Next Article in Journal
Eosin Y-Functionalized Upconverting Nanoparticles: Nanophotosensitizers and Deep Tissue Bioimaging Agents for Simultaneous Therapeutic and Diagnostic Applications
Next Article in Special Issue
Nano-Electrochemical Characterization of a 3D Bioprinted Cervical Tumor Model
Previous Article in Journal
Perioperative Blood Transfusion Is Dose-Dependently Associated with Cancer Recurrence and Mortality after Head and Neck Cancer Surgery
Previous Article in Special Issue
Free and Poly-Methyl-Methacrylate-Bounded BODIPYs: Photodynamic and Antimigratory Effects in 2D and 3D Cancer Models
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Current Status of Research on Small Extracellular Vesicles for the Diagnosis and Treatment of Urological Tumors

1
The First School of Clinical Medicine, Gannan Medical University, Ganzhou 341000, China
2
Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
3
Department of Nephrology, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(1), 100; https://doi.org/10.3390/cancers15010100
Submission received: 5 November 2022 / Revised: 17 December 2022 / Accepted: 20 December 2022 / Published: 23 December 2022
(This article belongs to the Special Issue Cancer Nanotherapy and Nanodiagnostic)

Abstract

:

Simple Summary

Small extracellular vesicles (sEVs) play an important role in the occurrence and development of various diseases, exhibiting the characteristics of wide acquisition and strong specificity. Due to the difficulties in the diagnosis and treatment of urological tumors, this article mainly reviews the function of sEVs in urinary tumors and their related application in the diagnosis and treatment of such tumors. The contents herein can provide new directions for the early diagnosis and individualized treatment of urinary tumors.

Abstract

Extracellular vesicles (EVs) are important mediators of communication between tumor cells and normal cells. These vesicles are rich in a variety of contents such as RNA, DNA, and proteins, and can be involved in angiogenesis, epithelial-mesenchymal transition, the formation of pre-metastatic ecological niches, and the regulation of the tumor microenvironment. Small extracellular vesicles (sEVs) are a type of EVs. Currently, the main treatments for urological tumors are surgery, radiotherapy, and targeted therapy. However, urological tumors are difficult to diagnose and treat due to their high metastatic rate, tendency to develop drug resistance, and the low sensitivity of liquid biopsies. Numerous studies have shown that sEVs offer novel therapeutic options for tumor treatment, such as tumor vaccines and tumor drug carriers. sEVs have attracted a great deal of attention owing to their contribution to in intercellular communication, and as novel biomarkers, and role in the treatment of urological tumors. This article reviews the research and applications of sEVs in the diagnosis and treatment of urological tumors.

1. Background

Urological tumors can occur in any part of the urinary system. Common malignant forms of these tumors include renal cell carcinoma (RCC), prostate cancer (PCa), and bladder cancer (BC). According to the 2019 American Cancer Society, the number of new cases of urological tumors in the United States in 2019 was as high as 158,220, and while estimated number of deaths resulting from them was 33,420 [1]. These tumors are a significant cause of death in men worldwide [2]. Currently, the main treatments for urological tumors include surgery, radiotherapy, and targeted therapy, but the monitoring and treatment of these cancers yield poor outcomes [3]. The deep location of urologic tumors within the body makes renders them difficult to be diagnosed early [4]. Therefore, there exists a need for novel biomarkers and therapeutic tools to improve the diagnostic efficacy and survival of urologic tumors. Small extracellular vesicles (sEVs) have the potential to be developed as biomarkers. All cells release sEVs, which is a feature that makes them highly heterogeneous throughout the body and different cell types [5,6]. Currently, research on the development of sEVs as diagnostic markers for urological tumors has primarily focused on the development of sEVs from serum, urine, and plasma sources, especially for sEVs carrying microRNAs. Bodily fluid-derived sEVs can be used for the early diagnosis of tumors and prognostic monitoring. In particular, sEVs harboring RNAs for PCa detection have been included in the National Comprehensive Cancer Network guidelines as biomarkers for early prostate cancer detection [7]. Due to the characteristic membrane structure of sEVs, they can effectively block RNAases and proteases. Thus, sEVs can also be used as drug delivery vehicles to deliver anti-tumor drugs or genes associated with drug resistance to target tumors, thereby reducing their resistance to drugs and assisting in tumor therapy; the development of novel tumor vaccines can also inhibit tumor growth [8,9]. This article briefly describes the existing literature on sEVs and relevance and potential application value in the diagnosis and treatment of urological malignancies.

2. Overview of Small Extracellular Vesicles

Extracellular vesicles (EVs) are small lipid membrane-bound vesicles with diameters between 30–2000 nm that are secreted by different cells into the extracellular space [10,11]. The MISEV 2018 guidelines update the nomenclature rules to classify EVs into small extracellular vesicles and medium/large extracellular vesicles, depending on their size [12,13,14]. Of these three types, research has primarily focused on sEVs, and this is mainly because their formation pathways have been more thoroughly studied and they are more stable in bodily fluids. Moreover, sEVs can act as bridges for cellular communication between donor and recipient cells [15]. sEVs contain a variety of contents including nucleic acids and proteins, which allows them to perform important roles in the occurrence and development of various diseases [16].

2.1. Composition of Small Extracellular Vesicles

sEVs are largely comprised of EVs proteins, nucleic acids, and EVs lipids [17]. They are found in a variety of bodily fluids, including blood, urine, cerebrospinal fluid, ascites, and milk, among others [18]. Their contained proteins include heat shock proteins (hsp 70, hsp 90, etc.) [19], membrane proteins (CD 63, CD81, CD9, etc.) [20], multivesicular body (MVB)-forming proteins (Alix, TSG101, etc.) [21], and cytoskeletal proteins, which are involved in the formation and release of sEVs. Their contained lipids include cholesterol [21], ceramides [22], and sphingolipids [23], which can regulate the biological activity of sEVs. Their contained nucleic acids include DNA, mRNA, microRNA, circRNA, and lncRNA [24,25]. sEVs largely contribute to the development of diseases through their mechanisms involving these cargo substances.

2.2. Formation of Small Extracellular Vesicles

The mechanism of small extracellular vesicle formation is still not fully understood, with the most classical pathway being that involving the endosomal sorting complex required for transport (ESRCT) [26]. The specific formation process of sEVs includes the following. First, the cell membrane inverts to form endosomes, which carry genetic material and fuse with each other to form early nuclear endosomes. Subsequently, these early endosomes form MVBs via the ESRCT pathway, which are then released into the extracellular space by traction of the Ras-associated GTP-binding proteins 27a and 27b (Rab 27a and Rab 27b) (Figure 1) [27]. Also, sEVs can form through budding from the membrane.

2.3. Isolation Techniques for Small Extracellular Vesicles

There are currently many methods used for the isolation of small extracellular vesicles, but there is no consensus on the best extraction method. Commonly used methods include: differential centrifugation, size exclusion, precipitation, and immunoaffinity separation [28]. Each method features advantages and disadvantages, and the most suitable separation method should be selected by analyzing the downstream experimental needs [13,29]. (i) Currently, differential ultracentrifugation is the most commonly used method for isolating and concentrating sEVs. It usually involves low-speed centrifugation to remove cells and large vesicles, followed by ultra-high-speed centrifugation to collect sEVs. (ii) Density gradient centrifugation is a more rigorous form of ultra-centrifugation, where vesicles of different densities settle at different rates on a gradient. (iii) Chemicals such as polyethylene glycol can reduce the solubility of sEVs thereby causing them to precipitate, followed by low-speed centrifugation to obtain sEVs. (iv) Immunocapture involves the separation of vesicles coated with magnetic beads containing the target protein. (v) Size exclusion chromatography allows for the separation of EVs on a column depending on size, which may contain a certain amount of free protein etc. [13,30,31]. The advantages and disadvantages of the various separation methods are summarized in Table 1. Today, these methods still inevitably recover soluble impurities in some samples, such as lipoproteins in plasma [32].

3. General Function of Small Extracellular Vesicles in Urological Tumors

sEVs are secreted by a variety of different cell types and can play an important role in the development of diseases by transporting proteins, nucleic acids, and lipids. sEVs are involved in cell-to-cell communication, participate in the formation of the tumor microenvironment, affect inflammation and immune regulation, and affect angiogenesis and blood clotting, among numerous other roles [33,34,35,36]. In tumor diseases, sEVs can promote the epithelial-mesenchymal transformation (EMT) process of tumor cells by carrying miRNAs involved in the process [37]. In addition, sEVs plays an important role in tumor metastasis, immune evasion, and tumor resistance mechanisms (Figure 2) [38,39].

3.1. sEVs Promotes Angiogenesis in Urological Tumors

Angiogenesis refers to the formation of new blood vessels derived from the original vascular network, a process that is triggered by pro-angiogenic factors [40,41]. Tumor progression is a dynamic process that requires adequate nutrition and oxygen, and angiogenesis is an important mechanism underlying tumor progression [42]. The generation of new blood vessels in the primary tumor lesion can promote the growth and spread of tumor cells. Tumor cells can, in turn, promote angiogenesis by activating endothelial cells. Several studies have shown that miRNAs carried by sEVs can target vascular endothelial growth factor (VEGF), matrix metalloproteinase 2 (MMP2), and MMP9 to regulate angiogenesis [43,44,45,46,47,48]. A previous study by Zhang and coworkers demonstrated that RCC cell-derived sEVs promote the transformation of macrophages to the M2 phenotype, increase the expression of cytokines (such as TGFβ1), enhance the phagocytic ability of macrophages, and induce angiogenesis in RCC by transferring lncARSR, thereby promoting the occurrence and development of RCC [49]. Moreover, αvβ6 integrins in PCa-derived sEVs can be transferred to endothelial cells, which activates TGFβ1 and, results in the inhibition of STAT1 signaling, thereby promoting angiogenesis [50]. Similarly, sEVs secreted by PCa cells have been shown to enhance the angiogenesis and invasion capacity of human umbilical vein endothelial cells, and the same study also suggests that miR-27a-3p may be involved in this phenotypic change [51]. The active cathepsin B protein, which is carried by sEVs can be taken up by endothelial cells by mediating AKT axis phosphorylation, thereby increasing the expression of VEGF and promoting angiogenesis in BC [52]. Li and coworkers found that BC cells in a nutrient-deficient environment secreted sEVs containing glutamine-fructose-6-phosphate aminotransferase 1 (GFAT1) to enhance O-GlcNAcylation in endothelial cells, thus promoting angiogenesis, which indicates a new research direction for developing anti-angiogenic treatments in BC [53]. These studies have jointly shown that angiogenesis promotes tumor growth and infiltration.

3.2. sEVs Promotes Epithelial—Mesenchymal Transformation in Urological Tumors

EMT is a process in cancer by which tumor cells acquire invasive and migratory capabilities [54]. EMT primarily manifests by the decreased expression of epithelial markers (such as E-cadherin and β-catenin) and the increased expression of acquired mesenchymal phenotypes (such as vimentin, N-cadherin, and fibronectin) [55,56]. Several studies have shown that tumor-derived sEVs can promote the activation of cancer-associated cells and promote cellular EMT by carrying miRNAs [57,58,59]. A study by Wang and coworkers found that sEVs secreted by CD103+ cancer stem cells could inhibit phosphatase and tensin homolog (PTEN) expression through the delivery of miR-19b-3p, thereby inducing EMT in RCC cells. The migration and invasion ability of RCC tumor cells has also been found to become greatly enhanced after stem cell sEVs treatment [60]. Similarly, sEVs secreted by PC3 cells overexpressing prostate-specific G protein-coupled receptors were shown to promote PCa EMT, thereby promoting migration between PCa cells and normal cells. The exogenous elevation of prostate-specific G protein-coupled receptors (PSGR) occurs in PCa cells, whose secretion of PSGR-carrying sEVs promotes EMT in both PCa and normal prostate epithelial cells [61]. Cancer-associated fibroblasts induce EMT through the secretion of IL-6-containing sEVs, thereby promoting the invasive phenotype of BC [62]. KRT6B expression was found to be elevated in BC-derived sEVs compared to normal tissue-derived sEVs, which promotes EMT in BC, and its high expression results in shorter survival cycles that can be used to predict poor prognosis [63]. Therefore, the results of these studies reveal that EVs can greatly enhance the migration and invasion ability of tumor cells by promoting EMT in urinary tumors, which ultimately promotes the progression of cancer.

3.3. Involvement in the Occurrence of Pre-Metastatic Niches in Urological Tumors

Tumor-derived sEVs play a key role in promoting the formation of pre-metastatic niches [64]. Pre-metastatic niches are pro-oncogenic microenvironments that are created by the release of some molecules by the primary tumor including soluble factors, sEVs, and bone marrow-derived cells; these molecules function to regulate the microenvironment, thus making it easier for tumor cells to colonize and spread to distant organs [65,66]. Numerous studies have shown that tumor-derived sEVs, which can alter the function of target cells through the substances they carry, can migrate to target organs via vascular spillover. For example, colorectal cancer (CRC)-derived sEVs are enriched in integrin beta-like 1 (ITGBL1), and when released into circulation, activate fibroblasts to promote the formation of pre-metastatic niches [67,68]. Bone metastasis is an important cause of death in PCa. A study by Wang and coworkers found that the expression of microRNA-378a-3p was significantly elevated in the serum-derived sEVs of patients with bone metastatic PCa, and that miR-378a-3p-containing cells secreted by PC3 cells promoted osteolysis mechanisms via the Dyrk1a/Nfatc1 pathway to enhance bone metastasis in PCa [69]. In addition, it has been shown that tenascin-C is highly expressed in the lymph nodes of patients with BC metastases, and that BC-derived sEVs can induce tenascin-C expression to promote the formation of premetastatic niches [70]. The formation of a premetastatic niche provides many prerequisites for tumor metastasis.

3.4. sEVs Regulates the Tumor Microenvironment

The cellular and cell-free components of the tumor microenvironment (TME) can influence tumor development and response to therapy [71]. Key components of the TME include immune cells, stromal cells, blood vessels, and extracellular matrix. sEVs have the effect of promoting inflammatory factor production, tumor angiogenesis, and metastasis in theTME [72]. A study by Yin and coworkers found that colorectal cancer cell-derived sEVs could upregulate PD-L1 in macrophages to promote tumor immune evasion [73]. It has also been shown that sEV-loaded miRNAs can regulate the communication between cancer cells and hepatic stellate cells in the hepatocellular carcinoma tumor microenvironment [74]. Several studies have shown that sEVs can mediate communication between the tumor and the microenvironment via the Notch pathway, for example, in multiple myeloma [75]. Therefore, sEVs play an important role in TME.

3.5. Antitumor Effects of sEVs in Urological Tumors

Stem cell-derived sEVs have therapeutic anti-tumor effects. Human liver stem cell-derived sEVs loaded with antitumor miR-145 attenuate the invasive effects of renal stem cells [76]. Similarly, it has been shown that human hepatic stem cell-derived sEVs treated tumor endothelial cells to downregulate the expression of miR-15a, miR-181b, miR-320c, and miR-874, thereby inhibiting the angiogenic effects of tumor endothelial cells [77]. Thus, new prospects for the treatment of urological tumors could focus on stem cell-derived sEVs.

4. Application of Small Extracellular Vesicles in the Diagnosis of Urinary Tumors

Compared to traditional tumor markers, sEVs can cross the blood-brain barrier and enter the circulation. This particular feature renders sEVs easily detectable in patients’ biological fluids. sEVs carry an abundance of genetic material, are highly stable and non-degradable in bodily fluids, and have the advantages of having high specificity and readily undergoing extraction. These features have led to the significant adoption of sEV-based biomarkers in the clinical field. The first sEV-based RNA test for prostate cancer has been included in the National Comprehensive Cancer Network guidelines for the early detection of prostate cancer (Table 2) [7,78,79].

4.1. miRNAs in Small Extracellular Vesicles

MicroRNAs (miRNAs) are endogenous non-coding RNAs with an average length of 22 nt [107]. They can regulate changes in gene expression by targeting one or more mRNAs, thereby regulating cell growth, coordinating differentiation, and causing functional changes. In sEVs, the content of miRNAs is the highest among all types of RNA. Due to the lipid membrane that envelopes sEVs, miRNAs are protected from degradation by RNA enzymes. Therefore, the miRNA content in sEVs is much higher than that of free miRNA in cells and bodily fluids [108]. These characteristics make miRNAs in sEVs more suitable as biomarkers for tumors than those present in bodily fluids.
There have been numerous studies on sEVs that are obtainable from the urine, serum, and plasma as diagnostic markers of urological tumors. Despite breakthroughs in the advanced treatment of renal cell carcinoma (RCC), the mortality rate of kidney cancer remains high. Currently, there are no reliable biomarkers available for the early diagnosis and prognostic monitoring of RCC. One study that conducted a microarray analysis of sEVs extracted from the serum of patients with advanced RCC showed that miR-4525 contained by sEVs was significantly elevated compared to healthy controls. The investigators hypothesized that miR-4525 in serum-derived sEVs could serve as a potential biomarker for advanced RCC [80]. Meng and coworkers found that sEV extracted from the serum of RCC patients contained higher miR-155 compared to healthy people [81]. In addition to serum-derived sEVs, plasma-derived sEVs also comprise an important source for the diagnosis of RCC. Dias and coworkers found that the two plasma-containing sEV-derived miRNAs hsa-miR-301a-3p and hsa-miR-1293 were expressed at higher levels in patients with metastatic RCC compared to patients with non-metastatic RCC, leading them to propose that the levels of these molecules might serve as biomarkers for metastatic RCC [82]. Similarly, Xiao and coworkers sequenced plasma-derived sEVs from RCC patients and healthy individuals, from which they found that has-miR-92a-1-5p, has-miR-149-3p, and has-miR-424-3p were differentially expressed and of diagnostic value [83]. Urine-derived sEVs also have diagnostic value; Qin and coworkers identified the overexpression of miR-224-5p in urine-derived sEVs from RCC patients, which can be used as a biomarker for immunotherapy [84]. miR-204-5p and miR-30c-5p in urinary sEVs have great potential as biomarkers for the early diagnosis of RCC [85,86].
Prostate cancer (PCa) is the most common male malignancy other than lung cancer. Due to the lack of treatment modalities, advanced PCa is difficult to cure and has a low survival rate compared to early PCa, which has a higher cure rate using surgery combined with hormone therapy [109]. Therefore, the identification of early diagnostic markers for PCa is crucial. sEVs are increasingly being studied as diagnostic markers for tumors. Linuma and coworkers found that miR-93 in serum-derived sEVs was significantly lower in patients after radiotherapy and had a role in monitoring treatment efficacy [87]. Similarly, miR-181a-5p in serum-derived sEVs can be used as a marker of bone metastatic PCa [88]. Plasma-derived sEVs also contain miR-145, miR-221, miR-451a, and miR-141, which have diagnostic potential in PCa [89]. Among them, miR-221 has the ability to sort benign and malignant tumors [90]. For the differential diagnosis of PCa and benign prostatic hyperplasia (BPH), Davey and coworkers identified miR-375 and miR-574 in urine-derived sEVs, the combination of which yield the best diagnostic power out of all the contained miRNAs for the screening of benign and malignant tumors [91]. Similarly, Matsuzaki and coworkers found that miR-30b and miR-126 in urine-derived sEVs predicted PCa with much higher sensitivity and specificity than serum prostate-specific antigen (PSA) [92].
In the diagnosis of bladder cancer (BC), sEVs obtained from blood, urine, and tissue sources are also of diagnostic value. The differential expression of miR-185, miR-106a, and miR-10b in plasma-derived sEVs can predict BC survival [93]. miR-96-5p and miR-183-5p in urinary sEVs can be used in BC diagnosis and follow-up [94].

4.2. lncRNA in Small Extracellular Vesicles

Long-stranded non-coding RNAs (lncRNAs) are RNAs longer than 200 nucleotides that do not encode proteins and can be used as tumor markers. Patients with prostate cancer were found to have increased expression of the lncRNA PCA3 in sEVs obtained from their urine; therefore, PCA3 in urine-derived sEVs can be used as a marker for the early diagnosis of PCa [95]. Similarly, there was a significant difference found in the expression of lncRNA-p21 in urine-derived sEVs between BPH and PCa, suggesting that it can be used as a diagnostic marker to distinguish between benign and malignant tumors [96]. The lncRNA HOXD-AS1 in serum-derived sEVs can promote the distant metastasis of PCa, therefore, have predictive value in the metastasis of PCa [97]. The reduced expression of lncRNA PTENP1 in plasma-derived sEVs in patients with bladder cancer may serve as a potential marker for BC [98]. Similarly, the expression of the lncRNAs ANRIL in urine-derived sEVs is significantly elevated in BC patients, and therefore, can be used as a non-invasive diagnostic marker for BC [99].

4.3. CircRNAs in Small Extracellular Vesicles

Circular RNAs (circRNAs) are endogenous, non-coding RNAs that are highly conserved and stable. Currently, there have been few studies on circRNAs in sEVs. Xiao and coworkers showed that plasma-derived sEVs harboring circ_400068 were significantly more highly expressed in kidney cancer patients, suggesting that this RNA is associated with kidney cancer progression and has potential as a diagnostic marker [100]. Another study by Li and coworkers found that circ_0044516 expression was upregulated in the sEVs of prostate cancer patients, which could promote PCa proliferation and metastasis. They concluded that this circRNA has diagnostic value for PCa [101].

4.4. Proteins in Small Extracellular Vesicles

Tsuruda and coworkers detected the increased expression of RAB27Bin sEVs derived from RCC cells and found it to have a positive correlation with sunitinib resistance, suggesting that this protein could be used as a prognostic marker in RCC [102]. Iliuk and coworkers performed a proteomic analysis of plasma-derived sEVs obtained from RCC patients and found that the phosphorylated form of the protein LYRIC (MTDH) could potentially be used as a biomarker [103]. Polymerase I and transcript release factor (PTRF) expression was found to be higher in the urine-derived sEVs of RCC patients compared to normal human urine-derived sEVs, suggesting that PTRF could be used as a potential diagnostic marker in RCC [104]. Carbonic anhydrase 9 (CA IX) is highly expressed and active in the plasma-derived sEVs of PCa patients compared to normal subjects; therefore, CA IX in sEVs may be a biomarker of PCa progression [105]. It has been shown that heat shock protein 90 (Hsp 90) is significantly upregulated in the urine-derived sEVs of BCa patients and can be used as a diagnostic marker for BCa [106]. In addition, Igami and coworkers found elevated expression of carcinoembryonic antigen-associated adhesion molecule protein (CEACAM) in the urinary sEVs of BCa patients, and that these sEVs could be a new target for liquid biopsy testing [110].

5. Investigations of Small Extracellular Vesicles in the Treatment of Urological Tumors

5.1. Small Extracellular Vesicles and Tumor Drug Resistance

In clinical practice, one of the major challenges in tumor treatment is tumor drug resistance. Drug-resistant tumors can secrete numerous sEVs that contain resistance-associated proteins, and these sEVs can in turn promote the development of drug resistance. Tinibs (a chemotherapy drug for RCC) are the first-line drugs used for kidney cancer treatment. He and coworkers showed that many advanced RCCs are resistant to sorafenib (a chemotherapy drug for RCC), leading to poor disease treatment outcomes. They also found that sEVs could target MutL homolog 1 (MLH1) by delivering miR-31-5p, thereby leading to sorafenib resistance in RCC. Similarly, they detected the significant upregulation of miR-31-5p in the plasma-derived sEVs of drug-resistant RCC patients [111]. On the other hand, it has been suggested that ketoconazole can inhibit the formation of sEVs in kidney cancer cells as a way to suppress the proliferation and migration function of the tumor. The combination of sunitinib and ketoconazole may improve the therapeutic efficacy of sunitinib [112]. Research by Guang and coworkers showed that miR-423-5p contained in sEVs secreted by cancer-associated fibroblasts targeted GREM2 via the TGFβ pathway, thereby promoting PCa drug resistance [113]. Similarly, sEV-derived miR-27a produced by prostate fibroblasts improved PCa chemoresistance by suppressing P53 gene expression [114]. A study by Shan and coworkers found that cancer-associated fibroblasts secreting sEVs could directly transport miR-148b-3p into bladder cancer cells, thereby promoting BC metastasis, proliferation, and drug resistance [115]. The above studies jointly show that sEV-mediated tumor drug resistance is a phenomenon that can provide numerous new targets for the targeted therapy of urinary tumors, promote the personalized treatment of urinary tumors, and improve treatment efficiency.

5.2. Small Extracellular Vesicles as Drug Carriers

Most of the tumor treatment drugs used clinically have the disadvantage that only a small fraction of their dose can reach the lesion to achieve a therapeutic effect. This makes the drug less effective and may cause stronger toxicity and side effects [116]. sEVs can carry a variety of therapeutic substances and easily cross the blood-brain barrier. Numerous studies have shown that macrophage-derived hybrid sEVs can be used to target tumors by carrying relevant antitumor drugs such as Adriamycin [117]. Macrophage-derived sEVs can also serve as a drug delivery system for triple-negative breast cancer by carrying paclitaxel and Adriamycin [118]. Currently, there are no studies on the development of such drug delivery systems in RCC, which still needs to be explored in depth. In contrast, there have been more studies on sEV drug delivery systems in PCa. One study used genetic engineering techniques to design an anti-prostate specific membrane antigen (PMSA) sEV that could target late-stage PCa to organize cellular internalization [119]. Similarly, Wang and coworkers used genetic engineering techniques to reverse the tumor microenvironment of PCa by encapsulating the sonosensitizers Chlorin e6 and the immune adjuvant R848 into sEV [120]. Zhou and coworkers designed a drug delivery system for macrophage-derived sEVs harboring CD73 inhibitors and monoclonal antibodies target to programmed cell death ligand 1. The combination of this complex significantly inhibited the activation and infiltration of cytotoxic T lymphocytes in BC [121]. Pelvic radiotherapy is an important treatment modality for prostate cancer, where acute radiation cystitis is a common response to radiotherapy. It has been shown that mesenchymal stem cells (MSCs) can target fibrosis, inflammation, and angiogenesis in cystitis to achieve a therapeutic effect [122]. Zhao and coworkers induced that nano-sEVs released from MSCs standardized for pluripotent stem cells could be used to treat prostate cancer [123]. Today, in addition to the therapeutic benefits of stem cell and immune cell-derived sEVs, milk-derived sEVs also have corresponding therapeutic benefits. For example, milk-derived sEVs are stable, can be absorbed by the intestine, and remain intact and improve the intestinal barrier [124]. In addition, sEVs from sources such as goat and donkey milk also have anti-inflammatory and immunomodulatory abilities and can therefore be used extensively for the regulation of chronic diseases [125]. The above studies clearly indicate that sEVs have not yet been well studied in the treatment of urological tumors, but their unique physiological properties make them very promising for such research. On the other hand, food-derived sEVs can enhance cell targeting by mild modification, which is one of the prospects for oral treatment of tumors (Figure 2).

5.3. Small Extracellular Vesicles and Tumor Vaccines

Vaccines developed using cancer-associated cell-derived sEVs have higher affinities than conventional vaccines. Numerous studies have shown that dendritic cell (DC)-derived sEVs can be used as effective anti-tumor vaccines. sEVs have previously been designed to function as an in-situ DC-initiated vaccine to boost anti-tumor immunity in breast cancer [126]. Similarly, Lu and coworkers demonstrated that antigen-modified DC-derived sEVs could inhibit tumor regression in hepatocellular carcinoma [127]. In contrast, there have been fewer studies conducted on urological tumors. Xu and coworkers found that Reca cell-derived sEVs could stimulate CD8+ T cells to enhance the anti-renal cortical adenocarcinoma effect via the Fas ligand (FasL) signaling pathway [128]. In addition, oral particulate vaccines encapsulating tumor-associated antigens derived from mouse prostate cancer cell lines were combined with cyclophosphamide to significantly reduce the tumor volume of PCa [129]. PCa-derived sEVs modified with interferon-γ into a tumor vaccine were shown to increase the number of M1 macrophages and thus significantly inhibit tumor growth [130]. Currently, there is insufficient research on tumor vaccines, which comprise a highly promising new approach to tumor treatment. It is not difficult to hypothesize that sEVs loaded with tumor suppressor genes and tumor chemotherapy drugs will contribute greatly to the development of such tumor vaccines.

6. Conclusions

The early diagnosis of urological tumors is one of the key factors in improving patient survival and prognosis. sEVs are novel liquid biopsy markers for urological tumors. They are present in a variety of bodily fluids and tissues and are highly heterogeneous. Therefore, sEVs can potentially be used as biomarkers for non-invasive screening. Because of their phospholipid bilayer, which transports and protects various bioactive substances within, and ease in crossing the blood-brain barrier, sEVs have become a focus of research in the development, diagnosis, and treatment of diseases. Tumor cell-derived sEVs regulate angiogenesis, epithelial-mesenchymal transition, and the microenvironment of urological tumors by carrying substances such as nucleic acids and proteins. An increasing number of studies have focused on sEVs in the diagnosis of urological tumors. sEVs also contribute to EMT, angiogenesis, and drug resistance in urological tumors. Such studies related to sEVs provide new ideas for the diagnosis and treatment of urological tumors and hold great promise for further research. Research on sEV in the diagnosis of urological tumors has become more extensive and focused over time, but most of the current research remains only at the laboratory stage, rather than the clinical stage. This is mainly due to the lack of a definitive molecule that has been repeatedly confirmed to be useful across different studies, as well as the lack of clinical data. Some studies have focused on the therapeutic resistance of sEVs in urological tumors. However, there is a paucity of studies on the application of sEVs in the treatment of urological tumors, particularly in renal cancer, concurrent with a lack of data on translational investigations of their clinical application. Therefore, further studies are needed to investigate sEV-mediated tumor vaccines and tumor drug carriers, as well as to understand the impact and mechanisms of sEV-mediated tumor resistance on targeted therapy in urological tumors.

Author Contributions

M.Z. collected and analyzed the relevant literature, the writing of the first draft, future design, and comment revision and feedback. L.W. performed analyses and manuscript revision. Y.L. helped write the first draft, graph design, and date collection. Y.M. and X.H. revised the manuscript. Z.C. identified the topic, helped with diagramming, and made grammatical corrections to the article. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Natural Science Foundation of Jiangxi Province, grant number 20202BABL206119 and Health Commission of Jiangxi Province, grant number 202210860 and number 202310839.

Acknowledgments

The authors deeply appreciate the supports by all participants.

Conflicts of Interest

The authors state that there is no conflict of interest.

References

  1. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2019. CA Cancer J. Clin. 2019, 69, 7–34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Safiri, S.; Kolahi, A.A.; Naghavi, M.; Global Burden of Disease Bladder Cancer Collaborators. Global, regional and national burden of bladder cancer and its attributable risk factors in 204 countries and territories, 1990–2019: A systematic analysis for the Global Burden of Disease study 2019. BMJ Glob. Health 2021, 6, e004128. [Google Scholar] [CrossRef] [PubMed]
  3. Teo, M.Y.; Rathkopf, D.E.; Kantoff, P. Treatment of Advanced Prostate Cancer. Annu. Rev. Med. 2019, 70, 479–499. [Google Scholar] [CrossRef]
  4. Gray, R.E.; Harris, G.T. Renal Cell Carcinoma: Diagnosis and Management. Am. Fam. Phys. 2019, 99, 179–184. [Google Scholar]
  5. Zebrowska, A.; Widlak, P.; Whiteside, T.; Pietrowska, M. Signaling of Tumor-Derived sEV Impacts Melanoma Progression. Int. J. Mol. Sci. 2020, 21, 5066. [Google Scholar] [CrossRef] [PubMed]
  6. Tan, T.T.; Lai, R.C.; Padmanabhan, J.; Sim, W.K.; Choo, A.B.H.; Lim, S.K. Assessment of Tumorigenic Potential in Mesenchymal-Stem/Stromal-Cell-Derived Small Extracellular Vesicles (MSC-sEV). Pharmaceutics 2021, 14, 345. [Google Scholar] [CrossRef] [PubMed]
  7. Yu, W.; Hurley, J.; Roberts, D.; Chakrabortty, S.K.; Enderle, D.; Noerholm, M.; Breakefield, X.O.; Skog, J.K. Exosome-based liquid biopsies in cancer: Opportunities and challenges. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2021, 32, 466–477. [Google Scholar] [CrossRef]
  8. Takenaka, M.; Yabuta, A.; Takahashi, Y.; Takakura, Y. Interleukin-4-carrying small extracellular vesicles with a high potential as anti-inflammatory therapeutics based on modulation of macrophage function. Biomaterials 2021, 278, 121160. [Google Scholar] [CrossRef]
  9. Takenaka, M.; Takahashi, Y.; Takakura, Y. Intercellular delivery of NF-κB inhibitor peptide utilizing small extracellular vesicles for the application of anti-inflammatory therapy. J. Control. Release Off. J. Control. Release Soc. 2020, 328, 435–443. [Google Scholar] [CrossRef]
  10. Urabe, F.; Kosaka, N.; Ito, K.; Kimura, T.; Egawa, S.; Ochiya, T. Extracellular vesicles as biomarkers and therapeutic targets for cancer. Am. J. Physiol. Cell Physiol. 2020, 318, C29–C39. [Google Scholar] [CrossRef]
  11. Zhang, Y.; Bi, J.; Huang, J.; Tang, Y.; Du, S.; Li, P. Exosome: A Review of Its Classification, Isolation Techniques, Storage, Diagnostic and Targeted Therapy Applications. Int. J. Nanomed. 2020, 15, 6917–6934. [Google Scholar] [CrossRef] [PubMed]
  12. Doyle, L.M.; Wang, M.Z. Overview of Extracellular Vesicles, Their Origin, Composition, Purpose, and Methods for Exosome Isolation and Analysis. Cells 2019, 8, 727. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Huang, D.; Chen, J.; Hu, D.; Xie, F.; Yang, T.; Li, Z.; Wang, X.; Xiao, Y.; Zhong, J.; Jiang, Y.; et al. Advances in Biological Function and Clinical Application of Small Extracellular Vesicle Membrane Proteins. Front. Oncol. 2021, 11, 675940. [Google Scholar] [CrossRef] [PubMed]
  15. Lu, Y.; Liu, D.; Feng, Q.; Liu, Z. Diabetic Nephropathy: Perspective on Extracellular Vesicles. Front. Immunol. 2020, 11, 943. [Google Scholar] [CrossRef]
  16. Javeed, N. Shedding Perspective on Extracellular Vesicle Biology in Diabetes and Associated Metabolic Syndromes. Endocrinology 2019, 160, 399–408. [Google Scholar] [CrossRef] [Green Version]
  17. Kalluri, R.; LeBleu, V.S. The biology, function, and biomedical applications of exosomes. Science 2020, 367, eaau6977. [Google Scholar] [CrossRef]
  18. Alberro, A.; Iparraguirre, L.; Fernandes, A.; Otaegui, D. Extracellular Vesicles in Blood: Sources, Effects, and Applications. Int. J. Mol. Sci. 2021, 22, 8163. [Google Scholar] [CrossRef]
  19. Caruso Bavisotto, C.; Marino Gammazza, A.; Campanella, C.; Bucchieri, F.; Cappello, F. Extracellular heat shock proteins in cancer: From early diagnosis to new therapeutic approach. Semin. Cancer Biol. 2021, 86, 36–45. [Google Scholar] [CrossRef]
  20. Mathieu, M.; Névo, N.; Jouve, M.; Valenzuela, J.I.; Maurin, M.; Verweij, F.J.; Palmulli, R.; Lankar, D.; Dingli, F.; Loew, D.; et al. Specificities of exosome versus small ectosome secretion revealed by live intracellular tracking of CD63 and CD9. Nat. Commun. 2021, 12, 4389. [Google Scholar] [CrossRef]
  21. Pietrowska, M.; Zebrowska, A.; Gawin, M.; Marczak, L.; Sharma, P.; Mondal, S.; Mika, J.; Polańska, J.; Ferrone, S.; Kirkwood, J.M.; et al. Proteomic profile of melanoma cell-derived small extracellular vesicles in patients’ plasma: A potential correlate of melanoma progression. J. Extracell. Vesicles 2021, 10, e12063. [Google Scholar] [CrossRef] [PubMed]
  22. Dasgupta, D.; Nakao, Y.; Mauer, A.S.; Thompson, J.M.; Sehrawat, T.S.; Liao, C.Y.; Krishnan, A.; Lucien, F.; Guo, Q.; Liu, M.; et al. IRE1A Stimulates Hepatocyte-Derived Extracellular Vesicles That Promote Inflammation in Mice with Steatohepatitis. Gastroenterology 2020, 159, 1487–1503. [Google Scholar] [CrossRef] [PubMed]
  23. McVey, M.J.; Weidenfeld, S.; Maishan, M.; Spring, C.; Kim, M.; Tabuchi, A.; Srbely, V.; Takabe-French, A.; Simmons, S.; Arenz, C.; et al. Platelet extracellular vesicles mediate transfusion-related acute lung injury by imbalancing the sphingolipid rheostat. Blood 2021, 137, 690–701. [Google Scholar] [CrossRef]
  24. Elzanowska, J.; Semira, C.; Costa-Silva, B. DNA in extracellular vesicles: Biological and clinical aspects. Mol. Oncol. 2021, 15, 1701–1714. [Google Scholar] [CrossRef]
  25. Li, Y.; Zhao, J.; Yu, S.; Wang, Z.; He, X.; Su, Y.; Guo, T.; Sheng, H.; Chen, J.; Zheng, Q.; et al. Extracellular Vesicles Long RNA Sequencing Reveals Abundant mRNA, circRNA, and lncRNA in Human Blood as Potential Biomarkers for Cancer Diagnosis. Clin. Chem. 2019, 65, 798–808. [Google Scholar] [CrossRef] [PubMed]
  26. Wei, D.; Zhan, W.; Gao, Y.; Huang, L.; Gong, R.; Wang, W.; Zhang, R.; Wu, Y.; Gao, S.; Kang, T. RAB31 marks and controls an ESCRT-independent exosome pathway. Cell Res. 2021, 31, 157–177. [Google Scholar] [CrossRef] [PubMed]
  27. Zhang, M.; Wang, L.; Chen, Z. Research progress of extracellular vesicles in type 2 diabetes and its complications. Diabet. Med. J. Br. Diabet. Assoc. 2022, 39, e14865. [Google Scholar] [CrossRef]
  28. Lu, Y.; Wang, L.; Zhang, M.; Chen, Z. Mesenchymal Stem Cell-Derived Small Extracellular Vesicles: A Novel Approach for Kidney Disease Treatment. Int. J. Nanomed. 2022, 17, 3603–3618. [Google Scholar] [CrossRef]
  29. Wu, Z.; Zhang, Z.; Xia, W.; Cai, J.; Li, Y.; Wu, S. Extracellular vesicles in urologic malignancies-Implementations for future cancer care. Cell Prolif. 2019, 52, e12659. [Google Scholar] [CrossRef] [Green Version]
  30. Vitorino, R.; Ferreira, R.; Guedes, S.; Amado, F.; Thongboonkerd, V. What can urinary exosomes tell us? Cell. Mol. Life Sci. 2021, 78, 3265–3283. [Google Scholar] [CrossRef]
  31. Burkova, E.E.; Sedykh, S.E.; Nevinsky, G.A. Human Placenta Exosomes: Biogenesis, Isolation, Composition, and Prospects for Use in Diagnostics. Int. J. Mol. Sci. 2021, 22, 2158. [Google Scholar] [CrossRef] [PubMed]
  32. Karimi, N.; Cvjetkovic, A.; Jang, S.C.; Crescitelli, R.; Hosseinpour Feizi, M.A.; Nieuwland, R.; Lötvall, J.; Lässer, C. Detailed analysis of the plasma extracellular vesicle proteome after separation from lipoproteins. Cell. Mol. Life Sci. CMLS 2018, 75, 2873–2886. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Ha, D.H.; Kim, H.K.; Lee, J.; Kwon, H.H.; Park, G.H.; Yang, S.H.; Jung, J.Y.; Choi, H.; Lee, J.H.; Sung, S.; et al. Mesenchymal Stem/Stromal Cell-Derived Exosomes for Immunomodulatory Therapeutics and Skin Regeneration. Cells 2020, 9, 1157. [Google Scholar] [CrossRef]
  34. Lazar, S.; Goldfinger, L.E. Platelets and extracellular vesicles and their cross talk with cancer. Blood 2021, 137, 3192–3200. [Google Scholar] [CrossRef]
  35. Zhao, M.; Liu, S.; Wang, C.; Wang, Y.; Wan, M.; Liu, F.; Gong, M.; Yuan, Y.; Chen, Y.; Cheng, J.; et al. Mesenchymal Stem Cell-Derived Extracellular Vesicles Attenuate Mitochondrial Damage and Inflammation by Stabilizing Mitochondrial DNA. ACS Nano 2021, 15, 1519–1538. [Google Scholar] [CrossRef] [PubMed]
  36. Li, Q.; Xu, Y.; Lv, K.; Wang, Y.; Zhong, Z.; Xiao, C.; Zhu, K.; Ni, C.; Wang, K.; Kong, M.; et al. Small extracellular vesicles containing miR-486-5p promote angiogenesis after myocardial infarction in mice and nonhuman primates. Sci. Transl. Med. 2021, 13, eabb0202. [Google Scholar] [CrossRef] [PubMed]
  37. Yang, C.; Dou, R.; Wei, C.; Liu, K.; Shi, D.; Zhang, C.; Liu, Q.; Wang, S.; Xiong, B. Tumor-derived exosomal microRNA-106b-5p activates EMT-cancer cell and M2-subtype TAM interaction to facilitate CRC metastasis. Mol. Ther. J. Am. Soc. Gene Ther. 2021, 29, 2088–2107. [Google Scholar] [CrossRef]
  38. Ding, J.; Zhang, Y.; Cai, X.; Zhang, Y.; Yan, S.; Wang, J.; Zhang, S.; Yin, T.; Yang, C.; Yang, J. Extracellular vesicles derived from M1 macrophages deliver miR-146a-5p and miR-146b-5p to suppress trophoblast migration and invasion by targeting TRAF6 in recurrent spontaneous abortion. Theranostics 2021, 11, 5813–5830. [Google Scholar] [CrossRef]
  39. Hu, J.L.; Wang, W.; Lan, X.L.; Zeng, Z.C.; Liang, Y.S.; Yan, Y.R.; Song, F.Y.; Zhu, X.H.; Liao, W.J.; Liao, W.T.; et al. CAFs secreted exosomes promote metastasis and chemotherapy resistance by enhancing cell stemness and epithelial-mesenchymal transition in colorectal cancer. Mol. Cancer 2019, 18, 91. [Google Scholar] [CrossRef] [Green Version]
  40. Huang, M.; Lei, Y.; Zhong, Y.; Chung, C.; Wang, M.; Hu, M.; Deng, L. New Insights Into the Regulatory Roles of Extracellular Vesicles in Tumor Angiogenesis and Their Clinical Implications. Front. Cell Dev. Biol. 2021, 9, 791882. [Google Scholar] [CrossRef]
  41. Ren, W.; Hou, J.; Yang, C.; Wang, H.; Wu, S.; Wu, Y.; Zhao, X.; Lu, C. Extracellular vesicles secreted by hypoxia pre-challenged mesenchymal stem cells promote non-small cell lung cancer cell growth and mobility as well as macrophage M2 polarization via miR-21-5p delivery. J. Exp. Clin. Cancer Res. 2019, 38, 62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Guarino, B.; Katari, V.; Adapala, R.; Bhavnani, N.; Dougherty, J.; Khan, M.; Paruchuri, S.; Thodeti, C. Tumor-Derived Extracellular Vesicles Induce Abnormal Angiogenesis via TRPV4 Downregulation and Subsequent Activation of YAP and VEGFR2. Front. Bioeng. Biotechnol. 2021, 9, 790489. [Google Scholar] [CrossRef] [PubMed]
  43. Gangadaran, P.; Rajendran, R.L.; Lee, H.W.; Kalimuthu, S.; Hong, C.M.; Jeong, S.Y.; Lee, S.W.; Lee, J.; Ahn, B.-C. Extracellular vesicles from mesenchymal stem cells activates VEGF receptors and accelerates recovery of hindlimb ischemia. J. Control. Release Off. J. Control. Release Soc. 2017, 264, 112–126. [Google Scholar] [CrossRef] [PubMed]
  44. Ge, L.; Xun, C.; Li, W.; Jin, S.; Liu, Z.; Zhuo, Y.; Duan, D.; Hu, Z.; Chen, P.; Lu, M. Extracellular vesicles derived from hypoxia-preconditioned olfactory mucosa mesenchymal stem cells enhance angiogenesis via miR-612. J. Nanobiotechnol. 2021, 19, 380. [Google Scholar] [CrossRef] [PubMed]
  45. Zhang, H.; Wu, J.; Wu, J.; Fan, Q.; Zhou, J.; Wu, J.; Liu, S.; Zang, J.; Ye, J.; Xiao, M.; et al. Exosome-mediated targeted delivery of miR-210 for angiogenic therapy after cerebral ischemia in mice. J. Nanobiotechnol. 2019, 17, 29. [Google Scholar] [CrossRef] [Green Version]
  46. Zhao, P.; Cheng, J.; Li, B.; Nie, D.; Li, C.; Gui, S.; Wang, H.; Zhang, Y. Up-regulation of the expressions of MiR-149-5p and MiR-99a-3p in exosome inhibits the progress of pituitary adenomas. Cell Biol. Toxicol. 2021, 37, 633–651. [Google Scholar] [CrossRef] [PubMed]
  47. Zhou, X.; Yan, T.; Huang, C.; Xu, Z.; Wang, L.; Jiang, E.; Wang, H.; Chen, Y.; Liu, K.; Shao, Z.; et al. Melanoma cell-secreted exosomal miR-155-5p induce proangiogenic switch of cancer-associated fibroblasts via SOCS1/JAK2/STAT3 signaling pathway. J. Exp. Clin. Cancer Res. 2018, 37, 242. [Google Scholar] [CrossRef] [Green Version]
  48. Ma, Z.; Wei, K.; Yang, F.; Guo, Z.; Pan, C.; He, Y.; Wang, J.; Li, Z.; Chen, L.; Chen, Y.; et al. Tumor-derived exosomal miR-3157-3p promotes angiogenesis, vascular permeability and metastasis by targeting TIMP/KLF2 in non-small cell lung cancer. Cell Death Dis. 2021, 12, 840. [Google Scholar] [CrossRef]
  49. Zhang, W.; Zheng, X.; Yu, Y.; Zheng, L.; Lan, J.; Wu, Y.; Liu, H.; Zhao, A.; Huang, H.; Chen, W. Renal cell carcinoma-derived exosomes deliver lncARSR to induce macrophage polarization and promote tumor progression via STAT3 pathway. Int. J. Biol. Sci. 2022, 18, 3209–3222. [Google Scholar] [CrossRef]
  50. Krishn, S.R.; Salem, I.; Quaglia, F.; Naranjo, N.M.; Agarwal, E.; Liu, Q.; Sarker, S.; Kopenhaver, J.; McCue, P.A.; Weinreb, P.H.; et al. The αvβ6 integrin in cancer cell-derived small extracellular vesicles enhances angiogenesis. J. Extracell. Vesicles 2020, 9, 1763594. [Google Scholar] [CrossRef]
  51. Prigol, A.N.; Rode, M.P.; Silva, A.H.; Cisilotto, J.; Creczynski-Pasa, T.B. Pro-angiogenic effect of PC-3 exosomes in endothelial cells in vitro. Cell. Signal. 2021, 87, 110126. [Google Scholar] [CrossRef] [PubMed]
  52. Li, X.; Wei, Z.; Yu, H.; Xu, Y.; He, W.; Zhou, X.; Gou, X. Secretory autophagy-induced bladder tumour-derived extracellular vesicle secretion promotes angiogenesis by activating the TPX2-mediated phosphorylation of the AURKA-PI3K-AKT axis. Cancer Lett. 2021, 523, 10–28. [Google Scholar] [CrossRef] [PubMed]
  53. Li, X.; Peng, X.; Zhang, C.; Bai, X.; Li, Y.; Chen, G.; Guo, H.; He, W.; Zhou, X.; Gou, X. Bladder Cancer-Derived Small Extracellular Vesicles Promote Tumor Angiogenesis by Inducing HBP-Related Metabolic Reprogramming and SerRS O-GlcNAcylation in Endothelial Cells. Adv. Sci. Weinh. Baden-Wurtt. Ger. 2022, 9, e2202993. [Google Scholar] [CrossRef]
  54. Zhang, X.; Sai, B.; Wang, F.; Wang, L.; Wang, Y.; Zheng, L.; Li, G.; Tang, J.; Xiang, J. Hypoxic BMSC-derived exosomal miRNAs promote metastasis of lung cancer cells via STAT3-induced EMT. Mol. Cancer 2019, 18, 40. [Google Scholar] [CrossRef] [Green Version]
  55. Qin, Y.; Zhang, J.; Avellán-Llaguno, R.D.; Zhang, X.; Huang, Q. DEHP-elicited small extracellular vesicles miR-26a-5p promoted metastasis in nearby normal A549 cells. Environ. Pollut. 2021, 272, 116005. [Google Scholar] [CrossRef] [PubMed]
  56. Wang, D.; Wang, X.; Si, M.; Yang, J.; Sun, S.; Wu, H.; Cui, S.; Qu, X.; Yu, X. Exosome-encapsulated miRNAs contribute to CXCL12/CXCR4-induced liver metastasis of colorectal cancer by enhancing M2 polarization of macrophages. Cancer Lett. 2020, 474, 36–52. [Google Scholar] [CrossRef]
  57. Cui, Y.; Wang, D.; Xie, M. Tumor-Derived Extracellular Vesicles Promote Activation of Carcinoma-Associated Fibroblasts and Facilitate Invasion and Metastasis of Ovarian Cancer by Carrying miR-630. Front. Cell Dev. Biol. 2021, 9, 652322. [Google Scholar] [CrossRef]
  58. Leal-Orta, E.; Ramirez-Ricardo, J.; Garcia-Hernandez, A.; Cortes-Reynosa, P.; Salazar, E.P. Extracellular vesicles from MDA-MB-231 breast cancer cells stimulated with insulin-like growth factor 1 mediate an epithelial-mesenchymal transition process in MCF10A mammary epithelial cells. J. Cell Commun. Signal. 2021, 16, 531–546. [Google Scholar] [CrossRef]
  59. Wang, T.; Wang, X.; Wang, H.; Li, L.; Zhang, C.; Xiang, R.; Tan, X.; Li, Z.; Jiang, C.; Zheng, L.; et al. High TSPAN8 expression in epithelial cancer cell-derived small extracellular vesicles promote confined diffusion and pronounced uptake. J. Extracell. Vesicles 2021, 10, e12167. [Google Scholar] [CrossRef]
  60. Wang, L.; Yang, G.; Zhao, D.; Wang, J.; Bai, Y.; Peng, Q.; Wang, H.; Fang, R.; Chen, G.; Wang, Z.; et al. CD103-positive CSC exosome promotes EMT of clear cell renal cell carcinoma: Role of remote MiR-19b-3p. Mol. Cancer 2019, 18, 86. [Google Scholar] [CrossRef] [Green Version]
  61. Li, Y.; Li, Q.; Li, D.; Gu, J.; Qian, D.; Qin, X.; Chen, Y. Exosome carrying PSGR promotes stemness and epithelial-mesenchymal transition of low aggressive prostate cancer cells. Life Sci. 2021, 264, 118638. [Google Scholar] [CrossRef] [PubMed]
  62. Goulet, C.R.; Champagne, A.; Bernard, G.; Vandal, D.; Chabaud, S.; Pouliot, F.; Bolduc, S. Cancer-associated fibroblasts induce epithelial-mesenchymal transition of bladder cancer cells through paracrine IL-6 signalling. BMC Cancer 2019, 19, 137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Song, Q.; Yu, H.; Cheng, Y.; Han, J.; Li, K.; Zhuang, J.; Lv, Q.; Yang, X.; Yang, H. Bladder cancer-derived exosomal KRT6B promotes invasion and metastasis by inducing EMT and regulating the immune microenvironment. J. Transl. Med. 2022, 20, 308. [Google Scholar] [CrossRef]
  64. Zhang, C.; Wang, X.Y.; Zhang, P.; He, T.C.; Han, J.H.; Zhang, R.; Lin, J.; Fan, J.; Lu, L.; Zhu, W.W.; et al. Cancer-derived exosomal HSPC111 promotes colorectal cancer liver metastasis by reprogramming lipid metabolism in cancer-associated fibroblasts. Cell Death Dis. 2022, 13, 57. [Google Scholar] [CrossRef] [PubMed]
  65. Wortzel, I.; Dror, S.; Kenific, C.M.; Lyden, D. Exosome-Mediated Metastasis: Communication from a Distance. Dev. Cell 2019, 49, 347–360. [Google Scholar] [CrossRef] [PubMed]
  66. Gong, Z.; Li, Q.; Shi, J.; Wei, J.; Li, P.; Chang, C.H.; Shultz, L.D.; Ren, G. Lung fibroblasts facilitate pre-metastatic niche formation by remodeling the local immune microenvironment. Immunity 2022, 55, 1483–1500.e1489. [Google Scholar] [CrossRef]
  67. Ji, Q.; Zhou, L.; Sui, H.; Yang, L.; Wu, X.; Song, Q.; Jia, R.; Li, R.; Sun, J.; Wang, Z.; et al. Primary tumors release ITGBL1-rich extracellular vesicles to promote distal metastatic tumor growth through fibroblast-niche formation. Nat. Commun. 2020, 11, 1211. [Google Scholar] [CrossRef] [Green Version]
  68. Yuan, X.; Qian, N.; Ling, S.; Li, Y.; Sun, W.; Li, J.; Du, R.; Zhong, G.; Liu, C.; Yu, G.; et al. Breast cancer exosomes contribute to pre-metastatic niche formation and promote bone metastasis of tumor cells. Theranostics 2021, 11, 1429–1445. [Google Scholar] [CrossRef]
  69. Wang, J.; Du, X.; Wang, X.; Xiao, H.; Jing, N.; Xue, W.; Dong, B.; Gao, W.Q.; Fang, Y.-X. Tumor-derived miR-378a-3p-containing extracellular vesicles promote osteolysis by activating the Dyrk1a/Nfatc1/Angptl2 axis for bone metastasis. Cancer Lett. 2022, 526, 76–90. [Google Scholar] [CrossRef]
  70. Silvers, C.R.; Messing, E.M.; Miyamoto, H.; Lee, Y.-F. Tenascin-C expression in the lymph node pre-metastatic niche in muscle-invasive bladder cancer. Br. J. Cancer 2021, 125, 1399–1407. [Google Scholar] [CrossRef]
  71. Jin, M.Z.; Jin, W.-L. The updated landscape of tumor microenvironment and drug repurposing. Signal Transduct. Target. Ther. 2020, 5, 166. [Google Scholar] [CrossRef] [PubMed]
  72. Anderson, N.M.; Simon, M.C. The tumor microenvironment. Curr. Biol. CB 2020, 30, R921–R925. [Google Scholar] [CrossRef] [PubMed]
  73. Yin, Y.; Liu, B.; Cao, Y.; Yao, S.; Liu, Y.; Jin, G.; Qin, Y.; Chen, Y.; Cui, K.; Zhou, L.; et al. Colorectal Cancer-Derived Small Extracellular Vesicles Promote Tumor Immune Evasion by Upregulating PD-L1 Expression in Tumor-Associated Macrophages. Adv. Sci. Weinh. Baden-Wurtt. Ger. 2022, 9, 2102620. [Google Scholar] [CrossRef] [PubMed]
  74. Li, J.; Yan, Y.; Ang, L.; Li, X.; Liu, C.; Sun, B.; Lin, X.; Peng, Z.; Zhang, X.; Zhang, Q.; et al. Extracellular vesicles-derived OncomiRs mediate communication between cancer cells and cancer-associated hepatic stellate cells in hepatocellular carcinoma microenvironment. Carcinogenesis 2020, 41, 223–234. [Google Scholar] [CrossRef] [PubMed]
  75. Giannandrea, D.; Platonova, N.; Colombo, M.; Mazzola, M.; Citro, V.; Adami, R.; Maltoni, F.; Ancona, S.; Dolo, V.; Giusti, I.; et al. Extracellular vesicles mediate the communication between multiple myeloma and bone marrow microenvironment in a NOTCH dependent way. Haematologica 2022, 107, 2183–2194. [Google Scholar] [CrossRef] [PubMed]
  76. Brossa, A.; Tapparo, M.; Fonsato, V.; Papadimitriou, E.; Delena, M.; Camussi, G.; Bussolati, B. Coincubation as miR-Loading Strategy to Improve the Anti-Tumor Effect of Stem Cell-Derived EVs. Pharmaceutics 2021, 13, 76. [Google Scholar] [CrossRef] [PubMed]
  77. Lopatina, T.; Grange, C.; Fonsato, V.; Tapparo, M.; Brossa, A.; Fallo, S.; Pitino, A.; Herrera-Sanchez, M.B.; Kholia, S.; Camussi, G.; et al. Extracellular vesicles from human liver stem cells inhibit tumor angiogenesis. Int. J. Cancer 2019, 144, 322–333. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Gagliardi, D.; Bresolin, N.; Comi, G.P.; Corti, S. Extracellular vesicles and amyotrophic lateral sclerosis: From misfolded protein vehicles to promising clinical biomarkers. Cell. Mol. Life Sci. 2021, 78, 561–572. [Google Scholar] [CrossRef]
  79. Li, W.; Liu, J.B.; Hou, L.K.; Yu, F.; Zhang, J.; Wu, W.; Tang, X.M.; Sun, F.; Lu, H.M.; Deng, J.; et al. Liquid biopsy in lung cancer: Significance in diagnostics, prediction, and treatment monitoring. Mol. Cancer 2022, 21, 25. [Google Scholar] [CrossRef]
  80. Muramatsu-Maekawa, Y.; Kawakami, K.; Fujita, Y.; Takai, M.; Kato, D.; Nakane, K.; Kato, T.; Tsuchiya, T.; Koie, T.; Miura, Y.; et al. Profiling of Serum Extracellular Vesicles Reveals miRNA-4525 as a Potential Biomarker for Advanced Renal Cell Carcinoma. Cancer Genom. Proteom. 2021, 18, 253–259. [Google Scholar] [CrossRef]
  81. Meng, L.; Xing, Z.; Guo, Z.; Qiu, Y.; Liu, Z. Hypoxia-induced microRNA-155 overexpression in extracellular vesicles promotes renal cell carcinoma progression by targeting FOXO3. Aging 2021, 13, 9613–9626. [Google Scholar] [CrossRef] [PubMed]
  82. Dias, F.; Teixeira, A.L.; Nogueira, I.; Morais, M.; Maia, J.; Bodo, C.; Ferreira, M.; Silva, A.; Vilhena, M.; Lobo, J.; et al. Extracellular Vesicles Enriched in hsa-miR-301a-3p and hsa-miR-1293 Dynamics in Clear Cell Renal Cell Carcinoma Patients: Potential Biomarkers of Metastatic Disease. Cancers 2020, 12, 1450. [Google Scholar] [CrossRef] [PubMed]
  83. Xiao, C.T.; Lai, W.J.; Zhu, W.A.; Wang, H. MicroRNA Derived from Circulating Exosomes as Noninvasive Biomarkers for Diagnosing Renal Cell Carcinoma. OncoTargets Ther. 2020, 13, 10765–10774. [Google Scholar] [CrossRef] [PubMed]
  84. Qin, Z.; Hu, H.; Sun, W.; Chen, L.; Jin, S.; Xu, Q.; Liu, Y.; Yu, L.; Zeng, S. miR-224-5p Contained in Urinary Extracellular Vesicles Regulates PD-L1 Expression by Inhibiting Cyclin D1 in Renal Cell Carcinoma Cells. Cancers 2021, 13, 618. [Google Scholar] [CrossRef] [PubMed]
  85. Kurahashi, R.; Kadomatsu, T.; Baba, M.; Hara, C.; Itoh, H.; Miyata, K.; Endo, M.; Morinaga, J.; Terada, K.; Araki, K.; et al. MicroRNA-204-5p: A novel candidate urinary biomarker of Xp11.2 translocation renal cell carcinoma. Cancer Sci. 2019, 110, 1897–1908. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Song, S.; Long, M.; Yu, G.; Cheng, Y.; Yang, Q.; Liu, J.; Wang, Y.; Sheng, J.; Wang, L.; Wang, Z.; et al. Urinary exosome miR-30c-5p as a biomarker of clear cell renal cell carcinoma that inhibits progression by targeting HSPA5. J. Cell. Mol. Med. 2019, 23, 6755–6765. [Google Scholar] [CrossRef] [Green Version]
  87. Iinuma, K.; Kawakami, K.; Mizutani, K.; Fujita, Y.; Yamaguchi, T.; Ito, M.; Kumano, T.; Matsuo, M.; Nakano, M.; Koie, T.; et al. miRNA-93 in Serum Extracellular Vesicles Before and After Low Dose Rate Prostate Brachytherapy. Anticancer Res. 2021, 41, 2411–2418. [Google Scholar] [CrossRef]
  88. Wang, Y.; Fang, Y.X.; Dong, B.; Du, X.; Wang, J.; Wang, X.; Gao, W.Q.; Xue, W. Discovery of extracellular vesicles derived miR-181a-5p in patient’s serum as an indicator for bone-metastatic prostate cancer. Theranostics 2021, 11, 878–892. [Google Scholar] [CrossRef]
  89. Zabegina, L.; Nazarova, I.; Nikiforova, N.; Slyusarenko, M.; Sidina, E.; Knyazeva, M.; Tsyrlina, E.; Novikov, S.; Reva, S.; Malek, A. A New Approach for Prostate Cancer Diagnosis by miRNA Profiling of Prostate-Derived Plasma Small Extracellular Vesicles. Cells 2021, 10, 2372. [Google Scholar] [CrossRef]
  90. Kim, J.; Cho, S.; Park, Y.; Lee, J.; Park, J. Evaluation of micro-RNA in extracellular vesicles from blood of patients with prostate cancer. PLoS ONE 2021, 16, e0262017. [Google Scholar] [CrossRef]
  91. Davey, M.; Benzina, S.; Savoie, M.; Breault, G.; Ghosh, A.; Ouellette, R.J. Affinity Captured Urinary Extracellular Vesicles Provide mRNA and miRNA Biomarkers for Improved Accuracy of Prostate Cancer Detection: A Pilot Study. Int. J. Mol. Sci. 2020, 21, 8330. [Google Scholar] [CrossRef] [PubMed]
  92. Matsuzaki, K.; Fujita, K.; Tomiyama, E.; Hatano, K.; Hayashi, Y.; Wang, C.; Ishizuya, Y.; Yamamoto, Y.; Hayashi, T.; Kato, T.; et al. MiR-30b-3p and miR-126-3p of urinary extracellular vesicles could be new biomarkers for prostate cancer. Transl. Androl. Urol. 2021, 10, 1918–1927. [Google Scholar] [CrossRef]
  93. Sabo, A.A.; Birolo, G.; Naccarati, A.; Dragomir, M.P.; Aneli, S.; Allione, A.; Oderda, M.; Allasia, M.; Gontero, P.; Sacerdote, C.; et al. Small Non-Coding RNA Profiling in Plasma Extracellular Vesicles of Bladder Cancer Patients by Next-Generation Sequencing: Expression Levels of miR-126-3p and piR-5936 Increase with Higher Histologic Grades. Cancers 2020, 12, 1507. [Google Scholar] [CrossRef] [PubMed]
  94. El-Shal, A.S.; Shalaby, S.M.; Abouhashem, S.E.; Elbary, E.H.A.; Azazy, S.; Rashad, N.M.; Sarhan, W. Urinary exosomal microRNA-96-5p and microRNA-183-5p expression as potential biomarkers of bladder cancer. Mol. Biol. Rep. 2021, 48, 4361–4371. [Google Scholar] [CrossRef] [PubMed]
  95. Li, Y.; Ji, J.; Lyu, J.; Jin, X.; He, X.; Mo, S.; Xu, H.; He, J.; Cao, Z.; Chen, X.; et al. A Novel Urine Exosomal lncRNA Assay to Improve the Detection of Prostate Cancer at Initial Biopsy: A Retrospective Multicenter Diagnostic Feasibility Study. Cancers 2021, 13, 4075. [Google Scholar] [CrossRef] [PubMed]
  96. Işın, M.; Uysaler, E.; Özgür, E.; Köseoğlu, H.; Şanlı, Ö.; Yücel, Ö.B.; Gezer, U.; Dalay, N. Exosomal lncRNA-p21 levels may help to distinguish prostate cancer from benign disease. Front. Genet. 2015, 6, 168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Jiang, Y.; Zhao, H.; Chen, Y.; Li, K.; Li, T.; Chen, J.; Zhang, B.; Guo, C.; Qing, L.; Shen, J.; et al. Exosomal long noncoding RNA HOXD-AS1 promotes prostate cancer metastasis via miR-361-5p/FOXM1 axis. Cell Death Dis. 2021, 12, 1129. [Google Scholar] [CrossRef]
  98. Zheng, R.; Du, M.; Wang, X.; Xu, W.; Liang, J.; Wang, W.; Lv, Q.; Qin, C.; Chu, H.; Wang, M.; et al. Exosome-transmitted long non-coding RNA PTENP1 suppresses bladder cancer progression. Mol. Cancer 2018, 17, 143. [Google Scholar] [CrossRef]
  99. Abbastabar, M.; Sarfi, M.; Golestani, A.; Karimi, A.; Pourmand, G.; Khalili, E. Tumor-derived urinary exosomal long non-coding RNAs as diagnostic biomarkers for bladder cancer. EXCLI J. 2020, 19, 301–310. [Google Scholar] [CrossRef]
  100. Xiao, H.; Shi, J. Exosomal circular RNA_400068 promotes the development of renal cell carcinoma via the miR-210-5p/SOCS1 axis. Mol. Med. Rep. 2020, 22, 4810–4820. [Google Scholar] [CrossRef]
  101. Li, T.; Sun, X.; Chen, L. Exosome circ_0044516 promotes prostate cancer cell proliferation and metastasis as a potential biomarker. J. Cell. Biochem. 2020, 121, 2118–2126. [Google Scholar] [CrossRef] [PubMed]
  102. Tsuruda, M.; Yoshino, H.; Okamura, S.; Kuroshima, K.; Osako, Y.; Sakaguchi, T.; Sugita, S.; Tatarano, S.; Nakagawa, M.; Enokida, H. Oncogenic effects of RAB27B through exosome independent function in renal cell carcinoma including sunitinib-resistant. PLoS ONE 2020, 15, e0232545. [Google Scholar] [CrossRef]
  103. Iliuk, A.; Wu, X.; Li, L.; Sun, J.; Hadisurya, M.; Boris, R.S.; Tao, W.A. Plasma-Derived Extracellular Vesicle Phosphoproteomics through Chemical Affinity Purification. J. Proteome Res. 2020, 19, 2563–2574. [Google Scholar] [CrossRef] [PubMed]
  104. Zhao, Y.; Wang, Y.; Zhao, E.; Tan, Y.; Geng, B.; Kang, C.; Li, X. PTRF/CAVIN1, regulated by SHC1 through the EGFR pathway, is found in urine exosomes as a potential biomarker of ccRCC. Carcinogenesis 2020, 41, 274–283. [Google Scholar] [CrossRef] [PubMed]
  105. Logozzi, M.; Mizzoni, D.; Capasso, C.; Del Prete, S.; Di Raimo, R.; Falchi, M.; Angelini, D.F.; Sciarra, A.; Maggi, M.; Supuran, C.T.; et al. Plasmatic exosomes from prostate cancer patients show increased carbonic anhydrase IX expression and activity and low pH. J. Enzym. Inhib. Med. Chem. 2020, 35, 280–288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Tomiyama, E.; Matsuzaki, K.; Fujita, K.; Shiromizu, T.; Narumi, R.; Jingushi, K.; Koh, Y.; Matsushita, M.; Nakano, K.; Hayashi, Y.; et al. Proteomic analysis of urinary and tissue-exudative extracellular vesicles to discover novel bladder cancer biomarkers. Cancer Sci. 2021, 112, 2033–2045. [Google Scholar] [CrossRef] [PubMed]
  107. Long, K.; Zeng, Q.; Dong, W. The clinical significance of microRNA-409 in pancreatic carcinoma and associated tumor cellular functions. Bioengineered 2021, 12, 4633–4642. [Google Scholar] [CrossRef]
  108. Liu, J.; Ren, L.; Li, S.; Li, W.; Zheng, X.; Yang, Y.; Fu, W.; Yi, J.; Wang, J.; Du, G. The biology, function, and applications of exosomes in cancer. Acta Pharm. Sin. B 2021, 11, 2783–2797. [Google Scholar] [CrossRef]
  109. Ma, X.; Guo, J.; Liu, K.; Chen, L.; Liu, D.; Dong, S.; Xia, J.; Long, Q.; Yue, Y.; Zhao, P.; et al. Identification of a distinct luminal subgroup diagnosing and stratifying early stage prostate cancer by tissue-based single-cell RNA sequencing. Mol. Cancer 2020, 19, 147. [Google Scholar] [CrossRef]
  110. Igami, K.; Uchiumi, T.; Shiota, M.; Ueda, S.; Tsukahara, S.; Akimoto, M.; Eto, M.; Kang, D. Extracellular vesicles expressing CEACAM proteins in the urine of bladder cancer patients. Cancer Sci. 2022, 113, 3120–3133. [Google Scholar] [CrossRef]
  111. He, J.; He, J.; Min, L.; He, Y.; Guan, H.; Wang, J.; Peng, X. Extracellular vesicles transmitted miR-31-5p promotes sorafenib resistance by targeting MLH1 in renal cell carcinoma. Int. J. Cancer 2020, 146, 1052–1063. [Google Scholar] [CrossRef] [PubMed]
  112. Greenberg, J.W.; Kim, H.; Moustafa, A.A.; Datta, A.; Barata, P.C.; Boulares, A.H.; Abdel-Mageed, A.B.; Krane, L.S. Repurposing ketoconazole as an exosome directed adjunct to sunitinib in treating renal cell carcinoma. Sci. Rep. 2021, 11, 10200. [Google Scholar] [CrossRef] [PubMed]
  113. Shan, G.; Gu, J.; Zhou, D.; Li, L.; Cheng, W.; Wang, Y.; Tang, T.; Wang, X. Cancer-associated fibroblast-secreted exosomal miR-423-5p promotes chemotherapy resistance in prostate cancer by targeting GREM2 through the TGF-β signaling pathway. Exp. Mol. Med. 2020, 52, 1809–1822. [Google Scholar] [CrossRef] [PubMed]
  114. Cao, Z.; Xu, L.; Zhao, S. Exosome-derived miR-27a produced by PSC-27 cells contributes to prostate cancer chemoresistance through p53. Biochem. Biophys. Res. Commun. 2019, 515, 345–351. [Google Scholar] [CrossRef]
  115. Shan, G.; Zhou, X.; Gu, J.; Zhou, D.; Cheng, W.; Wu, H.; Wang, Y.; Tang, T.; Wang, X. Downregulated exosomal microRNA-148b-3p in cancer associated fibroblasts enhance chemosensitivity of bladder cancer cells by downregulating the Wnt/β-catenin pathway and upregulating PTEN. Cell. Oncol. Dordr. 2021, 44, 45–59. [Google Scholar] [CrossRef]
  116. Niu, W.; Xiao, Q.; Wang, X.; Zhu, J.; Li, J.; Liang, X.; Peng, Y.; Wu, C.; Lu, R.; Pan, Y.; et al. A Biomimetic Drug Delivery System by Integrating Grapefruit Extracellular Vesicles and Doxorubicin-Loaded Heparin-Based Nanoparticles for Glioma Therapy. Nano Lett. 2021, 21, 1484–1492. [Google Scholar] [CrossRef]
  117. Rayamajhi, S.; Nguyen, T.D.T.; Marasini, R.; Aryal, S. Macrophage-derived exosome-mimetic hybrid vesicles for tumor targeted drug delivery. Acta Biomater. 2019, 94, 482–494. [Google Scholar] [CrossRef]
  118. Haney, M.J.; Zhao, Y.; Jin, Y.S.; Li, S.M.; Bago, J.R.; Klyachko, N.L.; Kabanov, A.V.; Batrakova, E.V. Macrophage-Derived Extracellular Vesicles as Drug Delivery Systems for Triple Negative Breast Cancer (TNBC) Therapy. J. Neuroimmune Pharmacol. Off. J. Soc. NeuroImmune Pharmacol. 2020, 15, 487–500. [Google Scholar] [CrossRef]
  119. Severic, M.; Ma, G.; Pereira, S.G.T.; Ruiz, A.; Cheung, C.C.L.; Al-Jamal, W.T. Genetically-engineered anti-PSMA exosome mimetics targeting advanced prostate cancer in vitro and in vivo. J. Control. Release Off. J. Control. Release Soc. 2021, 330, 101–110. [Google Scholar] [CrossRef]
  120. Wang, D.; Wan, Z.; Yang, Q.; Chen, J.; Liu, Y.; Lu, F.; Tang, J. Sonodynamical reversion of immunosuppressive microenvironment in prostate cancer via engineered exosomes. Drug Deliv. 2022, 29, 702–713. [Google Scholar] [CrossRef]
  121. Zhou, Q.; Ding, W.; Qian, Z.; Zhu, Q.; Sun, C.; Yu, Q.; Tai, Z.; Xu, K. Immunotherapy Strategy Targeting Programmed Cell Death Ligand 1 and CD73 with Macrophage-Derived Mimetic Nanovesicles to Treat Bladder Cancer. Mol. Pharm. 2021, 18, 4015–4028. [Google Scholar] [CrossRef] [PubMed]
  122. Helissey, C.; Guitard, N.; Théry, H.; Goulinet, S.; Mauduit, P.; Girleanu, M.; Favier, A.L.; Drouet, M.; Parnot, C.; Chargari, C.; et al. Two New Potential Therapeutic Approaches in Radiation Cystitis Derived from Mesenchymal Stem Cells: Extracellular Vesicles and Conditioned Medium. Biology 2022, 11, 980. [Google Scholar] [CrossRef] [PubMed]
  123. Zhao, Q.; Hai, B.; Kelly, J.; Wu, S.; Liu, F. Extracellular vesicle mimics made from iPS cell-derived mesenchymal stem cells improve the treatment of metastatic prostate cancer. Stem Cell Res. Ther. 2021, 12, 29. [Google Scholar] [CrossRef]
  124. Aarts, J.; Boleij, A.; Pieters, B.C.H.; Feitsma, A.L.; van Neerven, R.J.J.; Ten Klooster, J.P.; M’Rabet, L.; Arntz, O.J.; Koenders, M.I.; van de Loo, F.A.J. Flood Control: How Milk-Derived Extracellular Vesicles Can Help to Improve the Intestinal Barrier Function and Break the Gut-Joint Axis in Rheumatoid Arthritis. Front. Immunol. 2021, 12, 703277. [Google Scholar] [CrossRef]
  125. Mecocci, S.; Pietrucci, D.; Milanesi, M.; Pascucci, L.; Filippi, S.; Rosato, V.; Chillemi, G.; Capomaccio, S.; Cappelli, K. Transcriptomic Characterization of Cow, Donkey and Goat Milk Extracellular Vesicles Reveals Their Anti-Inflammatory and Immunomodulatory Potential. Int. J. Mol. Sci. 2021, 22, 12759. [Google Scholar] [CrossRef]
  126. Huang, L.; Rong, Y.; Tang, X.; Yi, K.; Qi, P.; Hou, J.; Liu, W.; He, Y.; Gao, X.; Yuan, C.; et al. Engineered exosomes as an in situ DC-primed vaccine to boost antitumor immunity in breast cancer. Mol. Cancer 2022, 21, 45. [Google Scholar] [CrossRef]
  127. Lu, Z.; Zuo, B.; Jing, R.; Gao, X.; Rao, Q.; Liu, Z.; Qi, H.; Guo, H.; Yin, H. Dendritic cell-derived exosomes elicit tumor regression in autochthonous hepatocellular carcinoma mouse models. J. Hepatol. 2017, 67, 739–748. [Google Scholar] [CrossRef] [PubMed]
  128. Xu, H.Y.; Li, N.; Yao, N.; Xu, X.F.; Wang, H.X.; Liu, X.Y.; Zhang, Y. CD8+ T cells stimulated by exosomes derived from RenCa cells mediate specific immune responses through the FasL/Fas signaling pathway and, combined with GM-CSF and IL-12, enhance the anti-renal cortical adenocarcinoma effect. Oncol. Rep. 2019, 42, 866–879. [Google Scholar] [CrossRef] [PubMed]
  129. Parenky, A.C.; Akalkotkar, A.; Mulla, N.S.; D’Souza, M.J. Harnessing T-cell activity against prostate cancer: A therapeutic microparticulate oral cancer vaccine. Vaccine 2019, 37, 6085–6092. [Google Scholar] [CrossRef]
  130. Shi, X.; Sun, J.; Li, H.; Lin, H.; Xie, W.; Li, J.; Tan, W. Antitumor efficacy of interferon-γ-modified exosomal vaccine in prostate cancer. Prostate 2020, 80, 811–823. [Google Scholar] [CrossRef]
Figure 1. Mechanism of sEVs formation. This figure summarizes the formation process of sEVs. sEVs form MVBs via the endosomal pathway, which are then released into the extracellular space by binding MVBs to Rab 27a and 27b. (This figure was created with Biorender.com).
Figure 1. Mechanism of sEVs formation. This figure summarizes the formation process of sEVs. sEVs form MVBs via the endosomal pathway, which are then released into the extracellular space by binding MVBs to Rab 27a and 27b. (This figure was created with Biorender.com).
Cancers 15 00100 g001
Figure 2. The general function of sEV in tumors and its use in therapy. This figure summarizes the role of sEVs in urological tumors and their application in the treatment. Urological tumor cells and sEVs released by urological cancer stem cells can promote tumor cell EMT, endothelial cell angiogenesis, macrophage activation, and pre-metastatic niches formation. sEVs can be used as drug delivery systems to treat urological tumors. (This figure was created with Biorender.com).
Figure 2. The general function of sEV in tumors and its use in therapy. This figure summarizes the role of sEVs in urological tumors and their application in the treatment. Urological tumor cells and sEVs released by urological cancer stem cells can promote tumor cell EMT, endothelial cell angiogenesis, macrophage activation, and pre-metastatic niches formation. sEVs can be used as drug delivery systems to treat urological tumors. (This figure was created with Biorender.com).
Cancers 15 00100 g002
Table 1. Separation methods for sEVs.
Table 1. Separation methods for sEVs.
MethodMechanismAdvantagesDisadvantages
UltracentrifugationDensityGold standard; Low costTime consuming; low specificity
Density gradientsDensityGold standard; High specificityLow production; Time consuming
PrecipitationSolubilityQuiklyLow specificity, presence of protein
Immuno-captureAntigenQuiklyHigh cost; High specificity
Size exclusion chromatographySizeQuiklyContaminated protein
Table 2. Possible candidate markers of EVs in Urological tumors.
Table 2. Possible candidate markers of EVs in Urological tumors.
TypeDiseaseSourceCargoesReference
miRNARCCSerummiR-4525, miR-155Muramatsu-Maekawa et al., Meng et al. [80,81]
PlasmamiR-301a-3p, miR-1293, miR-92a-1-5p, miR-149-3p, miR-424-3pDias et al., Xiao et al. [82,83]
UrinemiR-224-5p, miR-204-5p, miR-30c-5pQin et al., Kurahashi et al., Song et al. [84,85,86]
PCaSerummiR-93, miR-181a-5pIinuma et al., Wang et al. [87,88]
PlasmamiR-145, miR-221, mIR-451a, miR-141Zabegina et al., Kim et al. [89,90]
UrinemiR-375, miR-574, miR-30b, miR-126Davey et al., Matsuzaki et al. [91,92]
BCPlasmamiR-185, miR-106a, miR-10bSabo et al. [93]
UrinemiR-96-5p, miR-183-5pEl-Shal et al. [94]
lncRNAPCaUrinelncRNA PCA3, lncRNA-p21Li et al., Işın et al. [95,96]
SerumlncRNA HOXD-AS1Jiang et al. [97]
BCPlasmalncRNA PTENP1Zheng et al. [98]
UrinelncRNA ANRILAbbastabar et al. [99]
CircRNARCCPlasmacirc_400068Xiao et al. [100]
PCaBloodcirc_0044516Li et al. [101]
ProteinRCCCellsRab 27bTsuruda et al. [102]
PlasmaMTDHIliuk et al. [103]
UrinePTRFZhao et al. [104]
PCaPlasmaCA IXLogozzi et al. [105]
BCaUrineHsp 90, CEACAMTomiyama et al. [106]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zhang, M.; Lu, Y.; Wang, L.; Mao, Y.; Hu, X.; Chen, Z. Current Status of Research on Small Extracellular Vesicles for the Diagnosis and Treatment of Urological Tumors. Cancers 2023, 15, 100. https://doi.org/10.3390/cancers15010100

AMA Style

Zhang M, Lu Y, Wang L, Mao Y, Hu X, Chen Z. Current Status of Research on Small Extracellular Vesicles for the Diagnosis and Treatment of Urological Tumors. Cancers. 2023; 15(1):100. https://doi.org/10.3390/cancers15010100

Chicago/Turabian Style

Zhang, Mengting, Yukang Lu, Lanfeng Wang, Yiping Mao, Xinyi Hu, and Zhiping Chen. 2023. "Current Status of Research on Small Extracellular Vesicles for the Diagnosis and Treatment of Urological Tumors" Cancers 15, no. 1: 100. https://doi.org/10.3390/cancers15010100

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop