Abstract
Prostate cancer is one of the most common cancers affecting men, and finding better ways to detect and monitor it remains a top priority in oncology. In recent years, scientists have focused their attention on different classes of extracellular bodies, among them the small ones called exosomes. Exosomes are nanoscale extracellular vesicles (30–200 nm) released into body fluids, where they transport molecular cargo reflective of their cell of origin. Instead of serving as liquid biopsies themselves, exosomes present in accessible fluids such as plasma and urine can be analyzed as part of minimally invasive liquid biopsy strategies without the need for surgery or tissue sampling. In prostate cancer, exosomes are not just passive carriers: they actively influence how cancer grows, spreads, and responds to treatment. Exosomes can be extracted from simple fluid samples, opening the door to faster, safer, and more personalised approaches to diagnosis and care. Exosome content is analysed for the molecular profiling of tumours, including genomics, transcriptomics, proteomics, and metabolomics. This has led to the discovery of new biomarkers that may help detect prostate cancer earlier, predict its aggressiveness, and monitor the effectiveness of treatment. This review synthesizes current multi-omics data on exosomal cargo in prostate cancer, highlighting diagnostic, prognostic, and therapeutic implications as well as existing challenges to clinical translation.
1. Introduction
Prostate cancer (PCa) is one of the most prevalent malignancies affecting men worldwide and represents a major global health concern. It is estimated that there were 1.4 million new cases and 375,000 deaths from PCa worldwide in 2020 [,]. It ranks as the second most frequently diagnosed cancer in men worldwide, yet in Europe, it is the most commonly diagnosed malignancy and the third leading cause of cancer-related death among men [], highlighting the urgent need for improved strategies in early detection, risk stratification, treatment and monitoring strategies. The incidence and mortality rates have been shown to be influenced by various factors, such as age, ethnicity, genetic background and staging [].
Despite advances in screening and treatment, clinical challenges continue, primarily due to the heterogeneous nature of PCa and the limitations of existing diagnostic tools []. For decades, prostate-specific antigen (PSA) testing has formed the basis of screening and early detection of PCa. PSA is an enzyme in the glycoprotein family that is secreted by prostate epithelial cells, and small amounts are normally present in the bloodstream []. However, PSA testing is not cancer-specific: elevated PSA levels can also occur in conditions such as benign prostatic hyperplasia or prostatitis and can be caused by urological procedures []. This lack of specificity can result in false-positive findings, leading to unnecessary biopsies, patient anxiety, and the potential for overtreatment of indolent disease [].
Over the past decade, extracellular vesicles (EVs) have emerged as a potential source of biomarkers for various diseases, among them PCa []. EVs represent a heterogeneous population of membrane-bound vesicles. Exosomes are a subtype of small EVs originating from endosomal multivesicular bodies and should be referred to as ‘small EVs’ when biogenesis cannot be confirmed.
Microenvironmental acidity has been shown to increase the secretion of PSA-positive and CD81-positive small extracellular vesicles, indicating that acidic tumor conditions promote vesicle release. This condition leads to the spill-over of nanovesicles into the blood, where the levels of tumour biomarkers expressed by these nanovesicles may represent a novel, non-invasive clinical tool for the screening and early diagnosis of PCa []. CA IX-positive exosomes were found to be much higher in PCa patients’ plasma samples than in those of healthy controls. Analysis of PCa-linked exosomes could help distinguish PCa patients from those with non-malignant prostatic disease, but exosome analysis is not yet standardised or cheap enough for large-scale use [].
Defining an ‘abnormal’ PSA threshold is challenging due to the lack of universally standardised cut-off values []. PCa suspicion generally increases with serum PSA levels above age- and risk-adjusted thresholds (commonly 3–4 ng/mL). However, clinically significant cancers can occur even within these ranges, demonstrating the limitations of PSA-based screening []. When PCa is suspected based on elevated PSA levels or clinical findings, further evaluation typically involves multiparametric magnetic resonance imaging (mpMRI) to localise suspicious lesions, followed by a prostate biopsy for histopathological confirmation. They may also fail to capture tumour heterogeneity adequately. Setting a PI-RADS score of ≥4 as the threshold for biopsy indication significantly reduced the number of unnecessary biopsies and the detection of clinically insignificant PCa without compromising the detection of clinically significant PCa []. However, biopsies are invasive procedures that carry a risk of bleeding, infection, and urinary complications. MRI-based target biopsy can reduce the rate of adverse events by decreasing the number of cores obtained through omitting the systematic biopsy []. These diagnostic limitations highlight the need to develop new, minimally invasive biomarkers that enhance diagnostic accuracy, improve patient risk stratification, and inform personalized treatment strategies.
One promising approach is to integrate urine- and serum-based biomarkers alongside tissue-derived molecular markers to improve the early detection and classification of PCa. Using these advanced biomarkers could reduce unnecessary biopsies, more accurately identify clinically significant disease, and support the development of personalised management plans for patients [].
Exosomes are nanoscale extracellular vesicles (30–200 nm) released into body fluids, where they transport molecular cargo reflective of their cell of origin. These organelles consist of different chemicals, including proteins, lipids, nucleic acids, and other substances such as amino acids and metabolites []. Exosomes play a key role in a variety of cellular processes and can transfer information between cells. Beit-Yannai et al. showed that several different cell types can secrete these exosomes which are enclosed by a single lipid bilayer membrane, which maintains cargo stability and facilitates intercellular transfer [].
Exosomes play a role in regulating cancer growth by facilitating communication between cells. These cellular messengers transfer proteins and other biological substances through tissue fluids, thereby influencing the development of cancer []. Exosomes have the capacity to react to the growth and progression of tumor cells and can also influence the metastasis of tumor cells [].
Recent progress in high-throughput sequencing, proteomic profiling, and exosome isolation technologies has fueled major interest in exosomes as components of liquid biopsy platforms. Instead of serving as liquid biopsies themselves, exosomes present in accessible fluids such as plasma and urine can be analyzed as part of minimally invasive liquid biopsy strategies.
These minimally invasive diagnostic approaches have the potential to complement, or in some cases even replace, traditional PSA testing and invasive tissue biopsies []. Exosome-based profiling has the potential to detect PCa at an earlier stage, predict how aggressive the disease is, help to decide on treatment, and monitor how the patient responds to treatment more accurately than conventional tools [].
This review synthesizes current multi-omics data on exosomal cargo in PCa, highlighting diagnostic, prognostic, and therapeutic implications as well as existing challenges to clinical translation.
2. Exosomes
Exosomes are a prominent class of extracellular vesicles (EVs) measuring 30–200 nm. They are secreted by both eukaryotic and prokaryotic cells and are present in nearly all biological fluids [,]. They originate from the endosomal pathway, which allows for the incorporation of various biomolecules, including proteins (e.g., tetraspanin CD63, CD81, CD82, CD53 and CD37, as well as cytosolic proteins), lipids (e.g., sphingomyelin, cholesterol and generally saturated fats), nucleic acids, metabolites, and small molecules, either within their lumen or onto their membrane. This process facilitates intercellular communication []. Although the exact mechanisms of selective cargo loading are not fully understood, evidence suggests that the composition of exosomes varies depending on their cellular origin and the physiological or pathological state of the source cell []. This cargo variability reflects the status of the parent cells, highlighting exosomes as key players in disease modulation and promising biomarkers. Among their contents, membrane- and cytoskeleton-derived proteins [] play a crucial role in cancer progression [] and serve as diagnostic and prognostic markers []. Lipids, concentrated in the exosomal membrane, are vital for vesicle formation and stability in recipient cells, and their distinct profiles have also been linked to cancer regulation and diagnostics [,]. The size of exosomes influences the variation in their nucleic acids, including DNA and RNA. Larger exosomes tend to carry longer DNA fragments, while smaller vesicles are enriched in shorter nucleotide sequences []. EVs can deliver their cargo to recipient cells, thereby modulating signaling pathways []. For instance, exosomes from metastatic PCa cells are enriched with noncoding RNAs like miR-21 and miR-141, which regulate osteoclastogenesis and osteoblastogenesis. Tumor-derived exosomes also contribute to epithelial–mesenchymal transition (EMT) through miRNAs like miR-21 and miR-409, facilitating the progression from benign to malignant states []. Moreover, they influence drug resistance, as seen with miR-34 in PCa cells and exosomes targeting Bcl-2, which affect the response to docetaxel []. These findings underscore the pivotal role of EVs in PCa progression and therapy resistance (Figure 1).
Figure 1.
Role of exosomes in prostate cancer.
3. Exosome Isolation and Identification
Exosomes are found in all body fluids, including blood, urine, lymph, bile, saliva, milk, and amniotic fluid []. Their unique physicochemical features, including size, density, shape, charge, and surface antigens, allow for isolation through various methods such as size-exclusion chromatography, magnetic bead-based immunoaffinity, ultrafiltration, heparin affinity, and centrifugation techniques (Figure 2). Ultracentrifugation is the most commonly used method due to its simplicity, though it has both advantages and limitations. Despite significant advances in isolation strategies, achieving fully purified exosome preparations remains challenging [].
Figure 2.
Methods for exosomes extraction.
After exosomes are isolated, various techniques are used to confirm their identity. These techniques are typically based on size, density, and specific markers []. Common approaches include dynamic light scattering (DLS) and nanoparticle tracking analysis (NTA) to determine size distribution [], as well as electron microscopy methods, such as transmission electron microscopy (TEM), which is often combined with immunogold labeling [], freezing electron microscopy, scanning electron microscopy (SEM), and atomic force microscopy (AFM) to characterize structure and morphology []. Biochemical assays such as Western blotting, the enzyme-linked immunosorbent assay (ELISA), and the photosensitizer magnetic bead detection system (ExoScreen) further assess purity and enrichment [], while flow cytometry provides qualitative and quantitative insights, though it is limited by poor resolution for vesicles smaller than 100 nm []. Since each method has its own strengths and limitations, combining two or more techniques is often necessary for accurately characterizing exosomes.
A persistent barrier to clinical implementation is methodological heterogeneity in exosome research. Variability in isolation methods leads to inconsistent vesicle purity, yield, and molecular cargo composition, making it difficult to compare results across studies or establish reproducible biomarker signatures. Commonly used isolation techniques—including differential ultracentrifugation, density gradients, size-exclusion chromatography, polymer-based precipitation, and immunoaffinity capture—differ markedly in recovery efficiency and degree of contaminant co-isolation (e.g., lipoproteins, protein aggregates, apoptotic bodies). These differences directly influence downstream RNA, protein, and metabolite profiles, which in turn affect diagnostic and mechanistic interpretations. Compounding this challenge, pre-analytical factors such as sample collection time, anticoagulant choice, centrifugation speed, storage duration, and freeze–thaw cycles can introduce further variability in vesicle integrity and cargo stability. To advance toward clinical translation, the field must converge on standardized operating procedures for sample processing and vesicle isolation, incorporate certified reference materials to calibrate analytical assays, and adopt shared reporting frameworks such as those recommended by MISEV 2023. Harmonization of these methodological parameters, combined with multi-center validation and transparent experimental documentation, is essential to ensure the reproducibility, comparability, and ultimately, the clinical reliability of exosome-based biomarkers in PCa [,,].
4. Tumor Microenvironment
The tumor microenvironment (TME) is a complex network consisting of tumor cells, infiltrating immune cells (e.g., macrophages, dendritic cells, and lymphocytes), cancer-associated fibroblasts, endothelial cells, lipids, the extracellular matrix, and signaling molecules []. The TME plays a critical role in drug resistance, immune suppression, and metastasis, making it a major focus of cancer research []. miR-203 in PCa cells’ exosome can change M0 macrophages to anti-tumour M1. This inhibits LNCAP cells proliferation, migration and invasion, whilst promoting apoptosis. In vivo studies show miR-203 as a therapeutic target. Its upregulation is associated with reduced tumour growth and increased M1 macrophages in the TME [].
Within the TME, exosomal miRNAs act as key mediators of intercellular signaling that drive tumor progression, immune suppression, and metastatic niche remodeling. Beyond miR-21, miR-203 and miR-409 [,], several additional exosomal miRNAs have been implicated in PCa TME modulation. Exosomal miR-141 and miR-375 are enriched in metastatic and castration-resistant prostate cancer (CRPC) and promote osteoblast differentiation, facilitating osteotropic metastasis []. miR-92a and miR-27a, released from tumor cells and cancer-associated fibroblasts (CAFs), enhance angiogenesis through upregulation of VEGF and PI3K/AKT signaling in endothelial cells [,]. miR-146a and miR-125b act as immune regulatory miRNAs, suppressing T-cell–mediated cytotoxic responses and promoting M2 macrophage polarization. Additionally, exosomal miR-423-5p and miR-21 have been shown to induce fibroblast reprogramming into tumor-supportive CAFs, increasing extracellular matrix deposition and invasive potential [,]. Together, these exosomal miRNAs reprogram stromal, endothelial, and immune cells toward a tumor-permissive microenvironment and play a crucial role in metastatic dissemination, particularly to bone.
The tumor immune microenvironment (TIME) is closely related and strongly influences patient prognosis []. TIME consists mainly of bone marrow–derived cells and lymphocytes, and its activity depends on the types and functions of infiltrating immune cells, immune checkpoint expression, and matrix alterations []. Yang et al. investigated how PCa cells are attracted to osteoblasts by exosomes. Higher concentrations of CCL5 attract more PCa cells to the bone environment. Disrupting the circ-DHPS/miR-214-3p/CCL5 interaction may reduce cancer cell migration []. Dai et al. reported that high levels of miR183 enhance the invasion and migration of PCa cells by downregulating tropomyosin 1 (TPM1) []. miR-888 has been identified as a key player in promoting PCa cell growth and movement. PC3-ML cells contain high levels of exosomal miR-888, which downregulates proteins such as Krüppel-like factor 5 (KLF5), retinoblastoma-like protein 1 (RBL1), tissue Inhibitor of metalloproteinases 2 (TIMP2), and SMAD family member 4 (SMAD4), strengthening tumour cell abilities []. Osteoblasts play a pivotal role in PCa bone metastasis and are controlled by mechanisms found in exosomes. Studies have revealed a new mechanism involving exosomal miR-92a-1-5p in different PCa subtypes. It promotes osteoclast differentiation and suppresses osteogenesis. The bone-destructive potential of these subtypes correlates with levels of other osteogenic (miR-148a-3p, miR-375) and osteoclastic (miR-92a-1-5p) miRNAs []. Furthermore, PCa exosome-containing miR-26a-5p, miR-27a-3p, and miR-30e-5p suppress BMP-2-induced bone formation and osteoblast activity [].
Both the TME and the TIME facilitate tumor progression, in part, through exosomes, which serve as key mediators of communication within these environments.
The role of exosomes in the tumor microenvironment directly informs their clinical potential as biomarkers for liquid biopsies. Because tumor-derived vesicles continuously circulate and mirror dynamic molecular changes occurring within the tumor niche, their cargo reflects ongoing processes such as immune evasion, epithelial–mesenchymal transition, and metabolic reprogramming. Therefore, mechanistic insights into exosome-mediated microenvironmental interactions serve as the biological foundation for their diagnostic and prognostic utility [,].
5. Exosomes’ Potential for PCa Diagnostics
Most studies on exosomal biomarkers for PCa focused on miRNAs, with the majority reporting their overexpression, though some identified reduced levels of specific miRNAs in patients (Figure 3). In evaluations of diagnostic performance, exosomal miRNAs demonstrated superior diagnostic potential compared to the current gold standard, serum PSA analysis []. For example, high plasma exosomal expression of miR-141-3p and low expression of miR-125a-5p in PCa patients’ plasma exosomes might help identify specific tumour traits associated with PCa. Furthermore, the miR-125a-5p/miR-141-3p ratio appears to be a more effective marker than either value in isolation []. Exosomal miR-21, miR-141, miR-200c, and miR-375 have been consistently linked to disease progression across multiple studies and show great promise as non-invasive biomarkers for PCa, with potential applications in diagnosis, prognosis, therapy optimization, and prediction of clinical outcomes []. Urinary exosomal ERG, PCA3, PSMA, CK19, and EpCAM were significantly upregulated in PCa patients compared with healthy controls. Moreover, the expression levels of urinary exosomal ERG, ARV7, and PSMA showed a strong correlation with Gleason scores []. Urinary exosomal mRNA profiling, with an emphasis on RAB5B and WWP1, holds significant promise as a strategy to enhance the early detection of PCa [].
Figure 3.
Role of miRNAs involved in exosomes cargo in different steps of tumor progression.
Peng et al. identified four novel piRNAs with significant AUC values in ROC analysis [], while Merkert et al. examined both piRNAs and miRNAs, reporting AUCs above 0.7 for most markers []. Other studies analyzed lncRNAs in blood and urine samples (SAP30L-AS1, SChLAP1, AC015987.1, CTD-2589M5.4, RP11-363E6.3), all showing significant overexpression in PCa [,]. Urinary exosomal lncRNA assay combining PCA3 and MALAT1, which outperformed current clinical parameters by achieving a higher AUC for predicting biopsy outcomes and detecting high-grade disease []. Protein expression was assessed using Western blot, ELISA, or SDS-PAGE, focusing mainly on cancer-related proteins (PSMA, PCA3, TMPRSS2:EGR, PSA) or exosomal surface proteins (CD9, CD3, EpCAM, CD81) []. Cytokines were identified as promising diagnostic markers [,], and elevated levels of enzymes such as carbonic anhydrase IX and gamma-glutamyltransferase were also reported in PCa patients [,]. Clinical studies have also examined EV-derived RNAs as biomarkers: Yang et al., through meta-analysis, confirmed the strong diagnostic value of plasma exosomal miRNAs in PCa []. The early diagnosis of PCa can be facilitated by the combined prediction models, which consist of plasma exosomal miRNAs (hsa-miR-320c and hsa-miR-944), US radiomics, and clinical tPSA []. The use of advanced tools, including AI- and nanotechnology-based sensors, improves the accuracy of PCa diagnostics centred on exosomes, resulting in more sensitive detection and superior clinical outcomes [].
A growing number of studies have investigated exosomal biomarkers in different biofluids, particularly plasma, urine, and semen, reflecting the accessibility and clinical relevance of these sample types in PCa, as summarized in Table 1. Plasma-derived exosomal miRNAs such as miR-141 and miR-375 have been repeatedly associated with metastatic and castration-resistant disease, while urinary exosomal lncRNAs such as PCA3 and PCAT-1, and protein cargo such as PSMA, have demonstrated utility in distinguishing PCa from benign prostatic hyperplasia. Exosomal miRNAs, lncRNAs, proteins, and metabolites and their clinical implications in PCa are represented in Table 2. Seminal fluid exosomes are especially enriched in prostate-derived vesicles and therefore provide a more tissue-specific signal; however, systematic clinical evaluation remains limited. Importantly, while these biomarkers show promising diagnostic and prognostic performance within individual studies, direct comparisons across studies reveal substantial variability in sensitivity, specificity, and threshold values. These discrepancies are primarily driven by inconsistencies in pre-analytical handling (e.g., centrifugation steps, storage time, and temperature), isolation method (ultracentrifugation vs. size-exclusion chromatography vs. polymer-based precipitation), and downstream quantification platforms (qPCR vs. NGS vs. digital PCR). Moreover, the absence of universally accepted endogenous reference controls for exosomal RNA normalization significantly limits reproducibility and cross-cohort comparability. Notably, biomarker panels that perform well in discovery cohorts often lose predictive power in external validation settings, underscoring the need for multi-center standardization. Therefore, while exosomal cargo holds strong potential for clinical integration as a non-invasive diagnostic platform, rigorous standardization of workflows, transparent reporting of analytical parameters, and validation in large, prospectively collected cohorts are essential to establish clinical reliability and regulatory feasibility.
Table 1.
Potential miRNA biomarkers for prostate cancer in clinical settings.
Table 2.
Exosomal miRNAs, lncRNAs, proteins, and metabolites to clinical implications in PCa.
In the coming years, collaborative efforts among clinicians, researchers, and bioinformaticians will be crucial in determining which patients should undergo molecular profiling and when, designing more effective, biomarker-driven clinical trials, addressing technical challenges, and ultimately integrating liquid biopsy into the routine clinical care of PCa patients [,].
6. Exosomes’ Potential as a PCa Therapy
Two glycoproteins, P-glycoprotein and oncofetal glycoprotein 5T4, were among the quantified proteins and were linked to docetaxel resistance and the persistence of malignant cells, respectively []. Vardaki et al. reported that the immune checkpoint protein PD-L1 was associated with shorter overall survival in patients treated with Radium-223 []. Exosomal microRNAs play a role in PCa chemoresistance [] and may serve as surrogate biomarkers for tumor response to taxane-based therapies []. Studies have shown that exosomes—particularly those carrying MDR-1/P-gp—from docetaxel-resistant cell lines can transfer resistance to sensitive PCa cell lines such as DU145, 22Rv1, and LNCap []. Similarly, Kawakami et al. identified β4 (ITGB4) and vinculin within exosomes as potential markers of PCa progression, closely linked to taxane resistance []. Notably, elevated serum exosomal P-glycoprotein levels correlate with resistance to docetaxel but not to cabazitaxel, suggesting its value as a biomarker to guide treatment decisions in PCa management []. CAF-derived exosomal miR-196b-5p, upregulated after androgen deprivation therapy, promotes epithelial–mesenchymal transition in PCa cells via the HOXC8/NF-κB pathway, revealing a mechanism of metastasis and potential therapeutic targets for post-castration management []. Liu et al. reported that PCa cell–derived exosomes upregulated PD-1 and TIM-3 expression in CD8+ T cells, promoted the release of cytokines associated with T cell exhaustion, and markedly reduced their cytotoxic activity against PCa cells. Treatment with GW4869 reversed these effects by inhibiting exosome production, thereby restoring CD8+ T cell function and suppressing PCa cell growth both in vivo and in vitro. These results suggest that GW4869 may hold therapeutic potential for PCa []. DU145 cells are more radioresistant than PC3 cells, and exosomes may contribute to this resistance []. Therefore, investigating exosome functions is key to understanding carcinogenesis and improving radiotherapy. One study showed that urinary miR-664a-5p was significantly upregulated in patients responding to PARP inhibitors []. In addition, Pukha et al. identified specific metabolites (glucuronate, D-ribose 5-phosphate, and isobutyryl-L-carnitine) as potential markers of successful prostatectomy [], while Macías et al. demonstrated that lncRNAs (CCL2, CXCL5, and S100A9) could predict surgical efficacy and recovery []. Androgen receptor splice variant 7 (AR-V7) expression resulted in a strong predictor of response to ARSIs and hormone therapy []. Malla and colleagues reported that miRNAs let-7a-5p and miR-21-5p were elevated in high-risk PCa patients undergoing radiotherapy compared to those with intermediate-risk disease []. Considering the limited effectiveness of current immunotherapies for PCa, targeting PD-1–carrying exosome secretion or inhibiting USP7 function could represent promising immunostimulatory strategies for treatment [].
7. Exosomes’ Potential as a PCa Prognostic
Exosomal cargo detected through liquid biopsy has strong prognostic potential, as it can indicate disease grade, metastatic risk, overall survival, and biochemical recurrence-free survival. Among these, specific miRNAs and lncRNAs stand out, showing significant associations with tumor aggressiveness and recurrence. Gao et al. similarly highlighted the value of exosomal miRNAs in tracking PCa invasion and metastasis []. Using exosomal biomarkers to stratify patients by Gleason score and predict biochemical recurrence could transform active surveillance, enabling more personalized PCa management []. In a large cohort, Wang et al. used the Sentinel™ platform to identify three sncRNAs with superior sensitivity and specificity for diagnosing and predicting high-grade PCa []. Additionally, circRNAs were evaluated, all of which showed significant associations with prognosis []. Zavridou et al. linked GSTP1 and RASSF1A gene methylation with overall survival [], while Tao et al. identified several lncRNAs (AC015987.1, CTD-2589M5.4, and RP11-363E6.3) as potential tools for guiding active surveillance []. Similarly, Kretschmer et al., using the ExoDx test in over 2000 patients, demonstrated its value in stratifying disease from grade group 1 to 3 and supporting surveillance strategies []. Overall, most studies reported that elevated exosomal biomarker expression correlates with poor outcomes or higher-grade disease, except Ruiz-Plazas et al., who observed reduced miRNA expression linked to high-risk cases []. Wang et al. reported that exosome-associated genes, including AQP2 and ZNF114, show strong potential as non-invasive biomarkers for predicting PCa status and prognosis, offering an alternative to highly invasive diagnostic procedures []. Urinary exosomal FAM153C-RPL19 demonstrated greater diagnostic value than PSA, particularly in gray-zone cases, with elevated levels linked to poor prognosis [].
While exosome-derived biomarkers show promising diagnostic and prognostic potential in PCa, translation into routine clinical use remains limited by several methodological constraints. A key challenge is the lack of standardization across pre-analytical workflows, including biological fluid selection, time-to-processing, and freeze–thaw conditions, all of which influence vesicle integrity and detected cargo profiles. Divergent exosome isolation methods (ultracentrifugation, size-exclusion chromatography, polymer-based precipitation, and immunoaffinity capture) yield heterogeneous vesicle populations with variable purity. Moreover, many studies rely on limited patient cohorts without external validation, reducing reproducibility and generalizability. Multi-center validation studies paired with MISEV 2023-aligned reporting standards are necessary to establish clinically robust biomarker signatures [].
8. Conclusions and Perspectives
Although exosomal biomarkers hold significant promise for improving prostate cancer detection, risk stratification, and treatment monitoring, their translation into routine clinical practice remains challenging. A critical barrier is the lack of standardized isolation and characterization workflows, which results in substantial variability in vesicle purity, yield, and molecular cargo profiles across laboratories. This methodological heterogeneity complicates biomarker comparison, hinders reproducibility, and weakens the strength of evidence required for regulatory approval. Furthermore, many studies have been conducted in small or demographically narrow patient cohorts, often without external or prospective validation, limiting generalizability across disease stages and populations. Comprehensive multi-center studies using harmonized analytical pipelines and clinically annotated biospecimen collections will be essential to define clinically robust exosomal signatures. In addition, integration of exosome-based biomarkers with existing clinical frameworks—including risk calculators, imaging modalities, and molecular classifiers—will be necessary to determine their incremental predictive value over current standards of care. Advances in digital PCR, single-vesicle analysis, and machine learning–based biomarker modeling may further enhance precision and interpretability. Ultimately, the successful clinical adoption of exosome-based diagnostics will require a convergence of methodological standardization, large-scale validation, and incorporation into evidence-based clinical decision-support systems.
Author Contributions
Conceptualization, R.A.C. and I.B.-N.; methodology, R.A.C.; software, A.N.; validation, L.B. and I.B.-N.; investigation, L.B.; resources, R.A.C.; data curation, R.A.C.; writing—original draft preparation, R.A.C.; writing—review and editing, I.B.-N.; visualization, S.S.; supervision, I.B.-N. All authors have read and agreed to the published version of the manuscript.
Funding
This research received no external funding.
Institutional Review Board Statement
Not applicable.
Informed Consent Statement
Not applicable.
Data Availability Statement
No new data were created or analyzed in this study.
Conflicts of Interest
The authors declare no conflicts of interest.
References
- Culp, M.B.; Soerjomataram, I.; Efstathiou, J.A.; Bray, F.; Jemal, A. Recent Global Patterns in Prostate Cancer Incidence and Mortality Rates. Eur. Urol. 2020, 77, 38–52. [Google Scholar] [CrossRef] [PubMed]
- Bergengren, O.; Pekala, K.R.; Matsoukas, K.; Fainberg, J.; Mungovan, S.F.; Bratt, O.; Bray, F.; Brawley, O.; Luckenbaugh, A.N.; Mucci, L.; et al. 2022 Update on Prostate Cancer Epidemiology and Risk Factors-A Systematic Review. Eur. Urol. 2023, 84, 191–206. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- European Commission. Prostate Cancer Burden in EU-27. 2021. Available online: https://ecis.jrc.ec.europa.eu (accessed on 20 August 2025).
- Rebello, R.J.; Oing, C.; Knudsen, K.E.; Loeb, S.; Johnson, D.C.; Reiter, R.E.; Gillessen, S.; Van der Kwast, T.; Bristow, R.G. Prostate cancer. Nat. Rev. Dis. Primers 2021, 7, 9. [Google Scholar] [CrossRef] [PubMed]
- Ilic, D.; Djulbegovic, M.; Jung, J.H.; Hwang, E.C.; Zhou, Q.; Cleves, A.; Agoritsas, T.; Dahm, P. Prostate cancer screening with prostate-specific antigen (PSA) test: A systematic review and meta-analysis. BMJ 2018, 362, k3519. [Google Scholar] [CrossRef] [PubMed]
- Stamey, T.A.; Yang, N.; Hay, A.R.; McNeal, J.E.; Freiha, F.S.; Redwine, E. Prostate-specific antigen as a serum marker for adenocarcinoma of the prostate. N. Engl. J. Med. 1987, 317, 909–916. [Google Scholar] [CrossRef] [PubMed]
- Hoffman, R.M. Clinical practice. Screening for prostate cancer. N. Engl. J. Med. 2011, 365, 2013–2019. [Google Scholar] [CrossRef] [PubMed]
- Van der Kwast, T.H.; Roobol, M.J. Defining the threshold for significant versus insignificant prostate cancer. Nat. Rev. Urol. 2013, 10, 473–482. [Google Scholar] [CrossRef] [PubMed]
- Ludwig, M.; Rajvansh, R.; Drake, J.M. Emerging Role of Extracellular Vesicles in Prostate Cancer. Endocrinology 2021, 162, bqab139. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Logozzi, M.; Angelini, D.F.; Iessi, E.; Mizzoni, D.; Di Raimo, R.; Federici, C.; Lugini, L.; Borsellino, G.; Gentilucci, A.; Pierella, F.; et al. Increased PSA expression on prostate cancer exosomes in in vitro condition and in cancer patients. Cancer Lett. 2017, 403, 318–329. [Google Scholar] [CrossRef] [PubMed]
- Salciccia, S.; Frisenda, M.; Bevilacqua, G.; Gobbi, L.; Bucca, B.; Moriconi, M.; Viscuso, P.; Gentilucci, A.; Mariotti, G.; Cattarino, S.; et al. Exosome Analysis in Prostate Cancer: How They Can Improve Biomarkers’ Performance. Curr. Issues Mol. Biol. 2023, 45, 6085–6096. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Semjonow, A.; Brandt, B.; Oberpenning, F.; Roth, S.; Hertle, L. Discordance of assay methods creates pitfalls for the interpretation of prostate-specific antigen values. Prostate Suppl. 1996, 7, 3–16. [Google Scholar] [CrossRef] [PubMed]
- Thompson, I.M.; Pauler, D.K.; Goodman, P.J.; Tangen, C.M.; Lucia, M.S.; Parnes, H.L.; Minasian, L.M.; Ford, L.G.; Lippman, S.M.; Crawford, E.D.; et al. Prevalence of prostate cancer among men with a prostate-specific antigen level ≤ 4.0 ng per milliliter. N. Engl. J. Med. 2004, 350, 2239–2246, Erratum in N. Engl. J. Med. 2004, 351, 1470. [Google Scholar] [CrossRef] [PubMed]
- Fazekas, T.; Shim, S.R.; Basile, G.; Baboudjian, M.; Kói, T.; Przydacz, M.; Abufaraj, M.; Ploussard, G.; Kasivisvanathan, V.; Rivas, J.G.; et al. Magnetic Resonance Imaging in Prostate Cancer Screening: A Systematic Review and Meta-Analysis. JAMA Oncol. 2024, 10, 745–754. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Wegelin, O.; Exterkate, L.; van der Leest, M.; Kelder, J.C.; Bosch, J.L.H.R.; Barentsz, J.O.; Somford, D.M.; van Melick, H.H.E. Complications and Adverse Events of Three Magnetic Resonance Imaging-based Target Biopsy Techniques in the Diagnosis of Prostate Cancer Among Men with Prior Negative Biopsies: Results from the FUTURE Trial, a Multicentre Randomised Controlled Trial. Eur. Urol. Oncol. 2019, 2, 617–624. [Google Scholar] [CrossRef] [PubMed]
- Kretschmer, A.; Tilki, D. Biomarkers in prostate cancer—Current clinical utility and future perspectives. Crit. Rev. Oncol. Hematol. 2017, 120, 180–193. [Google Scholar] [CrossRef] [PubMed]
- Pegtel, D.M.; Gould, S.J. Exosomes. Annu. Rev. Biochem. 2019, 88, 487–514. [Google Scholar] [CrossRef] [PubMed]
- Beit-Yannai, E.; Tabak, S.; Stamer, W.D. Physical exosome:exosome interactions. J. Cell. Mol. Med. 2018, 22, 2001–2006. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Terrasini, N.; Lionetti, V. Exosomes in Critical Illness. Crit. Care. Med. 2017, 45, 1054–1060. [Google Scholar] [CrossRef] [PubMed]
- Soung, Y.H.; Ford, S.; Zhang, V.; Chung, J. Exosomes in Cancer Diagnostics. Cancers 2017, 9, 8. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Kalluri, R.; LeBleu, V.S. The biology, function, and biomedical applications of exosomes. Science 2020, 367, eaau6977. [Google Scholar] [CrossRef] [PubMed]
- Boukouris, S.; Mathivanan, S. Exosomes in bodily fluids are a highly stable resource of disease biomarkers. Proteomics Clin. Appl. 2015, 9, 358–367. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Teng, F.; Fussenegger, M. Shedding Light on Extracellular Vesicle Biogenesis and Bioengineering. Adv. Sci. 2020, 8, 2003505. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Thakur, A.; Parra, D.C.; Motallebnejad, P.; Brocchi, M.; Chen, H.J. Exosomes: Small vesicles with big roles in cancer, vaccine development, and therapeutics. Bioact. Mater. 2021, 10, 281–294. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Mathivanan, S.; Fahner, C.J.; Reid, G.E.; Simpson, R.J. ExoCarta 2012: Database of exosomal proteins, RNA and lipids. Nucleic Acids Res. 2012, 40, D1241–D1244. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Vlaeminck-Guillem, V. Extracellular Vesicles in Prostate Cancer Carcinogenesis, Diagnosis, and Management. Front. Oncol. 2018, 8, 222. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Brzozowski, J.S.; Bond, D.R.; Jankowski, H.; Goldie, B.J.; Burchell, R.; Naudin, C.; Smith, N.D.; Scarlett, C.J.; Larsen, M.R.; Dun, M.D.; et al. Extracellular vesicles with altered tetraspanin CD9 and CD151 levels confer increased prostate cell motility and invasion. Sci. Rep. 2018, 8, 8822. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Mashouri, L.; Yousefi, H.; Aref, A.R.; Ahadi, A.M.; Molaei, F.; Alahari, S.K. Exosomes: Composition, biogenesis, and mechanisms in cancer metastasis and drug resistance. Mol. Cancer 2019, 18, 75. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Cheng, L.; Zhang, K.; Qing, Y.; Li, D.; Cui, M.; Jin, P.; Xu, T. Proteomic and lipidomic analysis of exosomes derived from ovarian cancer cells and ovarian surface epithelial cells. J. Ovarian Res. 2020, 13, 9. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Zhang, H.; Freitas, D.; Kim, H.S.; Fabijanic, K.; Li, Z.; Chen, H.; Mark, M.T.; Molina, H.; Martin, A.B.; Bojmar, L.; et al. Identification of distinct nanoparticles and subsets of extracellular vesicles by asymmetric flow field-flow fractionation. Nat. Cell Biol. 2018, 20, 332–343. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Pan, J.; Ding, M.; Xu, K.; Yang, C.; Mao, L.J. Exosomes in diagnosis and therapy of prostate cancer. Oncotarget 2017, 8, 97693–97700. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Corcoran, C.; Rani, S.; O’Driscoll, L. miR-34a is an intracellular and exosomal predictive biomarker for response to docetaxel with clinical relevance to prostate cancer progression. Prostate 2014, 74, 1320–1334. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.T.; Shi, T.; Srivastava, S.; Kagan, J.; Liu, T.; Rodland, K.D. Proteomic Analysis of Exosomes for Discovery of Protein Biomarkers for Prostate and Bladder Cancer. Cancers 2020, 12, 2335. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Szatanek, R.; Baj-Krzyworzeka, M.; Zimoch, J.; Lekka, M.; Siedlar, M.; Baran, J. The Methods of Choice for Extracellular Vesicles (EVs) Characterization. Int. J. Mol. Sci. 2017, 18, 1153. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Jung, M.K.; Mun, J.Y. Sample Preparation and Imaging of Exosomes by Transmission Electron Microscopy. J. Vis. Exp. 2018, 131, 56482. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Chuo, S.T.; Chien, J.C.; Lai, C.P. Imaging extracellular vesicles: Current and emerging methods. J. Biomed. Sci. 2018, 25, 91. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Yoshioka, Y.; Kosaka, N.; Konishi, Y.; Ohta, H.; Okamoto, H.; Sonoda, H.; Nonaka, R.; Yamamoto, H.; Ishii, H.; Mori, M.; et al. Ultra-sensitive liquid biopsy of circulating extracellular vesicles using ExoScreen. Nat. Commun. 2014, 5, 3591. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Libregts, S.F.W.M.; Arkesteijn, G.J.A.; Németh, A.; Nolte-‘t Hoen, E.N.M.; Wauben, M.H.M. Flow cytometric analysis of extracellular vesicle subsets in plasma: Impact of swarm by particles of non-interest. J. Thromb. Haemost. 2018, 16, 1423–1436. [Google Scholar] [CrossRef] [PubMed]
- Gardiner, C.; Di Vizio, D.; Sahoo, S.; Théry, C.; Witwer, K.W.; Wauben, M.; Hill, A.F. Techniques used for the isolation and characterization of extracellular vesicles: Results of a worldwide survey. J. Extracell. Vesicles. 2016, 5, 32945. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Jeppesen, D.K.; Fenix, A.M.; Franklin, J.L.; Higginbotham, J.N.; Zhang, Q.; Zimmerman, L.J.; Liebler, D.C.; Ping, J.; Liu, Q.; Evans, R.; et al. Reassessment of Exosome Composition. Cell 2019, 177, 428–445.e18. [Google Scholar] [CrossRef]
- Royo, F.; Théry, C.; Falcón-Pérez, J.M.; Nieuwland, R.; Witwer, K.W. Methods for Separation and Characterization of Extracellular Vesicles: Results of a Worldwide Survey Performed by the ISEV Rigor and Standardization Subcommittee. Cells 2020, 9, 1955. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Giraldo, N.A.; Sanchez-Salas, R.; Peske, J.D.; Vano, Y.; Becht, E.; Petitprez, F.; Validire, P.; Ingels, A.; Cathelineau, X.; Fridman, W.H.; et al. The clinical role of the TME in solid cancer. Br. J. Cancer 2019, 120, 45–53. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Paluskievicz, C.M.; Cao, X.; Abdi, R.; Zheng, P.; Liu, Y.; Bromberg, J.S. T Regulatory Cells and Priming the Suppressive Tumor Microenvironment. Front. Immunol. 2019, 10, 2453. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Zhang, L.S.; Chen, Q.C.; Zong, H.T.; Xia, Q. Exosome miRNA-203 promotes M1 macrophage polarization and inhibits prostate cancer tumor progression. Mol. Cell. Biochem. 2024, 479, 2459–2470. [Google Scholar] [CrossRef] [PubMed]
- Zedan, A.H.; Osther, P.J.S.; Assenholt, J.; Madsen, J.S.; Hansen, T.F. Circulating miR-141 and miR-375 are associated with treatment outcome in metastatic castration resistant prostate cancer. Sci. Rep. 2020, 10, 227. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Yanshen, Z.; Lifen, Y.; Xilian, W.; Zhong, D.; Huihong, M. miR-92a promotes proliferation and inhibits apoptosis of prostate cancer cells through the PTEN/Akt signaling pathway. Libyan J. Med. 2021, 16, 1971837, Erratum in Libyan J. Med. 2025, 20, 2475687. https://doi.org/10.1080/19932820.2025.2475687. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Wan, X.; Huang, W.; Yang, S.; Zhang, Y.; Zhang, P.; Kong, Z.; Li, T.; Wu, H.; Jing, F.; Li, Y. Androgen-induced miR-27A acted as a tumor suppressor by targeting MAP2K4 and mediated prostate cancer progression. Int. J. Biochem. Cell Biol. 2016, 79, 249–260. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed Central]
- Zennami, K.; Graham, M.; Chikara, S.; Sysa-Shah, P.; Rafiqi, F.H.; Wang, R.; Abel, B.; Zeng, Q.; Krueger, T.E.G.; Brennen, W.N.; et al. Cell Type-Specific Effects of miR-21 Loss Attenuate Tumor Progression in MYC-Driven Prostate Cancer. bioRxiv 2025. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Chen, Z.; Zhou, L.; Liu, L.; Hou, Y.; Xiong, M.; Yang, Y.; Hu, J.; Chen, K. Single-cell RNA sequencing highlights the role of inflammatory cancer-associated fibroblasts in bladder urothelial carcinoma. Nat. Commun. 2020, 11, 5077. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Zhang, Y.; Liu, Q.; Liao, Q. Long noncoding RNA: A dazzling dancer in tumor immune microenvironment. J. Exp. Clin. Cancer Res. 2020, 39, 231. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Yang, Z.; Chen, J.Q.; Liu, T.J.; Chen, Y.L.; Ma, Z.K.; Fan, Y.Z.; Wang, Z.X.; Xu, S.; Wang, K.; Wang, X.Y.; et al. Knocking down AR promotes osteoblasts to recruit prostate cancer cells by altering exosomal circ-DHPS/miR-214-3p/CCL5 pathway. Asian J. Androl. 2024, 26, 195–204. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Dai, Y.; Gao, X. Inhibition of cancer cell-derived exosomal microRNA-183 suppresses cell growth and metastasis in prostate cancer by upregulating TPM1. Cancer Cell Int. 2021, 21, 145. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Hasegawa, T.; Glavich, G.J.; Pahuski, M.; Short, A.; Semmes, O.J.; Yang, L.; Galkin, V.; Drake, R.; Esquela-Kerscher, A. Characterization and Evidence of the miR-888 Cluster as a Novel Cancer Network in Prostate. Mol. Cancer Res. 2018, 16, 669–681. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Yu, L.; Sui, B.; Fan, W.; Lei, L.; Zhou, L.; Yang, L.; Diao, Y.; Zhang, Y.; Li, Z.; Liu, J.; et al. Exosomes derived from osteogenic tumor activate osteoclast differentiation and concurrently inhibit osteogenesis by transferring COL1A1-targeting miRNA-92a-1-5p. J. Extracell Vesicles 2021, 10, e12056. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Furesi, G.; de Jesus Domingues, A.M.; Alexopoulou, D.; Dahl, A.; Hackl, M.; Schmidt, J.R.; Kalkhof, S.; Kurth, T.; Taipaleenmäki, H.; Conrad, S.; et al. Exosomal miRNAs from Prostate Cancer Impair Osteoblast Function in Mice. Int. J. Mol. Sci. 2022, 23, 1285. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Hoshino, A.; Kim, H.S.; Bojmar, L.; Gyan, K.E.; Cioffi, M.; Hernandez, J.; Zambirinis, C.P.; Rodrigues, G.; Molina, H.; Heissel, S.; et al. Extracellular Vesicle and Particle Biomarkers Define Multiple Human Cancers. Cell 2020, 182, 1044–1061.e18. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Hamid, Y.; Rabbani, R.D.; Afsara, R.; Nowrin, S.; Ghose, A.; Papadopoulos, V.; Sirlantzis, K.; Ovsepian, S.V.; Boussios, S. Exosomal Liquid Biopsy in Prostate Cancer: A Systematic Review of Biomarkers for Diagnosis, Prognosis, and Treatment Response. Int. J. Mol. Sci. 2025, 26, 802. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Li, W.; Dong, Y.; Wang, K.J.; Deng, Z.; Zhang, W.; Shen, H.F. Plasma exosomal miR-125a-5p and miR-141-5p as non-invasive biomarkers for prostate cancer. Neoplasma 2020, 67, 1314–1318. [Google Scholar] [CrossRef] [PubMed]
- de Nóbrega, M.; Dos Reis, M.B.; Pereira, É.R.; de Souza, M.F.; de Syllos Cólus, I.M. The potential of cell-free and exosomal microRNAs as biomarkers in liquid biopsy in patients with prostate cancer. J. Cancer Res. Clin. Oncol. 2022, 148, 2893–2910. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Gan, J.; Zeng, X.; Wang, X.; Wu, Y.; Lei, P.; Wang, Z.; Yang, C.; Hu, Z. Effective Diagnosis of Prostate Cancer Based on mRNAs From Urinary Exosomes. Front. Med. 2022, 9, 736110. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- 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] [PubMed] [PubMed Central]
- Peng, Q.; Chiu, P.K.; Wong, C.Y.; Cheng, C.K.; Teoh, J.Y.; Ng, C.F. Identification of piRNA Targets in Urinary Extracellular Vesicles for the Diagnosis of Prostate Cancer. Diagnostics 2021, 11, 1828. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Markert, L.; Holdmann, J.; Klinger, C.; Kaufmann, M.; Schork, K.; Turewicz, M.; Eisenacher, M.; Savelsbergh, A. Small RNAs as biomarkers to differentiate benign and malign prostate diseases: An alternative for transrectal punch biopsy of the prostate? PLoS ONE 2021, 16, e0247930. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Wang, Y.H.; Ji, J.; Wang, B.C.; Chen, H.; Yang, Z.H.; Wang, K.; Luo, C.L.; Zhang, W.W.; Wang, F.B.; Zhang, X.L. Tumor-Derived Exosomal Long Noncoding RNAs as Promising Diagnostic Biomarkers for Prostate Cancer. Cell. Physiol. Biochem. 2018, 46, 532–545. [Google Scholar] [CrossRef] [PubMed]
- Tao, W.; Wang, B.Y.; Luo, L.; Li, Q.; Meng, Z.A.; Xia, T.L.; Deng, W.M.; Yang, M.; Zhou, J.; Zhang, X.; et al. A urine extracellular vesicle lncRNA classifier for high-grade prostate cancer and increased risk of progression: A multi-center study. Cell Rep. Med. 2023, 4, 101240. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- 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] [PubMed Central]
- Soekmadji, C.; Corcoran, N.M.; Oleinikova, I.; Jovanovic, L.; Australian Prostate Cancer Collaboration BioResource; Ramm, G.A.; Nelson, C.C.; Jenster, G.; Russell, P.J. Extracellular vesicles for personalized therapy decision support in advanced metastatic cancers and its potential impact for prostate cancer. Prostate 2017, 77, 1416–1423. [Google Scholar] [CrossRef] [PubMed]
- Xu, F.; Wang, X.; Huang, Y.; Zhang, X.; Sun, W.; Du, Y.; Xu, Z.; Kou, H.; Zhu, S.; Liu, C.; et al. Prostate cancer cell-derived exosomal IL-8 fosters immune evasion by disturbing glucolipid metabolism of CD8 + T cell. Cell Rep. 2023, 42, 113424. [Google Scholar] [CrossRef] [PubMed]
- Macías, M.; García-Cortés, Á.; Torres, M.; Ancizu-Marckert, J.; Ignacio Pascual, J.; Díez-Caballero, F.; Enrique Robles, J.; Rosell, D.; Miñana, B.; Mateos, B.; et al. Characterisation of the perioperative changes of exosomal immune-related cytokines induced by prostatectomy in early-stage prostate cancer patients. Cytokine 2021, 141, 155471. [Google Scholar] [CrossRef]
- 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. Enzyme. Inhib. Med. Chem. 2020, 35, 280–288. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Huertas-Lárez, R.; Muñoz-Moreno, L.; Recio-Aldavero, J.; Román, I.D.; Arenas, M.I.; Blasco, A.; Sanchís-Bonet, Á.; Bajo, A.M. Induction of more aggressive tumoral phenotypes in LNCaP and PC3 cells by serum exosomes from prostate cancer patients. Int. J. Cancer 2023, 153, 1829–1841. [Google Scholar] [CrossRef] [PubMed]
- Yang, B.; Xiong, W.Y.; Hou, H.J.; Xu, Q.; Cai, X.L.; Zeng, T.X.; Ha, X.Q. Exosomal miRNAs as Biomarkers of Cancer: A Meta-Analysis. Clin. Lab. 2019, 65, e23956. [Google Scholar] [CrossRef] [PubMed]
- Wang, C.; Zhou, C.; Zhang, Y.F.; He, H.; Wang, D.; Lv, H.X.; Yang, Z.J.; Wang, J.; Ren, Y.Q.; Zhang, W.B.; et al. Integrating plasma exosomal miRNAs, ultrasound radiomics and tPSA for the diagnosis and prediction of early prostate cancer: A multi-center study. Clin. Transl. Oncol. 2025, 27, 1248–1262. [Google Scholar] [CrossRef] [PubMed]
- Sonar, S. Decoding the mysteries of prostate cancer via cutting-edge liquid biopsy-based urine exosomes profiling. J. Liq. Biopsy 2024, 6, 100162. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Rodríguez, M.; Bajo-Santos, C.; Hessvik, N.P.; Lorenz, S.; Fromm, B.; Berge, V.; Sandvig, K.; Linē, A.; Llorente, A. Identification of non-invasive miRNAs biomarkers for prostate cancer by deep sequencing analysis of urinary exosomes. Mol. Cancer 2017, 16, 156. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Foj, L.; Ferrer, F.; Serra, M.; Arévalo, A.; Gavagnach, M.; Giménez, N.; Filella, X. Exosomal and Non-Exosomal Urinary miRNAs in Prostate Cancer Detection and Prognosis. Prostate 2017, 77, 573–583. [Google Scholar] [CrossRef] [PubMed]
- Mirzaei, S.; Paskeh, M.D.A.; Okina, E.; Gholami, M.H.; Hushmandi, K.; Hashemi, M.; Kalu, A.; Zarrabi, A.; Nabavi, N.; Rabiee, N.; et al. Molecular Landscape of LncRNAs in Prostate Cancer: A focus on pathways and therapeutic targets for intervention. J. Exp. Clin. Cancer Res. 2022, 41, 214. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Endzeliņš, E.; Berger, A.; Melne, V.; Bajo-Santos, C.; Soboļevska, K.; Ābols, A.; Rodriguez, M.; Šantare, D.; Rudņickiha, A.; Lietuvietis, V.; et al. Detection of circulating miRNAs: Comparative analysis of extracellular vesicle-incorporated miRNAs and cell-free miRNAs in whole plasma of prostate cancer patients. BMC Cancer 2017, 17, 730. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Barceló, M.; Castells, M.; Bassas, L.; Vigués, F.; Larriba, S. Semen miRNAs Contained in Exosomes as Non-Invasive Biomarkers for Prostate Cancer Diagnosis. Sci. Rep. 2019, 9, 13772. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Tai, Y.L.; Chu, P.Y.; Lee, B.H.; Chen, K.C.; Yang, C.Y.; Kuo, W.H.; Shen, T.L. Basics and applications of tumor-derived extracellular vesicles. J. Biomed. Sci. 2019, 26, 35. [Google Scholar] [CrossRef]
- Wang, S.; Du, P.; Cao, Y.; Ma, J.; Yang, X.; Yu, Z.; Yang, Y. Cancer associated fibroblasts secreted exosomal miR-1290 contributes to prostate cancer cell growth and metastasis via targeting GSK3β. Cell Death Discov. 2022, 8, 371. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Vardaki, I.; Corn, P.; Gentile, E.; Song, J.H.; Madan, N.; Hoang, A.; Parikh, N.; Guerra, L.; Lee, Y.C.; Lin, S.C.; et al. Radium-223 Treatment Increases Immune Checkpoint Expression in Extracellular Vesicles from the Metastatic Prostate Cancer Bone Microenvironment. Clin. Cancer Res. 2021, 27, 3253–3264. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Aaltomaa, S.; Lipponen, P.; Ala-Opas, M.; Kosma, V.M. Expression and prognostic value of CD44 standard and variant v3 and v6 isoforms in prostate cancer. Eur. Urol. 2001, 39, 138–144. [Google Scholar] [CrossRef] [PubMed]
- Trujillo, B.; Wu, A.; Wetterskog, D.; Attard, G. Blood-based liquid biopsies for prostate cancer: Clinical opportunities and challenges. Br. J. Cancer 2022, 127, 1394–1402. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Coman, R.A.; Schitcu, V.H.; Budisan, L.; Raduly, L.; Braicu, C.; Petrut, B.; Coman, I.; Berindan-Neagoe, I.; Al Hajjar, N. Evaluation of miR-148a-3p and miR-106a-5p as Biomarkers for Prostate Cancer: Pilot Study. Genes 2024, 15, 584. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Kato, T.; Mizutani, K.; Kameyama, K.; Kawakami, K.; Fujita, Y.; Nakane, K.; Kanimoto, Y.; Ehara, H.; Ito, H.; Seishima, M.; et al. Serum exosomal P-glycoprotein is a potential marker to diagnose docetaxel resistance and select a taxoid for patients with prostate cancer. Urol. Oncol. 2015, 33, 385.e15–385.e20. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Yang, X.; Guan, H.; Mizokami, A.; Keller, E.T.; Xu, X.; Liu, X.; Tan, J.; Hu, L.; Lu, Y.; et al. Exosome-derived microRNAs contribute to prostate cancer chemoresistance. Int. J. Oncol. 2016, 49, 838–846. [Google Scholar] [CrossRef]
- Kharaziha, P.; Chioureas, D.; Rutishauser, D.; Baltatzis, G.; Lennartsson, L.; Fonseca, P.; Azimi, A.; Hultenby, K.; Zubarev, R.; Ullén, A.; et al. Molecular profiling of prostate cancer derived exosomes may reveal a predictive signature for response to docetaxel. Oncotarget 2015, 6, 21740–21754. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Corcoran, C.; Rani, S.; O’Brien, K.; O’Neill, A.; Prencipe, M.; Sheikh, R.; Webb, G.; McDermott, R.; Watson, W.; Crown, J.; et al. Docetaxel-resistance in prostate cancer: Evaluating associated phenotypic changes and potential for resistance transfer via exosomes. PLoS ONE 2012, 7, e50999. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Kawakami, K.; Fujita, Y.; Kato, T.; Mizutani, K.; Kameyama, K.; Tsumoto, H.; Miura, Y.; Deguchi, T.; Ito, M. Integrin β4 and vinculin contained in exosomes are potential markers for progression of prostate cancer associated with taxane-resistance. Int. J. Oncol. 2015, 47, 384–390. [Google Scholar] [CrossRef] [PubMed]
- Song, X.; Li, T.; Zhou, W.; Feng, C.; Zhou, Z.; Chen, Y.; Li, D.; Chen, L.; Zhao, J.; Zhang, Y.; et al. CAF-derived exosomal miR-196b-5p after androgen deprivation therapy promotes epithelial-mesenchymal transition in prostate cancer cells through HOXC8/NF-κB signaling pathway. Biol. Direct. 2025, 20, 80. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Liu, J.; Guo, H.; Liu, S.; Hu, Y.; Huang, Y.; Rong, J.; Yuan, F.; Wang, R.; Wang, Z. Blocking secretion of exosomes by GW4869 dampens CD8+ T cell exhaustion and prostate cancer progression. Hum. Cell 2025, 38, 131. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Pszczółkowska-Kępa, B.; Olejarz, W.; Głuszko, A.; Wałpuski, G.; Lorenc, T.; Brzozowska, B. Exosome-mediated modulation of radioresistance: The radiation-induced bystander effect in prostate cancer cells. PLoS ONE 2025, 20, e0330501. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Kim, M.Y.; Moon, H.W.; Jo, M.S.; Lee, J.Y. Exosomal miR-664a-5p as a therapeutic target biomarker for PARP inhibitor response in prostate cancer. Am. J. Cancer Res. 2024, 14, 3789–3799. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Puhka, M.; Takatalo, M.; Nordberg, M.E.; Valkonen, S.; Nandania, J.; Aatonen, M.; Yliperttula, M.; Laitinen, S.; Velagapudi, V.; Mirtti, T.; et al. Metabolomic Profiling of Extracellular Vesicles and Alternative Normalization Methods Reveal Enriched Metabolites and Strategies to Study Prostate Cancer-Related Changes. Theranostics 2017, 7, 3824–3841. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Del Re, M.; Conteduca, V.; Crucitta, S.; Gurioli, G.; Casadei, C.; Restante, G.; Schepisi, G.; Lolli, C.; Cucchiara, F.; Danesi, R.; et al. Androgen receptor gain in circulating free DNA and splicing variant 7 in exosomes predict clinical outcome in CRPC patients treated with abiraterone and enzalutamide. Prostate. Cancer Prostatic. Dis. 2021, 24, 524–531. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Malla, B.; Aebersold, D.M.; Dal Pra, A. Protocol for serum exosomal miRNAs analysis in prostate cancer patients treated with radiotherapy. J. Transl. Med. 2018, 16, 223. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Zhang, J.; Chen, W.; Zhang, C.; He, Q.; Wang, X.; Han, J.; Gao, P.; Wang, K.; Xie, H.; Gao, F.; et al. Prostate Cancer Cells Secrete PD-1 in Exosomes to Enhance Myeloid-Derived Suppressor Cell Activity and Promote Tumor Immune Evasion. Cancer Res. 2025, 85, 3435–3453. [Google Scholar] [CrossRef] [PubMed]
- Gao, Z.; Pang, B.; Li, J.; Gao, N.; Fan, T.; Li, Y. Emerging Role of Exosomes in Liquid Biopsy for Monitoring Prostate Cancer Invasion and Metastasis. Front. Cell Dev. Biol. 2021, 9, 679527. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Wang, J.; Ni, J.; Beretov, J.; Thompson, J.; Graham, P.; Li, Y. Exosomal microRNAs as liquid biopsy biomarkers in prostate cancer. Crit. Rev. Oncol. Hematol. 2020, 145, 102860. [Google Scholar] [CrossRef] [PubMed]
- Wang, W.W.; Sorokin, I.; Aleksic, I.; Fisher, H.; Kaufman, R.P., Jr.; Winer, A.; McNeill, B.; Gupta, R.; Tilki, D.; Fleshner, N.; et al. Expression of Small Noncoding RNAs in Urinary Exosomes Classifies Prostate Cancer into Indolent and Aggressive Disease. J. Urol. 2020, 204, 466–475. [Google Scholar] [CrossRef] [PubMed]
- Tao, W.; Luo, Z.H.; He, Y.D.; Wang, B.Y.; Xia, T.L.; Deng, W.M.; Zhang, L.X.; Tang, X.M.; Meng, Z.A.; Gao, X.; et al. Plasma extracellular vesicle circRNA signature and resistance to abiraterone in metastatic castration-resistant prostate cancer. Br. J. Cancer 2023, 128, 1320–1332. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Zavridou, M.; Strati, A.; Bournakis, E.; Smilkou, S.; Tserpeli, V.; Lianidou, E. Prognostic Significance of Gene Expression and DNA Methylation Markers in Circulating Tumor Cells and Paired Plasma Derived Exosomes in Metastatic Castration Resistant Prostate Cancer. Cancers 2021, 13, 780. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Kretschmer, A.; Tutrone, R.; Alter, J.; Berg, E.; Fischer, C.; Kumar, S.; Torkler, P.; Tadigotla, V.; Donovan, M.; Sant, G.; et al. Pre-diagnosis urine exosomal RNA (ExoDx EPI score) is associated with post-prostatectomy pathology outcome. World J. Urol. 2022, 40, 983–989. [Google Scholar] [CrossRef]
- Ruiz-Plazas, X.; Altuna-Coy, A.; Alves-Santiago, M.; Vila-Barja, J.; García-Fontgivell, J.F.; Martínez-González, S.; Segarra-Tomás, J.; Chacón, M.R. Liquid Biopsy-Based Exo-oncomiRNAs Can Predict Prostate Cancer Aggressiveness. Cancers 2021, 13, 250. [Google Scholar] [CrossRef]
- Wang, H.; Li, X.; Wu, L.; Zhai, Y.; Zhu, Y.; Wang, W.; Chen, D.; Xing, N.; Ye, X.; Yang, F. Identification and verification of exosome-related gene signature to predict the cancer status and prognosis of prostate cancer. Discov. Oncol. 2025, 16, 1680. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Cheng, B.; Luo, T.; Wu, Y.; Hu, J.; Yang, C.; Wu, J.; Luo, Y.; Shangguan, W.; Li, W.; Yang, L.; et al. Urinary exosomal FAM153C-RPL19 chimeric RNA as a diagnostic and prognostic biomarker for prostate cancer in Chinese patients. Cancer Lett. 2025, 631, 217938. [Google Scholar] [CrossRef] [PubMed]
- Welsh, J.A.; Goberdhan, D.C.I.; O’DRiscoll, L.; Buzas, E.I.; Blenkiron, C.; Bussolati, B.; Cai, H.; Di Vizio, D.; Driedonks, T.A.P.; Erdbrügger, U.; et al. Minimal information for studies of extracellular vesicles (MISEV2023): From basic to ad vanced approaches. J. Extracell. Vesicles 2024, 13, e12404, Erratum in J. Extracell. Vesicles 2024, 13, e12451. https://doi.org/10.1002/jev2.12451. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
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. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).