58)
Hsu et al, 2017miR-23aPatients Cell modelHuman serum Hypoxic CL1-5Stimulating the angiogenesis, intrava-sation and extravasation in lung cancerqPCRIncrease(59)
Fabbri et al, 2012miR-21/29aCell model, Animal modelA549, SK-MES WT B6 mice B6 TLR7−/−miceTriggering tumour growth and metastasisqPCRIncrease(60)
Qin et al, 2017miR-100-5pCell modelA549/DDPAltering the recipient lung cancer cells' resistance to DDPMicroarray, qPCRDecrease(61)
Yuwen et al, 2017miR-146a-5pPatients Cell modelHuman serum A549/DDPReversing the resistance of A549/DDPqPCRIncrease(62)

B, Liver cancer

Author, yearmiRNAsStudy designSampleClinical significance Approach Performance(Refs.)

Sohn et al, 2015 miR-18a/221/222/224Case-controlHuman serumDiscriminating HCC from LC or CHBqPCRIncrease(67)
Sohn et al, 2015 miR-101/106b/122/195Case-controlHuman serumDiscriminating HCC from CHBqPCRDecrease(67)
Wang et al, 2014; Liu et al, 2017miR-21/125bCase-controlHuman serumDiscriminating HCC from CHB or healthy controlsqPCRIncrease(68,69)
Wei et al, 2015Vps4A-related miRNAsCell modelSMMC-7721, Hep3B, Huh-7Regulating PI3K/AKT signaling pathway and promoting proliferation, invasion and metastasis of HCC cellsRNA sequencingIncrease(71)
Lou et al, 2015miR-122Cell modelAMSCEnhancing the effect 5-FU or sorafenib on HCC cellsqPCRIncrease(77)

C, Gastric cancer

Author, yearmiRNAsStudy designSampleClinical significance Approach Performance(Refs.)

Wang et al, 2017 miR-19b-3p/106a-5Case-controlHuman serumPotential biomarkers for the early diagnosis of GCqPCRIncrease(82)
Tokuhisa, et al, 2015miR-21/1225-5pPatients Cell modelPeritoneum lavage fluid, OCUM-2M OCUM-2MD3Prognostic biomarkers for peritoneal recurrence after curative GC resectionMicroarray, qPCRIncrease(83)
Huang et al, 2017 miR-10b-5p/miR-195-5p/miR-20a-3p/miR-296-5pCase-controlHuman serumDiscriminating GC patients from healthy controlsqPCRIncrease(84)
Wang et al, 2014miR-221Patients Cell model Animal modelHuman tissue GC-MSCs BALB/cnu/nu nude micePromoting HGC-27 cells proliferation and migrationMicroarray, qPCRIncrease(85)
Zheng et al, 2017miR-21Cell model Animal modelM2 macrophages athymic C57-nudemiceConferring DDP resistance in GC cellsMicroarray, qPCRIncrease(92)
Wang et al, 2018Anti-miR-214Cell model Animal modelSGC7901, SGC7901/DDP BALB/c-nude miceReversing the resistance of GC cells to DDPqPCRIncrease(93)

[i] NSCLC, non-small-cell lung cancer; CSE-transformed HBE cells, cigarette smoke extrac-transformed human bronchial epithelial cells. Hypoxic lung cancer cell, hypoxic CL1-5; DDP, cisplatin; HCC, hepatocellular carcinoma; LC, liver cirrhosis; CHB, chronic hepatitis B; AMSC, adipose tissue mesenchymal stem cell; 5-FU, 5-fluorouracil; GC, gastric cancer; GC-MSCs, gastric cancer tissue-derived mesenchymal stem cells; qPCR, quantitative polymerase chain reaction.

Table IV.

Exosomal proteins in the top three mortality cancer types.

Table IV.

Exosomal proteins in the top three mortality cancer types.

A, Lung cancer

Author, yearProteinStudy designSampleClinical significanceApproachPerformance(Refs.)
Yamashita et al, 2017EGFRCase-controlHuman plasmaPotential diagnostic biomarker for characterization of lung cancerELISAIncrease(54)
Sandfeld-Paulsen et al, 2016NY-ESO-1Case-controlHuman plasmaA strongly prognostic markers for poor survival of NSCLCMicroarrayIncrease(55)
Ueda et al, 2014CD91Case-controlHuman serumDiagnostic markers for ADCELISA Mass spectrometryIncrease(56)

B, Liver cancer

Author, yearProteinStudy designSampleClinical significance Approach Performance(Refs.)

He et al, 2015 CAV1/CAV2/S100A4Cell modelHKCI-C3, HKCI-8 MHCC97LEnhancing the invasive and migratory abilities of non-motile MIHA cellsWestern blot Mass spectrometryIncrease(75)
Qu et al, 2016HGFCell model Animal modelMHCC-97L, MHCC-97H BALB/c nu/nu miceImproving sorafenib resistance of HCC cellsELISA Western blotIncrease(79)

C, Gastric cancer

Author, yearProteinStudy designSampleClinical significance Approach Performance(Refs.)

Zhang et al, 2017EGFRPatients Animal model Cell modelHuman serum/tissue BALB/c-nu nude mice SGC7901Promoting GC liver metastasisELISA Western blotIncrease(86)
Li et al, 2015; Liu et al, 2016CD97Cell modelSGC-7901Promoting GC cells proliferation and invasionWestern blotIncrease(87,88)
Zhong et al, 2011Hsp70, Hsp60PatientsHeat-treated malignant ascitesInducing a CTL response in vitro and involving in the promotion of DC maturationWestern blotIncrease(89)

[i] NSCLC, non-small-cell lung cancer; ADC, lung adenocarcinoma; MIHA, motile immortalized hepatocyte; HCC, hepatocellular carcinoma; CTL, cytotoxic T lymphocyte; DC, dendritic cell.

On one hand, existing data indicate that the packaging of miRNAs into exosomes is a selective process and that the levels of specific exosomal miRNAs and proteins are changed in exosomes upon tumorigenesis. For these reasons, exosomal miRNAs and proteins can be served as a class of novel biomarkers for clinical applications in high-mortality cancers. Moreover, the specificity, sensitivity and diagnostic value of exosomal miRNAs and proteins may be superior to that of traditional tumor markers. On the other hand, exosomal miRNAs and proteins are delivered between tumor cells to transmit information and modulate signaling pathways. Taken together, exosomal miRNAs and proteins perform the essential function of promoting tumor progression and metastasis as well as mediating the immune response and sensitivity of tumor cells to chemotherapy drugs (Fig. 3).

In the future, more robust techniques, such as RNA-Seq and mass spectrometry, can be used for the detection, characterization and discovery of exosomal miRNAs and proteins. Moreover, exosomes could efficiently deliver chemotherapeutic agents to cells and tissues. Therefore, these bioengineered, drug-loaded exosomes can serve as promising exosome mimetics for effective chemotherapeutic agent delivery, which will be applied for the target treatment of malignant tumors. Currently, the majority of research on chemotherapy resistance and exosomal microRNAs focuses on cisplatin, and little is known about other drugs. To identify more sensitive and specific exosomal miRNAs and proteins to guide personal chemotherapy selection, future studies should further elucidate the role and underlying mechanism of exosomal miRNAs and proteins in more diverse cancers with more chemotherapy drugs.

Acknowledgements

Not applicable.

Funding

This study was supported by the National Natural Science Foundation of China (grant no. 81772276) and the disciplines group construction project of Pudong Health Bureau of Shanghai (grant no. PWZxq2017-15).

Availability of data and materials

The datasets used or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

LML was a major contributor in writing the manuscript. HL and XHL were responsible for the collection of the relevant literatures. HBH and SML revised the manuscript critically for important intellectual content. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

References

1 

Global Burden of Disease Cancer Collaboration, . Fitzmaurice C, Allen C, Barber RM, Barregard L, Bhutta ZA, Brenner H, Dicker DJ, Chimed-Orchir O, Dandona R, et al: Global, regional, and National cancer incidence, mortality, years of life lost, years lived with disability, and disability-adjusted life-years for 32 cancer groups, 1990 to 2015: A systematic analysis for the global burden of disease study. JAMA Oncol. 3:524–548. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Chen W, Zheng R, Zhang S, Zhao P, Zeng H and Zou X: Report of cancer incidence and mortality in China, 2010. Ann Transl Med. 2:612014.PubMed/NCBI

3 

Zheng R, Zeng H, Zhang S and Chen W: Estimates of cancer incidence and mortality in China, 2013. Chin J Cancer. 36:662017. View Article : Google Scholar : PubMed/NCBI

4 

Zhou L, Lv T, Zhang Q, Zhu Q, Zhan P, Zhu S, Zhang J and Song Y: The biology, function and clinical implications of exosomes in lung cancer. Cancer Lett. 407:84–92. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Qi H, Liu C, Long L, Ren Y, Zhang S, Chang X, Qian X, Jia H, Zhao J, Sun J, et al: Blood exosomes endowed with magnetic and targeting properties for cancer therapy. ACS Nano. 10:3323–3333. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Greening DW, Gopal SK, Xu R, Simpson RJ and Chen W: Exosomes and their roles in immune regulation and cancer. Semin Cell Dev Biol. 40:72–81. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Taylor DD and Gercel-Taylor C: Exosomes/microvesicles: Mediators of cancer-associated immunosuppressive microenvironments. Semin Immunopathol. 33:441–454. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Zhao L, Liu W, Xiao J and Cao B: The role of exosomes and ‘exosomal shuttle microRNA’ in tumorigenesis and drug resistance. Cancer Lett. 356:339–346. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Kalluri R: The biology and function of exosomes in cancer. J Clin Invest. 126:1208–1215. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Frydrychowicz M, Kolecka-Bednarczyk A, Madejczyk M, Yasar S and Dworacki G: Exosomes-structure, biogenesis and biological role in non-small-cell lung cancer. Scand J Immunol. 81:2–10. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Aqil F, Munagala R, Jeyabalan J, Agrawal AK and Gupta R: Exosomes for the enhanced tissue bioavailability and efficacy of curcumin. AAPS J. 19:1691–1702. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Simons M and Raposo G: Exosomes-vesicular carriers for intercellular communication. Curr Opin Cell Biol. 21:575–581. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Schneider A and Simons M: Exosomes: Vesicular carriers for intercellular communication in neurodegenerative disorders. Cell Tissue Res. 352:33–47. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Mathivanan S, Ji H and Simpson RJ: Exosomes: Extracellular organelles important in intercellular communication. J Proteomics. 73:1907–1920. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Villarroya-Beltri C, Baixauli F, Gutiérrez-Vázquez C, Sánchez-Madrid F and Mittelbrunn M: Sorting it out: Regulation of exosome loading. Semin Cancer Biol. 28:3–13. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Hannafon BN and Ding WQ: Intercellular communication by exosome-derived microRNAs in cancer. Int J Mol Sci. 14:14240–14269. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Keerthikumar S, Chisanga D, Ariyaratne D, Al Saffar H, Anand S, Zhao K, Samuel M, Pathan M, Jois M, Chilamkurti N, et al: ExoCarta: A web-based compendium of exosomal cargo. J Mol Biol. 428:688–692. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Ludwig AK and Giebel B: Exosomes: Small vesicles participating in intercellular communication. Int J Biochem Cell Biol. 44:11–15. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Théry C, Zitvogel L and Amigorena S: Exosomes: Composition, biogenesis and function. Nat Rev Immunol. 2:569–579. 2002. View Article : Google Scholar : PubMed/NCBI

20 

Iraci N, Leonardi T, Gessler F, Vega B and Pluchino S: Focus on extracellular vesicles: Physiological role and signalling properties of extracellular membrane vesicles. Int J Mol Sci. 17:1712016. View Article : Google Scholar : PubMed/NCBI

21 

Li W, Li C, Zhou T, Liu X, Liu X, Li X and Chen D: Role of exosomal proteins in cancer diagnosis. Mol Cancer. 16:1452017. View Article : Google Scholar : PubMed/NCBI

22 

Hannafon BN, Carpenter KJ, Berry WL, Janknecht R, Dooley WC and Ding WQ: Exosome-mediated microRNA signaling from breast cancer cells is altered by the anti-angiogenesis agent docosahexaenoic acid (DHA). Mol Cancer. 14:1332015. View Article : Google Scholar : PubMed/NCBI

23 

Kapetanakis NI, Baloche V and Busson P: Tumor exosomal microRNAs thwarting anti-tumor immune responses in nasopharyngeal carcinomas. Ann Transl Med. 5:1642017. View Article : Google Scholar : PubMed/NCBI

24 

Thomou T, Mori MA, Dreyfuss JM, Konishi M, Sakaguchi M, Wolfrum C, Rao TN, Winnay JN, Garcia-Martin R, Grinspoon SK, et al: Adipose-derived circulating miRNAs regulate gene expression in other tissues. Nature. 542:450–455. 2017. View Article : Google Scholar : PubMed/NCBI

25 

Ying W, Riopel M, Bandyopadhyay G, Dong Y, Birmingham A, Seo JB, Ofrecio JM, Wollam J, Hernandez-Carretero A, Fu W, et al: Adipose tissue macrophage-derived exosomal miRNAs can modulate in vivo and in vitro insulin sensitivity. Cell. 171:372–384.e12. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Shi R, Zhao L, Cai W, Wei M, Zhou X, Yang G and Yuan L: Maternal exosomes in diabetes contribute to the cardiac development deficiency. Biochem Biophys Res Commun. 483:602–608. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Milane L, Singh A, Mattheolabakis G, Suresh M and Amiji MM: Exosome mediated communication within the tumor microenvironment. J Control Release. 219:278–294. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Ge Q, Zhou Y, Lu J, Bai Y, Xie X and Lu Z: miRNA in plasma exosome is stable under different storage conditions. Molecules. 19:1568–1575. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Pfeffer SR, Grossmann KF, Cassidy PB, Yang CH, Fan M, Kopelovich L, Leachman SA and Pfeffer LM: Detection of exosomal miRNAs in the plasma of melanoma patients. J Clin Med. 4:2012–2027. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Watahiki A, Macfarlane RJ, Gleave ME, Crea F, Wang Y, Helgason CD and Chi KN: Plasma miRNAs as biomarkers to identify patients with castration-resistant metastatic prostate cancer. Int J Mol Sci. 14:7757–7770. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Xu R, Greening DW, Rai A, Ji H and Simpson RJ: Highly-purified exosomes and shed microvesicles isolated from the human colon cancer cell line LIM1863 by sequential centrifugal ultrafiltration are biochemically and functionally distinct. Methods. 87:11–25. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Subra C, Laulagnier K, Perret B and Record M: Exosome lipidomics unravels lipid sorting at the level of multivesicular bodies. Biochimie. 89:205–212. 2007. View Article : Google Scholar : PubMed/NCBI

33 

Record M, Carayon K, Poirot M and Silvente-Poirot S: Exosomes as new vesicular lipid transporters involved in cell-cell communication and various pathophysiologies. Biochim Biophys Acta. 1841:108–120. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Raposo G and Stoorvogel W: Extracellular vesicles: Exosomes, microvesicles, and friends. J Cell Biol. 200:373–383. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Li P, Kaslan M, Lee SH, Yao J and Gao Z: Progress in exosome isolation techniques. Theranostics. 7:789–804. 2017. View Article : Google Scholar : PubMed/NCBI

36 

Caradec J, Kharmate G, Hosseini-Beheshti E, Adomat H, Gleave M and Guns E: Reproducibility and efficiency of serum-derived exosome extraction methods. Clin Biochem. 47:1286–1292. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Baranyai T, Herczeg K, Onódi Z, Voszka I, Módos K, Marton N, Nagy G, Mäger I, Wood MJ, El Andaloussi S, et al: Isolation of exosomes from blood plasma: Qualitative and quantitative comparison of ultracentrifugation and size exclusion chromatography methods. PLoS One. 10:e01456862015. View Article : Google Scholar : PubMed/NCBI

38 

Peterson MF, Otoc N, Sethi JK, Gupta A and Antes TJ: Integrated systems for exosome investigation. Methods. 87:31–45. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Lu L and Risch HA: Exosomes: Potential for early detection in pancreatic cancer. Future Oncol. 12:1081–1090. 2016. View Article : Google Scholar : PubMed/NCBI

40 

Zeringer E, Barta T, Li M and Vlassov AV: Strategies for isolation of exosomes. Cold Spring Harb Protoc. 2015:319–323. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Taylor DD and Shah S: Methods of isolating extracellular vesicles impact down-stream analyses of their cargoes. Methods. 87:3–10. 2015. View Article : Google Scholar : PubMed/NCBI

42 

Ban JJ, Lee M, Im W and Kim M: Low pH increases the yield of exosome isolation. Biochem Biophys Res Commun. 461:76–79. 2015. View Article : Google Scholar : PubMed/NCBI

43 

Marczak S, Richards K, Ramshani Z, Smith E, Senapati S, Hill R, Go DB and Chang HC: Simultaneous isolation and preconcentration of exosomes by ion concentration polarization. Electrophoresis. Feb 27–2018.(Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

44 

Tauro BJ, Greening DW, Mathias RA, Ji H, Mathivanan S, Scott AM and Simpson RJ: Comparison of ultracentrifugation, density gradient separation, and immunoaffinity capture methods for isolating human colon cancer cell line LIM1863-derived exosomes. Methods. 56:293–304. 2012. View Article : Google Scholar : PubMed/NCBI

45 

Vaswani K, Koh YQ, Almughlliq FB, Peiris HN and Mitchell MD: A method for the isolation and enrichment of purified bovine milk exosomes. Reprod Biol. 17:341–348. 2017. View Article : Google Scholar : PubMed/NCBI

46 

Muller L, Hong CS, Stolz DB, Watkins SC and Whiteside TL: Isolation of biologically-active exosomes from human plasma. J Immunol Methods. 411:55–65. 2014. View Article : Google Scholar : PubMed/NCBI

47 

Witwer KW, Buzás EI, Bemis LT, Bora A, Lässer C, Lötvall J, Nolte-'t Hoen EN, Piper MG, Sivaraman S, Skog J, et al: Standardization of sample collection, isolation and analysis methods in extracellular vesicle research. J Extracell Vesicles. 2:2013. View Article : Google Scholar : PubMed/NCBI

48 

Kadota T, Yoshioka Y, Fujita Y, Kuwano K and Ochiya T: Extracellular vesicles in lung cancer-From bench to bedside. Semin Cell Dev Biol. 67:39–47. 2017. View Article : Google Scholar : PubMed/NCBI

49 

Nanavaty P, Alvarez MS and Alberts WM: Lung cancer screening: Advantages, controversies, and applications. Cancer Control. 21:9–14. 2014. View Article : Google Scholar : PubMed/NCBI

50 

Pletnikoff PP, Laukkanen JA, Tuomainen TP, Kauhanen J, Rauramaa R, Ronkainen K and Kurl S: Cardiorespiratory fitness, C-reactive protein and lung cancer risk: A prospective population-based cohort study. Eur J Cancer. 51:1365–1370. 2015. View Article : Google Scholar : PubMed/NCBI

51 

Rabinowits G, Gercel-Taylor C, Day JM, Taylor DD and Kloecker GH: Exosomal microRNA: A diagnostic marker for lung cancer. Clin Lung Cancer. 10:42–46. 2009. View Article : Google Scholar : PubMed/NCBI

52 

Munagala R, Aqil F and Gupta RC: Exosomal miRNAs as biomarkers of recurrent lung cancer. Tumour Biol. 37:10703–10714. 2016. View Article : Google Scholar : PubMed/NCBI

53 

Liu Q, Yu Z, Yuan S, Xie W, Li C, Hu Z, Xiang Y, Wu N, Wu L, Bai L and Li Y: Circulating exosomal microRNAs as prognostic biomarkers for non-small-cell lung cancer. Oncotarget. 8:13048–13058. 2017.PubMed/NCBI

54 

Yamashita T, Kamada H, Kanasaki S, Maeda Y, Nagano K, Abe Y, Inoue M, Yoshioka Y, Tsutsumi Y, Katayama S, et al: Epidermal growth factor receptor localized to exosome membranes as a possible biomarker for lung cancer diagnosis. Pharmazie. 68:969–973. 2013.PubMed/NCBI

55 

Sandfeld-Paulsen B, Aggerholm-Pedersen N, Bæk R, Jakobsen KR, Meldgaard P, Folkersen BH, Rasmussen TR, Varming K, Jørgensen MM and Sorensen BS: Exosomal proteins as prognostic biomarkers in non-small cell lung cancer. Mol Oncol. 10:1595–1602. 2016. View Article : Google Scholar : PubMed/NCBI

56 

Ueda K, Ishikawa N, Tatsuguchi A, Saichi N, Fujii R and Nakagawa H: Antibody-coupled monolithic silica microtips for highthroughput molecular profiling of circulating exosomes. Sci Rep. 4:62322014. View Article : Google Scholar : PubMed/NCBI

57 

Ostrowski K and Kinsner A: Inhibition of angiogenesis in the treatment of tumors. Arch Immunol Ther Exp (Warsz). 49:27–31. 2001.PubMed/NCBI

58 

Liu Y, Luo F, Wang B, Li H, Xu Y, Liu X, Shi L, Lu X, Xu W, Lu L, et al: STAT3-regulated exosomal miR-21 promotes angiogenesis and is involved in neoplastic processes of transformed human bronchial epithelial cells. Cancer Lett. 370:125–135. 2016. View Article : Google Scholar : PubMed/NCBI

59 

Hsu YL, Hung JY, Chang WA, Lin YS, Pan YC, Tsai PH, Wu CY and Kuo PL: Hypoxic lung cancer-secreted exosomal miR-23a increased angiogenesis and vascular permeability by targeting prolyl hydroxylase and tight junction protein ZO-1. Oncogene. 36:4929–4942. 2017. View Article : Google Scholar : PubMed/NCBI

60 

Fabbri M, Paone A, Calore F, Galli R, Gaudio E, Santhanam R, Lovat F, Fadda P, Mao C, Nuovo GJ, et al: MicroRNAs bind to Toll-like receptors to induce prometastatic inflammatory response. Proc Natl Acad Sci USA. 109:E2110–E2116. 2012. View Article : Google Scholar : PubMed/NCBI

61 

Qin X, Yu S, Zhou L, Shi M, Hu Y, Xu X, Shen B, Liu S, Yan D and Feng J: Cisplatin-resistant lung cancer cell-derived exosomes increase cisplatin resistance of recipient cells in exosomal miR-100-5p-dependent manner. Int J Nanomedicine. 12:3721–3733. 2017. View Article : Google Scholar : PubMed/NCBI

62 

Yuwen DL, Sheng BB, Liu J, Wenyu W and Shu YQ: MiR-146a-5p level in serum exosomes predicts therapeutic effect of cisplatin in non-small cell lung cancer. Eur Rev Med Pharmacol Sci. 21:2650–2658. 2017.PubMed/NCBI

63 

Lobb RJ, van Amerongen R, Wiegmans A, Ham S, Larsen JE and Möller A: Exosomes derived from mesenchymal non-small cell lung cancer cells promote chemoresistance. Int J Cancer. 141:614–620. 2017. View Article : Google Scholar : PubMed/NCBI

64 

Wang FS, Fan JG, Zhang Z, Gao B and Wang HY: The global burden of liver disease: The major impact of China. Hepatology. 60:2099–2108. 2014. View Article : Google Scholar : PubMed/NCBI

65 

Li X and Xu WF: China's efforts to shed its title of ‘Leader in liver disease’. Drug Discov Ther. 1:84–85. 2007.PubMed/NCBI

66 

Wong MC, Jiang JY, Goggins WB, Liang M, Fang Y, Fung FD, Leung C, Wang HH, Wong GL, Wong VW and Chan HL: International incidence and mortality trends of liver cancer: A global profile. Sci Rep. 7:458462017. View Article : Google Scholar : PubMed/NCBI

67 

Sohn W, Kim J, Kang SH, Yang SR, Cho JY, Cho HC, Shim SG and Paik YH: Serum exosomal microRNAs as novel biomarkers for hepatocellular carcinoma. Exp Mol Med. 47:e1842015. View Article : Google Scholar : PubMed/NCBI

68 

Wang H, Hou L, Li A, Duan Y, Gao H and Song X: Expression of serum exosomal microRNA-21 in human hepatocellular carcinoma. Biomed Res Int. 2014:8648942014.PubMed/NCBI

69 

Liu W, Hu J, Zhou K, Chen F, Wang Z, Liao B, Dai Z, Cao Y, Fan J and Zhou J: Serum exosomal miR-125b is a novel prognostic marker for hepatocellular carcinoma. Onco Targets Ther. 10:3843–3851. 2017. View Article : Google Scholar : PubMed/NCBI

70 

Kogure T, Lin WL, Yan IK, Braconi C and Patel T: Intercellular nanovesicle-mediated microRNA transfer: A mechanism of environmental modulation of hepatocellular cancer cell growth. Hepatology. 54:1237–1248. 2011. View Article : Google Scholar : PubMed/NCBI

71 

Wei JX, Lv LH, Wan YL, Cao Y, Li GL, Lin HM, Zhou R, Shang CZ, Cao J, He H, et al: Vps4A functions as a tumor suppressor by regulating the secretion and uptake of exosomal microRNAs in human hepatoma cells. Hepatology. 61:1284–1294. 2015. View Article : Google Scholar : PubMed/NCBI

72 

Mishra SK, Siddique HR and Saleem M: S100A4 calcium-binding protein is key player in tumor progression and metastasis: Preclinical and clinical evidence. Cancer Metastasis Rev. 31:163–172. 2012. View Article : Google Scholar : PubMed/NCBI

73 

Tse EY, Ko FC, Tung EK, Chan LK, Lee TK, Ngan ES, Man K, Wong AS, Ng IO and Yam JW: Caveolin-1 overexpression is associated with hepatocellular carcinoma tumourigenesis and metastasis. J Pathol. 226:645–653. 2012. View Article : Google Scholar : PubMed/NCBI

74 

Cokakli M, Erdal E, Nart D, Yilmaz F, Sagol O, Kilic M, Karademir S and Atabey N: Differential expression of Caveolin-1 in hepatocellular carcinoma: Correlation with differentiation state, motility and invasion. BMC Cancer. 9:652009. View Article : Google Scholar : PubMed/NCBI

75 

He M, Qin H, Poon TC, Sze SC, Ding X, Co NN, Ngai SM, Chan TF and Wong N: Hepatocellular carcinoma-derived exosomes promote motility of immortalized hepatocyte through transfer of oncogenic proteins and RNAs. Carcinogenesis. 36:1008–1018. 2015. View Article : Google Scholar : PubMed/NCBI

76 

Cheng AL, Kang YK, Chen Z, Tsao CJ, Qin S, Kim JS, Luo R, Feng J, Ye S, Yang TS, et al: Efficacy and safety of sorafenib in patients in the Asia-Pacific region with advanced hepatocellular carcinoma: A phase III randomised, double-blind, placebo-controlled trial. Lancet Oncol. 10:25–34. 2009. View Article : Google Scholar : PubMed/NCBI

77 

Lou G, Song X, Yang F, Wu S, Wang J, Chen Z and Liu Y: Exosomes derived from miR-122-modified adipose tissue-derived MSCs increase chemosensitivity of hepatocellular carcinoma. J Hematol Oncol. 8:1222015. View Article : Google Scholar : PubMed/NCBI

78 

You H, Ding W, Dang H, Jiang Y and Rountree CB: c-Met represents a potential therapeutic target for personalized treatment in hepatocellular carcinoma. Hepatology. 54:879–889. 2011. View Article : Google Scholar : PubMed/NCBI

79 

Qu Z, Wu J, Wu J, Luo D, Jiang C and Ding Y: Exosomes derived from HCC cells induce sorafenib resistance in hepatocellular carcinoma both in vivo and in vitro. J Exp Clin Cancer Res. 35:1592016. View Article : Google Scholar : PubMed/NCBI

80 

Ferro A, Peleteiro B, Malvezzi M, Bosetti C, Bertuccio P, Levi F, Negri E, La Vecchia C and Lunet N: Worldwide trends in gastric cancer mortality (1980–2011), with predictions to 2015, and incidence by subtype. Eur J Cancer. 50:1330–1344. 2014. View Article : Google Scholar : PubMed/NCBI

81 

Peleteiro B, Severo M, La Vecchia C and Lunet N: Model-based patterns in stomach cancer mortality worldwide. Eur J Cancer Prev. 23:524–531. 2014. View Article : Google Scholar : PubMed/NCBI

82 

Wang N, Wang L, Yang Y, Gong L, Xiao B and Liu X: A serum exosomal microRNA panel as a potential biomarker test for gastric cancer. Biochem Biophys Res Commun. 493:1322–1328. 2017. View Article : Google Scholar : PubMed/NCBI

83 

Tokuhisa M, Ichikawa Y, Kosaka N, Ochiya T, Yashiro M, Hirakawa K, Kosaka T, Makino H, Akiyama H, Kunisaki C and Endo I: Exosomal miRNAs from peritoneum lavage fluid as potential prognostic biomarkers of peritoneal metastasis in gastric cancer. PLoS One. 10:e01304722015. View Article : Google Scholar : PubMed/NCBI

84 

Huang Z, Zhu D, Wu L, He M, Zhou X, Zhang L, Zhang H, Wang W, Zhu J, Cheng W, et al: Six serum-based miRNAs as potential diagnostic biomarkers for gastric cancer. Cancer Epidemiol Biomarkers Prev. 26:188–196. 2017. View Article : Google Scholar : PubMed/NCBI

85 

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

86 

Zhang H, Deng T, Liu R, Bai M, Zhou L, Wang X, Li S, Wang X, Yang H, Li J, et al: Exosome-delivered EGFR regulates liver microenvironment to promote gastric cancer liver metastasis. Nat Commun. 8:150162017. View Article : Google Scholar : PubMed/NCBI

87 

Li C, Liu DR, Li GG, Wang HH, Li XW, Zhang W, Wu YL and Chen L: CD97 promotes gastric cancer cell proliferation and invasion through exosome-mediated MAPK signaling pathway. World J Gastroenterol. 21:6215–6228. 2015. View Article : Google Scholar : PubMed/NCBI

88 

Liu D, Li C, Trojanowicz B, Li X, Shi D, Zhan C, Wang Z and Chen L: CD97 promotion of gastric carcinoma lymphatic metastasis is exosome dependent. Gastric Cancer. 19:754–766. 2016. View Article : Google Scholar : PubMed/NCBI

89 

Zhong H, Yang Y, Ma S, Xiu F, Cai Z, Zhao H and Du L: Induction of a tumour-specific CTL response by exosomes isolated from heat-treated malignant ascites of gastric cancer patients. Int J Hyperthermia. 27:604–611. 2011. View Article : Google Scholar : PubMed/NCBI

90 

Broere F, van der Zee R and van Eden W: Heat shock proteins are no DAMPs, rather ‘DAMPERs’. Nat Rev Immunol. 11:5652011. View Article : Google Scholar : PubMed/NCBI

91 

van Eden W, Spiering R, Broere F and van der Zee R: A case of mistaken identity: HSPs are no DAMPs but DAMPERs. Cell Stress Chaperones. 17:281–292. 2012. View Article : Google Scholar : PubMed/NCBI

92 

Zheng P, Chen L, Yuan X, Luo Q, Liu Y, Xie G, Ma Y and Shen L: Exosomal transfer of tumor-associated macrophage-derived miR-21 confers cisplatin resistance in gastric cancer cells. J Exp Clin Cancer Res. 36:532017. View Article : Google Scholar : PubMed/NCBI

93 

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

94 

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

Related Articles

Journal Cover

January-2019
Volume 17 Issue 1

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li LM, Liu H, Liu XH, Hu HB and Liu SM: Clinical significance of exosomal miRNAs and proteins in three human cancers with high mortality in China (Review). Oncol Lett 17: 11-22, 2019
APA
Li, L., Liu, H., Liu, X., Hu, H., & Liu, S. (2019). Clinical significance of exosomal miRNAs and proteins in three human cancers with high mortality in China (Review). Oncology Letters, 17, 11-22. https://doi.org/10.3892/ol.2018.9631
MLA
Li, L., Liu, H., Liu, X., Hu, H., Liu, S."Clinical significance of exosomal miRNAs and proteins in three human cancers with high mortality in China (Review)". Oncology Letters 17.1 (2019): 11-22.
Chicago
Li, L., Liu, H., Liu, X., Hu, H., Liu, S."Clinical significance of exosomal miRNAs and proteins in three human cancers with high mortality in China (Review)". Oncology Letters 17, no. 1 (2019): 11-22. https://doi.org/10.3892/ol.2018.9631