Abstract
Exosomes are extracellular vesicles with a diameter of 30–150 nm that function in mediating intercellular communication and intercellular material exchange. The liposomal membrane of exosomes protects the cargo carried by exosomes from degradation and assists in transporting cargo to recipient cells to regulate a variety of physiological and pathological processes. The incidence of gynecologic cancers is increasing annually, which is extremely harmful to the lives and health of women because such cancers are challenging to detect at the early stage. Recently, exosomes have emerged as novel biomarkers for diagnosing and predicting the development of gynecologic cancers. In particular, non-coding RNAs (microRNAs [miRNAs], long non-coding RNAs [lncRNAs], and circular RNAs [circRNAs]) carried by exosomes have been extensively investigated in gynecologic cancers. Therefore, the purpose of this review is to focus on the potential roles of exosomes of different origins in ovarian cancer, cervical cancer, and endometrial cancer, which will help to determine the molecular mechanism of carcinogenesis.
Keywords: exosomes, nucleic acids, proteins, ovarian cancer, cervical cancer, endometrial cancer
Graphical abstract

Exosomes can mediate intercellular communication and material exchange. This review focuses on the potential roles of exosomes of different origins in ovarian cancer, cervical cancer, and endometrial cancer, which will determine the molecular mechanisms of carcinogenesis and discover the potential biomarkers for gynecologic cancers.
Introduction
Gynecologic cancers, such as ovarian, cervical, and endometrial cancers, significantly contribute to the global cancer burden.1 For instance, ovarian cancer is one of the leading causes of death in women. At all stages, the 5-year overall survival rate of ovarian cancer patients is approximately 47%, and more than 70% of ovarian cancer patients are diagnosed at advanced stages, and the 5-year overall survival rate is even lower.2,3 Cervical cancer is the fourth most common female cancer worldwide and remains a major health problem, especially for women in developing countries.4 Endometrial cancer is commonly referred to as a type of uterine cancer that develops from the inner lining of the uterus, mainly occurring in postmenopausal women.5 Advanced and recurrent gynecologic cancers are associated with poor prognosis and lack of effective clinical treatments.5 Hence, early detection of cancers can contribute to improving the survival rate of gynecologic cancer patients, and the use of novel available technologies to identify promising biomarkers for these cancer patients must be considered.
Extracellular vesicles (EVs) are small particles composed of lipid bilayer membranes that are released by diverse cells in both physiological and pathological states.6 EVs are primarily classified into three categories, namely, apoptotic bodies (1,000–5,000 nm in diameter), microvesicles (200–1,000 nm in diameter), and exosomes (30–150 nm in diameter), according to their biogenesis.7,8 Apoptotic bodies are fragments released from apoptotic cells containing fragmented DNA, and cell organelles from their host cells can be found.9 Microvesicles bud directly from the plasma membrane of various cells.10 Exosomes are secreted from multivesicular bodies (MVBs) after fusion with the plasma membrane.11,12 Exosomes have a double-membrane structure with an abundance of cargo contents, such as proteins, messenger RNAs (mRNAs), long non-coding RNAs (lncRNAs), microRNAs (miRNAs), lipids, and viral particles, and exhibit a dish-like or cup-shaped morphology under electron microscopy.13,14 Exosomes can be isolated from body fluids, such as blood plasma or serum, ascites, saliva, urine, ejaculate, and human breast milk, and are released by numerous cell types, including cancer cells, mesenchymal stem cells, and immune cells.15, 16, 17 At present, ultracentrifugation (UC) is a commonly applied technique to isolate exosomes and can be conducted in one of the two ways: differential based or density gradient based.18,19
Exosomes serve as an important tool for intercellular communication and intercellular material exchange and can transfer informative substances to neighboring cells or even distant cells.20 The exosome-based cargo delivery system functions in intercellular communication through endocytosis and receptor-ligand interactions, and subsequently participates in regulating a variety of physiological and pathological processes.21,22 In particular, compared with healthy cells, the cancer cells release more exosomes, the molecular compositions, cargo, and properties of which differ from those of exosomes secreted by healthy cells.23,24 To date, the essential role of exosomes in carcinogenesis and tumor progression has been studied extensively.25,26 Remarkably, cancer-cell-derived exosomes can provide a suitable microenvironment for cancer development via regulation of cell proliferation,27,28 angiogenesis and metastasis,29 immune modulation,30 and drug resistance.31 Moreover, exosomes released by primary ovarian cancer serve as coordinators for the establishment of a premetastatic niche by inducing immunosuppression, facilitating angiogenesis, and remodeling stromal cells within the premetastatic niche of ovarian cancer.32
The nucleic acids and proteins carried by exosomes are packaged in the liposomal membrane, which protects the cargo from degradation, and potentially provides an essential source of clinical information for research.33,34 Non-coding RNAs (ncRNAs) are critical components carried by exosomes, and positively function in diverse biological processes.35 ncRNAs consist of various RNA transcripts, encompassing miRNAs, lncRNAs, and circular RNAs (circRNAs).36 Notably, more than 50% of miRNA genes are localized in tumor-related genomic loci or fragile regions, exerting a regulatory effect on carcinogenesis.37 lncRNAs are characterized by a low expression level, poor conservation between species, and a high coefficient of variation.38 Additionally, lncRNAs are involved in the regulation of tumor angiogenesis, cancer cell stemness, and acquired resistance to chemotherapy by serving as a competitive endogenous RNAs (ceRNAs) and binding directly to related proteins or mRNAs.35,38 CircRNAs are novel ncRNA molecules with a closed-loop structure owing to the covalent linking of 5′ and 3′ termini, and the knowledge of their canonical role remains at a superficial level.39 Acting as an miRNA sponge is the potential mechanism by which circRNAs are currently believed to promote tumor progression.40 Therefore, the roles of exosome-carried nucleic acids and proteins in ovarian cancer, cervical cancer, and endometrial cancer have been summarized in this review (Figure 1). The effects of exosomes on cancer biology will contribute to determining the molecular mechanism of carcinogenesis and discovering the potential biomarkers for gynecologic cancers.32,41
Figure 1.
Exosome-carried nucleic acids and proteins as potential predictive biomarkers in gynecologic cancers
The role of exosomes in ovarian cancer
Ovarian cancer is considered the most aggressive gynecologic malignancy and exhibits high genetic complexity and heterogenicity, with epithelial ovarian cancer (EOC) accounting for 85% of all cases.42,43 Although serum cancer antigen 125 (CA125) detection and ultrasonography are routine methods for the clinical diagnosis of ovarian cancer, they are not suitable for early detection because of low specificity and sensitivity.44,45 In recent years, studies have revealed that exosome-mediated transport of nucleic acids (miRNAs, mRNA, mitochondrial DNA, lncRNAs, and circRNAs) is involved in ovarian carcinogenesis, especially nucleic acids isolated from the plasma or serum of patients.46,47
The role of exosome-carried nucleic acids in ovarian cancer
Role of ovarian cancer-cell-derived exosomes in ovarian cancer
Miharu et al.48 conducted a study on highly (SKOV-3) and weakly (OVCAR-3) invasive ovarian cancer cell lines, and determined that SKOV-3 cells released 2.7-fold more exosomes than OVCAR-3 cells. Moreover, SKOV-3-cell-derived exosomes had a higher expression of let-7 family transcripts than that in the OVCAR-3-cell-derived exosomes, while the expression of miR-200 family transcripts was detected only in OVCAR-3-cell-derived exosomes.48
Exosomal miR-99a-5p from EOC cells improved the invasive ability of EOC cells through upregulation of fibronectin and vitronectin in human peritoneal mesothelial cells.49 Ovarian cancer-cell-secreted exosomal miR-205 accelerated metastasis by promoting angiogenesis by regulating the phosphatase and tensin homolog (PTEN)/Akt pathway.50 Additionally, SKOV3-cell-derived exosomal miR-205 governed proliferation, migration, invasion, and apoptosis via inhibition of vascular endothelial growth factor A (VEGFA) in ovarian cancer cells.51 Similarly, exosomes originating from SKOV-3 cells strengthened the metastatic ability of ovarian cancer compared with the exosomes from OVCAR-3 cells, which was confirmed in an ovarian cancer cell xenograft mouse model.52 Furthermore, exosomal circPUM1 derived from CAOV3 ovarian cancer cells promoted tumor metastasis of peritoneal mesothelial cells via upregulation of nuclear factor (NF)-κB and matrix metallopeptidase 2 (MMP2) expression.53 CircWHSC1 promoted ovarian oncogenesis, and its exosome forms isolated from CAOV3 cells promoted metastasis by acting on peritoneal mesothelium by upregulating mucin 1 (MUC1).54 Taken together, the nucleic acids carried by exosomes derived from ovarian cancer cells potentially influence the invasive and metastatic abilities of ovarian cancer.
Exosomes derived from ovarian cancer cells promoted angiogenesis in serous ovarian cancers, especially in the high-grade ovarian cancers.55 Additionally, exosomes originating from SKOV3-DDP (cisplatin) cells were abundant in miR-130a, which facilitated angiogenesis.56 Higher miR-940 expression in exosomes isolated from EOC cells was induced by hypoxia, and exosomal miR-940 was also abundant in the ascites of EOC patients.57 Moreover, tumor-derived exosomal miR-940 exhibited a vital role in promoting the polarization of tumor-associated macrophages in EOC progression.57 Exosomal miR-1246 was highly expressed in ovarian cancer-cell-derived exosomes, which exerted oncogenic properties in the tumor microenvironment by transferring the miR-1246 to the M2-type macrophages and subsequently had the potential to downregulate the expression of caveolin 1 (Cav1).58 Exosomes derived from human ovarian surface epithelial cells transferred miR-124 to cancer-associated fibroblasts (CAFs), which suppressed the transition from normal fibroblasts to cancer-associated fibroblasts by repressing sphingosine kinase 1 (SPHK1) expression in ovarian cancer.59 The above studies suggested that nucleic acids carried by exosomes derived from ovarian cancer cells function in the regulation of the tumor microenvironment.
Role of non-tumor-cell-derived exosomes in ovarian cancer
Exosomes purified from primary-cultured omental fibroblasts of ovarian cancer patients were loaded with miR-199a-3p (miR-199a-3p-Exo), and miR-199a-3p-Exo downregulated the expression of c-Met, which subsequently suppressed the proliferation and invasion of ovarian cancer cells.60 In addition, miR-199a-3p-Exo decreased the peritoneal dissemination in an ovarian cancer mouse model, and reduced the expression of c-Met, extracellular signal-regulated kinase (ERK) phosphorylation, and MMP2 in cancers.60 Exosomal miR-7 purified from the tumor necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK)-stimulated macrophages (TMs) suppressed the metastatic ability of EOC cells and inhibited the epidermal growth factor receptor (EGFR)/AKT/ERK1/2 signaling pathway.61 Nevertheless, exosomes isolated from some cell types promoted the progression of ovarian cancer. Plasma cells conferred the mesenchymal identity of ovarian cancers by enhancing the transfer of exosome-derived miR-330-3p.62 Exosomal miR-98-5p from CAFs facilitated the proliferation and cell cycle entry and inhibited the apoptotic ability of ovarian cancer cells.63 In summary, the different characteristics of exosomes in ovarian cancer development are predominantly determined by the cell types from which they originate.
Role of exosomes derived from body fluids in ovarian cancer
Xu et al.64 found that the expression of miR-101 and miR-30a was downregulated, while the expression of miR-21 and miR-210 was upregulated, in ovarian cancer tissues versus the noncancerous tissues. However, only miR-101 expression was significantly downregulated in the serum exosomes from ovarian cancer patients, suggesting that miR-101 potentially serves as a biomarker for ovarian cancer.64 The expression of miR-146b-5p in exosomes from the serum of EOC patients was higher than that in exosomes from healthy individuals.65 Zhang et al.66 documented that plasma exosomal miR-106a-5p was upregulated, while plasma exosomal miR-122-5p and miR-185-5p were downregulated, in ovarian cancer patients compared with healthy individuals. One study using microarrays found that six circRNAs were increased and 37 circRNAs were decreased in serum exosomes from ovarian cancer patients.67 Among the six circRNAs, higher circ-0001068 expression in the serum exosomes was further confirmed in a large cohort.67 Circ-0001068 induced programmed death 1 (PD1) expression by targeting miR-28-5p after it was delivered into T cells by exosomes.67 Different expression levels of nucleic acids in serum or plasma exosomes were found not only between ovarian cancer patients and healthy individuals but also among ovarian cancer patients with different grades. A study revealed that serum exosomal miR-34a expression was elevated in early-stage ovarian cancer cases.68 Consistently, exosomal miR-34a in serum was downregulated in ovarian cancer patients with lymph node metastasis and recurrence.68 MiR-214-3p, which is an epigenetic regulator with an oncogenic role in EOC,69 was highly expressed in serum exosomes from patients with highly malignant EOC and platinum-resistant high-grade serous ovarian cancer (HGSOC).70 This study also demonstrated that the miR-214-3p and its target gene, LIM homeobox domain 6 (LHX6), in serum exosomes potentially acted as biomarkers for predicting EOC progression.70 Moreover, Keseru et al.71 detected the exosome-encapsulated mitochondrial DNA (mtDNA) copy number in the cell-free plasma of serous epithelial ovarian cancer patients, and indicated that the exosomal mtDNA copy number was increased in late-stage ( International Federation of Gynecology and Obstetrics [FIGO] stages III and IV) patients compared with the healthy individuals.
Elevated expression of miR-1307 and miR-375 was found in serum exosomes from ovarian cancer patients in contrast to that in patients with benign ovarian tumors and healthy individuals, which may improve the diagnostic efficiency of CA125 for ovarian cancer.72 One study detected miR-93, miR-145, and miR-200c upregulation in exosomes derived from the serum of ovarian cancer patients, and the triple combination of serum exosomal miR-145 and miR-200c and serum CA125 was the most effective biomarker, with a sensitivity of 100% for the differential diagnosis of ovarian masses (benign ovarian cysts/borderline ovarian neoplasms versus ovarian cancers), indicating the strong diagnostic potential of miRNAs carried by exosomes.73 Lower expression of serum exosomal miR-484 was found in ovarian cancer patients than in healthy individuals, and detection of serum exosomal miR-484 with CA125 exhibited a good diagnostic performance in discriminating ovarian cancer patients and healthy individuals, with an area under the receiver operating characteristic (ROC) curve (AUC) of 0.912.74 Taken together, the combination of serum CA125 and nucleic acids in blood exosomes of ovarian cancer patients potentially enhances the diagnostic efficiency in ovarian cancer.
Exosomal miR-21, miR-100, miR-200b, and miR-320 were highly expressed, while exosomal miR-16, miR-93, miR-126, and miR-223 were weakly expressed, in the plasma of EOC patients compared with healthy individuals.75 In particular, the elevated expression of exosomal miR-200b was associated with an increasing value of CA125 and a poor overall survival for EOC patients.75 Additionally, ovarian cancer patients simultaneously with a low serum exosomal miR-484 and a high serum CA125 expressions showed a trend toward worse clinical outcomes.74 The upregulation of serum exosomal lncRNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) in EOC patients was associated with an advanced and metastatic phenotype of EOC, which potentially predicted poor overall survival for EOC patients.76 Therefore, the nucleic acids carried by blood exosomes of ovarian cancer patients may act as biomarkers for predicting their clinical outcomes.
Apart from plasma or serum, exosomes in other body fluids have attracted the attention of many researchers. miRNA microarray data revealed that miR-30a-5p expression was upregulated in the urine of serous ovarian adenocarcinoma patients compared with healthy controls and demonstrated that the urinary miR-30a-5p originated from the exosomes excreted by ovarian cancer cells.77 Cappellesso et al.78 confirmed that the mRNA expression of programmed cell death 4 (PDCD4) was upregulated, while miR-21 expression was downregulated, in the cells and exosomes from peritoneal effusions of serous ovarian carcinoma in contrast to nonneoplastic peritoneal effusions. This study also indicated that miR-21 transfer by exosomes could facilitate carcinogenic transformation in target cells far from the primary tumor without direct colonization by cancer cells and can be regarded as a diagnostic tool for ovarian serous carcinoma.78 Exosomes from ascites of ovarian cancer patients transferred miR-6780b-5p to ovarian cancer cells, which facilitated epithelial-mesenchymal transition (EMT) and subsequently promoted ovarian cancer metastasis.79
The role of exosome-carried proteins in ovarian cancer
Exosomes can package and release the unique proteins associated with a disease; therefore, the identification of characteristic proteins in the exosomes originating from ovarian cancer patients holds promise for the diagnosis of ovarian cancer.80 High cluster of differentiation (CD) 24 expression in plasma exosome was reported in serous ovarian cancer patients.81 CD24 and epithelial cell adhesion molecule (EpCAM) proteins were identified in exosomes purified from ascites fluid of ovarian cancer patients.82 Microfluidic technology can realize the rapid separation of blood exosomes83, 84, 85; in particular, microfluidic technology can validate significant biomarkers, such as folate receptor alpha (FRα) in plasma exosomes from ovarian cancer patients86 and hepatocyte growth factor (HGF), STAT3, and interleukin 6 (IL-6) in serum exosome samples from early-stage HGSOC patients.87 Zhang et al.88 identified 294 proteins in plasma exosomes from EOC patients and healthy individuals, and demonstrated that the lipopolysaccharide-binding protein (LBP), gelsolin, fibrinogen gamma chain, and fibrinogen alpha chain proteins in plasma exosomes from EOC patients could be used as biomarkers for the diagnosis of ovarian cancer. Furthermore, the plasma of ovarian cancer patients carried higher levels of exosomal proteins, including melanoma-associated antigen 3/6 (MAGE 3/6) and transforming growth factor β 1 (TGF-β1), than that of ovarian benign tumor patients and healthy individuals, which provided a novel strategy to diagnose ovarian cancer patients.89
Studies have demonstrated that exosome-mediated transfer of key proteins plays an essential role in ovarian carcinogenesis.90,91 For instance, the migration and invasion of low-metastatic ovarian cancer cells were enhanced due to exosome-involved transfer of CD44 from high-metastatic ovarian cancer cells.90 Exosomes derived from ovarian cancer cells transported CD44 to human peritoneal mesothelial cells, thus increasing the invasive ability of ovarian cancer.92 Serum exosomal antisense hypoxia inducible factor (aHIF) was highly expressed in EOC patients and was associated with poor overall survival, indicating that exosomal aHIF might be a prognostic predictor for EOC.93
Recently, researchers applied proteomics techniques to identify the promising biomarkers in ovarian cancer. Bebelman et al.94 observed high levels of the proteins glucose-6-phosphate dehydrogenase, transketolase, and transaldolase, which are key regulatory enzymes in the pentose phosphate pathway, in exosomes originating from two late-stage ovarian cancer cell lines, OVCA429 and HO8910PM. Liang et al.95 conducted a nano liquid chromatography-tandem mass spectrometry (LC-MS/MS) workflow on the exosomes from OVCAR-3 and IGROV1 ovarian cancer cell lines, and identified 2,230 exosomal proteins in these two ovarian cancer cell lines. Moreover, another study used an ExoProfile chip to analyze the circulating exosomes from the plasma of ovarian cancer patients, and revealed that the combinations of multiple markers, including EGFR, human EGFR 2 (HER2), CA125, folate receptor α (FRα), CD24, EpCAM, CD9, and CD63, in circulating exosomes potentially improved the efficacy for differentiating early- and late-stage ovarian cancer, yielding the best diagnostic AUC value.96 In addition, another study revealed that 1,433 proteins and 1,227 lipid species were identified in exosomes from the SKOV3 ovarian cancer cell line and HOSEPiC ovarian surface epithelial cell line, and demonstrated that the lipids cholesterol ester (ChE) and zymosterol (ZyE) species and collagen type V alpha 2 chain (COL5A2) and lipoprotein lipase (LPL) proteins were more abundant in exosomes derived from SKOV3 ovarian cancer cells than those derived from HOSEPiC ovarian surface epithelial cells.97
The role of exosomes in regulating chemotherapy of ovarian cancer
The exosomal tumor suppressor miR-6126 was detected at high levels in both chemosensitive and chemoresistant ovarian cancer cells.98 Higher expression of lncRNA urothelial carcinoma-associated 1 (UCA1) was detected in the serum exosomes of cisplatin-resistant ovarian cancer patients than their cisplatin-sensitive counterparts.99 One study revealed a lower expression of the circRNA Cdr1as in the serum exosomes of cisplatin-resistant ovarian cancer patients than in cisplatin-sensitive patients.100 The above studies indicated that the expression levels of nucleic acids in exosomes differed between chemosensitive and chemoresistant ovarian cancers.
Serum exosomal circular forkhead box protein P1 (circFoxp1) was upregulated in EOC patients, especially in DDP-resistant EOC patients, which conferred cisplatin resistance in EOC cells.101 Exosomal DNA methyltransferase 1 (DNMT1) transcripts were highly expressed in exosomes derived from ovarian cancer cells and enhanced the cisplatin resistance of ovarian cancer cells.102 In addition, miR-21-3p, miR-21-5p, and miR-891-5p were abundant in exosomes derived from ovarian cancer cells, contributing to carboplatin resistance of ovarian cancer cells.103 In particular, ovarian cancer patients at risk of relapse showed higher miR-891-5p expression of exosomes.103 Macrophage-derived exosomes transferred miR-223 to EOC cells to trigger chemoresistance via regulation of the PTEN/PI3K/Akt pathway, and the circulating exosomal miR-223 expression was linked to EOC recurrence.104 Exosomal miR-98-5p from CAFs enhanced cisplatin (DDP) resistance via downregulation of cyclin-dependent kinase inhibitor 1A (CDKN1A) in subcutaneous-ovarian-cancer-bearing nude mice.63 Exosomal plasma gelsolin enhanced the survival of ovarian cancer cells via both autocrine and paracrine mechanisms to enhance chemoresistance.91 The enhanced expression of exosome-carried plasma gelsolin from ovarian cancer cells weakened the immunosurveillance and promoted the synthesis of glutathione, which subsequently caused chemoresistance in ovarian cancer.105 Moreover, miR-21 transferred by exosomes isolated from neighboring stromal cells conferred paclitaxel resistance to ovarian cancer cells via regulation of apoptotic peptidase activating factor 1 (APAF1).106
Exosomal miR-146a derived from human umbilical cord mesenchymal stem cells (hUCMSCs) inhibited the growth and ameliorated the chemoresistance of ovarian cancer cells.107 Moreover, silencing of miR-146a in hUCMSC-derived exosomes promoted the growth and chemoresistance of ovarian cancer cells through the phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway via laminin γ2.107 Exosomes released by miR-30a-5p mimic-transfected SKOV3/DDP cells inhibited the proliferative ability and enhanced the apoptotic ability of SKOV3 cells, and improved the chemosensitivity of SKOV3 cells to DDP by downregulating the expression of SRY-box 9 (SOX9).108 Taken together, the different expression levels of nucleic acids and proteins in exosomes potentially contributed to the chemoresistance or chemosensitivity of ovarian cancer, and these biomarkers may become targets for clinical therapy.
Cisplatin-loaded exosomes from umbilical cord blood-originated M1 macrophages exerted a superior inhibitory effect on the growth of the A2780 EOC cell line and cisplatin-resistant A2780/DDP cell line compared with free cisplatin, and ameliorated the cisplatin resistance of ovarian cancer cells.109 Exosomes derived from SKOV3 cells were loaded with triptolide, and the triptolide-loaded exosomes exerted a stronger inhibitory effect on proliferation and growth and exhibited weaker cytotoxic and apoptotic effects on SKOV3 cells than did free triptolide.110 The exosome-based drug delivery system showed better therapeutic effects than a single drug in ovarian cancer.
The role of exosomes in cervical cancer
Cervical cancer can be broadly classified into two categories: cervical squamous cell carcinoma (accounting for 80% of cases) and cervical adenocarcinoma (accounting for 9% of cases).111 Persistent high-risk human papillomavirus (HPV) infection is considered a leading cause of cervical cancer, and the HPV oncogenes E6 and E7 play an essential role in this process.112,113 However, a few patients with cervical cancer do not show HPV infection; thus, preventing high-grade cervical intraepithelial neoplasia (CIN) from developing into invasive cervical cancer through early screening is crucial.114
Role of cervical-cancer-cell-derived exosomes in cervical cancer
One study reported that miR-221-3p in exosomes originating from cervical cancer cells promoted the proliferation, invasion, migration, and angiogenesis of microvascular endothelial cells in cervical cancer by reducing the expression of mitogen-activated protein kinase 10 (MAPK10).115 Cervical squamous cell carcinoma-cell-derived exosomes delivered miR-221-3p from cancer cells to the human umbilical vein endothelial cells (HUVECs), and the exosomal miR-221-3p promoted angiogenesis via downregulation of Thrombospondin-2 in cervical squamous cell carcinoma.116 Cervical cancer cells exposed to TGF-β1 tended to secrete more miR-663b-containing exosomes, and exosomal miR-663b could be endocytosed by the adjacent or distant cervical cancer cells.117 Exosomal miR-663b reduced the expression of mannoside acetyl-glucosaminyltransferase 3 (MGAT3), which subsequently facilitated the EMT and enhanced the local and distant metastasis of cervical cancers.117 One study illustrated that knockdown of HPV E6/E7 expression in cervical cancer cells (HPV18-positive HeLa cells) contributed to elevated release of HeLa--cell-derived exosomes.118 Exosomal let-7d-5p, miR-20a-5p, miR-378a-3p, miR-423-3p, miR-7-5p, and miR-92a-3p were downregulated, while exosomal miR-21-5p was upregulated in exosomes isolated from HeLa cells with the silencing of E6/E7 silencing.119 Cervical cancer-cell-secreted exosomes carrying lncRNA HNF1A-AS1 facilitated the proliferation and DDP resistance and weakened the apoptosis of cervical cancer cells via the upregulation of Tuftelin1 and downregulation of miR-34b.120 Cervical cancer cells transfer the hedgehog pathway (Hh)-related proteins (Patched1, Smoothened, Sonic hedgehog, Indian hedgehog) in their exosomes.121 Exosomal Wnt2B protein from cervical cancer cells promoted the activation of fibroblasts, thereby facilitating the progression of cervical cancer.122 Taken together, nucleic acids and proteins carried by exosomes released by cervical cancer cells potentially exert a pro-oncogenic effect on cervical cancer.
Role of non-tumor-cell-derived exosomes in cervical cancer
MiR-155-5p in exosomes isolated from HIV-infected T cells facilitated the proliferative, migrative, and invasive abilities of cervical cancer cells, which may be mediated through the AT-rich interaction domain 2 (ARID2)-ERCC5-NF-κB pathway.123 Furthermore, miR-22-enriched exosomes originating from the pre-miR-22-transfected HEK293 cells improved the sensitivity of cervical cancers to radiotherapy.124 More research must be carried out to study the effects of non-tumor-cell-derived exosomes in cervical cancer.
Role of exosomes derived from body fluids in cervical cancer
One study indicated that the expression of miR-146a-5p, miR-151a-3p, and miR-2110 was elevated in plasma exosomes derived from cervical cancer patients in contrast to those from healthy individuals.125 Plasma exosomal miR-125a-5p showed higher expression in cervical cancer patients than in the healthy individuals and potentially acted as a marker for the differential diagnosis between cervical cancer patients and healthy individuals.126 Moreover, Zheng et al.127 demonstrated that eight differentially expressed plasma exosomal miRNAs (let-7a-3p, let-7d-3p, miR-30d-5p, miR-144-5p, miR-182-5p, miR-183-5p, miR-215-5p, and miR-4443) could discriminate CIN II+ patients (including advanced CIN II patients) from CIN I patients (including CIN I patients and healthy individuals). However, only let-7d-3p and miR-30d-5p showed significant differences between cervical cancer tissues and adjacent normal tissues,127 indicating that exosomal miRNAs might be selectively secreted by tumor cells. Higher expression of serum exosomal lncRNA DLX6-AS1 was detected in cervical cancer patients than in the CIN patients and healthy individuals, which was positively correlated with lymph node metastasis, differentiation, and FIGO stage, and predicted relapse and a worse clinical outcome for cervical cancer patients.128 Altogether, the exosomes isolated from the plasma or serum of cervical cancer patients are useful in cervical cancer screening and diagnosis.
One study documented that the expression of PI3k/Akt/mammalian target of rapamycin (mTOR) in cervical cancer tissues and in the exosomes derived from vaginal secretions was higher than that in the adjacent normal cervical tissues, but the expression levels did not significantly differ between cervical cancer tissues and the exosomes derived from vaginal secretions.129 Additionally, exosomal miR-146a and miR-21 separated from cervicovaginal lavage fluid were increased in cervical cancer patients compared with individuals with no (pre)cervical disease (with or without HPV infection).130 Compared with that in cancer-free volunteers (with or without HPV infection), the expression of exosomal lncRNAs hox transcript antisense intergenic RNA (HOTAIR) and metastasis-associated lung carcinoma transcript 1 (MALAT1) was upregulated, whereas the expression of exosomal lncRNA maternally expressed gene 3 (MEG3) was downregulated in the cervicovaginal lavage fluid of cervical cancer patients.131 Moreover, among the cancer-free volunteers, elevated expression of exosomal lncRNAs HOTAIR and MALAT1 and decreased expression of exosomal lncRNA MEG3 were detected in the cervicovaginal lavage fluid from HPV-positive individuals compared with those in HPV-negative individuals, indicating that lncRNAs HOTAIR, MALAT1, and MEG3 may have an important role in the development of cervical cancer.131 Additionally, 45 miRNAs were upregulated and 55 miRNAs were downregulated in exosomes purified from the cervical-vaginal fluid of HPV16-infected patients versus those in the HPV16-free individuals.132 The detection and analysis of cervicovaginal lavage samples and vaginal secretions of cervical cancer patients, which are rich in exosomes, provide a new idea for noninvasive cervical cancer screening.130
The role of exosomes in endometrial cancer
Endometrial cancer predominantly occurs in postmenopausal women and can be grouped into two clusters: estrogen dependent (type I) and estrogen independent (type II).133,134 miRNAs are aberrantly expressed in endometrial cancer, and dysregulated miRNAs potentially function as pro-oncogenesis factors or tumor suppressors in endometrial cancer progression.135,136
Role of exosomes derived from cells in endometrial cancer
Endometrial cancer cells can deliver small regulatory RNAs to endometrial fibroblasts via exosomes.137 Exosomal miR-133a was found in the exosomes derived from endometrial cancer cells and could be transferred to the normal endometrial cells.138 Exosomal lncRNA deleted in lymphocytic leukemia1 (DLEU1) derived from endometrial cancer cells improved the migrative and invasive abilities of endometrial cancer cells via the regulation of the miR-381-3p/E2F transcription factor 3 (E2F3) axis.139 Exosomes originating from the CAFs facilitated the invasive ability of endometrial cancer cells compared with the exosomes obtained from the normal fibroblasts (NFs), which was partially attributed to the low expression levels of miR-148b in exosomes of CAFs.140 Nevertheless, the proliferation of endometrial cancer cells and tube formation of endothelial cells were inhibited in vitro by the exosomal miR-499 derived from the mesenchymal stem cells, and the exosomal miR-499 also suppressed the tumor growth and angiogenesis in vivo.141 In summary, exosomes secreted by different cell types have diverse effects on endometrial cancer progression.
Role of exosomes derived from body fluids in endometrial cancer
A total of 114 miRNAs were dysregulated in exosomes purified from the peritoneal lavage fluid of endometrial cancer patients in contrast to miRNAs in the exosomes isolated from the ascitic fluid of control patients, and the downregulated expression of peritoneal lavage exosomal miRNA-383-5p, miRNA-10b-5p, miRNA-34c-3p, miRNA-449b-5p, miRNA-34c-5p, miRNA-200b-3p, miRNA-2110, and miRNA-34b-3p potentially served as individual diagnostic biomarkers for endometrial cancer.142 Compared with the exosomes isolated from the urine of patients with symptoms of endometrial cancer, but no diagnosis of endometrial cancer, elevated miRNA-200c expression was discovered in exosomes isolated from the urine of endometrial cancer patients.143 Increased expression of 209 circRNAs and decreased expression of 66 circRNAs were discovered in exosomes isolated from the serum of stage III endometrial adenocarcinoma patients versus exosomes isolated from the serum of healthy individuals.144 Thus, nucleic acids carried by exosomes isolated from the peritoneal lavage fluid, urine, and serum of endometrial cancer patients have potential as novel diagnostic biomarkers of endometrial cancer.
Coculture of endometrial cancer cells and the exosomes isolated from the serum of polycystic ovary syndrome (PCOS) patients increased the migrative and invasive abilities of endometrial cancer cells, which may be regulated by the serum exosomal miR-27a-5p.145 Exosomal lectin galactoside-binding soluble 3-binding protein (LGALS3BP) was detected in exosomes isolated from the plasma of endometrial cancer patients and was upregulated with endometrial cancer progression, promoted the growth of endometrial cancer cells and facilitated the angiogenesis of HUVECs.146 The nucleic acids and proteins carried by exosomes may be involved in the progression of endometrial cancer.
Conclusions and perspectives
The small molecules within exosomes (including nucleic acids and proteins) allowed the identification of potential molecular biomarkers for the diagnosis, disease progression monitoring, and chemotherapy efficiency of ovarian cancer (Table 1; Figure 2), cervical cancer (Table 2; Figure 3), and endometrial cancer (Table 3; Figure 4).
Table 1.
Exosome-carried nucleic acids and proteins as potential biomarkers for ovarian cancer
| Type of biomarkers | Potential biomarkers | Exosome derivation | Reference |
|---|---|---|---|
| Nucleic acids | let-7 family, miR-200 family | cells | Miharu Kobayashi et al.48 |
| miR-99a-5p | cells | Yoshimura et al.49 | |
| miR-205 | cells | He et al.50; Wang et al.51 | |
| circPUM1 | cells | Guan et al.53 | |
| circWHSC1 | cells | Zong et al.54 | |
| miR-940 | cells | Chen et al.57 | |
| miR-1246 | cells | Kanlikilicer et al.58 | |
| miR-124 | cells | Zhang et al.59 | |
| miR-199a-3p | cells | Kobayashi et al.60 | |
| miR-7 | cells | Hu et al.61 | |
| miR-330-3p | cells | Yang et al.62 | |
| miR-98-5p | cells | Guo et al.63 | |
| miR-101 | serum | Xu et al.64 | |
| miR-146b-5p | serum | Wu et al.65 | |
| miR-106a-5p, miR-122-5p, miR-185-5p | plasma | Zhang et al.66 | |
| circ-0001068 | serum | Wang et al.67 | |
| miR-34a | serum | Maeda et al.68 | |
| miR-214-3p | serum | Yang et al.70 | |
| MtDNA | plasma | Keseru et al.71 | |
| miR-1307, miR-375 | serum | Su et al.72 | |
| miR-145, miR-200c | serum | Kim et al.73 | |
| miR-484 | serum | Zhang et al.74 | |
| miR-200b | plasma | Panet al.75 | |
| lncRNA MALAT1 | serum | Qiu et al.76 | |
| miR-30a-5p | urine | Zhouet al.77 | |
| miR-21 | ascites | Cappellesso et al.78 | |
| miR-6780b-5p | ascites | Cai et al.79 | |
| miR-6126 | cells | Kanlikilicer et al.98 | |
| lncRNA UCA1 | serum | Li et al.99 | |
| circRNA Cdr1as | serum | Zhao et al.100 | |
| circFoxp1 | serum | Luo et al.101 | |
| miR-891-5p | cells | Alharbi et al.103 | |
| miR-223 | cells | Zhu et al.104 | |
| miR-21 | cells | Au Yeung et al.106 | |
| miR-146a | cells | Liya et al.107 | |
| miR-30a-5p | cells | Liu et al.108 | |
| Proteins | FRα | plasma | Zhang et al.86 |
| HGF, STAT3, IL-6 | serum | Dorayappan et al.87 | |
| LBP, fibrinogen gamma chain, fibrinogen alpha chain | plasma | Zhang et al.88 | |
| gelsolin | plasma | Zhang et al.88; Asare-Wereheneet al.91; Meshach Asare-Werehene et al.105 | |
| MAGE 3/6, TGF-β1 | plasma | Szajnik et al.89 | |
| CD44 | cells | Shen et al.90; Nakamura et al.92 | |
| aHIF | serum | Tang et al.93 | |
| glucose-6-phosphate dehydrogenase, transketolase, transaldolase | cells | Yi et al.94 | |
| EGFR, HER2, CA125, FRα, CD24, EpCAM, CD9, CD63 | plasma | Zhang et al.96 | |
| COL5A2, LPL | cells | Cheng et al.97 |
Figure 2.
The role of exosomes in ovarian cancer
Table 2.
Exosome-carried nucleic acids and proteins as potential biomarkers for cervical cancer
| Type of biomarkers | Potential biomarkers | Exosome derivation | Reference |
|---|---|---|---|
| Nucleic acids | miR-221-3p | cells | Zhang et al.115; Wu et al.116 |
| miR-663b | cells | You et al.117 | |
| let-7d-5p, miR-20a-5p, miR-378a-3p, miR-423-3p, miR-7-5p, miR-92a-3p, miR-21-5p | cells | Honegger et al.119 | |
| lncRNA HNF1A-AS1 | cells | Luo et al.120 | |
| miR-155-5p | cells | Li et al.123 | |
| miR-22 | cells | Konishi et al.124 | |
| miR-146a-5p, miR-151a-3p, miR-2110 | plasma | Ma et al.125 | |
| miR-125a-5p | plasma | Lv et al.126 | |
| let-7d-3p, miR-30d-5p | plasma | Zheng et al.127 | |
| lncRNA DLX6-AS1 | serum | Ding et al.128 | |
| miR-146a, miR-21 | cervicovaginal lavages | Liu et al.130 | |
| lncRNAs HOTAIR, MALAT1, MEG3 | cervicovaginal lavages | Zhang et al.131 | |
| Proteins | Patched1, Smoothened, Sonic hedgehog, Indian hedgehog | cells | Bhat et al.121 |
| Wnt2B | cells | Liang et al.122 | |
| PI3k/Akt/mTOR | vaginal secretions | Zhang et al.129 |
Figure 3.
The role of exosomes in cervical cancer
Table 3.
Exosome-carried nucleic acids as potential biomarkers for endometrial cancer
| Type of biomarkers | Potential biomarkers | Exosome derivation | Reference |
|---|---|---|---|
| Nucleic acids | miR-133a | cells | Shi et al.138 |
| lncRNA DLEU1 | cells | Jia et al.139 | |
| miR-148b | cells | Li et al.140 | |
| miR-499 | cells | Jing et al.141 | |
| miRNA-383-5p, miRNA-10b-5p, miRNA-34c-3p, miRNA-449b-5p, miRNA-34c-5p, miRNA-200b-3p, miRNA-2110, miRNA-34b-3p | peritoneal lavage | Roman-Canal et al.142 | |
| miRNA-200c | urine | Srivastava et al.143 | |
| miR-27a-5p | serum | Xiaoxia et al.145 | |
| Protein | LGALS3BP | plasma | Song et al.146 |
Figure 4.
The role of exosomes in endometrial cancer
In contrast to other nanoparticles, exosomes present considerable advantages, including low immunogenicity, low toxicity, biological barrier permeability, high stability, excellent biocompatibility, and targetability, which render exosomes strong alternative candidates for clinical treatment.147 However, several challenges may be encountered during the clinical application of exosomes. First, the extraction and storage processes of exosomes must be optimized, which will benefit large-scale production of exosomes for clinical use. Second, the efficiency of drug loading and drug release of the exosome-based drug delivery system needs to be improved. Third, exosomes derived from various sources exhibit different functions in patients with different physiological or pathological statuses, which require further exploration. Therefore, further in-depth understanding of the effects of exosomes derived from different cell types or body fluids on gynecologic cancers can provide a rational theoretical basis for the targeted clinical treatment of ovarian cancer, cervical cancer, and endometrial cancer.
Acknowledgments
This work was sponsored by the National Natural Science Foundation of China (no. 81671809).
Author contributions
M.Y., J.W., S.P., and L.Z. searched the literature. M.Y. and J.W. wrote the manuscript. M.Y. and S.P. made the figures and tables. Z.W. and X.Z. designed the study and revised the manuscript. All authors read and approved the final manuscript.
Declaration of interests
The authors declare no competing interests.
Contributor Information
Zhi-Wei Wang, Email: zhiweichina@126.com.
Xueqiong Zhu, Email: zjwzzxq@163.com.
References
- 1.Grywalska E., Sobstyl M., Putowski L., Roliński J. Current possibilities of gynecologic cancer treatment with the use of immune checkpoint inhibitors. Int. J. Mol. Sci. 2019;20:4705. doi: 10.3390/ijms20194705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Bowtell D.D., Böhm S., Ahmed A.A., Aspuria P.J., Bast R.C., Jr., Beral V., Berek J.S., Birrer M.J., Blagden S., Bookman M.A., et al. Rethinking ovarian cancer II: reducing mortality from high-grade serous ovarian cancer. Nat. Rev. Cancer. 2015;15:668–679. doi: 10.1038/nrc4019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Siegel, R.L.,Miller, K.D.,Fuchs, H.E.,Jemal, A., Cancer statistics, 2021 CA Cancer J. Clin. , 71 (1), 7-33. [DOI] [PubMed]
- 4.Sung H., Ferlay J., Siegel R.L., Laversanne M., Soerjomataram I., Jemal A., Bray F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2021;71:209–249. doi: 10.3322/caac.21660. [DOI] [PubMed] [Google Scholar]
- 5.Wang W.-L., Hong G.-C., Chien P.-J., Huang Y.-H., Lee H.-T., Wang P.-H., Lee Y.-C., Chang W.-W. Tribbles pseudokinase 3 contributes to cancer stemness of endometrial cancer cells by regulating β-catenin expression. Cancers. 2020;12:3785. doi: 10.3390/cancers12123785. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Wan Y., Liu B., Lei H., Zhang B., Wang Y., Huang H., Chen S., Feng Y., Zhu L., Gu Y., et al. Nanoscale extracellular vesicle-derived DNA is superior to circulating cell-free DNA for mutation detection in early-stage non-small-cell lung cancer. Ann. Oncol. 2018;29:2379–2383. doi: 10.1093/annonc/mdy458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Dias M.V., Teixeira B.L., Rodrigues B.R., Sinigaglia-Coimbra R., Porto-Carreiro I., Roffe M., Hajj G.N., Martins V.R. PRNP/prion protein regulates the secretion of exosomes modulating CAV1/caveolin-1-suppressed autophagy. Autophagy. 2016;12:2113–2128. doi: 10.1080/15548627.2016.1226735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Dioufa N., Clark A.M., Ma B., Beckwitt C.H., Wells A. Bi-directional exosome-driven intercommunication between the hepatic niche and cancer cells. Mol. Cancer. 2017;16:172. doi: 10.1186/s12943-017-0740-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Shen Z., Huang W., Liu J., Tian J., Wang S., Rui K. Effects of mesenchymal stem cell-derived exosomes on autoimmune diseases. Front Immunol. 2021;12:749192. doi: 10.3389/fimmu.2021.749192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Kano S.I., Dohi E., Rose I.V.L. Extracellular vesicles for research on psychiatric disorders. Schizophr. Bull. 2019;45:7–16. doi: 10.1093/schbul/sby127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Tan A., Rajadas J., Seifalian A.M. Exosomes as nano-theranostic delivery platforms for gene therapy. Adv. Drug Deliv. Rev. 2013;65:357–367. doi: 10.1016/j.addr.2012.06.014. [DOI] [PubMed] [Google Scholar]
- 12.Yin B., Ma Q., Song C., Zhao L., Yu F., Wang C., Shi Y., Ye L. Exosome-derived noncoding RNAs as a promising treatment of bone regeneration. Stem Cells Int. 2021;2021:6696894. doi: 10.1155/2021/6696894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Mathivanan S., Fahner C.J., Reid G.E., Simpson R.J. ExoCarta 2012: database of exosomal proteins, RNA and lipids. Nucleic Acids Res. 2012;40:D1241–D1244. doi: 10.1093/nar/gkr828. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Zhong Y., Li H., Li P., Chen Y., Zhang M., Yuan Z., Zhang Y., Xu Z., Luo G., Fang Y., et al. Exosomes: a new pathway for cancer drug resistance. Front Oncol. 2021;11:743556. doi: 10.3389/fonc.2021.743556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Lopez-Verrilli M.A., Court F.A. Exosomes: mediators of communication in eukaryotes. Biol. Res. 2013;46:5–11. doi: 10.4067/S0716-97602013000100001. [DOI] [PubMed] [Google Scholar]
- 16.Zabeo D., Cvjetkovic A., Lässer C., Schorb M., Lötvall J., Höög J.L. Exosomes purified from a single cell type have diverse morphology. J. Extracell. Vesicles. 2017;6:1329476. doi: 10.1080/20013078.2017.1329476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Soung Y.H., Nguyen T., Cao H., Lee J., Chung J. Emerging roles of exosomes in cancer invasion and metastasis. BMB Rep. 2016;49:18–25. doi: 10.5483/BMBRep.2016.49.1.239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Oliveira F.D., Castanho M., Neves V. Exosomes and brain metastases: a review on their role and potential applications. Int. J. Mol. Sci. 2021;22:10899. doi: 10.3390/ijms221910899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Li P., Kaslan M., Lee S.H., Yao J., Gao Z. Progress in exosome isolation techniques. Theranostics. 2017;7:789–804. doi: 10.7150/thno.18133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Mao W., Wang K., Wu Z., Xu B., Chen M. Current status of research on exosomes in general, and for the diagnosis and treatment of kidney cancer in particular. J. Exp. Clin. Cancer Res. 2021;40:305. doi: 10.1186/s13046-021-02114-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Bunggulawa E.J., Wang W., Yin T., Wang N., Durkan C., Wang Y., Wang G. Recent advancements in the use of exosomes as drug delivery systems. J. Nanobiotechnol. 2018;16:81. doi: 10.1186/s12951-018-0403-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Soliman H.M., Ghonaim G.A., Gharib S.M., Chopra H., Farag A.K., Hassanin M.H., Nagah A., Emad-Eldin M., Hashem N.E., Yahya G., et al. Exosomes in Alzheimer’s disease: from being pathological players to potential diagnostics and therapeutics. Int. J. Mol. Sci. 2021;22:10794. doi: 10.3390/ijms221910794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Jiang K., Dong C., Yin Z., Li R., Mao J., Wang C., Zhang J., Gao Z., Liang R., Wang Q., et al. Exosome-derived ENO1 regulates integrin alpha6beta4 expression and promotes hepatocellular carcinoma growth and metastasis. Cell Death Dis. 2020;11:972. doi: 10.1038/s41419-020-03179-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Logozzi M., Capasso C., Di Raimo R., Del Prete S., Mizzoni D., Falchi M., Supuran C.T., Fais S. Prostate cancer cells and exosomes in acidic condition show increased carbonic anhydrase IX expression and activity. J. Enzyme Inhib. Med. Chem. 2019;34(1):272–278. doi: 10.1080/14756366.2018.1538980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Wu M., Wang G., Hu W., Yao Y., Yu X.F. Emerging roles and therapeutic value of exosomes in cancer metastasis. Mol. Cancer. 2019;18:53. doi: 10.1186/s12943-019-0964-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Dai J., Su Y., Zhong S., Cong L., Liu B., Yang J., Tao Y., He Z., Chen C., Jiang Y. Exosomes: key players in cancer and potential therapeutic strategy. Signal Transduct. Target. Ther. 2020;5:145. doi: 10.1038/s41392-020-00261-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Kogure T., Lin W.L., Yan I.K., Braconi C., Patel T. Intercellular nanovesicle-mediated microRNA transfer: a mechanism of environmental modulation of hepatocellular cancer cell growth. Hepatology. 2011;54:1237–1248. doi: 10.1002/hep.24504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Zhang L., Zhang S., Yao J., Lowery F.J., Zhang Q., Huang W.C., Li P., Li M., Wang X., Zhang C., et al. Microenvironment-induced PTEN loss by exosomal microRNA primes brain metastasis outgrowth. Nature. 2015;527:100–104. doi: 10.1038/nature15376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Nobuyoshi K., Haruhisa I., Keitaro H., Yusuke Y., Fumitaka T., Takahiro O. Neutral sphingomyelinase 2 (nSMase2)-dependent exosomal transfer of angiogenic microRNAs regulate cancer cell metastasis. J. Biol. Chem. 2013;288:10849–10859. doi: 10.1074/jbc.M112.446831-59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Muller F., Alessio P., Federica C., Roberta G., Eugenio G., Ramasamy S., Francesca L., Paolo F., Charlene M., Gerard J.N., et al. MicroRNAs bind to toll-like receptors to induce prometastatic inflammatory response. Proc. Natl. Acad. Sci. U S A. 2012;109:E2110–E2116. doi: 10.1073/pnas.1209414109-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Kishore B.C., Petra M.W., Paolo N., Haritha C., Mariam M., Tong X., Rebekah K., Cristina I., Xinna Z., Ivan V., et al. Exosome-mediated transfer of microRNAs within the tumor microenvironment and neuroblastoma resistance to chemotherapy. J. Natl. Cancer Inst. 2015;107:djv135. doi: 10.1093/jnci/djv135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Feng W., Dean D.C., Hornicek F.J., Shi H., Duan Z. Exosomes promote pre-metastatic niche formation in ovarian cancer. Mol. Cancer. 2019;18:124. doi: 10.1186/s12943-019-1049-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Mikaelian I., Scicchitano M., Mendes O., Thomas R.A., Leroy B.E. Frontiers in preclinical safety biomarkers: microRNAs and messenger RNAs. Toxicol. Pathol. 2013;41:18–31. doi: 10.1177/0192623312448939. [DOI] [PubMed] [Google Scholar]
- 34.Koga Y., Yasunaga M., Moriya Y., Akasu T., Fujita S., Yamamoto S., Matsumura Y. Exosome can prevent RNase from degrading microRNA in feces. J. Gastrointest. Oncol. 2011;2:215–222. doi: 10.3978/j.issn.2078-6891.2011.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Cheng C., Zhang Z., Cheng F., Shao Z. Exosomal lncRNA RAMP2-AS1 derived from chondrosarcoma cells promotes angiogenesis through miR-2355-5p/VEGFR2 Axis. Onco Targets Ther. 2020;13:3291–3301. doi: 10.2147/OTT.S244652. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Li W., Xu C., Guo J., Liu K., Hu Y., Wu D., Fang H., Zou Y., Wei Z., Wang Z., et al. Cis- and trans-acting expression quantitative trait loci of long non-coding RNA in 2,549 cancers with potential clinical and therapeutic implications. Front Oncol. 2020;10:602104. doi: 10.3389/fonc.2020.602104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Gluud M., Willerslev-Olsen A., Gjerdrum L.M.R., Lindahl L.M., Buus T.B., Andersen M.H., Bonefeld C.M., Krejsgaard T., Litvinov I.V., Iversen L., et al. MicroRNAs in the pathogenesis, diagnosis, prognosis and targeted treatment of cutaneous T-cell lymphomas. Cancers (Basel) 2020;12:1229. doi: 10.3390/cancers12051229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Yuan L., Xu Z.Y., Ruan S.M., Mo S., Qin J.J., Cheng X.D. Long non-coding RNAs towards precision medicine in gastric cancer: early diagnosis, treatment, and drug resistance. Mol. Cancer. 2020;19:96. doi: 10.1186/s12943-020-01219-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.He L., Zhang A., Xiong L., Li Y., Huang R., Liao L., Zhu Z., Wang A.Y. Deep circular RNA sequencing provides insights into the mechanism underlying grass carp reovirus infection. Int. J. Mol. Sci. 2017;18:1977. doi: 10.3390/ijms18091977. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Wang Y., Mo Y., Gong Z., Yang X., Yang M., Zhang S., Xiong F., Xiang B., Zhou M., Liao Q., et al. Circular RNAs in human cancer. Mol. Cancer. 2017;16:25. doi: 10.1186/s12943-017-0598-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Nakamura K., Sawada K., Kobayashi M., Miyamoto M., Shimizu A., Yamamoto M., Kinose Y., Kimura T. Role of the exosome in ovarian cancer progression and its potential as a therapeutic target. Cancers (Basel) 2019;11:1147. doi: 10.3390/cancers11081147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Xiao Y., Lai Y., Yu Y., Jiang P., Li Y., Wang C., Zhang R. The exocrine differentiation and proliferation factor (EXDPF) gene promotes ovarian cancer tumorigenesis by up-regulating DNA replication pathway. Front. Oncol. 2021;11:669603. doi: 10.3389/fonc.2021.669603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Filippov-Levy N., Reich R., Davidson B. The biological and clinical role of the long non-coding RNA LOC642852 in ovarian carcinoma. Int. J. Mol. Sci. 2020;21:5237. doi: 10.3390/ijms21155237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Trinidad C.V., Tetlow A.L., Bantis L.E., Godwin A.K. Reducing ovarian cancer mortality through early detection: approaches using circulating biomarkers. Cancer Prev. Res. (Phila) 2020;13:241–252. doi: 10.1158/1940-6207.CAPR-19-0184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Chen Z., Liang Q., Zeng H., Zhao Q., Guo Z., Zhong R., Xie M., Cai X., Su J., He Z., et al. Exosomal CA125 as a promising biomarker for ovarian cancer diagnosis. J. Cancer. 2020;11:6445–6453. doi: 10.7150/jca.48531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Liu S., Zhan Y., Luo J., Feng J., Lu J., Zheng H., Wen Q., Fan S. Roles of exosomes in the carcinogenesis and clinical therapy of non-small cell lung cancer. Biomed. Pharmacother. 2019;111:338–346. doi: 10.1016/j.biopha.2018.12.088. [DOI] [PubMed] [Google Scholar]
- 47.Shimizu A., Sawada K., Kimura T. Pathophysiological role and potential therapeutic exploitation of exosomes in ovarian cancer. Cells. 2020;9:814. doi: 10.3390/cells9040814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Miharu Kobayashi C.S., Tapia J., Illanes S.E., Mitchell M.D., Rice G.E. Ovarian cancer cell invasiveness is associated with discordant exosomal sequestration of Let-7 miRNA and miR-200. J. Transl Med. 2014;12:4. doi: 10.1186/1479-5876-12-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Yoshimura A., Sawada K., Nakamura K., Kinose Y., Nakatsuka E., Kobayashi M., Miyamoto M., Ishida K., Matsumoto Y., Kodama M., et al. Exosomal miR-99a-5p is elevated in sera of ovarian cancer patients and promotes cancer cell invasion by increasing fibronectin and vitronectin expression in neighboring peritoneal mesothelial cells. BMC Cancer. 2018;18:1065. doi: 10.1186/s12885-018-4974-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.He L., Zhu W., Chen Q., Yuan Y., Wang Y., Wang J., Wu X. Ovarian cancer cell-secreted exosomal miR-205 promotes metastasis by inducing angiogenesis. Theranostics. 2019;9:8206–8220. doi: 10.7150/thno.37455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Wang L., Zhao F., Xiao Z., Yao L. Exosomal microRNA-205 is involved in proliferation, migration, invasion, and apoptosis of ovarian cancer cells via regulating VEGFA. Cancer Cell Int. 2019;19:281. doi: 10.1186/s12935-019-0990-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Alharbi M., Lai A., Guanzon D., Palma C., Zuniga F., Perrin L., He Y., Hooper J.D., Salomon C. Ovarian cancer-derived exosomes promote tumour metastasis in vivo: an effect modulated by the invasiveness capacity of their originating cells. Clin. Sci. (Lond) 2019;133:1401–1419. doi: 10.1042/CS20190082. [DOI] [PubMed] [Google Scholar]
- 53.Guan X., Zong Z.H., Liu Y., Chen S., Wang L.L., Zhao Y. circPUM1 promotes tumorigenesis and progression of ovarian cancer by sponging miR-615-5p and miR-6753-5p. Mol. Ther. Nucleic Acids. 2019;18:882–892. doi: 10.1016/j.omtn.2019.09.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Zong Z.H., Du Y.P., Guan X., Chen S., Zhao Y. CircWHSC1 promotes ovarian cancer progression by regulating MUC1 and hTERT through sponging miR-145 and miR-1182. J. Exp. Clin. Cancer Res. 2019;38:437. doi: 10.1186/s13046-019-1437-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Huan Y., Ye J., Yang X.M., Zhang L.W., Zhang Z.G., Chen Y.P. High-grade ovarian cancer secreting effective exosomes in tumor angiogenesis. Int. J. Clin. Exp. Pathol. 2015;8:5062–5070. [PMC free article] [PubMed] [Google Scholar]
- 56.Li Z., Yan-Qing W., Xiao Y., Shi-Yi L., Meng-Qin Y., Shu X., Dong-Yong Y., Ya-Jing Z., Yan-Xiang C. Exosomes secreted by chemoresistant ovarian cancer cells promote angiogenesis. J. Ovarian Res. 2021;14:7. doi: 10.1186/s13048-020-00758-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Chen X., Ying X., Wang X., Wu X., Zhu Q., Wang X. Exosomes derived from hypoxic epithelial ovarian cancer deliver microRNA-940 to induce macrophage M2 polarization. Oncol. Rep. 2017;38:522–528. doi: 10.3892/or.2017.5697. [DOI] [PubMed] [Google Scholar]
- 58.Kanlikilicer P., Bayraktar R., Denizli M., Rashed M.H., Ivan C., Aslan B., Mitra R., Karagoz K., Bayraktar E., Zhang X., et al. Exosomal miRNA confers chemo resistance via targeting Cav1/p-gp/M2-type macrophage axis in ovarian cancer. EBioMedicine. 2018;38:100–112. doi: 10.1016/j.ebiom.2018.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Zhang Y., Cai H., Chen S., Sun D., Zhang D., He Y. Exosomal transfer of miR-124 inhibits normal fibroblasts to cancer-associated fibroblasts transition by targeting sphingosine kinase 1 in ovarian cancer. J. Cell Biochem. 2019;120:13187–13201. doi: 10.1002/jcb.28593. [DOI] [PubMed] [Google Scholar]
- 60.Kobayashi M., Sawada K., Miyamoto M., Shimizu A., Yamamoto M., Kinose Y., Nakamura K., Kawano M., Kodama M., Hashimoto K., et al. Exploring the potential of engineered exosomes as delivery systems for tumor-suppressor microRNA replacement therapy in ovarian cancer. Biochem. Biophys. Res. Commun. 2020;527:153–161. doi: 10.1016/j.bbrc.2020.04.076. [DOI] [PubMed] [Google Scholar]
- 61.Hu Y., Li D., Wu A., Qiu X., Di W., Huang L., Qiu L. TWEAK-stimulated macrophages inhibit metastasis of epithelial ovarian cancer via exosomal shuttling of microRNA. Cancer Lett. 2017;393:60–67. doi: 10.1016/j.canlet.2017.02.009. [DOI] [PubMed] [Google Scholar]
- 62.Yang Z., Wang W., Zhao L., Wang X., Gimple R.C., Xu L., Wang Y., Rich J.N., Zhou S. Plasma cells shape the mesenchymal identity of ovarian cancers through transfer of exosome-derived microRNAs. Sci. Adv. 2021;7:eabb0737. doi: 10.1126/sciadv.abb0737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Guo H., Ha C., Dong H., Yang Z., Ma Y., Ding Y. Cancer-associated fibroblast-derived exosomal microRNA-98-5p promotes cisplatin resistance in ovarian cancer by targeting CDKN1A. Cancer Cell Int. 2019;19:347. doi: 10.1186/s12935-019-1051-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Xu Y., Xu L., Zheng J., Geng L., Zhao S. MiR-101 inhibits ovarian carcinogenesis by repressing the expression of brain-derived neurotrophic factor. FEBS Open Bio. 2017;7:1258–1266. doi: 10.1002/2211-5463.12257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Wu Q., Wu X., Ying X., Zhu Q., Wang X., Jiang L., Chen X., Wu Y., Wang X. Suppression of endothelial cell migration by tumor associated macrophage-derived exosomes is reversed by epithelial ovarian cancer exosomal lncRNA. Cancer Cell Int. 2017;17:62. doi: 10.1186/s12935-017-0430-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Zhang H., Xu S., Liu X. MicroRNA profiling of plasma exosomes from patients with ovarian cancer using high-throughput sequencing. Oncol. Lett. 2019;17:5601–5607. doi: 10.3892/ol.2019.10220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Wang X., Yao Y., Jin M. Circ-0001068 is a novel biomarker for ovarian cancer and inducer of PD1 expression in T cells. Aging (Albany NY) 2020;12:19095–19106. doi: 10.18632/aging.103706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Maeda K., Sasaki H., Ueda S., Miyamoto S., Terada S., Konishi H., Kogata Y., Ashihara K., Fujiwara S., Tanaka Y., et al. Serum exosomal microRNA-34a as a potential biomarker in epithelial ovarian cancer. J. Ovarian Res. 2020;13:47. doi: 10.1186/s13048-020-00648-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Wang C., Qi S., Xie C., Li C., Wang P., Liu D. Upregulation of long non-coding RNA XIST has anticancer effects on epithelial ovarian cancer cells through inverse downregulation of hsa-miR-214-3p. J. Gynecol. Oncol. 2018;29:e99. doi: 10.3802/jgo.2018.29.e99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Yang C., Kim H.S., Park S.J., Lee E.J., Kim S.I., Song G., Lim W. Inhibition of miR-214-3p aids in preventing epithelial ovarian cancer malignancy by increasing the expression of LHX6. Cancers (Basel) 2019;11:1917. doi: 10.3390/cancers11121917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Keseru J.S., Soltesz B., Lukacs J., Marton E., Szilagyi-Bonizs M., Penyige A., Poka R., Nagy B. Detection of cell-free, exosomal and whole blood mitochondrial DNA copy number in plasma or whole blood of patients with serous epithelial ovarian cancer. J. Biotechnol. 2019;298:76–81. doi: 10.1016/j.jbiotec.2019.04.015. [DOI] [PubMed] [Google Scholar]
- 72.Su Y.Y., Sun L., Guo Z.R., Li J.C., Bai T.T., Cai X.X., Li W.H., Zhu Y.F. Upregulated expression of serum exosomal miR-375 and miR-1307 enhance the diagnostic power of CA125 for ovarian cancer. J. Ovarian Res. 2019;12:6. doi: 10.1186/s13048-018-0477-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Kim S., Choi M.C., Jeong J.Y., Hwang S., Jung S.G., Joo W.D., Park H., Song S.H., Lee C., Kim T.H., et al. Serum exosomal miRNA-145 and miRNA-200c as promising biomarkers for preoperative diagnosis of ovarian carcinomas. J. Cancer. 2019;10:1958–1967. doi: 10.7150/jca.30231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Zhang W., Su X., Li S., Liu Z., Wang Q., Zeng H. Low serum exosomal miR-484 expression predicts unfavorable prognosis in ovarian cancer. Cancer Biomark. 2020;27:485–491. doi: 10.3233/CBM-191123. [DOI] [PubMed] [Google Scholar]
- 75.Pan C., Stevic I., Muller V., Ni Q., Oliveira-Ferrer L., Pantel K., Schwarzenbach H. Exosomal microRNAs as tumor markers in epithelial ovarian cancer. Mol. Oncol. 2018;12:1935–1948. doi: 10.1002/1878-0261.12371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Qiu J.J., Lin X.J., Tang X.Y., Zheng T.T., Lin Y.Y., Hua K.Q. Exosomal metastasis associated lung adenocarcinoma transcript 1 promotes angiogenesis and predicts poor prognosis in epithelial ovarian cancer. Int. J. Biol. Sci. 2018;14:1960–1973. doi: 10.7150/ijbs.28048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Zhou J., Gong G., Tan H., Dai F., Zhu X., Chen Y., Wang J., Liu Y., Chen P., Wu X., et al. Urinary microRNA-30a-5p is a potential biomarker for ovarian serous adenocarcinoma. Oncol. Rep. 2015;33:2915–2923. doi: 10.3892/or.2015.3937. [DOI] [PubMed] [Google Scholar]
- 78.Cappellesso R., Tinazzi A., Giurici T., Simonato F., Guzzardo V., Ventura L., Crescenzi M., Chiarelli S., Fassina A. Programmed cell death 4 and microRNA 21 inverse expression is maintained in cells and exosomes from ovarian serous carcinoma effusions. Cancer Cytopathol. 2014;122:685–693. doi: 10.1002/cncy.21442. [DOI] [PubMed] [Google Scholar]
- 79.Cai J., Gong L., Li G., Guo J., Yi X., Wang Z. Exosomes in ovarian cancer ascites promote epithelial-mesenchymal transition of ovarian cancer cells by delivery of miR-6780b-5p. Cell Death Dis. 2021;12:210. doi: 10.1038/s41419-021-03490-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Bebelman M.P., Smit M.J., Pegtel D.M., Baglio S.R. Biogenesis and function of extracellular vesicles in cancer. Pharmacol. Ther. 2018;188:1–11. doi: 10.1016/j.pharmthera.2018.02.013. [DOI] [PubMed] [Google Scholar]
- 81.Soltesz B., Lukacs J., Szilagyi E., Marton E., Szilagyi Bonizs M., Penyige A., Poka R., Nagy B. Expression of CD24 in plasma, exosome and ovarian tissue samples of serous ovarian cancer patients. J. Biotechnol. 2019;298:16–20. doi: 10.1016/j.jbiotec.2019.03.018. [DOI] [PubMed] [Google Scholar]
- 82.Runz S., Keller S., Rupp C., Stoeck A., Issa Y., Koensgen D., Mustea A., Sehouli J., Kristiansen G., Altevogt P. Malignant ascites-derived exosomes of ovarian carcinoma patients contain CD24 and EpCAM. Gynecol. Oncol. 2007;107:563–571. doi: 10.1016/j.ygyno.2007.08.064. [DOI] [PubMed] [Google Scholar]
- 83.Zhao Z., Yang Y., Zeng Y., He M. A microfluidic ExoSearch chip for multiplexed exosome detection towards blood-based ovarian cancer diagnosis. Lab. Chip. 2016;16:489–496. doi: 10.1039/c5lc01117e. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Zhang P., He M., Zeng Y. Ultrasensitive microfluidic analysis of circulating exosomes using a nanostructured graphene oxide/polydopamine coating. Lab. Chip. 2016;16:3033–3042. doi: 10.1039/c6lc00279j. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Im H., Shao H., Park Y.I., Peterson V.M., Castro C.M., Weissleder R., Lee H. Label-free detection and molecular profiling of exosomes with a nano-plasmonic sensor. Nat. Biotechnol. 2014;32:490–495. doi: 10.1038/nbt.2886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Zhang P., Zhou X., He M., Shang Y., Tetlow A.L., Godwin A.K., Zeng Y. Ultrasensitive detection of circulating exosomes with a 3D-nanopatterned microfluidic chip. Nat. Biomed. Eng. 2019;3:438–451. doi: 10.1038/s41551-019-0356-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Dorayappan K.D.P., Gardner M.L., Hisey C.L., Zingarelli R.A., Smith B.Q., Lightfoot M.D.S., Gogna R., Flannery M.M., Hays J., Hansford D.J., et al. A microfluidic chip enables isolation of exosomes and establishment of their protein profiles and associated signaling pathways in ovarian cancer. Cancer Res. 2019;79:3503–3513. doi: 10.1158/0008-5472.CAN-18-3538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Zhang W., Ou X., Wu X. Proteomics profiling of plasma exosomes in epithelial ovarian cancer: a potential role in the coagulation cascade, diagnosis and prognosis. Int. J. Oncol. 2019;54:1719–1733. doi: 10.3892/ijo.2019.4742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Szajnik M., Derbis M., Lach M., Patalas P., Michalak M., Drzewiecka H., Szpurek D., Nowakowski A., Spaczynski M., Baranowski W., et al. Exosomes in plasma of patients with ovarian carcinoma: potential biomarkers of tumor progression and response to therapy. Gynecol. Obstet. (Sunnyvale) 2013;(Suppl 4):3. doi: 10.4172/2161-0932.S4-003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Shen X., Wang C., Zhu H., Wang Y., Wang X., Cheng X., Ge W., Lu W. Exosome-mediated transfer of CD44 from high-metastatic ovarian cancer cells promotes migration and invasion of low-metastatic ovarian cancer cells. J. Ovarian Res. 2021;14:38. doi: 10.1186/s13048-021-00776-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Asare-Werehene M., Nakka K., Reunov A., Chiu C.T., Lee W.T., Abedini M.R., Wang P.W., Shieh D.B., Dilworth F.J., Carmona E., et al. The exosome-mediated autocrine and paracrine actions of plasma gelsolin in ovarian cancer chemoresistance. Oncogene. 2020;39:1600–1616. doi: 10.1038/s41388-019-1087-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Nakamura K., Sawada K., Kinose Y., Yoshimura A., Toda A., Nakatsuka E., Hashimoto K., Mabuchi S., Morishige K.I., Kurachi H., et al. Exosomes promote ovarian cancer cell invasion through transfer of CD44 to peritoneal mesothelial cells. Mol. Cancer Res. 2017;15:78–92. doi: 10.1158/1541-7786.MCR-16-0191. [DOI] [PubMed] [Google Scholar]
- 93.Tang X., Liu S., Liu Y., Lin X., Zheng T., Liu X., Qiu J., Hua K. Circulating serum exosomal aHIF is a novel prognostic predictor for epithelial ovarian cancer. Onco Targets Ther. 2019;12:7699–7711. doi: 10.2147/OTT.S220533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Yi H., Zheng X., Song J., Shen R., Su Y., Lin D. Exosomes mediated pentose phosphate pathway in ovarian cancer metastasis: a proteomics analysis. Int. J. Clin. Exp. Pathol. 2015;8:15719–15728. [PMC free article] [PubMed] [Google Scholar]
- 95.Liang B., Peng P., Chen S., Li L., Zhang M., Cao D., Yang J., Li H., Gui T., Li X., et al. Characterization and proteomic analysis of ovarian cancer-derived exosomes. J. Proteomics. 2013;80:171–182. doi: 10.1016/j.jprot.2012.12.029. [DOI] [PubMed] [Google Scholar]
- 96.Zhang P., Zhou X., Zeng Y. Multiplexed immunophenotyping of circulating exosomes on nano-engineered ExoProfile chip towards early diagnosis of cancer. Chem. Sci. 2019;10:5495–5504. doi: 10.1039/c9sc00961b. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Cheng L., Zhang K., Qing Y., Li D., Cui M., Jin P., Xu T. Proteomic and lipidomic analysis of exosomes derived from ovarian cancer cells and ovarian surface epithelial cells. J. Ovarian Res. 2020;13:9. doi: 10.1186/s13048-020-0609-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Kanlikilicer P., Rashed M.H., Bayraktar R., Mitra R., Ivan C., Aslan B., Zhang X., Filant J., Silva A.M., Rodriguez-Aguayo C., et al. Ubiquitous release of exosomal tumor suppressor miR-6126 from ovarian cancer cells. Cancer Res. 2016;76:7194–7207. doi: 10.1158/0008-5472.CAN-16-0714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Li Z., Niu H., Qin Q., Yang S., Wang Q., Yu C., Wei Z., Jin Z., Wang X., Yang A., et al. lncRNA UCA1 mediates resistance to cisplatin by regulating the miR-143/FOSL2-signaling pathway in ovarian cancer. Mol. Ther. Nucleic Acids. 2019;17:92–101. doi: 10.1016/j.omtn.2019.05.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Zhao Z., Ji M., Wang Q., He N., Li Y. Circular RNA Cdr1as upregulates SCAI to suppress cisplatin resistance in ovarian cancer via miR-1270 suppression. Mol. Ther. Nucleic Acids. 2019;18:24–33. doi: 10.1016/j.omtn.2019.07.012. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 101.Luo Y., Gui R. Circulating exosomal circFoxp1 confers cisplatin resistance in epithelial ovarian cancer cells. J. Gynecol. Oncol. 2020;31:e75. doi: 10.3802/jgo.2020.31.e75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Cao Y.L., Zhuang T., Xing B.H., Li N., Li Q. Exosomal DNMT1 mediates cisplatin resistance in ovarian cancer. Cell Biochem Funct. 2017;35:296–303. doi: 10.1002/cbf.3276. [DOI] [PubMed] [Google Scholar]
- 103.Alharbi M., Sharma S., Guanzon D., Lai A., Zuniga F., Shiddiky M.J.A., Yamauchi Y., Salas-Burgos A., He Y., Pejovic T., et al. miRNa signature in small extracellular vesicles and their association with platinum resistance and cancer recurrence in ovarian cancer. Nanomedicine. 2020;28:102207. doi: 10.1016/j.nano.2020.102207. [DOI] [PubMed] [Google Scholar]
- 104.Zhu X., Shen H., Yin X., Yang M., Wei H., Chen Q., Feng F., Liu Y., Xu W., Li Y. Macrophages derived exosomes deliver miR-223 to epithelial ovarian cancer cells to elicit a chemoresistant phenotype. J. Exp. Clin. Cancer Res. 2019;38:81. doi: 10.1186/s13046-019-1095-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Meshach Asare-Werehene L.C., Carmona E., Han Y., Song Y.S., Burger D., Mes-Masson A.M., Tsang B.K. Plasma gelsolin inhibits CD8+ T-cell function and regulates glutathione production to confer chemoresistance in ovarian cancer. Cancer Res. 2020;80:3959–3971. doi: 10.1158/0008-5472.CAN-20-0788. [DOI] [PubMed] [Google Scholar]
- 106.Au Yeung C.L., Co N.N., Tsuruga T., Yeung T.L., Kwan S.Y., Leung C.S., Li Y., Lu E.S., Kwan K., Wong K.K., et al. Exosomal transfer of stroma-derived miR21 confers paclitaxel resistance in ovarian cancer cells through targeting APAF1. Nat. Commun. 2016;7:11150. doi: 10.1038/ncomms11150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Liya Q., Wang J., Chen M., Chen F., Tu W. Exosomal microRNA-146a derived from mesenchymal stem cells increases the sensitivity of ovarian cancer cells to docetaxel and taxane via a LAMC2-mediated PI3K/Akt axis. Int. J. Mol. Med. 2020;46:609–620. doi: 10.3892/ijmm.2020.4634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Liu R., Zhang Y., Sun P., Wang C. DDP-resistant ovarian cancer cells-derived exosomal microRNA-30a-5p reduces the resistance of ovarian cancer cells to DDP. Open Biol. 2020;10:190173. doi: 10.1098/rsob.190173. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 109.Zhang X., Liu L., Tang M., Li H., Guo X., Yang X. The effects of umbilical cord-derived macrophage exosomes loaded with cisplatin on the growth and drug resistance of ovarian cancer cells. Drug Dev. Ind. Pharm. 2020;46:1150–1162. doi: 10.1080/03639045.2020.1776320. [DOI] [PubMed] [Google Scholar]
- 110.Liu H., Shen M., Zhao D., Ru D., Duan Y., Ding C., Li H. The effect of triptolide-loaded exosomes on the proliferation and apoptosis of human ovarian cancer SKOV3 cells. Biomed. Res. Int. 2019;2019:1–14. doi: 10.1155/2019/2595801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Huang X., Wang B., Chen R., Zhong S., Gao F., Zhang Y., Niu Y., Li C., Shi G. The nuclear Farnesoid X receptor reduces p53 ubiquitination and inhibits cervical cancer cell proliferation. Front. Cell Dev. Biol. 2021;9:583146. doi: 10.3389/fcell.2021.583146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Liu C., Lu J., Tian H., Du W., Zhao L., Feng J., Yuan D., Li Z. Increased expression of PDL1 by the human papillomavirus 16 E7 oncoprotein inhibits anticancer immunity. Mol. Med. Rep. 2017;15:1063–1070. doi: 10.3892/mmr.2017.6102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Taguchi A., Hara K., Tomio J., Kawana K., Tanaka T., Baba S., Kawata A., Eguchi S., Tsuruga T., Mori M., et al. Multistate Markov model to predict the prognosis of high-risk human papillomavirus-related cervical lesions. Cancers (Basel) 2020;12:270. doi: 10.3390/cancers12020270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Cancer Genome Atlas Research Network, Albert Einstein College of Medicine, Analytical Biological Services, Barretos Cancer Hospital, Baylor College of Medicine, Beckman Research Institute of City of Hope, Buck Institute for Research on Aging, Canada's Michael Smith Genome Sciences Centre, Harvard Medical School, Helen F. Graham Cancer Center &Research Institut, et al. Integrated genomic and molecular characterization of cervical cancer. Nature. 2017;543:378–384. doi: 10.1038/nature21386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Zhang L., Li H., Yuan M., Li M., Zhang S. Cervical cancer cells-secreted exosomal microRNA-221-3p promotes invasion, migration and angiogenesis of microvascular endothelial cells in cervical cancer by down-regulating MAPK10 expression. Cancer Manag. Res. 2019;11:10307–10319. doi: 10.2147/CMAR.S221527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Wu X.G., Zhou C.F., Zhang Y.M., Yan R.M., Wei W.F., Chen X.J., Yi H.Y., Liang L.J., Fan L.S., Liang L., et al. Cancer-derived exosomal miR-221-3p promotes angiogenesis by targeting THBS2 in cervical squamous cell carcinoma. Angiogenesis. 2019;22:397–410. doi: 10.1007/s10456-019-09665-1. [DOI] [PubMed] [Google Scholar]
- 117.You X., Wang Y., Meng J., Han S., Liu L., Sun Y., Zhang J., Sun S., Li X., Sun W., et al. Exosomal miR663b exposed to TGFbeta1 promotes cervical cancer metastasis and epithelial‑mesenchymal transition by targeting MGAT3. Oncol. Rep. 2021;45:1. doi: 10.3892/or.2021.7963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Honegger A., Leitz J., Bulkescher J., Hoppe-Seyler K., Hoppe-Seyler F. Silencing of human papillomavirus (HPV) E6/E7 oncogene expression affects both the contents and the amounts of extracellular microvesicles released from HPV-positive cancer cells. Int. J. Cancer. 2013;133:1631–1642. doi: 10.1002/ijc.28164. [DOI] [PubMed] [Google Scholar]
- 119.Honegger A., Schilling D., Bastian S., Sponagel J., Kuryshev V., Sultmann H., Scheffner M., Hoppe-Seyler K., Hoppe-Seyler F. Dependence of intracellular and exosomal microRNAs on viral E6/E7 oncogene expression in HPV-positive tumor cells. PLoS Pathog. 2015;11:e1004712. doi: 10.1371/journal.ppat.1004712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Luo X., Wei J., Yang F.L., Pang X.X., Shi F., Wei Y.X., Liao B.Y., Wang J.L. Exosomal lncRNA HNF1A-AS1 affects cisplatin resistance in cervical cancer cells through regulating microRNA-34b/TUFT1 axis. Cancer Cell Int. 2019;19:323. doi: 10.1186/s12935-019-1042-4. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 121.Bhat A., Sharma A., Bharti A.C. Upstream Hedgehog signaling components are exported in exosomes of cervical cancer cell lines. Nanomedicine (Lond) 2018;13:2127–2138. doi: 10.2217/nnm-2018-0143. [DOI] [PubMed] [Google Scholar]
- 122.Liang L.J., Yang Y., Wei W.F., Wu X.G., Yan R.M., Zhou C.F., Chen X.J., Wu S., Wang W., Fan L.S. Tumor-secreted exosomal Wnt2B activates fibroblasts to promote cervical cancer progression. Oncogenesis. 2021;10:30. doi: 10.1038/s41389-021-00319-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Li H., Chi X., Li R., Ouyang J., Chen Y. HIV-1-infected cell-derived exosomes promote the growth and progression of cervical cancer. Int. J. Biol. Sci. 2019;15:2438–2447. doi: 10.7150/ijbs.38146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Konishi H., Hayashi M., Taniguchi K., Nakamura M., Kuranaga Y., Ito Y., Kondo Y., Sasaki H., Terai Y., Akao Y., et al. The therapeutic potential of exosomal miR-22 for cervical cancer radiotherapy. Cancer Biol. Ther. 2020;21:1128–1135. doi: 10.1080/15384047.2020.1838031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Ma G., Song G., Zou X., Shan X., Liu Q., Xia T., Zhou X., Zhu W. Circulating plasma microRNA signature for the diagnosis of cervical cancer. Cancer Biomark. 2019;26:491–500. doi: 10.3233/CBM-190256. [DOI] [PubMed] [Google Scholar]
- 126.Lv A., Tu Z., Huang Y., Lu W., Xie B. Circulating exosomal miR-125a-5p as a novel biomarker for cervical cancer. Oncol. Lett. 2021;21:54. doi: 10.3892/ol.2020.12316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Zheng M., Hou L., Ma Y., Zhou L., Wang F., Cheng B., Wang W., Lu B., Liu P., Lu W., et al. Exosomal let-7d-3p and miR-30d-5p as diagnostic biomarkers for non-invasive screening of cervical cancer and its precursors. Mol. Cancer. 2019;18:76. doi: 10.1186/s12943-019-0999-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Ding X.Z., Zhang S.Q., Deng X.L., Qiang J.H. Serum exosomal lncRNA DLX6-AS1 is a promising biomarker for prognosis prediction of cervical cancer. Technol. Cancer Res. Treat. 2021;20 doi: 10.1177/1533033821990060. 1533033821990060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Zhang W., Zhou Q., Wei Y., Da M., Zhang C., Zhong J., Liu J., Shen J. The exosome-mediated PI3k/Akt/mTOR signaling pathway in cervical cancer. Int. J. Clin. Exp. Pathol. 2019;12:2474–2484. [PMC free article] [PubMed] [Google Scholar]
- 130.Liu J., Sun H., Wang X., Yu Q., Li S., Yu X., Gong W. Increased exosomal microRNA-21 and microRNA-146a levels in the cervicovaginal lavage specimens of patients with cervical cancer. Int. J. Mol. Sci. 2014;15:758–773. doi: 10.3390/ijms15010758. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Zhang J., Liu S.C., Luo X.H., Tao G.X., Guan M., Yuan H., Hu D.K. Exosomal long noncoding RNAs are differentially expressed in the cervicovaginal lavage samples of cervical cancer patients. J. Clin. Lab. Anal. 2016;30:1116–1121. doi: 10.1002/jcla.21990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Wu Y., Wang X., Meng L., Li W., Li C., Li P., Xu S. Changes of miRNA expression profiles from cervical-vaginal fluid-derived exosomes in response to HPV16 infection. Biomed. Res. Int. 2020;2020:7046894. doi: 10.1155/2020/7046894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Healey G.D., Pan-Castillo B., Garcia-Parra J., Davies J., Roberts S., Jones E., Dhar K., Nandanan S., Tofazzal N., Piggott L., et al. Antibody drug conjugates against the receptor for advanced glycation end products (RAGE), a novel therapeutic target in endometrial cancer. J. Immunother. Cancer. 2019;7:280. doi: 10.1186/s40425-019-0765-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Rousset-Rouviere S., Rochigneux P., Chretien A.S., Fattori S., Gorvel L., Provansal M., Lambaudie E., Olive D., Sabatier R. Endometrial carcinoma: immune microenvironment and emerging treatments in immuno-oncology. Biomedicines. 2021;9:632. doi: 10.3390/biomedicines9060632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Xiong H., Li Q., Chen R., Liu S., Lin Q., Xiong Z., Jiang Q., Guo L. A multi-step miRNA-mRNA regulatory network construction approach identifies gene signatures associated with endometrioid endometrial carcinoma. Genes (Basel) 2016;7:26. doi: 10.3390/genes7060026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Yanokura M., Banno K., Aoki D. MicroRNA34b expression enhances chemosensitivity of endometrial cancer cells to paclitaxel. Int. J. Oncol. 2020;57:1145–1156. doi: 10.3892/ijo.2020.5127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Maida Y., Takakura M., Nishiuchi T., Yoshimoto T., Kyo S. Exosomal transfer of functional small RNAs mediates cancer-stroma communication in human endometrium. Cancer Med. 2016;5:304–314. doi: 10.1002/cam4.545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Shi S., Tan Q., Feng F., Huang H., Liang J., Cao D., Wang Z. Identification of core genes in the progression of endometrial cancer and cancer cell-derived exosomes by an integrative analysis. Sci. Rep. 2020;10:9862. doi: 10.1038/s41598-020-66872-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Jia J., Guo S., Zhang D., Tian X., Xie X. Exosomal-lncRNA DLEU1 accelerates the proliferation, migration, and invasion of endometrial carcinoma cells by regulating microRNA-E2F3. Onco Targets Ther. 2020;13:8651–8663. doi: 10.2147/OTT.S262661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Li B.L., Lu W., Qu J.J., Ye L., Du G.Q., Wan X.P. Loss of exosomal miR-148b from cancer-associated fibroblasts promotes endometrial cancer cell invasion and cancer metastasis. J. Cell. Physiol. 2019;234:2943–2953. doi: 10.1002/jcp.27111. [DOI] [PubMed] [Google Scholar]
- 141.Jing L., Hua X., Yuanna D., Rukun Z., Junjun M. Exosomal miR-499a-5p inhibits endometrial cancer growth and metastasis via targeting VAV3. Cancer Manag. Res. 2020;12:13541–13552. doi: 10.2147/CMAR.S283747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Roman-Canal B., Moiola C.P., Gatius S., Bonnin S., Ruiz-Miro M., Gonzalez E., Gonzalez-Tallada X., Llordella I., Hernandez I., Porcel J.M., et al. EV-associated miRNAs from peritoneal lavage are a source of biomarkers in endometrial cancer. Cancers (Basel) 2019;11:839. doi: 10.3390/cancers11060839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Srivastava A., Moxley K., Ruskin R., Dhanasekaran D.N., Zhao Y.D., Ramesh R. A non-invasive liquid biopsy screening of urine-derived exosomes for miRNAs as biomarkers in endometrial cancer patients. AAPS J. 2018;20:82. doi: 10.1208/s12248-018-0220-y. [DOI] [PubMed] [Google Scholar]
- 144.Hanzi X., Zhen G., Shen Y., Fang Y., Zhong S. Circular RNA expression in extracellular vesicles isolated from serum of patients with endometrial cancer. Epigenomics. 2018;10:187–197. doi: 10.2217/epi-2017-0109. [DOI] [PubMed] [Google Scholar]
- 145.Xiaoxia C., Jian F., Chen C., Chang L., Liu G., Feng W. PCOS serum-derived exosomal miR-27a-5p stimulates endometrial cancer cells migration and invasion. J. Mol. Endocrinol. 2020;64:1–12. doi: 10.1530/JME-19-0159. [DOI] [PubMed] [Google Scholar]
- 146.Song Y., Wang M., Tong H., Tan Y., Hu X., Wang K., Wan X. Plasma exosomes from endometrial cancer patients contain LGALS3BP to promote endometrial cancer progression. Oncogene. 2021;40:633–646. doi: 10.1038/s41388-020-01555-x. [DOI] [PubMed] [Google Scholar]
- 147.Li M.Y., Liu L.Z., Dong M. Progress on pivotal role and application of exosome in lung cancer carcinogenesis, diagnosis, therapy and prognosis. Mol. Cancer. 2021;20:22. doi: 10.1186/s12943-021-01312-y. [DOI] [PMC free article] [PubMed] [Google Scholar]




