Next Article in Journal
Humanized Monoclonal Antibody Against Citrullinated Histone H3 Attenuates Myocardial Injury and Prevents Heart Failure in Rodent Models
Previous Article in Journal
Study of the Lipophilicity of Tetracyclic Anticancer Azaphenothiazines
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Extracellular Vesicles Derived from Breast Cancer Cells: Emerging Biomarkers of Tumor Progression and Metastasis

1
Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
2
2nd Department of Oncology, Faculty of Medicine, Comenius University, Klenova 1, 833 10 Bratislava, Slovakia
*
Author to whom correspondence should be addressed.
Biomolecules 2025, 15(8), 1195; https://doi.org/10.3390/biom15081195
Submission received: 30 June 2025 / Revised: 31 July 2025 / Accepted: 12 August 2025 / Published: 19 August 2025

Abstract

Breast cancer (BC) remains one of the most prevalent and life-threatening malignancies worldwide, marked by significant heterogeneity and complex mechanisms of progression. Despite major advances in understanding its molecular and cellular basis, the processes driving tumor progression and metastasis continue to challenge effective treatment. Among the emerging research areas, extracellular vesicles (EVs) have gained considerable attention for their key role in intercellular communication and their contribution to cancer biology. In BC, tumor cell-derived EVs are implicated in multiple processes that promote disease progression, including tumor growth, remodeling of the tumor microenvironment, and facilitation of metastasis. By transferring oncogenic signals to recipient cells, EVs critically shape the metastatic niche and support the spread of cancer cells to distant organs. Recent studies highlight the diverse functions of BC-derived EVs in modulating immune responses, inducing angiogenesis, and enhancing cancer cell invasiveness. This review explores the role of BC-derived EVs in tumor progression and metastasis. We discuss their molecular composition, mechanisms of action, and impact on the tumor microenvironment, aiming to provide insights into their role in BC pathophysiology and discuss potential clinical applications. A deeper understanding of the complex interplay between EVs and cancer progression may pave the way for innovative strategies to combat BC and improve patient outcomes.

1. Introduction

In recent years, EVs have emerged as promising biomarkers for the early detection, prognosis, and monitoring of BC, owing to their unique molecular cargo and stability in bodily fluids [1,2,3]. Significant progress has been made in identifying and validating EV-derived biomarkers for BC. These advances are reshaping diagnostic paradigms by offering non-invasive, highly sensitive tools that complement—or even surpass—traditional biopsy-based approaches [4,5,6].
The ability to isolate and analyze EVs from easily accessible fluids enables non-invasive “liquid biopsies,” which are particularly valuable for early cancer detection and longitudinal disease monitoring. This approach not only reduces patient discomfort but also provides a more comprehensive representation of tumor heterogeneity, in contrast to tissue biopsies, which typically sample only a small portion of the tumor region [7].
Integrating EV-based biomarkers into personalized treatment strategies facilitates more precise and adaptive cancer management. As EV isolation and analytical technologies continue to evolve, and as additional biomarkers are validated in large-scale clinical trials, EV-based diagnostics are poised to become a cornerstone of precision oncology, offering renewed hope for improved patient outcomes.
Recent studies have identified a wide array of EV-derived biomarkers, including proteins, nucleic acids (e.g., DNA, microRNA (miRNA), messenger RNA (mRNA), circular RNA (circRNA), long non-coding RNA (lncRNA), small non-coding RNA (sncRNA), RNAs, transfer RNA-derived fragments (tRFs)), and metabolites. These molecules provide crucial insights into tumor biology, enabling early detection, subtype classification, and real-time monitoring of treatment response and disease progression [2,8,9,10].
The primary objective of this article is to comprehensively investigate the role of EVs derived from BC cells in tumor progression and metastasis, with a particular focus on their molecular cargo and their contribution to subtype-specific pathophysiology. The review also aims to explore the diagnostic, prognostic, and therapeutic potential of EVs in precision oncology.

2. Breast Cancer

BC is currently the most diagnosed cancer among women worldwide. Even when considering both sexes, it remains the second most prevalent cancer and the fourth leading cause of cancer-related death globally, as of 2022 [11].
Approximately one in eight to ten women will be diagnosed with BC during their lifetime [12], whereas less than 1% of men are diagnosed with the disease [13]. In total, there were 2.3 million new cases and 670,000 deaths due to female BC globally in 2022. Annual incidence rates are increasing by 1–5%.
A comparison between countries with a high and low Human Development Index (HDI and LDI) reveals that HDI countries show declining mortality rates. At the same time, LDI countries continue to experience an increase in BC-related deaths [14].
From a national perspective, in Slovakia, BC is the leading cause of cancer-related death among women, accounting for 27% of all female cancer cases. Regional disparities exist, with the capital city, Bratislava, reporting higher incidence rates compared to other regions. According to the European Cancer Information System, a 25% increase in total cancer cases is projected between 2022 and 2040, although this estimate is not specific to BC [15].
These data underline the ongoing global threat posed by BC and the pressing need for improvements in prevention, diagnostics, and treatment. As of now, PubMed lists 548,965 articles related to BC, with the earliest dating back to 1789—and the number continues to grow.
BC is not a singular disease with a single cause or outcome. It is a heterogeneous and multitype condition involving numerous initiating events and variations in biological and clinical behavior. This complexity is rooted in diverse genetic alterations that disrupt the function and regulation of specific genes and cellular processes [16,17,18].
Numerous factors contribute to BC oncogenesis, including genetic predisposition, environmental exposures, and lifestyle influences [19,20]. Interestingly, elements such as circadian rhythm disruptions, tumor-associated microbiota, and cancer stemness have also been identified as drivers of disease progression, adding further layers to the already intricate pathophysiology of BC [21].
According to the intrinsic classification proposed by Perou and Sørlie in 2000 [22], (BC) can be divided into four molecular subtypes: Luminal A and Luminal B (both characterized by estrogen receptor (ER) expression), basal-like (lacking ER, progesterone receptor (PR), and epidermal growth factor receptor 2 (HER2) expression—commonly referred to as triple-negative breast cancer (TNBC)), and HER2-enriched (lacking ER but overexpressing HER2).
The most common histological types of BC based on tissue origin are ductal carcinoma and lobular carcinoma, both of which are invasive lesions. Their non-invasive counterparts include ductal carcinoma in situ and lobular carcinoma in situ.
In current clinical practice, BC is typically classified into five subtypes based on histological and molecular characteristics, including the expression of ER, PR, HER2, and the proliferation marker Ki67. Tumors expressing ER and/or PR are classified as hormone receptor-positive, while those lacking ER, PR, and HER2 are categorized as triple-negative [23].
Based on the PAM50 gene expression classifier, which builds on these molecular features, five subtypes are identified: Luminal A, Luminal B, HER2-enriched, basal-like, and normal-like [24,25,26,27].
Currently, the World Health Organization (WHO) recognizes approximately 20 distinct histological types of breast carcinoma, in addition to invasive ductal carcinoma, which remains the most common subtype [28].
Despite these classification systems, further refinement is needed to better distinguish between the diverse forms of BC (Figure 1), thereby enabling more accurate diagnosis, prognosis, and personalized treatment strategies.

3. Extracellular Vesicles

EVs are a heterogeneous group of membrane-bound particles that include several subtypes based on their size and origin (Figure 2). These include apoptotic bodies (500 nm to 2 µm in diameter), microvesicles (100 nm to 1 µm), exosomes and small extracellular vesicles (sEVs) (30 to 150 nm) [29,30], and oncosomes (1 to 10 µm) [31,32]. However, current literature states different size limits for each category. In this regard, the Minimal Information for Studies of Extracellular Vesicles (MISEV) guidelines should be taken into consideration. An updated version was released in December 2023, which states the following EV size categories: sEVs under 200 nm in diameter, and large EVs over 200 nm in diameter. Other terms, such as exosomes, ectosomes or microvesicles are not recommended unless the subcellular origin is demonstrable [33].
In an alternative classification system, EVs are broadly divided into three main categories, with oncosomes considered a subtype of larger vesicles [34]. Other recognized subtypes in this framework include large dense core vesicles, membrane blebs, outer membrane vesicles, and prostasomes [35,36,37]. Additional specialized forms such as tolerosomes, proteasomes, prominosomes, migrasomes, exomeres, exophers, and mitovesicles have also been described [38,39].
EVs are found in prokaryotes, eukaryotes, and plants [36]. These vesicles formed by a lipid bilayer, are incapable of replication and do not contain a functional nucleus [37]. EVs are secreted into the extracellular space [30]. Microvesicles, also known as ectosomes [40], are formed directly from the plasma membrane and contain cargo from the cytoplasm [31,41]. Exosomes are formed by the fusion of multivesicular bodies (MVBs) with the plasma membrane, where MVB produces smaller vesicles (exosomes). Dying cells produce apoptotic bodies, which are present in higher quantities in the body compared to exosomes or microvesicles. Under specific conditions, their composition can vary across different biological fluids [31,42,43,44]. Oncosomes are large EVs formed from membrane protrusions and are primarily secreted by malignant cells [31,32,42]. Another type of EVs, argosomes, differs from exosomes in function. They carry morphogens—proteins that form concentration gradients within tissues and contribute to signal transduction processes, ensuring correct cell positioning during the development of multicellular organisms. Argosomes are believed to facilitate cargo transfer from donor to recipient cells [45,46]. Apoptotic blebs are membrane protrusions that arise when the plasma membrane detaches from the cortical cytoskeleton, usually due to increased hydrostatic pressure during cell contraction. These blebs, formed during programmed cell death, cover the entire cell surface and enclose chromatin and organelles. As apoptosis progresses, these blebs fragment into membrane-enclosed apoptotic bodies [46,47]. Tolerosomes are produced by intestinal endothelial cells [48], proteasomes are multistructural complexes designed for protein degradation [49,50], and prominosomes are associated with the plasma membrane, containing the prominin protein, and are considered organizational elements of the plasma membrane [51,52]. EVs are classified based on biogenesis, secretion pathways, size, content, and function [31,36,46]. Currently, markers specifying whether an EV originates from an endosome (exosomes) or the plasma membrane (ectosomes—microparticles/microvesicles) have not been precisely described. Assigning EVs to a specific subtype based on their biogenesis mechanism is complicated without real-time imaging [37,53,54]. Therefore, if EV identification based on precise markers of cellular origin is not possible, EVs should be identified according to the following categories: physical characteristics such as size: sEVs < 100 nm or <200 nm, and large or medium EVs > 200 nm; density—low, medium, or high, ranging from 1.06 to 1.21 g/mL [37,54], or 1.13 to 1.19 g/mL in sucrose [46,55]; biochemical content such as CD63+/CD81+-EVs, or EVs labeled with annexin V; or based on the description of the cell’s state and origin—EVs from podocytes, hypoxic EVs, large oncosomes, or apoptotic bodies [37]. As previously thought, EVs are a waste management system [56]; they have been identified as a communication interface delivering signaling biomolecules such as lipids, proteins, nucleic acids [57,58], thus driving key metabolic processes within functional physiology [59,60], creating a connection between EVs and functional pathways of various cancer physiologies including tumor progression and metastasis [61,62,63].

4. Extracellular Vesicles and Cancer

EVs are recognized as key mediators of intercellular communication, significantly influencing metabolic pathways in living organisms. As integral components of signaling networks, they are associated with a wide range of regulatory processes in biological systems. EVs play a critical role in cancer initiation, progression, and even regression of pathological states [64,65]. Their importance lies in their ability to transfer genetic and epigenetic regulatory factors between cells [66,67]. In addition to nucleic acids, EVs carry other bioactive molecules, such as proteins, which can be either enclosed within the vesicle or associated with its membrane [68,69]. Notably, lipids represent another major class of regulatory cargo, playing essential roles in cellular metabolism and the intricate mechanisms of EV-mediated communication [70]. These biomolecules, whether encapsulated in the EVs or present on their surface, are being intensively studied for their involvement in oncopathological processes, many of which are already known to contribute to cancer development.
In the following section, we highlight specific examples of EV cargo—comprising nucleic acids, proteins, and lipids—that contribute directly or indirectly to oncopathological processes (Table 1).

4.1. RNA EV Cargo Regulating Cancer Progression and Metastasis

According to Lei et al. [71], the lncRNA H19 is secreted into EVs, promoting resistance to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors in non-small-cell lung cancer cells. Additional evidence suggests that the presence of AGAP2 antisense RNA1 in EVs may contribute to resistance against human HER2 inhibitors [72]. Wang et al. [73] reported that in patients with colorectal cancer (CRC), EVs derived from the liver were enriched with miRNAs that regulate YAP signaling. These miRNAs suppressed LATS2 expression, thereby promoting CRC liver metastasis.
In renal cell carcinoma (RCC), EVs carrying circRNA circEHD2—found to be upregulated in tumor tissues—contribute to metastasis by inducing the secretion of the proinflammatory cytokine IL-6 [74]. Given that inflammation is a well-established contributor to cancer development [75,76,77], these findings further highlight the role of EVs in tumor-promoting inflammation. Another study focused on cancer-associated fibroblasts (CAFs), which are known to facilitate tumor growth and progression. A specific lncRNA was identified in EVs that correlates with CAF enrichment in bladder cancer [78]. Specifically, miR-181a-5p, present in EVs derived from CRC cells, has been shown to promote liver metastasis by activating hepatic stellate cells and modifying the tumor microenvironment [79]. In contrast, according to Luo et al. [80], miR-29a-3p-enriched EVs secreted by myocytes enhance anti-tumor immunity by downregulating collagen composition in tumors. Notably, the secretion of EVs containing miR-29a-3p is strongly upregulated by physical exercise.
There is also evidence for the presence of EV-associated cell-free mRNA (cf-mRNA) in various cancer types. Specifically, mRNAs 136, 199, 462, 151, and 105 were found to be significantly enriched in cancer samples compared to healthy controls [81]. Furthermore, miR-891b, which targets the phosphoserine aminotransferase 1 (PSAT1) gene, has been implicated in several cancers by regulating PSAT1 expression, potentially contributing to metastasis through EV-mediated signaling [82].
To this point, four types of RNA—miRNA, lncRNA, circRNA, and cf-mRNA—have been highlighted for their roles in EV cargo linked to oncopathology. However, additional RNA species are also present in EVs and are relevant to cancer biology, including mRNA [83], transfer RNA (tRNA) [84,85], small nuclear RNA (snRNA) [86], small nucleolar RNA (snoRNA) [87], mitochondrial RNA (mtRNA) [88,89], piwi-interacting RNA [90,91], Y RNAs [92], and retrotransposon-derived RNAs [93].
As these examples illustrate, RNA cargo in EVs is highly diverse and contributes through multiple mechanisms to tumor initiation, progression, and metastasis. Similar complexity and oncogenic potential have been observed in EV-associated proteins, DNA, lipids, and metabolites, further underscoring the multifaceted role of EVs in cancer biology.

4.2. DNA EV Cargo and Its Presence in Cancer

DNA cargo in EVs has emerged as a valuable tool in cancer diagnostics and prognostics. The phospholipid bilayer of EVs protects encapsulated nucleic acids from degradation by nucleases present in biological fluids. This protection makes EV-associated DNA a more stable and reliable candidate for cancer monitoring compared to freely circulating DNA (cfDNA) [94], enabling more precise and sensitive mutation detection [95,96,97,98,99,100]. DNA derived from EVs includes large-sized genomic fragments and tumor-specific mutant DNA [95]. In a study by Clancy and coworkers, a significant elevation of double-stranded DNA (dsDNA) was observed in EVs isolated from metastatic melanoma cell lines, compared to the primary A375P line and its metastatic variant A375-MA2 [99]. Another study described the role of oncogenic H-Ras—a GTPase involved in cell cycle regulation—in driving vesicle formation in intestinal epithelial cells. This process results in the release of sEVs containing chromatin-associated dsDNA, representing the host genome modified by oncogenic mutations. These EVs, which include DNA carrying the H-ras mutation, may interact with and influence normal cells [100]. Balaj et al. demonstrated that EVs released from tumors carry DNA reflecting tumor-specific features, such as amplification of the c-Myc oncogene [93]. Similarly, Kahlert et al. [101] identified dsDNA in serum-derived EVs from pancreatic cancer patients. These vesicles contained fragments over 10 kb in length, allowing for the detection of KRAS and TP53 mutations, thereby supporting the application of serum-based liquid biopsy for pancreatic cancer diagnosis.
Guescini and coworkers were the first to report the presence of mitochondrial DNA (mtDNA) in EVs secreted by glioblastoma cells and astrocytes. Their study showed that approximately 5% of total cellular mtDNA could be found in EVs, supporting the concept of horizontal mtDNA transfer between cells [102]. Overall, the field of EVs with DNA cargo is rapidly evolving, with a growing body of literature highlighting its complexity and diagnostic potential.

4.3. Protein EV Cargo in Cancer

Proteins involved in the regulation of oncopathology are found either within the intraluminal space of EVs or associated with their membrane surfaces [62]. According to Urabe et al., the EV-associated transmembrane protein CUB domain-containing protein 1 (CDCP1) acts as an inducer of osteoclastogenesis, thereby promoting metastasis in patients with prostate cancer [103]. A strong link between aging and melanoma progression was highlighted in the study by Hüser et al. [104]. The authors demonstrated that aging is associated with reduced expression of CD9—a key regulator of EV cargo sorting—which was diminished both in EVs and parental cells. This reduction was accompanied by an increase in angiopoietin-like protein 2, a potent pro-angiogenic factor, thereby facilitating angiogenesis and tumor progression. Further insights into the oncogenic potential of EVs were provided by McAtee et al. [105]. Their study revealed that EVs can induce the formation of filopodia, cellular structures essential for polarization, adhesion, and migration. The EVs responsible for this effect were enriched in the TGF-β family co-receptor endoglin, which modulates filopodia dynamics. Additionally, the presence of the transmembrane protein thrombospondin type 1 domain-containing 7A in EVs was shown to further enhance filopodia formation. In a pivotal study by Peinado et al. [106], several EV-associated proteins were proposed as potential biomarkers for melanoma progression. TYRP2, VLA-4, and HSP70 were significantly elevated in EVs from stage IV melanoma patients, while only TYRP2 showed significant elevation in stage III, correlating with metastatic potential. The study also demonstrated that the mesenchymal–epithelial transition (MET) oncoprotein, when transferred via EVs to bone marrow progenitor cells, could induce genetic and epigenetic changes associated with metastatic spread. Mass spectrometry analyses by Mears et al. [107] and Staubach et al. [108] confirmed that EVs secreted by melanoma and human breast carcinoma cells also carry mtDNA cargo, further supporting their role in horizontal transfer of oncogenic material. Overall, a growing body of literature underscores the presence and functional relevance of diverse protein molecules in EVs that are directly involved in cancer regulation and progression.

4.4. Lipid EV Cargo in Cancer

Lipids are one of the main components of EVs. EVs are rich in cholesterol (CHL), diglycerides, glycerophospholipids, sphingolipids and glycosylceramides [109,110]. In addition, they also contain bioactive lipids, such as phospholipase C and A2 [111,112,113]. These bioactive lipids, when transported by exosomes, can exert regulatory functions in recipient cells [114,115]. In a study by Skotland et al. [116], a specific combination and ratio of phosphatidylserine (PS) to ceramide (CER), as well as alterations in the levels of phosphatidylcholine (PC) and CHL within exosomes, were shown to reflect metabolic dysregulation in prostate cancer cells. These findings support the use of sEV-derived lipids from patient samples as potential biomarkers for prostate cancer diagnosis and therapy.
A separate study focusing on the metastatic prostate cancer cell line PC-3 quantified over 300 distinct lipids present in sEVs secreted by these cells. Glycosylated phospholipids were identified as promising biomarker candidates for malignant prostate pathology. Furthermore, the lipidome of sEVs differed significantly from that of the parent cells, with sEVs showing higher concentrations of glycosphingolipids, sphingomyelin SM, CHL, and PS [117]. Another study compared EV samples from patients with lung cancer, benign conditions, and healthy individuals. In EVs from lung cancer patients, elevated levels of lipids such as Cer(42:1), triglyceride(54:2), PC(24:0), PC-O(32:0), and lysophosphatidylcholine (LysoPC)(20:1) were detected. Increased levels of cholesteryl ester Cer(19:2) were found in patients with both benign and malignant conditions, compared to healthy controls. Conversely, higher levels of Cer(38:6), PC(42:2), PC(36:5), and PC(38:7) were observed in EVs from healthy individuals. The study also showed that lipid composition differs between serum and EV isolates, further emphasizing the functional specificity of EV cargo. However, due to methodological inconsistencies in EV isolation and normalization, as well as sample variability, it was not possible to construct a statistically robust multiplex lipid biomarker panel. Nonetheless, such a panel holds potential for future diagnostic or prognostic use [118]. In RCC, Del Boccio et al. [119] identified 48 EV-derived lipids with statistically significant alterations. Of these, 22 lipids were elevated and 26 were decreased under pathological conditions compared to healthy samples, suggesting their potential utility as biomarkers. Overall, growing evidence links lipid cargo variations in EVs with different types of cancer. These findings support the development of EV-based diagnostics, prognostics, and therapeutic strategies in oncology.

4.5. Metabolite EV Cargo in Cancer

In the study by Palviainen et al. [120], the authors provided the first evidence that metabolites carried by EVs reflect the altered metabolic state of various cancer pathologies. Metabolomic profiling of EVs derived from prostate, cutaneous T-cell lymphoma, and colon cancer cell lines revealed elevated levels of proline and succinate compared to normal control samples. Additionally, EVs from prostate and cutaneous T-cell lymphoma cell lines exhibited increased concentrations of creatinine and folate. Adenosine was identified as a functional EV cargo in the BC cell line MDA-MB-231-luc-D3H2LN. Acting as an immunosuppressive molecule, adenosine binds to its receptors on CD8+ cytotoxic T lymphocytes, impairing their immunogenic activity and thereby facilitating immune evasion by cancer cells [121]. A recent study on EV metabolomics in prostate cancer explored the potential of EV-derived metabolites for early diagnosis and risk assessment. Key metabolites identified included L-glutamic acid, pyruvic acid, lactic acid, and methylmalonic acid, all implicated in cancer-specific metabolic reprogramming [122]. In addition to these findings, glyoxalase enzymes, particularly glyoxalase 1 (GLO1) and glyoxalase 2 (GLO2), have recently been identified as components of tumor-derived EV cargo, including those from breast cancer cells [123,124]. These enzymes are central regulators of methylglyoxal detoxification, a reactive byproduct of glycolysis. Their encapsulation in EVs, alongside other glycolytic enzymes, suggests a role in intercellular metabolic signaling and adaptation. Moreover, MG-H1 (methylglyoxal-derived hydroimidazolone), a major advanced glycation end product produced from methylglyoxal, has been found in the same EV populations. This metabolite may act as a paracrine signal that facilitates metastatic niche formation and colonization [125]. These findings highlight glyoxalases and their byproducts as potential novel mediators of cancer progression through EV-based communication. Clos-García et al. [126] investigated urinary EVs’ metabolomic profiles in patients with prostate cancer compared to those with benign prostatic hyperplasia, proposing a non-invasive strategy for monitoring disease progression. They identified 76 metabolites with significantly different concentrations between the two groups. Alterations involved acylcarnitines, kynurenine, citrate, and PC, along with a notable increase in 3β-hydroxyandrost-5-en-17-one-3-sulfate in prostate cancer patients. In BC, EVs have been shown to transport elevated levels of uridine and guanosine compared to those from non-tumorigenic cell lines [121]. Similarly, sEVs from head and neck squamous cell carcinoma samples contained increased levels of purine metabolites such as adenosine and inosine—both known for their immunosuppressive functions [127]. In ovarian cancer, EVs were found to be enriched in L-kynurenine, a metabolite that promotes angiogenesis by inducing endothelial mitophagy, thereby contributing to metastatic potential [128]. Although research on the metabolite cargo of EVs in cancer is relatively recent, interest in this area is rapidly growing. The expanding list of EV-associated metabolites with diagnostic and prognostic relevance highlights their emerging role in oncological disease management. Nevertheless, further studies are needed to standardize methodologies and validate clinical applications.

5. EVs Derived from BC Cells

The diagnosis of BC remains challenging and often distressing for patients due to the invasive nature of tissue biopsies, which are essential for accurate disease classification and therapy planning. Imaging techniques such as sonography and mammography, followed by radiological assessment, can be time-consuming and are sometimes limited by suboptimal precision and the potential for human error. Current non-invasive prediagnostic methods, including the detection of biochemical markers in blood (e.g., Cancer Antigen 15-3, Carcinoembryonic antigen, Tissue/Mammary Cancer Antigen, etc.), are increasingly viewed as outdated and often lack sufficient accuracy.
In response, the scientific community is actively seeking diagnostic strategies that are simpler, faster, painless, more accurate, and—most importantly—less invasive. Among the most promising avenues is the analysis of EVs, which offer significant potential not only for early diagnosis but also for prognostic assessment and therapeutic decision-making in BC [129].

5.1. TNBC

TNBC is an aggressive subtype of BC defined by the absence of three key receptors: ER, PR, and human HER2. Due to the lack of these specific therapeutic targets, chemotherapy remains the primary treatment option, underscoring the urgent need for novel and more effective therapeutic strategies [130]. TNBC is further subclassified based on the expression of characteristic gene signatures (basal-like 1, basal-like 2, immunomodulatory, mesenchymal, mesenchymal stem-like and luminal androgen receptor).
The basal-like subtype, which represents approximately 10–20% of all BC cases, is defined by the expression of genes such as KRT5, CDH3, ID4, FABP7, KRT17, TRIM29, and LAMC2. This subtype is frequently associated with TP53 mutations and poor clinical prognosis. Another distinct subtype is claudin-low TNBC, which accounts for 12–14% of BC cases. In addition to lacking ER, PR, and HER2 expressions, this subtype is characterized by the expression of markers such as CD44 and SNAI3. Like the basal-like subtype, claudin-low TNBC exhibits a high frequency of TP53 mutations and is similarly associated with adverse clinical outcomes [131].

5.1.1. RNAs in TNBC EVs

EVs derived from TNBC cells contain bioactive molecules, including miRNAs, that contribute to tumor progression and metabolic reprogramming (Figure 3 and Table 2). One such molecule is miR-9-5p, which plays a significant role in CHL metabolism by targeting the genes INSIG1, INSIG2, and ATF3. In a BC mouse model, EV-associated miR-9-5p was found in significantly higher quantities compared to non-cancer controls, highlighting its potential as a cancer-specific metabolic regulator [132]. Another key molecule is miR-155-5p, a well-known oncomiR, which is upregulated in EVs released from BC cells. This miRNA is implicated in tumor initiation by activating the WNT signaling pathway through the downregulation of Adenomatous Polyposis Coli, a tumor suppressor gene. This suppression leads to pathway activation and promotes cancer progression. Additional targets of miR-155-5p include HSD17B12, MYC, SMAD1, SMAD3 and p53-inducible nuclear protein 1 (TP53INP1) all of which are downregulated, contributing to tumor advancement [133,134,135]. Furthermore, a study by Li et al. [136] demonstrated that miR-155-5p promotes both tumor progression and resistance to therapy by directly targeting TP53INP1, another tumor suppressor gene. In the study of Chen et al. [137], circHIF1A packed into EVs was identified, which plays a critical role in the growth and metastasis of TNBC. According to the study by Buschmann et al. [138], the overexpression of the glucocorticoid receptor significantly influences the miRNA composition of EVs derived from TNBC cells. In their analysis of EVs produced by the MDA-MB-231 and MDA-MB-468 TNBC cell lines, the most abundantly expressed miRNAs were identified as follows: MDA-MB-231: miR-100-5p, miR-21-5p, let-7f-5p, let-7i-5p, miR-486-5p, let-7a-5p, miR-92a-3p, let-7g-5p, miR-451a, and miR-27b-3p; MDA-MB-468: let-7f-2-3p, miR-103b, miR-4742-3p, let-7a-3p, miR-505-3p, let-7f-5p, let-7i-3p, miR-22-5p, let-7b-3p, and miR-196b-5p.
Additionally, miR-4516 was found to be significantly downregulated in CAF-derived EVs isolated from invasive ductal carcinoma samples, showing a fivefold decrease compared to normal tissue-derived EVs. This reduction suggests a potential tumor-suppressive role for miR-4516 in the TNBC microenvironment. Overexpression of miR-4516 in TNBC cells has been shown to inhibit cell proliferation. Its target gene, FOSL1, a transcription factor involved in tumor progression, was found to be overexpressed in TNBC cells, suggesting a tumor-suppressive role for miR-4516 [139]. A broader group of miRNAs associated with cancer progression and metastasis in TNBC-derived EVs includes miR-770, miR-9, miR-155, miR-221, miR-939, and the circRNA circPSMA1 [140]. In a study by Singh et al. [141], high levels of miR-10b were identified in EVs derived from TNBC cell lines. Upon uptake by recipient cells, miR-10b downregulated its target genes HOXD10 and KLF4, leading to enhanced cell invasion. Zhou et al. [142] further demonstrated that miR-105 is highly expressed in EVs derived from TNBC cells. This miRNA contributes to metastatic dissemination and vascular permeability in distant organs, promoting tumor progression. In a study by Ozawa et al. [143], the diagnostic potential of four EV-associated microRNAs—miR-142-5p, miR-150-5p, miR-320a, and miR-4433b-5p—was evaluated. These miRNAs were able to distinguish between Luminal A and TNBC subtypes, with miR-150-5p notably downregulated in EVs from TNBC samples. Furthermore, miR-421-5p was significantly overexpressed in TNBC-derived EVs and demonstrated high accuracy in differentiating TNBC from Luminal A cases, suggesting its value as a potential diagnostic biomarker. Another study investigated EV-derived miRNAs as prognostic indicators for recurrence in TNBC patients. miR-150-5p, miR-576-3p, and miR-4665-5p were found to be significantly upregulated in patients who experienced recurrence, highlighting their potential as non-invasive biomarkers for monitoring disease progression and guiding therapy decisions [144].
Table 2. TNBC EV-associated RNA bioactive molecules.
Table 2. TNBC EV-associated RNA bioactive molecules.
EV CargoTNBCFunctionReferences
RNAmiR-9-5pCHL metabolism[132]
RNAmiR-155-5pCancer initiation, WNT signaling pathway activation, tumor progression, drug resistance[133,134,135,136]
RNAmiR-4516Inhibition of cell proliferation[139]
RNAmiR-10bPromotion of cell invasion[141]
RNAmiR-105Metastatic and vascularization processes[142]
RNAmiR-142-5pDiagnostic potential for BC subtype differentiation[143]
RNAmiR-150-5pDiagnostic potential, downregulated in TNBC EVs compared to Luminal A[143]
RNAmiR-576-3pPrognostic marker for recurrence in TNBC[144]
RNAmiR-4665-5pPrognostic marker for recurrence in TNBC[144]
RNAmiR-421-5pDiscrimination between TNBC and Luminal A patients[143]
RNAmiR-100-5p, miR-21-5p, let-7f-5p, let-7i-5p, miR-486-5p, let-7a-5p, miR-92a-3p, let-7g-5p, miR-451a, miR-27b-3pSignificant expression in MDA-MB-231 cell line EVs, glucocorticoid receptor overexpression[138]
RNAlet-7f-2-3p, miR-103b, miR-4742-3p, let-7a-3p, miR-505-3p, let-7f-5p, let-7i-3p, miR-22-5p, let-7b-3p, miR-196b-5pSignificant expression in MDA-MB-468 cell line EVs, glucocorticoid receptor overexpression[138]
RNAmiR-770, miR-9, miR-155, miR-221, miR-939Metastatic processes[140]
RNAcircHIF1AGrowth and metastasis[137]
RNAcircPSMAMetastatic processes[140]

5.1.2. DNA in TNBC EVs

mtDNA is among the nucleic acid types identified in EVs derived from TNBC samples (Table 3). According to Vikramdeo et al. [145], EV-associated mtDNA in TNBC patients often harbors mutations, which may broaden current approaches to early diagnostics and preventive care. The study identified a total of 168 mtDNA mutations across all patient samples, with 73% located in coding regions and 27% in non-coding regions. Among these mutations, 80% were nucleotide transitions and 20% were transversions, with the G→A transition being the most frequently observed event. Each patient exhibited at least three distinct mutations. Mutations in non-coding regulatory regions, specifically RNR1 and RNR2, were detected in 15% of patients, while 12% of the mutations affected mitochondrial tRNA genes. Overall, mutation frequency in non-coding regions was low (around 3%), except for a few notable exceptions. For instance, the G1888A mutation was observed in 6% of patients. Among the tRNA-specific mutations, C5720T and T4434G were detected in 16% and 20% of patient samples, respectively. The remaining mutations were in coding regions encoding components of the mitochondrial respiratory chain:
-
Complex I: MT-ND1, ND2, ND3, ND4, ND4L, ND5, ND6;
-
Complex III: MT-CYTB;
-
Complex IV: MT-CO1, CO2, CO3;
-
Complex V: MT-ATP6, ATP8.
Mutations in these regions accounted for 44% of all mtDNA mutations. Notably, within Complex III, the T14894G mutation was found in 20% of the Complex III-specific mutations, far exceeding the average mutation rate of 3% in this region. In Complex IV, the T7953G mutation exhibited a remarkably high prevalence of 38%, compared to other mutations in the region, which ranged from 3% to 20% [145]. Another study demonstrated that EV-mediated transfer of mitochondria, specifically mutated mtND4, contributes to tumorigenesis and enhances chemoresistance in TNBC cells [146]. Furthermore, an intriguing study investigating the role of human papillomavirus (HPV) in TNBC found HPV DNA within EVs isolated from patient samples. These EVs could transfer viral DNA to stromal cells, which then exhibited an activated phenotype characterized by the expression of IL-6 and CD44. This transformation enhanced their ability to form mammospheres, suggesting a potential mechanistic link between HPV infection and TNBC initiation [147].

5.1.3. Proteins in TNBC EVs

According to Mashouri et al. [148], EVs originating from TNBC cells transport tumor-promoting proteins, such as EGFR and matrix metalloproteinases (MMPs), to target cells, boosting their ability to invade and migrate. The study by Raiter et al. [149] discusses the Gal3 binding protein in isolated sEVs in TNBC samples where the Gal3BP/Gal3 complex secreted via EVs induces immunosuppression through the CD45 receptor. According to the study by Desai et al. [150], Annexin A2, a calcium (Ca2+)-dependent phospholipid-binding protein which is transported within EV cargo, is involved in TNBC metastasis. In a different study, the cargo of EVs derived from MDA-MB-231 cells was investigated, revealing the presence of EGFR, ERBB2, and MAPK1, which are involved in the PI3K/Akt signaling pathway that promotes metabolism, proliferation, cell survival, and growth. These compounds may all phosphorylate PKM2, leading to the activation of aerobic glycolysis, more aggressive cell proliferation, and activation of the EMT [151]. In the article by Risha et al. [152], three potential extracellular vesicle membrane/surface proteins were identified as markers for TNBC. Specifically, these proteins are glypican-1, glucose transporter-1, and disintegrin and metalloproteinase domain-containing protein 10 (ADAM10). Xu et al. [153] identified various proteins isolated from EV cargo derived from TNBC, which are related to various cell metabolism regulatory processes, specifically, platelet activation-related proteins TLN2, VASP, and GNAS; antigen processing and presentation-related proteins B2M and PSMB9; regulation of actin cytoskeleton-related proteins CFL2, ITGB4, and GIT1; angiogenesis-related proteins CEACAM1, COL4A2, and CALD1; and cell motility-related proteins DST, ITGB4, and CFL2147 (Table 4).

5.1.4. Lipids in TNBC EVs

Lipids are a crucial component of cell signaling and, together with EVs, form a complex transport and signaling network in both physiological and pathological conditions within the organism (Table 5). Therefore, investigating their role in TNBC could be an essential factor in improving diagnostics, prognostics, and therapies for cancerous diseases. According to a study by Vikrameo et al. [145], the volume of cardiolipin associated with EVs from TNBC patient samples was significantly elevated. In another study, EVs derived from TNBC tumors were confirmed to traffic arachidonic acid, which plays a role in the reprogramming of neutrophils. This process leads to the accumulation of lipid droplets in cancer cells, thereby fostering an immunosuppressive tumor microenvironment [154]. In the study by D’Mello et al. [155], LysoPC 22:6/0:0 was proposed as an emerging biomarker for the diagnosis of TNBC.

5.1.5. Metabolites in TNBC EVs

Two metabolites, one of which is mentioned in the lipid TNBC section, were proposed as innovative biomarkers in relation to the diagnosis of TNBC, specifically the metabolite N-acetyl-L-phenylalanine, which was found to be unique to the MDA-MB-231 cell line investigated in the study [155].

5.2. Luminal A

Luminal A BC accounts for 50–60% of BC subtype prevalence worldwide. This molecular subtype is characterized by the expression of ER, PR, and the lack of expression of HER2-. Moreover, it is also characterized by the low expression of Ki-67. It is characterized by the following genes: ESR1, GATA3, KRT8, KRT18, XBP1, FOXA1, TFF3, CCND1, and LIV1. This subtype has a low prevalence of TP53 mutations, and patients generally have a good prognosis for recovery [131].

5.2.1. RNAs in Luminal A Subtype EVs

In the previously mentioned study by Ozawa et al. [143], it was found that miR-320a was overexpressed in Luminal A BC patient EVs compared to normal controls.

5.2.2. Proteins in Luminal A Subtype EVs

According to the study by Xu et al. [153], proteins isolated from EVs derived from Luminal A BC were characterized through proteomic analysis. A specific set of proteins was found to be associated with this BC subtype. The identified proteins include those related to proteolysis, such as SDCBP, COLEC11, and LTF; proteins involved in protein folding, including CCT2, HSP90AA1, and PPIG; proteins associated with the regulation of necroptotic cell death, namely HSP90AA1 and SDCBP; proteins linked to cellular stress responses and proteins associated with cellular responses to external stimuli such as HSBP1, IL6, and HSP90AA1 (Table 6).

5.3. Luminal B

Luminal B BC is characterized by the variable expression and absence of specific receptors in various combinations (ER+/−, PR+/−, HER2−/+), along with the expression of particular genes (ESR1, GATA3, KRT8, KRT18, XBP1, FOXA1, TFF3, SQLE, LAPTM4B). High expression of Ki-67 was also recorded. This subtype exhibits an intermediate incidence of TP53 mutations and accounts for 10–20% of all BC cases. Based on gene expression profiles and other molecular characteristics, Luminal B BC is associated with an intermediate to poor prognosis [129].

Proteins in Luminal B Subtype EVs

According to the study by Xu et al. [153], the following set of proteins isolated from EVs derived from Luminal B BC was characterized through proteomic analysis: MYL6, AFDN, tight junction-associated proteins, extracellular matrix-related receptor (ECM–receptor) interaction-associated proteins RELN and GP1BB, fructose and mannose metabolism-related proteins SLC2A14 and SLC2A3, glucose metabolism-related proteins SLC2A14 and SLC2A3, and insulin signaling pathway-associated proteins PKLR and PYGM (Table 7).

5.4. HER-2+

HER-2+ BC is characterized by expression of the HER receptor and accounts for 10–15% BC incidence. This subtype is characterized by the following specific genes: ERBB2, GRB7, and ITGA7. The rate of mutations in the TP53 region is low and prognosis of the disease is intermediate [129].

5.4.1. RNAs in HER-2+ Subtype EVs

According to the study by Chen et al. [9], several microRNAs are enriched in the EVs of the HER-2+ BC subtype. miR-29b-3p, miR-34c-5p, miR-203a-3p, miR-378g, and miR-382-5p are all involved in BC progression and, based on previous studies not focusing on EVs, are known as potential BC markers. However, this study had its limitations, as it utilized humanized mouse models, which may obscure the detection specificity for “only” human microRNAs. Nevertheless, the inclusion of next-generation sequencing and various non-coding RNAs, such as circular RNAs (circRNAs) and long non-coding RNAs (lncRNAs), may help address the speculated shortcomings of the method.

5.4.2. Proteins in HER-2+ Subtype EVs

Xu et al. [153] isolated the following proteins from EVs from HER-2+ BC: cellular response to hydrogen peroxide-related proteins SRC, ARG1, MAPK13, and ErbB2/ErbB3; signaling event-related proteins DOCK7 and SRC; keratinization-related proteins CDSN, KRT78, and KRT23; tyrosine metabolism-related proteins FAH and ADHFE1; and arginine and proline metabolism-related proteins ARG1 and RARS1 (Table 8).

6. Conclusions

  • EVs derived from BC cells have emerged as key players in tumor progression and metastasis, offering promising potential in modern diagnostics and therapeutic strategies. These vesicles, which carry diverse bioactive molecules such as RNA, DNA, proteins, lipids, and metabolites, are integral to intercellular communication and the modulation of the tumor microenvironment. EVs facilitate processes like immune evasion, angiogenesis, and metastatic niche formation, underscoring their significance in BC pathophysiology. Notably, EV cargo varies across BC subtypes, including TNBC, Luminal A, Luminal B, and HER-2+, reflecting unique molecular profiles that can be exploited for precision diagnostics and targeted therapies.
  • Despite their transformative potential, EV-based liquid biopsy diagnostics for BC remain experimental and unstandardized. Current findings highlight the relevance of EV cargo in subtype-specific BC characterization, yet comprehensive references for all bioactive molecules in each subtype are limited. Advances in EV research could pave the way for non-invasive diagnostic tools, enabling early detection, subtype differentiation, and real-time monitoring of disease progression. Continued exploration of EVs and their molecular cargo holds promise for revolutionizing BC management, offering hope for improved patient outcomes through precision oncology.
Future research must focus on refining EV-based diagnostics to enable non-invasive, precise, and personalized cancer management. By leveraging the unique molecular cargo of EVs, innovative diagnostic tools can be developed to classify BC subtypes with greater accuracy, monitor disease progression, and predict therapeutic responses.
  • As per the current understanding of BC and extracellular vesicle EV-based potential clinical applications, we recommend the following future directions for improved diagnostics, prognostics, and therapeutics:
    (1)
    Standardized EV isolation and profiling: Developing robust and reproducible protocols for EV isolation, such as advanced microfluidic systems and immunoaffinity techniques, will enhance the consistency and scalability of EV-based diagnostics. Improved characterization methods using next-generation sequencing, mass spectrometry, and metabolomics can uncover novel subtype-specific biomarkers.
    (2)
    Integration of artificial intelligence: Employing AI and machine learning algorithms to analyze EV molecular data could accelerate biomarker discovery, improve subtype classification, and predict patient outcomes with high precision. This approach will enable the identification of complex diagnostic patterns in EV cargo.
    (3)
    Multiplex panels for subtype differentiation: Comprehensive profiling of EV cargo across BC subtypes can facilitate the creation of multiplex panels for early detection, subtype classification, and therapy monitoring. These panels could combine RNA, DNA, protein, lipid, and metabolite biomarkers to improve diagnostic accuracy.
    (4)
    Therapeutic modulation of EV biogenesis: Investigating methods to modulate EV production and cargo content could lead to novel therapeutic strategies. For example, engineering EVs for targeted drug delivery or disrupting EV-mediated oncogenic signaling pathways could reduce tumor aggressiveness and metastasis.
    (5)
    Clinical validation and trials: Expanding large-scale clinical trials to validate EV-based biomarkers and diagnostic tools is critical for translating experimental findings into clinical practice. Collaborations between research institutions, industry, and healthcare providers can accelerate the development of EV-based technologies.
    (6)
    Subtype-specific therapeutic approaches: Tailoring therapies based on the molecular cargo of EVs in specific BC subtypes, such as TNBC or HER-2+, could improve treatment efficacy. For example, targeting EVs enriched with oncogenic RNAs or proteins in TNBC could offer new therapeutic avenues.
  • By addressing these research and development priorities, EVs could become central to precision oncology, transforming the landscape of BC management and improving patient outcomes.

Author Contributions

Conceptualization, L.D. and M.B.; methodology, S.B.; formal analysis, S.B., A.N. and M.C.; writing—original draft preparation, S.B. and A.N.; writing—review and editing, S.B., A.N., M.C., L.D. and M.B.; supervision, L.D.; project administration, S.B.; funding acquisition, S.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Comenius University, grant number UK/1256/2025.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

During the preparation of this manuscript, the authors used ChatGPT-4.0 and Grammarly for the purposes of writing assistance and grammar control. The authors have reviewed and edited the output and take full responsibility for the content of this publication.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
ADAM10Metalloproteinase domain-containing protein 10
BCBreast cancer
CAFsCancer-associated fibroblasts
CDCP1CUB domain-containing protein 1
CerCeramide
cfDNAFreely circulating DNA
cf-mRNACell-free mRNA
circRNACircular RNA
CRCColorectal cancer
dsDNADouble-stranded DNA
ECM-receptorExtracellular matrix-associated receptor
EGFREpidermal growth factor receptor
EMTEpithelial–mesenchymal transition
EREstrogen receptor
EVsExtracellular vesicles
HER2Epidermal growth factor receptor 2
HPVHuman papillomavirus
CHLCholesterol
lncRNALong non-coding RNA
LysoPCLysophosphatidylcholine
METMesenchymal–epithelial transition
miRNAMicroRNA
MISEVMinimal information for studies of extracellular vesicles
MMPsMatrix metalloproteinases
mRNAMessenger RNA
mtDNAMitochondrial DNA
MVEsMultivesicular endosome
PCPhosphatidylcholine
PRProgesterone receptor
PSPhosphatidylserine
RCCRenal cell carcinoma
sEVsSmall extracellular vesicles
sncRNASmall non-coding RNA
THSD7AThrombospondin type 1 domain-containing 7A
TNBCTriple negative breast cancer
TP53INP1p53-inducible nuclear protein 1
tRFstRNA-derived fragments
WHOWorld Health Organization

References

  1. Piombino, C.; Mastrolia, I.; Omarini, C.; Candini, O.; Dominici, M.; Piacentini, F.; Toss, A. The Role of Exosomes in Breast Cancer Diagnosis. Biomedicines 2021, 9, 312. [Google Scholar] [CrossRef] [PubMed]
  2. Zhang, X.; Wang, C.; Yu, J.; Bu, J.; Ai, F.; Wang, Y.; Lin, J.; Zhu, X. Extracellular Vesicles in the Treatment and Diagnosis of Breast Cancer: A Status Update. Front. Endocrinol. 2023, 14, 1202493. [Google Scholar] [CrossRef] [PubMed]
  3. Maqsood, Q.; Sumrin, A.; Saleem, Y.; Wajid, A.; Mahnoor, M. Exosomes in Cancer: Diagnostic and Therapeutic Applications. Clin. Med. Insights Oncol. 2024, 18, 11795549231215966. [Google Scholar] [CrossRef]
  4. Loric, S.; Denis, J.A.; Desbene, C.; Sabbah, M.; Conti, M. Extracellular Vesicles in Breast Cancer: From Biology and Function to Clinical Diagnosis and Therapeutic Management. Int. J. Mol. Sci. 2023, 24, 7208. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  5. Andre, M.; Caobi, A.; Miles, J.S.; Vashist, A.; Ruiz, M.A.; Raymond, A.D. Diagnostic potential of exosomal extracellular vesicles in oncology. BMC Cancer 2024, 24, 322. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  6. Saadh, M.J.; Al-Rihaymee, A.M.A.; Kaur, M.; Kumar, A.; Mutee, A.F.; Ismaeel, G.L.; Shomurotova, S.; Alubiady, M.H.S.; Hamzah, H.F.; Alhassan, Z.A.A.; et al. Advancements in Exosome Proteins for Breast Cancer Diagnosis and Detection: With a Focus on Nanotechnology. AAPS PharmSciTech 2024, 25, 276. [Google Scholar] [CrossRef]
  7. Wang, Z.; Wang, Q.; Qin, F.; Chen, J. Exosomes: A Promising Avenue for Cancer Diagnosis beyond Treatment. Front. Cell Dev. Biol. 2024, 12, 1344705. [Google Scholar] [CrossRef]
  8. Nakase, I.; Takatani-Nakase, T. Exosomes: Breast Cancer-Derived Extracellular Vesicles; Recent Key Findings and Technologies in Disease Progression, Diagnostics, and Cancer Targeting. Drug Metab. Pharmacokinet. 2022, 42, 100435. [Google Scholar] [CrossRef]
  9. Chen, I.-P.; Henning, S.; Bender, M.; Degenhardt, S.; Mhamdi Ghodbani, M.; Bergmann, A.K.; Volkmer, B.; Brockhoff, G.; Wege, A.K.; Greinert, R. Detection of Human Circulating and Extracellular Vesicle-Derived miRNAs in Serum of Humanized Mice Transplanted with Human Breast Cancer (HER2+ and TNBC) Cells—A Proof of Principle Investigation. Int. J. Mol. Sci. 2025, 26, 3629. [Google Scholar] [CrossRef]
  10. Zayakin, P.; Sadovska, L.; Eglītis, K.; Romanchikova, N.; Radoviča-Spalviņa, I.; Endzeliņš, E.; Liepniece-Karele, I.; Eglītis, J.; Linē, A. Extracellular Vesicles—A Source of RNA Biomarkers for the Detection of Breast Cancer in Liquid Biopsies. Cancers 2023, 15, 4329. [Google Scholar] [CrossRef]
  11. Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global Cancer Statistics 2022: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA A Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef]
  12. Houghton, S.C.; Hankinson, S.E. Cancer Progress and Priorities: Breast Cancer. Cancer Epidemiol. Biomark. Prev. 2021, 30, 822–844. [Google Scholar] [CrossRef]
  13. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer Statistics, 2019. CA A Cancer J. Clin. 2019, 69, 7–34. [Google Scholar] [CrossRef]
  14. Kim, J.; Harper, A.; McCormack, V.; Sung, H.; Houssami, N.; Morgan, E.; Mutebi, M.; Garvey, G.; Soerjomataram, I.; Fidler-Benaoudia, M.M. Global Patterns and Trends in Breast Cancer Incidence and Mortality across 185 Countries. Nat. Med. 2025, 31, 1154–1162. [Google Scholar] [CrossRef]
  15. OECD. European Commission EU Country Cancer Profile: Slovak Republic 2025; EU Country Cancer Profiles; OECD Publishing: Paris, France, 2025. [Google Scholar]
  16. Hanahan, D.; Weinberg, R.A. The Hallmarks of Cancer. Cell 2000, 100, 57–70. [Google Scholar] [CrossRef]
  17. Simpson, P.T.; Reis-Filho, J.S.; Gale, T.; Lakhani, S.R. Molecular Evolution of Breast Cancer. J. Pathol. 2005, 205, 248–254. [Google Scholar] [CrossRef]
  18. Reis-Filho, J.S.; Simpson, P.T.; Gale, T.; Lakhani, S.R. The Molecular Genetics of Breast Cancer: The Contribution of Com-parative Genomic Hybridization. Pathol.—Res. Pract. 2005, 201, 713–725. [Google Scholar] [CrossRef] [PubMed]
  19. Akram, M.; Iqbal, M.; Daniyal, M.; Khan, A.U. Awareness and Current Knowledge of Breast Cancer. Biol. Res. 2017, 50, 33. [Google Scholar] [CrossRef]
  20. Sun, Y.-S.; Zhao, Z.; Yang, Z.-N.; Xu, F.; Lu, H.-J.; Zhu, Z.-Y.; Shi, W.; Jiang, J.; Yao, P.-P.; Zhu, H.-P. Risk Factors and Preventions of Breast Cancer. Int. J. Biol. Sci. 2017, 13, 1387–1397. [Google Scholar] [CrossRef] [PubMed]
  21. Swanton, C.; Bernard, E.; Abbosh, C.; André, F.; Auwerx, J.; Balmain, A.; Bar-Sagi, D.; Bernards, R.; Bullman, S.; DeGregori, J.; et al. Embracing Cancer Complexity: Hallmarks of Systemic Disease. Cell 2024, 187, 1589–1616. [Google Scholar] [CrossRef] [PubMed]
  22. Perou, C.M.; Sørlie, T.; Eisen, M.B.; Van De Rijn, M.; Jeffrey, S.S.; Rees, C.A.; Pollack, J.R.; Ross, D.T.; Johnsen, H.; Akslen, L.A.; et al. Molecular Portraits of Human Breast Tumours. Nature 2000, 406, 747–752. [Google Scholar] [CrossRef]
  23. Harbeck, N.; Penault-Llorca, F.; Cortes, J.; Gnant, M.; Houssami, N.; Poortmans, P.; Ruddy, K.; Tsang, J.; Cardoso, F. Breast Cancer. Nat. Rev. Dis. Primers 2019, 5, 66. [Google Scholar] [CrossRef]
  24. Sørlie, T.; Perou, C.M.; Tibshirani, R.; Aas, T.; Geisler, S.; Johnsen, H.; Hastie, T.; Eisen, M.B.; Van De Rijn, M.; Jeffrey, S.S.; et al. Gene Expression Patterns of Breast Carcinomas Distinguish Tumor Subclasses with Clinical Implications. Proc. Natl. Acad. Sci. USA 2001, 98, 10869–10874. [Google Scholar] [CrossRef]
  25. Parker, J.S.; Mullins, M.; Cheang, M.C.U.; Leung, S.; Voduc, D.; Vickery, T.; Davies, S.; Fauron, C.; He, X.; Hu, Z.; et al. Supervised Risk Predictor of Breast Cancer Based on Intrinsic Subtypes. J. Clin. Oncol. 2009, 27, 1160–1167. [Google Scholar] [CrossRef] [PubMed]
  26. Nielsen, T.O.; Hsu, F.D.; Jensen, K.; Cheang, M.; Karaca, G.; Hu, Z.; Hernandez-Boussard, T.; Livasy, C.; Cowan, D.; Dressler, L.; et al. Immunohistochemical and Clinical Characterization of the Basal-Like Subtype of Invasive Breast Carcinoma. Clin. Cancer Res. 2004, 10, 5367–5374. [Google Scholar] [CrossRef] [PubMed]
  27. Prat, A.; Parker, J.S.; Karginova, O.; Fan, C.; Livasy, C.; Herschkowitz, J.I.; He, X.; Perou, C.M. Phenotypic and Molecular Characterization of the Claudin-Low Intrinsic Subtype of Breast Cancer. Breast Cancer Res. 2010, 12, R68. [Google Scholar] [CrossRef] [PubMed]
  28. Tan, P.H.; Ellis, I.; Allison, K.; Brogi, E.; Fox, S.B.; Lakhani, S.; Lazar, A.J.; Morris, E.A.; Sahin, A.; Salgado, R.; et al. The 2019 World Health Organization Classification of Tumours of the Breast. Histopathology 2020, 77, 181–185. [Google Scholar] [CrossRef] [PubMed]
  29. Shanmuganathan, M.; Vughs, J.; Noseda, M.; Emanueli, C. Exosomes: Basic Biology and Technological Advancements Suggesting Their Potential as Ischemic Heart Disease Therapeutics. Front. Physiol. 2018, 9, 1159. [Google Scholar] [CrossRef]
  30. Brennan, K.; Martin, K.; FitzGerald, S.P.; O’Sullivan, J.; Wu, Y.; Blanco, A.; Richardson, C.; Mc Gee, M.M. A Comparison of Methods for the Isolation and Separation of Extracellular Vesicles from Protein and Lipid Particles in Human Serum. Sci. Rep. 2020, 10, 1039. [Google Scholar] [CrossRef]
  31. Zaborowski, M.P.; Balaj, L.; Breakefield, X.O.; Lai, C.P. Extracellular Vesicles: Composition, Biological Relevance, and Methods of Study. BioScience 2015, 65, 783–797. [Google Scholar] [CrossRef]
  32. Di Vizio, D.; Morello, M.; Dudley, A.C.; Schow, P.W.; Adam, R.M.; Morley, S.; Mulholland, D.; Rotinen, M.; Hager, M.H.; Insabato, L.; et al. Large Oncosomes in Human Prostate Cancer Tissues and in the Circulation of Mice with Metastatic Disease. Am. J. Pathol. 2012, 181, 1573–1584. [Google Scholar] [CrossRef]
  33. Welsh, J.A.; Goberdhan, D.C.I.; O’Driscoll, L.; Buzas, E.I.; Blenkiron, C.; Bussolati, B.; Cai, H.; Di Vizio, D.; Driedonks, T.A.P.; Erdbrügger, U.; et al. Minimal Information for Studies of Extracellular Vesicles (MISEV2023): From Basic to Advanced Approaches. J. Extracell. Vesicle 2024, 13, e12404. [Google Scholar] [CrossRef]
  34. Raposo, G.; Nijman, H.W.; Stoorvogel, W.; Liejendekker, R.; Harding, C.V.; Melief, C.J.; Geuze, H.J. B Lymphocytes Secrete Antigen-Presenting Vesicles. J. Exp. Med. 1996, 183, 1161–1172. [Google Scholar] [CrossRef]
  35. Stegmayr, B.; Ronquist, G. Promotive Effect on Human Sperm Progressive Motility by Prostasomes. Urol. Res. 1982, 10, 253–257. [Google Scholar] [CrossRef] [PubMed]
  36. Yáñez-Mó, M.; Siljander, P.R.-M.; Andreu, Z.; Bedina Zavec, A.; Borràs, F.E.; Buzas, E.I.; Buzas, K.; Casal, E.; Cappello, F.; Carvalho, J.; et al. Biological Properties of Extracellular Vesicles and Their Physiological Functions. J. Extracell. Vesicle 2015, 4, 27066. [Google Scholar] [CrossRef] [PubMed]
  37. Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal Information for Studies of Extracellular Vesicles 2018 (MISEV2018): A Position Statement of the International Society for Extracellular Vesicles and Update of the MISEV2014 Guidelines. J. Extracell. Vesicle 2018, 7, 1535750. [Google Scholar] [CrossRef]
  38. Vlassov, A.V.; Magdaleno, S.; Setterquist, R.; Conrad, R. Exosomes: Current Knowledge of Their Composition, Biological Functions, and Diagnostic and Therapeutic Potentials. Biochim. Biophys. Acta (BBA)—Gen. Subj. 2012, 1820, 940–948. [Google Scholar] [CrossRef] [PubMed]
  39. Wee, I.; Syn, N.; Sethi, G.; Goh, B.C.; Wang, L. Role of Tumor-Derived Exosomes in Cancer Metastasis. Biochim. Biophys. Acta (BBA)—Rev. Cancer 2019, 1871, 12–19. [Google Scholar] [CrossRef]
  40. Cocucci, E.; Meldolesi, J. Ectosomes. Curr. Biol. 2011, 21, R940–R941. [Google Scholar] [CrossRef]
  41. Heijnen, H.F.G.; Schiel, A.E.; Fijnheer, R.; Geuze, H.J.; Sixma, J.J. Activated Platelets Release Two Types of Membrane Vesicles: Microvesicles by Surface Shedding and Exosomes Derived From Exocytosis of Multivesicular Bodies and-Granules. Blood 1999, 94, 3791–3799. [Google Scholar] [CrossRef]
  42. Morello, M.; Minciacchi, V.; De Candia, P.; Yang, J.; Posadas, E.; Kim, H.; Griffiths, D.; Bhowmick, N.; Chung, L.; Gan-dellini, P.; et al. Large Oncosomes Mediate Intercellular Transfer of Functional microRNA. Cell Cycle 2013, 12, 3526–3536. [Google Scholar] [CrossRef]
  43. El Andaloussi, S.; Mäger, I.; Breakefield, X.O.; Wood, M.J.A. Extracellular Vesicles: Biology and Emerging Therapeutic Opportunities. Nat. Rev. Drug. Discov. 2013, 12, 347–357. [Google Scholar] [CrossRef]
  44. Théry, C.; Boussac, M.; Véron, P.; Ricciardi-Castagnoli, P.; Raposo, G.; Garin, J.; Amigorena, S. Proteomic Analysis of Dendritic Cell-Derived Exosomes: A Secreted Subcellular Compartment Distinct from Apoptotic Vesicles. J. Immunol. 2001, 166, 7309–7318. [Google Scholar] [CrossRef]
  45. Greco, V.; Hannus, M.; Eaton, S. Argosomes: A potential vehicle for the spread of morphogens through epithelia. Cell 2001, 106, 633–645. [Google Scholar] [CrossRef]
  46. Borges, F.T.; Reis, L.A.; Schor, N. Extracellular Vesicles: Structure, Function, and Potential Clinical Uses in Renal Diseases. Braz. J. Med. Biol. Res. 2013, 46, 824–830. [Google Scholar] [CrossRef]
  47. Wickman, G.; Julian, L.; Olson, M.F. How Apoptotic Cells Aid in the Removal of Their Own Cold Dead Bodies. Cell Death Differ. 2012, 19, 735–742. [Google Scholar] [CrossRef]
  48. Karlsson, M.; Lundin, S.; Dahlgren, U.; Kahu, H.; Pettersson, I.; Telemo, E. “Tolerosomes” Are Produced by Intestinal Epithelial Cells. Eur. J. Immunol. 2001, 31, 2892–2900. [Google Scholar] [CrossRef] [PubMed]
  49. Rivett, A.J. Proteasomes: Multicatalytic Proteinase Complexes. Biochem. J. 1993, 291, 1–10. [Google Scholar] [CrossRef] [PubMed]
  50. Etlinger, J.D.; Goldberg, A.L. A Soluble ATP-Dependent Proteolytic System Responsible for the Degradation of Abnormal Proteins in Reticulocytes. Proc. Natl. Acad. Sci. USA 1977, 74, 54–58. [Google Scholar] [CrossRef] [PubMed]
  51. Karbanová, J.; Thamm, K.; Fargeas, C.A.; Deniz, I.A.; Lorico, A.; Corbeil, D. Prominosomes—A Particular Class of Extracellular Vesicles Containing Prominin-1/CD133? J. Nanobiotechnol. 2025, 23, 61. [Google Scholar] [CrossRef]
  52. Huttner, H.B.; Janich, P.; Köhrmann, M.; Jászai, J.; Siebzehnrubl, F.; Blümcke, I.; Suttorp, M.; Gahr, M.; Kuhnt, D.; Nimsky, C.; et al. The Stem Cell Marker Prominin-1/CD133 on Membrane Particles in Human Cerebrospinal Fluid Offers Novel Approaches for Studying Central Nervous System Disease. Stem Cells 2008, 26, 698–705. [Google Scholar] [CrossRef] [PubMed]
  53. Stein, J.M.; Luzio, J.P. Ectocytosis Caused by Sublytic Autologous Complement Attack on Human Neutrophils. The Sorting of Endogenous Plasma-Membrane Proteins and Lipids into Shed Vesicles. Biochem. J. 1991, 274, 381–386. [Google Scholar] [CrossRef] [PubMed]
  54. Cocucci, E.; Meldolesi, J. Ectosomes and Exosomes: Shedding the Confusion between Extracellular Vesicles. Trends Cell Biol. 2015, 25, 364–372. [Google Scholar] [CrossRef]
  55. Mathivanan, S.; Ji, H.; Simpson, R.J. Exosomes: Extracellular Organelles Important in Intercellular Communication. J. Proteom. 2010, 73, 1907–1920. [Google Scholar] [CrossRef] [PubMed]
  56. Kooijmans, S.A.A.; De Jong, O.G.; Schiffelers, R.M. Exploring Interactions between Extracellular Vesicles and Cells for Innovative Drug Delivery System Design. Adv. Drug Deliv. Rev. 2021, 173, 252–278. [Google Scholar] [CrossRef]
  57. Zhou, Y.; Xiao, Z.; Zhu, W. The Roles of Small Extracellular Vesicles as Prognostic Biomarkers and Treatment Approaches in Triple-Negative Breast Cancer. Front. Oncol. 2022, 12, 998964. [Google Scholar] [CrossRef]
  58. H. Rashed, M.; Bayraktar, E.; Helal, G.K.; Abd-Ellah, M.; Amero, P.; Chavez-Reyes, A.; Rodriguez-Aguayo, C. Exosomes: From Garbage Bins to Promising Therapeutic Targets. Int. J. Mol. Sci. 2017, 18, 538. [Google Scholar] [CrossRef]
  59. Colombo, M.; Raposo, G.; Théry, C. Biogenesis, Secretion, and Intercellular Interactions of Exosomes and Other Extracel-lular Vesicles. Annu. Rev. Cell Dev. Biol. 2014, 30, 255–289. [Google Scholar] [CrossRef]
  60. Meldolesi, J. Exosomes and Ectosomes in Intercellular Communication. Curr. Biol. 2018, 28, R435–R444. [Google Scholar] [CrossRef]
  61. Chang, W.-H.; Cerione, R.A.; Antonyak, M.A. Extracellular Vesicles and Their Roles in Cancer Progression. In Cancer Cell Signaling; Robles-Flores, M., Ed.; Methods in Molecular Biology; Springer: New York, NY, USA, 2021; Volume 2174, pp. 143–170. ISBN 978-1-0716-0758-9. [Google Scholar]
  62. Kalluri, R.; McAndrews, K.M. The Role of Extracellular Vesicles in Cancer. Cell 2023, 186, 1610–1626. [Google Scholar] [CrossRef]
  63. Lopez, K.; Lai, S.W.T.; Lopez Gonzalez, E.D.J.; Dávila, R.G.; Shuck, S.C. Extracellular Vesicles: A Dive into Their Role in the Tumor Microenvironment and Cancer Progression. Front. Cell Dev. Biol. 2023, 11, 1154576. [Google Scholar] [CrossRef] [PubMed]
  64. Wu, C.; Li, J.; Li, L.; Sun, J.; Fabbri, M.; Wayne, A.S.; Seeger, R.C.; Jong, A.Y. Extracellular Vesicles Derived from Natural Killer Cells Use Multiple Cytotoxic Proteins and Killing Mechanisms to Target Cancer Cells. J. Extracell. Vesicle 2019, 8, 1588538. [Google Scholar] [CrossRef]
  65. Cianciaruso, C.; Phelps, E.A.; Pasquier, M.; Hamelin, R.; Demurtas, D.; Alibashe Ahmed, M.; Piemonti, L.; Hirosue, S.; Swartz, M.A.; De Palma, M.; et al. Primary Human and Rat β-Cells Release the Intracellular Autoantigens GAD65, IA-2, and Proinsulin in Exosomes Together With Cytokine-Induced Enhancers of Immunity. Diabetes 2017, 66, 460–473. [Google Scholar] [CrossRef]
  66. Hánělová, K.; Raudenská, M.; Masařík, M.; Balvan, J. Protein Cargo in Extracellular Vesicles as the Key Mediator in the Progression of Cancer. Cell Commun. Signal. 2024, 22, 25. [Google Scholar] [CrossRef]
  67. Escola, J.-M.; Kleijmeer, M.J.; Stoorvogel, W.; Griffith, J.M.; Yoshie, O.; Geuze, H.J. Selective Enrichment of Tetraspan Proteins on the Internal Vesicles of Multivesicular Endosomes and on Exosomes Secreted by Human B-Lymphocytes. J. Biol. Chem. 1998, 273, 20121–20127. [Google Scholar] [CrossRef]
  68. Valadi, H.; Ekström, K.; Bossios, A.; Sjöstrand, M.; Lee, J.J.; Lötvall, J.O. Exosome-Mediated Transfer of mRNAs and mi-croRNAs Is a Novel Mechanism of Genetic Exchange between Cells. Nat. Cell Biol. 2007, 9, 654–659. [Google Scholar] [CrossRef]
  69. Ratajczak, J.; Miekus, K.; Kucia, M.; Zhang, J.; Reca, R.; Dvorak, P.; Ratajczak, M.Z. Embryonic Stem Cell-Derived Mi-crovesicles Reprogram Hematopoietic Progenitors: Evidence for Horizontal Transfer of mRNA and Protein Delivery. Leukemia 2006, 20, 847–856. [Google Scholar] [CrossRef]
  70. Fyfe, J.; Casari, I.; Manfredi, M.; Falasca, M. Role of Lipid Signalling in Extracellular Vesicles-Mediated Cell-to-Cell Com-munication. Cytokine Growth Factor Rev. 2023, 73, 20–26. [Google Scholar] [CrossRef] [PubMed]
  71. Lei, Y.; Guo, W.; Chen, B.; Chen, L.; Gong, J.; Li, W. Tumor-released lncRNA H19 Promotes Gefitinib Resistance via Packaging into Exosomes in Non-small Cell Lung Cancer. Oncol. Rep. 2018, 40, 3034–3446. [Google Scholar] [CrossRef]
  72. La Camera, G.; Gelsomino, L.; Caruso, A.; Panza, S.; Barone, I.; Bonofiglio, D.; Andò, S.; Giordano, C.; Catalano, S. The Emerging Role of Extracellular Vesicles in Endocrine Resistant Breast Cancer. Cancers 2021, 13, 1160. [Google Scholar] [CrossRef]
  73. Wang, Z.; Kim, S.Y.; Tu, W.; Kim, J.; Xu, A.; Yang, Y.M.; Matsuda, M.; Reolizo, L.; Tsuchiya, T.; Billet, S.; et al. Extracellular Vesicles in Fatty Liver Promote a Metastatic Tumor Microenvironment. Cell Metab. 2023, 35, 1209–1226.e13. [Google Scholar] [CrossRef] [PubMed]
  74. He, T.; Zhang, Q.; Xu, P.; Tao, W.; Lin, F.; Liu, R.; Li, M.; Duan, X.; Cai, C.; Gu, D.; et al. Extracellular Vesicle-circEHD2 Promotes the Progression of Renal Cell Carcinoma by Activating Cancer-Associated Fibroblasts. Mol. Cancer 2023, 22, 117. [Google Scholar] [CrossRef] [PubMed]
  75. Mantovani, A. Inflammation by Remote Control. Nature 2005, 435, 752–753. [Google Scholar] [CrossRef]
  76. Bonjoch, L.; Casas, V.; Carrascal, M.; Closa, D. Involvement of Exosomes in Lung Inflammation Associated with Experimental Acute Pancreatitis: Exosomes in Acute Pancreatitis. J. Pathol. 2016, 240, 235–245. [Google Scholar] [CrossRef]
  77. Choi, H.-I.; Choi, J.-P.; Seo, J.; Kim, B.J.; Rho, M.; Han, J.K.; Kim, J.G. Helicobacter Pylori-Derived Extracellular Vesicles Increased in the Gastric Juices of Gastric Adenocarcinoma Patients and Induced Inflammation Mainly via Specific Targeting of Gastric Epithelial Cells. Exp. Mol. Med. 2017, 49, e330. [Google Scholar] [CrossRef]
  78. Li, Y.; Zheng, H.; Luo, Y.; Lin, Y.; An, M.; Kong, Y.; Zhao, Y.; Yin, Y.; Ai, L.; Huang, J.; et al. An HGF-Dependent Positive Feedback Loop between Bladder Cancer Cells and Fibroblasts Mediates Lymphangiogenesis and Lymphatic Metastasis. Cancer Commun. 2023, 43, 1289–1311. [Google Scholar] [CrossRef]
  79. Zhao, S.; Mi, Y.; Zheng, B.; Wei, P.; Gu, Y.; Zhang, Z.; Xu, Y.; Cai, S.; Li, X.; Li, D. Highly-metastatic Colorectal Cancer Cell Released miR-181a-5p-rich Extracellular Vesicles Promote Liver Metastasis by Activating Hepatic Stellate Cells and Remodelling the Tumour Microenvironment. J. Extracell. Vesicle 2022, 11, e12186. [Google Scholar] [CrossRef]
  80. Luo, Z.; Mei, J.; Wang, X.; Wang, R.; He, Z.; Geffen, Y.; Sun, X.; Zhang, X.; Xu, J.; Wan, R.; et al. Voluntary Exercise Sensi-tizes Cancer Immunotherapy via the Collagen Inhibition-Orchestrated Inflammatory Tumor Immune Microenvironment. Cell Rep. 2024, 43, 114697. [Google Scholar] [CrossRef]
  81. Kim, H.J.; Rames, M.J.; Goncalves, F.; Kirschbaum, C.W.; Roskams-Hieter, B.; Spiliotopoulos, E.; Briand, J.; Doe, A.; Es-tabrook, J.; Wagner, J.T.; et al. Selective Enrichment of Plasma Cell-Free Messenger RNA in Cancer-Associated Extracellular Vesicles. Commun. Biol. 2023, 6, 885. [Google Scholar] [CrossRef] [PubMed]
  82. Yamamoto, T.; Nakayama, J.; Urabe, F.; Ito, K.; Nishida-Aoki, N.; Kitagawa, M.; Yokoi, A.; Kuroda, M.; Hattori, Y.; Yamamoto, Y.; et al. Aberrant Regulation of Serine Metabolism Drives Extracellular Vesicle Release and Cancer Progression. Cell Rep. 2024, 43, 114517. [Google Scholar] [CrossRef]
  83. Zhang, P.; Lim, S.B.; Jiang, K.; Chew, T.W.; Low, B.C.; Lim, C.T. Distinct mRNAs in Cancer Extracellular Vesicles Activate Angiogenesis and Alter Transcriptome of Vascular Endothelial Cells. Cancers 2021, 13, 2009. [Google Scholar] [CrossRef]
  84. Zhu, L.; Li, J.; Gong, Y.; Wu, Q.; Tan, S.; Sun, D.; Xu, X.; Zuo, Y.; Zhao, Y.; Wei, Y.-Q.; et al. Exosomal tRNA-Derived Small RNA as a Promising Biomarker for Cancer Diagnosis. Mol. Cancer 2019, 18, 74. [Google Scholar] [CrossRef]
  85. Kumar, S.R.; Kimchi, E.T.; Manjunath, Y.; Gajagowni, S.; Stuckel, A.J.; Kaifi, J.T. RNA Cargos in Extracellular Vesicles Derived from Blood Serum in Pancreas Associated Conditions. Sci. Rep. 2020, 10, 2800. [Google Scholar] [CrossRef]
  86. Liu, Y.; Gu, Y.; Han, Y.; Zhang, Q.; Jiang, Z.; Zhang, X.; Huang, B.; Xu, X.; Zheng, J.; Cao, X. Tumor Exosomal RNAs Promote Lung Pre-Metastatic Niche Formation by Activating Alveolar Epithelial TLR3 to Recruit Neutrophils. Cancer Cell 2016, 30, 243–256. [Google Scholar] [CrossRef] [PubMed]
  87. Driedonks, T.A.P.; Van Der Grein, S.G.; Ariyurek, Y.; Buermans, H.P.J.; Jekel, H.; Chow, F.W.N.; Wauben, M.H.M.; Buck, A.H.; Hoen, P.A.C.‘t.; Nolte-‘t Hoen, E.N.M. Immune Stimuli Shape the Small Non-Coding Transcriptome of Extracellular Vesi-cles Released by Dendritic Cells. Cell. Mol. Life Sci. 2018, 75, 3857–3875. [Google Scholar] [CrossRef] [PubMed]
  88. Sansone, P.; Savini, C.; Kurelac, I.; Chang, Q.; Amato, L.B.; Strillacci, A.; Stepanova, A.; Iommarini, L.; Mastroleo, C.; Daly, L.; et al. Packaging and Transfer of Mitochondrial DNA via Exosomes Regulate Escape from Dormancy in Hormonal Thera-py-Resistant Breast Cancer. Proc. Natl. Acad. Sci. USA 2017, 114, E9066–E9075. [Google Scholar] [CrossRef]
  89. Rabas, N.; Palmer, S.; Mitchell, L.; Ismail, S.; Gohlke, A.; Riley, J.S.; Tait, S.W.G.; Gammage, P.; Soares, L.L.; Macpherson, I.R.; et al. PINK1 Drives Production of mtDNA-Containing Extracellular Vesicles to Promote Invasiveness. J. Cell Biol. 2021, 220, e202006049. [Google Scholar] [CrossRef]
  90. Peng, Q.; Chiu, P.K.-F.; Wong, C.Y.-P.; Cheng, C.K.-L.; Teoh, J.Y.-C.; Ng, C.-F. Identification of piRNA Targets in Urinary Extracellular Vesicles for the Diagnosis of Prostate Cancer. Diagnostics 2021, 11, 1828. [Google Scholar] [CrossRef]
  91. Li, Y.; Dong, Y.; Zhao, S.; Gao, J.; Hao, X.; Wang, Z.; Li, M.; Wang, M.; Liu, Y.; Yu, X.; et al. Serum-Derived piR-Hsa-164586 of Extracellular Vesicles as a Novel Biomarker for Early Diagnosis of Non-Small Cell Lung Cancer. Front. Oncol. 2022, 12, 850363. [Google Scholar] [CrossRef] [PubMed]
  92. Chakrabortty, S.K.; Prakash, A.; Nechooshtan, G.; Hearn, S.; Gingeras, T.R. Extracellular Vesicle-Mediated Transfer of Processed and Functional RNY5 RNA. RNA 2015, 21, 1966–1979. [Google Scholar] [CrossRef] [PubMed]
  93. Balaj, L.; Lessard, R.; Dai, L.; Cho, Y.-J.; Pomeroy, S.L.; Breakefield, X.O.; Skog, J. Tumour Microvesicles Contain Re-trotransposon Elements and Amplified Oncogene Sequences. Nat. Commun. 2011, 2, 180. [Google Scholar] [CrossRef]
  94. Adashek, J.J.; Janku, F.; Kurzrock, R. Signed in Blood: Circulating Tumor DNA in Cancer Diagnosis, Treatment and Screening. Cancers 2021, 13, 3600. [Google Scholar] [CrossRef]
  95. Zocco, D.; Bernardi, S.; Novelli, M.; Astrua, C.; Fava, P.; Zarovni, N.; Carpi, F.M.; Bianciardi, L.; Malavenda, O.; Quaglino, P.; et al. Isolation of Extracellular Vesicles Improves the Detection of Mutant DNA from Plasma of Metastatic Melanoma Patients. Sci. Rep. 2020, 10, 15745. [Google Scholar] [CrossRef]
  96. Castellanos-Rizaldos, E.; Grimm, D.G.; Tadigotla, V.; Hurley, J.; Healy, J.; Neal, P.L.; Sher, M.; Venkatesan, R.; Karlovich, C.; Raponi, M.; et al. Exosome-Based Detection of EGFR T790M in Plasma from Non–Small Cell Lung Cancer Patients. Clin. Cancer Res. 2018, 24, 2944–2950. [Google Scholar] [CrossRef]
  97. Tutanov, O.; Shtam, T.; Grigor’eva, A.; Tupikin, A.; Tsentalovich, Y.; Tamkovich, S. Blood Plasma Exosomes Contain Cir-culating DNA in Their Crown. Diagnostics 2022, 12, 854. [Google Scholar] [CrossRef]
  98. Cui, S.; Cheng, Z.; Qin, W.; Jiang, L. Exosomes as a Liquid Biopsy for Lung Cancer. Lung Cancer 2018, 116, 46–54. [Google Scholar] [CrossRef]
  99. Clancy, J.W.; Sheehan, C.S.; Boomgarden, A.C.; D’Souza-Schorey, C. Recruitment of DNA to Tumor-Derived Microvesicles. Cell Rep. 2022, 38, 110443. [Google Scholar] [CrossRef] [PubMed]
  100. Lee, T.H.; Chennakrishnaiah, S.; Audemard, E.; Montermini, L.; Meehan, B.; Rak, J. Oncogenic Ras-Driven Cancer Cell Vesiculation Leads to Emission of Double-Stranded DNA Capable of Interacting with Target Cells. Biochem. Biophys. Res. Commun. 2014, 451, 295–301. [Google Scholar] [CrossRef] [PubMed]
  101. Kahlert, C.; Melo, S.A.; Protopopov, A.; Tang, J.; Seth, S.; Koch, M.; Zhang, J.; Weitz, J.; Chin, L.; Futreal, A.; et al. Identi-fication of Double-Stranded Genomic DNA Spanning All Chromosomes with Mutated KRAS and P53 DNA in the Serum Exo-somes of Patients with Pancreatic Cancer. J. Biol. Chem. 2014, 289, 3869–3875. [Google Scholar] [CrossRef] [PubMed]
  102. Guescini, M.; Genedani, S.; Stocchi, V.; Agnati, L.F. Astrocytes and Glioblastoma Cells Release Exosomes Carrying mtDNA. J. Neural Transm. 2010, 117, 1–4. [Google Scholar] [CrossRef]
  103. 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. Vesicle 2023, 12, 12312. [Google Scholar] [CrossRef]
  104. Hüser, L.; Chhabra, Y.; Gololobova, O.; Wang, V.; Liu, G.; Dixit, A.; Rocha, M.R.; Harper, E.I.; Fane, M.E.; Marino-Bravante, G.E.; et al. Aged Fibroblast-Derived Extracellular Vesicles Promote Angiogenesis in Melanoma. Cell Rep. 2024, 43, 114721. [Google Scholar] [CrossRef] [PubMed]
  105. McAtee, C.; Patel, M.; Hoshino, D.; Sung, B.H.; Von Lersner, A.; Shi, M.; Hong, N.H.; Young, A.; Krystofiak, E.; Zijlstra, A.; et al. Secreted Exosomes Induce Filopodia Formation. bioRxiv 2024. [Google Scholar] [CrossRef] [PubMed]
  106. Peinado, H.; Alečković, M.; Lavotshkin, S.; Matei, I.; Costa-Silva, B.; Moreno-Bueno, G.; Hergueta-Redondo, M.; Williams, C.; García-Santos, G.; Ghajar, C.; et al. Melanoma Exosomes Educate Bone Marrow Progenitor Cells toward a Pro-Metastatic Phenotype through MET. Nat. Med. 2012, 18, 883–891. [Google Scholar] [CrossRef]
  107. Mears, R.; Craven, R.A.; Hanrahan, S.; Totty, N.; Upton, C.; Young, S.L.; Patel, P.; Selby, P.J.; Banks, R.E. Proteomic Analysis of Melanoma-derived Exosomes by Two-dimensional Polyacrylamide Gel Electrophoresis and Mass Spectrometry. Proteomics 2004, 4, 4019–4031. [Google Scholar] [CrossRef]
  108. Staubach, S.; Razawi, H.; Hanisch, F. Proteomics of MUC1-containing Lipid Rafts from Plasma Membranes and Exosomes of Human Breast Carcinoma Cells MCF-7. Proteomics 2009, 9, 2820–2835. [Google Scholar] [CrossRef]
  109. Ghadami, S.; Dellinger, K. The Lipid Composition of Extracellular Vesicles: Applications in Diagnostics and Therapeutic Delivery. Front. Mol. Biosci. 2023, 10, 1198044. [Google Scholar] [CrossRef]
  110. Skotland, T.; Sagini, K.; Sandvig, K.; Llorente, A. An Emerging Focus on Lipids in Extracellular Vesicles. Adv. Drug Deliv. Rev. 2020, 159, 308–321. [Google Scholar] [CrossRef] [PubMed]
  111. Subra, C.; Grand, D.; Laulagnier, K.; Stella, A.; Lambeau, G.; Paillasse, M.; De Medina, P.; Monsarrat, B.; Perret, B.; Sil-vente-Poirot, S.; et al. Exosomes Account for Vesicle-Mediated Transcellular Transport of Activatable Phospholipases and Pros-taglandins. J. Lipid Res. 2010, 51, 2105–2120. [Google Scholar] [CrossRef]
  112. Record, M.; Carayon, K.; Poirot, M.; Silvente-Poirot, S. Exosomes as New Vesicular Lipid Transporters Involved in Cell–Cell Communication and Various Pathophysiologies. Biochim. Et Biophys. Acta (BBA)—Mol. Cell Biol. Lipids 2014, 1841, 108–120. [Google Scholar] [CrossRef]
  113. Keller, S.; Ridinger, J.; Rupp, A.-K.; Janssen, J.W.; Altevogt, P. Body Fluid Derived Exosomes as a Novel Template for Clinical Diagnostics. J. Transl. Med. 2011, 9, 86. [Google Scholar] [CrossRef]
  114. Zakharova, L.; Svetlova, M.; Fomina, A.F. T Cell Exosomes Induce Cholesterol Accumulation in Human Monocytes via Phosphatidylserine Receptor. J. Cell. Physiol. 2007, 212, 174–181. [Google Scholar] [CrossRef] [PubMed]
  115. Donoso-Quezada, J.; Ayala-Mar, S.; González-Valdez, J. The Role of Lipids in Exosome Biology and Intercellular Commu-nication: Function, Analytics and Applications. Traffic 2021, 22, 204–220. [Google Scholar] [CrossRef]
  116. Skotland, T.; Ekroos, K.; Kauhanen, D.; Simolin, H.; Seierstad, T.; Berge, V.; Sandvig, K.; Llorente, A. Molecular Lipid Species in Urinary Exosomes as Potential Prostate Cancer Biomarkers. Eur. J. Cancer 2017, 70, 122–132. [Google Scholar] [CrossRef]
  117. Hamed, M.A.; Wasinger, V.; Wang, Q.; Graham, P.; Malouf, D.; Bucci, J.; Li, Y. Prostate Cancer-Derived Extracellular Vesicles Metabolic Biomarkers: Emerging Roles for Diagnosis and Prognosis. J. Control. Release 2024, 371, 126–145. [Google Scholar] [CrossRef] [PubMed]
  118. Smolarz, M.; Kurczyk, A.; Jelonek, K.; Żyła, J.; Mielańczyk, Ł.; Sitkiewicz, M.; Pietrowska, M.; Polańska, J.; Rzyman, W.; Widłak, P. The Lipid Composition of Serum-Derived Small Extracellular Vesicles in Participants of a Lung Cancer Screening Study. Cancers 2021, 13, 3414. [Google Scholar] [CrossRef]
  119. Del Boccio, P.; Raimondo, F.; Pieragostino, D.; Morosi, L.; Cozzi, G.; Sacchetta, P.; Magni, F.; Pitto, M.; Urbani, A. A Hyphenated microLC-Q-TOF-MS Platform for Exosomal Lipidomics Investigations: Application to RCC Urinary Exosomes. Electrophoresis 2012, 33, 689–696. [Google Scholar] [CrossRef]
  120. 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. [Google Scholar] [CrossRef]
  121. Tadokoro, H.; Hirayama, A.; Kudo, R.; Hasebe, M.; Yoshioka, Y.; Matsuzaki, J.; Yamamoto, Y.; Sugimoto, M.; Soga, T.; Ochiya, T. Adenosine Leakage from Perforin-Burst Extracellular Vesicles Inhibits Perforin Secretion by Cytotoxic T-Lymphocytes. PLoS ONE 2020, 15, e0231430. [Google Scholar] [CrossRef]
  122. Hamed, M.A.; Wasinger, V.; Wang, Q.; Biazik, J.; Graham, P.; Malouf, D.; Bucci, J.; Li, Y. Optimising Extracellular Vesicle Metabolomic Methodology for Prostate Cancer Biomarker Discovery. Metabolites 2024, 14, 367. [Google Scholar] [CrossRef] [PubMed]
  123. Romani, R.; Talesa, V.N.; Antognelli, C. The Glyoxalase System Is a Novel Cargo of Amniotic Fluid Stem-Cell-Derived Extracellular Vesicles. Antioxidants 2022, 11, 1524. [Google Scholar] [CrossRef]
  124. Mc Cluskey, M.; Dubouchaud, H.; Nicot, A.-S.; Saudou, F. A Vesicular Warburg Effect: Aerobic Glycolysis Occurs on Axonal Vesicles for Local NAD+ Recycling and Transport. Traffic 2024, 25, e12926. [Google Scholar] [CrossRef]
  125. Xu, K.Y.; Becker, L.C. Ultrastructural Localization of Glycolytic Enzymes on Sarcoplasmic Reticulum Vesticles. J. Histochem. Cytochem. 1998, 46, 419–427. [Google Scholar] [CrossRef]
  126. Clos-Garcia, M.; Loizaga-Iriarte, A.; Zuñiga-Garcia, P.; Sánchez-Mosquera, P.; Rosa Cortazar, A.; González, E.; Torrano, V.; Alonso, C.; Pérez-Cormenzana, M.; Ugalde-Olano, A.; et al. Metabolic Alterations in Urine Extracellular Vesicles Are Associated to Prostate Cancer Pathogenesis and Progression. J. Extracell. Vesicle 2018, 7, 1470442. [Google Scholar] [CrossRef]
  127. Ludwig, N.; Gillespie, D.G.; Reichert, T.E.; Jackson, E.K.; Whiteside, T.L. Purine Metabolites in Tumor-Derived Exosomes May Facilitate Immune Escape of Head and Neck Squamous Cell Carcinoma. Cancers 2020, 12, 1602. [Google Scholar] [CrossRef]
  128. Ying, X.; Zheng, X.; Zhang, X.; Yin, Y.; Wang, X. Kynurenine in IDO1high Cancer Cell-Derived Extracellular Vesicles Promotes Angiogenesis by Inducing Endothelial Mitophagy in Ovarian Cancer. J. Transl. Med. 2024, 22, 267. [Google Scholar] [CrossRef]
  129. Su, Y.; Li, Y.; Guo, R.; Zhao, J.; Chi, W.; Lai, H.; Wang, J.; Wang, Z.; Li, L.; Sang, Y.; et al. Plasma Extracellular Vesicle Long RNA Profiles in the Diagnosis and Prediction of Treatment Response for Breast Cancer. npj Breast Cancer 2021, 7, 154. [Google Scholar] [CrossRef]
  130. Bhullar, A.S.; Jin, K.; Shi, H.; Jones, A.; Hironaka, D.; Xiong, G.; Xu, R.; Guo, P.; Binzel, D.W.; Shu, D. Engineered Extra-cellular Vesicles for Combinatorial TNBC Therapy: SR-SIM-Guided Design Achieves Substantial Drug Dosage Reduction. Mol. Ther. 2024, 32, 4467–4481. [Google Scholar] [CrossRef]
  131. Eroles, P.; Bosch, A.; Alejandro Pérez-Fidalgo, J.; Lluch, A. Molecular Biology in Breast Cancer: Intrinsic Subtypes and Signaling Pathways. Cancer Treat. Rev. 2012, 38, 698–707. [Google Scholar] [CrossRef]
  132. Li, M.-X.; Hu, S.; Lei, H.-H.; Yuan, M.; Li, X.; Hou, W.-K.; Huang, X.-J.; Xiao, B.-W.; Yu, T.-X.; Zhang, X.-H.; et al. Tu-mor-Derived miR-9-5p-Loaded EVs Regulate Cholesterol Homeostasis to Promote Breast Cancer Liver Metastasis in Mice. Nat. Commun. 2024, 15, 10539. [Google Scholar] [CrossRef]
  133. Yi, Y.; Wu, M.; Zeng, H.; Hu, W.; Zhao, C.; Xiong, M.; Lv, W.; Deng, P.; Zhang, Q.; Wu, Y. Tumor-Derived Exosomal Non-Coding RNAs: The Emerging Mechanisms and Potential Clinical Applications in Breast Cancer. Front. Oncol. 2021, 11, 738945. [Google Scholar] [CrossRef]
  134. Ozawa, P.M.M.; Alkhilaiwi, F.; Cavalli, I.J.; Malheiros, D.; de Souza Fonseca Ribeiro, E.M.; Cavalli, L.R. Extracellular Vesicles from Triple-Negative Breast Cancer Cells Promote Proliferation and Drug Resistance in Non-Tumorigenic Breast Cells. Breast Cancer Res. Treat. 2018, 172, 713–723. [Google Scholar] [CrossRef]
  135. Seillier, M.; Peuget, S.; Gayet, O.; Gauthier, C.; N’Guessan, P.; Monte, M.; Carrier, A.; Iovanna, J.L.; Dusetti, N.J. TP53INP1, a Tumor Suppressor, Interacts with LC3 and ATG8-Family Proteins through the LC3-Interacting Region (LIR) and Promotes Autophagy-Dependent Cell Death. Cell Death Differ. 2012, 19, 1525–1535. [Google Scholar] [CrossRef]
  136. Li, Y.; Zhang, L.; Dong, Z.; Xu, H.; Yan, L.; Wang, W.; Yang, Q.; Chen, C. MicroRNA-155-5p Promotes Tumor Progression and Contributes to Paclitaxel Resistance via TP53INP1 in Human Breast Cancer. Pathol.—Res. Pract. 2021, 220, 153405. [Google Scholar] [CrossRef]
  137. Chen, T.; Wang, X.; Li, C.; Zhang, H.; Liu, Y.; Han, D.; Li, Y.; Li, Z.; Luo, D.; Zhang, N.; et al. CircHIF1A Regulated by FUS Accelerates Triple-Negative Breast Cancer Progression by Modulating NFIB Expression and Translocation. Oncogene 2021, 40, 2756–2771. [Google Scholar] [CrossRef]
  138. Buschmann, D.; González, R.; Kirchner, B.; Mazzone, C.; Pfaffl, M.W.; Schelling, G.; Steinlein, O.; Reithmair, M. Glucocor-ticoid Receptor Overexpression Slightly Shifts microRNA Expression Patterns in Triple-Negative Breast Cancer. Int. J. Oncol. 2018, 52, 1765–1776. [Google Scholar] [CrossRef]
  139. Kim, J.E.; Kim, B.G.; Jang, Y.; Kang, S.; Lee, J.H.; Cho, N.H. The Stromal Loss of miR-4516 Promotes the FOSL1-Dependent Proliferation and Malignancy of Triple Negative Breast Cancer. Cancer Lett. 2020, 469, 256–265. [Google Scholar] [CrossRef]
  140. Dong, M.; Liu, Q.; Xu, Y.; Zhang, Q. Extracellular Vesicles: The Landscape in the Progression, Diagnosis, and Treatment of Triple-Negative Breast Cancer. Front. Cell Dev. Biol. 2022, 10, 842898. [Google Scholar] [CrossRef]
  141. Singh, R.; Pochampally, R.; Watabe, K.; Lu, Z.; Mo, Y.-Y. Exosome-Mediated Transfer of miR-10b Promotes Cell Invasion in Breast Cancer. Mol. Cancer 2014, 13, 256. [Google Scholar] [CrossRef]
  142. Zhou, W.; Fong, M.Y.; Min, Y.; Somlo, G.; Liu, L.; Palomares, M.R.; Yu, Y.; Chow, A.; O’Connor, S.T.F.; Chin, A.R.; et al. Cancer-Secreted miR-105 Destroys Vascular Endothelial Barriers to Promote Metastasis. Cancer Cell 2014, 25, 501–515. [Google Scholar] [CrossRef]
  143. Ozawa, P.M.M.; Vieira, E.; Lemos, D.S.; Souza, I.L.M.; Zanata, S.M.; Pankievicz, V.C.; Tuleski, T.R.; Souza, E.M.; Wowk, P.F.; Urban, C.D.A.; et al. Identification of miRNAs Enriched in Extracellular Vesicles Derived from Serum Samples of Breast Cancer Patients. Biomolecules 2020, 10, 150. [Google Scholar] [CrossRef]
  144. Wu, H.; Wang, Q.; Zhong, H.; Li, L.; Zhang, Q.; Huang, Q.; Yu, Z. Differentially Expressed microRNAs in Exosomes of Patients with Breast Cancer Revealed by Next-generation Sequencing. Oncol. Rep. 2019, 43, 240–250. [Google Scholar] [CrossRef]
  145. Vikramdeo, K.S.; Anand, S.; Sudan, S.K.; Pramanik, P.; Singh, S.; Godwin, A.K.; Singh, A.P.; Dasgupta, S. Profiling Mitochondrial DNA Mutations in Tumors and Circulating Extracellular Vesicles of Triple-negative Breast Cancer Patients for Potential Biomarker Development. FASEB BioAdvances 2023, 5, 412–426. [Google Scholar] [CrossRef]
  146. Abad, E.; Lyakhovich, A. Movement of Mitochondria with Mutant DNA through Extracellular Vesicles Helps Cancer Cells Acquire Chemoresistance. ChemMedChem 2022, 17, e202100642. [Google Scholar] [CrossRef]
  147. De Carolis, S.; Storci, G.; Ceccarelli, C.; Savini, C.; Gallucci, L.; Sansone, P.; Santini, D.; Seracchioli, R.; Taffurelli, M.; Fabbri, F.; et al. HPV DNA Associates With Breast Cancer Malignancy and It Is Transferred to Breast Cancer Stromal Cells by Extracellular Vesicles. Front. Oncol. 2019, 9, 860. [Google Scholar] [CrossRef]
  148. Mashouri, L.; Yousefi, H.; Aref, A.R.; Ahadi, A.M.; Molaei, F.; Alahari, S.K. Exosomes: Composition, Biogenesis, and Mechanisms in Cancer Metastasis and Drug Resistance. Mol. Cancer 2019, 18, 75. [Google Scholar] [CrossRef]
  149. Raiter, A.; Lipovetsky, J.; Stenbac, A.; Lubin, I.; Yerushalmi, R. TNBC-Derived Gal3BP/Gal3 Complex Induces Immuno-suppression through CD45 Receptor. Oncoimmunology 2023, 12, 2246322. [Google Scholar] [CrossRef]
  150. Desai, P.P.; Narra, K.; James, J.D.; Jones, H.P.; Tripathi, A.K.; Vishwanatha, J.K. Combination of Small Extracellular Vesi-cle-Derived Annexin A2 Protein and mRNA as a Potential Predictive Biomarker for Chemotherapy Responsiveness in Aggressive Triple-Negative Breast Cancer. Cancers 2022, 15, 212. [Google Scholar] [CrossRef]
  151. Kang, S.Y.; Lee, E.J.; Byun, J.W.; Han, D.; Choi, Y.; Hwang, D.W.; Lee, D.S. Extracellular Vesicles Induce an Aggressive Phenotype in Luminal Breast Cancer Cells Via PKM2 Phosphorylation. Front. Oncol. 2021, 11, 785450. [Google Scholar] [CrossRef]
  152. Risha, Y.; Minic, Z.; Ghobadloo, S.M.; Berezovski, M.V. The Proteomic Analysis of Breast Cell Line Exosomes Reveals Disease Patterns and Potential Biomarkers. Sci. Rep. 2020, 10, 13572. [Google Scholar] [CrossRef]
  153. Xu, G.; Huang, R.; Wumaier, R.; Lyu, J.; Huang, M.; Zhang, Y.; Chen, Q.; Liu, W.; Tao, M.; Li, J.; et al. Proteomic Profiling of Serum Extracellular Vesicles Identifies Diagnostic Signatures and Therapeutic Targets in Breast Cancer. Cancer Res. 2024, 84, 3267–3285. [Google Scholar] [CrossRef] [PubMed]
  154. Yu, L.; Liebenberg, K.; Shen, Y.; Liu, F.; Xu, Z.; Hao, X.; Wu, L.; Zhang, W.; Chan, H.L.; Wei, B.; et al. Tumor-Derived Arachidonic Acid Reprograms Neutrophils to Promote Immune Suppression and Therapy Resistance in Triple-Negative Breast Cancer. Immunity 2025, 58, 909–925.e7. [Google Scholar] [CrossRef] [PubMed]
  155. D’Mello, R.; Hüttmann, N.; Minic, Z.; Berezovski, M.V. Untargeted Metabolomic Profiling of Small Extracellular Vesicles Reveals Potential New Biomarkers for Triple Negative Breast Cancer. Metabolomics 2024, 20, 123. [Google Scholar] [CrossRef] [PubMed]
Figure 1. BC molecular subtypes and disease aggressiveness. BC is divided according to the presence or absence of specific receptor expression (Figure 1). Based on disease prognosis, we can rank the molecular subtypes from those with the best to those with the worst prognosis. Luminal A BC represents a molecular subtype characterized by the expression of ER and PR, the absence of HER2 receptor expression, low expression of Ki-67, and a relatively favorable disease prognosis. The second, more aggressive molecular subtype is Luminal B, characterized by the presence of ER and/or PR, and can be either HER2-positive or HER2-negative. It is also characterized by high Ki-67 expression. The third subtype is HER2-positive BC, characterized by the lack of progesterone and estrogen receptor expression and the presence of HER2 receptor expression. The molecular subtype with the worst disease prognosis is TNBC, which is characterized by the absence of estrogen, progesterone, and HER2 receptor expression.
Figure 1. BC molecular subtypes and disease aggressiveness. BC is divided according to the presence or absence of specific receptor expression (Figure 1). Based on disease prognosis, we can rank the molecular subtypes from those with the best to those with the worst prognosis. Luminal A BC represents a molecular subtype characterized by the expression of ER and PR, the absence of HER2 receptor expression, low expression of Ki-67, and a relatively favorable disease prognosis. The second, more aggressive molecular subtype is Luminal B, characterized by the presence of ER and/or PR, and can be either HER2-positive or HER2-negative. It is also characterized by high Ki-67 expression. The third subtype is HER2-positive BC, characterized by the lack of progesterone and estrogen receptor expression and the presence of HER2 receptor expression. The molecular subtype with the worst disease prognosis is TNBC, which is characterized by the absence of estrogen, progesterone, and HER2 receptor expression.
Biomolecules 15 01195 g001
Figure 2. Extracellular vesicle cargo, structure, and biogenesis. (1) EVs are particles formed from a lipid bilayer, characterized by various membrane-associated regulatory biomolecules and specific internalized cargo consisting of regulatory biomolecules. In the cell, EVs undergo a process of biogenesis, which begins with endocytosis (2), where various soluble biomolecules and EVs are internalized by the cell into the cytoplasm, leading to the formation of early endosomes. These then progress to form multivesicular endosomes (MVEs) consisting of intraluminal vesicles, where the packaging with various molecules occurs. The subsequent pathway continues either by the MVE content being tagged and fused with lysosomes, where degradation of unnecessary biomolecules takes place, or by the MVE content being secreted in the form of exosomes containing regulatory biomolecules through the process of exocytosis (3) into the extracellular space. Another method of EV genesis is ectocytosis (4), also known as membrane shedding, during which soluble bioactive molecules are packaged into the cell membrane and subsequently released into the extracellular space in the form of microvesicles or ectosomes.
Figure 2. Extracellular vesicle cargo, structure, and biogenesis. (1) EVs are particles formed from a lipid bilayer, characterized by various membrane-associated regulatory biomolecules and specific internalized cargo consisting of regulatory biomolecules. In the cell, EVs undergo a process of biogenesis, which begins with endocytosis (2), where various soluble biomolecules and EVs are internalized by the cell into the cytoplasm, leading to the formation of early endosomes. These then progress to form multivesicular endosomes (MVEs) consisting of intraluminal vesicles, where the packaging with various molecules occurs. The subsequent pathway continues either by the MVE content being tagged and fused with lysosomes, where degradation of unnecessary biomolecules takes place, or by the MVE content being secreted in the form of exosomes containing regulatory biomolecules through the process of exocytosis (3) into the extracellular space. Another method of EV genesis is ectocytosis (4), also known as membrane shedding, during which soluble bioactive molecules are packaged into the cell membrane and subsequently released into the extracellular space in the form of microvesicles or ectosomes.
Biomolecules 15 01195 g002
Figure 3. Simplified scheme of regulation and tumor progression via active regulatory biomolecules contained in EVs. (1) A breast lump represents a TNBC tumor, characterized by negative expression of PR, ER, and HER2+ receptors. (2) In the tumor cell, biogenesis of EVs occurs, involving the packaging of soluble regulatory factors directly into the cell membrane during membrane shedding (ectocytosis), resulting in ectosomes (microvesicles) that contain specific membrane molecules and cargo. (3) The biogenesis of EVs in cancer cells culminates in exocytosis, during which exosomes are secreted into the extracellular environment. (4) These exosomes are characterized by specific membrane molecules and contain cargo that includes regulatory molecules. Ectosomes, exosomes, and other EVs with cargo consisting of specific regulatory molecules such as miRNA, circRNA, and proteins are directed to their destinations, which may include normal cells. (5) Regulatory molecules target specific metabolic pathways and protein expression regulators in normal cells, thereby modulating metabolic pathways. (6) In this particular case, this modulation leads to cancer progression, EMT (7), and ultimately cell invasion (8), resulting in metastasis to distant organs (9).
Figure 3. Simplified scheme of regulation and tumor progression via active regulatory biomolecules contained in EVs. (1) A breast lump represents a TNBC tumor, characterized by negative expression of PR, ER, and HER2+ receptors. (2) In the tumor cell, biogenesis of EVs occurs, involving the packaging of soluble regulatory factors directly into the cell membrane during membrane shedding (ectocytosis), resulting in ectosomes (microvesicles) that contain specific membrane molecules and cargo. (3) The biogenesis of EVs in cancer cells culminates in exocytosis, during which exosomes are secreted into the extracellular environment. (4) These exosomes are characterized by specific membrane molecules and contain cargo that includes regulatory molecules. Ectosomes, exosomes, and other EVs with cargo consisting of specific regulatory molecules such as miRNA, circRNA, and proteins are directed to their destinations, which may include normal cells. (5) Regulatory molecules target specific metabolic pathways and protein expression regulators in normal cells, thereby modulating metabolic pathways. (6) In this particular case, this modulation leads to cancer progression, EMT (7), and ultimately cell invasion (8), resulting in metastasis to distant organs (9).
Biomolecules 15 01195 g003
Table 1. Overview of EV cargo in cancer.
Table 1. Overview of EV cargo in cancer.
EV Cargo TypeExamplesMechanisms/ImpactCancer Types
RNAlncRNA H19, miR-181a-5p, circEHD2, miR-29a-3p Regulation of metastasis, drug resistance, tumor-promoting inflammationLung, CRC, RCC, bladder
DNAdsDNA (KRAS, TP53), mtDNA, c-MycStable detection of mutations, tumor-specific DNA for liquid biopsyMelanoma, pancreatic, colorectal
ProteinsCDCP1, CD9, TGF-β co-receptors, TYRP2, HSP70 Modulation of angiogenesis, metastasis, immune evasionProstate, melanoma, breast, HNSCC
LipidsPhosphatidylserine, ceramide, glycosphingolipidsBiomarker potential, metabolic regulation, cancer-specific lipid signaturesProstate, RCC, lung
MetabolitesProline, adenosine, L-kynurenine, MG-H1, glyoxalase 1/2Immunosuppression, metabolic reprogramming, angiogenesisProstate, breast, ovarian, colon, HNSCC
Table 3. TNBC EV-associated DNA bioactive molecules.
Table 3. TNBC EV-associated DNA bioactive molecules.
EV CargoTNBCFunctionReferences
DNAmtDNAMutated mtDNA associated with early-stage cancer diagnostics and preventive care[145]
DNAmtDNA (RNR1, RNR2)Regulatory region mutations detected in 15% of patient samples[145]
DNAmtDNA (tRNA genes)Mutations specific to tRNA genes, impacting tumorigenesis and chemoresistance[145]
DNAmtDNA (G1888A mutation)Mutation detected in 6% of patient samples[145]
DNAmtDNA (C5720T mutation)Mutation present in 16% of patient samples[145]
DNAmtDNA (T4434G mutation)Mutation present in 20% of patient samples[145]
DNAmtDNA (Respiratory Complex I)Mutations in MT-ND1, ND2, ND3, ND4, ND4L, ND5, ND6 regions associated with mitochondrial respiratory chain[145]
DNAmtDNA (Respiratory Complex III)Mutations in MT-CYTB, specifically T14894G mutation accounting for 20%[145]
DNAmtDNA (Respiratory Complex IV)Mutations in MT-CO1, CO2, CO3 regions, with T7953G mutation exhibiting 38% prevalence[145]
DNAmtDNA (Respiratory Complex V)Mutations in MT-ATP6 and ATP8 subunits[145]
DNAmtND4 geneIncreased mtDNA levels mutated in mtND4 gene transported in EVs responsible for tumorigenesis and chemoresistance[146]
DNAHPV DNAPrevalence of HPV DNA in EVs linked to stromal cell activation and mammosphere formation[147]
Table 4. TNBC EV-associated protein bioactive molecules.
Table 4. TNBC EV-associated protein bioactive molecules.
EV CargoTNBCFunctionReferences
ProteinsEGFR, MMPsTumor-promoting proteins that enhance invasion and migration[148]
ProteinsGal3BP/Gal3 complexInduces immunosuppression via the CD45 receptor[149]
ProteinsAnnexin A2 Involved in TNBC metastasis[150]
ProteinsEGFR, ERBB2, MAPK1Promotes PI3K/Akt signaling pathway, metabolism, proliferation, cell survival, EMT activation[151]
ProteinsGlypican-1, glucose transporter-1, ADAM10Potential EV membrane/surface markers for TNBC[152]
ProteinsTLN2, VASP, GNASPlatelet activation-related proteins[153]
ProteinsB2M, PSMB9Antigen processing and presentation-related proteins[153]
ProteinsCFL2, ITGB4, GIT1Regulation of actin cytoskeleton-related proteins[153]
ProteinsCEACAM1, COL4A2, CALD1Angiogenesis-related proteins[153]
ProteinsDST, ITGB4, CFL2Cell motility-related proteins[153]
Table 5. TNBC EV-associated lipid bioactive molecules.
Table 5. TNBC EV-associated lipid bioactive molecules.
EV CargoTNBCFunction SpecificationReferences
LipidsCardiolipinElevated levels associated with TNBC patient samples[145]
LipidsArachidonic acidReprograms neutrophils, leading to lipid droplet accumulation and fostering an immunosuppressive tumor microenvironment[155]
LipidsLysoPC 22:6/0:0Proposed as a new biomarker for TNBC diagnostics[155]
Table 6. Luminal A-associated protein bioactive molecules.
Table 6. Luminal A-associated protein bioactive molecules.
EV CargoLuminal AFunctionReferences
ProteinsSDCBP, COLEC11, LTFProteolysis-related proteins[153]
ProteinsCCT2, HSP90AA1, PPIGProtein folding-related proteins[153]
ProteinsHSP90AA1, SDCBPRegulation of necroptotic cell death[153]
ProteinsHSBP1, IL6, HSP90AA1Cellular stress response and responses to external stimuli[153]
Table 7. Luminal B-associated protein bioactive molecules.
Table 7. Luminal B-associated protein bioactive molecules.
EV CargoLuminal BFunctionReferences
ProteinsMYL6, AFDNTight junction-associated proteins[153]
ProteinsRELN, GP1BBECM–receptor interaction-related proteins[153]
ProteinsSLC2A14, SLC2A3Fructose and mannose metabolism-related proteins[153]
ProteinsSLC2A14, SLC2A3Glucose metabolism-related proteins[153]
ProteinsPKLR, PYGMInsulin signaling pathway-associated proteins[153]
Table 8. HER-2+-associated protein bioactive molecules.
Table 8. HER-2+-associated protein bioactive molecules.
EV CargoHER-2+FunctionReferences
ProteinsSRC, ARG1, MAPK13Cellular response to hydrogen peroxide [153]
ProteinsDOCK7, SRCErbB2/ErbB3 signaling event-related proteins[153]
ProteinsCDSN, KRT78, KRT23Keratinization-related proteins[153]
ProteinsFAH, ADHFE1Tyrosine metabolism-related proteins[153]
ProteinsARG1, RARS1Arginine and proline metabolism-related proteins[153]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Bernatova, S.; Nicodemou, A.; Cehakova, M.; Danisovic, L.; Bohac, M. Extracellular Vesicles Derived from Breast Cancer Cells: Emerging Biomarkers of Tumor Progression and Metastasis. Biomolecules 2025, 15, 1195. https://doi.org/10.3390/biom15081195

AMA Style

Bernatova S, Nicodemou A, Cehakova M, Danisovic L, Bohac M. Extracellular Vesicles Derived from Breast Cancer Cells: Emerging Biomarkers of Tumor Progression and Metastasis. Biomolecules. 2025; 15(8):1195. https://doi.org/10.3390/biom15081195

Chicago/Turabian Style

Bernatova, Sona, Andreas Nicodemou, Michaela Cehakova, Lubos Danisovic, and Martin Bohac. 2025. "Extracellular Vesicles Derived from Breast Cancer Cells: Emerging Biomarkers of Tumor Progression and Metastasis" Biomolecules 15, no. 8: 1195. https://doi.org/10.3390/biom15081195

APA Style

Bernatova, S., Nicodemou, A., Cehakova, M., Danisovic, L., & Bohac, M. (2025). Extracellular Vesicles Derived from Breast Cancer Cells: Emerging Biomarkers of Tumor Progression and Metastasis. Biomolecules, 15(8), 1195. https://doi.org/10.3390/biom15081195

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

Article Metrics

Back to TopTop