Next Article in Journal
Impact of Preoperative CT-Measured Sarcopenia on Clinical, Pathological, and Oncological Outcomes After Elective Rectal Cancer Surgery
Next Article in Special Issue
A Cross-Sectional Comparative Study: Could Asprosin and Peptide Tyrosine-Tyrosine Be Used in Schizophrenia to Define the Disease and Determine Its Phases?
Previous Article in Journal
Transperineal Vulvar Ultrasound: A Review of Normal and Abnormal Findings with a Proposed Standardized Methodology
Previous Article in Special Issue
A Comparative Study on the Paradoxical Relationship Between Heavy Metal Exposure and Kidney Function
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Recent Exploration of Solid Cancer Biomarkers Hidden Within Urine or Blood Exosomes That Provide Fundamental Information for Future Cancer Diagnostics

1
Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan
2
Hirotsu Bio Science Inc., Chiyoda-Ku, Tokyo 102-0094, Japan
3
Department of Clinical and Molecular Medicine, University of Rome “Sapienza”, Santo Andrea Hospital, Via di Grottarossa, 1035-00189 Rome, Italy
*
Author to whom correspondence should be addressed.
Diagnostics 2025, 15(5), 628; https://doi.org/10.3390/diagnostics15050628
Submission received: 30 December 2024 / Revised: 27 January 2025 / Accepted: 12 February 2025 / Published: 5 March 2025
(This article belongs to the Special Issue Advances in Laboratory Markers of Human Disease)

Abstract

:
Cancer cells exhibit abnormal behavior compared to normal cells. They ignore growth arrest signals such as contact inhibition, a mechanism that stops their proliferation when they collide with surrounding cells, and proliferate in an uncontrolled manner, destroying tissue. Early detection and treatment of cancer are therefore important for healthy longevity. Cancer cells differ from normal cells in their characteristic gene expression due to their abnormalities. Cancer markers that reflect these characteristics have been searched for and applied to diagnosis. Although analysis of blood antigens has been the main method, further development of a diagnostic system is needed for early detection of cancer. Next-generation sequencers have improved gene expression analysis technology, making it possible to analyze detailed gene expression in cancer cells and nucleic acid molecules in blood or urine. In addition, cancer cells release extracellular vesicles, exosomes, which are known to contain molecules that may serve as cancer markers. This review summarizes the latest findings on exosomal cancer markers.

1. Introduction

Cancer cells behave in a selfish manner and use every means at their disposal for their own proliferation [1,2,3]. They change energy metabolism, adapt to hypoxia by enhancing the glycolytic system, promote proliferation by activating cell cycle signaling pathways, form new blood vessels to supply nutrition, acquire the ability to metastasize to other organs via blood and lymph fluids, acquire immune evasion ability by expressing immune checkpoint molecules such as PD-L1, and develop cancer stem cells that show resistance to chemotherapy [4,5,6,7]. It is a difficult task to achieve complete cures for the large amounts of cancerous tissues that have acquired all of these abilities. Therefore, early detection and treatment of cancerous tissues are important in completely curing cancer [8]. For this purpose, diagnosis of cancer using cancer markers in blood and urine is recommended [9]. Therefore, a search for cancer markers that can accurately diagnose early-stage cancer is being conducted [10,11]. In particular, exosomes released by cancer cells contain many cancer-specific molecules and are expected to be applied to cancer diagnosis [12]. Exosomes are extracellular vesicles of about 30–150 nm in diameter, with CD9, CD63, and CD81 as common exosome markers on their surface and various RNAs and proteins inside [13]. Exosomes are released when the cell membrane is entrapped to form endosomes, vesicles are formed within the endosomes, and these endosomes fuse with the cell membrane. Exosomes can be separated from blood and urine by ultracentrifugation, immunoaffinity, size exclusion, and precipitation [14]. Exosomes derived from cancer cells have been found to be involved in cancer growth, metastasis, angiogenesis, and immune escape, and are released into the blood and urine [15]. This review summarizes recent reports on candidate cancer markers present in the exosomes of cancer patients.

2. Search for Exosome-Derived Cancer Markers in Blood

Cancer diagnosis using blood cancer markers has become common [16,17]. Exosomes derived from cancer are circulating in the blood [18]. Therefore, collection of exosomes from blood and analysis of their contents can lead to the identification of useful cancer markers (Table 1).

2.1. Blood Exosomes in Breast Cancer Patients

This section describes the latest reports on blood exosome markers for breast cancer. Exosomes from the blood of 120 breast cancer patients and 60 healthy controls were collected and small RNA sequencing identified about 3500 small RNAs. A diagnostic model was constructed with six small RNAs (piR-36,340, piR-33,161, miR-484, miR-548ah-5p, miR-4282, and miR-6853-3p), and the area under the curve (AUC) was 0.972. The targets of these microRNAs (miRNAs) were involved in the chemokine signaling pathway [19]. miR-200c was quantified in blood exosomes from 51 breast cancer patients and 47 healthy controls. miR-200c expression was downregulated in breast cancer patients. The AUC of miR-200c was 0.854, whereas those of CEA, CA125, and CA153 were 0.615, 0.700, and 0.727, respectively. In those four combined models, the AUC was 0.914, sensitivity was 91.49%, and specificity was 76.6% [20]. Exosome sequencing was performed on the blood of three chemotherapy-sensitive patients and three drug-resistant patients with triple-negative breast cancer (TNBC). A total of 85 miRNAs were identified whose expression varied in relation to chemotherapy resistance. In addition, when six miRNAs (miR-182, miR-1246, miR-378-e, miR-6730-3p, miR-6831-5p, and miR-373) were quantified by RT-qPCR in 30 TNBC patients, only miR-6831-5P showed expression fluctuation, indicating that miR-6831-5P was downregulated in drug-resistant patients [21]. An integrated centrifugal disk chip (CD chip) was developed for rapid quantification of surface proteins of blood exosomes, and target proteins could be quantified within 10 min with 100 μL of blood. Exosomes from breast cancer cells (MCF-7) and normal mammalian epithelial cells (MCF-10A) were collected and Western blotting was used to detect EpCAM, PSMA, HER2, EGFR, CEA, and CA125, and a marked increase in expression of EGFR, CEA, and CA125 was observed in MCF-7. EpCAM was detected in MCF-7 and MCF-10A to the same extent, and PSMA and HER2 were slightly upregulated in MCF-7. Quantification of CEA, CA125, and EGFR on the surface of blood exosomes of six breast cancer patients and two healthy controls with a CD chip was able to identify breast cancer patients [22]. Quantification of CD9- and HER2-positive exosomes from pre- and postoperative blood samples of eight breast cancer patients showed a trend toward lower exosome levels after surgery [23]. Blood exosomes from 30 breast cancer patients, 30 colorectal cancer patients, 30 lung cancer patients, and 39 healthy controls were collected and quantified for PD-L1, EpCAM, and EGFR, showing that the expression of these genes was increased in cancer patients. Diagnostic modeling with PD-L1, EpCAM, and EGFR showed that the AUC was 0.978, sensitivity was 90.0%, specificity was 97.4%, and accuracy was 94.2% for breast cancer, AUC was 0.977, sensitivity was 96.7%, specificity was 100%, and accuracy was 98.6% for colorectal cancer, and AUC was 0.936, sensitivity was 80.0%, and accuracy was 98.6% for lung cancer [24]. The above miRNAs and proteins are expected to be used as exosome markers in breast cancer blood for diagnosis.

2.2. Blood Exosomes of Lung Cancer Patients

This section describes the latest reports on blood exosome markers for lung cancer. Blood exosomes from 24 patients with squamous-cell lung cancers (SQCLCs), 24 patients with lung adenocarcinomas (LUADs), and 24 healthy controls were collected for miRNA analysis, and eight differentially expressed miRNAs (miR-21-5p, miR-126-3 p, miR-210-3p, miR-221-3p, Let-7b-5p, miR-146a-5p, miR-222-3p, and miR-9-5p) were detected. Diagnostic models were constructed with each miRNA, and diagnostic tests were performed, showing that the AUC for SQCLC was 0.646–0.740, sensitivity was 62.5–83.3, and specificity was 66.7–87.5, and for LUAD, the AUC was 0.535–0.655, sensitivity was 41.7–79.2, and specificity was 4.2–91.7 [25]. Blood exosomes from 31 non-small-cell lung cancer (NSCLC) patients were collected and miR-29a-3p was quantified by RT-qPCR. miR-29a-3p expression was upregulated in the 20 patients in the non-relapse group [26]. Blood exosomes from 305 NSCLC patients and 226 healthy controls were collected and snoRNAs were quantified by RT-qPCR to identify those with altered expression. Among them, SNORD116 and SNORA21 were downregulated in NSCLC patients, with AUCs of 0.738 and 0.775, respectively. When traditional blood biomarkers CYFRA21-1 and carcinoembryonic antigen (CEA) were combined with SNORD116 and SNORA21, the AUC was 0.917. Furthermore, this diagnostic model was able to identify 132 metastatic NSCLC patients and 173 non-metastatic NSCLC patients with an AUC of 0.784 [27]. Blood exosomes from nine lung cancer patients and three healthy controls were collected and quantified for mRNAs, long non-coding RNAs (lncRNAs), miRNAs, and circular RNAs (circRNAs) by microarray and RNA-seq, showing that 591 mRNAs, 881 lncRNAs, 45 miRNAs, and 916 circRNAs were upregulated, while 108 mRNAs, 135 lncRNAs, and 480 circRNAs were downregulated in lung cancer patients. RT-qPCR of circ-0033861, circ-0043273, and circ-0011959 showed that they were significantly upregulated in lung cancer patients [28]. Blood exosomes of 79 patients with or without lung cancer metastasis were collected and gene expression analysis identified five genes (CXCL12, TFBR2, CD44v6, HIF1A, and KRT7) important for diagnosing metastasis. A diagnostic model was constructed with these genes and the AUC was 0.9488 [29]. Exosomes from the blood of 30 NSCLC patients were collected and nested PCR was used to detect EGFR mutations, showing a sensitivity of 76.6% [30]. Exosomes from the peripheral blood of 10 small-cell lung cancer (SCLC) patients were collected from peripheral blood mononuclear cells (PBMC-EXs) and analyzed for proteins. Protein analysis revealed increased expression of c-Myc and Snail and decreased expression of MAVS and STING in chemoimmunotherapy-treated non-responder cells. Furthermore, when responder exosomes were fed to SCLC cell lines, apoptosis was observed to be more pronounced than in the case of non-responder cells [31]. Exosome spectra of lung cancer cell lines (NCI-H226, HCC-827, and A549) and normal cell line (BEAS-2B) were analyzed by surface-enhanced Raman spectroscopy (SERS) and a diagnostic model was constructed using machine learning. Blood exosomes from 20 lung cancer patients and 20 healthy controls were collected for diagnostic testing, showing that the AUC was 0.84, sensitivity was 83.3%, and specificity was 83.3% [32]. Blood exosomes from 60 lung cancer patients, 95 breast cancer patients, and 78 healthy controls were collected for detection of alpha-linked Thomsen–Friedenreich glycoantigen (TF-Ag-α). TF-Ag-α was detected in patients with cancer and the diagnosis could be made with more than 95% accuracy. This indicates that TF-Ag-α is a new exosomal carbohydrate biomarker [33]. As described above, RNAs and proteins have been identified as exosome markers in lung cancer blood. It is interesting to note that circular RNAs and small nucleolar RNAs (snRNAs) involved in RNA modification also have potential diagnostic applications.

2.3. Blood Exosomes of Pancreatic Cancer Patients

This section describes the latest reports on blood exosome markers for pancreatic cancer. Blood exosomes of five pancreatic cancer patients and five healthy controls were collected and lncRNAs with altered expression were identified by RNA-seq. Four of them (LINC01268, LINC02802, AC124854.1, and AL132657.1) were further quantified in 78 pancreatic cancer patients and 70 healthy controls by RT-qPCR. When the diagnostic efficacy of these four lncRNAs was analyzed, the AUC, sensitivity, and specificity were 0.8476, 0.72, and 0.89, respectively [34]. Exosomes from the blood of 10 pancreatic cancer patients were collected and miRNAs were analyzed by microarray. miR-6855-5p was found to be associated with radioresistance. Further analysis of 28 pancreatic cancer patients confirmed a trend toward increased radioresistance in the group with higher expression of blood exosome-derived miR-6855-5p [35]. Blood exosomes from 18 pancreatic cancer patients and 16 healthy controls were collected for detection of miR-21, miR-191, and miR-451a by molecular beacon–peptide (MBP) probes, showing increased expression of these miRNAs in pancreatic cancer patients [36]. miRNA analysis of blood exosomes collected from 20 pancreatic cancer patients by microarray revealed that six miRNAs (miR-10394-5p, miR-6779-5p, miR-197-5p, miR-4327, miR-638, and miR-12117) were involved in gemcitabine-based preoperative treatment responder and three miRNAs (miR-6891-5p, miR-6732-5p, and miR-1234-3p) were involved in the poor responder. A diagnostic model with these miRNAs was further constructed and diagnostic tests were performed on 66 patients, showing that the AUC was 0.777 for responders and 0.685 for poor responders [37]. Blood exosomal gene expression data from 164 pancreatic cancer patients and 118 healthy controls in ExoRBase 2.0. were analyzed with the LASSO Cox regression model, showing that expression of ARNTL2, FHL2, KRT19, MMP1, CDCA5, and KIF11 was associated with pancreatic cancer. The prognosis was worse in the ARNTL2, KRT19, MMP1, CDCA5, and KIF11 high-expression group and the FHL2 low-expression group [38]. The above miRNAs, lncRNAs, and proteins were identified as exosome markers in pancreatic cancer blood. It was interesting that lncRNAs were detected as markers and they are increasingly expected to be studied for diagnostic applications.

2.4. Blood Exosomes in Colorectal Cancer Patients

This section describes the latest reports on blood exosome markers for colorectal cancer. Blood exosomes from 58 colorectal cancer (CRC) patients and 28 healthy controls were collected and NAMPT-Antisense (NAMPT-AS) and Nicotinamide phosphoribosyl-transferase (NAMPT) mRNA were quantified by RT-qPCR, showing that they were upregulated in patients with CRC. Elevated expression of serum NAMPT was also observed in patients with CRC. Diagnostic models of each showed an AUC of 0.65 for NAMPT-AS, 0.646 for NAMPT mRNA, and 0.632 for serum NAMPT [39]. Blood exosomes from 31 T1a-stage CRC patients, including 22 colon cancer (CC) patients and 9 rectum cancer (RC) patients, 19 precancerous advanced adenoma (AA) patients, and 10 healthy controls were collected and subjected to RNA-seq analysis to identify genes with variable expression. A CRC prediction model was constructed with eight RNAs (Let-7f-5p, C19orf43, TOP1, PPDPF, lnc-MKRN2-42:1, LNC-EV-9572, HIST2H2AA4, and miR-320a-3p), and the AUC was 0.76. An AA prediction model was also constructed with nine RNAs (miR-425-5p, Let-7f- 5p, C19orf43, TOP1, PPDPF, LNC-EV-9572, lnc-MKRN2-42:1, HIST2H2AA4, and MT-ND2), and the AUC was 0.88 [40]. Blood exosomes from six CRC patients and four healthy controls were collected and 5-methylcytosine miRNA-21 (m5C-miRNA-21) was quantified by DNAzyme-RCA-based colorimetric and lateral flow dipstick assays, showing that m5C-miRNA-21 was upregulated in patients with CRC [41]. Proteomic analysis of blood exosomes collected from 25 CRC patients and 12 healthy controls by mass spectrometry identified about 60 proteins with variable expression. A diagnostic model constructed using machine learning with ELISA data from 912 patients revealed that PF4 and AACT, which were upregulated in CRC patients, were important for CRC diagnosis, with an AUC of 0.963 in the diagnostic test [42]. The above miRNAs, lncRNAs, and proteins were identified as exosome markers in breast cancer blood, and the fact that RNA methylation was detected as a marker is intriguing. The application of the RNA modification perspective to diagnosis is increasingly expected.

2.5. Blood Exosomes in Other Cancer Patients

This section describes the latest reports on blood exosome markers for cancers other than those described in the previous sections. Analysis of blood RNA data (GSE164174) and gene expression data (TCGA) in gastric cancer (GC) identified four miRNAs (miR-21-5p, miR-320, miR-191-5p, and miR-451) for diagnosing metastasis. A diagnostic model was constructed for metastasis with these miRNAs and found that the AUC was 0.86, which is higher than that of the existing markers, CEA, CA19-9, CA125, and CA72-4 [43]. An SERS sensor was developed for exosome retrieval and target protein detection. When the MUC1 protein was quantified in blood exosomes of 15 gastric cancer patients and 5 healthy subjects, a significant increase in MUC1 expression was observed in gastric cancer patients [44]. When a diagnostic model was constructed using blood exosome gene expression data of hepatocellular carcinoma (HCC) patients extracted from exoRBase, RNA-seq data from cancer tissues from the TCGA database, and scRNA-seq data from the GEO database, the AUC was 0.847 in the exoRBase HCC cohort [45]. Blood exosomes from nine HCC patients were collected and proteomics analysis using mass spectrometry identified approximately 2500 proteins. In three of the HCC patients with post-hepatectomy liver failure (PHLF), 53 proteins were upregulated and 32 proteins were downregulated. The significantly upregulated proteins were DRAP1, GGCT, NSUN2, RAB13, PPCS, and SDHA, and the significantly downregulated proteins were CTSB, TIMM44, VTN, KATNAL2, and RPL27A [46]. Knockdown of lncRNA brain cytoplasmic RNA 1 (BCYRN1) in T24 and BOY bladder cancer cells suppressed proliferation, migration, and invasion. Blood exosomes from 31 bladder cancer patients and 19 healthy controls were collected and quantification of BCYRN1 showed increased expression of BCYRN1 in patients with bladder cancer. In addition, a decrease in BCYRN1 expression was observed in eight patients who underwent complete resection of bladder cancer [47]. Blood samples from 15 ovarian cancer (OC) patients, 14 ovarian cancer patients with anti-Yo-associated paraneoplastic cerebellar degeneration (PCD), and 15 healthy controls were collected and miRNA analysis identified about 100 miRNAs with variable expression. There was a marked upregulation of miR-483-5p, miR-4488, and miR-200c-3p in patients with OC compared to healthy controls. Furthermore, there was a significant upregulation of miR-451a, miR-486-5p, and miR-20b-5p in PCD patients compared to healthy controls, and miR-451a, miR-486-5p, and miR-15a-3p in PCD patients compared to OC patients [48]. Blood exosomes from 134 cervical cancer (CC) patients and 50 healthy controls were collected and lncRNA lymphocytic leukemia deletion gene 1 (DLEU1) was quantified by RT-qPCR. The diagnostic model of DLEU1 showed an AUC of 0.808, sensitivity of 63.4%, and specificity of 86.0%. In the case of CA-125, the AUC was 0.670, sensitivity was 56.0%, and specificity was 87.0%, and in the case of squamous-cell carcinoma (SCC), the AUC was 0.746, sensitivity was 56.7%, and specificity was 93.0%. The combined model of DLEU1, serum CA-125, and SCC showed an AUC of 0.878, sensitivity of 65.7%, and specificity of 94.0% [49]. Exosomes from patients with cholangiocarcinoma, patients suffering from an O. viverrini infection that was causing cholangiocarcinoma, and healthy controls were collected, and miRNA analysis by RNA-seq showed that miR-92a-3p, miR-203a-3p, miR-192-5p, miR-223-3p, miR-26a-5p, and miR-194-5p were significantly upregulated in cholangiocarcinoma patients, and in patients infected with O. viverrini, only miR-223-3p was significantly upregulated [50]. Blood exosomes from 30 laryngeal cancer (LCa) patients and 20 healthy controls were collected and exosomal surface proteins were quantified by flow cytometry multiplex analysis. Overexpression of CD1c, CD2, CD3, CD4, CD11c, CD14, CD20, CD44, CD56, CD105, CD146, and CD209 was observed in patients with LCa, while CD24, CD31, and CD40 were not overexpressed in patients with LCa, but were found to be associated with nodal involvement. The diagnostic model of CD56 had the highest AUC of 0.731 in LCa, and the diagnostic model of CD40 had the highest AUC of 0.80 in diagnosis of nodal involvement [51]. The above miRNAs, lncRNAs, and proteins are expected to be used as blood exosome markers for diagnosis.

3. Search for Exosome-Derived Cancer Markers in Urine

Urinalysis is very useful in that it can be performed in a noninvasive manner and is less burdensome to the patient. Exosomes are also found in urine, and new biomarkers are being identified from the analysis of cancer-derived urinary exosomes [52] (Table 2).

3.1. Urinary Exosomes in Patients with Bladder Cancer

This section describes the latest reports on urinary exosome markers for bladder cancer. RNA-seq of urinary exosomes from three bladder cancer patients and three healthy controls revealed that 145 lncRNAs were upregulated and 13 lncRNAs were downregulated in bladder cancer patients. Among them, RMRP was most significantly upregulated in patients with bladder cancer. Diagnostic testing with RMRP of urinary and plasma exosomes in bladder cancer patients showed an AUC of 0.720 for urine alone, 0.870 for plasma alone, and 0.890 for both combined. RMRP contributed to cancer progression by sponging miR-206 and increasing G6PD expression [53]. RT-RAA-CRISPR/Cas12a technology was developed to detect lncRNA mitochondrial RNA processing endoribonuclease (RMRP) in exosomes in about 30 min. When diagnostic tests were performed on RMRP of urinary exosomes from patients with bladder cancer and healthy controls, and the AUC was 0.946, sensitivity was 0.950, and specificity was 0.943 [54]. Urinary exosomes from 42 bladder cancer patients were collected and quantified for seven lncRNAs (UCA1, H19, MALAT1, TUG1, GAS5, RMRP, and LINC01517), showing increased expression of RMRP, UCA1, and MALAT1 in bladder cancer patients. The diagnostic model was constructed using these genes, and the AUC, sensitivity, and specificity were 0.875, 80.0%, and 81.4%, respectively [55]. Urinary exosomes from 42 bladder cancer patients and 42 healthy controls were collected and quantified for lncRNA SNHG16 by RT-qPCR, showing that lncRNA SNHG16 was highly expressed in bladder cancer patients. When lncRNA SNHG16 was used to construct a diagnostic model, the AUC was 0.791, which was higher than the AUC value (0.597) obtained by urinary cytology [56]. RT-qPCR quantification of miR-146a-5p, miR-93-5p, miR-663b, miR-21, and miR-4454 in urinary exosomes from 116 patients with bladder cancer and 116 healthy controls showed increased expression of these miRNAs in patients with bladder cancer. Furthermore, miR-21 was found to be associated with tumor-node-metastasis staging and grading [57]. Exosomes from 41 non-muscle-invasive bladder cancer patients and 15 healthy controls were collected and subjected to small RNA next-generation sequencing, which revealed 14 differentially expressed transfer RNA-derived fragments (tRFs) (tRF-16-F1R3WEE, tRF-17-8R6546J, tRF-17-I7XUK8N, tRF-17-D9W1X6K, tRF-18-HR1PF7D2, tRF-18-MBQ4NKDJ, tRF-20-40KK5Y93, tRF-21-86J8WPMNB, tRF-25-7P596VW631, tRF-26-IK9NJ4S2I7D, tRF-27-J87383RPD95, tRF-31-PER8YP9LON4VD, tRF-32-PER8YP9LON4V3, and tRF-38-PNR8YP9LON4VN18) [58]. RNA-seq of urinary exosomes from 12 bladder cancer patients and 6 healthy controls identified KLHDC7B as upregulated in bladder cancer patients. High-grade and low-grade bladder urothelial carcinoma (BLCA) could be distinguished by the expression level of KLHDC7B [59]. RT-qPCR quantification of KRT17, GPRC5A, SLC2A1, MDK, and CXCR2 in urinary exosomes from 236 bladder cancer patients and 42 healthy controls showed increased expression of these mRNAs in cancer patients. A diagnostic model of these mRNAs showed AUCs of 0.760 and 0.730, sensitivities of 0.633 and 0.620, and specificities of 0.786 and 0.810 for MDK and KRT17, respectively. KRT17 was significantly upregulated in patients with relapsed disease [60]. RNA-seq analysis of urinary exosomes from 60 bladder cancer patients and 40 healthy controls revealed that 33 genes were upregulated and 156 genes were downregulated in bladder cancer patients. These genes were related to the MAPK pathway, PPAP signaling pathway, PI3K Akt signaling pathway, and Hippo signaling pathway. ROC curves were constructed for tmeff1, SDPR, ACBD7, SCG2, and COL6A2, and the AUC was 0.6934, 0.7746, 0.7239, 0.6396, and 0.6610, respectively. When the ROC curves were constructed for SDPR and acbd7, the AUC was 0.7945, the sensitivity was 89.09%, and the sensitivity was 60.53% [61]. A magnetic 3D ordered macroporous zeolitic imidazolate framework-8 (magMZIF-8), constructed to efficiently separate urinary exosomes, enabled exosome metabolomics analysis using 50 mL of urine, and it took only 2 h to separate exosomes from 42 urine samples. Metabolite analyses in exosomes of bladder cancer patients and healthy controls were performed by LC-MS/MS, showing that arachiconic acid, docosahexaenoic acid, docosapentaenoic acid, and retinyl ester were elevated in bladder cancer patients. When a diagnostic model was constructed by applying machine learning algorithms to the analyzed data, the AUC was 0.875–1.00 [62]. An inorganic (Ti3AlC2)-based exosome collector (MXene@TiO2) was developed to collect urinary exosomes from 113 bladder cancer patients and 112 healthy controls, and metabolic profiles were constructed for 465 metabolites using mass spectrometry. The data were used to construct a diagnostic model using machine learning, resulting in an AUC of 0.867 [63]. When urinary exosomes from bladder cancer patients and healthy subjects were collected and analyzed for exosomal glycans, approximately 50 species could be identified, 16 of which showed differential expression. In particular, one upregulated bisecting N-acetylglucosamine (GlcNAc)-type glycan with core fucose, and two upregulated and two downregulated terminal-sialylated glycans were observed in bladder cancer patients [64]. Urinary exosomes from 39 urinary bladder cancer (UBC) patients and healthy controls were collected and analyzed by proximity extension assay, showing that MMP12, MMP7, HO-1, IL8, CD5, CCL20, CXCL13, MCP-1, CD8A, and TGF-beta-1 were upregulated in UBC patients. A model to diagnose muscle invasiveness was constructed from the analyzed data, and the accuracy was 92% [65]. Proteomic data from the urinary exosomes of 261 cancer patients (bladder cancer, prostate cancer, renal cancer, lung cancer, cervical cancer, colorectal cancer, esophageal, and gastric cancer) and 124 healthy controls identified 17 proteins (CD59, CDC42, ITM2B, CD81, PEBP1, VAT1, MYO1D, RAC1, DPP4, RAN, CAPG, PPIA, FOLR1, ANXA3, APOD, ANXA4, and AQP2) important for cancer diagnosis. When a diagnostic model was constructed by machine learning using the expression data of these 17 proteins, the AUC was 0.96 and accuracy was 0.90 [66]. The above miRNAs, lncRNAs, proteins, and metabolites were identified as urinary exosome markers for bladder cancer. It is interesting that metabolites were also detected as markers, and their diagnostic application is expected to increase.

3.2. Urinary Exosomes of Prostate Cancer Patients

This section describes the latest reports on urinary exosome markers of prostate cancer. A dumbbell dual-hairpin-triggered DNA nanonet that forms a net structure in the presence of miR-141 was developed to detect miR-141, enabling the detection of miR-141 at 57.6 pM [67]. Urinary exosomes were collected and quantified for miR-451 and miR-21 before and after surgery in 10 PCa patients, showing that both miRNAs were highly expressed preoperatively but low postoperatively [68]. RNA-seq analysis of urinary exosomes from 10 PCa patients and 10 healthy controls, and qRT-PCR analysis of urinary exosomes from 43 PCa patients and 92 healthy controls, revealed PCa patient-specific mRNAs. AUC values were between 0.799 and 0.906 for diagnosis by RAB5B, WWP1, HIST2H2BF, ZFY, MARK2, PASK, RBM10, and NRSN2. Diagnostic modeling with RAB5B and WWP1 showed an AUC of 0.923, 81.4% sensitivity, and 89.1% specificity [69]. Urinary exosomes of prostate cancer (PCa) patients were analyzed with a dual-gate field-effect transistor (DGFET)-based multimarker biosensor, and TMEM256 was upregulated in PCa patients [70]. Urine exosomes from 284 patients who underwent testing to determine prostate cancer were collected and quantified by ELISA for prostate-specific membrane antigen (PSMA), showing that PSMA was upregulated in PCa patients compared to benign patients, with an AUC of 0.876 [71]. In PCa patients, cancer-cell-derived exosomal PSM-E is upregulated in the serum and urine. When exosomal PSM-E is incorporated into the M0 macrophage, PSM-E binds to the fourth tryptophan aspartate repeat of RACK1 in the protease-associated domain and suppresses FAK and ERK signaling pathways, thereby inhibiting M2 macrophage polarization, resulting in the suppression of prostate cancer cell proliferation, invasion, and metastasis [72]. Urinary exosomes were collected from 272 prostate biopsy patients, and urinary exosomal prostate-specific antigen (UE-PSA) was quantified by ELISA, showing that increased expression of UE-PSA was observed in PCa patients compared to benign patients with an AUC of 0.953 [73]. The above miRNAs and proteins are expected to be used as urinary exosome markers for the diagnosis of prostate cancer.

3.3. Urinary Exosomes of Other Cancer Patients

This section describes the latest reports on urinary exosome markers for cancers other than those described in the previous sections. When exosomes were collected from the urine of 100 lung cancer patients and 100 healthy controls by nanowire, approximately 2500 miRNAs were detected, 48 of which were upregulated (miR-1250-5p, miR-1254, miR-1273f, miR-1910-3p, miR-3064-3p, miR-3164, miR-3591-3p, miR-3691-5p, miR-424-3p, miR-4296, miR-4300, miR-4306, miR-4311, miR-4321, miR-4428, miR-4429, miR-4436b-3p miR-4453, miR-4470, miR-4520-3p, miR-4520-5p, miR-4525, miR-4538, miR-4644, miR-4647, miR-4657, miR-4660, miR-4692, miR-4727-3p, miR-4784, miR-5093, miR-5189-5p, miR-551b-5p, miR-5698, miR-6076, miR-6131, miR-614, miR-650, miR-6515-5p, miR-6747-5p, miR-6760-5p, miR-6801-5p, miR-6815-5 p, miR-6828-5p, miR-7151-3p, miR-766-5p, miR-8057, and miR-921) and 6 of which were downregulated (miR-20a-3p, miR-374c-3c, miR-431-5p, miR-452-3p, miR-642a-3p, and miR-671-5p) in lung cancer patients. The upregulated miRNAs included those related to the MAPK signaling pathway and the PI3K-Akt signaling pathway. Furthermore, when analyzed data were applied to machine learning-based analysis to construct a diagnostic model, the AUC was 0.99 [74]. Through LC-MS/MS analysis of urinary exosomes from 30 healthy controls, 12 lymphocyte migration regulation-related proteins (WASL, STK10, SPNS2, STK10, PKD1, LCK, and GP2) were identified. Among them, WASL, STK10, and WNK1 were diagnosed in the urinary exosomes of 44 patients with lung cancer, with an AUC of 0.760 [75]. Urinary exosomes from nine pancreatic ductal adenocarcinoma (PDAC) patients and seven healthy controls were collected, and miRNA analysis showed that the miR-3940-5p/miR-8069 ratio was increased in PDAC patients. When the diagnosis was made using the miR-3940-5p/miR-8069 ratio and CA19-9, the sensitivity was 93.0% and the positive predictive value was 78.4% [76]. Exosomes were recovered from urine of five pancreatic cancer patients and five healthy controls using phosphatidylserine molecularly imprinted polymers (PS-MIPs), and proteomic analysis by mass spectrometry showed that SLC9A 3R1, SPAG9, and ferritin light chain (FTL) were upregulated in pancreatic cancer patients, suggesting their potential diagnostic markers for pancreatic cancer [77]. Mass spectrometry analysis of urinary exosomes from 10 epithelial ovarian cancer (EOC) patients and 10 healthy controls revealed increased expression of leucine-rich alpha-2-glycoprotein 1 (LRG1) in EOC patients. LRG1 expression is associated with poor prognosis and activates the focal adhesion kinase/protein kinase B (FAK/AKT) signaling pathway, which promotes cancer progression [78]. Urinary exosomes from four ovarian cancer patients and two healthy controls were collected and quantified for CD117. CD117 was not detected in healthy controls, but in the case of patients with high-grade serous ovarian carcinoma and papillary serous cystadenocarcinoma, CD117 was upregulated [79]. To rapidly detect miRNAs in exosomes, fusogenic nanoreactor (FNR)-encapsulating DNA-fueled molecular machines (DMMs) were constructed and miR-222, miR-200c, and miR-375 were quantified in the exosomes of breast cancer patients and healthy controls. Increased expression of these miRNAs was observed in breast cancer patients. In addition, a discrimination test between breast cancer patients and healthy controls was performed, and the diagnostic accuracy was 86.4% [80]. Surface-enhanced Raman spectroscopy (SERS) was performed to detect exosomal miRNAs, and it was able to detect exosomal miRNAs at concentrations as low as 0.5 nM. When exosomal miRNAs of cancer cells and normal cells were analyzed, exosomes derived from cancer cells were richer in miRNAs. In addition, exosomal miRNA levels were found to increase when cells were subjected to electrical stimulation [81]. miRNA-seq was performed on urinary exosomes from renal-cell carcinoma patients and healthy controls, and an increased expression of miR-542-5p and decreased expression of miR-320a were observed in cancer patients [82]. RNA-seq of urinary exosomes from 47 clear-cell renal-cell carcinoma (ccRCC) patients and 16 urolithiasis controls revealed that in ccRCC patients, SNORD99, SNORD22, SNORD26, and SNORA50C were downregulated in ccRCC patients. When a diagnostic model was constructed using SNORD99, SNORA50C, obesity, and hypertension, the accuracy was 0.811 [83]. Urinary exosomes from 30 Wilms’ tumor patients with a rare kidney cancer and 27 healthy controls were collected and quantified by qPCR for metastasis-associated lung adenocarcinoma transcript-1 (MALAT1), showing that MALAT1 expression was decreased in Wilms’ tumor patients [84]. PD-L1 and Alix were quantified in the urinary exosomes of 26 urothelial cancer patients and 12 healthy controls by ELISA and it was found that the expression of PD-L1 and Alix tended to be elevated in cancer patients [85]. Proteomic analysis of urinary exosomes from nine CRC patients and three healthy controls using label-free liquid chromatography–tandem mass spectrometry (LC-MS/MS) revealed the upregulation of 67 proteins and downregulation of 74 proteins in CRC patients. In particular, increased expression of CEACAM7 and CEACAM1, and decreased expression of CHMP4A, CHMP4B, CHMP2A, CHMP2B, and CHMP1B, were prominent in CRC patients [86]. When urinary exosomes from 21 patients with thyroid carcinoma were collected and analyzed for proteins by mass spectrometry, increased expression of tissue inhibitors of metalloproteinases (TIMPs) was observed in patients with lymph node metastasis [87]. Proteomic analysis of urinary exosomes from 18 hepatocellular carcinoma patients and healthy controls by mass spectrometry revealed increased expression of OLFM4, HDGF, and GDF15 in patients with hepatocellular carcinoma [88]. The above RNAs and proteins were identified as urinary exosome markers. It is interesting to note that changes were also observed in the spectral analysis of exosomes, which is expected to have diagnostic applications.

4. Conclusions and Future Directions

As described above, analysis of exosome contents in blood and urine has identified proteins, miRNAs, lncRNAs, and mRNAs that are promising cancer markers. It is interesting to note that the molecular species detected in the analysis of exosome contents differ depending on the report, even for the same cancer type. This may be due to differences in extraction methods, analysis methods, and sample storage methods. Therefore, it may be necessary to consider these conditions when applying exosomes to diagnosis. Since blood and urine samples are less burdensome for patients, diagnosis by blood and urine exosomes is a simple test for patients. Thus, regular testing may facilitate early detection of cancer. Exosomes are released from many cells, so ensuring the specificity of the marker will be a challenge in the future. If the issue of specificity in exosome diagnostics can be overcome, more cancer types could be addressed. In particular, its application to cancers that are difficult to detect at an early stage, such as pancreatic cancer, is highly promising. miR-21 is deeply involved in cancer progression in pancreatic cancer, and its use as a marker may be possible [89]. Exosomes contain information about cancer. Therefore, by analyzing exosomes, we may be able to learn the characteristics of cancer and use them not only for diagnosis but also for treatment selection. The use of exosomes may be cost-effective in that some information that cannot normally be obtained without resection of the cancer and analysis of the cancer tissue can be obtained by analysis of exosomes. Further analysis will continue in the future to achieve higher accuracy in cancer diagnosis. The discovery of new cancer markers through machine learning is particularly remarkable. As AI technology improves and the computational power of computers increases, it is expected that new indicators that have not been noticed by the human eye will become increasingly apparent. In addition, the improved capabilities of analyzers will yield big data. It will become possible to construct a system that can perform multifaceted cancer diagnosis by integrating big data obtained from patients and information on cancer markers identified so far. Such a system may already exist in certain organisms. In recent years, cancer diagnosis has become possible with nematodes [90]. Nematodes have hundreds of olfactory receptors that can detect cancer-derived substances in urine and the detection appears as chemotaxis [91]. In other words, they can answer the question of whether cancer is present or not from a large amount of input information. It is important to utilize this ability. RNA-seq technology has made it possible to analyze various RNA molecules. Recently, RNA modifications such as m6A have been found to be important for cancer growth [92,93]. Furthermore, it has been reported that modified RNA molecules are also present in exosomes, so it is likely that more and more modified RNA molecules will be found as cancer markers in the future as the measurement technology of modified RNA molecules improves [94]. In addition, since lncRNAs have recently been found to encode micropeptides, they may also be potential cancer marker candidates [95,96,97]. Surely a bright future awaits us in the field of cancer biomarker discovery.

Author Contributions

T.H. (Tomoaki Hara), S.M., A.H.A., H.H., E.d.L., T.H. (Takaaki Hirotsu) and H.I. contributed to conceptualization. T.H. (Tomoaki Hara), S.M., Y.A., Y.S. and K.I. collected references and made tables. T.H. (Tomoaki Hara), S.M., A.H.A., H.H., E.d.L., A.V., T.H. (Takaaki Hirotsu) and H.I. wrote the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported in part by a Grant-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology (grant nos. 19K22658, 20H00541, 21K19526, 22H03146, 22K19559, 23K19505, 23K18313, 23KK0153, 24K22144, and 16H06279 (PAGS)), and Japan Agency for Medical Research and Development (AMED) (grant nos. JP23ym0126809 and JP24ym0126809). Partial support was offered by the Princess Takamatsu Cancer Research Fund (2023) to H.I. and Suzuken Memorial Foundation, Japan (2024), to S.M.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors are grateful to all the lab members and Sarah Rennie, from the Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, DK2200 Copenhagen N, Denmark, for her advice.

Conflicts of Interest

Partial institutional endowments were received from Hirotsu Bio Science Inc. (Tokyo, Japan), Kinshu-kai Medical Corporation (Osaka, Japan), Kyowa-kai Medical Corporation (Kawanishi, Hyogo and Osaka, Japan), IDEA Consultants Inc. (Tokyo, Japan), and Unitech Co., Ltd. (Chiba, Japan). A.H.A., H.H., and E.d.L. are employees of Hirotsu Bio Science Inc. T.H. (Takaaki Hirotsu) is CEO of Hirotsu Bio Science Inc. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. Chigira, M. Selfish cells in altruistic cell society—A theoretical oncology. Int. J. Oncol. 1993, 3, 441–455. [Google Scholar] [CrossRef] [PubMed]
  2. Axelrod, R.; Axelrod, D.E.; Pienta, K.J. Evolution of cooperation among tumor cells. Proc. Natl. Acad. Sci. USA 2006, 103, 13474–13479. [Google Scholar] [CrossRef] [PubMed]
  3. Brutovský, B. Scales of Cancer Evolution: Selfish Genome or Cooperating Cells? Cancers 2022, 14, 3253. [Google Scholar] [CrossRef] [PubMed]
  4. Nong, S.; Han, X.; Xiang, Y.; Qian, Y.; Wei, Y.; Zhang, T.; Tian, K.; Shen, K.; Yang, J.; Ma, X. Metabolic reprogramming in cancer: Mechanisms and therapeutics. Medcomm 2023, 4, e218. [Google Scholar] [CrossRef]
  5. Schiliro, C.; Firestein, B.L. Mechanisms of Metabolic Reprogramming in Cancer Cells Supporting Enhanced Growth and Proliferation. Cells 2021, 10, 1056. [Google Scholar] [CrossRef]
  6. Chen, Z.; Han, F.; Du, Y.; Shi, H.; Zhou, W. Hypoxic microenvironment in cancer: Molecular mechanisms and therapeutic interventions. Signal Transduct. Target. Ther. 2023, 8, 70. [Google Scholar] [CrossRef]
  7. Zhang, H.; Li, S.; Wang, D.; Liu, S.; Xiao, T.; Gu, W.; Yang, H.; Wang, H.; Yang, M.; Chen, P.; et al. Metabolic reprogramming and immune evasion: The interplay in the tumor microenvironment. Biomark. Res. 2024, 12, 96. [Google Scholar] [CrossRef]
  8. Crosby, D.; Bhatia, S.; Brindle, K.M.; Coussens, L.M.; Dive, C.; Emberton, M.; Esener, S.; Fitzgerald, R.C.; Gambhir, S.S.; Kuhn, P.; et al. Early detection of cancer. Science 2022, 375, eaay9040. [Google Scholar] [CrossRef]
  9. Duffy, M.J. Tumor markers in clinical practice: A review focusing on common solid cancers. Med. Princ. Pract. 2013, 22, 4–11. [Google Scholar] [CrossRef]
  10. Prasanth, B.K.; Sawarkar, G.; Dharshini, B.D.; Prasanth, K.; Alkhowaiter, S.S.; Dharshini, D.; Baskaran, A.R. Unlocking Early Cancer Detection: Exploring Biomarkers, Circulating DNA, and Innovative Technological Approaches. Cureus 2023, 15, e51090. [Google Scholar] [CrossRef]
  11. Tenchov, R.; Sapra, A.K.; Sasso, J.; Ralhan, K.; Tummala, A.; Azoulay, N.; Zhou, Q.A. Biomarkers for Early Cancer Detection: A Landscape View of Recent Advancements, Spotlighting Pancreatic and Liver Cancers. ACS Pharmacol. Transl. Sci. 2024, 7, 586–613. [Google Scholar] [CrossRef] [PubMed]
  12. Wang, X.; Tian, L.; Lu, J.; Ng, I.O.-L. Exosomes and cancer—Diagnostic and prognostic biomarkers and therapeutic vehicle. Oncogenesis 2022, 11, 54. [Google Scholar] [CrossRef] [PubMed]
  13. Mathieu, M.; Martin-Jaular, L.; Lavieu, G.; Théry, C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat. Cell Biol. 2019, 21, 9–17. [Google Scholar] [CrossRef]
  14. Liu, J.; Zhang, Z.; Zhang, W.; Niraj, M.; Yang, F.; Guo, C.; Shen, L.; Xu, T.; Liu, S.; Zhang, J.; et al. Urinary exosomes: Potential diagnostic markers and application in bladder cancer. Heliyon 2024, 10, e32621. [Google Scholar] [CrossRef] [PubMed]
  15. Yu, D.; Li, Y.; Wang, M.; Gu, J.; Xu, W.; Cai, H.; Fang, X.; Zhang, X. Exosomes as a new frontier of cancer liquid biopsy. Mol. Cancer 2022, 21, 56. [Google Scholar] [CrossRef]
  16. Calero, J.B.; López, M.A.C.; Monge, P.G.C.; Portillo, J.D.; García, A.B.; Roldán, F.N. Analysis of blood markers for early colorectal cancer diagnosis. J. Gastrointest. Oncol. 2022, 13, 2259–2268. [Google Scholar] [CrossRef]
  17. Bayo, J.; Castaño, M.A.; Rivera, F.; Navarro, F. Analysis of blood markers for early breast cancer diagnosis. Clin. Transl. Oncol. 2018, 20, 467–475. [Google Scholar] [CrossRef]
  18. Ma, L.; Guo, H.; Zhao, Y.; Liu, Z.; Wang, C.; Bu, J.; Sun, T.; Wei, J. Liquid biopsy in cancer: Current status, challenges and future prospects. Signal Transduct. Target. Ther. 2024, 9, 336. [Google Scholar] [CrossRef]
  19. Mo, J.-L.; Li, X.; Lei, L.; Peng, J.; Liang, X.-S.; Zhou, H.-H.; Liu, Z.-Q.; Hong, W.-X.; Yin, J.-Y. A machine learning model revealed that exosome small RNAs may participate in the development of breast cancer through the chemokine signaling pathway. BMC Cancer 2024, 24, 1435. [Google Scholar] [CrossRef]
  20. Qiao, P.; Du, H.; Guo, X.; Yu, M.; Zhang, C.; Shi, Y. Serum exosomal miR-200c is a potential diagnostic biomarker for breast cancer. Biomarkers 2024, 29, 419–426. [Google Scholar] [CrossRef]
  21. Yang, L.M.; Fan, J.M.; Dong, C.M.; Wang, X.M.; Ma, B.M. Correlative expression of exosomal miRNAs in chemotherapy resistance of triple-negative breast cancer: An observational study. Medicine 2024, 103, e38549. [Google Scholar] [CrossRef] [PubMed]
  22. Wang, Y.; Gao, W.; Feng, B.; Shen, H.; Chen, X.; Yu, S. Surface protein analysis of breast cancer exosomes using visualized strategy on centrifugal disk chip. Int. J. Biol. Macromol. 2024, 280, 135651. [Google Scholar] [CrossRef] [PubMed]
  23. Inubushi, S.; Kunihisa, T.; Kuniyasu, M.; Inoue, S.; Yamamoto, M.; Yamashita, Y.; Miki, M.; Mizumoto, S.; Baba, M.; Hoffman, R.M.; et al. Serum Exosomes Expressing CD9, CD63 and HER2 From Breast-Cancer Patients Decreased After Surgery of the Primary Tumor: A Potential Biomarker of Tumor Burden. Cancer Genom. Proteom. 2024, 21, 580–584. [Google Scholar] [CrossRef] [PubMed]
  24. Park, M.; Lee, C.-H.; Noh, H.; Kang, G.; Lee, J.; Bae, J.-H.; Moon, H.; Park, J.; Kong, S.; Baek, M.-C.; et al. High-precision extracellular-vesicle isolation-analysis integrated platform for rapid cancer diagnosis directly from blood plasma. Biosens. Bioelectron. 2025, 267, 116863. [Google Scholar] [CrossRef]
  25. Hassanin, A.A.I.; Ramos, K.S. Circulating Exosomal miRNA Profiles in Non-Small Cell Lung Cancers. Cells 2024, 13, 1562. [Google Scholar] [CrossRef]
  26. Bafiti, V.; Thanou, E.; Ouzounis, S.; Kotsakis, A.; Georgoulias, V.; Lianidou, E.; Katsila, T.; Markou, A. Profiling Plasma Extracellular Vesicle Metabotypes and miRNAs: An Unobserved Clue for Predicting Relapse in Patients with Early-Stage NSCLC. Cancers 2024, 16, 3729. [Google Scholar] [CrossRef]
  27. Li, L.; Zhang, Z.; Xu, W.; Wang, J.; Feng, X. The diagnostic value of serum exosomal SNORD116 and SNORA21 for NSCLC patients. Clin. Transl. Oncol. 2024, 27, 650–659. [Google Scholar] [CrossRef]
  28. Zhu, W.; Zhang, H.; Tang, L.; Fang, K.; Lin, N.; Huang, Y.; Zhang, Y.; Le, H. Identification of a Plasma Exosomal lncRNA- and circRNA-Based ceRNA Regulatory Network in Patients With Lung Adenocarcinoma. Clin. Respir. J. 2024, 18, e70026. [Google Scholar] [CrossRef]
  29. Shah, K.A.; Rawal, R.M. A novel algorithm to differentiate between primary lung tumors and distant liver metastasis in lung cancers using an exosome based multi gene biomarker panel. Sci. Rep. 2024, 14, 13769. [Google Scholar] [CrossRef]
  30. Jahani, M.M.; Mashayekhi, P.; Omrani, M.D.; Khosravi, A.; Dehghanifard, A.; Manjiri, S.A.; Zahraie, M.; Mabani, M.; Seifi, S.; Salimi, B.; et al. Assessing the Sensitivity of Nested PCR Followed by Direct Sequencing on Exosomal DNA for EGFR Mutation Detection in NSCL. Iran. Biomed. J. 2024, 28, 208–215. [Google Scholar] [CrossRef]
  31. Amato, L.; De Rosa, C.; De Rosa, V.; Sheikhhossein, H.H.; Ariano, A.; Franco, P.; Nele, V.; Capaldo, S.; Di Guida, G.; Sepe, F.; et al. Immune-Cell-Derived Exosomes as a Potential Novel Tool to Investigate Immune Responsiveness in SCLC Patients: A Proof-of-Concept Study. Cancers 2024, 16, 3151. [Google Scholar] [CrossRef]
  32. Lu, D.; Shangguan, Z.; Su, Z.; Lin, C.; Huang, Z.; Xie, H. Artificial intelligence-based plasma exosome label-free SERS profiling strategy for early lung cancer detection. Anal. Bioanal. Chem. 2024, 416, 5089–5096. [Google Scholar] [CrossRef]
  33. Hsu, C.-C.; Su, Y.; Rittenhouse-Olson, K.; Attwood, K.M.; Mojica, W.; Reid, M.E.; Dy, G.K.; Wu, Y. Exosomal Thomsen–Friedenreich Glycoantigen: A New Liquid Biopsy Biomarker for Lung and Breast Cancer Diagnoses. Cancer Res. Commun. 2024, 4, 1933–1945. [Google Scholar] [CrossRef] [PubMed]
  34. He, X.; Chen, L.; Di, Y.; Li, W.; Zhang, X.; Bai, Z.; Wang, Z.; Liu, S.; Corpe, C.; Wang, J. Plasma-derived exosomal long noncoding RNAs of pancreatic cancer patients as novel blood-based biomarkers of disease. BMC Cancer 2024, 24, 961. [Google Scholar] [CrossRef]
  35. Ueda, H.; Takahashi, H.; Kobayashi, S.; Kubo, M.; Sasaki, K.; Iwagami, Y.; Yamada, D.; Tomimaru, Y.; Asaoka, T.; Noda, T.; et al. miR-6855-5p Enhances Radioresistance and Promotes Migration of Pancreatic Cancer by Inducing Epithelial-Mesenchymal Transition via Suppressing FOXA1: Potential of Plasma Exosomal miR-6855-5p as an Indicator of Radiosensitivity in Patients with Pancreatic Cancer. Ann. Surg. Oncol. 2025, 32, 720–735. [Google Scholar] [CrossRef] [PubMed]
  36. Liu, L.; Cai, J.; Yang, K.; Sun, B.; Liu, W.; Li, Y.; Hu, H. Molecular beacon-peptide probe based double recycling amplification for multiplexed detection of serum exosomal microRNAs. Anal. Methods 2024, 16, 5202–5211. [Google Scholar] [CrossRef] [PubMed]
  37. Ueda, H.; Takahashi, H.; Sakaniwa, R.; Kitamura, T.; Kobayashi, S.; Tomimaru, Y.; Kubo, M.; Sasaki, K.; Iwagami, Y.; Yamada, D.; et al. Preoperative treatment response prediction for pancreatic cancer by multiple microRNAs in plasma exosomes: Optimization using machine learning and network analysis. Pancreatology 2024, 24, 1097–1106. [Google Scholar] [CrossRef]
  38. Wang, Y.; Liang, C.; Liu, X.; Cheng, S.-Q. A novel tumor-derived exosomal gene signature predicts prognosis in patients with pancreatic cancer. Transl. Cancer Res. 2024, 13, 4324–4340. [Google Scholar] [CrossRef]
  39. Rizk, N.I.; Kassem, D.H.; Abulsoud, A.I.; AbdelHalim, S.; Yasser, M.B.; Kamal, M.M.; Hamdy, N.M. Revealing the role of serum exosomal novel long non-coding RNA NAMPT-AS as a promising diagnostic/prognostic biomarker in colorectal cancer patients. Life Sci. 2024, 352, 122850. [Google Scholar] [CrossRef]
  40. Min, L.; Bu, F.; Meng, J.; Liu, X.; Guo, Q.; Zhao, L.; Li, Z.; Li, X.; Zhu, S.; Zhang, S. Circulating small extracellular vesicle RNA profiling for the detection of T1a stage colorectal cancer and precancerous advanced adenoma. eLife 2024, 12, RP88675. [Google Scholar] [CrossRef]
  41. Zhang, H.; Tang, Y.; Zhou, Y.; Wang, Y.; Si, H.; Li, L.; Tang, B. DNAzyme-RCA-based colorimetric and lateral flow dipstick assays for the point-of-care testing of exosomal m5C-miRNA-21. Chem. Sci. 2024, 15, 9345–9352. [Google Scholar] [CrossRef]
  42. Yin, H.; Xie, J.; Xing, S.; Lu, X.; Yu, Y.; Ren, Y.; Tao, J.; He, G.; Zhang, L.; Yuan, X.; et al. Machine learning-based analysis identifies and validates serum exosomal proteomic signatures for the diagnosis of colorectal cancer. Cell Rep. Med. 2024, 5, 101689. [Google Scholar] [CrossRef] [PubMed]
  43. Wada, Y.; Nishi, M.; Yoshikawa, K.; Takasu, C.; Tokunaga, T.; Nakao, T.; Kashihara, H.; Yoshimoto, T.; Shimada, M. Circulating Exosomal MicroRNA Signature Predicts Peritoneal Metastasis in Patients with Advanced Gastric Cancer. Ann. Surg. Oncol. 2024, 31, 5997–6006. [Google Scholar] [CrossRef] [PubMed]
  44. Liu, X.; Zhang, J.; Chen, Z.; He, X.; Yan, C.; Lv, H.; Chen, Z.; Liu, Y.; Wang, L.; Song, C. Branched hybridization chain reaction and tetrahedral DNA-based trivalent aptamer powered SERS sensor for ultra-highly sensitive detection of cancer-derived exosomes. Biosens. Bioelectron. 2025, 267, 116737. [Google Scholar] [CrossRef] [PubMed]
  45. Wang, C.; Yang, G.; Feng, G.; Deng, C.; Zhang, Q.; Chen, S. Developing an advanced diagnostic model for hepatocellular carcinoma through multi-omics integration leveraging diverse cell-death patterns. Front. Immunol. 2024, 15, 1410603. [Google Scholar] [CrossRef] [PubMed]
  46. Resch, U.; Hackl, H.; Pereyra, D.; Santol, J.; Brunnthaler, L.; Probst, J.; Jankoschek, A.S.; Aiad, M.; Nolte, H.; Krueger, M.; et al. SILAC-Based Characterization of Plasma-Derived Extracellular Vesicles in Patients Undergoing Partial Hepatectomy. Int. J. Mol. Sci. 2024, 25, 10685. [Google Scholar] [CrossRef]
  47. Arima, J.; Yoshino, H.; Fukumoto, W.; Kawahara, I.; Saito, S.; Li, G.; Fukuda, I.; Iizasa, S.; Mitsuke, A.; Sakaguchi, T.; et al. LncRNA BCYRN1 as a Potential Therapeutic Target and Diagnostic Marker in Serum Exosomes in Bladder Cancer. Int. J. Mol. Sci. 2024, 25, 5955. [Google Scholar] [CrossRef]
  48. Solheim, E.T.; Thomsen, L.C.V.; Bjørge, L.; Anandan, S.; Peter, E.; Desestret, V.; Totland, C.; Vedeler, C.A. Altered exosomal miRNA profiles in patients with paraneoplastic cerebellar degeneration. Ann. Clin. Transl. Neurol. 2024, 11, 3255–3266. [Google Scholar] [CrossRef]
  49. Chen, Y.; Cui, F.; Wu, X.; Zhao, W.; Xia, Q. The expression and clinical significance of serum exosomal-long non-coding RNA DLEU1 in patients with cervical cancer. Ann. Med. 2025, 57, 2442537. [Google Scholar] [CrossRef]
  50. Supradit, K.; Wongprasert, K.; Tangphatsornruang, S.; Yoocha, T.; Sonthirod, C.; Pootakham, W.; Thitapakorn, V.; Butthongkomvong, K.; Phanaksri, T.; Kunjantarachot, A.; et al. microRNA profiling of exosomes derived from plasma and their potential as biomarkers for Opisthorchis viverrini-associated cholangiocarcinoma. Acta Trop. 2024, 258, 107362. [Google Scholar] [CrossRef]
  51. Bocchetti, M.; Luce, A.; Iannarone, C.; Pasquale, L.S.; Falco, M.; Tammaro, C.; Abate, M.; Ferraro, M.G.; Addeo, R.; Ricciardiello, F.; et al. Exosomes multiplex profiling, a promising strategy for early diagnosis of laryngeal cancer. J. Transl. Med. 2024, 22, 582. [Google Scholar] [CrossRef] [PubMed]
  52. Zhou, Z.; Zhang, D.; Wang, Y.; Liu, C.; Wang, L.; Yuan, Y.; Xu, X.; Jiang, Y. Urinary exosomes: A promising biomarker of drug-induced nephrotoxicity. Front. Med. 2023, 10, 1251839. [Google Scholar] [CrossRef] [PubMed]
  53. Gao, Y.; Wang, X.; Luo, H.; Chen, C.; Li, J.; Sun, R.; Li, D.; Sun, Z. Exosomal Long Non-Coding Ribonucleic Acid Ribonuclease Component of Mitochondrial Ribonucleic Acid Processing Endoribonuclease Is Defined as a Potential Non-Invasive Diagnostic Biomarker for Bladder Cancer and Facilitates Tumorigenesis via the miR-206/G6PD Axis. Cancers 2023, 15, 5305. [Google Scholar] [CrossRef] [PubMed]
  54. Gao, Y.; Zhang, X.; Wang, X.; Sun, R.; Li, Y.; Li, J.; Quan, W.; Yao, Y.; Hou, Y.; Li, D.; et al. The clinical value of rapidly detecting urinary exosomal lncRNA RMRP in bladder cancer with an RT-RAA-CRISPR/Cas12a method. Clin. Chim. Acta 2024, 562, 119855. [Google Scholar] [CrossRef]
  55. Qiu, T.; Xue, M.; Li, X.; Li, F.; Liu, S.; Yao, C.; Chen, W. Comparative evaluation of long non-coding RNA-based biomarkers in the urinary sediment and urinary exosomes for non-invasive diagnosis of bladder cancer. Mol. Omics 2022, 18, 938–947. [Google Scholar] [CrossRef]
  56. Liu, C.; Xu, P.; Shao, S.; Wang, F.; Zheng, Z.; Li, S.; Liu, W.; Li, G. The value of urinary exosomal lncRNA SNHG16 as a diagnostic biomarker for bladder cancer. Mol. Biol. Rep. 2023, 50, 8297–8304. [Google Scholar] [CrossRef]
  57. Yang, F.; Tian, C.; Zhou, L.; Guan, T.; Chen, G.; Zheng, Y.; Cao, Z. The value of urinary exosomal microRNA-21 in the early diagnosis and prognosis of bladder cancer. Kaohsiung J. Med. Sci. 2024, 40, 660–670. [Google Scholar] [CrossRef]
  58. Strømme, O.; Heck, K.A.; Brede, G.; Lindholm, H.T.; Otterlei, M.; Arum, C.-J. tRNA-Derived Fragments as Biomarkers in Bladder Cancer. Cancers 2024, 16, 1588. [Google Scholar] [CrossRef]
  59. Hou, J.; Huang, H.; Xie, J.; Yu, W.; Hao, H.; Li, H. KLHDC7B as a novel diagnostic biomarker in urine exosomal mRNA promotes bladder urothelial carcinoma cell proliferation and migration, inhibits apoptosis. Mol. Carcinog. 2024, 63, 286–300. [Google Scholar] [CrossRef]
  60. Murakami, T.; Minami, K.; Harabayashi, T.; Maruyama, S.; Takada, N.; Kashiwagi, A.; Miyata, H.; Sato, Y.; Matsumoto, R.; Kikuchi, H.; et al. Cross-sectional and longitudinal analyses of urinary extracellular vesicle mRNA markers in urothelial bladder cancer patients. Sci. Rep. 2024, 14, 6801. [Google Scholar] [CrossRef]
  61. Wang, X.; Song, D.; Zhu, B.; Jin, Y.; Cai, C.; Wang, Z. Urinary exosomal mRNA as a biomarker for the diagnosis of bladder cancer. AntiCancer Drugs 2024, 35, 362–370. [Google Scholar] [CrossRef] [PubMed]
  62. Cao, Y.; Feng, J.; Zhang, Q.; Deng, C.; Yang, C.; Li, Y. Magnetic 3D macroporous MOF oriented urinary exosome metabolomics for early diagnosis of bladder cancer. J. Nanobiotechnol. 2024, 22, 671. [Google Scholar] [CrossRef] [PubMed]
  63. Chen, H.; Qi, Y.; Yang, C.; Tai, Q.; Zhang, M.; Shen, X.-Z.; Deng, C.; Guo, J.; Jiang, S.; Sun, N. Heterogeneous MXene Hybrid-Oriented Exosome Isolation and Metabolic Profiling for Early Screening, Subtyping and Follow-up Evaluation of Bladder Cancer. ACS Nano 2023, 17, 23924–23935. [Google Scholar] [CrossRef]
  64. Chen, Y.; Wu, Y.; Li, J.; Deng, C.; Sun, N. Resol/triblock copolymer composite-guided smart fabrication of carbonized mesopores for efficiently decoding exosomal glycans. Microchim. Acta 2023, 190, 319. [Google Scholar] [CrossRef]
  65. Steiner, L.; Eldh, M.; Offens, A.; Veerman, R.E.; Johansson, M.; Hemdan, T.; Netterling, H.; Huge, Y.; Aljabery, F.A.-S.; Alamdari, F.; et al. Protein profile in urinary extracellular vesicles is a marker of malignancy and correlates with muscle invasiveness in urinary bladder cancer. Cancer Lett. 2025, 609, 217352. [Google Scholar] [CrossRef]
  66. Li, B.; Kugeratski, F.G.; Kalluri, R. A novel machine learning algorithm selects proteome signature to specifically identify cancer exosomes. eLife 2024, 12, RP90390. [Google Scholar] [CrossRef] [PubMed]
  67. Li, Y.; Tang, X.; Deng, R.; Feng, L.; Xie, S.; Chen, M.; Zheng, J.; Chang, K. Dumbbell Dual-Hairpin Triggered DNA Nanonet Assembly for Cascade-Amplified Sensing of Exosomal MicroRNA. ACS Omega 2024, 9, 19723–19731. [Google Scholar] [CrossRef]
  68. Chiou, Y.-E.; Yu, K.-J.; Pang, S.-N.; Yang, Y.-L.; Pang, S.-T.; Weng, W.-H. A Highly Sensitive Urinary Exosomal miRNAs Biosensor Applied to Evaluation of Prostate Cancer Progression. Bioengineering 2022, 9, 803. [Google Scholar] [CrossRef]
  69. Yu, J.; Yu, C.; Jiang, K.; Yang, G.; Yang, S.; Tan, S.; Li, T.; Liang, H.; He, Q.; Wei, F.; et al. Unveiling potential: Urinary exosomal mRNAs as non-invasive biomarkers for early prostate cancer diagnosis. BMC Urol. 2024, 24, 163. [Google Scholar] [CrossRef]
  70. Choi, J.Y.; Park, S.; Shim, J.S.; Park, H.J.; Kuh, S.U.; Jeong, Y.; Park, M.G.; Noh, T.I.; Yoon, S.G.; Park, Y.M.; et al. Explainable artificial intelligence-driven prostate cancer screening using exosomal multi-marker based dual-gate FET biosensor. Biosens. Bioelectron. 2025, 267, 116773. [Google Scholar] [CrossRef]
  71. Wang, C.-B.; Chen, S.-H.; Zhao, L.; Jin, X.; Chen, X.; Ji, J.; Mo, Z.-N.; Wang, F.-B. Urine-derived exosomal PSMA is a promising diagnostic biomarker for the detection of prostate cancer on initial biopsy. Clin. Transl. Oncol. 2023, 25, 758–767. [Google Scholar] [CrossRef] [PubMed]
  72. Qin, X.; Niu, R.; Tan, Y.; Huang, Y.; Ren, W.; Zhou, W.; Wu, H.; Zhang, J.; Xu, M.; Zhou, X.; et al. Exosomal PSM-E inhibits macrophage M2 polarization to suppress prostate cancer metastasis through the RACK1 signaling axis. Biomark. Res. 2024, 12, 138. [Google Scholar] [CrossRef]
  73. Wei, C.; Chen, X.; Ji, J.; Xu, Y.; He, X.; Zhang, H.; Mo, Z.; Wang, F. Urinary exosomal prostate-specific antigen is a noninvasive biomarker to detect prostate cancer: Not only old wine in new bottles. Int. J. Cancer 2023, 152, 1719–1727. [Google Scholar] [CrossRef] [PubMed]
  74. Yasui, T.; Natsume, A.; Yanagida, T.; Nagashima, K.; Washio, T.; Ichikawa, Y.; Chattrairat, K.; Naganawa, T.; Iida, M.; Kitano, Y.; et al. Early Cancer Detection via Multi-microRNA Profiling of Urinary Exosomes Captured by Nanowires. Anal. Chem. 2024, 96, 17145–17153. [Google Scholar] [CrossRef]
  75. Jin, S.; Liu, T.; Wang, W.; Li, T.; Liu, Z.; Zhang, M. Lymphocyte migration regulation related proteins in urine exosomes may serve as a potential biomarker for lung cancer diagnosis. BMC Cancer 2023, 23, 1125. [Google Scholar] [CrossRef]
  76. Yoshizawa, N.; Sugimoto, K.; Tameda, M.; Inagaki, Y.; Ikejiri, M.; Inoue, H.; Usui, M.; Ito, M.; Takei, Y. miR-3940-5p/miR-8069 ratio in urine exosomes is a novel diagnostic biomarker for pancreatic ductal adenocarcinoma. Oncol. Lett. 2020, 19, 2677–2684. [Google Scholar] [CrossRef]
  77. Cheng, X.; Yu, W.; Liu, Y.; Jia, S.; Wang, D.; Hu, L. Proteomic Characterization of Urinary Exosomes with Pancreatic Cancer by Phosphatidylserine Imprinted Polymer Enrichment and Mass Spectrometry Analysis. J. Proteome Res. 2024, 24, 111–120. [Google Scholar] [CrossRef]
  78. Wu, D.; Xie, W.; Chen, X.; Sun, H. LRG1 Is Involved in the Progression of Ovarian Cancer via Modulating FAK/AKT Signaling Pathway. Front. Biosci. 2023, 28, 101. [Google Scholar] [CrossRef]
  79. Shnaider, P.V.; Petrushanko, I.Y.; Aleshikova, O.I.; Babaeva, N.A.; Ashrafyan, L.A.; Borovkova, E.I.; Dobrokhotova, J.E.; Borovkov, I.M.; Shender, V.O.; Khomyakova, E. Expression level of CD117 (KIT) on ovarian cancer extracellular vesicles correlates with tumor aggressiveness. Front. Cell Dev. Biol. 2023, 11, 1057484. [Google Scholar] [CrossRef]
  80. Park, C.; Chung, S.; Kim, H.; Kim, N.; Son, H.Y.; Kim, R.; Lee, S.; Park, G.; Rho, H.W.; Park, M.; et al. All-in-One Fusogenic Nanoreactor for the Rapid Detection of Exosomal MicroRNAs for Breast Cancer Diagnosis. ACS Nano 2024, 18, 26297–26314. [Google Scholar] [CrossRef]
  81. Qi, G.; Diao, X.; Tian, Y.; Sun, D.; Jin, Y. Electroactivated SERS Nanoplatform for Rapid and Sensitive Detection and Identification of Tumor-Derived Exosome miRNA. Anal. Chem. 2024, 96, 18519–18527. [Google Scholar] [CrossRef] [PubMed]
  82. Walter-Rodriguez, B.; Ricketts, C.J.; Linehan, W.M.; Merino, M.J. Evaluating the Urinary Exosome microRNA Profile of von Hippel Lindau Syndrome Patients with Clear Cell Renal Cell Carcinoma. Genes 2024, 15, 905. [Google Scholar] [CrossRef]
  83. Grützmann, K.; Salomo, K.; Krüger, A.; Lohse-Fischer, A.; Erdmann, K.; Seifert, M.; Baretton, G.; Aust, D.; William, D.; Schröck, E.; et al. Identification of novel snoRNA-based biomarkers for clear cell renal cell carcinoma from urine-derived extracellular vesicles. Biol. Direct 2024, 19, 38. [Google Scholar] [CrossRef]
  84. Sharma, D.; Singh, A.; Wilson, C.; Swaroop, P.; Kumar, S.; Yadav, D.K.; Jain, V.; Agarwala, S.; Husain, M.; Sharawat, S.K. Exosomal long non-coding RNA MALAT1: A candidate of liquid biopsy in monitoring of Wilms’ tumor. Pediatr. Surg. Int. 2024, 40, 57. [Google Scholar] [CrossRef] [PubMed]
  85. Woo, H.; Park, J.; Kim, K.H.; Ku, J.Y.; Ha, H.K.; Cho, Y. Alix-normalized exosomal programmed death-ligand 1 analysis in urine enables precision monitoring of urothelial cancer. Cancer Sci. 2024, 115, 1602–1610. [Google Scholar] [CrossRef] [PubMed]
  86. Ma, L.; Yu, H.; Zhu, Y.; Xu, K.; Zhao, A.; Ding, L.; Gao, H.; Zhang, M. Isolation and proteomic profiling of urinary exosomes from patients with colorectal cancer. Proteome Sci. 2023, 21, 3. [Google Scholar] [CrossRef]
  87. Wang, C.-Y.; Shih, S.-R.; Chen, K.-Y.; Huang, P.-J. Urinary Exosomal Tissue TIMP and Angiopoietin-1 Are Preoperative Novel Biomarkers of Well-Differentiated Thyroid Cancer. Biomedicines 2022, 11, 24. [Google Scholar] [CrossRef]
  88. Feng, X.; Jia, S.; Ali, M.M.; Zhang, G.; Li, D.; Tao, W.A.; Hu, L. Proteomic Discovery and Array-Based Validation of Biomarkers from Urinary Exosome by Supramolecular Probe. J. Proteome Res. 2023, 22, 2516–2524. [Google Scholar] [CrossRef]
  89. Chen, C.; Demirkhanyan, L.; Gondi, C.S. The Multifaceted Role of miR-21 in Pancreatic Cancers. Cells 2024, 13, 948. [Google Scholar] [CrossRef]
  90. di Luccio, E.; Morishita, M.; Hirotsu, T. C. elegans as a Powerful Tool for Cancer Screening. Biomedicines 2022, 10, 2371. [Google Scholar] [CrossRef]
  91. Inaba, S.; Shimozono, N.; Yabuki, H.; Enomoto, M.; Morishita, M.; Hirotsu, T.; di Luccio, E. Accuracy evaluation of the C. elegans cancer test (N-NOSE) using a new combined method. Cancer Treat. Res. Commun. 2021, 27, 100370. [Google Scholar] [CrossRef] [PubMed]
  92. Hara, T.; Meng, S.; Arao, Y.; Saito, Y.; Inoue, K.; Rennie, S.; Ofusa, K.; Doki, Y.; Eguchi, H.; Kitagawa, T.; et al. Recent advances in noncoding RNA modifications of gastrointestinal cancer. Cancer Sci. 2024, 116, 8–20. [Google Scholar] [CrossRef] [PubMed]
  93. Hara, T.; Meng, S.; Sato, H.; Tatekawa, S.; Sasaki, K.; Takeda, Y.; Tsuji, Y.; Arao, Y.; Ofusa, K.; Kitagawa, T.; et al. High N6-methyladenosine-activated TCEAL8 mRNA is a novel pancreatic cancer marker. Cancer Sci. 2024, 115, 2360–2370. [Google Scholar] [CrossRef]
  94. Konno, M.; Koseki, J.; Asai, A.; Yamagata, A.; Shimamura, T.; Motooka, D.; Okuzaki, D.; Kawamoto, K.; Mizushima, T.; Eguchi, H.; et al. Distinct methylation levels of mature microRNAs in gastrointestinal cancers. Nat. Commun. 2019, 10, 3888. [Google Scholar] [CrossRef] [PubMed]
  95. Tornesello, A.L.; Cerasuolo, A.; Starita, N.; Amiranda, S.; Cimmino, T.P.; Bonelli, P.; Tuccillo, F.M.; Buonaguro, F.M.; Buonaguro, L.; Tornesello, M.L. Emerging role of endogenous peptides encoded by non-coding RNAs in cancer biology. Non Coding RNA Res. 2025, 10, 231–241. [Google Scholar] [CrossRef]
  96. Hara, T.; Meng, S.; Tsuji, Y.; Arao, Y.; Saito, Y.; Sato, H.; Motooka, D.; Uchida, S.; Ishii, H. RN7SL1 may be translated under oncogenic conditions. Proc. Natl. Acad. Sci. USA 2024, 121, e2312322121. [Google Scholar] [CrossRef]
  97. Chothani, S.P.; Adami, E.; Widjaja, A.A.; Langley, S.R.; Viswanathan, S.; Pua, C.J.; Zhihao, N.T.; Harmston, N.; D’agostino, G.; Whiffin, N.; et al. A high-resolution map of human RNA translation. Mol. Cell 2022, 82, 2885–2899.e8. [Google Scholar] [CrossRef]
Table 1. Cancer marker candidates in blood exosomes.
Table 1. Cancer marker candidates in blood exosomes.
Candidate Cancer MarkersMolecular TypesIsolation MethodsAnalysis MethodsCancer TypesReferences
piR-36,340, piR-33,161, miR-484, miR-548ah-5p, miR-4282, and miR-6853-3ppiRNA or miRNAexoEasy maxi kit
(QIAGEN, Hilden, Germany)
RT-qPCRBreast[19]
miR-200cmiRNAHhieff® quick exosome isolation kit
(YEASEN, Shanghai, China)
RT-qPCRBreast[20]
miR-6831-5PmiRNAExosome rapid extraction reagent kit
(YEASEN, Shanghai, China)
RT-qPCRBreast[21]
CEA, CA125, and EGFRGlycoprotein/proteinIntegrated centrifugal disk chipELISABreast[22]
CD9 and Her2ProteinAntibody-conjugated diskELISABreast[23]
PD-L1, EpCAM, and EGFRProteinDEP–ELISA chipELISABreast, colon, and lung[24]
miR-21-5p, miR-126-3p, miR-210-3p, miR-221-3p, Let-7b-5p, miR-146a-5p, miR-222-3p, and miR-9-5pmiRNAexoEasy maxi kit
(QIAGEN, Hilden, Germany)
RT-qPCRLung[25]
miR-29a-3pmiRNAMacherey-Nagel™ exosome precipitation solution for serum/plasma
(Fisher Scientific, Waltham, MA, USA)
RT-qPCRLung[26]
SNORD116 and SNORA21snRNAUltracentrifugationMicroarray Lung[27]
circ-0033861, circ-0043273, and circ-0011959circRNAUltracentrifugationMicroarray Lung[28]
CXCL12, TFBR2, CD44v6, HIF1A, and KRT7mRNATotal exosome isolation kit
(Invitogen, Carlsbad, CA, USA)
RT-qPCRLung[29]
EGFR mutationsDNAXCFTM Exosomal DNA isolation kit
(System Biosciences, Palo Alto, CA, USA)
RT-qPCRLung[30]
c-Myc, Snail, MAVS, and STINGProteinUltracentrifugationWestern blotLung[31]
Raman spectrumExosomeUltracentrifugationRaman spectrumLung[32]
TF-Ag-αGlycoproteinUltracentrifugationSurface plasmon resonance (SPR)Lung and breast[33]
LINC01268, LINC02802, AC124854.1, and AL132657.1lncRNAexoRNeasy midi kit
(QIAGEN, Hilden, Germany)
RNA-seqPancrea[34]
miR-6855-5pmiRNAqEVTM original 35 nm size exclusion column
(Izon Science, Christchurch, New Zealand)
RNA-seqPancrea[35]
miR-21, miR-191, and miR-451amiRNAExoQuick
(System Biosciences, Palo Alto, CA, USA)
RT-qPCRPancrea[36]
miR-6891-5p, miR-6732-5p, and miR-1234-3pmiRNAqEVTM original 35 nm size exclusion column
(Izon Science, Christchurch, New Zealand)
MicroarrayPancrea[37]
ARNTL2, FHL2, KRT19, MMP1, CDCA5, and KIF11mRNAExoRBase 2.0 databaseRNA-seqPancrea[38]
lncRNA NAMPT-ASlncRNAExosome isolation kit
(Miltenyi Biotec, Bergisch Gladbach, Germany)
RNA-seqColon[39]
miR-425–5 p, Let-7f-5p, C19orf43, TOP1, PPDPF, LNC-EV-9572, lnc-MKRN2-42:1, HIST2H2AA4, and MT-ND2miRNA, mRNA, and lncRNAUltracentrifugationRNA-seqColon[40]
5-methylcytosine miRNA-21Modified miRNAUltracentrifugationDNAzyme-triggered rolling circle amplificationColon[41]
PF4 and AACTProteinUltracentrifugationELISAColon[42]
miR-21-5p, miR-320, miR-191-5p, and miR-451miRNATotal exosome isolation kit
(Thermo Fisher Scientific, Waltham, MA, USA)
RNA-seqGastric[43]
MUC1ProteinUltracentrifugationRaman spectrumGastric[44]
TRIB3 and NQO1mRNAUltracentrifugationRNA-seqHepatocellular carcinoma[45]
DRAP1, GGCT, NSUN2, RAB13, PPCS, SDHA, CTSB, TIMM44, VTN, KATNAL2, and RPL27AProteinUltracentrifugationMass spectrometryHepatocellular carcinoma[46]
lncRNA brain cytoplasmic RNA 1 (BCYRN1)lncRNAexoEasy maxi Kit
(QIAGEN, Hilden, Germany)
RNA-seqBladder[47]
miR-483-5p, miR-4488, and miR-200c-3pmiRNAexoRNeasy midi kit
(QIAGEN, Hilden, Germany)
RNA-seqOvarian[48]
lncRNA DLEU1lncRNAUltracentrifugationRT-qPCRCervical[49]
miR-92a-3p, miR-203a-3p, miR-192–5p, miR-223–3p, miR-26a-5p, and miR-194–5pmiRNAexoRNeasy serum/plasma midi kit
(QIAGEN, Hilden, Germany)
RNA-seqCholangiocarcinoma[50]
CD1c, CD2, CD3, CD4, CD11c, CD14, CD20, CD44, CD56, CD105, CD146, and CD209ProteinExosome isolation kit
(Miltenyi Biotec, Bergisch Gladbach, Germany)
Flow cytometryLaryngeal[51]
Table 2. Cancer marker candidates in urine exosomes.
Table 2. Cancer marker candidates in urine exosomes.
Candidate Cancer MarkersMolecular TypesIsolation MethodsAnalysis MethodsCancer TypesReferences
lncRNA RMRPlncRNAExoQuick kit
(Bestbio, Shanghai, China)
RT-qPCRBladder[53,54]
lncRNA RMRP, UCA1, and MALAT1lncRNAUltracentrifugationRT-qPCRBladder[55]
lncRNA SNHG16lncRNAExosome RNA isolation kit
(Rengen Biosciences, Shenyang, China)
RT-qPCRBladder[56]
miR-21miRNAUltracentrifugationRT-qPCRBladder[57]
tRF-16-F1R3WEE, tRF-17-8R6546J, tRF-17-I7XUK8N, tRF-17-D9W1X6K, tRF-18-HR1PF7D2, tRF-18-MBQ4NKDJ, tRF-20-40KK5Y93, tRF-21-86J8WPMNB, tRF-25-7P596VW631, tRF-26-IK9NJ4S2I7D, tRF-27-J87383RPD95, tRF-31-PER8YP9LON4VD, tRF-32-PER8YP9LON4V3, and tRF-38-PNR8YP9LON4VN18Transfer RNA-derived fragmentexoRNeasy kit
(QIAGEN, Hilden, Germany)
RNA-seqBladder[58]
KLHDC7BmRNAUltracentrifugationRT-qPCRBladder[59]
KRT17, GPRC5A, SLC2A1, MDK, and CXCR2mRNAExoComplete tube kit
(Showa Denko Materials, Tokyo, Japan)
RT-qPCRBladder[60]
tmeff1, SDPR, ACBD7, SCG2, and COL6A2mRNAUltracentrifugationRNA-seqBladder[61]
Arachiconic acid, docosahexaenoic acid, docosapentaenoic acid, and retinyl esterMetabolitemagMZIF-8Mass spectrometryBladder[62]
Metabolic profilesMetaboliteMXene@TiO2/Fe3OMass spectrometryBladder[63]
GlycansSugarUltracentrifugationMass spectrometryBladder[64]
MMP12, MMP7, HO-1, IL8, CD5, CCL20, CXCL13, MCP-1, CD8A, and TGF-beta-1ProteinUltracentrifugationFlow cytometryBladder[65]
CD59, CDC42, ITM2B, CD81, PEBP1, VAT1, MYO1D, RAC1, DPP4, RAN, CAPG, PPIA, FOLR1, ANXA3, APOD, ANXA4, and AQP2ProteinPublic exosome proteomics dataMass spectrometryBladder, prostate, renal, lung, cervical, colorectal, esophageal and gastric[66]
miR-141miRNAexoEasy maxi kit
(QIAGEN, Hilden, Germany)
DNA nanonetProstate[67]
miR-451 and miR-21miRNAUrine microRNA purification kit
(Norgen Biotek, Thorold, ON, Canada)
ssDNA sensorProstate[68]
RAB5B, WWP1, HIST2H2BF, ZFY, MARK2, PASK, RBM10, and NRSN2mRNAUltracentrifugationRNA-seqProstate[69]
TMEM256ProteinExoQuick-TC™
(System Biosciences, Palo Alto, CA, USA)
ELISAProstate[70]
PSMAProteinExosome isolation kit
(Wayen, Shanghai, China)
ELISAProstate[71]
PSM-EProteinUltracentrifugationMass spectrometryProstate[72]
Urinary exosomal prostate-specific antigen (UE-PSA)GlycoproteinExosome isolation kit
(Wayen, Shanghai, China)
ELISAProstate[73]
54 miRNAsmiRNAZnO nanowiresMicroarray Lung[74]
CX3CL1,WNK1, GBA, CD58, WASL, LGALS8, MSN, SPNS2, STK10, PKD1, LCK, and GP2ProteinUltracentrifugationMass spectrometryLung[75]
miR-3940-5p/miR-8069miRNAExoQuick-TC
(System Biosciences, Palo Alto, CA, USA)
Microarray Pancrea[76]
SLC9A3R1, SPAG9, and ferritin light chain (FTL)ProteinPhosphatidylserine molecularly imprinted polymersMass spectrometryPancrea[77]
Leucine-rich alpha-2-glycoprotein 1(LRG1)GlycoproteinUltracentrifugationMass spectrometryOvarian[78]
CD117Protein100 kDa Amicon Ultra-15 centrifugal filter units
(Millipore, Boston, MA, USA)
Flow cytometryOvarian[79]
miR-222, miR-200c, and miR-375miRNAmiRNeasy serum/plasma kit
(QIAGEN, Hilden, Germany)
RT-qPCRBreast[80]
Raman spectrumExosomeSurface-enhanced Raman spectroscopy Raman spectrumMCF-7, HeLa, and H8 cell lines[81]
miR-542-5p and miR-320amiRNAExo-Urine™ EV isolation kit
(System Biosciences, Palo Alto, CA, USA)
RNA-seqRenal[82]
SNORD99 and SNORA50CsnRNAmiRCURY exosome cell/urine/CSF kit
(QIAGEN, Hilden, Germany)
RNA-seqRenal[83]
lncRNA MALAT1lncRNATotal exosome isolation kit
(Invitogen, Carlsbad, CA, USA)
RT-qPCRWilms’ tumor (a rare kidney cancer)[84]
PD-L1 and AlixProteinExoDisc
(LabSpinner, San Diego, CA, USA)
ELISAUrothelial[85]
CEACAM7, CEACAM1, CHMP4A, CHMP4B, CHMP2A, CHMP2B, and CHMP1BProteinUltracentrifugationMass spectrometryColorectal[86]
TIMPProteinExoQuick-TC
(System Biosciences, Palo Alto, CA, USA)
Mass spectrometryThyroid[87]
OLFM4, HDGF, and GDF15ProteinArray-based amphiphilic supramolecular probe (ADSP)-modified membranesELISAHepatocellular[88]
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

Hara, T.; Meng, S.; Alshammari, A.H.; Hatakeyama, H.; Arao, Y.; Saito, Y.; Inoue, K.; di Luccio, E.; Vecchione, A.; Hirotsu, T.; et al. Recent Exploration of Solid Cancer Biomarkers Hidden Within Urine or Blood Exosomes That Provide Fundamental Information for Future Cancer Diagnostics. Diagnostics 2025, 15, 628. https://doi.org/10.3390/diagnostics15050628

AMA Style

Hara T, Meng S, Alshammari AH, Hatakeyama H, Arao Y, Saito Y, Inoue K, di Luccio E, Vecchione A, Hirotsu T, et al. Recent Exploration of Solid Cancer Biomarkers Hidden Within Urine or Blood Exosomes That Provide Fundamental Information for Future Cancer Diagnostics. Diagnostics. 2025; 15(5):628. https://doi.org/10.3390/diagnostics15050628

Chicago/Turabian Style

Hara, Tomoaki, Sikun Meng, Aya Hasan Alshammari, Hideyuki Hatakeyama, Yasuko Arao, Yoshiko Saito, Kana Inoue, Eric di Luccio, Andrea Vecchione, Takaaki Hirotsu, and et al. 2025. "Recent Exploration of Solid Cancer Biomarkers Hidden Within Urine or Blood Exosomes That Provide Fundamental Information for Future Cancer Diagnostics" Diagnostics 15, no. 5: 628. https://doi.org/10.3390/diagnostics15050628

APA Style

Hara, T., Meng, S., Alshammari, A. H., Hatakeyama, H., Arao, Y., Saito, Y., Inoue, K., di Luccio, E., Vecchione, A., Hirotsu, T., & Ishii, H. (2025). Recent Exploration of Solid Cancer Biomarkers Hidden Within Urine or Blood Exosomes That Provide Fundamental Information for Future Cancer Diagnostics. Diagnostics, 15(5), 628. https://doi.org/10.3390/diagnostics15050628

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