Next Article in Journal
Phosphasilazanes as Inhibitors for Respirable Fiber Fragments Formed during Burning of Carbon-Fiber-Reinforced Epoxy Resins
Next Article in Special Issue
Chitosan-Coated Solid Lipid Nanoparticles as an Efficient Avenue for Boosted Biological Activities of Aloe perryi: Antioxidant, Antibacterial, and Anticancer Potential
Previous Article in Journal
Orbital Polarization-Dependent Fragment Twist-Induced Intramolecular Electric-Field-Driven Charge Transfer
Previous Article in Special Issue
Phytochemicals from Vanda bensonii and Their Bioactivities to Inhibit Growth and Metastasis of Non-Small Cell Lung Cancer Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Critical Review on the Different Roles of Exosomes in TNBC and Exosomal-Mediated Delivery of microRNA/siRNA/lncRNA and Drug Targeting Signalling Pathways in Triple-Negative Breast Cancer

by
Manosi Banerjee
and
Vijayarangan Devi Rajeswari
*
School of Bioscience and Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
*
Author to whom correspondence should be addressed.
Molecules 2023, 28(4), 1802; https://doi.org/10.3390/molecules28041802
Submission received: 26 November 2022 / Revised: 23 December 2022 / Accepted: 12 January 2023 / Published: 14 February 2023
(This article belongs to the Special Issue Natural Products in Anticancer Activity)

Abstract

:
Triple-negative breast cancer is the most potent metastatic type of breast cancer that can spread to other body parts. Chemotherapy and surgical intervention are the sole treatments for TNBC, owing to the scarcity of therapeutic targets. Manipulation of the membranes as per the desired targets of exosomes has recently gained much attention as a drug delivery method. Despite their known roles in different diseases, very few studies have focused on signalling that triggers the metastasis of triple-negative breast cancer to other body parts by exosomes. This article highlights the significant roles of exosomes associated with TNBC, the involvement of exosomes in breast cancer diagnosis, progression, and the treatment of triple-negative breast cancer by the exosomes as a drug delivery system. This review paper also illustrates the role of exosomes in initiating EMT in breast cancer, including novel signalling.

Graphical Abstract

1. Introduction

1.1. Triple Negative Breast Cancer

Breast cancer is the leading cancer in women, accounting for 27% of all cancers in India [1]. TNBC subtypes account for 15–20% of breast cancer [2]. Mutation in BRCA1/2 causes breast cancer. TNBC is aggressive and eventually transfers to other body organs such as the lungs, brain, heart, bone, and liver [3]. The survival rate (5 years) for triple-negative breast cancer patients is low (77% in contrast to 93% for other breast cancer subtypes) [4]. The receptor’s expression profile in TNBC is very complicated and contradictory to what its name suggests. The lack of phenotypic receptors, such as estrogen and progesterone, reduces the expression of human epidermal growth factor 2. Molecular subtypes of triple-negative breast cancer are (i) basal-like 1, (ii) basal-like 2, (iii) immunomodulatory, (iv) mesenchymal, (v) mesenchymal stem-like, and (vi) luminal androgen receptor [5,6]. The prediction of a 5-year survival rate can be more accurate depending on the subtyping of TNBC. The current statistics show that it differs in basal-like tumours (68%) in contrast to non-basal-like tumours (79%). The effect of chemotherapy is different in different subtypes of breast cancer [7].
Clinically, treatment for TNBC is very restricted due to the lack of specific targets [8]. The dearth of effective targeted delivery leads to poor prognosis and treatment outcomes [9]. Hence, side effects are associated with conventional chemotherapy, which can simultaneously be fatal and distressing, but it is the sole option available [10]. Chemotherapeutics have a small therapeutic window when used together [11]. Furthermore, most of them cannot penetrate the blood–brain barrier (BBB), limiting therapeutic options for brain tumours, particularly those caused by metastasis of TNBC [12,13]. Regrettably, even when the first line of therapy is there, the recurrent rate is much higher and occurs within three years.

1.2. Need of Delivery Vehicle—Exosomes as an Answer

Restrictions on anticancer drug potency are due to poor drug solubility, half-life, and a drug-resistant result of inadequate binding of the drugs with the specific ligands of the tumours [14]. To find a proper solution to all these clinical challenges, testing of nano-formulations such as hydrogels, microneedle patches, biodegradable polymers, microspheres, and nanoparticles has been under research [15]. The ability of the nano-formulations was proved in pre-clinical trials, but off-target effects and toxicity have disrupted their role in the clinical trials [16]. All challenges described above justify the need for new and improved nanocarriers with adequate properties such as low toxicity, longer half-life, and biocompatibility. Developing engineered nanocarriers can be a solution that can target specific ligands [17]. To fulfil these demands, scientists have devised extracellular vesicles, i.e., exosomes, as the delivery system for various multiple chemotherapeutics. The toxicity of other nano-formulations makes exosomes efficient delivery agents with a small size and more biocompatibility. Exosomes have better pharmacokinetics and pharmacodynamics compared to other nano-delivery systems [18]. The exosomes can even cross the blood barrier and can target tumours in the brain. Hence, when TNBC metastasizes to the brain, exosomes can quickly cross the BBB and target it [19]. The exosomes are derived from different biological sources and act not too similarly in vivo. The cellular origins of exosomes are significant [20]. Drug loading capacity also depends on the source from which exosomes are extracted. Extracted exosomes from macrophages show higher loading efficiency than those derived from pancreatic cells [21,22]. Exosomes separated from donor cells that are not the same as the recipient cells are more effective as delivery vehicles. Exosomes isolated from particular cell types exhibit organotropism toward various cancers, accumulating mostly in the organ’s primary cell type rather than in adjoining cell types [23].

2. Extracellular Vesicles

Intercellular communication in all cells results from the extracellular vesicles produced by almost all cell types. EVs can be divided into three categories, i.e., microvesicles (MVs), exosomes, and apoptotic bodies, depending upon the size and the generation of pathways [24]. Among them, exosomes regulate the normal functioning of cells. They are also involved in the pathology of several diseases, including neurodegenerative and autoimmune, cardiovascular, and cancer [25,26,27,28]. These features have drawn much attention to exosomes; therefore, they have been widely researched in cancer biology and therapy. The size of exosomes is 30–150 nm, synthesized by the endocytic process and produced by the fusion of multi-vesicular bodies (MVBs) with the plasma membrane [29].
Exosomes are overexpressed with distinguished membrane proteins called tetraspanins and proteins such as CD9, CD37, CD63, CD81, and CD82. Tumour susceptibility gene (TSG)-101 and Alix of the exosome sorting complex required for transport (ESCRT) are some of the exosome’s biomarkers [30]. The ever-increasing literature suggests that exosomes regulate the normal functioning of cells. The exosome components (lipids, nucleic acids, metabolites, proteins, and peptides) reflect their cellular origin. Various databases have all the detailed compositions of exosomes to characterize them properly, such as ExoCarta, Vesiclepedia, and EV miRNA [31,32,33].

3. Exosomes in TNBC

3.1. Exosomes Carry Signature Markers from Their Cells of Origin

The number of exosomes released by TNBC (triple-negative breast cancer) cells is more incredible than more significant normal healthy cells. The augmentation of TSAP6, a p53-regulated gene product that governs the exosomal secretion signalling transcription by activating p53, which is frequently aberrantly driven in cancer cells, could alter the rise in exosome shedding from cancer cells. The most associated biomarkers with exosomes are tetraspanins (CD9, CD63, CD81), proteins of the endosome system (TSG101, Rab-GTPase), and heat shock chaperones (HSP70, HSP90). These are all the characterizations of all exosomes [34,35,36].
Exosomes produced by cancer cells have different content than normal cells. The microRNA content in the exosomes extracted from cancer cells has different signature microRNA and acts similar to “fingerprints”, which can identify their origin. Hence, they can also serve as a biomarker. A protein senescence-associated secretory phenotype (SASP) that can enhance tumour growth is one of the constituents of exosomes [37].
Chemotherapeutics induce apoptosis. Some cancer cells experience therapeutic-induced senescence (TIS), which enables them to stay metabolically active but lose their capacity to metastasize. This condition makes them more resistant to chemotherapies. Kavanagh et al. found that exosomes collected from TIS TNBC CAL51 cells had much larger quantities than non-senescent cells. Concerning non-senescent TNBC cells, the researchers found a remarkable increase in the amounts of 142 proteins in exosomes from these TIS cells. Essential proteins, which are all involved in (i) cell proliferation, (ii) ATP depletion, (iii) apoptosis, and (iv) SASP factors, are more prevalent in exosomes from TIS cells, implying that the abolition of these proteins from TIS cells through exosomes permits cancer senescent cells to stay feasible [38].
Exosomes also play an important role as a messenger and can communicate between the recipient cells and donor cells through the microRNA, which they carry, which becomes translated into proteins. Communication can involve immune suppression, microenvironment changes, angiogenesis, immune escape and invasion, and tumour progression and repression.

3.2. Exosomes for Diagnosis of Triple-Negative Breast Cancer

With the advancement of nano-theragnostics, exosome profiling, diagnosis, and potential disease therapy are possible. A ‘liquid biopsy’ enables the study of EVs and exosomes. It also aids in analysing circulating DNA from tumours and cells from tumours. The exosomes carry specific biomarkers that help to diagnose triple-negative breast cancer. The content of the exosomes is different for patients with triple-negative breast cancer [39]. Proteins that act as biomarkers for TNBC are carcinoembryonic antigen (CEA), survivin, and CA 15-3(cancer antigen 15-3) [40]. The levels of these biomarkers are elevated in patients suffering from TNBC. The microRNA present inside the exosomes also plays a vital role in diagnosing. A specific microRNA identified as a biomarker is miR-373, which is elevated in TNBC patients and further downregulates the expression of estrogen [41]. The Wnt signalling is responsible for metastasis and chemoresistance in TNBC. Myotubularin-related protein 3 is downregulated by miR-1910-3p, which also stimulates the NF-κB and wnt/β-catenin signalling pathways and, in turn, accelerates the development of breast cancer. When combined with the conventional tumour marker CA153, serum miR-1910-3p in exosomes is a potent diagnostic sign that increases the sensitivity of breast cancer detection. In conclusion, miR-1910-3p in serum exosomes may be a new molecular indicator for the detection of breast cancer. Liquid biopsy investigates intra-tumour heterogenicity and the tumour microenvironment, while single-tissue biopsy does not include the microenvironment [42,43] Figure 1.

3.3. Exosomes Initiating the Epithelial-Mesenchymal Transition in Breast Cancer and TNBC Influencing the Metastasis to Other Organs

EMT is a process through which normal cells transfer to mesenchymal cells. Mesenchymal cells have the property of invasion and malignancy, leading to the worst prognosis and TNBC migrating to other organs. The EMT transcription factors involved are β-catenin, TWIST, zinc finger protein, SNAIL2 (zinc finger protein SNAI 1), SLUG (SNAI 2 gene), ZEB1 (zinc finger E-box-binding homeobox 1), and ZEB 2. Studies have proven that altering these transcription factors is not enough to cease the epithelial–mesenchymal transition. The exosomes are involved in intercommunication between the cancer cells. The exosomes have various contents, which can regulate the transcription involved in the epithelial-to-mesenchymal transition [44]. The literature, with various reports, shows that exosomes are involved in the epithelial–mesenchymal transition in breast cancer.
Synthesis of exosomes takes place when intercommunication between notch receptors, ligands (JAGs) (jagged protein), and regulators (ADAM 10/17) (A disintegrin and metalloproteinase) occurs. ASPH (aspartate beta-hydroxylase) initiates the Notch cascade to synthesis or release of pro-metastatic exosomes. Normal breast cells have muted ASPH. The activation of ASPH by the NOTCH signalling gives rise to aggressive tumour progression in breast cancer with the help of exosomes [45]. Hippo signalling pathways are essential in restricting organ size. They inhibit oncogenic co-activators such as YAP (yes-associated protein) [46]. Inactivation of the Hippo pathway initiates the TEA (transcriptional enhancer factor) domain transcription factor to bind with the unphosphorylated YAP/TAZ (tafazzin), which translocates it to the nucleus [47]. TEAD (transcriptional enhancer factor domain TEF1)-influences the target genes. The transcription enhances the expression of mesenchymal markers such as Vimentin and N-cadherin and suppresses the expression of epithelial markers including E-cadherin [48]. The YAP gene cross-communicates with transcription factors such as ZEB1, SNAIL, and SLUG, which initiates epithelial–mesenchymal transition in cancer cells. Exosomes from mesenchymal-stem-cell-derived adipocytes enhance Hippo-induced epithelial–mesenchymal transition in breast cancer cells [49].
The Wnt/β-catenin signalling pathway is involved in the epithelial–mesenchymal transition. Specifically, a β-catenin molecule is involved in the epithelial–mesenchymal transition [50,51]. Scientists reported that exosomes containing Wnt ligands could influence Wnt signalling, which is responsible for EMT synthesis. Macrophage-derived exosomes containing Wnt5a initiate the Wnt/β-catenin signalling pathway in breast cancer cells [52]. Exosomal Wnt7a is accountable for the metastasis of breast tumours to the lungs [53]. In human breast fibroblasts, the exosome-mediated release of miR-9 produces cancer-associated fibroblast-like characteristics. Hence, regulating the Wnt ligands encapsulated in exosomes can be reasonable therapeutic control for cancer cells. Modulation of Wnt signalling by proteins derived from exosomes can inhibit metastasis [54].
The lncRNAs MALAT1 (metastasis-associated lung adenocarcinoma transcript 1) and ID4 (inhibitor of DNA-binding 4) cause an upregulation of the circular RNA circ 0076611 in exosomes released by TNBC. This circular RNA circ 0076611, released from the exosomes of TNBC cells, interacts with MYC (proto-oncogenes) and VEGFA (vascular endothelial growth factor A) mRNAs, further initiating cell proliferation and migration of TNBC cells [55]. Exosomes secreted by the TNBC cell line containing MMP-1 (Matrix Metallopeptidase 1) interact with the PAR1( protenase-activated receptor 1), a G protein receptor that initiates the EMT, which enables the breast cancer tumour to metastasize to the lungs [56]. miR-939 in exosomes downregulates the expression of VE-cadherin, destroys the barrier function of endothelial monolayers, and initiates tumourigenesis in breast cancer [57]. miR-155 is upregulated in TNBC, induces epithelial–mesenchymal transition, and plays an essential role in resistance [58,59] Figure 2.

4. Exosomes—Production, Preservation, Loading, Surface Engineering, and Exosome Hybrids in Drug Delivery

The production extraction of exosomes from the cells includes ultrafiltration, ultracentrifugation and precipitation, and immunoaffinity [60]. All these processes often have low-quality yield with contamination. Acoustic and fluidic device-based purification methods are often used to improve the quality [60,61]. Tangential flow filtration is often used to extract exosomes from the cell lines, improving good quality yield. Laboratory and industrial-grade bioreactors are also used for large-scale productions [62]. On the other hand, these bioreactor-based approaches involve inherent procedures that alter and modify the phenotypics of the cells over time, lowering the exosome quality. When T cells are agitated at 180 rpm, interleukin (IL)-2 levels in their exosomes are lowered; as a result, it is critical to carefully identify and develop a process for huge-scale exosome manufacturing that ensures exosome quality without negatively affecting the exosome source [63]. External stimuli such as chemical and electrical stimulation, hypoxia conditions, and silencing of specific endo lysosomal traffic proteins have shown better exosome yield from the cells [64,65].
Preservation of the exosomes often involves storing them at −80 °C or 4 °C. Keeping them at −80 degrees Celsius shows perfect enzymatic activity of beta-glucuronidase [66]. Exosome aggregation during the lyophilization step is reduced in the B16BL6 cell line when exosomes are stored using cryoprotectants such as trehalose [67]. Contrasting results were seen by Akers et al., that lyophilization leads to a reduction in the number of exosomes and RNA content compared to when it is stored at room temperature [68]. Modifying the exosomes with the arginine, the cell retained its quality after lyophilization. Continuous freezing and thawing result in a decrease in the number of exosomes [69].
Loading the exosomes with the desired compound is important. The exosomes can be used as a delivery agent for molecular drugs such as microRNA and siRNA, which are digested by the nuclease present in the body [70]. Other biomolecules include nucleic acids, lipids, proteins, and chemotherapeutic drugs. At present, there are two methods of loading: (a) endogenous and (b) exogenous methods. In the endogenous methods, the exosomes already have the compound of interest with the content of the protein or nucleic acid pre-loaded by the overexpression of the protein of interest; they are secreted [71].
In the exogenous methods, the exosomes are loaded by the interest of protein or nucleic acids with the help of artificial processes such as electroporation, co-incubation, sonication, lipofection, pH gradient, and calcium chloride methods [72]. All loading methods have cons and pros associated with them. The type of molecule of interest is vital for loading. For example, the loading of mRNAs into exosomes is often done by endogenous methods by enhancing the overexpression by transfecting the cell with plasmid DNA to trigger the production of desired biomolecules, which an electric signal can further stimulate. This method is known as the nano-proration method, which increases the yield of exosomes by 50-fold and 1000-fold in the exosomes having mRNAs compared with the standard methods [70]. The loading of microRNA and siRNA is performed through exogenous methods. One of the methods of sonication was seen to be effective in the case of breast cancer, where siRNA against HER2 was loaded, and a further reduction in the HER2 (human epidermal receptor 2) mRNA expression in the recipient cell was observed, making it a viable option for breast cancer therapy. The exogenous loading is more efficient than the direct loading, as it leads to inefficient loading and no effect on the recipient cells [73].
Surface engineering of exosomes is significant, as exosomes may be helpful in delivering drugs in vitro, but, for the perfect delivery in vivo, the exosomes need to be engineered. Otherwise, it will be limited due to non-specific toxicity and off-target effects [74]. The exosomes loaded with drugs are engineered by altering the surface with tumour-targeted ligands. Ligands used on the surface are often those ligands that can bind to those overexpressed receptors in the cancer cells [74]. The recipient cells quickly take up the exosomes with antibodies and the ligands through endocytosis, and they can easily escape encapsulation and trapping. The exosome surfaces are often altered using lipophilic linkers to attach monoclonal antibodies [75] Figure 3.

5. Exosomes for the Delivery of Chemotherapeutic Drugs

Exosomes can be used as delivery agents to influence signalling pathways, silence genes, and regulate the expression of different proteins. Exosomes perform gene transfection because of their intrinsic role in endogenous gene transfer. The endogenous origin makes exosomes less toxic to the cells and has a minimal immune effect [76]. The human body can tolerate exosomes, as they are naturally present, enabling them to behave as a delivery agent for potent drugs. Doxorubicin loaded in exosomes was developed to target HER2+ tumour cells and showed less cytotoxicity compared with the free delivery of doxorubicin (non-capsulated with exosomes), and, hence, can reduce the side effects caused by the chemotherapeutics drugs [77].

6. Engineered Exosomes in Triple-Negative Breast Cancer Drug Delivery

HELA-exosomes (cervical cancer cell lines) are engineered exosomes that exhibit significant anticancer effects in both a mouse model and human breast cancer organoids by increasing the activation of cDC1s in situ and, hence, boosting the tumour-reactive CD8+ T cell responses. Exosomes extracted from mesothelin (MSLN) initiate CAR-T cells, which have abilities similar to T cells, including CARs (cysteinyl-TRNA-synthetase) and CD3 surface expression. The exosomes loaded with CARs are seen to inhibit the growth of both endogenous and exogenous MSLN + TNBC. With the help of proteins such as perforin and granzyme B, exosomes can terminate the tumour. The tumour inhibition rate is high, followed by fewer side effects. Exosomes that have been isolated from CAR-T cells that are mesothelin (MSLN)-targeted significantly hinder the proliferation of MSLN-positive TNBC cells. Exosomes extracted from CAR-T cells exhibit a form of cell-free therapy that is significantly less toxic and more effective at preventing tumour growth than live CAR-T cells [78].
The exosomes derived from the HEK293T (human embryonic kidney cells with adenovirus) cells have LipHA-hEVs (Lipid grafted hyaluronic acid) as a ligand loaded with doxorubicin to target those cells expressing CD44, which often escape from the macrophages in MCF7/ADR cell lines or breast cancer [79]. Therefore, in breast cancer, the tumour mass is reduced by 89 percent, and the animal survival rate is increased by 50 percent [76]. Engineered exosomes with a modified surface coating of poly (lactic-co-glycolic acid) can target C-met, the epithelial–mesenchymal transition factor. Overexpression of C-met is in TNBC cells. The coated exosomes enhance the efficiency of uptake and anti-tumour activity of doxorubicin. In vivo experiments also show that the anti-tumour activity is improved, which induces apoptosis in triple-negative breast cancer [80]. Anti-HER2 antibody-conjugated paclitaxel-loaded liposomes are then employed to deliver HER2-targeted drugs. In vitro and in vivo experiments show that HER2 grafting combined with targeted medication delivery can boost therapy success. This innovative strategy may open the way for the rising wave of targeted medicine and immunotherapy by providing a simple method of modifying cell membranes by antigen presentation via accessible exosome uptake [81].
It is well known that the recently-created chemotherapeutic medication erastin has the ability to cause ferroptosis in TNBC cells, but its use has been limited by its poor water solubility and side effects. To solve this problem, Yu et al. loaded erastin into exosomes labelled with folate (FA) to target the TNBC cells overexpressing folate receptors on the cell surface, thereby enhancing ferroptosis with the pre-depletion of intracellular glutathione and overgeneration of reactive oxygen species (ROS). This significantly reduced the proliferative and migratory abilities of MDA-MB-231 cells [82]. Anti-IL-3R-EV therapy suppresses lung metastasis caused by intravenous injection of MDA-MB-231 cell lines. Anti-IL-3R-EV lowers Vimentin, β-catenin, and TWIST1 expression and practically eliminates liver and lung invasion from primary tumours. Anti-IL-3R-EVs of miR-24-3p (antago-miR-24-3p-EVs) are eliminated in vivo to regulate TWIST1 and diminish proliferative damage. In MDA-MB-231 cells, anti-IL-3R-EVs and anti--24-3p-EVs stimulate the expression of the SPRY2 (Sprouty homolog 2) gene, which is compatible with network analysis of miR-24-3p gene targeting. Finally, SPRY2 knockdown demonstrates that SPRY2 is the target for anti-IL-3R-EVs and induces apoptosis by inhibiting apoptotic stimuli mediated by these molecules and antago-miR-24-3p-EV [83].
One possible option is to increase tissue-specific targeting using tissue-specific antibodies or homing peptides by engineering the innate homing ability of exosomes. A chimeric protein against HER2+ is delivered by engineered exosomes with pLEX-LAMP DARP on their surface [84,85]. Engineered exosomes can target different tissues by masking the exosomes with membrane and surface display with an extra protein. To create a reconfigurable transmembrane adhesion platform, streptavidin is linked to 1,2-bis(dimethylphosphino)ethane: polyethylene glycol 5 k (DMPE-PEG). This enables biotinylated molecules, such as antibodies, to be associated with the anchor and used to construct vesicle surfaces for targeted delivery [86] Tables 1 and 2.

7. Exosomes as a Delivery Agent for microRNA and siRNA in TNBC Modulating Signalling

Exosomes generated from adipose mesenchymal stem cells infused with miRNA-381-3p reduce the metastasis in TNBC cell lines. Additionally, it was observed that miRNA-381-3p downregulates the expression of Wnt signalling and elements linked to the epithelial–mesenchymal transition in MDA-MB-231 [87]. miR3182 conjugated with exosomes derived from the human umbilical cord induce apoptosis in the TNBC cell line through downregulating genes such as mTOR and S6KB1 [88]. Exosomal miR-27a-3p, through production by endoplasmic reticulum stress, enhances immunological escape in breast cancer through modulating PD-L1 levels in macrophage’s immune evasion via the PTEN/AKT/PI3K axis [89]. Exosome A15 derived from monocyte-derived macrophages by activating protein kinase C can deliver microRNA 159 and doxorubicin, causing a synergetic effect on the TCF-7 gene and improving the anticancer effects. Exosomes containing miRNA-127, miR-197, miR-222, and miR-223 from stroma also aid breast cancer cell quiescence and enhance anti-proliferation rates, but to a lower extent than miRNAs transferred through gap junctions [90]. The TPD52 gene is downregulated by 70% when engineered exosomes from HEK293T cells are directed to translocate to HER2 positive breast cancer cells [91].
Exosomal miR-335-5p suppresses TNBC immune escape mediated by PD-L1 by promoting the ubiquitination of PD-L1 by USP22 via downregulation of USP22 and PD-L2 [92]. miR-134 is downregulated in the TNBC Hs578Ts(i)8 cell line. miR-134′s expression is lowered, and it acts as the tumour suppressor. It targets the pathways such as MAPK/ERK (mitogen-activated protein kinase/extracellular signal-regulated kinase) and other signallings such as EGFR (epidermal growth factor receptor) and NOTCH [93]. When miRNA-134 is inserted into Hs578Ts(i)8 cells with the help of exosomes, it can downregulate genes such as STAT5B (signal transducer activator of transcription 5) and HSP90. These genes are anti-apoptotic proteins and regulate the targets of miRNA-134. The conjugated exosomes with the microRNA-134 control the migration and the invasion of recipient TNBC cells and enhance recipient cell’s sensitivity to anti-HSP90 inhibitors [94]. Upward expression of miR-134 also represses breast cancer metastasis by targeting KRAS (Kirsten rat sarcoma virus). Therefore, exosomes could be used as a delivery agent and can intervene with the signalling in TNBC [95,96].
The level of microRNA let-7a expression in TNBC cell lines and patients suffering from triple-negative breast cancer shows low expression. The let-7a conjugated with exosomes, when targeted in the TNBC mice model, reduces tumour growth, migration, and invasion. The let-7a binds to the 3’ UTR of the c-Myc gene and silences the gene expression [97]. miR-770, transferred with the help of exosomes, can alter the tumour microenvironment in TNBC. The transmission of miR-770 via exosomes was confirmed in the co-culture of tumour-derived exosomes and tumour-associated macrophages, as revealed by enhanced expression of miR-770 in the recipient cells. Doxorubicin sensitivity is increased in cancer cells by miR-770 and it enhances therapeutically-induced apoptosis [98].
In vivo tumour growth is aided by miR-9. miR-9 is an essential player in the cross-communication between cancer cells and stroma [99]. miR-9 and miR-155 target PTEN (phosphatase and tensin homologue) and DUSP14 (dual specificity phosphatase 14) tumour suppressor genes in MCF-7 cell lines extracted from MDA-MB-231 exosomes [100]. Anti-miR-142-3p can be delivered efficiently by MSCs-Exo, lowering miR-142-3p and miR-150 levels while increasing the expression of the regulatory target genes APC (adenomatous polyposis coli protein regulator of Wnt) and P2X7R (purinergic receptor) in TNBC [101]. miR-496 and miR-137 are loaded in exosomes from MCF10A cell line target Del-1 and modulate it by inhibiting tumour growth in the TNBC cell lines such as MCF10A and MDA-MB-231 [102]. Extracellular vesicles derived from adipose mesenchymal cells enhance apoptosis by suppressing PD-L1 signalling in MDA-MB-231 TNBC cells [103,104] Table 1 and Table 2 and Figure 4.

8. Exosomal-Associated Small Molecule Modulating Pathways Inhibiting Metastasis of Triple-Negative Breast Cancer to Other Organs

As per the current report, exosomes delivering lncRNA DARS-AS1 siRNA in TNBC inhibit mild stress induced by TNBC tumours, which causes metastasis [104]. The mesenchymal stem cells derived from exosomal miR-106a-5P are suppressed by LncRNA HAND2-AS1 (link RNA heart and neural crest derivatives expressed 2), influencing the growth of triple-negative breast cancer [105]. Zhao et al. discovered that cationic bovine serum albumin (CBSA) conjugated siS100A4 and exosome membrane-coated nanoparticles are biocompatible and can improve drug delivery to lung PMN (poly morpho nuclear leucocytes). They can silence the genes that inhibit breast cancer’s malignancy in the lungs and are essential for post-overactive breast cancer tumours [106]. The therapeutic index of exosomes loaded with doxorubicin is more than the free one in TNBC. The cardiac toxicity is lowered [107]. miR-4516 can be used as a therapeutic for TNBC, as the CAF-derived exosomes influence the FOSL1 (transcription factor subunit) and initiate TNBC progression in the absence of miR-4516 [108].

9. Exosomes Specifically Regulating TNBC Signallings

Endogenous exosomes’ capacity to transmit proteins or miRNA makes them particularly useful for delivering nucleic acid- or peptide-based therapies directly to cancer cells. The siS100A4 is coupled to cationic bovine serum albumin (CBSA). This approach has demonstrated a strong suppressive impact on post-surgery metastasis in TNBC. The expression of the S100A4 protein, which is involved in metastasis, is downregulated by exosome-mediated RNAi, and postoperative metastasis in TNBC is significantly suppressed as a result. These investigations show that an effective therapeutic method for the prevention and treatment of cancer is the administration of siS100A4 through exosome membrane-packed core-shell NPS [109]. (Table 1 and Table 2)
KLF4 (Kruppel-like factor 4) and HOXD4 (homeobox D4) are the transcription factors that play a major role in metastasis and proliferation. MicroRNA miR-10b (microRNA) can transfer in metastatic BC cells (MDA-MB-231) according to different research by Singh et al., 2014 [110]. The target genes are KLF4 (Kruppel-like factor 4) and HOXD10 (homeobox D10). Target genes such as KLF4 and HOXD10 that are connected to protein expression can be silenced by the miR-10b if present in the elevated form [111].
Gong et al. created metalloproteinase-15-rich nanoscale target-specific exosomes that can carry doxorubicin (DOX) and miR-159 that have been modified with cholesterol to TNBC cells. In both in vitro and in vivo scenarios, it has been demonstrated that exosomes co-loaded with DOX and cholesterol-miR159 efficiently inhibit the growth of MDA-MB-231 TNBC cells by downregulating the expression of TCF-7 (transcription factor 7) [112]. The exosome-coated nanoparticles’ precise targeting ability greatly improved cellular absorption efficiency and demonstrated extraordinary TNBC-targeted efficacy, causing cancer cells to die, and, thus, inhibiting the course of the disease. Exosomes made from macrophages and coated in poly (lactic-co-glycolic acid) were highly effective in treating TNBC with targeted chemotherapy [113].
PD-L1 (programmed death ligand 1) is the major biomarker for TNBC [114]. When exosome surface PD-L1 attaches to the anti-PD-L1 antibody, leaving the tumour PD-L1 exposed, or when exosome surface PD-L1 binds to PD-1 on effector T cells during monoclonal antibody therapy, tumour cells can specifically bypass immunosuppression [115]. Therefore, exogenous PD-L1 is required for exosomes to have an immunosuppressive function; this immunosuppression may be prevented by pre-clearing exosomes with an anti-PD-L1 antibody. Furthermore, immune checkpoint blockade (ICB) therapy for metastatic TNBC uses exosomes loaded with PD-L1 as a helpful and reliable biomarker [116].
Exosomes for immunotherapy in TNBC can also be produced by mesenchymal stem cells (MSCs), i.e., MSC-derived exosomes, in addition to lymphocytes. Exosomes made from MSCs play a crucial role in immunomodulation as well, primarily by controlling immune cell activity and modifying the release of inflammatory mediators such as TNF and IL-1 in inflammatory bowel disease [117]. Monocytic myeloid-derived suppressor cells (M-MDSCs) may be differentiated into highly immunosuppressive M2-polarized macrophages with the help of MSC-derived exosomes. Additionally, PD-L1 overexpression in myeloid cells and PD-1 downregulation in T cells in breast cancer in vivo are two additional ways that MSC-derived exosomes might compromise protective anti-tumour immunity [111]. Although several studies have shown exosomes may have a therapeutic use and numerous clinical trials are now using exosomes, no clinically exosome-based medicines have yet received approval. All early research indicates that exosomes are a unique, potential treatment option for TNBC, and there is a high likelihood of employing modified exosomes as multifunctional therapeutic agents for TNBC therapy. Exosomes from the TNBC tumour microenvironment may also be used as a target for vaccination in the future to enhance the treatment effectiveness of TNBC.

10. Exosomes from Plants and Bacteria as a Delivery Agent

Derivation of exosomes is also from many other sources such as bacteria and plants. Exosomes from plants and bacteria have shown anti-tumour function. Low toxicity and large production are why exosome usage is attractive in cancer therapy. The tumour microenvironment contains EVs or outer membrane vesicles in the size of 40 nm obtained from gram −ve and +ve bacteria. They can produce anti-tumour cytokines as well as interferon-γ and CXCL10 (C-X-C motif chemokine ligand 10) [118]. Plant-derived extra vesicles are well researched. EVs from grapes, ginger, and citrus fruit act as delivery agents that can deliver chemotherapeutic and small molecular drugs [119]. Exosomes derived from grapefruit loaded with miR-17 and folic acid were delivered by the intranasal route inside mice, which inhibited brain tumours. Hence, we can conclude that breast cancer, when metastasized to the brain, can easily be targeted by using the exosomes, as they can even pass through the blood–brain barrier [120]. More studies in the field of triple-negative breast cancers are yet to be done, and it can be a new therapeutic approach to treat TNBC.
No current therapy involves plant-derived exosomes for the treatment of TNBC. As per the studies after the characterization of exosomes extracted from plants, it can be used as a new therapeutic for TNBC by targeting the pathways such as Wnt signalling, NOTCH, HIPPO signalling, and genes involved in epithelial–mesenchymal transition, which can fulfil the present research gap as mentioned in this review paper. More exploration with plant exosomes is needed.

11. Exosome-Mediated Delivery Vehicles in Pre-Clinical and Clinical Trials (Vaccines) in Triple-Negative Breast Cancer

Vaccines against cancer help the human body to redirect the immune system to fight against cancer rather than to prevent it. Standard vaccination leads to the prevention of infection towards other viral or bacterial infections. As a result, it appears that developing preventative EV-based cancer vaccines against virally-produced cancers, such as HPV, and therapeutic EV-based cancer vaccines for non-virally induced cancers, such as TNBC, would be more appropriate. EVs can communicate between the cells and, therefore, can be used as a vaccine. Natural killer cells derived from exosomes have more potential cytotoxicity effects against tumour cells. Exosomes have the potential to be a drug delivery agent in vitro as well as in vivo. Clinically, exosomes are tested as a delivery vehicle in other cancers. Exosome delivery could be a hope for TNBC as well [121,122]. Exosomes loaded with curcumin are tested against colon cancer undergoing clinical phase 1 trials. (NCT01294072).
Exosomes are small vesicles secreted from dendritic cells consisting of MHC class 1 and class II molecules, along with other CD86 proteins and tetraspanins, stimulate an immune response and undergo phase 2 clinical trials [123,124]. The literature already reports that EV cancer vaccines derived from mesenchymal stem cells, macrophages, and tumour-derived exosomes are present. Tumour-derived exosomes can act as the vaccine against cancer. However, consideration of also having oncogenic materials is needed. TEV (tumour-derived extracellular vesicles)-based vaccines contain tumour rejection antigens and can effectively be represented to the dendritic cells by MHC-1 molecules for cancer treatment [125]. As the TEV-based vaccines have safety issues, other alternatives can be used, such as extracellular vesicles from healthy cell lines carrying TAAs (tumour-associated antigens) DNA, protein, or the mRNA [126]. Zitvogel showed that a single dose of a dendritic cell-derived vaccine against cancer could stimulate the immune response via activating T cell-mediated anti-tumour effects [127]. The dendritic cell-derived exosomes can inhibit tumours by activating CTLs (cytotoxic T lymphocytes) and natural killer cells [128]. They have also shown an immune response toward mammary adenocarcinoma tumour cells. DC-EV-based vaccines against cancer can trigger the immune system by presenting them with peptides or TAAs related to cancers [129]. They have also been proven safe and achievable in phase 1 of clinical studies. Advanced research is necessary for the long-lasting response of the immune system against tumour cells in the field of exosome-derived vaccines [130]. As shown in Table 3, [131] six active clinical trials test cancer vaccines for TNBC with chemotherapeutic medicines or immunotherapy treatments [131].

12. Conclusions and Perspectives

Exosomes have many promising characteristics—bio-feasibility, biocompatibility, good loading capacity, and less toxicity, being derivatives from biological sources—making them a very appropriate candidate for drug delivery. Exosomes can easily be manipulated accordingly by making changes to their surface with the addition of proteins or antibodies. Many methods are available for loading and also for extraction. Cost-effective production of exosomes is also needed, as there is a limitation with the available methods for the extensive quantity of extractions from the cells. Much scientific data showed how exosomes could reduce the metastatic rate of TNBC by delivering miRNA, siRNA, LncRNA, and chemotherapeutic drugs in breast cancer cell lines. The extraction of exosomes from plants has not been explored yet in the field of TNBC, but scientific proof is present with brain cancer. Hence, the exosomes from plants can also be used as a delivery agent for TNBC. More research is needed in the field to study the characteristics of TNBC and the constituents of TNBC. Exosomes also play a crucial role in the EMT transition in TNBC. Altering the secretions of exosomes by modulating the content or silencing some genes may be a solution for the metastasis of TNBC, which progresses to other organs. Exosomes can also be biomarkers for TNBC that are responsible for chemoresistance in cancers. Exosomes also target signalling such as Wnt, NOTCH, and PTEN/AKT/PI3K (phosphatase tensin homolog/phosphokinase pathway), etc., in the TNBC, which plays a vital role in the epithelial-to-mesenchymal transition. Exosomes can also target genes, making them a perfect target delivery agent. Therefore, a cure for TNBC with personalized therapy is possible if targeted therapy with the help of exosomes is studied more widely. An in-depth study can also pave the way for exosomes to emerge as a vaccine to treat triple-negative breast cancer.

Author Contributions

Conceptualization, software, Writing—Original draft preparation: M.B.; Validation, writing—review and editing, visualization, Supervision, project administration: V.D.R.; All authors have read and agreed to the published version of the manuscript.

Funding

This research received no specific grant from the public, commercial, or not-for-profit funding agencies.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Given by the authors and the institute.

Data Availability Statement

Not applicable.

Acknowledgments

We want to express our gratitude to the Vellore Institute of Technology for providing us with all facilities required for the completion of the review.

Conflicts of Interest

The authors declare no conflicts of Interest.

Abbreviations

TNBC: triple-negative breast cancer; BBB: the blood–brain barrier; HSP: heat shock protein; TIS: therapeutic-induced senescence; SNAIL2: zinc finger protein SNAI 1; SLUG: SNAI 2 gene; ZEB1: zinc finger E-box-binding homeobox 1, JAGs: jagged protein, ADAM 10/17: a disintegrin and metalloproteinase; ASPH: aspartate beta-hydroxylase; YAP: yes-associated protein; TEF1: transcriptional enhancer factor; TAZ: tafazzin; VEGFA: vascular endothelial growth factor A; MMP-1: matrix metallopeptidase 1; PAR1: proteinase-activated receptor 1; HER2: human epidermal receptor 2; MAPK/ERK: mitogen-activated protein kinase/extracellular signal regulated kinase; STAT5B: signal transducer activator of transcription 5; KRAS: Kirsten rat sarcoma virus; PTEN: phosphatase and tensin homolog; DUSP14: dual specificity phosphatase 14; APC: adenomatous polyposis coli protein regulator of Wnt; P2X7R: purinergic receptor; LncRNA HAND2-AS1: link RNA heart and neural crest derivatives expressed 2; PMN: poly morpho nuclear leucocytes; CXCL10: C-X-C motif chemokine ligand 10; TEV: tumour-derived extracellular vesicles; TAAs: tumour-associated antigens; CTLs: cytotoxic T lymphocytes; PTEN/AKT/PI3K: phosphatase tensin homolog/phosphokinase pathway.

References

  1. Prusty, R.K.; Begum, S.; Patil, A.; Naik, D.D.; Pimple, S.; Mishra, G. Knowledge of Symptoms and Risk Factors of Breast Cancer among Women: A Community Based Study in a Low Socio-Economic Area of Mumbai, India. BMC Womens Health 2020, 20, 106. [Google Scholar] [CrossRef]
  2. Yin, L.; Duan, J.-J.; Bian, X.-W.; Yu, S.-C. Triple-Negative Breast Cancer Molecular Subtyping and Treatment Progress. Breast Cancer Res. 2020, 22, 61. [Google Scholar] [CrossRef] [PubMed]
  3. O’Reilly, D.; Al Sendi, M.; Kelly, C.M. Overview of Recent Advances in Metastatic Triple Negative Breast Cancer. World J. Clin. Oncol. 2021, 12, 164. [Google Scholar] [CrossRef] [PubMed]
  4. Honda, Y.; Aruga, T.; Yamashita, T.; Miyamoto, H.; Horiguchi, K.; Kitagawa, D.; Idera, N.; Goto, R.; Kuroi, K. Prolonged Survival after Diagnosis of Brain Metastasis from Breast Cancer: Contributing Factors and Treatment Implications. Jpn. J. Clin. Oncol. 2015, 45, 713–718. [Google Scholar] [CrossRef] [PubMed]
  5. Prat, A.; Adamo, B.; Cheang, M.C.; Anders, C.K.; Carey, L.A.; Perou, C.M. Molecular Characterization of Basal-like and Non-basal-like Triple-negative Breast Cancer. Oncologist 2013, 18, 123–133. [Google Scholar] [CrossRef] [PubMed]
  6. Lehmann, B.; Bauer, J.; 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. Am. Soc. Clin. Investig. 2011, 121, 2750–2767. [Google Scholar] [CrossRef] [PubMed]
  7. Walsh, E.M.; Keane, M.M.; Wink, D.A.; Callagy, G.; Glynn, S.A. Review of Triple Negative Breast Cancer and the Impact of Inducible Nitric Oxide Synthase on Tumor Biology and Patient Outcomes. Crit. Rev. Oncog. 2016, 21, 333–351. [Google Scholar] [CrossRef] [PubMed]
  8. Yao, H.; He, G.; Yan, S.; Chen, C.; Song, L.; Rosol, T.J.; Deng, X. Triple-Negative Breast Cancer: Is There a Treatment on the Horizon? Oncotarget 2016, 8, 1913–1924. [Google Scholar] [CrossRef] [PubMed]
  9. Okumura, Y.; Nishimura, R.; Nakatsukasa, A.; Yoshida, N.; Masuda, M.; Tanabe, T.; Shien, S.; Tanaka, N.; Arima, Y.; Komoike, T.; et al. Change in Estrogen Receptor, HER2, and Ki-67 Status between Primary Breast Cancer and Ipsilateral Breast Cancer Tumor Recurrence. Eur. J. Surg. Oncol. 2015, 41, 548–552. [Google Scholar] [CrossRef]
  10. Nakashoji, A.; Matsui, A.; Nagayama, A.; Iwata, Y.; Sasahara, M.; Murata, Y. Clinical Predictors of Pathological Complete Response to Neoadjuvant Chemotherapy in Triple-Negative Breast Cancer. Oncol. Lett. 2017, 14, 4135–4141. [Google Scholar] [CrossRef] [Green Version]
  11. Aslam, M.S.; Naveed, S.; Ahmed, A.; Abbas, Z.; Gull, I.; Athar, M.A.; Aslam, M.S.; Naveed, S.; Ahmed, A.; Abbas, Z.; et al. Side Effects of Chemotherapy in Cancer Patients and Evaluation of Patients Opinion about Starvation Based Differential Chemotherapy. J. Cancer Ther. 2014, 5, 817–822. [Google Scholar] [CrossRef]
  12. He, Q.; Liu, J.; Liang, J.; Liu, X.; Li, W.; Liu, Z.; Ding, Z.; Tuo, D. Towards Improvements for Penetrating the Blood–Brain Barrier—Recent Progress from a Material and Pharmaceutical Perspective. Cells 2018, 7, 24. [Google Scholar] [CrossRef]
  13. Lin, N.U.; Claus, E.; Sohl, J.; Razzak, A.R.; Arnaout, A.; Winer, E.P. Sites of Distant Recurrence and Clinical Outcomes in Patients with Metastatic Triple-Negative Breast Cancer: High Incidence of Central Nervous System Metastases. Cancer 2008, 113, 2638–2645. [Google Scholar] [CrossRef] [PubMed]
  14. Livshits, Z.; Rao, R.B.; Smith, S.W. An Approach to Chemotherapy-Associated Toxicity. Emerg. Med. Clin. N. Am. 2014, 32, 167–203. [Google Scholar] [CrossRef] [PubMed]
  15. Hossen, S.; Hossain, M.K.; Basher, M.K.; Mia, M.N.H.; Rahman, M.T.; Uddin, M.J. Smart Nanocarrier-Based Drug Delivery Systems for Cancer Therapy and Toxicity Studies: A Review. J. Adv. Res. 2018, 15, 1–18. [Google Scholar] [CrossRef] [PubMed]
  16. Ventola, C.L. Progress in Nanomedicine: Approved and Investigational Nanodrugs. Pharm. Ther. 2017, 42, 742. [Google Scholar]
  17. Din, F.U.; Aman, W.; Ullah, I.; Qureshi, O.S.; Mustapha, O.; Shafique, S.; Zeb, A. Effective Use of Nanocarriers as Drug Delivery Systems for the Treatment of Selected Tumors. Int. J. Nanomed. 2017, 12, 7291. [Google Scholar] [CrossRef]
  18. Melzer, C.; Rehn, V.; Yang, Y.; Bähre, H.; von der Ohe, J.; Hass, R. Taxol-Loaded MSC-Derived Exosomes Provide a Therapeutic Vehicle to Target Metastatic Breast Cancer and Other Carcinoma Cells. Cancers 2019, 11, 798. [Google Scholar] [CrossRef]
  19. Chen, C.C.; Liu, L.; Ma, F.; Wong, C.W.; Guo, X.E.; Chacko, J.V.; Farhoodi, H.P.; Zhang, S.X.; Zimak, J.; Ségaliny, A.; et al. Elucidation of Exosome Migration across the Blood-Brain Barrier Model In Vitro. Cell. Mol. Bioeng. 2016, 9, 509–529. [Google Scholar] [CrossRef]
  20. Li, M.; Li, S.; Du, C.; Zhang, Y.; Li, Y.; Chu, L.; Han, X.; Galons, H.; Zhang, Y.; Sun, H.; et al. Exosomes from Different Cells: Characteristics, Modifications, and Therapeutic Applications. Eur. J. Med. Chem. 2020, 207, 112784. [Google Scholar] [CrossRef]
  21. Kim, H.; Jang, H.; Cho, H.; Choi, J.; Hwang, K.Y.; Choi, Y.; Kim, S.H.; Yang, Y. Recent Advances in Exosome-Based Drug Delivery for Cancer Therapy. Cancers 2021, 13, 4435. [Google Scholar] [CrossRef]
  22. Kanchanapally, R.; Deshmukh, S.K.; Chavva, S.R.; Tyagi, N.; Srivastava, S.K.; Patel, G.K.; Singh, A.P.; Singh, S. Drug-Loaded Exosomal Preparations from Different Cell Types Exhibit Distinctive Loading Capability, Yield, and Antitumor Efficacies: A Comparative Analysis. Int. J. Nanomed. 2019, 14, 531–541. [Google Scholar] [CrossRef]
  23. Xi, X.M.; Xi, X.M.; Xia, S.J.; Lu, R. Drug Loading Techniques for Exosome-Based Drug Delivery Systems. Pharmazie 2021, 76, 61–67. [Google Scholar] [CrossRef] [PubMed]
  24. Berumen Sánchez, G.; Bunn, K.E.; Pua, H.H.; Rafat, M. Extracellular Vesicles: Mediators of Intercellular Communication in Tissue Injury and Disease. Cell Commun. Signal. 2021, 19, 104. [Google Scholar] [CrossRef] [PubMed]
  25. Bei, Y.; Chen, T.; Banciu, D.D.; Cretoiu, D.; Xiao, J. Circulating Exosomes in Cardiovascular Diseases. Adv. Exp. Med. Biol. 2017, 998, 255–269. [Google Scholar] [CrossRef]
  26. Howitt, J.; Hill, A.F. Exosomes in the Pathology of Neurodegenerative Diseases. J. Biol. Chem. 2016, 291, 26589–26597. [Google Scholar] [CrossRef]
  27. Baharlooi, H.; Azimi, M.; Salehi, Z.; Izad, M. Mesenchymal Stem Cell-Derived Exosomes: A Promising Therapeutic Ace Card to Address Autoimmune Diseases. Int. J. Stem Cells 2020, 13, 13–23. [Google Scholar] [CrossRef]
  28. Steinbichler, T.B.; Dudás, J.; Riechelmann, H.; Skvortsova, I.I. The Role of Exosomes in Cancer Metastasis. Semin. Cancer Biol. 2017, 44, 170–181. [Google Scholar] [CrossRef]
  29. 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]
  30. Yoshioka, Y.; Konishi, Y.; Kosaka, N.; Katsuda, T.; Kato, T.; Ochiya, T. Comparative Marker Analysis of Extracellular Vesicles in Different Human Cancer Types. J. Extracell. Vesicles 2013, 2, 20424. [Google Scholar] [CrossRef] [PubMed]
  31. Liu, T.; Zhang, Q.; Zhang, J.; Li, C.; Miao, Y.R.; Lei, Q.; Li, Q.; Guo, A.Y. EVmiRNA: A Database of MiRNA Profiling in Extracellular Vesicles. Nucleic Acids Res. 2019, 47, D89–D93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Pathan, M.; Fonseka, P.; Chitti, S.V.; Kang, T.; Sanwlani, R.; Van Deun, J.; Hendrix, A.; Mathivanan, S. Vesiclepedia 2019: A Compendium of RNA, Proteins, Lipids and Metabolites in Extracellular Vesicles. Nucleic Acids Res. 2019, 47, D516–D519. [Google Scholar] [CrossRef]
  33. Keerthikumar, S.; Chisanga, D.; Ariyaratne, D.; Al Saffar, H.; Anand, S.; Zhao, K.; Samuel, M.; Pathan, M.; Jois, M.; Chilamkurti, N.; et al. ExoCarta: A Web-Based Compendium of Exosomal Cargo. J. Mol. Biol. 2016, 428, 688–692. [Google Scholar] [CrossRef] [PubMed]
  34. Tai, Y.L.; Chen, K.C.; Hsieh, J.T.; Shen, T.L. Exosomes in Cancer Development and Clinical Applications. Cancer Sci. 2018, 109, 2364–2374. [Google Scholar] [CrossRef]
  35. Zlotogorski-Hurvitz, A.; Dekel, B.Z.; Malonek, D.; Yahalom, R.; Vered, M. FTIR-Based Spectrum of Salivary Exosomes Coupled with Computational-Aided Discriminating Analysis in the Diagnosis of Oral Cancer. J. Cancer Res. Clin. Oncol. 2019, 145, 685–694. [Google Scholar] [CrossRef] [PubMed]
  36. Kavanagh, E.L.; Lindsay, S.; Halasz, M.; Gubbins, L.C.; Weiner-Gorzel, K.; Guang, M.H.Z.; McGoldrick, A.; Collins, E.; Henry, M.; Blanco-Fernández, A.; et al. Protein and Chemotherapy Profiling of Extracellular Vesicles Harvested from Therapeutic Induced Senescent Triple Negative Breast Cancer Cells. Oncogenesis 2017, 6, e388. [Google Scholar] [CrossRef]
  37. Jakhar, R.; Crasta, K. Exosomes as Emerging Pro-Tumorigenic Mediators of the Senescence-Associated Secretory Phenotype. Int. J. Mol. Sci. 2019, 20, 2547. [Google Scholar] [CrossRef]
  38. Campisi, J.; D’Adda Di Fagagna, F. Cellular Senescence: When Bad Things Happen to Good Cells. Nat. Rev. Mol. Cell Biol. 2007, 8, 729–740. [Google Scholar] [CrossRef]
  39. Lianidou, E.; Pantel, K. Liquid Biopsies. Genes Chromosomes Cancer 2019, 58, 219–232. [Google Scholar] [CrossRef]
  40. Toth, B.; Nieuwland, R.; Liebhardt, S.; Ditsch, N.; Steinig, K.; Stieber, P.; Rank, A.; Göhring, P.; Thaler, C.J.; Friese, K.; et al. Circulating Microparticles in Breast Cancer Patients: A Comparative Analysis with Established Biomarkers. Anticancer Res. 2008, 28, 1107–1112. [Google Scholar]
  41. Eichelser, C.; Stückrath, I.; Müller, V.; Milde-Langosch, K.; Wikman, H.; Pantel, K.; Schwarzenbach, H. Increased Serum Levels of Circulating Exosomal MicroRNA-373 in Receptor-Negative Breast Cancer Patients. OncoTargets Ther. 2014, 5, 9650–9663. [Google Scholar] [CrossRef]
  42. Dioufa, N.; Clark, A.M.; Ma, B.; Beckwitt, C.H.; Wells, A. Bi-Directional Exosome-Driven Intercommunication between the Hepatic Niche and Cancer Cells. Mol. Cancer 2017, 16, 172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Kim, J.E.; Cho, N.H. 261: Cancer-Associated Fibroblast (CAF)-Derived Exosome May Mediate Breast Cancer Progression by Reducing Exosomal MicroRNAs. Eur. J. Cancer 2014, 50, S61. [Google Scholar] [CrossRef]
  44. Kim, H.; Lee, S.; Shin, E.; Seong, K.M.; Jin, Y.W.; Youn, H.S.; Youn, B.H. The Emerging Roles of Exosomes as EMT Regulators in Cancer. Cells 2020, 9, 861. [Google Scholar] [CrossRef] [PubMed]
  45. Lin, Q.; Chen, X.; Meng, F.; Ogawa, K.; Li, M.; Song, R.; Zhang, S.; Zhang, Z.; Kong, X.; Xu, Q.; et al. ASPH-Notch Axis Guided Exosomal Delivery of Prometastatic Secretome Renders Breast Cancer Multi-Organ Metastasis. Mol. Cancer 2019, 18, 156. [Google Scholar] [CrossRef] [PubMed]
  46. Boopathy, G.T.K.; Hong, W. Role of Hippo Pathway-YAP/TAZ Signaling in Angiogenesis. Front. Cell Dev. Biol. 2019, 7, 49. [Google Scholar] [CrossRef]
  47. Huh, H.; Kim, D.; Jeong, H.; Park, H.W. Regulation of TEAD Transcription Factors in Cancer Biology. Cells 2019, 8, 600. [Google Scholar] [CrossRef]
  48. Loh, C.-Y.; Chai, J.Y.; Fang Tang, T.; Fen Wong, W.; Sethi, G.; Shanmugam, M.K.; Chong, P.P.; Looi, C.Y. The E-Cadherin and N-Cadherin Switch in Epithelial-to-Mesenchymal Transition: Signaling, Therapeutic Implications, and Challenges. Cells 2019, 8, 1118. [Google Scholar] [CrossRef] [PubMed]
  49. Wang, S.; Su, X.; Xu, M.; Xiao, X.; Li, X.; Li, H.; Keating, A.; Zhao, R.C. Exosomes Secreted by Mesenchymal Stromal/Stem Cell-Derived Adipocytes Promote Breast Cancer Cell Growth via Activation of Hippo Signaling Pathway. Stem Cell Res. Ther. 2019, 10, 117. [Google Scholar] [CrossRef]
  50. Wei, C.Y.; Zhu, M.X.; Yang, Y.W.; Zhang, P.F.; Yang, X.; Peng, R.; Gao, C.; Lu, J.C.; Wang, L.; Deng, X.Y.; et al. Downregulation of RNF128 Activates Wnt/β-Catenin Signaling to Induce Cellular EMT and Stemness via CD44 and CTTN Ubiquitination in Melanoma. J. Hematol. Oncol. 2019, 12, 21. [Google Scholar] [CrossRef]
  51. Yang, M.; Li, S.N.; Anjum, K.M.; Gui, L.X.; Zhu, S.S.; Liu, J.; Chen, J.K.; Liu, Q.F.; Ye, G.D.; Wang, W.J.; et al. A Double-Negative Feedback Loop between Wnt-β-Catenin Signaling and HNF4α Regulates Epithelial-Mesenchymal Transition in Hepatocellular Carcinoma. J. Cell Sci. 2013, 126, 5692–5703. [Google Scholar] [CrossRef]
  52. Ekström, E.J.; Bergenfelz, C.; von Bülow, V.; Serifler, F.; Carlemalm, E.; Jönsson, G.; Andersson, T.; Leandersson, K. WNT5A Induces Release of Exosomes Containing Pro-Angiogenic and Immunosuppressive Factors from Malignant Melanoma Cells. Mol. Cancer 2014, 13, 88. [Google Scholar] [CrossRef] [Green Version]
  53. Li, C.; Yoshimura, T.; Tian, M.; Wang, Y.; Kondo, T.; Yamamoto, K.I.; Fujisawa, M.; Ohara, T.; Sakaguchi, M.; Matsukawa, A. Exosomal Wnt7a from a Low Metastatic Subclone Promotes Lung Metastasis of a Highly Metastatic Subclone in the Murine 4t1 Breast Cancer. Breast Cancer Res. 2022, 24, 60. [Google Scholar] [CrossRef] [PubMed]
  54. Nagaharu, K.; Zhang, X.; Yoshida, T.; Katoh, D.; Hanamura, N.; Kozuka, Y.; Ogawa, T.; Shiraishi, T.; Imanaka-Yoshida, K. Tenascin C Induces Epithelial-Mesenchymal Transition-like Change Accompanied by SRC Activation and Focal Adhesion Kinase Phosphorylation in Human Breast Cancer Cells. Am. J. Pathol. 2011, 178, 754–763. [Google Scholar] [CrossRef] [PubMed]
  55. Turco, C.; Esposito, G.; Iaiza, A.; Goeman, F.; Benedetti, A.; Gallo, E.; Daralioti, T.; Perracchio, L.; Sacconi, A.; Pasanisi, P.; et al. MALAT1-Dependent Hsa_circ_0076611 Regulates Translation Rate in Triple-Negative Breast Cancer. Commun. Biol. 2022, 5, 598. [Google Scholar] [CrossRef] [PubMed]
  56. Zhu, Y.; Tao, Z.; Chen, Y.; Lin, S.; Zhu, M.; Ji, W.; Liu, X.; Li, T.; Hu, X. Exosomal MMP-1 Transfers Metastasis Potential in Triple-Negative Breast Cancer through PAR1-Mediated EMT. Breast Cancer Res. Treat. 2022, 193, 65–81. [Google Scholar] [CrossRef] [PubMed]
  57. 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]
  58. Gupta, I.; Sareyeldin, R.M.; Al-Hashimi, I.; Al-Thawadi, H.A.; Al Farsi, H.; Vranic, S.; Moustafa, A.E. Al Triple Negative Breast Cancer Profile, from Gene to MicroRNA, in Relation to Ethnicity. Cancers 2019, 11, 363. [Google Scholar] [CrossRef]
  59. Stevic, I.; Müller, V.; Weber, K.; Fasching, P.A.; Karn, T.; Marmé, F.; Schem, C.; Stickeler, E.; Denkert, C.; Van Mackelenbergh, M.; et al. Specific MicroRNA Signatures in Exosomes of Triple-Negative and HER2-Positive Breast Cancer Patients Undergoing Neoadjuvant Therapy within the GeparSixto Trial. BMC Med. 2018, 16, 179. [Google Scholar] [CrossRef]
  60. 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]
  61. Patel, G.K.; Zubair, H.; Khan, M.A.; Srivastava, S.K.; Ahmad, A.; Patton, M.C.; Singh, S.; Khushman, M.; Singh, A.P. Exosomes: Key Supporters of Tumor Metastasis. In Diagnostic and Therapeutic Applications of Exosomes in Cancer; Academic Press: Cambridge, MA, USA, 2018; pp. 261–283. [Google Scholar] [CrossRef]
  62. Ingato, D.; Edson, J.A.; Zakharian, M.; Kwon, Y.J. Cancer Cell-Derived, Drug-Loaded Nanovesicles Induced by Sulfhydryl-Blocking for Effective and Safe Cancer Therapy. ACS Nano 2018, 12, 9568–9577. [Google Scholar] [CrossRef] [PubMed]
  63. Carswell, K.S.; Papoutsakis, E.T. Culture of Human T Cells in Stirred Bioreactors for Cellular Immunotherapy Applications: Shear, Proliferation, and the IL-2 Receptor. Biotechnol. Bioeng. 2000, 68, 328–338. [Google Scholar] [CrossRef]
  64. Kore, R.A.; Edmondson, J.L.; Jenkins, S.V.; Jamshidi-Parsian, A.; Dings, R.P.M.; Reyna, N.S.; Griffin, R.J. Hypoxia-Derived Exosomes Induce Putative Altered Pathways in Biosynthesis and Ion Regulatory Channels in Glioblastoma Cells. Biochem. Biophys. Rep. 2018, 14, 104–113. [Google Scholar] [CrossRef]
  65. Fukuta, T.; Nishikawa, A.; Kogure, K. Low Level Electricity Increases the Secretion of Extracellular Vesicles from Cultured Cells. Biochem. Biophys. Rep. 2019, 21, 100713. [Google Scholar] [CrossRef]
  66. Frank, J.; Richter, M.; de Rossi, C.; Lehr, C.M.; Fuhrmann, K.; Fuhrmann, G. Extracellular Vesicles Protect Glucuronidase Model Enzymes during Freeze-Drying. Sci Rep. 2018, 8, 12377. [Google Scholar] [CrossRef]
  67. Charoenviriyakul, C.; Takahashi, Y.; Nishikawa, M.; Takakura, Y. Preservation of Exosomes at Room Temperature Using Lyophilization. Int. J. Pharm. 2018, 553, 1–7. [Google Scholar] [CrossRef]
  68. Akers, J.C.; Ramakrishnan, V.; Kim, R.; Skog, J.; Nakano, I.; Pingle, S.; Kalinina, J.; Hua, W.; Kesari, S.; Mao, Y.; et al. MiR-21 in the Extracellular Vesicles (EVs) of Cerebrospinal Fluid (CSF): A Platform for Glioblastoma Biomarker Development. PLoS ONE 2013, 8, e78115. [Google Scholar] [CrossRef]
  69. Noguchi, K.; Hirano, M.; Hashimoto, T.; Yuba, E.; Takatani-Nakase, T.; Nakase, I. Effects of Lyophilization of Arginine-Rich Cell-Penetrating Peptide-Modified Extracellular Vesicles on Intracellular Delivery. Anticancer Res. 2019, 39, 6701–6709. [Google Scholar] [CrossRef]
  70. Yang, Z.; Shi, J.; Xie, J.; Wang, Y.; Sun, J.; Liu, T.; Zhao, Y.; Zhao, X.; Wang, X.; Ma, Y.; et al. Large-Scale Generation of Functional MRNA-Encapsulating Exosomes via Cellular Nanoporation. Nat. Biomed. Eng. 2020, 4, 69–83. [Google Scholar] [CrossRef]
  71. Huang, L.; Wang, D.; Gu, N.; Zhang, X.E. Construction of Engineered Exosomes with High Loading Efficiency of Cellular Endogenous Proteins. Sheng Wu Gong Cheng Xue Bao 2019, 35, 1537–1545. [Google Scholar] [CrossRef] [PubMed]
  72. Kooijmans, S.A.A.; Stremersch, S.; Braeckmans, K.; De Smedt, S.C.; Hendrix, A.; Wood, M.J.A.; Schiffelers, R.M.; Raemdonck, K.; Vader, P. Electroporation-Induced SiRNA Precipitation Obscures the Efficiency of SiRNA Loading into Extracellular Vesicles. J. Control. Release 2013, 172, 229–238. [Google Scholar] [CrossRef]
  73. Lamichhane, T.N.; Jeyaram, A.; Patel, D.B.; Parajuli, B.; Livingston, N.K.; Arumugasaamy, N.; Schardt, J.S.; Jay, S.M. Oncogene Knockdown via Active Loading of Small RNAs into Extracellular Vesicles by Sonication. Cell. Mol. Bioeng. 2016, 9, 315. [Google Scholar] [CrossRef]
  74. Oppel, F.; Schurmann, M.; Goon, P.; Albers, A.E.; Sudhoff, H. Specific Targeting of Oncogenes Using CRISPR Technology. Cancer Res. 2018, 78, 5506–5512. [Google Scholar] [CrossRef]
  75. Si, Y.; Kim, S.; Zhang, E.; Tang, Y.; Jaskula-Sztul, R.; Markert, J.M.; Chen, H.; Zhou, L.; Liu, X. Targeted Exosomes for Drug Delivery: Biomanufacturing, Surface Tagging, and Validation. Biotechnol. J. 2020, 15, 1900163. [Google Scholar] [CrossRef]
  76. Rashed, M.H.; Bayraktar, E.; Helal, G.K.; Abd-Ellah, M.F.; Amero, P.; Chavez-Reyes, A.; Rodriguez-Aguayo, C. Exosomes: From Garbage Bins to Promising Therapeutic Targets. Int. J. Mol. Sci. 2017, 18, 538. [Google Scholar] [CrossRef]
  77. Bobrie, A.; Krumeich, S.; Reyal, F.; Recchi, C.; Moita, L.F.; Seabra, M.C.; Ostrowski, M.; Théry, C. Rab27a Supports Exosome-Dependent and -Independent Mechanisms That Modify the Tumor Microenvironment and Can Promote Tumor Progression. Cancer Res. 2012, 72, 4920–4930. [Google Scholar] [CrossRef]
  78. Yang, P.; Cao, X.; Cai, H.; Feng, P.; Chen, X.; Zhu, Y.; Yang, Y.; An, W.; Yang, Y.; Jie, J. The Exosomes Derived from CAR-T Cell Efficiently Target Mesothelin and Reduce Triple-Negative Breast Cancer Growth. Cell. Immunol. 2021, 360, 104262. [Google Scholar] [CrossRef]
  79. Liu, J.; Ye, Z.; Xiang, M.; Chang, B.; Cui, J.; Ji, T.; Zhao, L.; Li, Q.; Deng, Y.; Xu, L.; et al. Functional Extracellular Vesicles Engineered with Lipid-Grafted Hyaluronic Acid Effectively Reverse Cancer Drug Resistance. Biomaterials 2019, 223, 119475. [Google Scholar] [CrossRef]
  80. Li, S.; Wu, Y.; Ding, F.; Yang, J.; Li, J.; Gao, X.; Zhang, C.; Feng, J. Engineering Macrophage-Derived Exosomes for Targeted Chemotherapy of Triple-Negative Breast Cancer. Nanoscale 2020, 12, 10854–10862. [Google Scholar] [CrossRef]
  81. Quinn, Z.; Mao, W.; Xia, Y.; John, R.; Wan, Y. Conferring Receptors on Recipient Cells with Extracellular Vesicles for Targeted Drug Delivery. Bioact. Mater. 2021, 6, 749. [Google Scholar] [CrossRef]
  82. Yu, M.; Gai, C.; Li, Z.; Ding, D.; Zheng, J.; Zhang, W.; Lv, S.; Li, W. Targeted Exosome-Encapsulated Erastin Induced Ferroptosis in Triple Negative Breast Cancer Cells. Cancer Sci. 2019, 110, 3173–3182. [Google Scholar] [CrossRef] [Green Version]
  83. Lopatina, T.; Grange, C.; Cavallari, C.; Navarro-Tableros, V.; Lombardo, G.; Rosso, A.; Cedrino, M.; Pomatto, M.A.C.; Koni, M.; Veneziano, F.; et al. Targeting IL-3Rα on Tumor-Derived Endothelial Cells Blunts Metastatic Spread of Triple-Negative Breast Cancer via Extracellular Vesicle Reprogramming. Oncogenesis 2020, 9, 90. [Google Scholar] [CrossRef]
  84. Limoni, S.K.; Salimi, F.; Moghaddam, M.F. Designing PLEX-LAMP-DARPin Lentiviral Vector for Exression of HER2 Targeted DARPin on Exosome Surface. J. Maz. Univ. Med. Sci. 2017, 27, 12–23. [Google Scholar]
  85. Gomari, H.; Moghadam, M.F.; Soleimani, M. Targeted Cancer Therapy Using Engineered Exosome as a Natural Drug Delivery Vehicle. OncoTargets Ther. 2018, 11, 5753–5762. [Google Scholar] [CrossRef]
  86. Antes, T.J.; Middleton, R.C.; Luther, K.M.; Ijichi, T.; Peck, K.A.; Liu, W.J.; Valle, J.; Echavez, A.K.; Marbán, E. Targeting Extracellular Vesicles to Injured Tissue Using Membrane Cloaking and Surface Display. J. Nanobiotechnol. 2018, 16, 61. [Google Scholar] [CrossRef]
  87. 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]
  88. Khazaei-Poul, Y.; Shojaei, S.; Koochaki, A.; Ghanbarian, H.; Mohammadi-Yeganeh, S. Evaluating the Influence of Human Umbilical Cord Mesenchymal Stem Cells-Derived Exosomes Loaded with MiR-3182 on Metastatic Performance of Triple Negative Breast Cancer Cells. Life Sci. 2021, 286, 120015. [Google Scholar] [CrossRef]
  89. Yao, X.; Tu, Y.; Xu, Y.; Guo, Y.; Yao, F.; Zhang, X. Endoplasmic Reticulum Stress-Induced Exosomal MiR-27a-3p Promotes Immune Escape in Breast Cancer via Regulating PD-L1 Expression in Macrophages. J. Cell. Mol. Med. 2020, 24, 9560–9573. [Google Scholar] [CrossRef]
  90. 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]
  91. Limoni, S.K.; Moghadam, M.F.; Moazzeni, S.M.; Gomari, H.; Salimi, F. Engineered Exosomes for Targeted Transfer of SiRNA to HER2 Positive Breast Cancer Cells. Appl. Biochem. Biotechnol. 2018, 187, 352–364. [Google Scholar] [CrossRef]
  92. Chen, T.; Dong, Y.; Wu, X. Plasma Exosomal MiR-335-5p Serves as a Diagnostic Indicator and Inhibits Immune Escape in Triple-Negative Breast Cancer. Mol. Ther. Nucleic Acids 2022, 38, 347–356. [Google Scholar]
  93. O’Brien, K.; Lowry, M.C.; Corcoran, C.; Martinez, V.G.; Daly, M.; Rani, S.; Gallagher, W.M.; Radomski, M.W.; MacLeod, R.A.; O’Driscoll, L. miR-134 inhibits epithelial to mesenchymal transition by targeting FOXM1 in non-small cell lung cancer cells. FEBS Lett. 2012, 586, 3761–3765. [Google Scholar] [CrossRef]
  94. O’Brien, K.; Lowry, M.C.; Corcoran, C.; Martinez, V.G.; Daly, M.; Rani, S.; Gallagher, W.M.; Radomski, M.W.; MacLeod, R.A.F.; O’Driscoll, L. MiR-134 in Extracellular Vesicles Reduces Triple-Negative Breast Cancer Aggression and Increases Drug Sensitivity. Oncotarget 2015, 6, 32774–32789. [Google Scholar] [CrossRef] [Green Version]
  95. Su, X.; Zhang, L.; Li, H.; Cheng, P.; Zhu, Y.; Liu, Z.; Zhao, Y.; Xu, H.; Li, D.; Gao, H.; et al. MicroRNA-134 Targets KRAS to Suppress Breast Cancer Cell Proliferation, Migration and Invasion. Oncol. Lett. 2017, 13, 1932–1938. [Google Scholar] [CrossRef]
  96. Pan, J.Y.; Zhang, F.; Sun, C.C.; Li, S.J.; Li, G.; Gong, F.Y.; Bo, T.; He, J.; Hua, R.X.; Hu, W.D.; et al. MiR-134: A Human Cancer Suppressor? Mol. Ther. Nucleic Acids 2017, 6, 140. [Google Scholar] [CrossRef]
  97. Du, J.; Fan, J.J.; Dong, C.; Li, H.T.; Ma, B.L. Inhibition Effect of Exosomes-Mediated Let-7a on the Development and Metastasis of Triple Negative Breast Cancer by down-Regulating the Expression of c-Myc. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 5301–5314. [Google Scholar] [CrossRef]
  98. Li, Y.; Liang, Y.; Sang, Y.; Song, X.; Zhang, H.; Liu, Y.; Jiang, L.; Yang, Q. MiR-770 Suppresses the Chemo-Resistance and Metastasis of Triple Negative Breast Cancer via Direct Targeting of STMN1. Cell Death Dis. 2018, 9, 14. [Google Scholar] [CrossRef]
  99. Baroni, S.; Romero-Cordoba, S.; Plantamura, I.; Dugo, M.; D’Ippolito, E.; Cataldo, A.; Cosentino, G.; Angeloni, V.; Rossini, A.; Daidone, M.G.; et al. Exosome-Mediated Delivery of MiR-9 Induces Cancer-Associated Fibroblast-like Properties in Human Breast Fibroblasts. Cell Death Dis. 2016, 7, e2312. [Google Scholar] [CrossRef]
  100. 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]
  101. Naseri, Z.; Oskuee, R.K.; Jaafari, M.R.; Moghadam, M.F. Exosome-Mediated Delivery of Functionally Active MiRNA-142-3p Inhibitor Reduces Tumorigenicity of Breast Cancer In Vitro and In Vivo. Int. J. Nanomed. 2018, 13, 7727–7747. [Google Scholar] [CrossRef]
  102. Lee, S.J.; Jeong, J.H.; Lee, J.; Park, H.Y.; Jung, J.H.; Kang, J.; Kim, E.A.; Park, N.J.Y.; Park, J.Y.; Lee, I.H.; et al. MicroRNA-496 Inhibits Triple Negative Breast Cancer Cell Proliferation by Targeting Del-1. Medicine 2021, 100, e25270. [Google Scholar] [CrossRef]
  103. 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]
  104. Liu, X.; Zhang, G.; Yu, T.; He, J.; Liu, J.; Chai, X.; Zhao, G.; Yin, D.; Zhang, C. Exosomes Deliver LncRNA DARS-AS1 SiRNA to Inhibit Chronic Unpredictable Mild Stress-Induced TNBC Metastasis. Cancer Lett. 2022, 543, 215781. [Google Scholar] [CrossRef]
  105. 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]
  106. Zhao, L.; Gu, C.; Gan, Y.; Shao, L.; Chen, H.; Zhu, H. Exosome-Mediated SiRNA Delivery to Suppress Postoperative Breast Cancer Metastasis. J. Control. Release 2020, 318, 1–15. [Google Scholar] [CrossRef]
  107. 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]
  108. 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]
  109. 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]
  110. Xiao, H.; Li, H.; Yu, G.; Xiao, W.; Hu, J.; Tang, K.; Zeng, J.; He, W.; Zeng, G.; Ye, Z.; et al. MicroRNA-10b Promotes Migration and Invasion through KLF4 and HOXD10 in Human Bladder Cancer. Oncol. Rep. 2014, 31, 1832–1838. [Google Scholar] [CrossRef]
  111. Gong, C.; Tian, J.; Wang, Z.; Gao, Y.; Wu, X.; Ding, X.; Qiang, L.; Li, G.; Han, Z.; Yuan, Y.; et al. Functional Exosome-Mediated Co-Delivery of Doxorubicin and Hydrophobically Modified MicroRNA 159 for Triple-Negative Breast Cancer Therapy. J. Nanobiotechnol. 2019, 17, 93. [Google Scholar] [CrossRef]
  112. Kim, M.; Haney, M.; Zhao, Y.; Yuan, D.; Deygen, I.; Klyachko, N.L.; Kabanov, A.V.; Batrakova, E.V. Engineering Macrophage-Derived Exosomes for Targeted Paclitaxel Delivery to Pulmonary Metastases: In Vitro and In Vivo Evaluations. Nanomedicine 2018, 14, 195–204. [Google Scholar] [CrossRef]
  113. Cocco, S.; Piezzo, M.; Calabrese, A.; Cianniello, D.; Caputo, R.; di Lauro, V.; Fusco, G.; Gioia, G.D.; Licenziato, M.; de Laurentiis, M. Biomarkers in Triple-Negative Breast Cancer: State-of-the-Art and Future Perspectives. Int. J. Mol. Sci. 2020, 21, 4579. [Google Scholar] [CrossRef]
  114. Chen, J.; Song, Y.; Miao, F.; Chen, G.; Zhu, Y.; Wu, N.; Pang, L.; Chen, Z.; Chen, X. PDL1-positive Exosomes Suppress Antitumor Immunity by Inducing Tumor-specific CD8+ T Cell Exhaustion during Metastasis. Cancer Sci. 2021, 112, 3437–3454. [Google Scholar] [CrossRef]
  115. Shen, H.; Yang ES, H.; Conry, M.; Fiveash, J.; Contreras, C.; Bonner, J.A.; Shi, L.Z. Predictive Biomarkers for Immune Checkpoint Blockade and Opportunities for Combination Therapies. Genes Dis. 2019, 6, 232–246. [Google Scholar] [CrossRef]
  116. Liu, H.; Liang, Z.; Wang, F.; Zhou, C.; Zheng, X.; Hu, T.; He, X.; Wu, X.; Lan, P. Exosomes from Mesenchymal Stromal Cells Reduce Murine Colonic Inflammation via a Macrophage-Dependent Mechanism. JCI Insight 2019, 4, 131–273. [Google Scholar] [CrossRef]
  117. Biswas, S.; Mandal, G.; Chowdhury, S.R.; Purohit, S.; Payne, K.K.; Anadon, C.; Gupta, A.; Swanson, P.; Yu, X.; Conejo-Garcia, J.R.; et al. Exosomes Produced by Mesenchymal Stem Cells Drive Differentiation of Myeloid Cells into Immunosuppressive M2-Polarized Macrophages in Breast Cancer. J. Immunol. 2019, 203, 3447–3460. [Google Scholar] [CrossRef]
  118. Kim, O.Y.; Park, H.T.; Dinh, N.T.H.; Choi, S.J.; Lee, J.; Kim, J.H.; Lee, S.W.; Gho, Y.S. Bacterial Outer Membrane Vesicles Suppress Tumor by Interferon-γ-Mediated Antitumor Response. Nat. Commun. 2017, 8, 626. [Google Scholar] [CrossRef]
  119. Kameli, N.; Dragojlovic-kerkache, A.; Savelkoul, P.; Stassen, F.R. Plant-Derived Extracellular Vesicles: Current Findings, Challenges, and Future Applications. Membranes 2021, 11, 411. [Google Scholar] [CrossRef]
  120. Wang, G.; Hu, W.; Chen, H.; Shou, X.; Ye, T.; Xu, Y. Cocktail Strategy Based on NK Cell-Derived Exosomes and Their Biomimetic Nanoparticles for Dual Tumor Therapy. Cancers 2019, 11, 1560. [Google Scholar] [CrossRef]
  121. Somiya, M.; Yoshioka, Y.; Ochiya, T.; Somiya, M.; Yoshioka, Y.; Ochiya, T. Drug Delivery Application of Extracellular Vesicles; Insight into Production, Drug Loading, Targeting, and Pharmacokinetics. AIMS Bioeng. 2017, 4, 73–92. [Google Scholar] [CrossRef]
  122. Scherphof, G.L.; Dijkstra, J.; Spanjer, H.H.; Derksen, J.T.P.; Roerdink, F.H. Uptake and Intracellular Processing of Targeted and Nontargeted Liposomes by Rat Kupffer Cells In Vivo and In Vitroa. Ann. N. Y. Acad. Sci. 1985, 446, 368–384. [Google Scholar] [CrossRef] [PubMed]
  123. Besse, B.; Charrier, M.; Lapierre, V.; Dansin, E.; Lantz, O.; Planchard, D.; Le Chevalier, T.; Livartoski, A.; Barlesi, F.; Laplanche, A.; et al. Dendritic Cell-Derived Exosomes as Maintenance Immunotherapy after First Line Chemotherapy in NSCLC. Oncoimmunology 2016, 5, e1071008. [Google Scholar] [CrossRef]
  124. Tan, A.; de la Peña, H.; Seifalian, A.M. The Application of Exosomes as a Nanoscale Cancer Vaccine. Int. J. Nanomed. 2010, 5, 889–900. [Google Scholar] [CrossRef] [Green Version]
  125. Wolfers, J.; Lozier, A.; Raposo, G.; Regnault, A.; Théry, C.; Masurier, C.; Flament, C.; Pouzieux, S.; Faure, F.; Tursz, T.; et al. Tumor-Derived Exosomes Are a Source of Shared Tumor Rejection Antigens for CTL Cross-Priming. Nat. Med. 2001, 7, 297–303. [Google Scholar] [CrossRef]
  126. Zhang, X.; Yuan, X.; Shi, H.; Wu, L.; Qian, H.; Xu, W. Exosomes in Cancer: Small Particle, Big Player. J. Hematol. Oncol. 2015, 8, 83. [Google Scholar] [CrossRef]
  127. Zitvogel, L.; Regnault, A.; Lozier, A.; Wolfers, J.; Flament, C.; Tenza, D.; Ricciardi-Castagnoli, P.; Raposo, G.; Amigorena, S. Eradication of Established Murine Tumors Using a Novel Cell-Free Vaccine: Dendritic Cell Derived Exosomes. Nat. Med. 1998, 4, 594–600. [Google Scholar] [CrossRef]
  128. Dong Gao, L.J. Exosomes in Cancer Therapy: A Novel Experimental Strategy—PubMed. Am. J. Cancer Res. 2018, 8, 2165–2175. [Google Scholar]
  129. 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]
  130. St-Denis-bissonnette, F.; Khoury, R.; Mediratta, K.; El-Sahli, S.; Wang, L.; Lavoie, J.R. Applications of Extracellular Vesicles in Triple-Negative Breast Cancer. Cancers 2022, 14, 451. [Google Scholar] [CrossRef]
  131. Mediratta, K.; El-Sahli, S.; D’costa, V.; Wang, L. Current Progresses and Challenges of Immunotherapy in Triple-Negative Breast Cancer. Cancers 2020, 12, 3529. [Google Scholar] [CrossRef]
Figure 1. Exosomes as a biomarker for TNBC—showing different protein contents inside exosomes, which are used for identifying TNBC and also in ‘liquid biopsy’. Carcinoembryonic antigen (CEA), survivin, CA 15-3(Cancer antigen 15-3), miR-373, mi-134, miR-1910-3p, and miR-155 are some of the biomarkers (https://biorender.com/, accessed on 30 October 2022).
Figure 1. Exosomes as a biomarker for TNBC—showing different protein contents inside exosomes, which are used for identifying TNBC and also in ‘liquid biopsy’. Carcinoembryonic antigen (CEA), survivin, CA 15-3(Cancer antigen 15-3), miR-373, mi-134, miR-1910-3p, and miR-155 are some of the biomarkers (https://biorender.com/, accessed on 30 October 2022).
Molecules 28 01802 g001
Figure 2. Exosomes targeting different pathways in TNBC and breast cancer leading to epithelial–mesenchymal transition of breast tumours to other parts of the body (exosomes have circ_0076611 target MYC and VEGFA genes and initiate epithelial–mesenchymal transition). NOTCH signalling initiates ASPH in breast cancer cell lines and triggers epithelial–mesenchymal transition. Exosomes extracted from adipose mesenchymal stem cells inhibit Hippo signalling and initiate the TEA domain transcription factor to bind with the unphosphorylated YAP/TAZ, which translocates it to the nucleus. TEAD-influenced target gene transcription enhances the expression of mesenchymal markers such as Vimentin and N-cadherin and suppresses the expression of epithelial markers including E-cadherin. The YAP gene cross-communicates with transcription factors such as ZEB1, SNAIL, and SLUG, which initiates epithelial–mesenchymal transition in cancer cells. MMP-1 interacts with PAR1, a G protein receptor that initiates the EMT. Macrophage-derived exosomes containing Wnt5a and Wnt7a activate Wnt/β signalling, which, in turn, activates SNAIL, SLUG, and TWIST and initiates EMT. miR-939 in exosomes downregulates the expression of VE-cadherin, destroys the barrier function of endothelial monolayers, and initiates tumourigenesis in breast cancer. (Created with the help of Biorender).
Figure 2. Exosomes targeting different pathways in TNBC and breast cancer leading to epithelial–mesenchymal transition of breast tumours to other parts of the body (exosomes have circ_0076611 target MYC and VEGFA genes and initiate epithelial–mesenchymal transition). NOTCH signalling initiates ASPH in breast cancer cell lines and triggers epithelial–mesenchymal transition. Exosomes extracted from adipose mesenchymal stem cells inhibit Hippo signalling and initiate the TEA domain transcription factor to bind with the unphosphorylated YAP/TAZ, which translocates it to the nucleus. TEAD-influenced target gene transcription enhances the expression of mesenchymal markers such as Vimentin and N-cadherin and suppresses the expression of epithelial markers including E-cadherin. The YAP gene cross-communicates with transcription factors such as ZEB1, SNAIL, and SLUG, which initiates epithelial–mesenchymal transition in cancer cells. MMP-1 interacts with PAR1, a G protein receptor that initiates the EMT. Macrophage-derived exosomes containing Wnt5a and Wnt7a activate Wnt/β signalling, which, in turn, activates SNAIL, SLUG, and TWIST and initiates EMT. miR-939 in exosomes downregulates the expression of VE-cadherin, destroys the barrier function of endothelial monolayers, and initiates tumourigenesis in breast cancer. (Created with the help of Biorender).
Molecules 28 01802 g002
Figure 3. Extraction of exosomes from different sources such as ginger and grapes, stem cells, and bacterial cells using different methods such as ultracentrifugation; the influence of external agents on the production of exosomes; the loading of the exosomes with miRNA, siRNA, and drugs; engineering of exosomes by protein, antibody, nanoparticle, or hydrazine linker, finally using it as drug delivery; and its effect on the tumour (https://biorender.com/, accessed on 30 October 2022).
Figure 3. Extraction of exosomes from different sources such as ginger and grapes, stem cells, and bacterial cells using different methods such as ultracentrifugation; the influence of external agents on the production of exosomes; the loading of the exosomes with miRNA, siRNA, and drugs; engineering of exosomes by protein, antibody, nanoparticle, or hydrazine linker, finally using it as drug delivery; and its effect on the tumour (https://biorender.com/, accessed on 30 October 2022).
Molecules 28 01802 g003
Figure 4. Exosomes generated from adipose mesenchymal stem cells infused with miRNA-381-3p downregulate the expression of Wnt signalling and inhibit EMT. miR3182 conjugated with exosomes derived from the human umbilical cord induce apoptosis in the TNBC cell line through downregulating genes such as mTOR and S6KB1. Exosomal miR-27a-3p, through production by endoplasmic reticulum stress, enhances immunological escape in breast cancer through modulating PD-L1 levels in macrophage’s immune evasion via the PTEN/AKT/PI3K axis. microRNA 159 and doxorubicin cause a synergetic effect on the TCF-7 gene and improve the anticancer effects. Exosomes containing miRNA-127, miR-197, miR-222, and miR-223 from stroma also aid breast cancer cell quiescence and enhance anti-proliferation rates. miR-134 targets the pathways such as MAPK/ERK and other signallings such as EGFR and NOTCH. Upward expression of miR-134 also represses breast cancer metastasis by targeting KRAS. miR-9 and miR-155 target PTEN and DUSP14 tumour suppressor genes in MCF-7 cell lines extracted from MDA-MB-231 exosomes. miR-496 and miR-137 loaded in exosomes from the MCF10A cell line target Del-1 and modulate it by inhibiting tumour growth in the TNBC cell lines such as MCF10A and MDA-MB-231. Extracellular vesicles derived from adipose mesenchymal cells enhance apoptosis by suppressing PD-L1 signalling in MDA-MB-231 TNBC cells.
Figure 4. Exosomes generated from adipose mesenchymal stem cells infused with miRNA-381-3p downregulate the expression of Wnt signalling and inhibit EMT. miR3182 conjugated with exosomes derived from the human umbilical cord induce apoptosis in the TNBC cell line through downregulating genes such as mTOR and S6KB1. Exosomal miR-27a-3p, through production by endoplasmic reticulum stress, enhances immunological escape in breast cancer through modulating PD-L1 levels in macrophage’s immune evasion via the PTEN/AKT/PI3K axis. microRNA 159 and doxorubicin cause a synergetic effect on the TCF-7 gene and improve the anticancer effects. Exosomes containing miRNA-127, miR-197, miR-222, and miR-223 from stroma also aid breast cancer cell quiescence and enhance anti-proliferation rates. miR-134 targets the pathways such as MAPK/ERK and other signallings such as EGFR and NOTCH. Upward expression of miR-134 also represses breast cancer metastasis by targeting KRAS. miR-9 and miR-155 target PTEN and DUSP14 tumour suppressor genes in MCF-7 cell lines extracted from MDA-MB-231 exosomes. miR-496 and miR-137 loaded in exosomes from the MCF10A cell line target Del-1 and modulate it by inhibiting tumour growth in the TNBC cell lines such as MCF10A and MDA-MB-231. Extracellular vesicles derived from adipose mesenchymal cells enhance apoptosis by suppressing PD-L1 signalling in MDA-MB-231 TNBC cells.
Molecules 28 01802 g004
Table 1. Engineered exosomes with different proteins embedded their membranes, and their effects on triple-negative breast cancer.
Table 1. Engineered exosomes with different proteins embedded their membranes, and their effects on triple-negative breast cancer.
Engineered Exosomes Element in Membrane/SourceEffectReported Effect and OutcomesReferences
pLEX-LAMP DARPin for expression of DARPinHER2+ cellsInhibit metastasis[84,85]
Mesothelin MSLNCAR-T cells with CARs and CD3 surface expressionInhibit metastasis[78]
Poly (lactic-co-glycolic acid)MDA-MB-231Target c-Met and inhibit metastasis[80]
Anti-HER2 antibody conjugated paclitaxel-loaded liposomesMDA-MB-231Inhibit metastasis, boost the therapy, increase apoptosis[81]
Exosome A15 derived from monocyte-derived macrophagesMDA-MB-231Effect on TCF-7 gene leading to improved anticancer effects[90]
Table 2. Engineered exosomes loaded with different molecular drugs, microRNA, siRNA targeting different signalling, and genes in triple-negative breast cancer.
Table 2. Engineered exosomes loaded with different molecular drugs, microRNA, siRNA targeting different signalling, and genes in triple-negative breast cancer.
CargoEffectReported Effect and OutcomesRefs.
miR-134Hs578Ts(i)8Downregulate STAT5B, HSP90, and KRAS[93,94,95,96]
let-7aMDA-MB-231Silence c-Myc gene[97]
miRNA-770 and DoxorubicinMDA-MB-231HER2+ tumour[98]
CARMSLN + TNBCPerforin and Granzyme B mechanism [78]
ErastinMDA-MB-231Ferroptosis[82]
Anti-IL-3R-EVMetastasis of TNBC to other parts, liver, lungsLower Vimentin, β-catenin, and TWIST1[83]
Anti-IL-3R-EVs and antago-miR-24-3p-EVsMDA-MB-231Upregulate SPRY2, enhance apoptosis [83]
miRNA-381-3p from adipose mesenchymal stem cellsMDA-MB-231Decrease Wnt signalling and factors related to EMT[87]
miR3182MDA-MB-231Apoptosis in TNBC to downregulate mTOR and S6KB1[88]
miR-27a-3pMDA-MB-231Modulate PD-L1 levels in macrophages causing immune evasion via the PTEN/AKT/PI3K axis[89]
microRNA 159MDA-MB-231Activate protein kinase C further TCF-7[90]
miRNA-127, miR-197, miR-222, and miR-223MDA-MB-231Enhance apoptosis, metastasis [90]
siRNA from HEK293T cellsHER2 Positive Breast CancerTPD52 gene downregulated by 70 percent[91]
miR-9 and miR-155 from MDA-MB-231MCF-7PTEN and DUSP14 tumour suppressor gene[100]
Anti-miR-142-3pMDA-MB-231Lower miR-142-3p and miR-150 levels while increasing the level of target genes APC and P2X7R in TNBC[101]
miR-496 and miR-137 from MCF10AMDA-MB-231Del-1[102]
miR-424-5pMDA-MB-231Suppress PD-L1 enhancing apoptosis[103]
Table 3. List of all clinical trials with exosomes loaded with drugs in triple-negative breast cancer [131].
Table 3. List of all clinical trials with exosomes loaded with drugs in triple-negative breast cancer [131].
Trial (National Clinical Trial ID)PhaseConditionInterventions
NCT03362060 1TNBC, Metastatic TNBCPembrolizumab Biological: PVX-410
NCT041055821TNBC BC Biological: Neo-antigen pulsed DCs
NCT03199040 1TNBC Metastatic TNBCDrug: Durvalumab Biological: Neoantigen DNA vaccine Device: TDS-IM system (Anchor Medical Systems)
NCT02316457 I TNBCBiological: IVAC_W_bre1_uID Biological: IVAC_W_bre1_uID/IVAC_M_uID
NCT04024800 II TNBC Biological: AE37 Peptide vaccine
NCT028264341BCBiological: PVX-410 Biological: Durvalumab Drug: Hilton
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

Banerjee, M.; Rajeswari, V.D. Critical Review on the Different Roles of Exosomes in TNBC and Exosomal-Mediated Delivery of microRNA/siRNA/lncRNA and Drug Targeting Signalling Pathways in Triple-Negative Breast Cancer. Molecules 2023, 28, 1802. https://doi.org/10.3390/molecules28041802

AMA Style

Banerjee M, Rajeswari VD. Critical Review on the Different Roles of Exosomes in TNBC and Exosomal-Mediated Delivery of microRNA/siRNA/lncRNA and Drug Targeting Signalling Pathways in Triple-Negative Breast Cancer. Molecules. 2023; 28(4):1802. https://doi.org/10.3390/molecules28041802

Chicago/Turabian Style

Banerjee, Manosi, and Vijayarangan Devi Rajeswari. 2023. "Critical Review on the Different Roles of Exosomes in TNBC and Exosomal-Mediated Delivery of microRNA/siRNA/lncRNA and Drug Targeting Signalling Pathways in Triple-Negative Breast Cancer" Molecules 28, no. 4: 1802. https://doi.org/10.3390/molecules28041802

Article Metrics

Back to TopTop