Next Article in Journal
Genome-Wide DNA Methylation Profiling as Frontline Diagnostics for Central Nervous System Embryonal Tumors in Hong Kong
Previous Article in Journal
The Relationship between Furin and Chronic Inflammation in the Progression of Cervical Intraepithelial Neoplasia to Cancer: A Cross-Sectional Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Extracellular Vesicles in Triple–Negative Breast Cancer: Immune Regulation, Biomarkers, and Immunotherapeutic Potential

1
Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, TX 75708, USA
2
School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700012, India
3
Department of Biotechnology, GLA University, Mathura 281406, India
4
School of Medicine, The University of Texas at Tyler Health Science Center, Tyler, TX 75708, USA
5
Department of Neurobiology, The University of Texas Medical Branch, Galveston, TX 77555, USA
6
Division of Hematology & Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
7
Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
8
South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2023, 15(19), 4879; https://doi.org/10.3390/cancers15194879
Submission received: 5 August 2023 / Revised: 25 September 2023 / Accepted: 28 September 2023 / Published: 7 October 2023
(This article belongs to the Special Issue Emerging Trends in Immunotherapy for Triple Negative Breast Cancer)

Abstract

:

Simple Summary

Triple–negative breast cancer (TNBC), an aggressive phenotype, is commonly attributed to the loss of estrogen receptor, progesterone receptor, and human epidermal growth factor receptor, thereby posing unique challenges in treatment via conventional targeted therapies. Although present chemotherapeutic regimens show promise against TNBC morbidity; the variability in treatment outcomes among patients and emerging resistance limit their potential. Moreover, due to significant mutational burden, TNBC is considered as highly immunogenic. Extracellular vesicles (EVs) have shown to regulate multiple human pathologies including cancer due to their role in transferring bioactive molecules between cells, evading immune surveillance. Conversely, the biological novelties of EVs are also exploited to develop experimental therapies. Here, we review studies in which EVs contribute to the progression of human TNBC and their immune regulation. Understanding these mechanistic details may open up avenues for repurposing EV–based immunotherapeutic strategies in the context of human TNBC treatment.

Abstract

Triple–negative breast cancer (TNBC) is an aggressive subtype accounting for ~10–20% of all human BC and is characterized by the absence of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) amplification. Owing to its unique molecular profile and limited targeted therapies, TNBC treatment poses significant challenges. Unlike other BC subtypes, TNBC lacks specific molecular targets, rendering endocrine therapies and HER2–targeted treatments ineffective. The chemotherapeutic regimen is the predominant systemic treatment modality for TNBC in current clinical practice. However, the efficacy of chemotherapy in TNBC is variable, with response rates varying between a wide range of patients, and the emerging resistance further adds to the difficulties. Furthermore, TNBC exhibits a higher mutational burden and is acknowledged as the most immunogenic of all BC subtypes. Consequently, the application of immune checkpoint inhibition has been investigated in TNBC, yielding promising outcomes. Recent evidence identified extracellular vesicles (EVs) as an important contributor in the context of TNBC immunotherapy. In view of the extraordinary ability of EVs to transfer bioactive molecules, such as proteins, lipids, DNA, mRNAs, and small miRNAs, between the cells, EVs are considered a promising diagnostic biomarker and novel drug delivery system among the prospects for immunotherapy. The present review provides an in–depth understanding of how EVs influence TNBC progression, its immune regulation, and their contribution as a predictive biomarker for TNBC. The final part of the review focuses on the recent key advances in immunotherapeutic strategies for better understanding the complex interplay between EVs and the immune system in TNBC and further developing EV–based targeted immunotherapies.

1. Introduction

1.1. Extracellular Vesicles: More Than Cellular Dust

Extracellular vesicles (EVs) were first identified in 1946 by Chargaff and West as platelet–derived procoagulant particles in plasma [1]. More than two decades later, Wolf renamed these procoagulant plasma–derived bodies “platelet dust” [2]. A further decade passed before the intriguing discovery of Harding et al. [3] that plasma particles serve as an important mode of cell–cell communication via autocrine, paracrine, or endocrine mechanisms via the transfer of bioactive molecules [4]. In the late 2000s, the advancement of EV research further established the biological implications of EVs [5,6]. In the last decade, EVs have come up as an important contributor in various pathophysiological conditions [7,8,9,10,11,12,13,14], basically via passing on bioactive molecules in the form of DNA, RNAs, miRNAs, proteins, lipids, etc., between the cells [15].
EVs are membrane–enclosed nanoparticles that are released from almost every cell type into the extracellular milieu [4,16]. They are abundantly found in body fluids like blood, saliva, urine, and amniotic fluid, and their presence is detectable within the interstitial spaces between the cells [17,18,19]. Sometimes, the number and composition of EVs are significantly altered in various disease conditions, often rendering them novel biomarkers for different pathophysiological settings [20,21]. However, depending on their biogenesis, release mechanisms, and functions, the EVs are broadly classified into microparticles, exosomes, and apoptotic bodies.

1.2. Microparticles

Microparticles (MPs), microvesicles (MVs), or ectosomes are produced by the external budding of the plasma membrane [14]. Initially, the loss of membrane asymmetry results in the externalization of phosphatidylserine (PS), which usually resides in the inner leaflet of the plasma membrane, to the outer leaflet and, hence, it is enriched on the MPs’ surfaces. This is followed by the reorganization of cytoskeletal components to induce membrane curvature, ultimately leading to the liberation of the MPs outside the cells [22,23,24]. Therefore, cytosolic and membrane–associated proteins, such as tetraspanins, which are the proteins clustered at the plasma membrane, are enriched into the MPs, which often serve as an MP marker, regardless of the originating cells [25,26]. However, cytoskeleton–associated proteins, like heat shock proteins (HSPs), integrins, and proteins associated with post–translational modifications (phosphorylation, glycosylation, etc.), are also abundantly found in MPs [27,28,29]. MPs appear to be larger at ~100–1000 nm in diameter [14,19].

1.3. Exosomes

Exosomes, on the other hand, are 30–150 nm in diameter [30] and are basically of endocytic origin [14]. Additionally, exosomes are nanovesicles that facilitate cell–to–cell communication, altering the tumor microenvironment through paracrine signaling [31]. Exosomes play an important role in promoting the EMT phenotype and increasing chemoresistance [32]. Conventionally, early endosomal membranes invaginate to produce such exosomes, which mature into multivesicular bodies (MVBs) [14]. Further, these MVBs fuse with the plasma membrane to shed the exosomes outside the cell [14]. The endosomal sorting complexes required for the transport (ESCRT) pathway play a major role in the biogenesis of exosomes [14], thereby ESCRT proteins, including TSG101, Alix, HSP90β, and HSC70, are abundantly found in the exosomes [33,34], which often serve as exosomal markers. In contrast, exosome biogenesis also involves ESCRT–independent mechanisms, which were shown to be mediated by the sphingolipid ceramide [35].

1.4. Autophagic EVs

An increasing body of evidence identified an unconventional mechanism, namely, “autophagy”, which is the lysosomal mechanism of bulk degradation of the dysfunctional and unusable components of the cells [36] that helps cellular metabolism and homeostasis. Autophagy is induced by different factors, such as nutrient starvation and the accumulation of old or damaged materials, such as proteins, lipids, and organelles, and plays an important role in tumor progression or suppression. Autophagy can regulate the biogenesis of the exosomes. During autophagy, the cytoplasmic portion is sequestered by an organelle, namely, the phagophore, resulting in the formation of “autophagosomes” that eventually fuse with the MVBs to form “amphisomes” [37,38]. Therefore, both the endosome, as well as autophagosome markers, for example, LC3 and CD63, are found in amphisomes and also found to be enriched with nucleosomes and cytosolic DNA. During starvation, these endosomes can be recycled with the assistance of Rab11, Rab27, and Rab35 [39].
There are 10 members in the Rab GTPases family that facilitate the autophagic process, with the majority functioning at the autophagosome level [40]. The Rab GTPases maintain intracellular trafficking, regulating the traffic between organelles by recruiting effector proteins. Therefore, Rab GTPases are considered the “master regulators” of intracellular trafficking [41]. During early apoptosis in cells, cells secrete apoptotic exosome–like vesicles (AEVs). Moreover, AEVs biogenesis is proteasome–dependent, while AEVs secrete in a caspase–3–dependent manner. The major difference between apoptotic bodies and AEVs is that AEVs lack apoptotic markers, such as GM130, calreticulin, and tubulin [42]. When the autophagosome fuses with lysosomes, which is facilitated by Rab7, this results in the degradation of their contents. As a result, the cell receives nutrients and energy. Second, amphisomes can also fuse with the plasma membrane, leading to the release of exosomes outside of the cell via exophagy [41]. Both MPs and exosomes play crucial roles, not only in maintaining homeostasis but also in the progression and development of various pathophysiological conditions, for example, cancer, due to the transfer of bioactive cargo [43]. MPs and exosomes are readily taken up by the recipient cells, either via direct fusion with the plasma membrane or an endocytic mechanism; in either case, the vesicles’ contents are released into the cytosol of the recipient cells [44,45,46,47]. Figure 1 briefly summarizes the mechanism of autophagic EVs’ biogenesis and the impact on the tumor microenvironment.

1.5. Apoptotic Bodies (ApoBDs)

ApoBDs are membrane blebs that are fairly variable in size and diameter, ranging from 50 nm to 5 µm, and are produced by the cells undergoing apoptosis [48]. The induction of apoptosis produces a significant hydrostatic pressure, which is generated during the contraction of the cells, thereby segregating the plasma membrane from the cytoskeleton, as a result of which the apoptotic bodies are released [49]. However, unlike MPs and exosomes, apoptotic bodies carry cellular organelles, chromatin, and a few glycosylated proteins [48,50,51,52]. Consequently, markers for cellular organelles, such as HSP60 for mitochondria, GRP78 for Golgi and endoplasmic reticulum, and histones for nuclear markers, are in the apoptotic bodies [49]. Additionally, these ApoBDs have external features (“Eat–me” signals, as well as nuclear autoantigens) that induce phagocytosis [48,50,51,52] as schematically depicted in Figure 2.
Table 1 summarizes the size distribution, markers, and biogenetic mechanisms of different forms of EVs.

2. EVs and Diseases

Increasing evidence indicates that EVs, due to the transfer of heterogeneous cargos between the cells, often influence various pathophysiological conditions. For example, in the case of a urinary tract infection (UTI), mediated by Gram–negative bacteria–induced sepsis, higher expression of procoagulant tissue factor (TF) is often observed on the plasma EVs, which reflects the hypercoagulative states in UTI patients [60]. On the other hand, activated platelet–derived EVs are shown to develop anticoagulant states [61]. In atherosclerosis, negatively charged phospholipids, such as phosphatidylserine (PS), allow for the uptake of intercellular adhesion molecule 1 (ICAM–1)–bearing plaque–derived EVs by the endothelial cells, thereby facilitating the recruitment of pro–inflammatory cells, leading to plaque progression [62]. Furthermore, the expression of fetuin–A is shown to be significantly upregulated, whereas aquaporin 1 (AQP1) is downregulated in the urinary EVs of acute kidney injury (AKI) patients, and hence, fetuin–A+ and AQP1+ EVs are considered biomarkers for AKI [63]. The upregulation of ten signature miRNA molecules (miR–199a–5p, miR–4745–3p, miR–143–3p, miR–4532, miR–193b–3p, miR–199b–3p, miR–25–3p, miR–199a–3p, miR–629–5p, and miR–6087), as well as the downregulation of another ten (miR–23b–3p, miR–141–3p, miR–10a–5p, miR–200c–3p, miR–98–5p, miR–382–5p, miR–200a–3p, miR–483–3p, miR–483–5p, and miR–3911) are often seen in the human follicular fluid (HFF)–derived EVs of PCOS biomarkers [64]. α–synuclein +EVs are shown to be enriched in the cerebrospinal fluid (CSF) of Parkinson’s disease (PD) patients and contribute to PD progression via facilitating α–synuclein aggregation in healthy cells [65]. In congenital myopathies (CMs), EVs from the differentiating fibroblasts were shown to be released into circulation, which enhances the regeneration of muscles; therefore, an elevated level of circulating EVs is considered a diagnostic biomarker of CM [66]. Moreover, it was observed that with respect to healthy controls, the composition of microbial EVs in the blood, urine, and feces of patients with gastrointestinal tract disease is significantly altered, which also serves as a diagnostic and therapeutic biomarker for the progression of the disease [67]. On numerous occasions, EVs were shown to be involved in the development and progression of various types of cancer. For example, miR–144 expression in the EVs of nasopharyngeal carcinoma (NPC) is significantly upregulated, which promotes the migration, invasion, and angiogenesis of endothelial cells [68]. Another study indicated that hypoxic–lung–cancer–cell–derived EVs are enriched with miR–103a, which targets PTEN in macrophages, leading to M2 polarization via the activation of AKT and STAT3, which further contributes to the release of immunosuppressive and pro–angiogenic factors to promote lung cancer progression [69]. Moreover, hepatocellular carcinoma (HCC)–derived EVs are abundant with miR–3129, which promotes the growth and metastasis of HCC via targeting TXNIP [70]. Furthermore, human colorectal cancer (CRC)–derived EVs facilitate tumor progression and metastasis via miR–25–mediated downregulation of SIRT6 [71]. The following section delineates how EVs contribute to the pathogenesis of human breast cancer, specifically human TNBC, in different ways. This is followed by a brief discussion on how EVs contribute as biomarkers for TNBC. The final part of the review discusses how EVs act as an immunotherapeutic mediator in the treatment of human TNBC.

3. Breast Cancer and Its Subtypes

Breast cancer is known as a pathological condition in which the malignant cells in the breast start growing out of control [72,73,74]. Breast cancer is considered the second–leading cause of cancer–related death among women worldwide [75], and in 2022, ~287,850 new cases of invasive breast cancer were predicted to arise in the United States, with a predictable death rate of 43,250 [75].

3.1. Breast Cancer Subtypes

However, the modern classification of breast cancer on the basis of primary markers (ER, PR, and HER2) [76,77], the Ki–67 proliferation index [78], and basal markers (EGFR, CK5/6) [79] differentiates breast cancer into luminal A, luminal B, HER2 high, normal–like, basal–like, and claudin low. The luminal subtype of breast tumor involves the inner (luminal) breast epithelial cells [80], which was first discovered in 2000 using ER+ immunohistochemical profiling [81]. Depending on HER2 expression, the luminal subtype is further classified into luminal A and luminal B, luminal A is shown to be HER2−, whereas luminal B is HER2+ [82]. Moreover, luminal B is characterized by a worse prognosis and propagates at a faster rate than luminal A [82]. In contrast with luminal subtypes, the HER2 high subtype is ER− and PR− but HER2+ and is characterized by a higher growth rate with a worse prognosis [83]. Interestingly, the HER2 high breast cancer subtype is shown to be highly sensitive to the chemotherapeutic drug trastuzumab [84]. The normal breast cancer subtype is characterized by the loss of expression of the proliferation gene, which is carried by a low percentage of cancer cells [80]. The basal–like subtype is ER, PR, and HER2 negative (HER2−), and is so named because of the similarity with cytokeratins (CKs) 5/6, and EGFR expression [81,85,86,87,88]. The basal–like breast cancer subtype is also known as TNBC, which was shown to be associated with a mutation in the breast cancer gene 1 (BRCA1) [89]. However, recent evidence indicates that not all TNBC can be considered basal types. Prat et al. identified ~70–80% of all TNBC as basal type [90]. Due to the deficiencies of ER, PR, and HER2 expressions, TNBC cells often show resistance against various hormones and targeted therapy and, therefore, can only be treated with cytotoxic drugs [91]. The claudin–low subtype, sometimes also referred to as TNBC, is characterized by lower expressions of E–cadherin, mucin1, EpCAM, and claudins (3, 4, 7) and limited expression of Ki–67 and luminal markers compared with the other breast cancer subtypes [90]. In contrast, an elevated expression of genes associated with the epithelial–to–mesenchymal transition (EMT), such as vav1, CD79b, and CD14, and cancer stemness are often observed in the claudin–low subtype [92,93,94]. Table 2 briefly illustrates the immunohistochemistry–based subtyping of various breast cancers.

3.2. Subtypes of TNBC and Their Treatment Measures

Based on the gene expression profile [95], TNBC is a type of breast cancer that has a lot of different kinds of cells. It can be further broken down into basal–like 1 (BL–1), basal–like 2 (BL–2), immunomodulatory (IM), luminal androgen receptor (LAR), mesenchymal (M), and mesenchymal stem–like (MSL). The BL–1 and BL–2 subtypes have similar expression profiles of cell cycle and cell division–associated genes.
However, BL–1 exerts a relatively higher expression of DNA replication and repair–associated genes, whereas BL–2 shows an elevated expression of growth–factor–signaling genes [95]. The treatment measures for BL–1 include PARP inhibitors [96] and platinum compounds [97,98], whereas the inhibition of growth signaling was shown to be effective against BL–2 [99]. In contrast, the IM subtype often demonstrates enhanced expression of immune–response–related genes, such as genes associated with the NK–cell pathway, Th1/Th2 pathway, B–cell receptor (BCR), antigen processing, and cytokine signaling; therefore, immune checkpoint inhibition is often found to be an effective measure against IM [100]. The LAR subtype, on the other hand, shows an over–expression of genes related to androgen receptors, as well as several hormonally regulated pathways [95,101]. The androgen receptor agonist bicalutamide was shown to be effective in treating the LAR subtype [102]. As the names suggest, both M and MSL are associated with the over–expression of genes related to cell motility, extracellular receptor interaction, and cellular differentiation pathways. However, subtle differences exist, for example, the expression of claudins (3, 4, 7) was shown to be significantly lower in MSL [95], which is often considered claudin–low breast cancer [103]. The treatment measures for the M subtype include targeting the Wnt/β–catenin pathway [104], PI3K/mTOR pathway [95,105], and TGFβ receptor kinase [106]. Figure 3 summarizes the different subtypes of TNBC, their general characteristics, and treatment measures.

4. The Role of EVs in the Progression of TNBC

Numerous studies identified the active participation of EVs in the progression of human TNBC; in some instances, EVs are associated with the growth of TNBC, whereas in other cases, they influence the metastatic dissemination of TNBC. Table 3 briefly discusses how EVs contribute to the growth and metastasis of TNBC in different ways.

4.1. EVs in TNBC Growth

Due to the extraordinary capability of the EVs to transfer bioactive molecules between the cells, EVs are often shown to promote TNBC growth, thereby contributing to the progression of TNBC. For example, EVs from HCC1806 TNBC cells induce the proliferation of and drug resistance in the normal breast epithelial cells MCF10A by involving PI3K/AKT, MAPK, and HIF1α pathways [107]. Moreover, plant–derived sesquiterpene lactone deoxyelephantopin (DET) and its derivative, namely, DET derivative 35 (DETD–35), were shown to evoke the release of EVs from the human TNBC cells MDA–MB–231, which, in turn, inhibit the proliferation of MDA–MB–231 cells via the down–regulation of cell adhesion, migration, and angiogenesis [108]. Cancer–associated fibroblasts (CAFs) in the tumor microenvironment (TME) often trigger the growth of tumor cells [14]. A recent study showed that integrin β4 (ITGB4) + EVs that are released from MDA–MB–231 cells promote CAFs’ growth by triggering the glycolytic pathway via BNI3PL–dependent mitophagy and lactate production, which, in turn, induce the growth of the TNBC tumor [109]. Furthermore, mesenchymal stem cell (MSC)–derived EVs, via the transfer of miR–106a–5p, promote TNBC tumor progression upon downregulating the expression of HAND2–AS1 [110]. In contrast, the expression of miR–4516 is shown to be downregulated in CAFs compared with normal fibroblasts and CAF–derived EVs often promote the proliferation of TNBC cells via upregulating the expression of the miR–4516 target FOSL1 [111]. Circular RNA HIF1A (circHIF1A) is enriched in the plasma EVs of TNBC patients and was shown to promote tumor growth via the activation of the PI3K/AKT pathway and downregulation of p21 [112]. Figure 4 illustrates how EVs play their part in the growth of TNBC.

4.2. EVs in TNBC Metastasis

EVs also participate in the metastatic dissemination of human TNBC. For example, in response to chemotherapy, TNBC cells release a significant number of PS + EVs, which trigger an increase in vascular permeability, leading to the transendothelial migration (TEM) of cancer cells [113]. Moreover, the serum EVs of TNBC patients are enriched with tetraspanins, including CD151, which was shown to stimulate the migration and invasion of TNBC cells [114]. Again, circulating EVs of TNBC patients are observed to be abundant with the cancer testis antigen SPANXB1, which downregulates the SH3GL2 expression in TNBC cells, thereby upregulating the expression of Rac1, FAK, and α–actinin, leading to TNBC metastasis [115]. Furthermore, the level of miR–9 and miR–155 is well–elevated in the TNBC cells MDA–MB–231–derived EVs, which target PTEN and DUSP14 in less metastatic cells, namely, MCF–7, leading to the enhancement of MCF–7 metastatic potential [116]. In addition, EVs from TNBC cells often show an upregulation of miR–939, which targets VE–cadherin in endothelial cells, thereby triggering increased vascular leakage to promote transendothelial migration of the tumor [117]. Serum EVs of TNBC patients are overexpressed with circPSMA1, which acts as a “miRNA sponge” to absorb miR–637, thereby releasing the inhibitory effect of miR–637 on AKT1 to stimulate β–catenin–driven expression of cyclin D1, ultimately leading to the enhanced proliferation, migration, and metastasis of TNBC cells [118]. Figure 4 also delineates how EVs contribute to the metastasis of human TNBC.

5. Blood Coagulation Is Associated with TNBC Progression

Blood coagulation and cancer are intrinsically related; enhanced thrombosis is often observed in various types of cancer, which contributes to the morbidity and mortality of cancer patients [119]. On the other hand, our studies, for the first time, found that coagulation proteases trigger the progression of human TNBC via the secretion of EVs. We demonstrated that coagulation protease factor VIIa (FVIIa) binds to its primary receptors, namely, TF and TF–FVIIa, via the activation of protease–activated receptor 2 (PAR2) [120], which stimulates the release of EVs from the human TNBC cells MDA–MB–231 [18]. The study indicates that TF–FVIIa–PAR2–mediated generation of EVs is dependent on three independent signaling pathways: the PI3K/AKT/p38–MAPK/MK2/HSP27 pathway, MEK1/2/ERK1/2/MLCK/MLC2 pathway, and ROCKII pathway [18]. The crosstalk of these three pathways promotes actomyosin reorganization, which critically regulates the release of EVs from the TNBC cells [18]. The study also revealed that TNBC–EVs are readily taken up by the non–metastatic breast cancer cells MCF–7, thereby triggering the induction of metastatic potential to EVs–fused recipient MCF–7 cells [18]. In a separate study, we investigated that another protease, namely, trypsin, which is well–known for the activation of PAR2, also promotes the generation of EVs from human TNBC cells via AKT/Rab5–dependent actin rearrangements and these EVs induce metastasis to MCF–7 cells [17]. In a continuation study, we revealed that FVIIa–EVs from TNBC cells are enriched with miR–221, which targets PTEN in the recipient MCF–7 cells, leading to the activation of AKT/NF–ĸB pathway to promote the expression of the EMT–associated transcription factors snail and slug [11]. EMT–transcription factors, in turn, trigger the induction of the mesenchymal markers N–cadherin and vimentin, while prohibiting the expression of the epithelial marker E–cadherin, ultimately leading to EMT [11]. The induction of EMT not only enhances the proliferation, migration, and invasion of EVs–fused recipient MCF–7 but also confers resistance of MCF–7 against the chemotherapeutic apoptosis–inducing drug cisplatin [11]. The incorporation of an anti–miR–221 inhibitor into EVs not only reverses the EV–triggered induction of EMT in MCF–7 but also downregulates EV–mediated MCF–7 proliferation, migration, invasion, and drug resistance [11]. Consistent with our in vitro observations, our in vivo data also show that FVIIa–EVs from TNBC cells promote the growth and metastasis of MCF–7 tumors, and the introduction of anti–miR–221 significantly attenuates EV–induced tumor growth and metastatic dissemination [11]. These studies were further strengthened by our human patients’ data, which indicate that compared with normal healthy controls, EVs from TNBC patients’ plasma also stimulate the miR–221–dependent growth and metastasis of non–metastatic cells in both in vitro and in vivo settings [11]. Figure 5 summarizes how EVs from FVIIa–treated TNBC cells promote the growth and metastasis of non–metastatic breast cancer cells, depending on the miR–221 transfer.

6. Role of EVs as a Biomarker for TNBC

As EVs often bear the cargo molecules of the cells from which they arise, they often serve as effective biomarkers in various pathophysiological conditions, including TNBC. The present section briefly summarizes the role of EVs as a biomarker in different types of TNBC (Table 4). Figure 6 also briefly delineates how EVs serve as a biomarker for TNBC in different ways.

6.1. EVs’ Proteins as a TNBC Biomarker

EVs from the TNBC cells MDA–MB–231 were shown to be enriched with extracellular growth factor receptor (EGFR) ligands, such as amphiregulin (AREG), which promote the invasiveness of recipient breast cancer cells, thereby being considered a biomarker for TNBC [121]. The expression of the deubiquitinase UCHL1 is significantly higher in EVs derived from TNBC cells and plasma, which stimulates the migration and extravasation of the breast cancer cells by upregulating TGFβ signaling via protecting both TGFβ type I receptors and SMAD2 from ubiquitination [122]. Moreover, the chemotherapeutic drug paclitaxel (PTX) induces the release of Survivin + EVs from MDA–MB–231 cells, which promote the growth and survivability of PTX–exposed fibroblasts and other breast cancer cells, which essentially serve as a TNBC biomarker [123]. In addition, serum EVs of TNBC patients are enriched with CD151, which was shown to induce the migration and invasion of TNBC cells and is thereby considered a predictive biomarker for TNBC [114].

6.2. EVs’ miRNAs as a Biomarker for TNBC

miRNAs are small (~22 nt long) non–coding RNA molecules that play a major role in tumor progression including TNBC [128]. Moreover, these miRNAs, which are carried by the EVs, serve as a prognostic and diagnostic biomarker for TNBC. For example, cisplatin (DDP)–resistant MDA–MB–231–cell–derived EVs transfer miR–423–5p to other breast cancer cells, leading to the induction of P–glycoprotein (P–gp), enhancement of migration and invasion, and downregulation of apoptosis [124]. Moreover, TNBC–associated macrophages release miR–223 + EVs, which promote the invasion of breast cancer via the Mef2c–β–catenin pathway, and thus, miR–223–loaded EVs are considered a prognostic and diagnostic biomarker for TNBC [125]. In addition, TNBC–EVs often show a higher expression of miR–10b, which induces the invasiveness of non–malignant cells by targeting HOXD10 and KLF4, and miR–10b–enriched TNBC–EVs often serve as a predictive biomarker for TNBC [126]. Furthermore, the EV–mediated transfer of miR–105 from the TNBC cells MDA–MB–231 to endothelial cells induces endothelial barrier permeability via targeting the expression of the tight junction protein ZO–1, thereby facilitating TNBC metastasis [127]. Hence, miR–105–containing TNBC–EVs are often considered a biomarker for early–stage TNBC.

7. Emerging Role of EVs in TNBC Immune Regulation

EVs from TNBC cells are often shown to actively participate in the immune regulation of TNBC. However, EVs released from different cells of the TME, such as stromal cells and activated immune cells, also contribute to TNBC immune regulation. Table 5 briefly summarizes EVs’ contribution to immune regulation in the context of TNBC. For example, TNBC–EVs are enriched with CCL5, which is readily transferred to naive macrophages, leading to their transformation into tumor EV–educated macrophages (TEMs) [129]. TEMs, in turn, release various factors, including IFNγ, CXCL1, OPN, CTLA–4, TGFβ, HGF, and CCL19, thereby inducing stromal remodeling and immune infiltration to facilitate TNBC metastasis [129]. Moreover, TNBC–EVs are also reported to induce macrophage polarization into type 2 (M2) phenotypes, thereby creating a favorable environment for the lymph node (LN) metastasis of TNBC [130]. Again, activated T–cells that express programmed death 1 (PD–1) often release PD–1+ EVs that interact with programmed death–ligand 1 (PD–L1)–containing TNBC cells or TNBC–EVs, thereby preventing direct interaction between T–cells/TNBC cells via PD–1/PD–L1, ultimately attenuating the PD–L1–triggered suppression of activated T–cells by TNBC cells [131]. On the other hand, oscillatory strain (OS) enhances the release of TNBC–EVs that are positive for PD–L1, which not only promotes the enrichment of myeloid–derived suppressor cells (MDSCs) and M2 macrophages in the TME but also reduces the abundance of CD8+ T–cells [132]. Figure 7 depicts how EVs are associated with the immune regulation of TNBC.

8. Emerging Role of EVs in TNBC Immunotherapy

Immunotherapy is the major segment of cancer therapies that involves the immune cells of the body fighting against the cancer cells [133]. The advantages of cancer immunotherapy over other therapeutic approaches include fewer side effects, long–term protection, targeting each mutation in the cells, and being potentially effective against every type of cancer [134]. Although immunotherapy has made significant promises against different cancer types, its role in the treatment of human TNBC is still in the initial stages. However, a few successful implementations of immunotherapeutic approaches against human NBC are ongoing.

8.1. Immunotherapeutic Approaches against Human TNBC

Although chimeric antigen receptor T–cell (CAR–T) therapy has become more promising against different types of hematological tumors, its application against TNBC has been far more challenging [135]. On numerous occasions, the PD–1/PD–L1 mechanism was shown to be an effective immunotherapeutic target against TNBC. Pembrolizumab, which is the FDA–approved immune checkpoint inhibitor, and monoclonal antibody, which targets PD–1, were in a phase I clinical trial against TNBC [100]. Atezolizumab, which is an anti–PD–L1 antibody, was also shown to be tolerated with durable clinical benefits against metastatic TNBC [136]. However, in the phase III clinical trial IMpassion131, atezolizumab did not pass through due to its limited clinical benefits [137]. Another FDA–approved immunotherapeutic agent, namely, sacituzumab govitecan (IMMU–132), which is a tumor–associated calcium signal transducer 2 (TROP2) antibody, in conjugation with SN–38, which is the topoisomerase/inhibitor chemotherapy, showed promising effects in phase I and II clinical trials against metastatic TNBC and is now in clinical trial III [138,139]. At present, thirteen immunotherapy–based approaches are registered against TNBC in clinical trials, and among them, ten are in phase I. In addition, fifty–five clinical trials have been registered against TNBC that involve different chemotherapeutic regimens, in combination with immunotherapy [140].

8.2. EVs in TNBC Immunotherapy

Cytotoxic immune cells like natural killer (NK) cells and CD8+ cytotoxic T–lymphocytes (CTLs) have an amazing ability to find and kill cancer cells [141]. This has been used to make immunotherapies for TNBC [142]. However, due to their low penetrance into the tumor to reach the cancer cells, as well as their restricted efficacy, these mechanisms have limited therapeutic applications [142,143]. The advent of EVs, which can penetrate even solid tumors, has led researchers to employ EVs derived from NK–cells or CTLs in the treatment of different TNBC types. TNBC tumors often express PD–L1 on the cell surface, whose receptor PD–1 is expressed on tumor–infiltrating lymphocytes (TILs). The interaction of tumor cells with TILs via PD–L1/PD–1 not only attenuates TIL proliferation but also leads to the apoptosis of TILs, thereby contributing to the immune evasion mechanisms of the TNBC [142]. Qiu et al. demonstrated that PD–1+ EVs are released from TILs, which interact with either the cell surface or EVs’ PD–L1, thereby preventing the TILs–TNBC cells interaction through PD–L1/PD–1, ultimately leading to the attenuation of the PD–L1–mediated suppression of TILs activity [131]. This mechanism opens a new immunotherapeutic strategy to enhance TIL activity, thereby facilitating the killing of TNBC cells (Figure 7).
Unlike CTLs, NK–cells, independent of major histocompatibility complex I (MHC–I) recognition, exert cytolytic activity via the downregulation of tumor growth and metastasis [143]. Similar to NK–cells, NK–EVs were also shown to be positive for killer proteins like FasL, perforin, and granzyme [144], and elicit cytotoxicity against solid tumors [145], via the transfer of killer proteins (Figure 8A) [146]. Although, so far, NK–EVs have been shown to be effective against various solid tumors, their effects still need to be validated in the context of TNBC.
On several occasions, EVs carrying bioactive molecules, such as miRNAs, were shown to be effective therapeutic targets in the treatment of TNBC [147]. For example, let–7, encapsulated within aptamer–AS1411–modified EVs, is readily taken up by nucleolin–positive TNBC cells, leading to a significant reduction in tumor growth [147]. Again, bone marrow stromal EVs enriched with signature miRNA molecules (miR–222, miR–197, miR–127, and miR–223) often reduce the proliferation of TNBC cells [148]. Furthermore, adipose–tissue–derived MSCs promote the apoptosis of TNBC cells via the transfer of miR–424–5p–enriched EVs [149]. In addition, MSC–EVs also bear miR–381, which decreases the viability, migration, and invasion of TNBC cells via the downregulation of the Wnt pathway, expression of Twist and Snail, and subsequent inhibition of EMT [150]. Figure 8B summarizes how EVs’ miRNAs serve in the therapeutic implications of human TNBC.
In addition to the above, bioengineered EVs from the TNBC cells themselves often turn out to be more effective against TNBC [151]. For example, TNBC–exosome–encapsulated doxorubicin was shown to be more effective against TNBC than its free form, with reduced cardiac toxicity [151]. On the other hand, miR–9 and miR–155–loaded EVs from TNBC cells often transfer their metastatic phenotype to the non–metastatic cells MCF7 via the downregulation of the respective targets PTEN and DSUP14 [116]. Furthermore, regarding TNBC cells, HCC1806–derived EVs not only promote the proliferation but also confer resistance in the normal breast epithelial cells MCF10A against the therapeutic drugs docetaxel and doxorubicin [107]. A recent study indicated that EVs released from TNBC cells that were bioengineered to express a high–affinity variant of the human PD–1 protein (havPD–1) and knockdown intrinsic PD–L1 and β2–microglobulin reduce tumor proliferation and promote apoptosis via the downregulation of PD–L1–dependent T–cell suppression [152]. The addition of a PARP inhibitor with PD–1 + EVs further improves the treatment efficacy [152]. Moreover, EVs from HEK293F cells are also shown to target TNBC tumors when bioengineered to contain verrucarin A (Ver–A) followed by surface tagging with anti–EGFR/CD47 mAbs [153].

9. EVs in Clinical Trials: The Translational Significance in Cancer

Due to significant success in preclinical studies indicating the enormous diagnostic potential of EVs, EVs have been registered in several clinical trials in the context of different cancers, as discussed in Table 6 (www.clinicaltrials.gov, accessed on 18 September 2023).

10. Conclusions, Future Direction and Challenges

TNBC is considered the most aggressive form of human breast cancer and accounts for ~10–20% of all breast cancer phenotypes. The paucity of PR and HER2 on the TNBC cells renders conventional therapeutic regimens ineffective against human TNBC. A growing body of evidence indicates that EVs play a pivotal role in the treatment of various pathophysiological conditions due to their extraordinary capability to deliver bioactive molecules to a wide variety of cells. Additionally, bioengineered EVs show potential in the fight against different types of human cancer. However, the functional application of EVs in the treatment of human TNBC is still in its initial stages. The present review highlighted the in–depth understanding of the crucial roles of EVs in the growth and metastatic dissemination of human TNBC. Moreover, how the EVs contribute as biomarkers for the early detection and progression of human TNBC is also illustrated. In addition to these, the immune regulation of EVs in the context of human TNBC is emerging and was shown to be highly effective. EV–based immunotherapeutic interventions also delineate beneficial effects in combating human TNBC. Despite these EV–associated immunotherapeutic advancements, the mechanism and function of the EVs in TNBC tumorigenesis are yet to be completely explored. A thorough and significant study needs to be performed to understand the diagnostic, prognostic, and therapeutic values of EVs in clinical settings. Moreover, EV–based combinational therapy alongside immunotherapy could be highly effective against human TNBC.
Despite the abovementioned promises, EV–based therapy against TNBC still faces significant challenges. The conventional protocols for the isolation and characterization of EVs often result in EV yields with high variability. Moreover, contamination by cellular fragments, impurities, and similar–sized particles further add to the variability. Again, the role of EVs in the epigenetic regulation of the TNBC needs to be further investigated. Additionally, in the TME, several cells communicate with the tumor cells and vice versa via the release of EVs; hence, it is very difficult to understand the crucial roles of the EVs in a particular cell type under a given physiologic condition. However, despite these shortcomings, EVs are still considered a promising biomarker and therapeutic means against TNBC [167].
In addition to the above, the treatment of human TNBC itself has a few significant limitations. The major metabolic pathways involved in the progression of TNBC that could be targeted converge with one or a few HUB proteins and bottleneck proteins that include PTEN, AKT1, MAPK1, MAPK3, EGFR, TP53, UBC, HRAS, RPS27A, GRB2, UBA52, and SRC. These proteins also play pivotal roles in the normal physiological functions of almost every cell; hence, targeting these proteins often interferes with the normal physiological functions of the system. Therefore, at present, the identification of specific molecules is a prerequisite and of prime importance in targeting TNBC tumors without affecting normal physiology. Without knowledge of specific molecular mechanisms, designing the drugs that combat TNBC becomes almost impossible. Moreover, several post–translational modifications (PTMs) affecting protein functions and activity further add to the complexity. The heterogeneity of TNBC can also be influenced by PTMs, which again complicates the matter for the development of specific targeted therapies, including emerging immunotherapies. The lack of a comprehensive and dynamic view of protein interactions, PTMs, and metabolic functions not only impedes understanding the complex behavior of human TNBC but also restricts the development of specific therapeutic targets. However, the emerging development of alternating approaches and technologies, as well as the advancement of genomics, proteomics, metabolomics, and other disciplines, certainly hold promise for a better understanding of TNBC complexity, thereby developing potential therapeutic targets.

Author Contributions

K.D. and D.P. contributed to the conceptualization, literature search, study design, critical review, and preparation of the final version of the manuscript. S.P., A.G., S.G., T.M., P.S., A.G., S.K. and A.S. contributed to the literature search, data collection, and preparation of the manuscript. S.C.C. and V.K.K. contributed to the study design, critical review, and preparation of the final version of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

All the images in the manuscript were created with BioRender.com on 7 July 2023. We acknowledge BioRender for providing us with their platform for the preparation of images.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

EVextracellular vesicle
miRmicroRNA
MPmicroparticle
MVmicrovesicle
PSphosphatidylserine
HSPheat–shock protein
MVBmultivesicular body
ESCRTendosomal sorting complexes required for transport
TSG101tumor susceptibility gene 101
AlixALG–2–interacting protein X
HSC70heat–shock cognate 70 kDa protein
LC3microtubule–associated proteins 1A/1B light chain 3B
CDcluster of differentiation
GRP78glucose–regulated protein 78
UTIurinary tract infection
TFtissue factor
ICAM–1intercellular adhesion molecule 1
AKIacute kidney injury
AQP1aquaporin 1
HFFhuman follicular fluid
PCOSpolycystic ovary syndrome
CSFcerebrospinal fluid
PDParkinson’s disease
CMcongenital myopathies
NPCnasopharyngeal carcinoma
TNBCtriple–negative breast cancer
PTENphosphatase and tensin homolog
STAT3signal transducer and activator of transcription 3
M2type 2 macrophage
HCChepatocellular carcinoma
TXNIPthioredoxin–interacting protein
IHCimmunohistochemistry
ERestrogen receptor
PRprogesterone receptor
HER2human epidermal growth factor receptor–2
EGFRextracellular growth factor receptor
CKcytokeratin
BRCA1breast cancer gene 1
EpCAMepithelial cellular adhesion molecule
EMTepithelial to mesenchymal transition
BLbasal–like
IMimmunomodulatory
LARluminal androgen receptor
Mmesenchymal
MSLmesenchymal stem like
NKnatural killer
BCRB–cell receptor
PI3Kphosphoinositide 3–kinase
mTORmammalian target of rapamycin
Wntwingless/integrated
TGFβtransforming growth factor β
MAPKmitogen–activated protein kinase
HIF1Ahypoxia inducible factor 1 subunit α
DETdeoxyelephantopin
DETD–35DET derivative 35
CAFcancer–associated fibroblast
TMEtumor microenvironment
ITGB4integrin β4
BNI3PLbcl2 interacting protein 3 like
MSCmesenchymal stem cell
HAND2–AS1heart and neural crest derivatives expressed 2 antisense RNA 1
FOSL1Fos like 1
SPANXB1sperm protein associated with the nucleus on the X chromosome (SPANX) family member B1
SH3GL2SH3 domain containing GRB2 like 2
DUSP14dual specificity phosphatase 14
VE cadherinvascular endothelial cadherin
PSMA1proteasome 20S subunit α1
FVIIaactivated factor VII
PAR2protease–activated receptor 2
MK2MAPK–activated protein kinase 2
MEKMAPK kinase
ERKextracellular signal–regulated kinase
MLCKmyosin light chain kinase
MLC2myosin light chain 2
RabRas–associated binding
NF–ĸBnuclear factor ĸB
E–cadherinepithelial cadherin
N–cadherinneuronal cadherin
AREGamphiregulin
UCHL1ubiquitin C–terminal hydrolase 1
SMAD2mothers against decapentaplegic homolog 2
PTXpaclitaxel
P–gpP–glycoprotein
Mef2cmyocyte–specific enhancer factor 2C
HOXD10homeobox D10
KLF4Krüppel–like factor 4
ZO–1zonula occludens 1
CCLchemokine (C–C motif) ligand
TEMtumor EV–educated macrophage
IFNGinterferon γ
CXCL1CXC motif chemokine ligand 1
OPNosteopontin
CTLA–4cytotoxic T–lymphocyte–associated protein 4
HGFhepatocyte growth factor
LNlymph node
PD–1programmed death 1
PD–L1programmed death ligand 1
OSoscillatory strain
MDSCmyeloid–derived suppressor cell
CAR–Tchimeric antigen receptor T
FDAFood and Drug Administration
TROP2tumor–associated calcium signal transducer 2
TILtumor–infiltrating lymphocyte
MHCmajor histocompatibility complex
FasLFas ligand
AS141126–mer G–rich DNA oligonucleotide
Ver–Averrucarin A
NSCLCnon–small cell lung cancer
CRCcolorectal cancer
BALFbronchoalveolar fluid
TP53tumor protein p53
UBCubiquitin C
HRASHarvey Rat sarcoma virus
RPS27Aribosomal protein S27A
GRB2growth factor receptor–bound protein 2
UBA52ubiquitin A–52 residue ribosomal protein fusion product 1
SRCSrc family kinase

References

  1. Chargaff, E.; West, R. The biological significance of the thromboplastic protein of blood. J. Biol. Chem. 1946, 166, 189–197. [Google Scholar] [CrossRef]
  2. Wolf, P. The nature and significance of platelet products in human plasma. Br. J. Haematol. 1967, 13, 269–288. [Google Scholar] [CrossRef] [PubMed]
  3. Harding, C.; Heuser, J.; Stahl, P. Endocytosis and intracellular processing of transferrin and colloidal gold-transferrin in rat reticulocytes: Demonstration of a pathway for receptor shedding. Eur. J. Cell Biol. 1984, 35, 256–263. [Google Scholar] [PubMed]
  4. Yanez-Mo, M.; Siljander, P.R.; Andreu, Z.; Zavec, A.B.; Borras, 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. Vesicles 2015, 4, 27066. [Google Scholar] [CrossRef]
  5. Stranford, D.M.; Leonard, J.N. Delivery of Biomolecules via Extracellular Vesicles: A Budding Therapeutic Strategy. Adv. Genet. 2017, 98, 155–175. [Google Scholar] [PubMed]
  6. Couch, Y.; Buzas, E.I.; Di Vizio, D.; Gho, Y.S.; Harrison, P.; Hill, A.F.; Lotvall, J.; Raposo, G.; Stahl, P.D.; Thery, C.; et al. A brief history of nearly Everything—The rise and rise of extracellular vesicles. J. Extracell. Vesicles 2021, 10, e12144. [Google Scholar] [CrossRef] [PubMed]
  7. Das, K.; Keshava, S.; Ansari, S.A.; Kondreddy, V.; Esmon, C.T.; Griffin, J.H.; Pendurthi, U.R.; Rao, L.V.M. Factor VIIa induces extracellular vesicles from the endothelium: A potential mechanism for its hemostatic effect. Blood 2021, 137, 3428–3442. [Google Scholar] [CrossRef]
  8. Das, K.; Keshava, S.; Pendurthi, U.R.; Rao, L.V.M. Factor VIIa suppresses inflammation and barrier disruption through the release of EEVs and transfer of microRNA 10a. Blood 2022, 139, 118–133. [Google Scholar] [CrossRef]
  9. Das, K.; Pendurthi, U.R.; Manco-Johnson, M.; Martin, E.J.; Brophy, D.F.; Rao, L.V.M. Factor VIIa treatment increases circulating extracellular vesicles in hemophilia patients: Implications for the therapeutic hemostatic effect of FVIIa. J. Thromb. Haemost. 2022, 20, 1928–1933. [Google Scholar] [CrossRef]
  10. Das, K.; Rao, L.V.M. The Role of microRNAs in Inflammation. Int. J. Mol. Sci. 2022, 23, 15479. [Google Scholar] [CrossRef]
  11. Das, K.; Paul, S.; Singh, A.; Ghosh, A.; Roy, A.; Ansari, S.A.; Prasad, R.; Mukherjee, A.; Sen, P. Triple-negative breast cancer-derived microvesicles transfer microRNA221 to the recipient cells and thereby promote epithelial-to-mesenchymal transition. J. Biol. Chem. 2019, 294, 13681–13696. [Google Scholar] [CrossRef] [PubMed]
  12. Shetty, A.K.; Upadhya, R. Extracellular Vesicles in Health and Disease. Aging Dis. 2021, 12, 1358–1362. [Google Scholar] [CrossRef] [PubMed]
  13. Akbar, N.; Azzimato, V.; Choudhury, R.P.; Aouadi, M. Extracellular vesicles in metabolic disease. Diabetologia 2019, 62, 2179–2187. [Google Scholar] [CrossRef]
  14. Das, K.; Mukherjee, T.; Shankar, P. The Role of Extracellular Vesicles in the Pathogenesis of Hematological Malignancies: Interaction with Tumor Microenvironment; a Potential Biomarker and Targeted Therapy. Biomolecules 2023, 13, 897. [Google Scholar] [CrossRef] [PubMed]
  15. Buzas, E.I.; Gyorgy, B.; Nagy, G.; Falus, A.; Gay, S. Emerging role of extracellular vesicles in inflammatory diseases. Nat. Rev. Rheumatol. 2014, 10, 356–364. [Google Scholar] [CrossRef] [PubMed]
  16. Das, K.; Paul, S.; Mukherjee, T.; Ghosh, A.; Sharma, A.; Shankar, P.; Gupta, S.; Keshava, S.; Parashar, D. Beyond Macromolecules: Extracellular Vesicles as Regulators of Inflammatory Diseases. Cells 2023, 12, 1963. [Google Scholar] [CrossRef] [PubMed]
  17. Das, K.; Prasad, R.; Roy, S.; Mukherjee, A.; Sen, P. The Protease Activated Receptor2 Promotes Rab5a Mediated Generation of Pro-metastatic Microvesicles. Sci. Rep. 2018, 8, 7357. [Google Scholar] [CrossRef] [PubMed]
  18. Das, K.; Prasad, R.; Singh, A.; Bhattacharya, A.; Roy, A.; Mallik, S.; Mukherjee, A.; Sen, P. Protease-activated receptor 2 promotes actomyosin dependent transforming microvesicles generation from human breast cancer. Mol. Carcinog. 2018, 57, 1707–1722. [Google Scholar] [CrossRef]
  19. Raposo, G.; Stoorvogel, W. Extracellular vesicles: Exosomes, microvesicles, and friends. J. Cell Biol. 2013, 200, 373–383. [Google Scholar] [CrossRef]
  20. Bonsergent, E.; Grisard, E.; Buchrieser, J.; Schwartz, O.; Thery, C.; Lavieu, G. Quantitative characterization of extracellular vesicle uptake and content delivery within mammalian cells. Nat. Commun. 2021, 12, 1864. [Google Scholar] [CrossRef]
  21. van Niel, G.; Carter, D.R.F.; Clayton, A.; Lambert, D.W.; Raposo, G.; Vader, P. Challenges and directions in studying cell-cell communication by extracellular vesicles. Nat. Rev. Mol. Cell Biol. 2022, 23, 369–382. [Google Scholar] [CrossRef] [PubMed]
  22. Bodega, G.; Alique, M.; Puebla, L.; Carracedo, J.; Ramirez, R.M. Microvesicles: ROS scavengers and ROS producers. J. Extracell. Vesicles 2019, 8, 1626654. [Google Scholar] [CrossRef] [PubMed]
  23. Lai, C.P.; Mardini, O.; Ericsson, M.; Prabhakar, S.; Maguire, C.; Chen, J.W.; Tannous, B.A.; Breakefield, X.O. Dynamic biodistribution of extracellular vesicles in vivo using a multimodal imaging reporter. ACS Nano 2014, 8, 483–494. [Google Scholar] [CrossRef] [PubMed]
  24. Xiong, J.; Miller, V.M.; Li, Y.; Jayachandran, M. Microvesicles at the crossroads between infection and cardiovascular diseases. J. Cardiovasc. Pharmacol. 2012, 59, 124–132. [Google Scholar] [CrossRef] [PubMed]
  25. 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] [PubMed]
  26. Zoller, M. Tetraspanins: Push and pull in suppressing and promoting metastasis. Nat. Rev. Cancer 2009, 9, 40–55. [Google Scholar] [CrossRef] [PubMed]
  27. 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] [PubMed]
  28. Heijnen, H.F.; 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 alpha-granules. Blood 1999, 94, 3791–3799. [Google Scholar] [CrossRef]
  29. Morello, M.; Minciacchi, V.R.; de Candia, P.; Yang, J.; Posadas, E.; Kim, H.; Griffiths, D.; Bhowmick, N.; Chung, L.W.; Gandellini, P.; et al. Large oncosomes mediate intercellular transfer of functional microRNA. Cell Cycle 2013, 12, 3526–3536. [Google Scholar] [CrossRef]
  30. Kowal, J.; Tkach, M.; Thery, C. Biogenesis and secretion of exosomes. Curr. Opin. Cell Biol. 2014, 29, 116–125. [Google Scholar] [CrossRef]
  31. Gupta, P.; Kadamberi, I.P.; Mittal, S.; Tsaih, S.W.; George, J.; Kumar, S.; Vijayan, D.K.; Geethadevi, A.; Parashar, D.; Topchyan, P.; et al. Tumor Derived Extracellular Vesicles Drive T Cell Exhaustion in Tumor Microenvironment through Sphingosine Mediated Signaling and Impacting Immunotherapy Outcomes in Ovarian Cancer. Adv. Sci. 2022, 9, e2104452. [Google Scholar] [CrossRef] [PubMed]
  32. Parashar, D.; Geethadevi, A.; McAllister, D.; Ebben, J.; Peterson, F.C.; Jensen, D.R.; Bishop, E.; Pradeep, S.; Volkman, B.F.; Dwinell, M.B.; et al. Targeted biologic inhibition of both tumor cell-intrinsic and intercellular CLPTM1L/CRR9-mediated chemotherapeutic drug resistance. NPJ Precis. Oncol. 2021, 5, 16. [Google Scholar] [CrossRef] [PubMed]
  33. Geminard, C.; De Gassart, A.; Blanc, L.; Vidal, M. Degradation of AP2 during reticulocyte maturation enhances binding of hsc70 and Alix to a common site on TFR for sorting into exosomes. Traffic 2004, 5, 181–193. [Google Scholar] [CrossRef] [PubMed]
  34. van Niel, G.; Porto-Carreiro, I.; Simoes, S.; Raposo, G. Exosomes: A common pathway for a specialized function. J. Biochem. 2006, 140, 13–21. [Google Scholar] [CrossRef] [PubMed]
  35. Trajkovic, K.; Hsu, C.; Chiantia, S.; Rajendran, L.; Wenzel, D.; Wieland, F.; Schwille, P.; Brugger, B.; Simons, M. Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science 2008, 319, 1244–1247. [Google Scholar] [CrossRef] [PubMed]
  36. Glick, D.; Barth, S.; Macleod, K.F. Autophagy: Cellular and molecular mechanisms. J. Pathol. 2010, 221, 3–12. [Google Scholar] [CrossRef]
  37. Fader, C.M.; Colombo, M.I. Autophagy and multivesicular bodies: Two closely related partners. Cell Death Differ. 2009, 16, 70–78. [Google Scholar] [CrossRef] [PubMed]
  38. Thery, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef]
  39. Krylova, S.V.; Feng, D. The Machinery of Exosomes: Biogenesis, Release, and Uptake. Int. J. Mol. Sci. 2023, 24, 1337. [Google Scholar] [CrossRef]
  40. Brunel, A.; Bégaud, G.; Auger, C.; Durand, S.; Battu, S.; Bessette, B.; Verdier, M. Autophagy and Extracellular Vesicles, Connected to rabGTPase Family, Support Aggressiveness in Cancer Stem Cells. Cells 2021, 10, 1330. [Google Scholar] [CrossRef]
  41. Park, J.S.; Perl, A. Endosome Traffic Modulates Pro-Inflammatory Signal Transduction in CD4(+) T Cells-Implications for the Pathogenesis of Systemic Lupus Erythematosus. Int. J. Mol. Sci. 2023, 24, 10749. [Google Scholar] [CrossRef] [PubMed]
  42. Arya, S.B.; Collie, S.P.; Parent, C.A. The ins-and-outs of exosome biogenesis, secretion, and internalization. Trends Cell Biol. 2023. [Google Scholar] [CrossRef] [PubMed]
  43. Doyle, L.M.; Wang, M.Z. Overview of Extracellular Vesicles, Their Origin, Composition, Purpose, and Methods for Exosome Isolation and Analysis. Cells 2019, 8, 727. [Google Scholar] [CrossRef] [PubMed]
  44. Mulcahy, L.A.; Pink, R.C.; Carter, D.R. Routes and mechanisms of extracellular vesicle uptake. J. Extracell. Vesicles 2014, 3, 24641. [Google Scholar] [CrossRef] [PubMed]
  45. O’Brien, K.; Ughetto, S.; Mahjoum, S.; Nair, A.V.; Breakefield, X.O. Uptake, functionality, and re-release of extracellular vesicle-encapsulated cargo. Cell Rep. 2022, 39, 110651. [Google Scholar] [CrossRef] [PubMed]
  46. Montecalvo, A.; Larregina, A.T.; Shufesky, W.J.; Stolz, D.B.; Sullivan, M.L.; Karlsson, J.M.; Baty, C.J.; Gibson, G.A.; Erdos, G.; Wang, Z.; et al. Mechanism of transfer of functional microRNAs between mouse dendritic cells via exosomes. Blood 2012, 119, 756–766. [Google Scholar] [CrossRef] [PubMed]
  47. Parolini, I.; Federici, C.; Raggi, C.; Lugini, L.; Palleschi, S.; De Milito, A.; Coscia, C.; Iessi, E.; Logozzi, M.; Molinari, A.; et al. Microenvironmental pH is a key factor for exosome traffic in tumor cells. J. Biol. Chem. 2009, 284, 34211–34222. [Google Scholar] [CrossRef] [PubMed]
  48. 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]
  49. 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]
  50. Thery, C.; Boussac, M.; Veron, 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]
  51. Escrevente, C.; Keller, S.; Altevogt, P.; Costa, J. Interaction and uptake of exosomes by ovarian cancer cells. BMC Cancer 2011, 11, 108. [Google Scholar] [CrossRef] [PubMed]
  52. Kerr, J.F.; Wyllie, A.H.; Currie, A.R. Apoptosis: A basic biological phenomenon with wide-ranging implications in tissue kinetics. Br. J. Cancer 1972, 26, 239–257. [Google Scholar] [CrossRef]
  53. Jeppesen, D.K.; Zhang, Q.; Franklin, J.L.; Coffey, R.J. Extracellular vesicles and nanoparticles: Emerging complexities. Trends Cell Biol. 2023, 33, 667–681. [Google Scholar] [CrossRef] [PubMed]
  54. Rother, N.; Yanginlar, C.; Pieterse, E.; Hilbrands, L.; van der Vlag, J. Microparticles in Autoimmunity: Cause or Consequence of Disease? Front. Immunol. 2022, 13, 822995. [Google Scholar] [CrossRef]
  55. Jeppesen, D.K.; Fenix, A.M.; Franklin, J.L.; Higginbotham, J.N.; Zhang, Q.; Zimmerman, L.J.; Liebler, D.C.; Ping, J.; Liu, Q.; Evans, R.; et al. Reassessment of Exosome Composition. Cell 2019, 177, 428–445.e18. [Google Scholar] [CrossRef]
  56. Baietti, M.F.; Zhang, Z.; Mortier, E.; Melchior, A.; Degeest, G.; Geeraerts, A.; Ivarsson, Y.; Depoortere, F.; Coomans, C.; Vermeiren, E.; et al. Syndecan-syntenin-ALIX regulates the biogenesis of exosomes. Nat. Cell Biol. 2012, 14, 677–685. [Google Scholar] [CrossRef] [PubMed]
  57. Ostrowski, M.; Carmo, N.B.; Krumeich, S.; Fanget, I.; Raposo, G.; Savina, A.; Moita, C.F.; Schauer, K.; Hume, A.N.; Freitas, R.P.; et al. Rab27a and Rab27b control different steps of the exosome secretion pathway. Nat. Cell Biol. 2010, 12, 19–30. [Google Scholar] [CrossRef] [PubMed]
  58. Dieudé, M.; Bell, C.; Turgeon, J.; Beillevaire, D.; Pomerleau, L.; Yang, B.; Hamelin, K.; Qi, S.; Pallet, N.; Béland, C.; et al. The 20S proteasome core, active within apoptotic exosome-like vesicles, induces autoantibody production and accelerates rejection. Sci. Transl. Med. 2015, 7, 318ra200. [Google Scholar] [CrossRef] [PubMed]
  59. Hristov, M.; Erl, W.; Linder, S.; Weber, P.C. Apoptotic bodies from endothelial cells enhance the number and initiate the differentiation of human endothelial progenitor cells in vitro. Blood 2004, 104, 2761–2766. [Google Scholar] [CrossRef]
  60. Woei, A.J.F.J.; van der Starre, W.E.; Tesselaar, M.E.; Garcia Rodriguez, P.; van Nieuwkoop, C.; Bertina, R.M.; van Dissel, J.T.; Osanto, S. Procoagulant tissue factor activity on microparticles is associated with disease severity and bacteremia in febrile urinary tract infections. Thromb. Res. 2014, 133, 799–803. [Google Scholar] [CrossRef]
  61. Tans, G.; Rosing, J.; Thomassen, M.C.; Heeb, M.J.; Zwaal, R.F.; Griffin, J.H. Comparison of anticoagulant and procoagulant activities of stimulated platelets and platelet-derived microparticles. Blood 1991, 77, 2641–2648. [Google Scholar] [CrossRef] [PubMed]
  62. Rautou, P.E.; Leroyer, A.S.; Ramkhelawon, B.; Devue, C.; Duflaut, D.; Vion, A.C.; Nalbone, G.; Castier, Y.; Leseche, G.; Lehoux, S.; et al. Microparticles from human atherosclerotic plaques promote endothelial ICAM-1-dependent monocyte adhesion and transendothelial migration. Circ. Res. 2011, 108, 335–343. [Google Scholar] [CrossRef] [PubMed]
  63. Zhang, W.; Zhou, X.; Zhang, H.; Yao, Q.; Liu, Y.; Dong, Z. Extracellular vesicles in diagnosis and therapy of kidney diseases. Am. J. Physiol. Ren. Physiol. 2016, 311, F844–F851. [Google Scholar] [CrossRef] [PubMed]
  64. Chua, R.L.; Lukassen, S.; Trump, S.; Hennig, B.P.; Wendisch, D.; Pott, F.; Debnath, O.; Thurmann, L.; Kurth, F.; Volker, M.T.; et al. COVID-19 severity correlates with airway epithelium-immune cell interactions identified by single-cell analysis. Nat. Biotechnol. 2020, 38, 970–979. [Google Scholar] [CrossRef] [PubMed]
  65. Herman, S.; Djaldetti, R.; Mollenhauer, B.; Offen, D. CSF-derived extracellular vesicles from patients with Parkinson’s disease induce symptoms and pathology. Brain 2023, 146, 209–224. [Google Scholar] [CrossRef] [PubMed]
  66. Murphy, C.; Withrow, J.; Hunter, M.; Liu, Y.; Tang, Y.L.; Fulzele, S.; Hamrick, M.W. Emerging role of extracellular vesicles in musculoskeletal diseases. Mol. Asp. Med. 2018, 60, 123–128. [Google Scholar] [CrossRef] [PubMed]
  67. Yang, J.; Shin, T.S.; Kim, J.S.; Jee, Y.K.; Kim, Y.K. A new horizon of precision medicine: Combination of the microbiome and extracellular vesicles. Exp. Mol. Med. 2022, 54, 466–482. [Google Scholar] [CrossRef] [PubMed]
  68. Tian, X.; Liu, Y.; Wang, Z.; Wu, S. miR-144 delivered by nasopharyngeal carcinoma-derived EVs stimulates angiogenesis through the FBXW7/HIF-1alpha/VEGF-A axis. Mol. Ther. Nucleic Acids 2021, 24, 1000–1011. [Google Scholar] [CrossRef]
  69. Hsu, Y.L.; Hung, J.Y.; Chang, W.A.; Jian, S.F.; Lin, Y.S.; Pan, Y.C.; Wu, C.Y.; Kuo, P.L. Hypoxic Lung-Cancer-Derived Extracellular Vesicle MicroRNA-103a Increases the Oncogenic Effects of Macrophages by Targeting PTEN. Mol. Ther. 2018, 26, 568–581. [Google Scholar] [CrossRef]
  70. Yang, Y.; Mao, F.; Guo, L.; Shi, J.; Wu, M.; Cheng, S.; Guo, W. Tumor cells derived-extracellular vesicles transfer miR-3129 to promote hepatocellular carcinoma metastasis by targeting TXNIP. Dig. Liver Dis. 2021, 53, 474–485. [Google Scholar] [CrossRef]
  71. Wang, S.; Zhang, Z.; Gao, Q. Transfer of microRNA-25 by colorectal cancer cell-derived extracellular vesicles facilitates colorectal cancer development and metastasis. Mol. Ther. Nucleic Acids 2021, 23, 552–564. [Google Scholar] [CrossRef] [PubMed]
  72. Smolarz, B.; Nowak, A.Z.; Romanowicz, H. Breast Cancer-Epidemiology, Classification, Pathogenesis and Treatment (Review of Literature). Cancers 2022, 14, 2569. [Google Scholar] [CrossRef] [PubMed]
  73. Jagadish, N.; Gupta, N.; Agarwal, S.; Parashar, D.; Sharma, A.; Fatima, R.; Topno, A.P.; Kumar, V.; Suri, A. Sperm-associated antigen 9 (SPAG9) promotes the survival and tumor growth of triple-negative breast cancer cells. Tumour Biol. J. Int. Soc. Oncodevelopmental Biol. Med. 2016, 37, 13101–13110. [Google Scholar] [CrossRef] [PubMed]
  74. Sinha, A.; Agarwal, S.; Parashar, D.; Verma, A.; Saini, S.; Jagadish, N.; Ansari, A.S.; Lohiya, N.K.; Suri, A. Down regulation of SPAG9 reduces growth and invasive potential of triple-negative breast cancer cells: Possible implications in targeted therapy. J. Exp. Clin. Cancer Res. CR 2013, 32, 69. [Google Scholar] [CrossRef] [PubMed]
  75. Giaquinto, A.N.; Sung, H.; Miller, K.D.; Kramer, J.L.; Newman, L.A.; Minihan, A.; Jemal, A.; Siegel, R.L. Breast Cancer Statistics, 2022. CA Cancer J. Clin. 2022, 72, 524–541. [Google Scholar] [CrossRef] [PubMed]
  76. Sorlie, 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]
  77. Vallejos, C.S.; Gomez, H.L.; Cruz, W.R.; Pinto, J.A.; Dyer, R.R.; Velarde, R.; Suazo, J.F.; Neciosup, S.P.; Leon, M.; de la Cruz, M.A.; et al. Breast cancer classification according to immunohistochemistry markers: Subtypes and association with clinicopathologic variables in a peruvian hospital database. Clin. Breast Cancer 2010, 10, 294–300. [Google Scholar] [CrossRef]
  78. Urruticoechea, A.; Smith, I.E.; Dowsett, M. Proliferation marker Ki-67 in early breast cancer. J. Clin. Oncol. 2005, 23, 7212–7220. [Google Scholar] [CrossRef]
  79. Abd El-Rehim, D.M.; Pinder, S.E.; Paish, C.E.; Bell, J.; Blamey, R.W.; Robertson, J.F.; Nicholson, R.I.; Ellis, I.O. Expression of luminal and basal cytokeratins in human breast carcinoma. J. Pathol. 2004, 203, 661–671. [Google Scholar] [CrossRef]
  80. Yersal, O.; Barutca, S. Biological subtypes of breast cancer: Prognostic and therapeutic implications. World J. Clin. Oncol. 2014, 5, 412–424. [Google Scholar] [CrossRef]
  81. Perou, C.M.; Sorlie, 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] [PubMed]
  82. Ellis, M.J.; Tao, Y.; Luo, J.; A’Hern, R.; Evans, D.B.; Bhatnagar, A.S.; Chaudri Ross, H.A.; von Kameke, A.; Miller, W.R.; Smith, I.; et al. Outcome prediction for estrogen receptor-positive breast cancer based on postneoadjuvant endocrine therapy tumor characteristics. J. Natl. Cancer Inst. 2008, 100, 1380–1388. [Google Scholar] [CrossRef] [PubMed]
  83. Fragomeni, S.M.; Sciallis, A.; Jeruss, J.S. Molecular Subtypes and Local-Regional Control of Breast Cancer. Surg. Oncol. Clin. N. Am. 2018, 27, 95–120. [Google Scholar] [CrossRef] [PubMed]
  84. von Minckwitz, G.; Procter, M.; de Azambuja, E.; Zardavas, D.; Benyunes, M.; Viale, G.; Suter, T.; Arahmani, A.; Rouchet, N.; Clark, E.; et al. Investigators, Adjuvant Pertuzumab and Trastuzumab in Early HER2-Positive Breast Cancer. N. Engl. J. Med. 2017, 377, 122–131. [Google Scholar] [CrossRef] [PubMed]
  85. 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]
  86. Collins, L.C.; Martyniak, A.; Kandel, M.J.; Stadler, Z.K.; Masciari, S.; Miron, A.; Richardson, A.L.; Schnitt, S.J.; Garber, J.E. Basal cytokeratin and epidermal growth factor receptor expression are not predictive of BRCA1 mutation status in women with triple-negative breast cancers. Am. J. Surg. Pathol. 2009, 33, 1093–1097. [Google Scholar] [CrossRef] [PubMed]
  87. Kanapathy Pillai, S.K.; Tay, A.; Nair, S.; Leong, C.O. Triple-negative breast cancer is associated with EGFR, CK5/6 and c-KIT expression in Malaysian women. BMC Clin. Pathol. 2012, 12, 18. [Google Scholar] [CrossRef] [PubMed]
  88. Cheang, M.C.; Voduc, D.; Bajdik, C.; Leung, S.; McKinney, S.; Chia, S.K.; Perou, C.M.; Nielsen, T.O. Basal-like breast cancer defined by five biomarkers has superior prognostic value than triple-negative phenotype. Clin. Cancer Res. 2008, 14, 1368–1376. [Google Scholar] [CrossRef]
  89. Rakha, E.A.; Elsheikh, S.E.; Aleskandarany, M.A.; Habashi, H.O.; Green, A.R.; Powe, D.G.; El-Sayed, M.E.; Benhasouna, A.; Brunet, J.S.; Akslen, L.A.; et al. Triple-negative breast cancer: Distinguishing between basal and nonbasal subtypes. Clin. Cancer Res. 2009, 15, 2302–2310. [Google Scholar] [CrossRef]
  90. 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]
  91. Nounou, M.I.; ElAmrawy, F.; Ahmed, N.; Abdelraouf, K.; Goda, S.; Syed-Sha-Qhattal, H. Breast Cancer: Conventional Diagnosis and Treatment Modalities and Recent Patents and Technologies. Breast Cancer 2015, 9 (Suppl. 2), 17–34. [Google Scholar] [CrossRef] [PubMed]
  92. Hennessy, B.T.; Gonzalez-Angulo, A.M.; Stemke-Hale, K.; Gilcrease, M.Z.; Krishnamurthy, S.; Lee, J.S.; Fridlyand, J.; Sahin, A.; Agarwal, R.; Joy, C.; et al. Characterization of a naturally occurring breast cancer subset enriched in epithelial-to-mesenchymal transition and stem cell characteristics. Cancer Res. 2009, 69, 4116–4124. [Google Scholar] [CrossRef] [PubMed]
  93. Creighton, C.J.; Li, X.; Landis, M.; Dixon, J.M.; Neumeister, V.M.; Sjolund, A.; Rimm, D.L.; Wong, H.; Rodriguez, A.; Herschkowitz, J.I.; et al. Residual breast cancers after conventional therapy display mesenchymal as well as tumor-initiating features. Proc. Natl. Acad. Sci. USA 2009, 106, 13820–13825. [Google Scholar] [CrossRef] [PubMed]
  94. Taube, J.H.; Herschkowitz, J.I.; Komurov, K.; Zhou, A.Y.; Gupta, S.; Yang, J.; Hartwell, K.; Onder, T.T.; Gupta, P.B.; Evans, K.W.; et al. Core epithelial-to-mesenchymal transition interactome gene-expression signature is associated with claudin-low and metaplastic breast cancer subtypes. Proc. Natl. Acad. Sci. USA 2010, 107, 15449–15454. [Google Scholar] [CrossRef]
  95. Lehmann, B.D.; Bauer, J.A.; Chen, X.; Sanders, M.E.; Chakravarthy, A.B.; Shyr, Y.; Pietenpol, J.A. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J. Clin. Investig. 2011, 121, 2750–2767. [Google Scholar] [CrossRef]
  96. De Soto, J.A.; Wang, X.; Tominaga, Y.; Wang, R.H.; Cao, L.; Qiao, W.; Li, C.; Xu, X.; Skoumbourdis, A.P.; Prindiville, S.A.; et al. The inhibition and treatment of breast cancer with poly (ADP-ribose) polymerase (PARP-1) inhibitors. Int. J. Biol. Sci. 2006, 2, 179–185. [Google Scholar] [CrossRef] [PubMed]
  97. Hu, X.C.; Zhang, J.; Xu, B.H.; Cai, L.; Ragaz, J.; Wang, Z.H.; Wang, B.Y.; Teng, Y.E.; Tong, Z.S.; Pan, Y.Y.; et al. Cisplatin plus gemcitabine versus paclitaxel plus gemcitabine as first-line therapy for metastatic triple-negative breast cancer (CBCSG006): A randomised, open-label, multicentre, phase 3 trial. Lancet Oncol. 2015, 16, 436–446. [Google Scholar] [CrossRef] [PubMed]
  98. O’Shaughnessy, J.; Schwartzberg, L.; Danso, M.A.; Miller, K.D.; Rugo, H.S.; Neubauer, M.; Robert, N.; Hellerstedt, B.; Saleh, M.; Richards, P.; et al. Phase III study of iniparib plus gemcitabine and carboplatin versus gemcitabine and carboplatin in patients with metastatic triple-negative breast cancer. J. Clin. Oncol. 2014, 32, 3840–3847. [Google Scholar] [CrossRef]
  99. Carey, L.A.; Rugo, H.S.; Marcom, P.K.; Mayer, E.L.; Esteva, F.J.; Ma, C.X.; Liu, M.C.; Storniolo, A.M.; Rimawi, M.F.; Forero-Torres, A.; et al. TBCRC 001: Randomized phase II study of cetuximab in combination with carboplatin in stage IV triple-negative breast cancer. J. Clin. Oncol. 2012, 30, 2615–2623. [Google Scholar] [CrossRef]
  100. Nanda, R.; Chow, L.Q.; Dees, E.C.; Berger, R.; Gupta, S.; Geva, R.; Pusztai, L.; Pathiraja, K.; Aktan, G.; Cheng, J.D.; et al. Pembrolizumab in Patients With Advanced Triple-Negative Breast Cancer: Phase Ib KEYNOTE-012 Study. J. Clin. Oncol. 2016, 34, 2460–2467. [Google Scholar] [CrossRef]
  101. Gucalp, A.; Traina, T.A. Triple-negative breast cancer: Role of the androgen receptor. Cancer J. 2010, 16, 62–65. [Google Scholar] [CrossRef] [PubMed]
  102. Gucalp, A.; Tolaney, S.; Isakoff, S.J.; Ingle, J.N.; Liu, M.C.; Carey, L.A.; Blackwell, K.; Rugo, H.; Nabell, L.; Forero, A.; et al. Phase II trial of bicalutamide in patients with androgen receptor-positive, estrogen receptor-negative metastatic Breast Cancer. Clin. Cancer Res. 2013, 19, 5505–5512. [Google Scholar] [CrossRef]
  103. Herschkowitz, J.I.; Simin, K.; Weigman, V.J.; Mikaelian, I.; Usary, J.; Hu, Z.; Rasmussen, K.E.; Jones, L.P.; Assefnia, S.; Chandrasekharan, S.; et al. Identification of conserved gene expression features between murine mammary carcinoma models and human breast tumors. Genome Biol. 2007, 8, R76. [Google Scholar] [CrossRef] [PubMed]
  104. Xu, J.; Prosperi, J.R.; Choudhury, N.; Olopade, O.I.; Goss, K.H. beta-Catenin is required for the tumorigenic behavior of triple-negative breast cancer cells. PLoS ONE 2015, 10, e0117097. [Google Scholar]
  105. Basho, R.K.; Gilcrease, M.; Murthy, R.K.; Helgason, T.; Karp, D.D.; Meric-Bernstam, F.; Hess, K.R.; Herbrich, S.M.; Valero, V.; Albarracin, C.; et al. Targeting the PI3K/AKT/mTOR Pathway for the Treatment of Mesenchymal Triple-Negative Breast Cancer: Evidence From a Phase 1 Trial of mTOR Inhibition in Combination With Liposomal Doxorubicin and Bevacizumab. JAMA Oncol. 2017, 3, 509–515. [Google Scholar] [CrossRef] [PubMed]
  106. Bhola, N.E.; Balko, J.M.; Dugger, T.C.; Kuba, M.G.; Sanchez, V.; Sanders, M.; Stanford, J.; Cook, R.S.; Arteaga, C.L. TGF-beta inhibition enhances chemotherapy action against triple-negative breast cancer. J. Clin. Investig. 2013, 123, 1348–1358. [Google Scholar] [CrossRef]
  107. 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]
  108. Shiau, J.Y.; Chang, Y.Q.; Nakagawa-Goto, K.; Lee, K.H.; Shyur, L.F. Phytoagent Deoxyelephantopin and Its Derivative Inhibit Triple Negative Breast Cancer Cell Activity through ROS-Mediated Exosomal Activity and Protein Functions. Front. Pharmacol. 2017, 8, 398. [Google Scholar] [CrossRef]
  109. Sung, J.S.; Kang, C.W.; Kang, S.; Jang, Y.; Chae, Y.C.; Kim, B.G.; Cho, N.H. ITGB4-mediated metabolic reprogramming of cancer-associated fibroblasts. Oncogene 2020, 39, 664–676. [Google Scholar] [CrossRef]
  110. Xing, L.; Tang, X.; Wu, K.; Huang, X.; Yi, Y.; Huan, J. LncRNA HAND2-AS1 suppressed the growth of triple negative breast cancer via reducing secretion of MSCs derived exosomal miR-106a-5p. Aging 2020, 13, 424–436. [Google Scholar] [CrossRef]
  111. 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] [PubMed]
  112. 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] [PubMed]
  113. Zhang, C.; Yang, Z.; Zhou, P.; Yu, M.; Li, B.; Liu, Y.; Jin, J.; Liu, W.; Jing, H.; Du, J.; et al. Phosphatidylserine-exposing tumor-derived microparticles exacerbate coagulation and cancer cell transendothelial migration in triple-negative breast cancer. Theranostics 2021, 11, 6445–6460. [Google Scholar] [CrossRef] [PubMed]
  114. Li, S.; Li, X.; Yang, S.; Pi, H.; Li, Z.; Yao, P.; Zhang, Q.; Wang, Q.; Shen, P.; Li, X.; et al. Proteomic Landscape of Exosomes Reveals the Functional Contributions of CD151 in Triple-Negative Breast Cancer. Mol. Cell Proteom. 2021, 20, 100121. [Google Scholar] [CrossRef] [PubMed]
  115. Kannan, A.; Philley, J.V.; Hertweck, K.L.; Ndetan, H.; Singh, K.P.; Sivakumar, S.; Wells, R.B.; Vadlamudi, R.K.; Dasgupta, S. Cancer Testis Antigen Promotes Triple Negative Breast Cancer Metastasis and is Traceable in the Circulating Extracellular Vesicles. Sci. Rep. 2019, 9, 11632. [Google Scholar] [CrossRef] [PubMed]
  116. Kia, V.; Paryan, M.; Mortazavi, Y.; Biglari, A.; Mohammadi-Yeganeh, S. Evaluation of exosomal miR-9 and miR-155 targeting PTEN and DUSP14 in highly metastatic breast cancer and their effect on low metastatic cells. J. Cell. Biochem. 2019, 120, 5666–5676. [Google Scholar] [CrossRef] [PubMed]
  117. Di Modica, M.; Regondi, V.; Sandri, M.; Iorio, M.V.; Zanetti, A.; Tagliabue, E.; Casalini, P.; Triulzi, T. Breast cancer-secreted miR-939 downregulates VE-cadherin and destroys the barrier function of endothelial monolayers. Cancer Lett. 2017, 384, 94–100. [Google Scholar] [CrossRef] [PubMed]
  118. Yang, S.J.; Wang, D.D.; Zhong, S.L.; Chen, W.Q.; Wang, F.L.; Zhang, J.; Xu, W.X.; Xu, D.; Zhang, Q.; Li, J.; et al. Tumor-derived exosomal circPSMA1 facilitates the tumorigenesis, metastasis, and migration in triple-negative breast cancer (TNBC) through miR-637/Akt1/beta-catenin (cyclin D1) axis. Cell Death Dis. 2021, 12, 420. [Google Scholar] [CrossRef]
  119. Abdol Razak, N.B.; Jones, G.; Bhandari, M.; Berndt, M.C.; Metharom, P. Cancer-Associated Thrombosis: An Overview of Mechanisms, Risk Factors, and Treatment. Cancers 2018, 10, 380. [Google Scholar] [CrossRef]
  120. Das, K.; Prasad, R.; Ansari, S.A.; Roy, A.; Mukherjee, A.; Sen, P. Matrix metalloproteinase-2: A key regulator in coagulation proteases mediated human breast cancer progression through autocrine signaling. Biomed. Pharmacother. 2018, 105, 395–406. [Google Scholar] [CrossRef]
  121. Higginbotham, J.N.; Demory Beckler, M.; Gephart, J.D.; Franklin, J.L.; Bogatcheva, G.; Kremers, G.J.; Piston, D.W.; Ayers, G.D.; McConnell, R.E.; Tyska, M.J.; et al. Amphiregulin exosomes increase cancer cell invasion. Curr. Biol. 2011, 21, 779–786. [Google Scholar] [CrossRef] [PubMed]
  122. Liu, S.; Gonzalez-Prieto, R.; Zhang, M.; Geurink, P.P.; Kooij, R.; Iyengar, P.V.; van Dinther, M.; Bos, E.; Zhang, X.; Le Devedec, S.E.; et al. Deubiquitinase Activity Profiling Identifies UCHL1 as a Candidate Oncoprotein That Promotes TGFbeta-Induced Breast Cancer Metastasis. Clin. Cancer Res. 2020, 26, 1460–1473. [Google Scholar] [CrossRef] [PubMed]
  123. Kreger, B.T.; Johansen, E.R.; Cerione, R.A.; Antonyak, M.A. The Enrichment of Survivin in Exosomes from Breast Cancer Cells Treated with Paclitaxel Promotes Cell Survival and Chemoresistance. Cancers 2016, 8, 111. [Google Scholar] [CrossRef] [PubMed]
  124. Wang, B.; Zhang, Y.; Ye, M.; Wu, J.; Ma, L.; Chen, H. Cisplatin-resistant MDA-MB-231 Cell-derived Exosomes Increase the Resistance of Recipient Cells in an Exosomal miR-423-5p-dependent Manner. Curr. Drug Metab. 2019, 20, 804–814. [Google Scholar] [CrossRef] [PubMed]
  125. Yang, M.; Chen, J.; Su, F.; Yu, B.; Su, F.; Lin, L.; Liu, Y.; Huang, J.D.; Song, E. Microvesicles secreted by macrophages shuttle invasion-potentiating microRNAs into breast cancer cells. Mol. Cancer 2011, 10, 117. [Google Scholar] [CrossRef] [PubMed]
  126. 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]
  127. Zhou, W.; Fong, M.Y.; Min, Y.; Somlo, G.; Liu, L.; Palomares, M.R.; Yu, Y.; Chow, A.; O’Connor, S.T.; 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] [PubMed]
  128. Parashar, D.; Geethadevi, A.; Aure, M.R.; Mishra, J.; George, J.; Chen, C.; Mishra, M.K.; Tahiri, A.; Zhao, W.; Nair, B.; et al. miRNA551b-3p Activates an Oncostatin Signaling Module for the Progression of Triple-Negative Breast Cancer. Cell Rep. 2019, 29, 4389–4406.e10. [Google Scholar] [CrossRef]
  129. Rabe, D.C.; Walker, N.D.; Rustandy, F.D.; Wallace, J.; Lee, J.; Stott, S.L.; Rosner, M.R. Tumor Extracellular Vesicles Regulate Macrophage-Driven Metastasis through CCL5. Cancers 2021, 13, 3459. [Google Scholar] [CrossRef]
  130. Piao, Y.J.; Kim, H.S.; Hwang, E.H.; Woo, J.; Zhang, M.; Moon, W.K. Breast cancer cell-derived exosomes and macrophage polarization are associated with lymph node metastasis. Oncotarget 2018, 9, 7398–7410. [Google Scholar] [CrossRef]
  131. Qiu, Y.; Yang, Y.; Yang, R.; Liu, C.; Hsu, J.M.; Jiang, Z.; Sun, L.; Wei, Y.; Li, C.W.; Yu, D.; et al. Activated T cell-derived exosomal PD-1 attenuates PD-L1-induced immune dysfunction in triple-negative breast cancer. Oncogene 2021, 40, 4992–5001. [Google Scholar] [CrossRef] [PubMed]
  132. Wang, Y.; Goliwas, K.F.; Severino, P.E.; Hough, K.P.; Van Vessem, D.; Wang, H.; Tousif, S.; Koomullil, R.P.; Frost, A.R.; Ponnazhagan, S.; et al. Mechanical strain induces phenotypic changes in breast cancer cells and promotes immunosuppression in the tumor microenvironment. Lab. Invest. 2020, 100, 1503–1516. [Google Scholar] [CrossRef]
  133. Naran, K.; Nundalall, T.; Chetty, S.; Barth, S. Principles of Immunotherapy: Implications for Treatment Strategies in Cancer and Infectious Diseases. Front. Microbiol. 2018, 9, 3158. [Google Scholar] [CrossRef] [PubMed]
  134. Koury, J.; Lucero, M.; Cato, C.; Chang, L.; Geiger, J.; Henry, D.; Hernandez, J.; Hung, F.; Kaur, P.; Teskey, G.; et al. Immunotherapies: Exploiting the Immune System for Cancer Treatment. J. Immunol. Res. 2018, 2018, 9585614. [Google Scholar] [CrossRef] [PubMed]
  135. Nasiri, F.; Kazemi, M.; Mirarefin, S.M.J.; Mahboubi Kancha, M.; Ahmadi Najafabadi, M.; Salem, F.; Dashti Shokoohi, S.; Evazi Bakhshi, S.; Safarzadeh Kozani, P.; Safarzadeh Kozani, P. CAR-T cell therapy in triple-negative breast cancer: Hunting the invisible devil. Front. Immunol. 2022, 13, 1018786. [Google Scholar] [CrossRef] [PubMed]
  136. Emens, L.A.; Cruz, C.; Eder, J.P.; Braiteh, F.; Chung, C.; Tolaney, S.M.; Kuter, I.; Nanda, R.; Cassier, P.A.; Delord, J.P.; et al. Long-term Clinical Outcomes and Biomarker Analyses of Atezolizumab Therapy for Patients With Metastatic Triple-Negative Breast Cancer: A Phase 1 Study. JAMA Oncol. 2019, 5, 74–82. [Google Scholar] [CrossRef] [PubMed]
  137. Miles, D.; Gligorov, J.; Andre, F.; Cameron, D.; Schneeweiss, A.; Barrios, C.; Xu, B.; Wardley, A.; Kaen, D.; Andrade, L.; et al. Primary results from IMpassion131, a double-blind, placebo-controlled, randomised phase III trial of first-line paclitaxel with or without atezolizumab for unresectable locally advanced/metastatic triple-negative breast cancer. Ann. Oncol. 2021, 32, 994–1004. [Google Scholar] [CrossRef]
  138. Weiss, J.; Glode, A.; Messersmith, W.A.; Diamond, J. Sacituzumab govitecan: Breakthrough targeted therapy for triple-negative breast cancer. Expert. Rev. Anticancer. Ther. 2019, 19, 673–679. [Google Scholar] [CrossRef]
  139. Bardia, A.; Mayer, I.A.; Diamond, J.R.; Moroose, R.L.; Isakoff, S.J.; Starodub, A.N.; Shah, N.C.; O’Shaughnessy, J.; Kalinsky, K.; Guarino, M.; et al. Efficacy and Safety of Anti-Trop-2 Antibody Drug Conjugate Sacituzumab Govitecan (IMMU-132) in Heavily Pretreated Patients With Metastatic Triple-Negative Breast Cancer. J. Clin. Oncol. 2017, 35, 2141–2148. [Google Scholar] [CrossRef]
  140. Lyons, T.G. Targeted Therapies for Triple-Negative Breast Cancer. Curr. Treat. Options Oncol. 2019, 20, 82. [Google Scholar] [CrossRef]
  141. Rosenberg, J.; Huang, J. CD8(+) T Cells and NK Cells: Parallel and Complementary Soldiers of Immunotherapy. Curr. Opin. Chem. Eng. 2018, 19, 9–20. [Google Scholar] [CrossRef] [PubMed]
  142. Mittendorf, E.A.; Philips, A.V.; Meric-Bernstam, F.; Qiao, N.; Wu, Y.; Harrington, S.; Su, X.; Wang, Y.; Gonzalez-Angulo, A.M.; Akcakanat, A.; et al. PD-L1 expression in triple-negative breast cancer. Cancer Immunol. Res. 2014, 2, 361–370. [Google Scholar] [CrossRef] [PubMed]
  143. Batista, I.A.; Quintas, S.T.; Melo, S.A. The Interplay of Exosomes and NK Cells in Cancer Biology. Cancers 2021, 13, 473. [Google Scholar] [CrossRef] [PubMed]
  144. Lugini, L.; Cecchetti, S.; Huber, V.; Luciani, F.; Macchia, G.; Spadaro, F.; Paris, L.; Abalsamo, L.; Colone, M.; Molinari, A.; et al. Immune surveillance properties of human NK cell-derived exosomes. J. Immunol. 2012, 189, 2833–2842. [Google Scholar] [CrossRef] [PubMed]
  145. Zhu, L.; Kalimuthu, S.; Gangadaran, P.; Oh, J.M.; Lee, H.W.; Baek, S.H.; Jeong, S.Y.; Lee, S.W.; Lee, J.; Ahn, B.C. Exosomes Derived From Natural Killer Cells Exert Therapeutic Effect in Melanoma. Theranostics 2017, 7, 2732–2745. [Google Scholar] [CrossRef] [PubMed]
  146. Wu, C.H.; 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. Vesicles 2019, 8, 1588538. [Google Scholar] [CrossRef] [PubMed]
  147. Wang, Y.; Chen, X.; Tian, B.; Liu, J.; Yang, L.; Zeng, L.; Chen, T.; Hong, A.; Wang, X. Nucleolin-targeted Extracellular Vesicles as a Versatile Platform for Biologics Delivery to Breast Cancer. Theranostics 2017, 7, 1360–1372. [Google Scholar] [CrossRef]
  148. Lim, P.K.; Bliss, S.A.; Patel, S.A.; Taborga, M.; Dave, M.A.; Gregory, L.A.; Greco, S.J.; Bryan, M.; Patel, P.S.; Rameshwar, P. Gap junction-mediated import of microRNA from bone marrow stromal cells can elicit cell cycle quiescence in breast cancer cells. Cancer Res. 2011, 71, 1550–1560. [Google Scholar] [CrossRef]
  149. Zhou, Y.; Yamamoto, Y.; Takeshita, F.; Yamamoto, T.; Xiao, Z.; Ochiya, T. Delivery of miR-424-5p via Extracellular Vesicles Promotes the Apoptosis of MDA-MB-231 TNBC Cells in the Tumor Microenvironment. Int. J. Mol. Sci. 2021, 22, 844. [Google Scholar] [CrossRef]
  150. Shojaei, S.; Hashemi, S.M.; Ghanbarian, H.; Sharifi, K.; Salehi, M.; Mohammadi-Yeganeh, S. Delivery of miR-381-3p Mimic by Mesenchymal Stem Cell-Derived Exosomes Inhibits Triple Negative Breast Cancer Aggressiveness; an In Vitro Study. Stem Cell Rev. Rep. 2021, 17, 1027–1038. [Google Scholar] [CrossRef]
  151. Hadla, M.; Palazzolo, S.; Corona, G.; Caligiuri, I.; Canzonieri, V.; Toffoli, G.; Rizzolio, F. Exosomes increase the therapeutic index of doxorubicin in breast and ovarian cancer mouse models. Nanomedicine 2016, 11, 2431–2441. [Google Scholar] [CrossRef] [PubMed]
  152. Chen, Y.; Wang, L.; Zheng, M.; Zhu, C.; Wang, G.; Xia, Y.; Blumenthal, E.J.; Mao, W.; Wan, Y. Engineered extracellular vesicles for concurrent Anti-PDL1 immunotherapy and chemotherapy. Bioact. Mater. 2022, 9, 251–265. [Google Scholar] [CrossRef] [PubMed]
  153. Si, Y.; Chen, K.; Ngo, H.G.; Guan, J.S.; Totoro, A.; Zhou, Z.; Kim, S.; Kim, T.; Zhou, L.; Liu, X. Targeted EV to Deliver Chemotherapy to Treat Triple-Negative Breast Cancers. Pharmaceutics 2022, 14, 146. [Google Scholar] [CrossRef] [PubMed]
  154. Kretschmer, A.; Tutrone, R.; Alter, J.; Berg, E.; Fischer, C.; Kumar, S.; Torkler, P.; Tadigotla, V.; Donovan, M.; Sant, G.; et al. Pre-diagnosis urine exosomal RNA (ExoDx EPI score) is associated with post-prostatectomy pathology outcome. World J. Urol. 2022, 40, 983–989. [Google Scholar] [CrossRef]
  155. Panigrahi, A.R.; Srinivas, L.; Panda, J. Exosomes: Insights and therapeutic applications in cancer. Transl. Oncol. 2022, 21, 101439. [Google Scholar] [CrossRef]
  156. Buscail, E.; Alix-Panabières, C.; Quincy, P.; Cauvin, T.; Chauvet, A.; Degrandi, O.; Caumont, C.; Verdon, S.; Lamrissi, I.; Moranvillier, I.; et al. High Clinical Value of Liquid Biopsy to Detect Circulating Tumor Cells and Tumor Exosomes in Pancreatic Ductal Adenocarcinoma Patients Eligible for Up-Front Surgery. Cancers 2019, 11, 1656. [Google Scholar] [CrossRef]
  157. Aheget, H.; Mazini, L.; Martin, F.; Belqat, B.; Marchal, J.A.; Benabdellah, K. Exosomes: Their Role in Pathogenesis, Diagnosis and Treatment of Diseases. Cancers 2020, 13, 84. [Google Scholar] [CrossRef]
  158. Janockova, J.; Slovinska, L.; Harvanova, D.; Spakova, T.; Rosocha, J. New therapeutic approaches of mesenchymal stem cells-derived exosomes. J. Biomed. Sci. 2021, 28, 39. [Google Scholar] [CrossRef]
  159. McKiernan, J.; Donovan, M.J.; Margolis, E.; Partin, A.; Carter, B.; Brown, G.; Torkler, P.; Noerholm, M.; Skog, J.; Shore, N.; et al. A Prospective Adaptive Utility Trial to Validate Performance of a Novel Urine Exosome Gene Expression Assay to Predict High-grade Prostate Cancer in Patients with Prostate-specific Antigen 2-10ng/ml at Initial Biopsy. Eur. Urol. 2018, 74, 731–738. [Google Scholar] [CrossRef]
  160. Zhao, Z.; Fan, J.; Hsu, Y.S.; Lyon, C.J.; Ning, B.; Hu, T.Y. Extracellular vesicles as cancer liquid biopsies: From discovery, validation, to clinical application. Lab. A Chip 2019, 19, 1114–1140. [Google Scholar] [CrossRef]
  161. Chanteloup, G.; Cordonnier, M.; Isambert, N.; Bertaut, A.; Marcion, G.; Garrido, C.; Gobbo, J. Membrane-bound exosomal HSP70 as a biomarker for detection and monitoring of malignant solid tumours: A pilot study. Pilot. Feasibility Stud. 2020, 6, 35. [Google Scholar] [CrossRef] [PubMed]
  162. Tian, J.; Casella, G.; Zhang, Y.; Rostami, A.; Li, X. Potential roles of extracellular vesicles in the pathophysiology, diagnosis, and treatment of autoimmune diseases. Int. J. Biol. Sci. 2020, 16, 620–632. [Google Scholar] [CrossRef]
  163. Kumar, P.; Kumawat, R.K.; Uttam, V.; Behera, A.; Rani, M.; Singh, N.; Barwal, T.S.; Sharma, U.; Jain, A. The imminent role of microRNAs in salivary adenoid cystic carcinoma. Transl. Oncol. 2023, 27, 101573. [Google Scholar] [CrossRef] [PubMed]
  164. Snyder, M.; Iraola-Guzmán, S.; Saus, E.; Gabaldón, T. Discovery and Validation of Clinically Relevant Long Non-Coding RNAs in Colorectal Cancer. Cancers 2022, 14, 3866. [Google Scholar] [CrossRef] [PubMed]
  165. Mo, Z.; Cheong, J.Y.A.; Xiang, L.; Le, M.T.N.; Grimson, A.; Zhang, D.X. Extracellular vesicle-associated organotropic metastasis. Cell Prolif. 2021, 54, e12948. [Google Scholar] [CrossRef] [PubMed]
  166. Srivastava, A.; Amreddy, N.; Pareek, V.; Chinnappan, M.; Ahmed, R.; Mehta, M.; Razaq, M.; Munshi, A.; Ramesh, R. Progress in extracellular vesicle biology and their application in cancer medicine. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2020, 12, e1621. [Google Scholar] [CrossRef] [PubMed]
  167. Du, R.; Wang, C.; Zhu, L.; Yang, Y. Extracellular Vesicles as Delivery Vehicles for Therapeutic Nucleic Acids in Cancer Gene Therapy: Progress and Challenges. Pharmaceutics 2022, 14, 2236. [Google Scholar] [CrossRef]
Figure 1. The biogenesis of different multivesicular bodies (MVBs) and composition of EVs and their immunoregulatory roles in tumor microenvironment. (A). Biogenesis of various types of multivesicular bodies (MVBs). (1) The canonical MVB biogenesis starts at the plasma membrane, where cargos are sorted efficiently in an early endosome via endocytosis, which further matures into the multivesicular appearance of late endosomes (MVBs). (2) The recycling of endosomes during glutamine deprivation. (3) After sequestering a portion of the cytosol called a phagophore, it develops further into CD63–positive and LC3–positive MVBs called autophagosomes that fuse with multivesicular bodies (MVBs) that originate from late endosome through endocytosis of plasma membrane and form amphisomes (4,5) that release into extracellular space via exocytosis. (6) Autophagosomes fuse with lysosomes to form autolysosomes and release AEVs into the extracellular space in a caspase–3–dependent manner. (7) The budding of mitochondria into mitochondria–derived vesicles (MDVs) and their fusion with MVBs, followed by the release of mitochondria–derived vesicle (MDV) exosomes containing mitochondrial proteins and mitochondrial DNA (mtDNA). (8) Nuclear–envelope–derived MVBs (NEMVBs) are produced via the outward budding of the inner nuclear envelope, which is followed by the release of exosomes that contain leukotriene B4 (LTB4). (B) General structure of exosomes with the surface markers of an exosome. Exosomes originate from MVBs and carry different noncoding RNA, including miRNA, lncRNA, and circRNA; proteins (such as HSP 60, 70, and 90); and so on. Additionally, exosomes also carry different molecules that provide them immune tolerance (i.e., help them avoid being destroyed by the immune system and complement), such as CD47, CD55, and CD59, which prevent exosomes from being destroyed by the complement cascade. HLA–G and Fas/FasL molecules also protect exosomes from being destroyed by cytotoxic T and NK cells. These contents have a significant impact on tumor development. (C) In various microenvironments (e.g., low pH, hypoxic conditions, starvation, and ER stress), cancer cells release exosomes and may induce autophagy in recipient cells, which can further induce cell proliferation, EMT, and chemoresistance due to a significant impact on different components of the tumor microenvironment, like neutrophils activation, macrophage polarization, fibroblast differentiation into CAFs, and promote premetastatic niche formation. Red arrow indicate increased (upward) level.
Figure 1. The biogenesis of different multivesicular bodies (MVBs) and composition of EVs and their immunoregulatory roles in tumor microenvironment. (A). Biogenesis of various types of multivesicular bodies (MVBs). (1) The canonical MVB biogenesis starts at the plasma membrane, where cargos are sorted efficiently in an early endosome via endocytosis, which further matures into the multivesicular appearance of late endosomes (MVBs). (2) The recycling of endosomes during glutamine deprivation. (3) After sequestering a portion of the cytosol called a phagophore, it develops further into CD63–positive and LC3–positive MVBs called autophagosomes that fuse with multivesicular bodies (MVBs) that originate from late endosome through endocytosis of plasma membrane and form amphisomes (4,5) that release into extracellular space via exocytosis. (6) Autophagosomes fuse with lysosomes to form autolysosomes and release AEVs into the extracellular space in a caspase–3–dependent manner. (7) The budding of mitochondria into mitochondria–derived vesicles (MDVs) and their fusion with MVBs, followed by the release of mitochondria–derived vesicle (MDV) exosomes containing mitochondrial proteins and mitochondrial DNA (mtDNA). (8) Nuclear–envelope–derived MVBs (NEMVBs) are produced via the outward budding of the inner nuclear envelope, which is followed by the release of exosomes that contain leukotriene B4 (LTB4). (B) General structure of exosomes with the surface markers of an exosome. Exosomes originate from MVBs and carry different noncoding RNA, including miRNA, lncRNA, and circRNA; proteins (such as HSP 60, 70, and 90); and so on. Additionally, exosomes also carry different molecules that provide them immune tolerance (i.e., help them avoid being destroyed by the immune system and complement), such as CD47, CD55, and CD59, which prevent exosomes from being destroyed by the complement cascade. HLA–G and Fas/FasL molecules also protect exosomes from being destroyed by cytotoxic T and NK cells. These contents have a significant impact on tumor development. (C) In various microenvironments (e.g., low pH, hypoxic conditions, starvation, and ER stress), cancer cells release exosomes and may induce autophagy in recipient cells, which can further induce cell proliferation, EMT, and chemoresistance due to a significant impact on different components of the tumor microenvironment, like neutrophils activation, macrophage polarization, fibroblast differentiation into CAFs, and promote premetastatic niche formation. Red arrow indicate increased (upward) level.
Cancers 15 04879 g001
Figure 2. An apoptotic bodies (between 0.05–5 µm in diameter), which is produced by dying cells during the later stages of apoptosis, is released into the extracellular space, and eliminated by neighboring cells.
Figure 2. An apoptotic bodies (between 0.05–5 µm in diameter), which is produced by dying cells during the later stages of apoptosis, is released into the extracellular space, and eliminated by neighboring cells.
Cancers 15 04879 g002
Figure 3. TNBC subtypes, their general characteristics, and treatment measures. Based on gene expression profiles, TNBC is categorized into six subtypes, i.e., BL–1, BL–2, IM, LAR, M, and MSL. Higher expressions of genes related to the cell cycle and the DNA damage pathway are linked to BL–1. PARP inhibitors and platinum compounds are used to treat BL–1 (dark green arrow). BL–2 is accompanied by higher expressions of growth–factor–signaling and metabolic–pathway–associated genes and can be treated with growth–signaling inhibitors (dark red arrow). IM is related to elevated expression of genes associated with immune cellular processes; the treatment measures include immune checkpoint inhibitors (blue arrow). LAR, on the other hand, is associated with higher relative expression of hormonal regulation and metabolic pathway genes and can be targeted by androgen receptor agonists (purple arrow). Elevated expressions of cell motility, differentiation, and EMT genes are observed in MC, which can be treated with Wnt/β–catenin pathway, PI3K/mTOR pathway, and TGFβ receptor kinase inhibitors (dark yellow arrow). MSL is very similar to M; however, claudin expression was shown to be significantly lower in MSL compared with M (violet arrow).
Figure 3. TNBC subtypes, their general characteristics, and treatment measures. Based on gene expression profiles, TNBC is categorized into six subtypes, i.e., BL–1, BL–2, IM, LAR, M, and MSL. Higher expressions of genes related to the cell cycle and the DNA damage pathway are linked to BL–1. PARP inhibitors and platinum compounds are used to treat BL–1 (dark green arrow). BL–2 is accompanied by higher expressions of growth–factor–signaling and metabolic–pathway–associated genes and can be treated with growth–signaling inhibitors (dark red arrow). IM is related to elevated expression of genes associated with immune cellular processes; the treatment measures include immune checkpoint inhibitors (blue arrow). LAR, on the other hand, is associated with higher relative expression of hormonal regulation and metabolic pathway genes and can be targeted by androgen receptor agonists (purple arrow). Elevated expressions of cell motility, differentiation, and EMT genes are observed in MC, which can be treated with Wnt/β–catenin pathway, PI3K/mTOR pathway, and TGFβ receptor kinase inhibitors (dark yellow arrow). MSL is very similar to M; however, claudin expression was shown to be significantly lower in MSL compared with M (violet arrow).
Cancers 15 04879 g003
Figure 4. The role of EVs in the growth and metastasis of human TNBC. Growth: (1) TNBC–EVs induce growth and drug resistance in normal epithelial cells. (2) Plant derivative–induced TNBC cell–derived EVs inhibit TNBC proliferation. (3) TNBC–EVs also enhance glycolysis in CAFs. (4) MSC–EVs promote TNBC progression. (5) CAF–EVs also induce TNBC proliferation. (6) TNBC–EVs often trigger the growth of TNBC in an autocrine manner. Metastasis: (7) In response to chemotherapy, TNBC cells release EVs, which act on endothelial cells to promote transendothelial migration of TNBC cells. (8) Serum–EVs of TNBC patients promote TNBC metastasis. (9) TNBC–EVs also promote metastasis of non–metastatic breast cancer cells. (10) TNBC–EVs also enhance vascular permeability upon acting on endothelial cells. (11) Serum–EVs of TNBC patients again trigger proliferation, migration, and metastasis of TNBC cells. The green dotted arrows indicate phenomena associated with TNBC progression, whereas red dotted arrow denotes prevention of TNBC progression.
Figure 4. The role of EVs in the growth and metastasis of human TNBC. Growth: (1) TNBC–EVs induce growth and drug resistance in normal epithelial cells. (2) Plant derivative–induced TNBC cell–derived EVs inhibit TNBC proliferation. (3) TNBC–EVs also enhance glycolysis in CAFs. (4) MSC–EVs promote TNBC progression. (5) CAF–EVs also induce TNBC proliferation. (6) TNBC–EVs often trigger the growth of TNBC in an autocrine manner. Metastasis: (7) In response to chemotherapy, TNBC cells release EVs, which act on endothelial cells to promote transendothelial migration of TNBC cells. (8) Serum–EVs of TNBC patients promote TNBC metastasis. (9) TNBC–EVs also promote metastasis of non–metastatic breast cancer cells. (10) TNBC–EVs also enhance vascular permeability upon acting on endothelial cells. (11) Serum–EVs of TNBC patients again trigger proliferation, migration, and metastasis of TNBC cells. The green dotted arrows indicate phenomena associated with TNBC progression, whereas red dotted arrow denotes prevention of TNBC progression.
Cancers 15 04879 g004
Figure 5. TF–FVIIa–driven PAR2 activation triggers EV generation from human TNBC cells, which promotes EMT in non–metastatic breast cancer cells, leading to the induction of proliferation, migration, invasion, and anti–apoptosis. TF–FVIIa–mediated cleavage of PAR2 in human TNBC cells leads to the intracellular activation of AKT, ROCK, and ERK1/2 pathways independently, which promotes the release of EVs packaged with miR–221 via actomyosin reorganization. TNBC–EVs are taken up by non–metastatic breast cancer cells, thereby delivering miR–221 into the recipient cells. In recipient cells, miR–221 targets PTEN, leading to the activation of AKT/p65–signaling pathway in which the activated (via phosphorylation) p65 enters the nucleus to stimulate the upregulation of N–cadherin and vimentin while downregulating the expression of E–cadherin. As a result of this, EMT is induced, which ultimately leads to the promotion of proliferation, migration, invasion, and anti–apoptosis of EV–fused recipient cells. Red arrows indicate decreased and green arrow indicate increased levels.
Figure 5. TF–FVIIa–driven PAR2 activation triggers EV generation from human TNBC cells, which promotes EMT in non–metastatic breast cancer cells, leading to the induction of proliferation, migration, invasion, and anti–apoptosis. TF–FVIIa–mediated cleavage of PAR2 in human TNBC cells leads to the intracellular activation of AKT, ROCK, and ERK1/2 pathways independently, which promotes the release of EVs packaged with miR–221 via actomyosin reorganization. TNBC–EVs are taken up by non–metastatic breast cancer cells, thereby delivering miR–221 into the recipient cells. In recipient cells, miR–221 targets PTEN, leading to the activation of AKT/p65–signaling pathway in which the activated (via phosphorylation) p65 enters the nucleus to stimulate the upregulation of N–cadherin and vimentin while downregulating the expression of E–cadherin. As a result of this, EMT is induced, which ultimately leads to the promotion of proliferation, migration, invasion, and anti–apoptosis of EV–fused recipient cells. Red arrows indicate decreased and green arrow indicate increased levels.
Cancers 15 04879 g005
Figure 6. The role of EVs as a biomarker for TNBC. EV–carried proteins often serve as TNBC biomarkers. (1) AREF + TNBC–EVs promote breast cancer invasiveness. (2) UCHL1 + TNBC–EVs trigger migration and extravasation of breast cancer cells. (3) PTX–treated TNBC cells release Survivin + EVs, which promote the growth and survival of fibroblasts and breast cancer cells. (4) TNBC serum is enriched with CD151 + EVs, which induce migration and invasion of breast cancer. EV–transported miRNAs also contribute as a biomarker for TNBC. (5) Cisplatin–resistant TNBC–EVs carry miR–423–5p, which promotes migration, invasion, and anti–apoptosis of breast cancer cells. (6) TNBC–associated macrophage–derived EVs are positive for miR–223, which stimulates invasion of TNBC cells. (7) TNBC–EVs enriched with miR–10b also promote invasion of non–malignant cells. (8) miR–105–loaded TNBC–EVs increase endothelial permeability, thereby contributing to TNBC metastasis.
Figure 6. The role of EVs as a biomarker for TNBC. EV–carried proteins often serve as TNBC biomarkers. (1) AREF + TNBC–EVs promote breast cancer invasiveness. (2) UCHL1 + TNBC–EVs trigger migration and extravasation of breast cancer cells. (3) PTX–treated TNBC cells release Survivin + EVs, which promote the growth and survival of fibroblasts and breast cancer cells. (4) TNBC serum is enriched with CD151 + EVs, which induce migration and invasion of breast cancer. EV–transported miRNAs also contribute as a biomarker for TNBC. (5) Cisplatin–resistant TNBC–EVs carry miR–423–5p, which promotes migration, invasion, and anti–apoptosis of breast cancer cells. (6) TNBC–associated macrophage–derived EVs are positive for miR–223, which stimulates invasion of TNBC cells. (7) TNBC–EVs enriched with miR–10b also promote invasion of non–malignant cells. (8) miR–105–loaded TNBC–EVs increase endothelial permeability, thereby contributing to TNBC metastasis.
Cancers 15 04879 g006
Figure 7. The role of EVs in TNBC immune regulation. (1) TNBC–EVs are positive for CCL5, which triggers the conversion of macrophages into TEMs, leading to stromal remodeling and immune infiltration, ultimately contributing to TNBC metastasis. (2) TNBC–EVs promote M2 polarization of macrophages, which leads to lymph node metastasis of TNBC. (3) Activated T–cells release PD–1+ EVs that bind to PD–L1, either on TNBC cells or TNBC–EVs (defined as green dotted arrows), thereby preventing the PD–1/PD–L1–dependent interaction of T–cells and TNBC cells (shown as red dotted arrows), because of which TNBC–mediated T–cell suppression is reduced. (4) OS triggers the release of PD–L1+ EVs from TNBC cells, which not only enrich MDSCs and M2 macrophages in the TME but also reduce the population of CD8+ T–cells.
Figure 7. The role of EVs in TNBC immune regulation. (1) TNBC–EVs are positive for CCL5, which triggers the conversion of macrophages into TEMs, leading to stromal remodeling and immune infiltration, ultimately contributing to TNBC metastasis. (2) TNBC–EVs promote M2 polarization of macrophages, which leads to lymph node metastasis of TNBC. (3) Activated T–cells release PD–1+ EVs that bind to PD–L1, either on TNBC cells or TNBC–EVs (defined as green dotted arrows), thereby preventing the PD–1/PD–L1–dependent interaction of T–cells and TNBC cells (shown as red dotted arrows), because of which TNBC–mediated T–cell suppression is reduced. (4) OS triggers the release of PD–L1+ EVs from TNBC cells, which not only enrich MDSCs and M2 macrophages in the TME but also reduce the population of CD8+ T–cells.
Cancers 15 04879 g007
Figure 8. The role of EVs in TNBC immunotherapy. (A) NK–EVs in TNBC immunotherapy. EVs are shown to be enriched with FasL, Perforin, and Granzyme, which trigger the killing of TNBCs. (B) EVs’ miRNAs in TNBC immunotherapy. (1) let–7–containing EVs with aptamer (AS1411) modification target nucleolin + TNBC cells, resulting in the downregulation of TNBC growth. (2) Bone marrow stromal cell–derived EVs are abundant with miR–127, –197, –222, and –223, which reduce TNBC proliferation. (3) Adipose–tissue–derived MSCs release miR–424–5p–bearing EVs, which promote TNBC apoptosis. (4) MSC–EVs are rich in miR–381, which reduces the metastatic potential of TNBC.
Figure 8. The role of EVs in TNBC immunotherapy. (A) NK–EVs in TNBC immunotherapy. EVs are shown to be enriched with FasL, Perforin, and Granzyme, which trigger the killing of TNBCs. (B) EVs’ miRNAs in TNBC immunotherapy. (1) let–7–containing EVs with aptamer (AS1411) modification target nucleolin + TNBC cells, resulting in the downregulation of TNBC growth. (2) Bone marrow stromal cell–derived EVs are abundant with miR–127, –197, –222, and –223, which reduce TNBC proliferation. (3) Adipose–tissue–derived MSCs release miR–424–5p–bearing EVs, which promote TNBC apoptosis. (4) MSC–EVs are rich in miR–381, which reduces the metastatic potential of TNBC.
Cancers 15 04879 g008
Table 1. General characteristics of different types of EVs.
Table 1. General characteristics of different types of EVs.
Type of EVsMarker/sSize (Diameter)Biogenetic MechanismReference
Microparticles (MPs)Tetraspanins, CD41, CD63100 nm–1 µmEvagination of the plasma membrane followed by pinching to release the MPs outside the cells.[53,54]
ExosomesAlix, TSG101, HSP90β, HSC70, CD63, Syntenin 130–150 nmConventional mechanism of exosome biogenesis includes the invagination of the early endosomal membrane to form exosomes, which fuse with the plasma membrane to release the exosomes from the cells.[55,56,57]
Apoptotic bodiesHSP60, GRP78, Annexin V (phosphatidylserine)50 nm–5 µmCellular contraction is induced during apoptosis, which produces a hydrodynamic force that triggers the separation of the plasma membrane from the cytoskeleton, leading to the release of apoptotic bodies from the cells.[55,58,59]
Abbreviations: TSG101, tumor susceptibility gene 101; Alix, ALG–2–interacting protein X; HSP, heat shock protein; HSC70, heat shock cognate protein 70; CD, cluster of differentiation; GRP78, glucose–regulated protein 78.
Table 2. Subtyping of breast cancer, based on immunohistochemistry analysis.
Table 2. Subtyping of breast cancer, based on immunohistochemistry analysis.
SubtypeImmunohistochemistry ProfileOther FeaturesReferences
Luminal AER+, PR+, HER2−Ki–67−[76]
Luminal BER+, PR+, HER2+Ki–67+[76]
HER2 highER−, PR−, HER2+Ki–67+[81]
Normal–likeER+, PR+, HER2−Ki–67−[81]
Basal–like (TNBC)ER−, PR−, HER2−, basal markersKi–67+, (EGFR, CK5/6)[76]
Claudin low (TNBC)ER−, PR−, HER2−Low Ki–67, E–cadherin, Claudin 3, 4, 7[93]
Abbreviations: ER, estrogen receptor; PR, progesterone receptor; HER2, human epidermal growth factor receptor–2; TNBC, triple–negative breast cancer; EGFR, extracellular growth factor receptor; CK, cytokeratin; −, negative; +, positive.
Table 3. The role of EVs in the growth and metastasis of human TNBC.
Table 3. The role of EVs in the growth and metastasis of human TNBC.
TNBC PropertyMechanism of ActionReferences
GrowthHCC1806–EVs promote the growth and drug resistance to MCF10A cells via the involvement of PI3K/AKT, MAPK, and HIF1A pathways.[107]
DET– and DETD–35–treated MDA–MB–231–derived EVs inhibit the proliferation of MDA–MB–231 cells by downregulating cell adhesion, migration, and angiogenesis.[108]
ITGB4 + EVs from MDA–MB–231 cells trigger the glycolytic pathway of CAFs via BNI3PL–dependent mitophagy and lactate production, which, in turn, promote tumor growth.[109]
MSC–EVs transfer miR–106a–5p to the TNBC tumor and induce tumor progression via downregulating HAND2–AS1.[110]
CAF–EVs show a relatively lower expression of miR–4516, which promotes the proliferation of TNBC cells by upregulating the miR–4516 target, namely, FOSL1 expression.[111]
CircHIF1A + EVs from human TNBC promote tumor growth via the upregulation of the PI3K/AKT pathway while downregulating p21.[112]
MetastasisIn response to chemotherapy, TNBC cells release a significant number of PS + EVs, which induce endothelial barrier permeability, hence helping with the transendothelial migration of cancer cells, contributing to TNBC metastasis.[113]
Serum EVs of TNBC patients are enriched with CD151, which triggers the migration and invasion of TNBC cells.[114]
Circulating EVs of TNBC patients are enriched with SPANXB1, which downregulates SH3GL2 expression in TNBC cells, and upregulates Rac1, FAK, and α–actinin expression, leading to TNBC metastasis.[115]
MDA–MB–231–EVs are abundant with miR–9 and miR–155, which target PTEN and DUSP14 in MCF–7, thereby inducing the metastatic potential of recipient MCF–7 cells.[116]
TNBC–EVs are enriched with miR–393, which targets VE–cadherin in endothelial cells, leading to enhanced vascular leakage, and thereby facilitating transendothelial migration of tumor cells.[117]
Serum EVs of TNBC patients are enriched with circPSMA1, which absorbs miR–637 to release the inhibitory function on AKT1 and leads to the downstream activation of β–catenin and cyclin D1 to trigger the proliferation, migration, and metastasis of TNBC cells.[118]
Abbreviations: PI3K, phosphoinositide 3–kinase; MAPK, mitogen–activated protein kinase; HIF1A, hypoxia–inducible factor 1α; DET, deoxyelephantopin; DETD–35, DET derivative 35; ITGB4, integrin β4; CAF, cancer–associated fibroblast; BNI3PL, B–cell leukemia/lymphoma 2 protein (Bcl–2) interacting protein 3 like; MSC, mesenchymal stem cell; HAND2–AS1; heart and neural crest derivatives expressed 2 antisense RNA 1; FOSL1, Fos like 1; PS, phosphatidylserine; CD, cluster of differentiation; SPANXB1, sperm protein associated with the nucleus on the X chromosome (SPANX) family member B1; SH3GL2, SH3 domain containing GRB2 like 2; Rac1, Ras–related C3 botulinum toxin substrate 1; FAK, focal adhesion kinase; PTEN, phosphatase and tensin homolog; DUSP14; dual specificity phosphatase 14; VE–cadherin, vascular endothelial cadherin; PSMA1, proteasome 20S subunit α1.
Table 4. The role of EVs as a biomarker for human TNBC.
Table 4. The role of EVs as a biomarker for human TNBC.
EVs’ Cargo TypeName of the CargoOriginFunctionReferences
ProteinEGFR ligandsMDA–MB–231EVs’ EGFR ligands, such as AREG, promote the invasiveness of recipient breast cancer cells.[121]
UCHL1TNBC cells, TNBC plasmaEV–carried UCHL1 protects the TGFβ type I receptor and SMAD2 from ubiquitination, stimulating the migration and extravasation of the breast cancer cells.[122]
SurvivinMDA–MB–231PTX–treated TNBC–derived EVs are enriched with Survivin, which promotes the growth and survivability of PTX–exposed fibroblasts and other breast cancer cells.[123]
CD151TNBC serumEVs from TNBC serum are enriched with CD151, which promotes the migration and invasion of TNBC cells.[114]
miRNAmiR–423–5pMDA–MB–231EVs from cisplatin–resistant TNBC cells are positive for miR–423–5p, which induces P–gp expression, migration, invasion, and anti–apoptosis in other breast cancer cells.[124]
miR–223MDA–MB–231TNBC–associated macrophages release miR–223–containing EVs, which promote the invasion of breast cancer.[125]
miR–10bTNBC cellsmiR–10b + TNBC–EVs target HOXD10 and KLF4 in non–malignant cells to promote cell invasion.[126]
miR–105MDA–MB–231miR–105 + EVs target ZO–1 in endothelial cells, leading to increased vascular permeability and facilitating the metastasis of TNBC.[127]
Abbreviations: EGFR, extracellular growth factor receptor; AREG, amphiregulin; UCHL1, ubiquitin C–terminal hydrolase 1; TGFβ, transforming growth factor beta; SMAD2, mothers against decapentaplegic homolog 2; PTX, paclitaxel; CD, cluster of differentiation; HOXD10, homeobox D10; KLF4, Krüppel–like factor 4; ZO–1, zonula occludens protein 1.
Table 5. The role of EVs in immune regulation of TNBC.
Table 5. The role of EVs in immune regulation of TNBC.
Donor CellsRecipient CellsEVs’ CargoFunctionReferences
TNBCMacrophagesCCL5TNBC–EVs transport CCL5 to macrophages leading to the development of TEMs, which induce stromal remodeling and immune infiltration to facilitate TNBC metastasis.[129]
TNBCMacrophages TNBC–EVs trigger macrophage polarization into M2 phenotypes, leading to LN metastasis.[130]
Activated T–cellsTNBC cells or TNBC–EVsPD–1Activated T–cell–derived EVs release PD–1 + EVs, which bind to PD–L1 + TNBC cells or EVs, thereby preventing T–cell/TNBC cell interaction and attenuating immune suppression of T–cells by TNBC cells.[131]
TNBCMDSCs, M2– macrophages, CD8 + T–cellsPD–L1OS stimulates the release of PD–L1 + EVs from TNBC cells, which enriches MDSCs and M2 macrophages in the TME and reduces CD8 + T–cells.[132]
Abbreviations: CCL5, C–C motif chemokine ligand 5; M2 macrophage, type 2 macrophage; LN, lymph node; PD–1, programmed death 1; PD–L1, programmed death ligand 1; MDSC; myeloid–derived suppressor cell; OS, oscillatory strain; TME, tumor microenvironment; CD, cluster of differentiation.
Table 6. EVs in clinical trials in different types of cancer.
Table 6. EVs in clinical trials in different types of cancer.
Trial NameNCT NumberCharacteristicsCancer TypesEVs’ SourceStatusOutcome MeasuresRefs.
Clinical Validation of an Urinary Exosome Gene Signature in Men Presenting for Suspicion of Prostate CancerNCT02702856Duration: May 2014–June 2015
Population: 2000 men
Age: >50
Prostate cancerUrineCompletedThe ExoDx Prostate IntelliScore (EPI) EPI score was associated with low–risk pathology post–RP, with potential implications on informing AS decisions.[154,155]
Diagnostic Accuracy of Circulating Tumor Cells (CTCs) And Onco–exosome Quantification in the Diagnosis of Pancreatic Cancer—PANC–CTC (PANC–CTC)NCT03032913Duration: February 2017–November 2017
Population: 20 with PDAC and 20 controls
Age: 18 years and older
Pancreatic ductal adenocarcinomaCirculationCompleted [156]
microRNAs Role in Pre–eclampsia
Diagnosis
NCT03562715Duration: November 2016–December 2017
Population:
100 patients and 100 controls Females
Age: 23 years to 35 years
PreeclampsiaCirculationCompletedLiquid biopsy combining several biomarkers could provide a rapid, reliable, noninvasive decision–making tool in early, potentially curable pancreatic cancer.[157,158]
Clinical Evaluation of the “ExoDx Prostate IntelliScore” (EPI)NCT03031418Duration: September 2016–September 2018
Population: 532 Patients
Age: above 50 years of age
Males
Prostate cancerUrineCompletedThe expression of miRNAs 136, 494, and 495 in exosomes of peripheral blood and UCMSCs conditioned media.[159]
Olmutinib Trial in T790M(+)NSCLC Patients Detected by Liquid Biopsy Using BALF Extracellular Vesicular DNANCT03228277Duration: July 2017–July 2019
Population: 25
Males or females, aged at least 19 years.
NSCLCBALFCompletedExoDx Prostate (IntelliScore) test can predict ≥ GG2 PCa at initial biopsy and defer unnecessary biopsies better than existing risk calculators and standard clinical data.[160]
Pilot Study with the Aim to Quantify a Stress Protein in the
Blood and in the Urine for the Monitoring and Early Diagnosis of Malignant Solid Tumors (EXODIAG)
NCT02662621Duration: December 2015–April 2019
Population: 71 patients
Age: 18 years and older
CancerCirculationCompletedAssess the anti–tumor efficacy via objective response rate (ORR), disease control rate (DCR), and progression–free survival (PFS).[161]
Pimo Study: Extracellular Vesicle–based Liquid Biopsy to Detect cancer Hypoxia in TumorsNCT03262311Duration: November 2017–September 2019
Population: 21
Age: ≥18 years
Hypoxia–induced cancerBloodCompletedHSP70 exosomes could be a powerful tool to diagnose cancer and guide.[162]
The Sensitivity and Specificity of Using Salivary miRNAs in Detection of Malignant
Transformation of Oral Lesions
NCT04913545Duration: January 2020–August 2020
Population: 18
Age: 35 years to 70 years
Oral premalignant lesionsSalivaCompletedClinicians in therapeutic decision–making, improving patient care.[163]
Clinical Evaluation of ExoDx Prostate (IntelliScore) in Men Presenting for Initial Prostate BiopsyNCT04720599Duration: June 2020–June 2021
Population: 120 males
Age: 50 years and older
Urologic cancerUrineCompletedNot available.
Serum Exosomal Long Noncoding RNAs as Potential Biomarkers for Lung Cancer DiagnosisNCT03830619Duration: Jan 2017–July 2021
Population: 1000
Age: 18 years to 75 years
Location: Hubei, China
Lung cancerSerumCompletedMeasuring the sensitivity and specificity of using the salivary miRNAs (412,512) to detect the malignant transformation in potentially malignant lesions.[164]
Identification of New Diagnostic Protein Markers for Colorectal Cancer (EXOSCOL01)NCT03895216Duration: December 2018–December 2021
Population: 34
Age: 18 years and older
Bone metastasisPlasmaCompletedEPI–CE provides information beyond standard clinical parameters and provides a better risk assessment prior to MRI of patients suspected of prostate cancer than the commonly used multiparametric risk calculators.[165]
Exosomes Implication in PD1–PD–L1 Activation in OSAS (ExoSAS)NCT03811600Duration: March 2019–October 2020
Population: 90
Age: 18 years and older
Cancer, obstructive
sleep apnea
PlasmaCompletedNo study results are posted on ClinicalTrials.gov for this study.[166]
Abbreviations: NSCLC, non–small–cell lung cancer; BALF, bronchoalveolar fluid; CRC, colorectal cancer.
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

Das, K.; Paul, S.; Ghosh, A.; Gupta, S.; Mukherjee, T.; Shankar, P.; Sharma, A.; Keshava, S.; Chauhan, S.C.; Kashyap, V.K.; et al. Extracellular Vesicles in Triple–Negative Breast Cancer: Immune Regulation, Biomarkers, and Immunotherapeutic Potential. Cancers 2023, 15, 4879. https://doi.org/10.3390/cancers15194879

AMA Style

Das K, Paul S, Ghosh A, Gupta S, Mukherjee T, Shankar P, Sharma A, Keshava S, Chauhan SC, Kashyap VK, et al. Extracellular Vesicles in Triple–Negative Breast Cancer: Immune Regulation, Biomarkers, and Immunotherapeutic Potential. Cancers. 2023; 15(19):4879. https://doi.org/10.3390/cancers15194879

Chicago/Turabian Style

Das, Kaushik, Subhojit Paul, Arnab Ghosh, Saurabh Gupta, Tanmoy Mukherjee, Prem Shankar, Anshul Sharma, Shiva Keshava, Subhash C. Chauhan, Vivek Kumar Kashyap, and et al. 2023. "Extracellular Vesicles in Triple–Negative Breast Cancer: Immune Regulation, Biomarkers, and Immunotherapeutic Potential" Cancers 15, no. 19: 4879. https://doi.org/10.3390/cancers15194879

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