Skip to main content

Prostate cancer-derived small extracellular vesicle proteins: the hope in diagnosis, prognosis, and therapeutics

Abstract

Current diagnostic tools for prostate cancer (PCa) diagnosis and risk stratification are insufficient. The hidden onset and poor efficacy of traditional therapies against metastatic PCa make this disease a heavy burden in global men’s health. Prostate cancer-derived extracellular vesicles (PCDEVs) have garnered attention in recent years due to their important role in communications in tumor microenvironment. Recent advancements have demonstrated PCDEVs proteins play an important role in PCa invasion, progression, metastasis, therapeutic resistance, and immune escape. In this review, we briefly discuss the applications of sEV proteins in PCa diagnosis and prognosis in liquid biopsy, focus on the roles of the PCa-derived small EVs (sEVs) proteins in tumor microenvironment associated with cancer progression, and explore the therapeutic potential of sEV proteins applied for future metastatic PCa therapy.

Graphical Abstract

Introduction

Prostate cancer (PCa) is the most common cancer in men worldwide. It was estimated that there would be 288,300 new cases of PCa in the United States in 2023, accounting for 29% of new cases of male cancers, with an estimated 34,700 deaths, accounting for 11% of male cancer deaths [1]. The incidence of PCa in Asia–pacific areas such as China is increasing with more mortality [2]. Tumor staging has a significant impact on treatment and prognosis. While generally, the non-metastatic localized PCa patients have a good prognosis, with a 5-year survival rate of 60–90%, distant metastatic PCa patients lead to irreversible disease progression, with a 5-year survival rate of only 30–40% [3]. The proportion of patients diagnosed with metastatic disease has witnessed a significant 25% increase in the past decade [4]. The elevated mortality rate is closely linked to the extensive spread of metastasis. Although surgical procedures and radiation therapy can effectively address localized disease, up to 30% of PCa patients treated in such a manner experience relapse and metastasis. Unfortunately, there is currently no curative therapy available for metastatic PCa, and the median survival period is approximately 1 year.

A major challenge in PCa clinical management lies in the limitations of current diagnostic tests, such as serum prostate specific antigen (PSA) testing, digital rectal examination, and histopathologic grading of tissues, to discern between indolent and aggressive disease [5]. The only existing screening biomarker, PSA, has been a subject of controversy as a screening assay due to its limitations, including false negatives and a high positive rate [6]. PSA cannot be used for early diagnosis and stratifying progression risk groups. It often results in over-diagnosis and over-treatment ranging from 20 to 42%, which can potentially cause more harm than good to patients. Furthermore, PSA has demonstrated little or no benefit in terms of PCa-specific survival [7, 8]. Therefore, improving the screening and diagnosis of PCa-especially metastatic PCa, and in-depth research on the mechanism of PCa metastasis are of great importance to design reasonable treatment plans, and improve the prognosis of patients. There is an urgent need to find biomarkers that can replace the current diagnostic approaches for more accurate diagnosis tools and monitoring PCa progression [9].

Liquid biopsy has recently emerged as an appealing non-invasive strategy to support early cancer diagnosis, selection for biopsy, active surveillance for low-risk cancer and post-treatment for recurrence. Liquid biopsy is a blood test that detects cancer cells or DNA or extracellular vesicles (EVs) that are circulating in the blood. It has demonstrated unparalleled advantages over conventional tissue biopsy and may complement or even replace medical imaging as a first- or second-line screening tool for earlier cancer detection and better surveillance of cancer metastasis and prognosis [10,11,12]. Liquid biopsy includes the analysis of circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), and EVs [13, 14]. However, the utilization of CTCs or ctDNA as cancer biomarkers encounter various technical and translational challenges including difficult isolation and characterization, a short half-life, high fragmentation, low abundance, and low stability. EVs can be classified to different subtypes based on different biogenesis pathways (exosome, microvesicles), physical characteristics [small EVs (sEVs), large EVs (lEVs),)], biochemical composition (CD81+/syntenin+-EVs), or descriptions of origin (large oncosomes, podocyte EVs). EVs possess several advantages including abundance (108–13 exosomes/mL in plasma), stability (capable of being stored at – 80 ℃ for months and even years), easy accessibility and having tumor-specific surface signatures.

All cells release EVs that are also classified into different types based on their morphology and contents, such as exosomes (sEVs), microvesicles (lEVs) and apoptotic bodies. Figure 1 describes the two classifications of EVs. Exosomes are released from all kinds of live cells after multivesicular body (MVB) fused with the cell membrane and can be detected through a range of methods, including high-resolution electron microscopy (EM) and mass spectrometry (MS) [15]. Exosomes biogenesis originates from the endocytic pathway, which involves several steps: (1) the plasma membrane buds inward to form early endosomes, (2) early endosomes develop into late endosomes and MVBs containing intraluminal vesicles (ILVs), (3) MVBs are transported to the plasma membrane, and (4) some MVBs are degraded after fusing with lysosomes, while others, which have specific surface proteins, are released in vesicular form after fusion with the plasma membrane, forming exosomes [16]. Multiple mechanisms are involved in each step, such as the endosomal sorting complex required for transport (ESCRT) or non-ESCRT mechanisms, which have been shown to depend on different MVBs to achieve the biogenesis of ILVs. ESCRT consists of four complexes: ESCRT-0, ESCRT-I, ESCRT-II, and ESCRT-III. ESCRT-0, made up of hepatocyte growth factor-regulated tyrosine kinase substrate (Hrs) and signal transducing adaptor molecule (STAM), recognizes mono- or polyubiquitinated cargo proteins through its ubiquitin-binding domain, while ESCRT-I and ESCRT-II complexes are responsible for inward budding of the endosomal membrane. The ESCRT-III complex then mediates the fusion of ILVs, ultimately generating ILVs. Research has shown that even if all four key subunits of the ESCRT complexes are simultaneously silenced, ILVs still form in MVBs, indicating the existence of ESCRT-independent mechanisms [17]. These mechanisms include two pathways: (1) Tetraspanins play a crucial role in clustering membrane cargos and localizing to specific membrane domains that facilitate the budding of ILVs; and (2) The recruitment of factors such as flotillins and the autophagy-related protein LC3 occurs in lipid domains that are enriched in ceramide and cholesterol [18]. Various molecules, including Pmel17 and RAB31, have been found to be involved in ESCRT-independent biogenesis [19, 20].

Fig. 1
figure 1

This figure was created with BioRender.com

Two classifications of EVs. A According to the origin of EVs, they can be classified as exosomes, microvesicles and apoptotic bodies. Exosomes are formed by early endosome formation, MVB development, plasma membrane transportation fusion and exocytosis. Microvesicles are formed by ectocytosis of the plasma membrane and apoptotic bodies are formed directly by means of membrane blebbing of apoptotic cells. B According to the particle size, EVs are divided into sEVs and lEVs

sEVs are nano-sized extracellular vesicles (40–200 nm) in diameter, one of the most common investigated EV subpopulations released by cells into biological fluids that contain a plethora of biomolecules such as proteins and nucleic acids [21, 22]. sEV may share some pathways mentioned above with exosomes. Due to their high representation of the cell of origin, sEVs serve as potent and clinically valuable tools for early diagnosis and prognosis of PCa [23]. Accumulating studies indicate that the levels of sEVs in body fluids have significant clinical relevance to cancer status [24, 25]. sEVs were reported to promote tumor growth by transporting their contents to recipient cells to regulate their cellular functions, and these biological active molecules have caught great attention in cancer research [26]. Compared to genetic information changes, the key regulatory proteins found in sEVs offer more direct information regarding disease progression. The communication of sEVs proteins plays a critical role in tumor progression and metastasis not only by regulating the tumor microenvironment (TME) around in situ tumor cells, but also by affecting distant metastatic sites and inducing the formation of pre-metastatic niches (PMN) [27]. This review discusses the biogenesis and contents of sEVs and summarizes the methods for isolation and characterization of sEVs. Furthermore, this article primarily focuses on the role of sEV proteins in the TME of PCa, as well as their application as biomarkers for PCa diagnosis and prognosis. Finally, the potential prospects of using sEV protein-based therapeutic approaches for PCa are also discussed.

sEV isolation and characterization

Despite the significant potential of sEVs in cancer diagnosis, prognosis and therapeutics, the biggest challenge we face since their discovery is how to efficiently isolate and purify sEVs. So far, various methods have been developed for the separation of sEVs and the detection of sEV proteins and nucleic acids.

Ultracentrifugation (UC)

UC is currently the most used separation method and includes differential ultracentrifugation and gradient density ultracentrifugation. Differential ultracentrifugation separates different extracellular components of the fluid sample according to their density, size, and shape under a certain centrifugal force. Due to its convenience, low technical requirements, and the ability to be used with large scale and without complex sample pre-treatment, differential ultracentrifugation has been widely regarded as the gold standard technology for the purification of sEVs for the past 30 years. However, the density gradient centrifugation can maximize the removal of interference caused by proteins. sEV samples prepared by differential ultracentrifugation often have low purity compared to density gradient ultracentrifugation [28]. Density gradient ultracentrifugation separates particles by layering them on different densities of biocompatible media (such as sucrose). Although sEVs obtained by density gradient ultracentrifugation have higher purity than those obtained by differential ultracentrifugation [29], this method requires large sample volumes, and is time-consuming, uses expensive equipment, and involves complex steps [30]. Currently, extensive research has successfully isolated sEVs from both PCa cells and the plasma of PCa patients using UC. These sEVs were utilized as a benchmark to evaluate the pros and cons of novel isolation techniques [31, 32]. One study employed UC to identify over 3000 sEV proteins extracted from urine samples of PCa patients. Significantly, the majority of these proteins exhibited the close links with sEVs, thus validating the reliability of EV extraction from urine samples [33]. Another study demonstrated that further refinement of UC methods significantly enhanced the enrichment of pertinent proteins in sEVs derived from urine samples of PCa patients, presenting an encouraging strategy for future investigations [34]. In a separate study, Dhondt et al. devised a separation strategy utilizing Optiprep density gradient centrifugation to enrich sEVs from urine samples of patients with benign prostatic hyperplasia (BPH) and PCa. Through this approach, they identified a total of 3686 proteins, including several hundred proteins not previously documented in known human urine sEV proteomes [35]. All these results suggest that UC is still a valuable standard tool for PCa EV isolation.

Size-based methods

Ultrafiltration techniques and size exclusion chromatography (SEC) are two size-based methods. Ultrafiltration techniques are similar to traditional filtration methods. Ultrafiltration employs ultrafine nanomembranes with various critical molecular weight cut-off (MWCO) values to separate sEVs from clinical samples or cell culture media. This is a size-based method for sEV separation. Compared to UC, ultrafiltration is less time-consuming and doesn’t require special equipment. However, filtration-based methods easily have blockage issues, and unavoidable absorption on membrane would result in a loss of sEVs and possibility of false negative molecular result. SEC is a method used to separate biological molecules based on the size of the sample and the pore size of the gel. Smaller molecules briefly pause in the matrix, resulting in a longer elution time through the chromatography column, while larger molecules are eluted first. Unlike filtration and centrifugation methods, SEC is a gentler technique that can preserve the biophysical and bioactive characteristics of sEVs [36]. This method is commonly used in human plasma samples for downstream analysis. A study successfully isolated sEVs from normal prostate RWPE1 cells using ultrafiltration and characterized their associated proteins [37]. Another study found that SEC demonstrated advantages of high purity and low protein contamination in the extraction of sEVs from the plasma and urine of PCa patients, ensuring the quality requirements for subsequent downstream analysis [38, 39]. Chen et al. developed a highly efficient sEV isolation platform called Exosome detection via the ultrafast-isolation system (EXDUS) that is based on ultrafiltration, which achieves ultrafast and efficient purification of sEVs through negative pressure oscillation and double-coupled harmonic oscillator-enabled membrane vibration. EXDUS enables maximum enrichment of sEVs from different types and volumes of biological fluids by changing the oscillation mode. Currently, the application of EXDUS in urine samples from bladder cancer and kidney cancer patients within the urogenital system has demonstrated significantly higher yields and purities of isolated sEVs compared to other isolation techniques. Successful downstream studies have been conducted using EXDUS for the satisfied results, indicating promising prospects for its wide application in PCa research in the near future [40].

Immunoaffinity capture

This method relies on specific immune interactions between target proteins (antigens) on the surface of sEVs and antibodies to obtain highly purified sEVs [41]. Immunoaffinity capture methods are an ideal platform for separating subsets of EVs with specific origins due to their ability to recognize specific biomarkers. This results in a high level of purity and selectivity. However, the high cost, low yield, and the need for additional elution steps limit their application [28]. The reliability of immunoaffinity technology has been substantiated in a PCa study. This method facilitates the high-purity isolation of sEV products from both PCa cells and patients’ plasma. Through continuous optimization of reagent selection and protocols, this method can further enhance the capture efficiency and sensitivity for downstream protein and RNA analysis. Immunoaffinity technology demonstrated applicability to complex and low-volume samples [42]. Moreover, a comparative investigation utilizing atomic force microscopy (AFM) and nanoscale flow cytometry (NFC) evaluated immunoaffinity approaches and commercial kits, indicating that immunoaffinity approaches more effectively eliminate the impact of plasma proteins on sEV purity in the plasma of PCa patients. This finding is very important in the pursuit of PCa-specific biomarkers, as it helps to minimize or diminish plasma protein interference in sEV products [43]. This approach holds promise in prostate cancer-derived extracellular vesicles (PCDEVs) biomarker research.

Commercial kits

Precipitation method mainly relies on using polymers to precipitate sEVs, typically using polyethylene glycol (PEG) as a medium. After co-incubating sample with PEG solution at 4 ℃ overnight, sEVs are collected by low-speed centrifugation. This method is simple and doesn’t require special equipment, often producing high yields of sEVs suitable for processing large samples [44]. However, it was found that nucleic acids, protein contaminants, and some polymer residues inevitably appear in the products, resulting in lower purity and the potential for false positives [45]. Nevertheless, this method is a promising, inexpensive, and fast strategy for sEV isolation.

Several studies have conducted evaluations on the effectiveness of different methods for isolating sEVs from PCa cells and patient plasma. These studies have found that the purity of sEVs obtained through precipitation methods is consistently the lowest [38, 46]. Furthermore, precipitation methods have been widely applied in the exploration of diagnostic biomarkers for PCa. Multiple studies have identified the diagnostic value of sEVs prostate cancer specific antigen (PSMA) and lncRNA in the urine of PCa patients using precipitation methods [47, 48]. Mercadal et al. discovered that the miRNA expression profile of sEVs extracted from the semen of PCa patients using various isolation methods, including precipitation, exhibited variations. The possible reason for these variations is that different extraction methods yield different sEV subtypes. However, these differences have minimal impact on the performance of the proposed novel diagnostic model using combined semen sEV miRNA and blood PSA levels for improving PCa diagnosis [49]. Generally, commercial kits are applicable in cell line sEV isolation or plasma and urine isolation for sEV miRNA analysis in PCa research.

Microfluidics

Microfluidics-based techniques are important tools for integrating the isolation and detection of sEV. They combine microfluidic technology with traditional isolation techniques and rely on the physical and chemical characteristics of sEVs, such as size, density, and surface antigens, to achieve rapid and high-purity separation. The resulting detection technology meets the requirements for high-throughput and high-precision [50]. However, this technology is still in the early stages of development and improvements are needed in the future. Microfluidics has garnered increasing attention in the realm of PCa research. A groundbreaking advancement comes in the form of a 3D Self-Assembled Nanostructured SiO2 Microfluidic Chip. This innovative chip facilitates the efficient separation and detection of sEVs from PCa cells and the plasma of PCa patients, while significantly reducing sample consumption compared to conventional methods. The chip has several notable advantages, including straightforward preparation, enhanced affinity between sEVs and the chip surface, and highly sensitive analysis. By utilizing the 3D-SiO2 porous chip equipped with three markers (CD81, EpCAM, PSMA), early diagnosis and risk stratification of PCa patients become feasible [51]. Additionally, a study has successfully developed an acoustic microfluidics-based technique for enriching sEVs from urine samples of PCa patients, demonstrating exceptional efficiency and continuous enrichment of sEVs. Notably, certain differentially expressed miRNAs identified in these sEVs potentially serve as biomarkers for PCa diagnosis [52]. This approach is promising and applicable in clinical settings due to its small sample volume, more sensitive and specific characteristics, portable and point of care for patients.

Combination approach

Individual isolation methods often have their inevitable drawbacks. In recent years, some researchers have proposed combining two or more separation techniques. Currently, microfluidics-based techniques occupy a prominent position in combined separation techniques. In addition to combining with different traditional isolation methods, individual traditional isolation method is also combined with acoustic, microfluidic viscoelasticity, and other techniques, having shown promising results in some studies [50, 53]. Furthermore, one study found that the combination of UC and SEC (qEV column) was more effective in enriching sEVs than either method alone, resulting in improved yield and efficiency in animal plasma sample [54]. With the growing interest in sEVs, future research will undoubtedly require a large number of purer sEV samples for mechanism investigation and therapeutic purpose. Whether combined separation techniques can overcome limitations in sEV isolation and achieve or approach our ideal requirements remains to be further studied. In PCa-related study, it was discovered that combined approach demonstrated greater value in sEV isolation. The combination of ultrafiltration and SEC in the separation of PCa urine samples significantly reduced contamination from non-EV proteins and ensured the maximum purity of sEVs [55]. It is worthwhile testing different combination approaches in clinical samples such as blood and urine to obtain purer sEVs for downstream analysis in PCa biomarker research. Table 1 summarizes each isolation method along with its respective advantages and disadvantages.

Table 1 Summary of the current EV isolation methods with advantages and disadvantages

Other new approaches recently developed

In recent years, several research teams have been dedicated to developing alternative novel methods for sEV isolation, which have garnered significant attention due to their unique double-layer lipid structure. Sun et al. discovered a novel and effective technique for sEV enrichment. This method utilizes the lipid bilayer structure of sEVs to selectively label the lipid components with trans-cyclooctene (TCO). The labeled sEVs are subsequently immobilized onto tetrazine (Tz)-grafted microbeads through a bioorthogonal click chemistry reaction, enabling the straightforward separation and collection of the captured sEVs. This approach shares some similarities with immunocapture techniques. Immunocapture relies on the availability of specific antigen molecules, and this method effectively overcomes the limitations of immunocapture, demonstrating a higher sEV capture rate in cancer cells compared to UC, ExoQuick, and other methods. Notably, this method has shown promising results in the downstream mRNA analysis of sEVs isolated from plasma samples of patients with Ewing sarcoma or pancreatic cancer, suggesting its diagnostic and monitoring potential [56]. Another study focuses on phosphatidylserine (PS), a lipid molecule present in the lipid bilayer structure. By exploiting the calcium-dependent binding of PS to TIM4, a high-purity isolation of intact sEVs was achieved [57].

An innovative approach utilizing dip-pen nanolithography creates microscale arrays of lipid patches for sEV capture, ensuring maximal preservation of RNA components within sEVs for subsequent analysis. This method demonstrates strong sensitivity and specificity [58]. Field flow fractionation (FFF), a sEV separation technique based on charge, leverages the differential charges carried by sEV subtypes to achieve efficient and high-purity label-free separation [59]. Asymmetrical flow field-flow fractionation (AF4) in combination with capillary electrophoresis (CE) is another label-free sEV isolation method. AF4 separates sEVs based on size, while CE further distinguishes sEVs of the same size but with different charge characteristics from abundant matrix components such as lipoproteins. The combination of AF4 and CE enhances separation efficiency and holds great value in studying sEVs and their subgroups. However, the carrying capacity of CE is limited, and its effectiveness for continuous processing of large samples remains a subject of debate, thereby limiting its widespread clinical application [60]. The emergence of these novel technologies has provided deeper insights into the physicochemical properties of sEVs and holds the potential to become routine isolation methods with ongoing advancements in the field. These novel EV isolation approaches have a great chance to be used in PCa research in the future.

Characterization of sEVs

sEVs possess the property mainly based on their distinctive characteristics such as their size, morphology, floatation density, and the presence of marker proteins. According to the Minimal Information for Studies of Extracellular Vesicles 2018 (MISEV2018) recommendations, EV identification includes western blot verification of EV-specific markers such as CD9, CD81, CD63, electron microscopy (EM) or nanoparticle tracking analysis (NTA) and at least two characterization methods needs to be performed [64]. The methods used to characterize sEVs are in line with their distinct features and can be broadly categorized into three types: quantitative, qualitative and single-vesicle characterization [65]. The summary of sEV characterizations applied in PCa is shown in Fig. 2. It is important to note that these classifications are intended to simplify the understanding and categorization of sEV characterization techniques, and are not strict definitions, as some techniques have overlapping features between quantitative characterization, qualitative characterization and single EV characterization. In practical applications, it is often necessary to combine multiple techniques to obtain comprehensive and accurate characterization results of sEVs.

Fig. 2
figure 2

This figure was created with BioRender.com

Small extracellular vesicle (sEV) quantitative, qualitative, and single EV characterizations. sEVs isolated from prostate cancer (PCa) blood or urine samples and cell lines can be characterized by size, morphology, concentration, and sEV markers. A quantitative characterization. This is performed by enzyme-linked immunosorbent assay (ELISA), dynamic light scattering (DLS), electron microscopy (EM), nanoparticle tracking analysis (NTA), resistive pulse sensing (RPS), multiple reaction monitoring (MRM), parallel reaction monitoring (PRM), liquid chromatography with tandem mass spectrometry (LC–MS/MS), surface plasmon resonance microscopy (SPRM), fluorescence correlation spectroscopy (FCS) and flow cytometry (FCM). B qualitative characterization. This is completed by Western blot (WB), next-generation sequencing (NGS), quantitative polymerase chain reaction (qPCR), MRM, PRM, LC–MS/MS, SPRM, FCS, FCM, stimulated emission depletion (STED) microscopy, RS and Atomic force microscopy (AFM). C single EV characterization. This is done by small-angle X-ray scattering (SAXS), DLS, EM, NTA, RPS, SPRM, FCS, FCM, Nano flow cytometry (nFCM), high-sensitivity flow cytometry (HSFCM), and high-resolution flow cytometry (hFCM), RS and AFM

Quantitative methods

Quantitative characterization is used to assess the success of sEV isolation and the quality of the products, including the yield and purity of biomolecules such as proteins, nucleic acids, and lipids. Techniques such as EM, NTA, flow cytometry (FCM), fluorescence correlation spectroscopy (FCS), dynamic light scattering (DLS), resistive pulse sensing (RPS) are primarily used to identify the total number of sEVs and quantify their yield by measuring the total number of particles within a certain size range [66]. Among these, NTA and FCS are the most used methods. Protein content is also an important indicator of purity, and enzyme-linked immunosorbent assay (ELISA) and mass spectrometry (MS) are representative techniques for quantifying it. In addition, surface plasmon resonance microscopy (SPRM) converts SPR response into surface binding mass for quantitative analysis of sEVs [67]. By combining with specific capture of target sEV proteins, it allows for the analysis of specific protein concentrations [68]. The ratio of protein content to total sEV particle count can help determine sample purity.

Qualitative methods

Qualitative methods are primarily employed to identify sEVs, validate proteomic and lipidomic identifications, and sequence coverage of DNA/RNA. Atomic Force Microscopy (AFM) is used to reveal the surface properties and structural features of sEVs for preliminary qualitative analysis [69]. Techniques such as Western blot (WB), FCM, stimulated emission depletion (STED) microscopy, and surface plasmon resonance microscopy (SPRM) are utilized to qualitatively characterize sEVs based on their protein markers [70]. Meanwhile, FCS labeled lipid-binding proteins, Raman spectroscopy (RS), and liquid chromatography with tandem mass spectrometry (LC–MS/MS) are used to identify lipid and protein markers, while multiple reaction monitoring (MRM), and parallel reaction monitoring (PRM) are used to validate identified peptides or lipids. Microarray technology, next-generation sequencing (NGS) and quantitative polymerase chain reaction (qPCR) are employed to analyze the genomic content of sEVs.

Single EV methods

The single EV characterization method focuses on individual sEV characteristics, including size, structure, and chemical composition. Atomic force microscopy (AFM), STED, SPRM, small-angle X-ray scattering (SAXS), and scanning electron microscopy (SEM) identify the typical cup-shaped structure of sEVs. NTA, DLS, EM, SAXS, FCM, and tunable resistive pulse sensing (TRPS) help identify the size of individual sEVs, while RS can identify their chemical composition. Nano flow cytometry (nFCM), High-sensitivity flow cytometry (HSFCM) and high-resolution flow cytometry (hFCM) are used to identify very small EVs and EV subpopulations.

These techniques help determine the purity and yield of products and choose the most appropriate separation method, as well as evaluate the therapeutic value of sEVs as drug carriers [71].

Mechanisms of sEV proteins in tumor microenvironment and prostate cancer pathology advancements

Increasing evidence has demonstrated that PCa cells release a higher amount of sEVs compared to normal cells [72]. sEV concentrations were increased as PCa progressed from low-grade to high-grade [73], indicating PCa-derived sEVs play an important role in cancer progression and metastasis. These PCa-derived sEVs may play a significant role in regulating communication between tumor cells, the tumor microenvironment (TME), and distant metastasis sites through both autocrine and paracrine mechanisms [74]. This promotes signal transduction and the formation of pre-metastasis niches (PMNs), ultimately contributing to tumor development and metastasis [75]. Notably, cancer cells require sufficient oxygen to grow, and studies indicate that cancer-derived sEVs serve as carriers of various proteins, RNAs and lipids, which re-program the secretion of growth factors and cytokines by endothelial cells, activate signaling pathways, and prompt perivascular cell migration and the formation of new blood vessels [76]. Such conditions create a favorable circumstance for cancer invasion and metastasis [77]. Furthermore, cancer cell-derived sEV proteins predominantly inhibit anti-cancer immune responses by impacting T cells, dendritic cells (DCs), NK cells, macrophages, myeloid-derived suppressor cells (MDSCs), and regulatory B cells, thus facilitating immune evasion [78]. Accumulated studies reveal that cancer-derived sEV proteins mediate therapeutic resistance in cancer cells through various mechanisms, such as pumping anti-cancer drugs out of cells, promoting epithelial-mesenchymal transition (EMT), and regulating signaling molecules to promote anti-apoptotic pathways [79, 80]. Additionally, due to their high abundance, strong stability, and specificity, cancer-derived sEV proteins can serve as promising biomarkers for diagnosis and prognosis in liquid biopsy. Overall, cancer-derived sEV proteins play critical roles in facilitating carcinogenesis, angiogenesis, and drug resistance, and enable cancer cells to evade the host immune system, and act as valuable diagnostic and/or prognostic biomarkers as shown in Fig. 3. In this section, we focus on discussing the roles and applications of sEV proteins in PCa TME.

Fig. 3
figure 3

This figure was created with BioRender.com

Overview of sEV proteins in prostate cancer angiogenesis, tumor growth, metastasis, drug resistance, immune suppression in the tumor microenvironment. A PCa cell-derived sEV proteins including Integrin α2, PSGR, EBAG9, Cav-1, AR-V7, CXCL14, Integrin αvβ6, Src, LRG1 promote angiogenesis, tumor growth, and metastasis in tumor cells, thereby accelerating tumor progression. B PCa cell-derived sEV proteins including Syntaxin 6, YAP1, P-gp converts drug sensitive PCa cells to drug resistant PCa cells, leading to the development of drug resistance. C PCa cell-derived sEV proteins including PGE2 and PD-L1 mediate the communication with CD8+ T cells, thereby participating in immune suppression. AR-V7 androgen receptor-v7, Cav-1 caveolin-1, CXCL4 Chemokine (C-X-C motif) ligand 14, EBAG9 estrogen receptor-binding fragment-associated antigen 9, LRG1 leucine-rich alpha-2-glycoprotein 1, MSCs mesenchymal stem cells, PCa prostate cancer, PSGR Prostate-specific G-protein coupled receptor, YAP1 Yes-associated protein 1, P-gp P-Glycoprotein, PGE2 Prostaglandin E2, PD-L1 programmed cell death ligand 1.

sEV proteins promote prostate cancer growth and metastasis

sEV proteins induce EMT in prostate cancer

Cancer progression is a dynamic and multistep process, and sEV proteins play a significant role in the development of PCa by regulating the physiological functions of surrounding cancer cells and TME. EMT is a process whereby epithelial cells transition into a mesenchymal stem cell state, and in cancer, EMT is linked to tumor occurrence, invasion, metastasis, and resistance to therapy [81]. Recent studies have investigated the role of sEV proteins in EMT in PCa. For instance, the sEV-mediated integrin α2 subunit enhances the activity of Vimentin, FAK, and ERK1/2 in PCa cells, inducing EMT and ultimately promoting the development of PCa towards a more invasive form [82]. sEVs carrying Prostate-specific G-protein coupled receptor (PSGR) were found to induce low-invasive PCa cells to complete EMT, leading to a more invasive and metastatic phenotype and knocking down PSGR inhibited tumor cell proliferation and cloning [83]. Tumor-derived sEV-mediated EBAG9 protein is a critical factor that promotes EMT of PCa cells while inhibiting cytotoxic T cells, thereby facilitating tumor progression [84]. Additionally, PCa-derived sEVs containing Cav-1 were reported to promote EMT in NEPC via the NF-κB signaling pathway [85].

sEV proteins promote prostate cancer proliferation

The proliferation of cancer cells is a major driving force behind tumor progression. Fast-dividing cancer cells require a high level of energy to sustain their growth, which triggers the transition from oxidative phosphorylation to glycolysis. Intercellular communication between cancer cells and surrounding normal cells involves energy-requiring processes such as internalization, making this communication process particularly important. Recent research has shown that sEVs secreted by normal prostate epithelial cells and PC3 PCa cells exhibit inconsistent ATPase activity. The ATPase activity of PC3 sEVs is low comparing to that in normal prostate cells, resulting in the more ATP production, which increases intercellular communication and promotes tumor cell proliferation [86]. Moreover, sEV AR-V7 was shown to positively regulate AR signaling, promoting the proliferation of PCa cells, and affecting tumor growth [87]. sEV protein CXCL14 was found to promote M2 macrophage polarization through the NF-κB signaling pathway, thereby facilitating EMT in PCa cells. Its downregulation inhibited the proliferation and invasion of PCa cells, but did not affect apoptosis in PCa cells [88]. Studies have also shown that sEVs from adipocytes induced glycolysis and increased the growth rate of PCa cells through the activation of Akt and subsequent stabilization of hypoxia inducible factor-1α (HIF-1α) [89]. Additionally, a study has identified 1474 proteins in seminal sEVs that overlap with the PCDEVs database. Some of these proteins are involved in biological processes such as metabolic reprogramming, energy pathways, cell growth and maintenance, and transport, laying the foundation for further screening of their role in PCa growth [90]. Apoptosis and cellular senescence are crucial signaling pathways regulated by sEV proteins in PCa. Apoptosis is the major form of cell death in physiological conditions while senescence results in permanent cell cycle arrest. Both pathways serve as obstacles to tumor development [91]. A recent study revealed that sEV miR-143, derived from MSCs, inhibits the expression of TFF3, thereby suppressing the proliferation of PCa cells and promoting apoptosis [92]. Additionally, cellular senescence is defined as a stable cell cycle arrest in the G1 phase, providing a mechanism to inhibit cancer cell growth. Research has found that PCa cells release an increased amount of sEVs through associated mechanisms during treatment-induced cellular senescence, which transfers protein and genetic information between cells, indicating a certain connection between sEVs and cell apoptosis [93]. Further study has found that sEVs isolated from mock-treated senescent human lung epithelial carcinoma (A549) cells contain functional PTEN, and their transfer to PTEN-deficient PC3 cells leads to growth arrest [94]. However, the interaction between cellular aging and apoptosis suggests that inhibiting cellular aging to slow down cell growth may not always suppress tumor growth. Senescent cells exhibit enhanced metabolic activity, which may potentially contribute to tumor progression and recurrence. Moreover, if the immune system fails to clear senescent cells, or cancer cells escape senescence, which may become resistant to apoptosis stimuli [91].

sEV proteins associate with prostate cancer angiogenesis

Balancing the interaction between angiogenesis and tumor growth in the future is a challenge we must face. Angiogenesis is a multi-step process in which tumors form a new vascular system that is crucial for their growth [95]. Studies have shown that sEV αvβ6 integrin can regulate the level of survivin, increase the negative regulatory factor of angiogenesis pSTAT1, and promote PCa angiogenesis [96]. Additionally, it was found that the sEV Src protein in PCa cells activates focal adhesion kinase (FAK) through integrins, leading to angiogenesis and metastasis [97]. Moreover, high expression of α2-glycoprotein 1 (LRG1) was detected in the plasma sEVs of CRPC patients, further confirming sEV proteins play an important role in promoting angiogenesis and are associated with PCa progression [98]. These findings suggest that sEV proteins are a key player in promoting angiogenesis and the development of PCa.

sEV proteins influence prostate cancer fibroblasts and ECM remodeling

In the TME of PCa, the extracellular matrix (ECM) plays a crucial but often overlooked role. Remodeling of the ECM significantly impacts tumor cell proliferation and metastasis. In a DU145 and fibroblast co-culture model, the dynamic interaction between cancer cells and fibroblasts leads to pronounced changes in the ECM, thereby facilitating the invasive potential of DU145 cells [99]. During the progression of PCa, the prostate stroma undergoes phenotypic alterations, gradually transforming into cancer-associated fibroblasts (CAFs). This transformation triggers ECM remodeling, ultimately enhancing the invasive and progressive capabilities of PCa [100]. Consequently, CAFs assume a central role in the TME, gradually reshaping the biological functions of cancer cells through frequent communication. Recent research has shed light on the pivotal role of sEV TGFβ1 in driving fibroblast differentiation into myofibroblast-like cells, thus promoting the formation of a tumor-supportive stromal phenotype. These findings underscore the indispensable contribution of sEV TGFβ1 to the development of a pro-tumorigenic ECM [101]. Moreover, investigations have demonstrated that treating primary rat prostate fibroblasts with sEVs derived from metastatic PCa, as opposed to those derived from indolent non-metastatic PCa, significantly upregulates the mRNA expression of growth factors and cytokines, indicating fibroblast activation [102]. Furthermore, treatment of prostate fibroblasts with sEVs isolated from urine samples of PCa patients and healthy individuals elicits distinct transcriptional responses, whereas the transcriptional response in foreskin fibroblasts exhibits remarkable similarity. These findings provide further evidence that sEV derived from PCa patients exert a profound influence on the phenotypic and functional changes of fibroblasts [103]. There are reasons to speculate that sEV proteins of PCa cells play a significant role in this process.

sEV proteins associate with inflammation and immune escape in prostate cancer

The inflammasome is an important component of the TME, and various subsets of cells in the TME activate the inflammasome to regulate the progression of malignant tumors. Currently, researchers generally believe that the role of the TME in promoting/inhibiting tumor progression mainly depends on the type of tumor and inflammasome [104]. Inflammation is also a significant risk factor in the development of PCa. For example, the inflammasome NLRP3 was shown to enhance the proliferation and migration ability of PCa cells by activating caspase-1, thereby promoting the malignant progression of PCa [105]. Further studies have found that PC3-derived sEV induced the activation of NLRP3 inflammasome and caspase-1 through the ERK1/2 pathway, thereby creating a PCa inflammatory microenvironment [106]. Recently, it was discovered that through proteomic analysis and subsequent Ingenuity pathway analysis (IPA) of the sEVs derived from PCa cells of African American individuals, it was found that proteins loaded in sEVs correlate strongly with acute inflammatory response signalling pathways. In addition, Filamin A is closely associated with the occurrence of this inflammatory response. However, further investigation is still needed to elucidate the specific role of Filamin A in the transmission of inflammatory pathways [107].

Cancer cells invade various organs and tissues and spread to the body through metastasis, leading patients to an irreversible terminal stage of the disease. This process involves multiple mechanisms and steps [27]. In PCa, the primary sites of metastasis are the lymph node, bone, lungs, liver, and brain. The selection of metastatic sites by tumor cells is highly correlated with integrin avβ6, which is found in PCa-derived sEVs [108]. Further study shown that sEV integrin avβ6 inhibited the STAT1/MX1/2 signal transduction and reprogramed mononuclear cells into M2 tumor-supportive phenotypes, promoting the progression of PCa to the CRPC phenotype during metastasis [109]. The sEV protein PLD2 derived from C4-2B cells was reported not only to stimulate sEV secretion but also increase osteoblast activity, thus participating in the communication between PCa cells and the bone metastatic microenvironment [110]. PMN formation refers to the communication between tumor cells and cells in distant pre-metastatic organs, which helps to establish a tumor environment conducive to invasion, immune escape, and metastasis formation. sEVs serve as an effective medium in this process. The regulation of matrix metalloproteinases (MMPs) plays a crucial role in cancer metastasis [111]. Studies have found that under hypoxic conditions, sEVs secreted by PCa cells enhance the activity of MMPs in the hypothesized metastatic site, thereby reshaping the PMN [112]. Furthermore, it has been reported that the cholesterol homeostasis in bone marrow cells affects the reception and transduction of premetastatic signals mediated by sEVs released by PCa cells [113]. In recent years, there has also been a growing recognition of the role of sEV proteins in the PMN of PCa. A recent study has shown that sEVs released from C4-2B PCa cells stimulate the formation of PMN via the HIF1α-dependent pathway, which is mediated by pyruvate kinase M2 (PKM2) [114]. Furthermore, the EMT process is also a key factor in tumor cell metastasis, with the current consensus being that EMT is required during local invasion while the mesenchymal-epithelial transformation (MET) process is necessary during distant metastasis [115]. Previous studies demonstrated that the sEV protein Tspan8 in breast cancer cells plays a role in facilitating the progression of metastasis by mediating phenotypic changes in MET [116]. PCa and breast cancer share some similarities in inducing MET [117], Further research is still needed to support the involvement of sEV proteins in mediating MET occurrence in PCa.

sEVs promote pre-metastatic niche formation in prostate cancer

Although PCa bone metastasis is predominantly associated with osteoblastic changes, studies have found that osteoclasts still play a crucial role in PCa bone metastasis. One study revealed that sEVs derived from PCa cells are essential mediators in maintaining bone homeostasis. These sEVs promote osteoclast differentiation and inhibit the formation of osteoblasts in vivo and in vitro, thus creating a pre-metastatic osteolytic niche for PCa metastasis [118]. A recent study by Urabe et al. confirmed this finding, demonstrating that the protein CDCP1 on sEVs derived from metastatic PCa cells promotes the formation of mature osteoclasts. Furthermore, compared to localized PCa patients, CDCP1 showed high expression on plasma sEVs in patients with bone metastasis [119]. These results provide new insights for elucidating the mechanisms of osteoclast involvement in bone metastasis and identifying potential protein biomarkers of PCa bone metastasis for monitoring progression and evaluating treatment effects.

Overall, the progression and metastasis of PCa rely on the formation of the TME, wherein sEV proteins mediate cellular communication between cancer cells and host cells, influencing the phenotype or function of immune cells, fibroblasts, epithelial cells, inflammasomes, and endothelial cells. Figure 4 illustrates the alterations in each component regulated by sEV proteins in the PCa TME, including the polarization of CAFs, EMT, angiogenesis, disruption of immune homeostasis leading to an immune-evading TME, the promotion of cancer cell proliferation and invasion, proliferation, accelerated activation of inflammasomes, and establishment of a PMN in distant sites.

Fig. 4
figure 4

This figure was created with BioRender.com

Outline of the alterations in each component regulated by sEV proteins in the PCa TME. In addition to exerting pro-tumorigenic effects at the primary site of PCa, sEVs proteins also play a role in creating conditions for metastasis at distant sites. A sEV proteins promote the differentiation of fibroblasts into cancer-associated fibroblasts (CAFs) and remodel the extracellular matrix (ECM). B sEV proteins promote epithelial-mesenchymal transition (EMT) in PCa cells. C sEV proteins facilitate abnormal angiogenesis, providing nutrients for tumor growth. D sEV proteins promote the transformation of macrophages into the M2 phenotype, recruit pro-tumorigenic regulatory T cells (Tregs), induce CD8+ T cell apoptosis, and inhibit natural killer (NK) cell cytotoxicity, thus facilitating immune evasion. E sEV proteins enhance PCa cell proliferation. F sEV proteins promote the activation of inflammasomes, leading to the occurrence of inflammation in the tumor microenvironment (TME). G sEV proteins act on distant sites to establish pre-metastatic niches (PMNs)

sEV proteins in PCa drug resistance

Drug resistance is a major challenge in cancer treatment, and sEV proteins play a significant role in cell communication during the drug resistance process of cancer cells. The molecular mechanisms by which sEV mediate cancer resistance are generally categorized into three groups: (1) cancer cells excrete chemotherapeutic drugs through sEVs, (2) sEVs carry drug-resistant cargo and communicate with drug-sensitive tumor cells, and (3) sEVs act as bait targets in therapeutic drug [120]. Accumulating data shown that the mechanism of PCa drug resistance primarily belongs to the second category, where sEV proteins mediate intercellular communication between drug-resistant and drug-sensitive cells, modifying gene expression in the drug-sensitive cell population, enabling them to resist apoptosis when exposed to drugs [121]. One recent report suggested that sEVs mediated enzalutamide resistance (EnzaR) and treatment induced PCa neuroendocrine differentiation in PCa cells, and inhibiting sEV release partially restored the sensitivity of EnzaR PCa cells [122]. Another study also demonstrated the involvement of sEV release in the mechanism of EnzaR PCa. The inhibition of sEVs significantly suppressed the survival ability of resistant cells, but further research support is needed to determine whether sEV proteins are involved in the development of resistance [123]. In addition, increased expression of YAP1 was observed in sEVs isolated from EnzaR PCa patients’ serum, revealing that EnzaR sEVs enhanced LNCaP cell resistance by increasing YAP1 function [124]. Kharaziha et al. discovered that docetaxel-resistant DU145 cells release more sEVs than docetaxel-sensitive DU145 cells. Additionally, they identified a group of enriched proteins in the sEVs secreted by docetaxel-resistant DU145 cells [125]. sEV proteins also play a role in chemoresistance, as PCa cells carrying Cav-1 in metastatic CRPC (mCRPC)-derived sEVs acquire resistance to radiotherapy and docetaxel treatment [85]. Corcoran et al. reported that docetaxel-sensitive PCa cells (DU145, 22Rv1, and LNCaP) became resistant to docetaxel after exposure to docetaxel-resistant DU145 and 22Rv1 variants (DU145RD and 22Rv1RD), possibly due to the release of MDR-1/P-gp transporter proteins in sEVs. Similarly, Kato et al. found that the sEV p-gp level in docetaxel-resistant PC3 cells was higher than that in PC3 cells. Furthermore, downregulation of sEV p-gp reduced PC3 cell resistance to docetaxel, indicating that sEV-mediated protein transport plays a critical role in the development of PCa cell resistance [126]. Although the specific mechanism by which sEV proteins contribute to drug resistance in PCa remains unclear, inhibiting their formation and release may provide a new therapeutic strategy for treating this disease. Furthermore, targeting the PCa-derived sEV proteins through precision medicine may offer to overcome drug resistance and improve the current therapy efficacy. The role of sEV proteins in regulation of PCa drug resistance in TME is shown in Fig. 3.

sEV proteins modulate immune escape during PCa progression

Cancer-derived sEV proteins impact the function and composition of immune cells within TME. Studies have shown that cancer-derived sEVs inhibited innate immune responses by mobilizing MDSCs, regulating/activating tumor-associated macrophages (TAMs), and neutrophils [127]. There is also direct evidence supporting the involvement of sEVs in immune evasion by PCa cells. For instance, sEVs derived from PCas were found to weaken the cytotoxic function of NK cells and CD8+ T cells by downregulating the activating receptor NKG2D, thus promoting cancer immune escape [128]. One recent report further demonstrated that avβ6 integrin in PCa is essential for monocytes to differentiate into M2-type immune suppressive cells. Interestingly, in another study, inhibiting Rab27a to block sEV release on DU145 cells significantly enhanced the tumor antigen-specific T cell response in dendritic cells (DCs). sEVs carrying PGE2 was found to induce the CD73 expression in DCs, resulting in T cell dysfunction in an adenosine-dependent manner [129]. Additionally, MDSCs are an important immunosuppressive cell type that mediates immune escape in the TME. sEVs derived from PCa cells were found to upregulate chemokine receptor 4 (CXCR4) by activating the TLR2/NF-κB signaling pathway, promoting the migration of MDSCs to the TME, and disrupting the balance of immune suppression [130]. The high expression of programmed cell death-ligand 1 (PD-L1) in PCa has recently received considerable attention, leading to the development of immune checkpoint inhibitors. The latest research has found that PD-L1 is also expressed on sEVs in some cancers, and inhibiting PD-L1 on sEVs through the Ca2+ channel ORAI1 induced anti-cancer responses [131]. In PCa, when PD-L1 binds to programmed cell death protein-1 (PD-1) on T cells, the anti-cancer activity of T cells was suppressed, and apoptosis was enhanced [132]. As PCa is a “cold” tumor, the ability of immunotherapy for inducing immune response is low compared with other “hot” tumors such as melanoma, lung cancer, breast cancer. So, it will be important and interesting to investigate sEV proteins as a therapeutic target for developing new immunotherapy to overcome immune escape in PCa patients, especially in CRPC and bone metastasis. The role of sEV proteins in the regulation of PCa immune escape is shown in Figs. 3 and 4.

sEV proteins as diagnostic or prognostic biomarkers in PCa liquid biopsy

Liquid biopsy is a comprehensive and non-invasive diagnostic tool that retrieves tumor-related biomarkers from biological fluids, mainly including CTCs, ctDNA and EVs. Liquid biopsy plays a significant role in early diagnosis, risk stratification, residual monitoring, and recurrence of cancer. Among the three types of biomarkers, sEVs have gradually become a more widely used platform due to their numerous advantages. The dynamic variability of the mechanisms determines the heterogeneity of sEVs, which communicate between cells by carrying various contents such as proteins, lipids, and nucleic acids (mRNA, LncRNA, microRNAs, and DNA) [133]. The Exocarta database has so far identified 9769 proteins, 3408 mRNAs, 2838 miRNAs, and 1116 lipids in sEVs, and the content and type of these materials vary in different cell lines, indicating their complexity and potential functional diversity [134]. sEVs exhibit high heterogeneity, which is manifested in their size, content, function, and cell origin. Some sEV proteins display cell and tissue specificity, providing a basis for the study of related disease markers. Additionally, some surface proteins serve as sEV markers and play an important role in biogenesis. Table 2 summarizes the sEV protein markers related to PCa and discovered in the last 5 years for their source, application, and function, which will be discussed in the following.

Table 2 Summary of PCa-related sEV proteins in source, applications, and function

Recently, potential sEV protein biomarkers have been identified in both blood and urine in PCa patients (Table 2). These biomarkers have demonstrated a better performance and accuracy in determining cancer progression compared with traditional cancer markers such as prostate specific antigen (PSA). For instance, using MS-based proteomics, Bhagirath et al. discovered that sEV protein TSP1 is a highly sensitive and non-invasive biomarker for diagnosing neuroendocrine prostate cancer (NEPC) [135]. Additionally, another report showed that plasma sEV CLD3 has a good diagnostic value in distinguishing PCa patients with a Gleason score ≥ 8 from those with scores of 6–7 and benign prostatic hyperplasia (BPH) patients [137]. Park et al. recently demonstrated the potential use of sEV protein PSMA as a diagnostic marker in PCa [138]. Plasma/serum levels of STEAP1[140], CA IX [141], Del-1[143], and ephrin A2 [144] were also reported to distinguish between PCa patients and healthy individuals and/or BPH patients. Gamma-glutamyltransferase 1 was shown to be expressed at higher levels in urinary sEVs and tissues of PCa patients [145]. Zhang reported that urine sEV protein markers (SERPINA3, LRG1, and SCGB3A1) were higher in PCa patients (n = 20) than in healthy controls (n = 20), with SERPINA3 showing the highest correlation in distinguishing patients from healthy individuals [147]. Additionally, FABP5 was found to be not only higher in the PCa group compared with the control group, but also significantly correlated with Gleason grading in urinary sEVs [146].

sEV proteins are also used as non-invasive prognostic biomarkers of PCa. In a study conducted by Krishn et al., sEV αvβ3 integrin was demonstrated to be served as a non-invasive biomarker in the blood of PCa patients, and act as a molecule of intercellular communication that leads to disease progression [142]. Elevated expression of ACTN4 was found in the plasma sEVs of castration resistant prostate cancer (CRPC) patients and untreated PCa group, as well as in DU145 and PC3 cells. Downregulation of ACTN4 expression inhibited cancer cell growth, indicating that sEV ACTN4 may be a prognostic marker for evaluating tumor burden [136]. Increased expression of sEV PD-L1 was found to be associated with Radium-223 radiotherapy, indicating this sEV protein may aid in monitoring the response of PCa to radiation therapy [139].

Currently, the exploration of PCa-derived sEV proteins as biomarkers is still in infancy. Although these studies have shown promising results in screening or as diagnostic/prognostic biomarkers, further clinical translation faces significant obstacles. Firstly, there is a lack of large set of clinical samples or reliable cutoff values to evaluate its real clinical diagnostic values. Secondly, the potential sEV protein biomarkers identified need to be compared with traditional clinical diagnostic tools such as blood PSA test, MRI examination and tissue biopsies for sensitivity and specificity. Thirdly, there is lack of functional and mechanistic studies to investigate their roles in PCa progression and metastasis. If these issues can be addressed, and clinical challenges may be overcome, sEV proteins have the good potential to become new diagnostic tools for PCa diagnosis and prognosis in the future.

sEV protein based potential therapy in prostate cancer

Due to the natural intercellular communication function, good biocompatibility, low immunogenicity, low toxicity, long blood circulation capability, biodegradability, and the ability to cross various biological barriers, sEVs have been emerging as a promising drug delivery approach in cancer treatment [148]. Due to the widespread adoption of sEV proteomics analysis, the types and functions of proteins derived from sEVs obtained from cell lines or bodily fluids are continuously being discovered. This has provided a new avenue of hope for utilizing sEV proteins in the treatment of cancer. Currently, there are three potential directions for cancer therapy based on sEVs proteins:

Carrying therapeutic protein for targeted cancer therapy

Considerable efforts have been made to investigate the capacity of sEVs as carriers for cargo delivery while ensuring their toxicity towards cancer cells. sEVs have been a focus of research as carriers of chemotherapy drugs. It was shown that sEVs derived from PC3 cells increased the solubility of paclitaxel (PTX) in aqueous solutions compared to the control group of pure water, indicating their potential as a carrier [71]. Saari et al. discovered that sEVs carrying PTX entered LNCaP and PC3 cells through endocytosis and exerted cytotoxic effects on the cells. However, a new challenge has emerged: Certain types of sEVs without drugs can increase the vitality of cancer cells and reduce the cytotoxicity of PTX [149]. Furthermore, there were reports indicating that engineered sEVs, Exo Ce6+R848, created by co-cultivating the photosensitizer Chlorin e6 (Ce6), the immune adjuvant R848, and HEK 293 T cell-derived sEVs, synergistically converted M2 macrophages into M1 macrophages upon ultrasound irradiation, further activating effector T cells and creating an immune-promoting microenvironment [150]. Furthermore, a study discovered that the binding of serum-derived sEVs with TGFβRI kinase inhibitors or TLR7/8 agonists effectively inhibited the migration of PC3 cells through endocytosis. This binding also enhanced the release of pro-inflammatory cytokines from macrophages and DCs, ultimately leading to the occurrence of anti-tumor immune responses. The results of this study suggest that it is possible to achieve engineered sEV-based therapy for PCa in the future using TGFβRI kinase inhibitors and TLR7/8 agonists [151]. Recent research has found that sEV shows promising results and biocompatibility in the treatment of PCa. For example, sEVs derived from Akkermansia muciniphila when intravenously injected into PCa mouse Xenograft mode, resulted in an increased proportion of CD8+ T cells for granzyme B (GZMB) and interferon-gamma (IFN-γ) lymphocytes, leading to an increased M1 macrophages and a decreased M2 macrophages. Further in vitro experiments demonstrated that sEVs derived from Akkermansia muciniphila inhibited the proliferation and invasion of PCa cells [152]. PCa arises from a cancer stem or progenitor cell with homogeneous characteristics. Treatment with miR-let-7c loaded onto mesenchymal stem cell (MSC) sEVs resulted in a significant reduction in the proliferation and migration of CRPC-like PC3 and CWR22Rv1 cells, achieving a possibility of sEV therapy for CRPC [153]. These studies indicate that regardless of whether the sEVs are compatible with biomacromolecules or drugs, they demonstrate strong loading capacity. This lays the foundation for further research on their ability to carry therapeutic proteins and counteract the progression of PCa.

A study found that in a syngeneic mouse model, sEVs carrying the surface membrane protein SIRPα were transported to tumor sites, disrupting the CD47-SIRPα interaction and increasing the ability of macrophages to engulf tumor cells. This led to the inhibition of tumor growth. Additionally, it enhanced T cell infiltration and increased the possibility of releasing innate and adaptive anti-tumor responses through CD47-targeted therapy [154]. Currently, research on using sEV as carriers to deliver sEV proteins for the treatment of PCa is still relatively limited. The current research on sEV proteins as therapeutic carriers is still in the early stage. However, the aforementioned studies provide a new approach for PCa therapy. Regardless of the source of sEVs or the different endogenous contents and exogenous drugs they carry, they demonstrate a good ability to target tumor cells while ensuring sufficient safety and efficacy. The diversity of sEVs and their associated cargo set a solid foundation for personalized cancer treatment.

sEV proteins as novel targets for therapy

sEVs can function as mediators of intercellular communication through paracrine or autocrine signaling. As mentioned earlier in this article, recent proteomic studies on sEV proteins in PCa cell lines, plasma, tissues, and other sources have revealed their involvement in sEV biogenesis, cargo loading, transport, and their subsequent impact on PCa cell development, progression, and prognosis. At each of these stages, sEV proteins play a crucial role. Therefore, the sEV proteins released by PCa cell lines, as depicted in Fig. 3, represent potential therapeutic targets. For instance, PD-L1, frequently overexpressed in PCa, poses challenges for immune checkpoint inhibitors due to the presence in the TME. In recent years, it has been discovered that tumor cells exert immune suppression by secreting sEVs carrying PD-L1, which binds to PD-1 on T cells. Consequently, this phenomenon has gained significant attention. One study found that Sulfisoxazole effectively inhibited plasma sEV PD-L1 levels in tumor-bearing mice, preventing CD8+ T cells from being depleted by PD-L1. This approach successfully overcame immune evasion mechanisms employed by cancer cells, offering a new therapeutic option for PCa treatment [155]. Furthermore, during the development of PCa, the role of immunosuppressive cells has been observed. M2 macrophages, commonly believed to communicate with tumor cells through paracrine signaling, were found to be associated with poorer prognosis and survival rates [156]. A study revealed that co-culturing M2 macrophages with PCa cells unveiled the M2 cells’ ability to enhance the NOTCH signaling, promoting PCa invasiveness. The intercellular communication mediated by M2 macrophages has received significant attention [157]. In a recent study by Cui et al. co-culturing M2 macrophages and colon cancer cells demonstrated that sEV protein ferritin heavy chain (FTH1) was transferred from M2 macrophages to colon cancer cells, promoting their proliferation. This finding highlights the importance of sEVs as essential mediators in intercellular communication. The sEV target protein FTH1 on M2 macrophages provides us with a potential therapeutic approach [158]. In a recent study using a preclinical tumor model, researchers discovered that sEVs derived from M2 macrophages downregulated tumor cell MHC-I expression through the delivery of apolipoprotein E (ApoE). This suppression inhibited tumor intrinsic immunogenicity, ultimately leading to resistance to immune checkpoint blockade (ICB). However, the ApoE ligand EZ-482 reversed the M2-sEV-induced ICB resistance. The results of this study suggest that ApoE ligand EZ-482 could be beneficial for tumor patients, particularly those with M2-rich tumors such as PCa, providing a theoretical basis for combined immunotherapy with ICB.

Modifications of sEV proteins

Current research primarily focuses on modifying sEV proteins to develop sEV vaccines and enhance the loading and delivery capacity of sEV-mediated biomolecular cargo. Sipuleucel-T, an FDA-approved cancer vaccine targeting prostate acid phosphatase (PAP) antigen in PCa, consists of autologous peripheral blood mononuclear cells enriched with autologous DCs and has shown the survival benefits in patients with CRPC [159]. Studies have found that sEVs derived from DCs activate CD4+ and CD8+ T cells, triggering anti-tumor immune responses [160]. Furthermore, the composition and function of sEVs can be altered by adjusting cell types, isolation, and purification conditions to meet the personalized treatment demands of vaccines. Therefore, researchers are exploring the use of sEVs as vaccines for PCa treatment. As early as 2011, a high attenuated vaccine called MVA-BN was selectively engineered to incorporate PSA or PAP. Targeting of sEVs was achieved by fusing the antigens to the milk fat globule-EGF factor 8 (MFG-E8) C1C2 domain. Results from treating PCa mouse models with these vaccines showed that the group treated with MVA-BN-PSA-C1C2 exhibited increased immunogenicity against PSA compared to the control group, and it improved the efficacy of anti-tumor therapy [161]. Shi et al. anchored IFN-γ fusion protein to the surface of sEVs derived from PCa cells to develop a novel sEV vaccine. This vaccine demonstrated the ability to increase the population of M1 macrophages in the body and downregulate the expression of vascular endothelial growth factor receptor 2 (VEGFR2), further induced the production of specific antibodies against sEVs which weakened the pro-tumorigenic effects of PCDEVs, and inhibited tumor growth [162]. The development of tumor vaccines based on sEVs still faces many challenges. For example, cold tumors like PCa have a unique tumor immune-suppressive microenvironment, making them insensitive to immunotherapy [163]. Additionally, they exhibit different antigen expression patterns [164], raising concerns about whether sEV-based vaccines can effectively cover antigen variations. Furthermore, there are difficulties in production, optimization of delivery, and determining the therapeutic efficacy and dosage, all of which are inevitable obstacles. Nevertheless, understanding the immune evasion mechanisms of PCa is our primary goal in developing sEV-based vaccines. Since the discovery that functional sEV RNAs and proteins can mediate communication between EVs and tumor cells, there has been great interest in understanding how bioactive molecules are loaded into sEVs and delivered to tumor cells.

Understanding the regulatory mechanisms that influence loading and delivery efficiency is crucial to harnessing the full potential of sEVs as a tool for tumor treatment. Current research reveals the involvement of sEV proteins in these processes, suggesting that modulation of loading and delivery can be achieved through modifications of sEV proteins. Es-Haghi et al. developed a fusion protein (hCD9.hAGO2) that combines sEV membrane protein CD9 with RNA-binding protein AGO2. Compared to sEVs isolated solely by overexpressing the desired miRNA or shRNA, this fusion protein exhibited a higher level of miRNA and shRNA loading capacity and was effectively delivered to recipient cells [165]. Another study utilized a targeted and modular EV loading (TAMEL) system, which involved fusing the MS2 bacteriophage capsid protein with sEV-associated proteins. This approach resulted in a six-fold increase in RNA loading compared to the control group. However, when this system was applied for delivery to PC3 cells, it was found that this modification reduced the RNA payload in sEVs and restricted their release in the cytoplasm. This could be due to the modification altering the subcellular localization, thereby affecting the interaction between sEVs and recipient cells and limiting endosomal escape. This suggests the need for further optimization of the modification strategy to gradually enhance the cargo RNA loading capacity and release efficiency [166].

All these studies indicate that sEVs are promising drug delivery candidates, although their use also has some limitations, such as insufficient targeting ability, which limits their clinical application as therapeutic carriers. Currently, researchers are exploring ways to enhance the natural targeting abilities of sEVs through modifications. This involves modifying sEVs to improve their existing targeting capabilities or to enable them to target cells that they normally wouldn’t. One promising application of such modifications is the construction of a new type of cell-derived, surface-modified liposome carrier for nanodrug therapy. For example, Pan et al. developed a nanovector called Exo-PMA/Fe-HSA@DOX that was able to block the EGFR/AKT/NF-kB/IkB signaling pathway and achieve targeted therapy for PCa [167]. Altanerova et al. showed that MSCs-Exo modified with superparamagnetic iron oxide nanoparticles and exposed to an external alternating magnetic field induced toxicity in PC3 cells [168]. More recently, a novel nano-platform that mimics sEVs has been developed for loading tumor therapeutic drugs. This platform exhibits similar drug loading capacity and targeting ability for tumor cells as nature sEVs but is more controllable and productive [169]. Currently, there have been studies on the generation of sEV-mimetic nanovesicles from standardized MSCs derived from human induced pluripotent stem cells (iPSCs). A comparison between these nanovesicles carrying docetaxel and free docetaxel in a mouse model of PCa revealed that the docetaxel-loaded sEV-mimetic nanovesicles exhibited enhanced tumor targeting ability and significantly suppressed tumor growth [170]. In addition, Severic et al. engineered prostate specific membrane antigen (PSMA)-targeting peptides onto the surface of sEV mimetics, which effectively targeted PSMA-positive PCa cell lines (specifically LNCaP and C4-2B). These engineered vesicles exhibited excellent targeting ability in both in vitro and in vivo experiments [171].

Overall, modifying sEVs holds great potential for advancing nanodrug therapy. Although endogenous delivery vehicles, including sEVs, are safe and efficient drug delivery system (DDS), they still face significant challenges, such as the unfavorable characteristics of sEVs, low volume, high heterogeneity, complex cargo, and difficulties in characterization, which may hinder their clinical translation. Figure 5 illustrates three current potential sEV proteins-based PCa treatments.

Fig. 5
figure 5

This figure was created with BioRender.com

Three potential sEV protein-based prostate cancer treatments. A sEVs exert cytotoxic effects by delivering therapeutic sEV proteins to cancer cells. B Cancer-derived sEV proteins serve as therapeutic targets. C Modification of sEV proteins can be employed to develop vaccines and enhance the payload capacity of sEV-RNA

Conclusions and perspectives

PCa-derived proteins are important for intercellular communication and transport, and they play a critical role in the progression of the disease. As we gain a deeper understanding of the roles and signaling pathways mediated by sEV proteins, we can further elucidate the mechanisms underlying PCa progression. sEV proteins are stable and present in much higher quantities in patients than in healthy individuals, making them a promising biomarker for diagnosing and prognosing PCa progression and treatment response. The EMT mediates changes in multiple steps during PCa deterioration, highlighting the need to identify common upstream pathways that regulate sEV protein mediated EMT. Developing sEV-based therapies that target multiple pathways may be necessary. sEV proteins participate in PCa metastasis through multiple mechanisms, including promoting tumor cell growth and proliferation, inducing angiogenesis abnormalities, facilitating EMT, remodeling the TME, and facilitating PMN formation. Understanding the specific roles of these sEV proteins in PCa metastasis may help us identify therapeutic targets and ultimately inhibit their pro-metastatic functions, thereby facilitating the development of precision treatment strategies based on sEV proteins.

PD-1/PD-L1 are crucial negative co-stimulatory molecules in PCa that mediate tumor immune evasion, and their role has been validated in sEVs. Their expression also increases after radiotherapy, suggesting that developing combination therapies with sEV immune checkpoint inhibitors is a promising new approach. In this review, we innovatively summarize three main therapeutic strategies of sEV proteins in PCa. sEV proteins provide insights into the mechanisms of sEV biogenesis, protein–protein interactions, and receptor targeting, offering therapeutic potential. sEV protein-based therapy may provide a new avenue for the treatment of ENPC and CRPC. However, unfortunately, the application of sEV protein-based therapy in PCa is not yet widespread. This is primarily due to the lack of protein carrier amplification procedures in sEV proteomics analysis, which adds clinical complexity. Additionally, the immunosuppressive microenvironment in PCa hinders the development of immunotherapy. Currently, all sEV protein-based treatments involve the immune microenvironment composed of immune cells and immune suppressor cells.

Overall, sEV proteins play a pivotal role in prostate carcinogenesis, and have the great potential to be used as diagnostic, prognostic, and therapeutic agents in PCa. The emergence of single-vesicle analysis allows us to explore the heterogeneity and functions of different subpopulations of sEV and their cargo proteins in the TME. It has made significant contributions in revealing the diversity of EV subpopulations and their protein content, as well as their potential roles in the progression and treatment of PCa. While progress has been made, there is still a long way to go for the translation of sEVs from laboratory research to clinical applications. It is important to acknowledge that the lack of standardized techniques for sEV isolation and characterization is a significant limitation, and large-scale prospective studies are necessary to validate the diagnostic accuracy of sEV proteins. Despite these challenges, we believe that the value of sEV proteins in the field of cancer will be realized in the near future with the establishment of standardized processes from production to effective application.

Availability of data and materials

Not applicable.

References

  1. Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA Cancer J Clin. 2023;73:17–48.

    Article  PubMed  Google Scholar 

  2. Ju W, Zheng R, Zhang S, Zeng H, Sun K, Wang S, Chen R, Li L, Wei W, He J. Cancer statistics in Chinese older people, 2022: current burden, time trends, and comparisons with the US, Japan, and the Republic of Korea. Sci China Life Sci. 2022. https://doi.org/10.1007/s11427-022-2218-x.

    Article  PubMed  Google Scholar 

  3. Rebello RJ, Oing C, Knudsen KE, Loeb S, Johnson DC, Reiter RE, Gillessen S, Van der Kwast T, Bristow RG. Prostate cancer. Nat Rev Dis Primers. 2021;7:9.

    Article  PubMed  Google Scholar 

  4. Bandini M, Mazzone E, Preisser F, Nazzani S, Zaffuto E, Marchioni M, Tian Z, Pompe RS, Tilki D, Graefen M, et al. Increase in the annual rate of newly diagnosed metastatic prostate cancer: a contemporary analysis of the surveillance, epidemiology and end results database. Eur Urol Oncol. 2018;1:314–20.

    Article  PubMed  Google Scholar 

  5. Cary KC, Cooperberg MR. Biomarkers in prostate cancer surveillance and screening: past, present, and future. Ther Adv Urol. 2013;5:318–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Prensner JR, Rubin MA, Wei JT, Chinnaiyan AM. Beyond PSA: the next generation of prostate cancer biomarkers. Sci Transl Med. 2012;4:127rv123.

    Article  Google Scholar 

  7. Medd JCC, Stockler MR, Collins R, Lalak A. Measuring men’s opinions of prostate needle biopsy. ANZ J Surg. 2005;75:662–4.

    Article  PubMed  Google Scholar 

  8. Schröder FH, Hugosson J, Roobol MJ, Tammela TLJ, Zappa M, Nelen V, Kwiatkowski M, Lujan M, Määttänen L, Lilja H, et al. Screening and prostate cancer mortality: results of the European randomised study of screening for prostate cancer (ERSPC) at 13 years of follow-up. Lancet. 2014;384:2027–35.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Etzioni R, Gulati R, Cooperberg MR, Penson DM, Weiss NS, Thompson IM. Limitations of basing screening policies on screening trials: the US preventive services task force and prostate cancer screening. Med Care. 2013;51:295–300.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Cohen JD, Li L, Wang Y, Thoburn C, Afsari B, Danilova L, Douville C, Javed AA, Wong F, Mattox A, et al. Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science. 2018;359:926–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Mellby LD, Nyberg AP, Johansen JS, Wingren C, Nordestgaard BG, Bojesen SE, Mitchell BL, Sheppard BC, Sears RC, Borrebaeck CAK. Serum biomarker signature-based liquid biopsy for diagnosis of early-stage pancreatic cancer. J Clin Oncol. 2018;36:2887–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. De Rubis G, Rajeev Krishnan S, Bebawy M. Liquid biopsies in cancer diagnosis, monitoring, and prognosis. Trends Pharmacol Sci. 2019;40:172–86.

    Article  PubMed  Google Scholar 

  13. Fitts CA, Ji N, Li Y, Tan C. Exploiting exosomes in cancer liquid biopsies and drug delivery. Adv Healthc Mater. 2019;8:e1801268.

    Article  PubMed  Google Scholar 

  14. Chaffer CL, Weinberg RA. A perspective on cancer cell metastasis. Science. 2011;331:1559–64.

    Article  CAS  PubMed  Google Scholar 

  15. Azmi AS, Bao B, Sarkar FH. Exosomes in cancer development, metastasis, and drug resistance: a comprehensive review. Cancer Metastasis Rev. 2013;32:623–42.

    Article  CAS  PubMed  Google Scholar 

  16. Han Q-F, Li W-J, Hu K-S, Gao J, Zhai W-L, Yang J-H, Zhang S-J. Exosome biogenesis: machinery, regulation, and therapeutic implications in cancer. Mol Cancer. 2022;21:207.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Stuffers S, Sem Wegner C, Stenmark H, Brech A. Multivesicular endosome biogenesis in the absence of ESCRTs. Traffic. 2009;10:925–37.

    Article  CAS  PubMed  Google Scholar 

  18. Kenific CM, Zhang H, Lyden D. An exosome pathway without an ESCRT. Cell Res. 2021;31:105–6.

    Article  PubMed  Google Scholar 

  19. Wei D, Zhan W, Gao Y, Huang L, Gong R, Wang W, Zhang R, Wu Y, Gao S, Kang T. RAB31 marks and controls an ESCRT-independent exosome pathway. Cell Res. 2021;31:157–77.

    Article  CAS  PubMed  Google Scholar 

  20. Juan T, Fürthauer M. Biogenesis and function of ESCRT-dependent extracellular vesicles. Semin Cell Dev Biol. 2018;74:66–77.

    Article  CAS  PubMed  Google Scholar 

  21. Dong X, Bai X, Ni J, Zhang H, Duan W, Graham P, Li Y. Exosomes and breast cancer drug resistance. Cell Death Dis. 2020;11:987.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  23. 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.

    Article  PubMed  Google Scholar 

  24. Yang KS, Im H, Hong S, Pergolini I, del Castillo AF, Wang R, Clardy S, Huang C-H, Pille C, Ferrone S, et al. Multiparametric plasma EV profiling facilitates diagnosis of pancreatic malignancy. Sci Transl Med. 2017;9:eaal3226.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Chen G, Huang AC, Zhang W, Zhang G, Wu M, Xu W, Yu Z, Yang J, Wang B, Sun H, et al. Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature. 2018;560:382–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Jurj A, Pop-Bica C, Slaby O, Ştefan CD, Cho WC, Korban SS, Berindan-Neagoe I. tiny actors in the big cellular world: extracellular vesicles playing critical roles in cancer. Int J Mol Sci. 2020;21:7688.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Wortzel I, Dror S, Kenific CM, Lyden D. Exosome-mediated metastasis: communication from a distance. Dev Cell. 2019;49:347–60.

    Article  CAS  PubMed  Google Scholar 

  28. Ulivi P, Indraccolo S. Liquid biopsies in cancer diagnosis monitoring and prognosis. Biomedicines. 2022;10:2748.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Paolini L, Zendrini A, Di Noto G, Busatto S, Lottini E, Radeghieri A, Dossi A, Caneschi A, Ricotta D, Bergese P. Residual matrix from different separation techniques impacts exosome biological activity. Sci Rep. 2016;6:23550.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Chen B-Y, Sung CW-H, Chen C, Cheng C-M, Lin DP-C, Huang C-T, Hsu M-Y. Advances in exosomes technology. Clinica Chimica Acta Int J Clin Chem. 2019;493:14–9.

    Article  CAS  Google Scholar 

  31. Palviainen M, Laukkanen K, Tavukcuoglu Z, Velagapudi V, Kärkkäinen O, Hanhineva K, Auriola S, Ranki A, Siljander P. Cancer alters the metabolic fingerprint of extracellular vesicles. Cancers. 2020;12:3292.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Sunkara V, Kim C-J, Park J, Woo H-K, Kim D, Ha HK, Kim M-H, Son Y, Kim J-R, Cho Y-K. Fully automated, label-free isolation of extracellular vesicles from whole blood for cancer diagnosis and monitoring. Theranostics. 2019;9:1851–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Bijnsdorp IV, Maxouri O, Kardar A, Schelfhorst T, Piersma SR, Pham TV, Vis A, van Moorselaar RJ, Jimenez CR. Feasibility of urinary extracellular vesicle proteome profiling using a robust and simple, clinically applicable isolation method. J Extracell Vesicles. 2017;6:1313091.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Correll VL, Otto JJ, Risi CM, Main BP, Boutros PC, Kislinger T, Galkin VE, Nyalwidhe JO, Semmes OJ, Yang L. Optimization of small extracellular vesicle isolation from expressed prostatic secretions in urine for in-depth proteomic analysis. J Extracell Vesicles. 2022;11:e12184.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Dhondt B, Geeurickx E, Tulkens J, Van Deun J, Vergauwen G, Lippens L, Miinalainen I, Rappu P, Heino J, Ost P, et al. Unravelling the proteomic landscape of extracellular vesicles in prostate cancer by density-based fractionation of urine. J Extracell Vesicles. 2020;9:1736935.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Guan S, Yu H, Yan G, Gao M, Sun W, Zhang X. Characterization of urinary exosomes purified with size exclusion chromatography and ultracentrifugation. J Proteome Res. 2020;19:2217–25.

    Article  CAS  PubMed  Google Scholar 

  37. Brzozowski JS, Bond DR, Jankowski H, Goldie BJ, Burchell R, Naudin C, Smith ND, Scarlett CJ, Larsen MR, Dun MD, et al. Extracellular vesicles with altered tetraspanin CD9 and CD151 levels confer increased prostate cell motility and invasion. Sci Rep. 2018;8:8822.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Pang B, Zhu Y, Ni J, Ruan J, Thompson J, Malouf D, Bucci J, Graham P, Li Y. Quality assessment and comparison of plasma-derived extracellular vesicles separated by three commercial kits for prostate cancer diagnosis. Int J Nanomedicine. 2020;15:10241–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. García-Flores M, Sánchez-López CM, Ramírez-Calvo M, Fernández-Serra A, Marcilla A, López-Guerrero JA. Isolation and characterization of urine microvesicles from prostate cancer patients: different approaches, different visions. BMC Urol. 2021;21:137.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Chen Y, Zhu Q, Cheng L, Wang Y, Li M, Yang Q, Hu L, Lou D, Li J, Dong X, et al. Exosome detection via the ultrafast-isolation system: EXODUS. Nat Methods. 2021;18:212–8.

    Article  CAS  PubMed  Google Scholar 

  41. Morani M, Taverna M, Krupova Z, Alexandre L, Defrenaix P, Mai TD. Development of a microfluidic droplet platform with an antibody-free magnetic-bead-based strategy for high through-put and efficient EVs isolation. Talanta. 2022;249:123625.

    Article  CAS  PubMed  Google Scholar 

  42. Foroni C, Zarovni N, Bianciardi L, Bernardi S, Triggiani L, Zocco D, Venturella M, Chiesi A, Valcamonico F, Berruti A. When less is more: specific capture and analysis of tumor exosomes in plasma increases the sensitivity of liquid biopsy for comprehensive detection of multiple androgen receptor phenotypes in advanced prostate cancer patients. Biomedicines. 2020;8:131.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Brett SI, Lucien F, Guo C, Williams KC, Kim Y, Durfee PN, Brinker CJ, Chin JI, Yang J, Leong HS. Immunoaffinity based methods are superior to kits for purification of prostate derived extracellular vesicles from plasma samples. Prostate. 2017;77:1335–43.

    Article  CAS  PubMed  Google Scholar 

  44. Patel GK, Khan MA, Zubair H, Srivastava SK, Khushman Md, Singh S, Singh AP. Comparative analysis of exosome isolation methods using culture supernatant for optimum yield, purity and downstream applications. Sci Rep. 2019;9:5335.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Shtam T, Evtushenko V, Samsonov R, Zabrodskaya Y, Kamyshinsky R, Zabegina L, Verlov N, Burdakov V, Garaeva L, Slyusarenko M, et al. Evaluation of immune and chemical precipitation methods for plasma exosome isolation. PLoS ONE. 2020;15:e0242732.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Dong L, Zieren RC, Horie K, Kim C-J, Mallick E, Jing Y, Feng M, Kuczler MD, Green J, Amend SR, et al. Comprehensive evaluation of methods for small extracellular vesicles separation from human plasma, urine and cell culture medium. J Extracell Vesicles. 2020;10:e12044.

    Article  CAS  PubMed  Google Scholar 

  47. 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;25:758–67.

    Article  CAS  PubMed  Google Scholar 

  48. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Mercadal M, Herrero C, López-Rodrigo O, Castells M, de la Fuente A, Vigués F, Bassas L, Larriba S. Impact of extracellular vesicle isolation methods on downstream mirna analysis in semen: a comparative study. Int J Mol Sci. 2020;21:5949.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Lin S, Yu Z, Chen D, Wang Z, Miao J, Li Q, Zhang D, Song J, Cui D. Progress in microfluidics-based exosome separation and detection technologies for diagnostic applications. Small. 2020;16:e1903916.

    Article  PubMed  Google Scholar 

  51. Li Q, Wang Y, Xue Y, Qiao L, Yu G, Liu Y, Yu S. Ultrasensitive analysis of exosomes using a 3D Self-assembled nanostructured SiO2 microfluidic chip. ACS Appl Mater Interfaces. 2022;14:14693–702.

    Article  CAS  PubMed  Google Scholar 

  52. Ku A, Fredsøe J, Sørensen KD, Borre M, Evander M, Laurell T, Lilja H, Ceder Y. High-throughput and automated acoustic trapping of extracellular vesicles to identify microRNAs with diagnostic potential for prostate cancer. Front Oncol. 2021;11:631021.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Kang Y-T, Kim YJ, Bu J, Cho Y-H, Han S-W, Moon B-I. High-purity capture and release of circulating exosomes using an exosome-specific dual-patterned immunofiltration (ExoDIF) device. Nanoscale. 2017;9:13495–505.

    Article  CAS  PubMed  Google Scholar 

  54. Koh YQ, Almughlliq FB, Vaswani K, Peiris HN, Mitchell MD. Exosome enrichment by ultracentrifugation and size exclusion chromatography. Front Biosci (Landmark Ed). 2018;23:865–74.

    Article  CAS  PubMed  Google Scholar 

  55. Lee H, Kang SJ, Lee J, Park KH, Rhee WJ. Isolation and characterization of urinary extracellular vesicles from healthy donors and patients with castration-resistant prostate cancer. Int J Mol Sci. 2022;23:7134.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Sun N, Tran BV, Peng Z, Wang J, Zhang C, Yang P, Zhang TX, Widjaja J, Zhang RY, Xia W, et al. Coupling lipid labeling and click chemistry enables isolation of extracellular vesicles for noninvasive detection of oncogenic gene alterations. Adv Sci (Weinh). 2022;9:e2105853.

    Article  PubMed  Google Scholar 

  57. Yoshida T, Hanayama R. TIM4-Affinity methods targeting phosphatidylserine for isolation or detection of extracellular vesicles. Methods Mol Biol. 2022;2466:23–36.

    Article  CAS  PubMed  Google Scholar 

  58. Liu H-Y, Kumar R, Zhong C, Gorji S, Paniushkina L, Masood R, Wittel UA, Fuchs H, Nazarenko I, Hirtz M. Rapid capture of cancer extracellular vesicles by lipid patch microarrays. Adv Mater. 2021;33:e2008493.

    Article  PubMed  Google Scholar 

  59. Petersen KE, Shiri F, White T, Bardi GT, Sant H, Gale BK, Hood JL. Exosome isolation: cyclical electrical field flow fractionation in low-ionic-strength fluids. Anal Chem. 2018;90:12783–90.

    Article  CAS  PubMed  Google Scholar 

  60. Gao Z, Hutchins Z, Li Z, Zhong W. Offline coupling of asymmetrical flow field-flow fractionation and capillary electrophoresis for separation of extracellular vesicles. Anal Chem. 2022;94:14083–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Bryzgunova OE, Zaripov MM, Skvortsova TE, Lekchnov EA, Grigor’eva AE, Zaporozhchenko IA, Morozkin ES, Ryabchikova EI, Yurchenko YB, Voitsitskiy VE, Laktionov PP. Comparative study of extracellular vesicles from the urine of healthy individuals and prostate cancer patients. PLOS ONE. 2016;11:e0157566.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Rood IM, Deegens JKJ, Merchant ML, Tamboer WPM, Wilkey DW, Wetzels JFM, Klein JB. Comparison of three methods for isolation of urinary microvesicles to identify biomarkers of nephrotic syndrome. Kidney Int. 2010;78:810–6.

    Article  CAS  PubMed  Google Scholar 

  63. Lewis JM, Vyas AD, Qiu Y, Messer KS, White R, Heller MJ. Integrated analysis of exosomal protein biomarkers on alternating current electrokinetic chips enables rapid detection of pancreatic cancer in patient blood. ACS Nano. 2018;12:3311–20.

    Article  CAS  PubMed  Google Scholar 

  64. Théry C, Witwer KW, Aikawa E, Alcaraz MJ, Anderson JD, Andriantsitohaina R, Antoniou A, Arab T, Archer F, Atkin-Smith GK, et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the international society for extracellular vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles. 2018;7:1535750.

    Article  PubMed  PubMed Central  Google Scholar 

  65. Schey KL, Luther JM, Rose KL. Proteomics characterization of exosome cargo. Methods. 2015;87:75–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Crescitelli R, Lässer C, Jang SC, Cvjetkovic A, Malmhäll C, Karimi N, Höög JL, Johansson I, Fuchs J, Thorsell A, et al. Subpopulations of extracellular vesicles from human metastatic melanoma tissue identified by quantitative proteomics after optimized isolation. J Extracell Vesicles. 2020;9:1722433.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Rupert DLM, Lässer C, Eldh M, Block S, Zhdanov VP, Lotvall JO, Bally M, Höök F. Determination of exosome concentration in solution using surface plasmon resonance spectroscopy. Anal Chem. 2014;86:5929–36.

    Article  CAS  PubMed  Google Scholar 

  68. Liu C, Zeng X, An Z, Yang Y, Eisenbaum M, Gu X, Jornet JM, Dy GK, Reid ME, Gan Q, Wu Y. Sensitive detection of exosomal proteins via a compact surface plasmon resonance biosensor for cancer diagnosis. ACS Sens. 2018;3:1471–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Li M, Xi N, Wang Y-C, Liu L-Q. Atomic force microscopy for revealing micro/nanoscale mechanics in tumor metastasis: from single cells to microenvironmental cues. Acta Pharmacol Sin. 2021;42:323–39.

    Article  CAS  PubMed  Google Scholar 

  70. Corso G, Heusermann W, Trojer D, Görgens A, Steib E, Voshol J, Graff A, Genoud C, Lee Y, Hean J, et al. Systematic characterization of extracellular vesicle sorting domains and quantification at the single molecule - single vesicle level by fluorescence correlation spectroscopy and single particle imaging. J Extracell Vesicles. 2019;8:1663043.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Fisher WS, Tchounwou C, Wei S, Roberts L, Ewert KK, Safinya CR. Exosomes are secreted at similar densities by M21 and PC3 human cancer cells and show paclitaxel solubility. Biochim Biophys Acta Biomembr. 2022;1864:183841.

    Article  CAS  PubMed  Google Scholar 

  72. Bertokova A, Svecova N, Kozics K, Gabelova A, Vikartovska A, Jane E, Hires M, Bertok T, Tkac J. Exosomes from prostate cancer cell lines: Isolation optimisation and characterisation. Biomed Pharmacother. 2022;151:113093.

    Article  CAS  PubMed  Google Scholar 

  73. Choi WWY, Sánchez C, Li JJ, Dinarvand M, Adomat H, Ghaffari M, Khoja L, Vafaee F, Joshua AM, Chi KN, et al. Extracellular vesicles from biological fluids as potential markers in castration resistant prostate cancer. J Cancer Res Clin Oncol. 2022. https://doi.org/10.1007/s00432-022-04391-6.

    Article  PubMed  PubMed Central  Google Scholar 

  74. Ma Q, Liang M, Wu Y, Dou C, Xu J, Dong S, Luo F. Small extracellular vesicles deliver osteolytic effectors and mediate cancer-induced osteolysis in bone metastatic niche. J Extracell Vesicles. 2021;10:e12068.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Linxweiler J, Kolbinger A, Himbert D, Zeuschner P, Saar M, Stöckle M, Junker K. Organ-specific uptake of extracellular vesicles secreted by urological cancer cells. Cancers. 2021;13:4937.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Chen K, Wang Q, Liu X, Wang F, Yang Y, Tian X. Hypoxic pancreatic cancer derived exosomal miR-30b-5p promotes tumor angiogenesis by inhibiting GJA1 expression. Int J Biol Sci. 2022;18:1220–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Todorova D, Simoncini S, Lacroix R, Sabatier F, Dignat-George F. Extracellular vesicles in angiogenesis. Circ Res. 2017;120:1658–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Whiteside TL. Tumor-derived exosomes and their role in tumor-induced immune suppression. Vaccines. 2016;4:35.

    Article  PubMed  PubMed Central  Google Scholar 

  79. Tai Y-L, Chen K-C, Hsieh J-T, Shen T-L. Exosomes in cancer development and clinical applications. Cancer Sci. 2018;109:2364–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Mashouri L, Yousefi H, Aref AR, Ahadi AM, Molaei F, Alahari SK. Exosomes: composition, biogenesis, and mechanisms in cancer metastasis and drug resistance. Mol Cancer. 2019;18:75.

    Article  PubMed Central  Google Scholar 

  81. Pastushenko I, Blanpain C. EMT transition states during tumor progression and metastasis. Trends Cell Biol. 2019;29:212–26.

    Article  CAS  PubMed  Google Scholar 

  82. Gaballa R, Ali HEA, Mahmoud MO, Rhim JS, Ali HI, Salem HF, Saleem M, Kandeil MA, Ambs S, Abd Elmageed ZY. Exosomes-mediated transfer of Itga2 promotes migration and invasion of prostate cancer cells by inducing epithelial-mesenchymal transition. Cancers. 2020;12:2300.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Li Y, Li Q, Li D, Gu J, Qian D, Qin X, Chen Y. Exosome carrying PSGR promotes stemness and epithelial-mesenchymal transition of low aggressive prostate cancer cells. Life Sci. 2021;264:118638.

    Article  CAS  PubMed  Google Scholar 

  84. Miyazaki T, Ikeda K, Sato W, Horie-Inoue K, Inoue S. Extracellular vesicle-mediated EBAG9 transfer from cancer cells to tumor microenvironment promotes immune escape and tumor progression. Oncogenesis. 2018;7:7.

    Article  PubMed  PubMed Central  Google Scholar 

  85. Lin C-J, Yun E-J, Lo UG, Tai Y-L, Deng S, Hernandez E, Dang A, Chen Y-A, Saha D, Mu P, et al. The paracrine induction of prostate cancer progression by caveolin-1. Cell Death Dis. 2019;10:834.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Ronquist KG, Sanchez C, Dubois L, Chioureas D, Fonseca P, Larsson A, Ullén A, Yachnin J, Ronquist G, Panaretakis T. Energy-requiring uptake of prostasomes and PC3 cell-derived exosomes into non-malignant and malignant cells. J Extracell Vesicles. 2016;5:29877.

    Article  PubMed  Google Scholar 

  87. Read J, Ingram A, Al Saleh HA, Platko K, Gabriel K, Kapoor A, Pinthus J, Majeed F, Qureshi T, Al-Nedawi K. Nuclear transportation of exogenous epidermal growth factor receptor and androgen receptor via extracellular vesicles. Eur J Cancer. 2017;70:62–74.

    Article  CAS  PubMed  Google Scholar 

  88. Tian H-Y, Liang Q, Shi Z, Zhao H. Exosomal CXCL14 contributes to M2 macrophage polarization through NF-κB signaling in prostate cancer. Oxid Med Cell Longev. 2022;2022:7616696.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Fontana F, Anselmi M, Carollo E, Sartori P, Procacci P, Carter D, Limonta P. Adipocyte-derived extracellular vesicles promote prostate cancer cell aggressiveness by enabling multiple phenotypic and metabolic changes. Cells. 2022;11:2388.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Yang C, Guo WB, Zhang WS, Bian J, Yang JK, Zhou QZ, Chen MK, Peng W, Qi T, Wang CY, Liu CD. Comprehensive proteomics analysis of exosomes derived from human seminal plasma. Andrology. 2017;5:1007–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Basu A. The interplay between apoptosis and cellular senescence: Bcl-2 family proteins as targets for cancer therapy. Pharmacol Ther. 2022;230:107943.

    Article  CAS  PubMed  Google Scholar 

  92. Che Y, Shi X, Shi Y, Jiang X, Ai Q, Shi Y, Gong F, Jiang W. Exosomes derived from miR-143-overexpressing MSCs inhibit cell migration and invasion in human prostate cancer by downregulating TFF3. Mol Ther Nucleic Acids. 2019;18:232–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Lehmann BD, Paine MS, Brooks AM, McCubrey JA, Renegar RH, Wang R, Terrian DM. Senescence-associated exosome release from human prostate cancer cells. Cancer Res. 2008;68:7864–71.

    Article  CAS  PubMed  Google Scholar 

  94. Chowdhury SG, Ray R, Bhattacharya D, Karmakar P. DNA damage induced cellular senescence and it’s PTEN-armed exosomes-the warriors against prostate carcinoma cells. Med Oncol. 2022;39:34.

    Article  CAS  PubMed  Google Scholar 

  95. Jiang X, Wang J, Deng X, Xiong F, Zhang S, Gong Z, Li X, Cao K, Deng H, He Y, et al. The role of microenvironment in tumor angiogenesis. J Exp Clin Cancer Res. 2020;39:204.

    Article  PubMed  PubMed Central  Google Scholar 

  96. Krishn SR, Salem I, Quaglia F, Naranjo NM, Agarwal E, Liu Q, Sarker S, Kopenhaver J, McCue PA, Weinreb PH, et al. The αvβ6 integrin in cancer cell-derived small extracellular vesicles enhances angiogenesis. J Extracell Vesicles. 2020;9:1763594.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. DeRita RM, Zerlanko B, Singh A, Lu H, Iozzo RV, Benovic JL, Languino LR. c-Src, Insulin-like growth factor I receptor, G-protein-coupled receptor kinases and focal adhesion kinase are enriched into prostate cancer cell exosomes. J Cell Biochem. 2017;118:66–73.

    Article  CAS  PubMed  Google Scholar 

  98. Liu P, Wang W, Wang F, Fan J, Guo J, Wu T, Lu D, Zhou Q, Liu Z, Wang Y, et al. Alterations of plasma exosomal proteins and motabolies are associated with the progression of castration-resistant prostate cancer. J Transl Med. 2023;21:40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Ojalill M, Virtanen N, Rappu P, Siljamäki E, Taimen P, Heino J. Interaction between prostate cancer cells and prostate fibroblasts promotes accumulation and proteolytic processing of basement membrane proteins. Prostate. 2020;80:715–26.

    Article  CAS  PubMed  Google Scholar 

  100. Bedeschi M, Marino N, Cavassi E, Piccinini F, Tesei A. Cancer-associated fibroblast: role in prostate cancer progression to metastatic disease and therapeutic resistance. Cells. 2023;12:802.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Webber JP, Spary LK, Sanders AJ, Chowdhury R, Jiang WG, Steadman R, Wymant J, Jones AT, Kynaston H, Mason MD, et al. Differentiation of tumour-promoting stromal myofibroblasts by cancer exosomes. Oncogene. 2015;34:290–302.

    Article  CAS  PubMed  Google Scholar 

  102. Halin Bergström S, Hägglöf C, Thysell E, Bergh A, Wikström P, Lundholm M. Extracellular vesicles from metastatic rat prostate tumors prime the normal prostate tissue to facilitate tumor growth. Sci Rep. 2016;6:31805.

    Article  PubMed  PubMed Central  Google Scholar 

  103. Sadovska L, Zayakin P, Bajo-Santos C, Endzeliņš E, Auders J, Keiša L, Jansons J, Lietuvietis V, Linē A. Effects of urinary extracellular vesicles from prostate cancer patients on the transcriptomes of cancer-associated and normal fibroblasts. BMC Cancer. 2022;22:1055.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Zhang Z, Li X, Wang Y, Wei Y, Wei X. Involvement of inflammasomes in tumor microenvironment and tumor therapies. J Hematol Oncol. 2023;16:24.

    Article  PubMed  PubMed Central  Google Scholar 

  105. Xu Z, Wang H, Qin Z, Zhao F, Zhou L, Xu L, Jia R. NLRP3 inflammasome promoted the malignant progression of prostate cancer via the activation of caspase-1. Cell Death Discov. 2021;7:399.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Mezzasoma L, Talesa VN, Costanzi E, Bellezza I. Natriuretic peptides regulate prostate cells inflammatory behavior: potential novel anticancer agents for prostate cancer. Biomolecules. 2021;11:794.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Panigrahi GK, Praharaj PP, Kittaka H, Mridha AR, Black OM, Singh R, Mercer R, van Bokhoven A, Torkko KC, Agarwal C, et al. Exosome proteomic analyses identify inflammatory phenotype and novel biomarkers in African American prostate cancer patients. Cancer Med. 2019;8:1110–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Urabe F, Patil K, Ramm GA, Ochiya T, Soekmadji C. Extracellular vesicles in the development of organ-specific metastasis. J Extracell Vesicles. 2021;10:e12125.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Lu H, Bowler N, Harshyne LA, Craig Hooper D, Krishn SR, Kurtoglu S, Fedele C, Liu Q, Tang H-Y, Kossenkov AV, et al. Exosomal αvβ6 integrin is required for monocyte M2 polarization in prostate cancer. Matrix Biol. 2018;70:20–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Borel M, Lollo G, Magne D, Buchet R, Brizuela L, Mebarek S. Prostate cancer-derived exosomes promote osteoblast differentiation and activity through phospholipase D2. Biochim Biophys Acta Mol Basis Dis. 2020;1866:165919.

    Article  CAS  PubMed  Google Scholar 

  111. Shi Y, Pu K, Yao H, Chen Y, Zheng X, Zhao L, Ma X, Ge C. Gold nanorods inhibit tumor metastasis by regulating MMP-9 activity: implications for radiotherapy. ACS Appl Mater Interfaces. 2023. https://doi.org/10.1021/acsami.2c20944.

    Article  PubMed  PubMed Central  Google Scholar 

  112. Deep G, Jain A, Kumar A, Agarwal C, Kim S, Leevy WM, Agarwal R. Exosomes secreted by prostate cancer cells under hypoxia promote matrix metalloproteinases activity at pre-metastatic niches. Mol Carcinog. 2020;59:323–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Henrich SE, McMahon KM, Plebanek MP, Calvert AE, Feliciano TJ, Parrish S, Tavora F, Mega A, De Souza A, Carneiro BA, Thaxton CS. Prostate cancer extracellular vesicles mediate intercellular communication with bone marrow cells and promote metastasis in a cholesterol-dependent manner. J Extracell Vesicles. 2020;10:e12042.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Dai J, Escara-Wilke J, Keller JM, Jung Y, Taichman RS, Pienta KJ, Keller ET. Primary prostate cancer educates bone stroma through exosomal pyruvate kinase M2 to promote bone metastasis. J Exp Med. 2019;216:2883–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Li D, Xia L, Huang P, Wang Z, Guo Q, Huang C, Leng W, Qin S. Heterogeneity and plasticity of epithelial-mesenchymal transition (EMT) in cancer metastasis: focusing on partial EMT and regulatory mechanisms. Cell Prolif. 2023;56:e13423.

    Article  PubMed  PubMed Central  Google Scholar 

  116. Voglstaetter M, Thomsen AR, Nouvel J, Koch A, Jank P, Navarro EG, Gainey-Schleicher T, Khanduri R, Groß A, Rossner F, et al. Tspan8 is expressed in breast cancer and regulates E-cadherin/catenin signalling and metastasis accompanied by increased circulating extracellular vesicles. J Pathol. 2019;248:421–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Liu J, Wu Q, Wang Y, Wei Y, Wu H, Duan L, Zhang Q, Wu Y. Ovol2 induces mesenchymal-epithelial transition via targeting ZEB1 in osteosarcoma. Onco Targets Ther. 2018;11:2963–73.

    Article  PubMed  PubMed Central  Google Scholar 

  118. Yu L, Sui B, Fan W, Lei L, Zhou L, Yang L, Diao Y, Zhang Y, Li Z, Liu J, Hao X. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Urabe F, Kosaka N, Yamamoto Y, Ito K, Otsuka K, Soekmadji C, Egawa S, Kimura T, Ochiya T. Metastatic prostate cancer-derived extracellular vesicles facilitate osteoclastogenesis by transferring the CDCP1 protein. J Extracell Vesicles. 2023;12:e12312.

    Article  PubMed  Google Scholar 

  120. Yang Q, Xu J, Gu J, Shi H, Zhang J, Zhang J, Chen Z-S, Fang X, Zhu T, Zhang X. Extracellular vesicles in cancer drug resistance: roles, mechanisms, and implications. Adv Sci (Weinh). 2022;9:e2201609.

    Article  PubMed  Google Scholar 

  121. Cui X, Fu Q, Wang X, Xia P, Cui X, Bai X, Lu Z. Molecular mechanisms and clinical applications of exosomes in prostate cancer. Biomark Res. 2022;10:56.

    Article  PubMed  PubMed Central  Google Scholar 

  122. Bhagirath D, Yang TL, Tabatabai ZL, Majid S, Dahiya R, Tanaka Y, Saini S. BRN4 Is a novel driver of neuroendocrine differentiation in castration-resistant prostate cancer and is selectively released in extracellular vesicles with BRN2. Clin Cancer Res. 2019;25:6532–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Peak TC, Panigrahi GK, Praharaj PP, Su Y, Shi L, Chyr J, Rivera-Chávez J, Flores-Bocanegra L, Singh R, Vander Griend DJ, et al. Syntaxin 6-mediated exosome secretion regulates enzalutamide resistance in prostate cancer. Mol Carcinog. 2020;59:62–72.

    Article  CAS  PubMed  Google Scholar 

  124. Lee H-C, Ou C-H, Huang Y-C, Hou P-C, Creighton CJ, Lin Y-S, Hu C-Y, Lin S-C. YAP1 overexpression contributes to the development of enzalutamide resistance by induction of cancer stemness and lipid metabolism in prostate cancer. Oncogene. 2021;40:2407–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. 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;6:21740–54.

    Article  PubMed  PubMed Central  Google Scholar 

  126. 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.e315-385.e320.

    Article  Google Scholar 

  127. Greening DW, Xu R, Ale A, Hagemeyer CE, Chen W. Extracellular vesicles as next generation immunotherapeutics. Semin Cancer Biol. 2023. https://doi.org/10.1016/j.semcancer.2023.02.002.

    Article  PubMed  Google Scholar 

  128. Lundholm M, Schröder M, Nagaeva O, Baranov V, Widmark A, Mincheva-Nilsson L, Wikström P. Prostate tumor-derived exosomes down-regulate NKG2D expression on natural killer cells and CD8+ T cells: mechanism of immune evasion. PLoS ONE. 2014;9:e108925.

    Article  PubMed  PubMed Central  Google Scholar 

  129. Salimu J, Webber J, Gurney M, Al-Taei S, Clayton A, Tabi Z. Dominant immunosuppression of dendritic cell function by prostate-cancer-derived exosomes. J Extracell Vesicles. 2017;6:1368823.

    Article  PubMed  PubMed Central  Google Scholar 

  130. Li N, Wang Y, Xu H, Wang H, Gao Y, Zhang Y. Exosomes derived from RM-1 cells promote the recruitment of MDSCs into tumor microenvironment by upregulating CXCR4 via TLR2/NF-κB pathway. J Oncol. 2021;2021:5584406.

    Article  PubMed  PubMed Central  Google Scholar 

  131. Chen X, Li J, Zhang R, Zhang Y, Wang X, Leung EL-H, Ma L, Wong VKW, Liu L, Neher E, Yu H. Suppression of PD-L1 release from small extracellular vesicles promotes systemic anti-tumor immunity by targeting ORAI1 calcium channels. J Extracell Vesicles. 2022;11:e12279.

    Article  PubMed  Google Scholar 

  132. Liu J, He D, Cheng L, Huang C, Zhang Y, Rao X, Kong Y, Li C, Zhang Z, Liu J, et al. p300/CBP inhibition enhances the efficacy of programmed death-ligand 1 blockade treatment in prostate cancer. Oncogene. 2020;39:3939–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Zhang L, Yu D. Exosomes in cancer development, metastasis, and immunity. Biochim Biophys Acta Rev Cancer. 2019;1871:455–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Keerthikumar S, Chisanga D, Ariyaratne D, Saffar H, Anand S, Zhao KN, Samuel M, Pathan M, Jois M, Chilamkurti N, et al. ExoCarta: a web-based compendium of exosomal cargo. J Mol Biol. 2016;428:688–92.

    Article  CAS  PubMed  Google Scholar 

  135. Bhagirath D, Liston M, Akoto T, Lui B, Bensing BA, Sharma A, Saini S. Novel, non-invasive markers for detecting therapy induced neuroendocrine differentiation in castration-resistant prostate cancer patients. Sci Rep. 2021;11:8279.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Ishizuya Y, Uemura M, Narumi R, Tomiyama E, Koh Y, Matsushita M, Nakano K, Hayashi Y, Wang C, Kato T, et al. The role of actinin-4 (ACTN4) in exosomes as a potential novel therapeutic target in castration-resistant prostate cancer. Biochem Biophys Res Commun. 2020;523:588–94.

    Article  CAS  PubMed  Google Scholar 

  137. Worst TS, von Hardenberg J, Gross JC, Erben P, Schnölzer M, Hausser I, Bugert P, Michel MS, Boutros M. Database-augmented mass spectrometry analysis of exosomes identifies claudin 3 as a putative prostate cancer biomarker. Mol Cell Proteomics. 2017;16:998–1008.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Park YH, Shin HW, Jung AR, Kwon OS, Choi Y-J, Park J, Lee JY. Prostate-specific extracellular vesicles as a novel biomarker in human prostate cancer. Sci Rep. 2016;6:30386.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Vardaki I, Corn P, Gentile E, Song JH, 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;27:3253–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Khanna K, Salmond N, Lynn KS, Leong HS, Williams KC. Clinical significance of STEAP1 extracellular vesicles in prostate cancer. Prostate Cancer Prostatic Dis. 2021;24:802–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Logozzi M, Mizzoni D, Capasso C, Del Prete S, Di Raimo R, Falchi M, Angelini DF, Sciarra A, Maggi M, Supuran CT, Fais S. Plasmatic exosomes from prostate cancer patients show increased carbonic anhydrase IX expression and activity and low pH. J Enzyme Inhib Med Chem. 2020;35:280–8.

    Article  CAS  PubMed  Google Scholar 

  142. Krishn SR, Singh A, Bowler N, Duffy AN, Friedman A, Fedele C, Kurtoglu S, Tripathi SK, Wang K, Hawkins A, et al. Prostate cancer sheds the αvβ3 integrin in vivo through exosomes. Matrix Biol. 2019;77:41–57.

    Article  CAS  PubMed  Google Scholar 

  143. Chung J-W, Kim HT, Ha Y-S, Lee EH, Chun SY, Lee C-H, Byeon KH, Choi SH, Lee JN, Kim BS, et al. Identification of a novel non-invasive biological marker to overcome the shortcomings of PSA in diagnosis and risk stratification for prostate cancer: initial prospective study of developmental endothelial locus-1 protein. PLoS ONE. 2021;16:e0250254.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Li S, Zhao Y, Chen W, Yin L, Zhu J, Zhang H, Cai C, Li P, Huang L, Ma P. Exosomal ephrinA2 derived from serum as a potential biomarker for prostate cancer. J Cancer. 2018;9:2659–65.

    Article  PubMed  PubMed Central  Google Scholar 

  145. Kawakami K, Fujita Y, Matsuda Y, Arai T, Horie K, Kameyama K, Kato T, Masunaga K, Kasuya Y, Tanaka M, et al. Gamma-glutamyltransferase activity in exosomes as a potential marker for prostate cancer. BMC Cancer. 2017;17:316.

    Article  PubMed  PubMed Central  Google Scholar 

  146. Fujita K, Kume H, Matsuzaki K, Kawashima A, Ujike T, Nagahara A, Uemura M, Miyagawa Y, Tomonaga T, Nonomura N. Proteomic analysis of urinary extracellular vesicles from high gleason score prostate cancer. Sci Rep. 2017;7:42961.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Zhang H, Zhang G-Y, Su W-C, Chen Y-T, Liu Y-F, Wei D, Zhang Y-X, Tang Q-Y, Liu Y-X, Wang S-Z, et al. High throughput isolation and data independent acquisition mass spectrometry (DIA-MS) of urinary extracellular vesicles to improve prostate cancer diagnosis. Molecules. 2022;27:8155.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Boukouris S, Mathivanan S. Exosomes in bodily fluids are a highly stable resource of disease biomarkers. Proteomics Clin Appl. 2015;9:358–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Saari H, Lázaro-Ibáñez E, Viitala T, Vuorimaa-Laukkanen E, Siljander P, Yliperttula M. Microvesicle- and exosome-mediated drug delivery enhances the cytotoxicity of paclitaxel in autologous prostate cancer cells. J Control Release. 2015;220:727–37.

    Article  CAS  PubMed  Google Scholar 

  150. Wang D, Wan Z, Yang Q, Chen J, Liu Y, Lu F, Tang J. Sonodynamical reversion of immunosuppressive microenvironment in prostate cancer via engineered exosomes. Drug Deliv. 2022;29:702–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Lee JH, Song J, Kim IG, You G, Kim H, Ahn J-H, Mok H. Exosome-mediated delivery of transforming growth factor-β receptor 1 kinase inhibitors and toll-like receptor 7/8 agonists for combination therapy of tumors. Acta Biomater. 2022;141:354–63.

    Article  CAS  PubMed  Google Scholar 

  152. Luo Z-W, Xia K, Liu Y-W, Liu J-H, Rao S-S, Hu X-K, Chen C-Y, Xu R, Wang Z-X, Xie H. Extracellular vesicles from akkermansia muciniphila elicit antitumor immunity against prostate cancer via modulation of CD8+ T cells and macrophages. Int J Nanomedicine. 2021;16:2949–63.

    Article  PubMed  PubMed Central  Google Scholar 

  153. Kurniawati I, Liu M-C, Hsieh C-L, Do AD, Sung S-Y. Targeting castration-resistant prostate cancer using mesenchymal stem cell exosomes for therapeutic MicroRNA-let-7c delivery. Front Biosci (Landmark Ed). 2022;27:256.

    Article  CAS  PubMed  Google Scholar 

  154. Koh E, Lee EJ, Nam G-H, Hong Y, Cho E, Yang Y, Kim I-S. Exosome-SIRPα, a CD47 blockade increases cancer cell phagocytosis. Biomaterials. 2017;121:121–9.

    Article  CAS  PubMed  Google Scholar 

  155. Shin JM, Lee C-H, Son S, Kim CH, Lee JA, Ko H, Shin S, Song SH, Park S-S, Bae J-H, et al. Sulfisoxazole elicits robust antitumour immune response along with immune checkpoint therapy by inhibiting exosomal PD-L1. Adv Sci (Weinh). 2022;9:e2103245.

    Article  PubMed  Google Scholar 

  156. Chen S, Zhu G, Yang Y, Wang F, Xiao Y-T, Zhang N, Bian X, Zhu Y, Yu Y, Liu F, et al. Single-cell analysis reveals transcriptomic remodellings in distinct cell types that contribute to human prostate cancer progression. Nat Cell Biol. 2021;23:87–98.

    Article  CAS  PubMed  Google Scholar 

  157. Shi F, Sun M-H, Zhou Z, Wu L, Zhu Z, Xia S-J, Han B-M, Zhao Y-Y, Jing Y-F, Cui D. Tumor-associated macrophages in direct contact with prostate cancer cells promote malignant proliferation and metastasis through NOTCH1 pathway. Int J Biol Sci. 2022;18:5994–6007.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Cui Z, Li W, Wang Y, Zhao M, Liu K, Yang Y, Teng S, Zhang N, Min L, Li P, et al. M2 macrophage-derived exosomal ferritin heavy chain promotes colon cancer cell proliferation. Biol Trace Elem Res. 2022. https://doi.org/10.1007/s12011-022-03488-w.

    Article  PubMed  PubMed Central  Google Scholar 

  159. Tanimoto T, Hori A, Kami M. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. New Engl J Med. 2010;363:1966 (1966; author reply 1967-1966; author reply 1968).

    Article  CAS  PubMed  Google Scholar 

  160. Zuo B, Zhang Y, Zhao K, Wu L, Qi H, Yang R, Gao X, Geng M, Wu Y, Jing R, et al. Universal immunotherapeutic strategy for hepatocellular carcinoma with exosome vaccines that engage adaptive and innate immune responses. J Hematol Oncol. 2022;15:46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Rountree RB, Mandl SJ, Nachtwey JM, Dalpozzo K, Do L, Lombardo JR, Schoonmaker PL, Brinkmann K, Dirmeier U, Laus R, Delcayre A. Exosome targeting of tumor antigens expressed by cancer vaccines can improve antigen immunogenicity and therapeutic efficacy. Cancer Res. 2011;71:5235–44.

    Article  CAS  PubMed  Google Scholar 

  162. Shi X, Sun J, Li H, Lin H, Xie W, Li J, Tan W. Antitumor efficacy of interferon-γ-modified exosomal vaccine in prostate cancer. Prostate. 2020;80:811–23.

    Article  CAS  PubMed  Google Scholar 

  163. Rathi N, McFarland TR, Nussenzveig R, Agarwal N, Swami U. Evolving role of immunotherapy in metastatic castration refractory prostate cancer. Drugs. 2021;81:191–206.

    Article  CAS  PubMed  Google Scholar 

  164. Mentink A, Isebia KT, Kraan J, Terstappen LWMM, Stevens M. Measuring antigen expression of cancer cell lines and circulating tumour cells. Sci Rep. 2023;13:6051.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Es-Haghi M, Neustroeva O, Chowdhury I, Laitinen P, Väänänen M-A, Korvenlaita N, Malm T, Turunen MP, Turunen TA. Construction of fusion protein for enhanced small RNA loading to extracellular vesicles. Genes. 2023;14:261.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Hung ME, Leonard JN. A platform for actively loading cargo RNA to elucidate limiting steps in EV-mediated delivery. J Extracell Vesicles. 2016;5:31027.

    Article  PubMed  Google Scholar 

  167. Pan S, Zhang Y, Huang M, Deng Z, Zhang A, Pei L, Wang L, Zhao W, Ma L, Zhang Q, Cui D. Urinary exosomes-based engineered nanovectors for homologously targeted chemo-chemodynamic prostate cancer therapy via abrogating EGFR/AKT/NF-kB/IkB signaling. Biomaterials. 2021;275:120946.

    Article  CAS  PubMed  Google Scholar 

  168. Altanerova U, Babincova M, Babinec P, Benejova K, Jakubechova J, Altanerova V, Zduriencikova M, Repiska V, Altaner C. Human mesenchymal stem cell-derived iron oxide exosomes allow targeted ablation of tumor cells via magnetic hyperthermia. Int J Nanomedicine. 2017;12:7923–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Vázquez-Ríos AJ, Molina-Crespo Á, Bouzo BL, López-López R, Moreno-Bueno G, de la Fuente M. Exosome-mimetic nanoplatforms for targeted cancer drug delivery. J Nanobiotechnol. 2019;17:85.

    Article  Google Scholar 

  170. Zhao Q, Hai B, Kelly J, Wu S, Liu F. Extracellular vesicle mimics made from iPS cell-derived mesenchymal stem cells improve the treatment of metastatic prostate cancer. Stem Cell Res Ther. 2021;12:29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Severic M, Ma G, Pereira SGT, Ruiz A, Cheung CCL, Al-Jamal WT. Genetically-engineered anti-PSMA exosome mimetics targeting advanced prostate cancer in vitro and in vivo. J Control Release. 2021;330:101–10.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work is mainly supported by the by the Basic Public Welfare Research Project of Zhejiang Province (LGF22H050001-Han M, LTGY23H050002-Zhou C), Zhejiang Provincial Foundation for Medical and Health Sciences (Grant No. 2019KY575-Zhou C), Ningbo Top Medical and Health Research Program (No.2022020203) and Medical Health Science and Technology Project of Zhejiang Provincial Health Commission (2021KY1017-Han M), Ningbo Science Foundation (202002N3193-Jiang JH), St George Hospital Cancer Care Research Trust Fund and SSMRF. Haotian Chen is supported by Health Science Center, Ningbo University.

Author information

Authors and Affiliations

Authors

Contributions

YL, BP and JJ conceived the manuscript. HC conducted literature collection, article writing, and created the figures and tables. YL provided guidance throughout the preparation of this manuscript. BP, YL and JJ conducted paper revisions. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Yong Li or Junhui Jiang.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, H., Pang, B., Zhou, C. et al. Prostate cancer-derived small extracellular vesicle proteins: the hope in diagnosis, prognosis, and therapeutics. J Nanobiotechnol 21, 480 (2023). https://doi.org/10.1186/s12951-023-02219-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12951-023-02219-0

Keywords