Next Article in Journal
Senescent Markers Expressed by Periodontal Ligament-Derived Stem Cells (PDLSCs) Harvested from Patients with Periodontitis Can Be Rejuvenated by RG108
Next Article in Special Issue
Red Blood Cell-Derived Extracellular Vesicles: An Overview of Current Research Progress, Challenges, and Opportunities
Previous Article in Journal
Processing Adipose Tissue Samples in a GMP Environment Standardizes the Use of SVF in Cell Therapy Treatments: Data on 302 Patients
Previous Article in Special Issue
Advances in the Therapeutic Applications of Plant-Derived Exosomes in the Treatment of Inflammatory Diseases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Communicator Extraordinaire: Extracellular Vesicles in the Tumor Microenvironment Are Essential Local and Long-Distance Mediators of Cancer Metastasis

Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA
*
Author to whom correspondence should be addressed.
Biomedicines 2023, 11(9), 2534; https://doi.org/10.3390/biomedicines11092534
Submission received: 16 June 2023 / Revised: 30 August 2023 / Accepted: 3 September 2023 / Published: 14 September 2023
(This article belongs to the Special Issue Exosomes and Their Role in Diseases)

Abstract

:
Human tumors are increasingly being described as a complex “ecosystem”, that includes many different cell types, secreted growth factors, extracellular matrix (ECM) components, and microvessels, that altogether create the tumor microenvironment (TME). Within the TME, epithelial cancer cells control the function of surrounding stromal cells and the non-cellular ECM components in an intricate orchestra of signaling networks specifically designed for cancer cells to exploit surrounding cells for their own benefit. Tumor-derived extracellular vesicles (EVs) released into the tumor microenvironment are essential mediators in the reprogramming of surrounding stromal cells, which include cancer-associated fibroblasts (CAFs), tumor-associated macrophages (TAMs), tumor-infiltrating lymphocytes (TILs), and tumor endothelial cells (TECs), which are responsible for the promotion of neo-angiogenesis, immune cell evasion, and invasion which are essential for cancer progression. Perhaps most importantly, tumor-derived EVs play critical roles in the metastatic dissemination of tumor cells through their two-fold role in initiating cancer cell invasion and the establishment of the pre-metastatic niche, both of which are vital for tumor cell migration, homing, and colonization at secondary tumor sites. This review discusses extracellular vesicle trafficking within the tumor microenvironment and pre-metastatic niche formation, focusing on the complex role that EVs play in orchestrating cancer-to-stromal cell communication in order to promote the metastatic dissemination of cancer cells.

1. Introduction

Although solid tumors are generally composed of monoclonal cancerous epithelial cells, they are a complex “ecosystem” that incorporates different cell types, secreted factors, extracellular matrix (ECM), and microvessels, all contributing to this biological entity [1]. The infiltration of stromal cells such as inflammatory immune cells (i.e., macrophages, dendritic cells, neutrophils, and myeloid-derived suppressor cells), adipocytes, fibroblasts, and endothelial cells in addition to non-cancerous and cancerous epithelial cells, all contribute to what is termed the tumor microenvironment (TME) [2,3,4]. The TME comprises cancerous epithelial cells that have gained the ability to control the function of surrounding stromal cells and the secretion/composition of non-cellular ECM components in an intricate orchestra of signaling networks designed for these cancer cells to exploit surrounding non-malignant cells to their benefit [5]. The “reverse Warburg effect” is one classic example wherein epithelial cancer cells induce the metabolic reprogramming of surrounding stromal cells to enhance aerobic glycolysis and their production of lactate [6]. Cancer cell metabolism is unique when compared to that of normal cells in that even in the presence of sufficient oxygen, tumor cells rely predominantly on oxidative phosphorylation (OXPHOS) rather than glycolysis for the production of adenosine triphosphate (ATP) (i.e., energy) [7,8]. This process was described in 1927 by Otto Warburg and has been termed the “Warburg effect” [9]. In addition to the enhanced energy supply that the recruitment of stromal cells into the TME has, the infiltration of stromal cells has also extensively been shown to promote neo-angiogenesis [10], epithelial cell migration [11,12], and extracellular matrix remodeling [13,14]. Additionally, stromal influences are known to contribute significantly to both the evasion of immune surveillance and enhanced chemotherapeutic resistance of cancer cells [15,16], which are largely attributed to the increased production of several stromal derived chemokines, cytokines [17,18], that include tumor necrosis factor alpha (TNF-α) [19], interleukin-6 (IL-6) [20], transforming growth factor beta (TGF-β) [21], and interleukin-10 (IL-10) [22], and growth factors. In recent years, however, circulating extracellular vesicles and other types of extracellular vesicles have gained in importance with regards to the delivery and biological effects of their packaged cargo [23,24,25]. Indeed, these membrane-bound biological nanoparticles have been shown to deliver a variety of important signals locally as well as at long distances in the organism to enable cell-to-cell communication. To date, the known ways of cellular communication that have been clearly identified for extracellular vesicles during metastasis include (1) their transfer from one cancer cell to another, to promote epithelial-to-mesenchymal transition (EMT) and regulate metastasis, and (2) their transfer from cancer cells to stromal cells and vice versa [26,27,28].
Extracellular vesicles (EVs), a term which is collectively used to describe nanosomes, exosomes, microvesicles (MVs), and apoptotic bodies, are small bi-layered membrane-bound vesicles, typically ranging from 8 to 12 nm, 30 to 150 nm, 200 to 1000 nm, and 500+ nm in diameter, respectively [29]. Apoptotic bodies are formed during the process of apoptotic cell death when the cytoskeleton of cells breaks down resulting in the outward blebbing of the membrane and the splitting of cellular content into distinct membrane-enclosed vesicles [30]. Microvesicles and nanovesicles are formed via the outward blebbing and detachment of the plasma membrane. While not completely understood, it is believed that the mechanism governing the formation and release of microvesicles is a controlled process which utilizes the endosomal machinery for vesicle formation [31]. Exosome formation, which is distinct from the formation of both apoptotic bodies and microvesicles, originates from the inward budding of the plasma membrane with the subsequent formation of multivesicular bodies (MVBs) through the endosomal pathway which are ultimately released into the extracellular matrix upon the fusion of MVBs with the plasma membrane [32] (Figure 1). The international society for extracellular vesicles (ISEV) published the minimal information for studies of extracellular vesicles 2018 (MISEV2018) guidelines wherein they endorse the utilization of the term “extracellular vesicle” (EV) for any naturally released particles from cells which are enclosed by a double lipid membrane and cannot self-replicate. This is because the assignment of EVs to a particular biogenesis pathway remains extraordinarily difficult; as such, the ISEV urges authors to make use of the nomenclature “EVs” in place of terms such as “exosomes” and “microvesicles” [33]. In accordance with these guidelines in this review we utilize the term EV for all descriptions; however, where appropriate if cited papers have utilize the term “exosome” and the manuscript contains sufficient data that include multiple experimentation methods demonstrating biophysical characteristics of EVs (i.e., including the composition of tetraspanin markers, transmission electron microscopy, and/or nanoparticle tracking), and descriptions of cellular origin, we refer to these vesicles as “exosome-like vesicles” in this manuscript. Exosome-like vesicles are known to carry common surface molecules that include several tetraspanin transmembrane proteins such as Alix, CD63, CD81, and CD9 [34,35,36], but EVs in general can also carry these plasma membrane markers in addition to other cell-type specific surface markers that are acquired during their normal biogenesis. Particular examples include: prostate specific antigen (PSA) for exosome-like vesicles produced by prostate cells [37], asialoglycoprotein receptor 1 (ASGR1) for EVs produced by liver cells [38], microglial proteins (CD11b and CD45) for exosome-like vesicles found in cerebral spinal fluid (CSF) [39], the Clara cell protein 16 (CC16) for exosome-like vesicles present in bronchoalveolar lavage fluid (BALF) [40], ACE2 for SARS-CoV-2 spike protein-guided EV isolation from plasma [41] and the placental alkaline phosphatase (PLAP) for exosome-like vesicles produced by placental cells [42].
The size, membrane composition, and content of EVs are known to be heterogeneous and highly dependent on the cellular state, source, and the environmental conditions of the cells from which they originate [43]. As such, the composition of EV cargo (i.e., their miRNA, lncRNA, DNA, and proteins) has been shown to partially reflect the metabolic status of their cells of origin. Specifically, during periods of enhanced cellular stress and disease progression a high degree of cellular sorting into EVs is observed [44]. Although the content of EVs provides an indirect reflection of the state of the cell from which they are derived, it does not represent a mirror image of the cellular content, as EVs may be enriched in low-expressed RNAs or proteins from their cells of origin, while being depleted in others [45]. The molecular mechanisms involved in packaging their cargo are highly intricate and specialized processes [32]. The packaging of EV cargo, particularly in cancer cells, appears to favor the enrichment of specific proteins, lipids, and RNAs over others [46]. Proteins enriched within EVs derived from tumor cells have been shown to include heat shock proteins (HSP70, HSP90), export molecules (RAB27a/b, Alix, TSG101), proteases (ADAM10), and metabolic enzymes (enolase-α, glyceraldehyde 3 phosphate dehydrogenase, ATP synthase) [47]. EVs are also enriched with a variety of both short and long RNA species [48], including tRNA, vault RNA, and miRNAs [49].
The biogenesis and release of EVs is affected during disease, and in particular EV secretion from tumor cells is known to be exacerbated, which in turn enhances tumorigenesis and cancer progression [50]. The enhanced secretion and subsequent fusion of tumor-derived EVs with target cells, either locally within the TME or at a long distance in pre-metastatic tissues, facilitates the horizontal transfer of both their cell surface receptors and their internal cargos, which exhibit multifaceted and oftentimes double-edged functionality in recipient cells [51]. The complex composition of EV cargo during tumor progression coupled with their enhanced ability to travel through the body by diffusion, through the circulation, or within biofluids, further highlights that they are well adapted devices to establish and maintain the control of cancer cells over other normal cells. Additionally, because their cargo represents an encapsulated molecular snapshot of their cellular origin, they are potent reservoirs of biomarkers not only for initial cancer diagnosis but also for the monitoring of metastasis. Indeed, several studies have already demonstrated that EVs derived from tumor cells can play a significant role in promoting angiogenesis [52], stromal remodeling [53], enhanced immune surveillance [54], and most notably metastasis [55,56]. While the acidity of the tumor microenvironment is known to play a critical role in several cellular processes prominent in cancer, including chemotherapeutic resistance [57,58,59,60] and metastatic dissemination [61], several studies have suggested that the TME may itself contribute to the regulation of EV trafficking within the tumor mass [62]. For example, several studies have shown that the acidity of the tumor microenvironment contributes to the enhanced EV release and that it also facilitates the fusion of EVs with their recipient cells, through lipid–lipid interactions [62,63].
Considering the potent roles of extracellular vesicles and their local release in the tumor microenvironment, in this review we provide an extensive overview of EV trafficking within the tumor microenvironment, focusing primarily on the complex role that EVs play in orchestrating cancer-to-stromal cell communication within the tumor microenvironment and the pre-metastatic niche for the purpose of promoting the metastatic dissemination of cancer cells.

2. The Tumor Microenvironment

It is increasingly recognized that the complex heterogeneous nature of the tumor microenvironment plays a critical role in the evolution and progression of cancer. The communication between matrix cells within the TME, via extracellular vesicles, serves as an essential mediator for the development, recurrence, and metastatic dissemination of several cancer types [64]. The role of EVs within the tumor microenvironment appears to be multifaceted and bidirectional as the initial release of EVs from cancer cells leads to the recruitment and activation of stromal cells, which includes cancer-associated fibroblasts (CAFs), tumor-associated macrophages (TAM), cancer-associated endothelial cells (CAEC), and mesenchymal stem cells (MSC) [65]. As tumor growth progresses, evidence suggests that EVs released from surrounding stromal cells drive the epithelial-to-mesenchymal transition (EMT) of cancer cells, and thus their progression to more metastatic phenotypes [66,67]. Complementarily, as the release of EVs is enhanced during tumor growth, for certain tumors it allows their widespread diffusion and provides them with the potential to establish the pre-metastatic niche, which is necessary for the successful dissemination, colonization, and expansion of these cells to distant organ sites [68].

2.1. Mechanisms of Extracellular Vesicle-Mediated Metabolic Reprogramming and TME Remodeling in Metastasis

2.1.1. Cancer-Associated Fibroblasts (CAFs)

Fibroblasts stem from a mesenchymal origin and are considered to be the most abundant cell type present in connective tissue (i.e., the supportive tissue of an organ) [69,70]. Through their secretion of collagen proteins, elastins, adhesive proteins (e.g., laminin and fibronectin), and ground substance (e.g., glycosaminoglycans), they maintain integrity of the extracellular matrix (ECM), thereby providing structural support to tissues and organs [71]. Under normal homeostatic conditions, fibroblasts remain quiescent as spindle-shaped single cells embedded within the ECM in the interstitial space, however, in response to tissue damage/injury they become reversibly “activated” to promote tissue repair during the wound healing process [72]. Once “activated” these fibroblasts display enhanced expression of α-smooth muscle actin (αSMA) and vimentin, and, together with increased ECM production and cytoskeleton remodeling, become stellate in shape (i.e., star-like) and gain contractile properties [6], at which point they are identified as being mesenchymal stem cell-like with characteristic signs of smooth muscle that has granted them the name of myofibroblasts.
In many instances, tumors are analogous to a wound as the expansion of new cells and chronic inflammation results in a continuous state of tissue injury, and the chronic activation of a wound healing/fibrotic response, and as such tumors are often referred to as wounds that do not heal [73,74,75]. Within the TME, fibroblasts that are “activated” by tumor cells are termed cancer-associated fibroblasts (CAFs), which typically constitute the most abundant stromal cell type present within the tumor microenvironment of solid tumors, including those of the colon, breast, and pancreas [76]. In pancreatic cancer for example, the tumor mass typically comprises 60–70% stromal CAFs, enhanced collagen, and other ECM components [77]. The intercellular communication between CAFs and tumor cells occurs via multiple ways, by direct cell–cell contact, by the transfer of secreted molecules, and by secreted extracellular vesicles that include EVs. It has been recently shown that the secretion of EVs is an important way for CAFs to influence the behavior of cancer cells (and vice versa) [78,79].
Understanding the role that EVs play in the communication between surrounding stromal cells and epithelial cancer cells, and vice versa, is critical for elucidating the influence of the tumor microenvironment during cancer cell progression and/or metastasis. The inherent mechanisms involved in the “activation” of CAFs within the tumor microenvironment appear to be related to EMT-induced differentiation of both resident stromal cells and recruited bone marrow-derived stem cells, which result in the formation of myofibroblasts. The initiation of CAF “activation” is predominantly driven by transforming growth factor beta 1 (TGF-β1) and fibroblast growth factor-2 (FGF-2) [80]. Interestingly, it has been recently shown that EV-derived TGF-β1 supplied by cancer cells is the only essential requirement for the differentiation of fibroblasts into CAFs [81], supporting the role of EVs for maintenance and modification of the tumor microenvironment. Similarly, ovarian and breast cancer-derived EVs have been shown to induce the conversion of adipose-derived mesenchymal stem cells into myofibroblast-like cells [82,83]. Within the TME, CAFs are predominantly responsible for the production of essential extracellular matrix proteins (such as fibronectin and collagens) and proteases [84]. It has been shown that increased production of these proteins leads to the progressive stiffening of the extracellular matrix, thereby facilitating tumor progression, vascularization, and metastasis [85]. The EV release of bioactive molecules, such as platelet-derived growth factor (PDGF), hepatocyte growth factor (HGF), interleukin-6 (IL6), proteases, and miRNAs [86] by CAFs into the extracellular matrix further supports their importance in cell–cell communication within the tumor microenvironment. In breast cancer for example, a study by Luga et al. showed that EVs released from surrounding stromal cells can promote breast cancer cell motility and metastasis through the mobilization of Wnt11-induced planar cell polarity [87]. Indeed, Luga and colleagues demonstrated that EVs released from cancer-associated fibroblasts (CAFs) promote the increased motility and metastatic capability of breast cancer cells, which is dependent on the interaction of breast cancer cell-produced Wnt11 and CAF-derived CD81 [87]. Another study on lung cancer cells also showed that microvesicles containing the extracellular matrix MMPs inducer (EMMPRIN) can stimulate the expression of matrix metalloproteinases (MMPs) in CAFs, which results in enhanced tumor metastization [88]. Other studies, performed on prostate cancer cell-derived EVs, which contain significantly increased levels of IL6, TGF-β, MMPs, carbonic anhydrase IX, and tumor necrosis factor 1α (TNF-1α) reported the induction of a stem cell-like phenotype thus enhancing metastasis under hypoxic conditions [89,90,91].

2.1.2. Tumor-Associated Macrophages (TAMs)

Macrophages are a key population of innate immune cells responsible for executing a broad spectrum of functions ranging from the modulation of tissue homeostasis to the defense against pathogens and the facilitation of wound healing [91]. Macrophages that have infiltrated into the microenvironment of solid tumors are termed tumor-associated macrophages (TAMs) and form a critical component of the TME. Many of the TAMS located within the TME originate from bone marrow monocyte precursors and are recruited into the TME as a result of the tumor-derived chemoattractants that are continuously present within tumors. TAMs affect tumor growth, angiogenesis, metastasis, and chemoresistance. Within tumors, most TAMs gather at the leading edge and within hypoxic avascular areas [92], while a few also align along the abluminal side (i.e., away from the lumen) of the vessels [93]. They are recruited and activated by various signals in the TME and can have dramatic impacts on tumor progression and metastasis. TAMs have been demonstrated to perform a diverse range of immune regulatory functions and tumor progression, including that of promoting cancer cell proliferation and invasive capacity. In fact, tumor-elicited inflammation promotes tumor growth via the presence of the TAM-derived inflammatory cytokines interleukin (IL)-23 and IL-17 [94]. Moreover, an increase in TAM-derived IL-6 has been shown to contribute to STAT3 signaling induced hepatocellular carcinoma development and progression [95].
Macrophages can display different and even opposing phenotypes, depending on the microenvironment which they are embedded in. Once activated, macrophages are often classified as having either an M1 (classical-activated macrophages) or M2 (alternative-activated macrophages) phenotype [96]. M1 macrophages typically promote an inflammatory response against invading pathogens and tumor cells, whereas M2 macrophages tend to exert an immune-suppressive phenotype, which favors tissue repair and tumor progression. Each polarized macrophage type displays distinct expression profiles of genes, cytokines, and cell-surface markers [97]. Among those factors, colony-stimulating factor 1 (CSF-1) and C-C motif ligand 2 (CCL2) are the two most well-documented macrophage recruiters and M2-stimulating factors. CSF-1 is a potent determinant factor of macrophage polarization, as CSF-1 overexpression is often observed at the invasive edge of various tumors and correlates with a significant increase in metastasis [98].
Many studies have attempted to elucidate the crosstalk that exists between tumor and immune cells within the TME. Studies have shown that tumor-derived EVs play a vital role in the conversion of monocyte-derived macrophages into regulatory macrophages and in the mediation of cancer-related inflammation and tumor development [99,100] through the transfer of their cargos to recipient cells within the TME [101,102]. These cargos include proteins, nucleic acids, and lipids. Several studies have shown that depletion of EVs can disrupt the communication between tumor cells and TAMs, which reverses some of the harmful effects that EVs exert during tumor progression, restoring chemotherapeutic drug sensitivity [103,104,105].
In recent years, several studies have shown that EV miRNAs can play a crucial role in tumor progression, through their ability to regulate angiogenesis and facilitate metastasis by interfering with normal cellular programs of recipient cells [106,107]. Tumor-derived EV miRNAs have been shown to polarize recipient macrophages by targeting several signaling pathways, which can positively or negatively impact tumor progression [65,108]. It has been shown that tumor-derived EV miRNAs can promote cancer metastasis by regulating the crosstalk between cancer cells and TAMs, also providing a therapeutic strategy for cancer therapy. For example, colorectal-derived EVs carrying miR-203 are incorporated into monocytes [109], while EVs carrying miR-145 [110] and exosome-like vesicles containing miR-934 [108] are taken up by macrophages, which leads to their polarization into the M2 phenotype. M2 polarization induced by EVs derived from oral squamous cell carcinoma harboring miR-29a-3p has been shown to target (SOCS)1/STAT6 signaling, directly promoting tumor growth [111]. Whereas, hypoxia in ovarian cancer induces the production of EVs enriched in miR-940, miR-21-3p, miR-125b-5p, miR-181d-5p, and miR-222-3p stimulates macrophage M2 phenotype polarization and enhanced tumor growth [112,113]. Similarly, hypoxia-induced miR-301a-3p-enrichment in lung cancer-derived exosome-like vesicles results in a HIF1a/2a-dependent polarization of TAMs to the M2 phenotype, facilitating enhanced cell invasion, migration, and epithelial–mesenchymal transition (EMT) during lung metastasis [114].

2.1.3. Tumor Endothelial Cells (TECs)

Angiogenesis, also known as new blood vessel formation, is essential for tumor progression and metastasis. The onset of angiogenesis occurs at any stage of tumor progression and depends on the type of tumor and its microenvironment. Endothelial cells, in the majority of solid tumors, are located within the inner layer of blood vessels and compared to normal endothelial cells have an altered morphology and molecular phenotype. Tumor blood vessels are characteristically unorganized, where they are often thin, fragile, and defective in barrier function resulting in leakiness of tumor blood vessels, whereas normal vasculature shows a hierarchal branching pattern of arteries, veins, and capillaries [115]. The unorganized nature of tumor blood vessels means that within specific focal regions they lack endothelial cells or basement membrane [116], which results in their exhibited chaotic blood flow (often termed leaky/hemorrhagic) [117]. Additionally, the high interstitial fluid pressure that is present within solid tumors causes blood vessels to collapse and further impedes blood flow. As such, hypoxic regions within the tumor tissue develop, despite the high level of vascularization [118]. During tumor formation and progression epithelial cancer cells actively secrete several pro-angiogenic factors which result in the excessive formation of abnormal blood vessels. Several studies have highlighted the differences between normal endothelial cells and tumor endothelial cells (TECs) [119]. In particular, the release of vascular endothelial growth factor (VEGF) in addition to other growth factors belonging to the ephrin and angiopoietin families from TECs is essential in promoting the formation of tumor blood vessels [120]. Hida et al. demonstrated that when compared to normal endothelial cells, TECs display several abnormalities [121,122] including differences in their responsiveness to epidermal growth factor (EGF) [123], adrenomedullin [124], and VEGF [125]. Ultimately, the differences in their response to these growth factors are important in the proangiogenic phenotype of TECs [122]. In particular, VEGF has been shown to stimulate cell migration and enhances survival of TECs in an autocrine manner.
Several studies have demonstrated that like CAFs and TAMs, the infiltration and metabolic switch of endothelial cells to TECs within the tumor microenvironment supports and enhances angiogenesis during tumor growth and morphologically abnormal tumor vasculature promotes tumor cell intravasation during metastasis (Figure 2). VEGF–VEGF receptor signaling loosens the tight junctions that interconnect adjacent endothelial cells which renders blood vessels permeable to leakage. Additionally, high interstitial fluid pressures coupled with the immature structure of tumor blood vessels enhances the ease by which tumor cells permeate through tumor blood vessels [126]. Several researchers have demonstrated that TECs release specific growth factors, called angiocrine factors [127] into the TEM which convert indolent tumor cells to more aggressive cells displaying greater tumorigenicity, extranodal invasion, and chemoresistance [128,129]. Cao et al. showed that fibroblast growth factor 4 (FGF4), produced by B-cell lymphoma cells, activates fibroblast growth factor receptor 1 (FGFR1) in neighboring endothelial cells resulting in the upregulation of the Notch ligand Jagged1. In turn, Jagged1 on endothelial cells reciprocally induces Notch2–Hey1 signaling in lymphoma cells [128]. Mao and colleagues demonstrated that exosome-like vesicles derived from esophageal squamous cell carcinoma (ESCC) cells are mediators of intracellular communication between epithelial cancer cells and vascular endothelial cells within the TME. Specifically, they showed that hypoxic ESCC-derived EVs resulted in increased proliferation, improved capillary-like structure formation, and increased invasive ability of human umbilical endothelial cells (HUVEC), concluding that hypoxic EVs derived from cancer cells alter TECs within the TME, enhancing tumor angiogenesis [130]. To date, many studies have demonstrated that the miRNA, mRNA, and other non-coding RNA cargos of EVs released from tumor cells are responsible for the metabolic reprogramming of stromal cells, including TECs. For example, Chen et al. showed that exosome-like vesicles isolated from the serum of colorectal cancer (CRC) with metastases contain circTUBCGP4 which leads to miR-146b-3p inhibition in HUVEC cells, leading to Akt signaling pathway activation, which results in enhanced cell migration and angiogenic tube formation [131]. Biagoni et al. showed that urokinase plasminogen activator surface receptor (uPAR) containing exosome-like vesicles released from melanoma cells led to an increase in pro-angiogenesis of both human microvascular endothelial cells (HMVECs) and endothelial colony-forming cells (ECFCs), which they demonstrated was as a result of tumor exosome-like vesicle-mediated induction of vascular endothelial cadherin (VE-Cadherin), uPAR, and EGFR protein expression in endothelial cells [132]. In colorectal cancer cell-derived exosome-like vesicles, miR-25-3p has been shown to promote angiogenesis and the disruption of vein endothelial cell tight junctions within distant sites, including the lung and liver, helping to establish the pre-metastatic niche [133].

2.1.4. Tumor-Infiltrating Lymphocytes (TILs)

Tumor-infiltrating lymphocytes (TILs) are defined as all lymphatic cell populations that invade into solid tumors. They consist primarily of cytotoxic CD8+ T cells and CD4+ helper T cells [134] in addition to smaller proportions of natural killer (NK) and B cells [135]. In all solid tumors TILs play distinct roles in modulating the TME, and for decades their role in tumor progression had been widely debated. In fact, the infiltration of immune cells into the TME and their role in cancer immunosurveillance is one of the hallmarks of cancer [136]. Although TILs have been shown to serve as somewhat of a double-edged sword as their infiltration into tumors can promote the initial establishment of a TME which is more susceptible to enhanced tumor progression [137], they can also attack tumor cells and in that way serve as potent tumor suppressors [138]. During the initial stages of tumor development, the infiltration of TILs and their prolonged interaction with surrounding tumor cells primes the hosts immune system against tumor cell elimination and in that way the tumor immunosurveillance results in the promotion of tumor growth [139]. However, in the long run the constant infiltration of TILs into the TME results in their exhaustion, characterized by sustained expression of inhibitory receptors distinct from functional effector and memory T cells, ultimately resulting in their failure to arrest tumor progression [138,140]. In fact, in a variety of tumor types (i.e., breast, colon, lung, and ovarian) the infiltration of immune cells into the TME has been shown to provide value as a predictive prognostic marker [141,142,143]. The treatment of metastatic cancer remains challenging, and in fact the study by Haj-Shomaly et al., 2022, suggests that paclitaxel chemotherapy, while effective in some cancer instances, may also promote tumor metastasis in the lung through its ability to rapidly induce ECM remodeling mediated by CD8+ T cells expressing lysyl oxidase (LOX), a potent ECM remodeling enzyme [144]. As such, in recent years tumor immunotherapy has become an attractive and effective treatment strategy for many solid tumor types. Several studies highlight the benefits of external expansion of TILs as an immunotherapeutic strategy, whereby TILs are harvested directly from tumor biopsies, expanded ex vivo, and then readministered to patients, referred to as adoptive cell therapy (ACT), with the main goal being to restore and enhance TIL anti-tumoral responses and the direct elimination of tumor cells [145,146,147],
Tumor-derived EVs are known to promote tumor progression through their direct modulation and suppression of the host immune response and chemoresistance and peripheral tolerance in cancer patients [148,149]. The cargo carried in tumor-derived exosome-like vesicles has been shown to include immunosuppressive molecules which influence the development, progression, and anti-tumor activity of immune cells either directly or indirectly [150]. Nakazawa et al., 2021, demonstrated that tumor-derived EVs containing CD300a are taken up by dendritic cells resulting in their inhibited secretion of interferon-β (IFN-β) leading to enhanced tumor immunity via the decreased activation of regulatory T cells [151]. Tumor-derived EV-dependent modulation of TIL activity occurs through the inhibition of proliferation and signaling activity of CD8+ T cells resulting in their apoptotic cell death [152]. Contrarily, dendritic cell-derived EVs promote the proliferation of T cells within the TME [153]. Cancer-derived CD8+ T cells from patients with head and neck cancer when co-cultured with tumor-derived EVs has been shown to induce the loss of CD27 expression in CD8+ T-cell and thus resulting in their change from the anti-tumor phenotype towards a more potent tumor suppressor phenotype [154,155,156]. Additionally, several studies have also suggested that tumor-derived EVs expressing the transmembrane protein FasL isolated from the plasma of oral cancer patients have the ability to induce apoptosis of CD8+ T cells [157,158]. Since regulatory T cells (Tregs) are critical for immune system suppression and the infiltration of Tregs into the TME, and their elevated presence in circulation is a strong prognostic marker in cancer [159], several studies have demonstrated that tumor-derived EVs promote the expansion of CD4/CD25/FOXP3 triple-positive Tregs and apoptotic induction of TILs [160]. Based on the immunoregulatory effects of EVs, which include the modulation of antigen presentation and immune activation and surveillance [161,162,163], multiple studies have investigated the ability of exosome-like vesicles to participate in tumor regression, demonstrating that immune cell-derived EVs display potent cytotoxic effects in hepatocellular carcinoma when administered as a cell-free anti-tumor vaccine [164,165].
Collectively, when considering the multifaceted and intricate role that EVs play in cell-to-cell communication between tumor cells, CAFs, TAMs, TECs, and TILs within the TME, it is no wonder that extensive research has been conducted to understand the mechanisms underlying EVs in tumor promotion and metastasis. Additionally, their enhanced secretion from tumors and the altered content of tumor-derived EVs when compared to EVs secreted from normal cells offer the potential to not only diagnose and monitor cancer progression, but also the generation of bioengineering EVs which interrupt the communication between tumor cells with the surrounding TME, thereby preventing EMT initiation, is an exciting and promising avenue in anti-metastatic therapy in cancer.

3. Extracellular Vesicle-Mediated Pre-Metastatic Niche Formation

The release of EVs from primary tumors selectively and favorably modifies the microenvironment of distant organs prior to the dissemination of metastatic cancer cells [166], termed pre-metastatic niche (PMN) formation. These changes to secondary organs, most commonly involving the lung followed by the liver, bone, and brain [167], have been shown to be caused by matrix metalloproteinase 9 (MMP-9), which is specifically induced in pre-metastatic lung endothelial cells and macrophages [168]. Hiratsuka et al. (2008) also demonstrated that in the lung, inflammatory PMN formation was promoted by the inflammatory mediator S100A8/A9 via a toll-like receptor (TLR)-dependent mechanism of induction of serum amyloid A (SAA) and the recruitment of Mac1+ myeloid cells [169]. Kaplan et al. (2005) demonstrated that the migration of VEGFR-1-expressing bone marrow-derived hematopoietic progenitor cells (BMDCs) into organ-specific pre-metastatic sites was induced by tumor-derived conditioned media and that BMDC cluster formation, which create a favorable microenvironment for incoming tumor cells, preceded tumor cell arrival [166]. Additionally, cell-free conditioned medium harvested from hypoxic breast cancer cells has been shown to drive the infiltration of CD11b+/Ly6Cmed/Ly6G+ myeloid-derived suppressor cells (MDSCs) and the reduction in cytotoxic natural killer (NK) cell populations, within the lung PMN of immune-competent mice [170]. In addition to the contribution of BMDCs to the generation of pre-metastatic niches, multiple studies have identified a large number of cytokines, chemokines, and growth factors which are connected to specific pre-metastatic niche-related processes [171,172,173]. In recent studies, EVs secreted from primary tumor cells have been described to have unique and central functions during PMN establishment and maintenance [174,175,176].
Successful dissemination of tumor cells from primary tumor sites and the progression of metastasis is highly dependent on the establishment of a favorable pre-metastatic niche microenvironment through both the modulation of vascular permeability and the stimulation of neo-angiogenesis within secondary organ sites (Figure 3). It is precisely this modulation of endothelial activity that serves as one critical element of the pro-metastatic role that tumor-derived EVs play in the successful invasion and colonization of tumor cells during metastasis. Recently, the miRNAs contained within tumor-derived EVs have been extensively correlated with vascular remodeling and neo-angiogenesis. For example, miR-105 carried within EVs derived from the metastatic MDA-MB-231 breast cancer cell line increases metastasis by facilitating endothelial cell barrier destruction via the downregulation of the tight junction zonula occludens 1 (ZO-1) protein [177] and Golgi integral membrane protein 4 (GOLIM4) [178]. Hannafon et al. also demonstrated that EVs derived from the breast cancer cell lines MDA-MB-231, MCF-7, and BT20 displayed significantly different miRNA profiles when these cells were treated with anti-angiogenic docosahexaenoic acid (DHA) compared to those isolated from untreated cells [179]. They showed that the expression levels of 83 miRNAs were altered in MCF-7 cell-derived EVs following DHA treatment and that the EVs they isolated from DHA-treated MDA-MB-231 cancer cells contained abundant miRNAs associated with anti-angiogenesis, including let-7a, miR-21, miR-23b, miR27a/b, and miR-320b, which when taken up by endothelial cells reduced their angiogenic potential [179]. Indeed, miR-105 and miR-939 carried within EVs derived from metastatic breast cancer cells have been shown to increase vascular permeability, accelerating metastasis by facilitating colonization in new tissues [177,180]. Kosaka et al. demonstrated that exosome-like vesicles derived from breast cancer cells have also been shown to contain miR-210 which when taken up by endothelial cells induced a pro-angiogenic response and caused increased TEC migration and capillary-like network formation [181]. The importance of vascular remodeling of pre-metastatic organ tissue sites is supported by observations that EVs purified from CD105+ renal cancer stem cell supernatants enhance lung metastasis in vivo via the up-regulation of VEGF and MMP-2 in lung endothelial cells [182]. Collectively, these studies demonstrate that tumor-derived EVs strongly modify endothelial cell function, which contributes to the remodeling of both the primary tumor microenvironment and the PMN to promote tumor progression and metastatic dissemination. However, it is noteworthy to understand that although vascular remodeling is considered an essential step during the metastatic cascade, alone is not enough to promote tumor colonization and progression within secondary sites.
Cellular survival can be seen as the sole driving force of cancer cells; it is therefore not surprising that during cancer development and progression clonal selection is facilitated by the acquisition of beneficial mutations that downregulate tumor suppressors and/or upregulate oncogenes [183]. Tumor cells have therefore developed mechanisms to increase their resistance to chemotherapeutic drugs, which in the case of doxorubicin can occur through the upregulation of ATP-binding cassette (ABC) efflux pumps, enhanced nuclear export, continued topoisomerase IIa activity, and/or suppression of downstream apoptosis signaling [184]. Since EVs play critical roles in facilitating intercellular communication, cancer cells have been shown to package chemotherapeutic drugs within their cargo, and as such they actively participate in the increased resistance of cancer cells to chemotherapeutic treatment [185,186,187,188]. Zheng et al. (2017) demonstrated that the exosome-like vesicles that were isolated from M2 macrophages had the ability to confer cisplatin resistance to gastric cancer cells through the transfer of miR-21, which resulted in phosphatase and tensin homolog (PTEN) downregulation and the inhibition of PI3K/Akt signaling [189]. Ji et al. (2015) showed that in gastric cancer mesenchymal stem cell (MSC)-derived EVs induced fluorouracil resistance through the activation of the CaMK/Raf/MEK/ERK signaling pathway [190]. Additionally, several studies have also demonstrated that, in addition to the role that tumor-derived EVs play in establishing a suitable PMN, they also have distinct local effects within the primary TME wherein they promote the transformation of MSCs into tumor-like cells [191,192]. Boelens et al. (2014) demonstrated that stromal cells transfer EV-derived 5′-triphosphate RNA to breast cancer cells resulting in increased radiation resistance through the activation of retinoic acid-inducible gene I (RIG-I)-dependent NOTCH3 signaling [193].
Overall, considering the role that EVs play in the establishment of the PMN coupled with the fact a hospitable environment specifically tailored to the needs of each tumor cell type is critical for successful metastatic dissemination, the prospect of tailoring/targeting EVs to inhibit and prevent PMN formation in addition to the prevention of EMT initiation as described previously holds significant potential as a future gold-standard anticancer therapeutic avenue.

4. Concluding Remarks

EV studies that have individually investigated the different types of cells constituting primary tumors, as well as their interactions, have highlighted how tumor-derived EVs, their cargos, and their surface signaling molecules can directly reprogram their TME as well as the microenvironment of PMN to enhance tumor cell survival, proliferation, and particularly metastatic, invasive, and colonizing abilities. Furthermore, it is well established that tumor-derived EVs are also capable of modulating the extracellular matrix (ECM) in order to remodel both primary tumor sites, where metastasis can be successfully initiated, and generate secondary tumor sites, where they educate normal cells and establish metastatic niches. It is the precise and timely orchestration of these intricate signaling interactions between tumor-derived EVs and PMN cells that create hospitable environments, known as cellular education, which direct colonization and systemic expansion of cancer cells. Tumor-derived EVs have gained research interest in the quest to decipher the metastatic process, but they are very complex and independent cellular entities with unique multi-omic compound ratios, which uniquely orchestrate the cellular programs of tumor cells that maintain survival and direct expansion. Additionally, within the EV community it is widely accepted that tumor cells acquire an increase in output of EVs when compared to normal cells, and it is believed that this increase in tumor-derived EVs within the circulation is essential for successful PMN formation, metastatic dissemination, and secondary tumor expansion. This enhanced biological process has potential to provide detectable EV-based biomarkers for early diagnosis but also suggests that targeted therapeutic strategies aimed at reducing EV concentrations in the circulation may help control (reduce or redirect) metastatic processes in the human body. One may imagine the development of novel devices, which may help not only filter out metastatic EVs from the circulation by targeted selection, but also allow the circulation of EVs that may help reduce primary and secondary tumor growth in cancer patients.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare that there are no conflict of interest regarding the publication of this paper.

References

  1. Hanahan, D.; Coussens, L.M. Accessories to the crime: Functions of cells recruited to the tumor microenvironment. Cancer Cell 2012, 21, 309–322. [Google Scholar] [CrossRef]
  2. Kidd, S.; Spaeth, E.; Watson, K.; Burks, J.; Lu, H.; Klopp, A.; Andreeff, M.; Marini, F.C. Origins of the Tumor Microenvironment: Quantitative Assessment of Adipose-Derived and Bone Marrow–Derived Stroma. PLoS ONE 2012, 7, e30563. [Google Scholar] [CrossRef]
  3. Xiong, Y.; McDonald, L.T.; Russell, D.L.; Kelly, R.R.; Wilson, K.R.; Mehrotra, M.; Soloff, A.C.; LaRue, A.C. Hematopoietic stem cell-derived adipocytes and fibroblasts in the tumor microenvironment. World J. Stem Cells 2015, 7, 253–265. [Google Scholar] [CrossRef]
  4. Sakurai, M.; Miki, Y.; Takagi, K.; Suzuki, T.; Ishida, T.; Ohuchi, N.; Sasano, H. Interaction with adipocyte stromal cells induces breast cancer malignancy via S100A7 upregulation in breast cancer microenvironment. Breast Cancer Res. 2017, 19, 70. [Google Scholar] [CrossRef]
  5. Hastings, J.F.; Skhinas, J.N.; Fey, D.; Croucher, D.R.; Cox, T.R. The extracellular matrix as a key regulator of intracellular signalling networks. Br. J. Pharmacol. 2018, 176, 82–92. [Google Scholar] [CrossRef]
  6. Mitchell, M.I.; Engelbrecht, A.-M. Metabolic hijacking: A survival strategy cancer cells exploit? Crit. Rev. Oncol. Hematol. 2017, 109, 1–8. [Google Scholar] [CrossRef]
  7. Xu, X.D.; Shao, S.X.; Jiang, H.P.; Cao, Y.W.; Wang, Y.H.; Yang, X.C.; Wang, Y.L.; Wang, X.S.; Niu, H.T. Warburg Effect or Reverse Warburg Effect? A Review of Cancer Metabolism. Oncol. Res. Treat. 2015, 38, 117–122. [Google Scholar] [CrossRef]
  8. Fukushi, A.; Kim, H.-D.; Chang, Y.-C.; Kim, C.-H. Revisited Metabolic Control and Reprogramming Cancers by Means of the Warburg Effect in Tumor Cells. Int. J. Mol. Sci. 2022, 23, 10037. [Google Scholar] [CrossRef]
  9. Warburg, O.; Wind, F.; Negelein, E. The metabolism of tumors in the body. J. Gen. Physiol. 1927, 8, 519–530. [Google Scholar] [CrossRef]
  10. Lugano, R.; Ramachandran, M.; Dimberg, A. Tumor angiogenesis: Causes, consequences, challenges and opportunities. Cell. Mol. Life Sci. 2020, 77, 1745–1770. [Google Scholar] [CrossRef]
  11. Karnoub, A.E.; Dash, A.B.; Vo, A.P.; Sullivan, A.; Brooks, M.W.; Bell, G.W.; Richardson, A.L.; Polyak, K.; Tubo, R.; Weinberg, R.A. Mesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature 2007, 449, 557–563. [Google Scholar] [CrossRef]
  12. Polyak, K.; Weinberg, R.A. Transitions between epithelial and mesenchymal states: Acquisition of malignant and stem cell traits. Nat. Rev. Cancer 2009, 9, 265–273. [Google Scholar] [CrossRef] [PubMed]
  13. Tuxhorn, J.A.; Ayala, G.E.; Smith, M.J.; Smith, V.C.; Dang, T.D.; Rowley, D.R. Reactive stroma in human prostate cancer: Induction of myofibroblast phenotype and extracellular matrix remodeling. Clin. Cancer Res. 2002, 8, 2912–2923. [Google Scholar] [PubMed]
  14. Larsen, M.; Artym, V.V.; Green, J.A.; Yamada, K.M. The matrix reorganized: Extracellular matrix remodeling and integrin signaling. Curr. Opin. Cell Biol. 2006, 18, 463–471. [Google Scholar] [CrossRef]
  15. Mantovani, A.; Romero, P.; Palucka, A.K.; Marincola, F.M. Tumour immunity: Effector response to tumour and role of the microenvironment. Lancet 2008, 371, 771–783. [Google Scholar] [CrossRef] [PubMed]
  16. Wei, Y.; Au, J.L.-S. Role of Tumour Microenvironment in Chemoresistance. In Integration/Interaction of Oncologic Growth. Cancer Growth and Progression; Meadows, G.G., Ed.; Springer: Dordrecht, The Netherlands, 2005; Volume 15. [Google Scholar] [CrossRef]
  17. Straussman, R.; Morikawa, T.; Shee, K.; Barzily-Rokni, M.; Qian, Z.R.; Du, J.; Davis, A.; Mongare, M.M.; Gould, J.; Frederick, D.T.; et al. Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion. Nature 2012, 487, 500–504. [Google Scholar] [CrossRef] [PubMed]
  18. Nakasone, E.S.; Askautrud, H.A.; Kees, T.; Park, J.-H.; Plaks, V.; Ewald, A.J.; Fein, M.; Rasch, M.G.; Tan, Y.-X.; Qiu, J.; et al. Imaging Tumor-Stroma Interactions during Chemotherapy Reveals Contributions of the Microenvironment to Resistance. Cancer Cell 2012, 21, 488–503. [Google Scholar] [CrossRef] [PubMed]
  19. Cruceriu, D.; Baldasici, O.; Balacescu, O.; Berindan-Neagoe, I. The dual role of tumor necrosis factor-alpha (TNF-α) in breast cancer: Molecular insights and therapeutic approaches. Cell. Oncol. 2020, 43, 1–18. [Google Scholar] [CrossRef] [PubMed]
  20. Zeng, J.; Chen, S.; Li, C.; Ye, Z.; Lin, B.; Liang, Y.; Wang, B.; Ma, Y.; Chai, X.; Zhang, X.; et al. Mesenchymal stem/stromal cells-derived IL-6 promotes nasopharyngeal carcinoma growth and resistance to cisplatin via upregulating CD73 expression. J. Cancer 2020, 11, 2068–2079. [Google Scholar] [CrossRef] [PubMed]
  21. Angioni, R.; Sánchez-Rodríguez, R.; Viola, A.; Molon, B. TGF-β in Cancer: Metabolic Driver of the Tolerogenic Crosstalk in the Tumor Microenvironment. Cancers 2021, 13, 401. [Google Scholar] [CrossRef]
  22. Huang, W.-H.; Liu, C.-D.; Chang, C.-C. The perspectives of interleukin-10 in the pathogenesis and therapeutics of multiple myeloma. Tzu Chi Med. J. 2021, 33, 257–262. [Google Scholar] [CrossRef] [PubMed]
  23. Kok, V.C.; Yu, C.-C. Cancer-Derived Exosomes: Their Role in Cancer Biology and Biomarker Development. Int. J. Nanomed. 2020, 15, 8019–8036. [Google Scholar] [CrossRef] [PubMed]
  24. da Costa, V.R.; Araldi, R.P.; Vigerelli, H.; D’ámelio, F.; Mendes, T.B.; Gonzaga, V.; Policíquio, B.; Colozza-Gama, G.A.; Valverde, C.W.; Kerkis, I. Exosomes in the Tumor Microenvironment: From Biology to Clinical Applications. Cells 2021, 10, 2617. [Google Scholar] [CrossRef] [PubMed]
  25. Liu, J.; Ren, L.; Li, S.; Li, W.; Zheng, X.; Yang, Y.; Fu, W.; Yi, J.; Wang, J.; Du, G. The biology, function, and applications of exosomes in cancer. Acta Pharm. Sin. B 2021, 11, 2783–2797. [Google Scholar] [CrossRef]
  26. Antonyak, M.A.; Li, B.; Boroughs, L.K.; Johnson, J.L.; Druso, J.E.; Bryant, K.L.; Holowka, D.A.; Cerione, R.A. Cancer cell-derived microvesicles induce transformation by transferring tissue transglutaminase and fibronectin to recipient cells. Proc. Natl. Acad. Sci. USA 2011, 108, 4852–4857, Erratum in Proc. Natl. Acad. Sci. USA 2011, 108, 17569. [Google Scholar] [CrossRef]
  27. Li, I.; Nabet, B.Y. Exosomes in the tumor microenvironment as mediators of cancer therapy resistance. Mol. Cancer 2019, 18, 32. [Google Scholar] [CrossRef]
  28. Zhang, H.; Deng, T.; Liu, R.; Ning, T.; Yang, H.; Liu, D.; Zhang, Q.; Lin, D.; Ge, S.; Bai, M.; et al. CAF secreted miR-522 suppresses ferroptosis and promotes acquired chemo-resistance in gastric cancer. Mol. Cancer 2020, 19, 43. [Google Scholar] [CrossRef]
  29. Sedgwick, A.E.; D'Souza-Schorey, C. The biology of extracellular microvesicles. Traffic 2018, 19, 319–327. [Google Scholar] [CrossRef]
  30. Battistelli, M.; Falcieri, E. Apoptotic Bodies: Particular Extracellular Vesicles Involved in Intercellular Communication. Biology 2020, 9, 21. [Google Scholar] [CrossRef]
  31. Tricarico, C.; Clancy, J.; D'Souza-Schorey, C. Biology and biogenesis of shed microvesicles. Small GTPases 2016, 8, 220–232. [Google Scholar] [CrossRef]
  32. Mitchell, M.I.; Ma, J.; Carter, C.L.; Loudig, O. Circulating Exosome Cargoes Contain Functionally Diverse Cancer Biomarkers: From Biogenesis and Function to Purification and Potential Translational Utility. Cancers 2022, 14, 3350. [Google Scholar] [CrossRef]
  33. Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef] [PubMed]
  34. Christianson, H.C.; Svensson, K.J.; van Kuppevelt, T.H.; Li, J.-P.; Belting, M. Cancer cell exosomes depend on cell-surface heparan sulfate proteoglycans for their internalization and functional activity. Proc. Natl. Acad. Sci. USA 2013, 110, 17380–17385. [Google Scholar] [CrossRef] [PubMed]
  35. Zhao, B.; Zhang, Y.; Han, S.; Zhang, W.; Zhou, Q.; Guan, H.; Liu, J.; Shi, J.; Su, L.; Hu, D. Exosomes derived from human amniotic epithelial cells accelerate wound healing and inhibit scar formation. Histochem. J. 2017, 48, 121–132. [Google Scholar] [CrossRef] [PubMed]
  36. Miki, Y.; Yashiro, M.; Okuno, T.; Kitayama, K.; Masuda, G.; Hirakawa, K.; Ohira, M. CD9-positive exosomes from cancer-associated fibroblasts stimulate the migration ability of scirrhous-type gastric cancer cells. Br. J. Cancer 2018, 118, 867–877. [Google Scholar] [CrossRef] [PubMed]
  37. Logozzi, M.; Angelini, D.F.; Giuliani, A.; Mizzoni, D.; Di Raimo, R.; Maggi, M.; Gentilucci, A.; Marzio, V.; Salciccia, S.; Borsellino, G.; et al. Increased Plasmatic Levels of PSA-Expressing Exosomes Distinguish Prostate Cancer Patients from Benign Prostatic Hyperplasia: A Prospective Study. Cancers 2019, 11, 1449. [Google Scholar] [CrossRef]
  38. Newman, L.A.; Useckaite, Z.; Johnson, J.; Sorich, M.J.; Hopkins, A.M.; Rowland, A. Selective Isolation of Liver-Derived Extracellular Vesicles Redefines Performance of miRNA Biomarkers for Non-Alcoholic Fatty Liver Disease. Biomedicines 2022, 10, 195. [Google Scholar] [CrossRef]
  39. Chiasserini, D.; van Weering, J.R.; Piersma, S.R.; Pham, T.V.; Malekzadeh, A.; Teunissen, C.E.; de Wit, H.; Jiménez, C.R. Proteomic analysis of cerebrospinal fluid extracellular vesicles: A comprehensive dataset. J. Proteom. 2014, 106, 191–204. [Google Scholar] [CrossRef]
  40. Choudhary, I.; Vo, T.; Paudel, K.; Wen, X.; Gupta, R.; Kesimer, M.; Patial, S.; Saini, Y. Vesicular and extravesicular protein analyses from the airspaces of ozone-exposed mice revealed signatures associated with mucoinflammatory lung disease. Sci. Rep. 2021, 11, 23203. [Google Scholar] [CrossRef]
  41. Mimmi, S.; Zimbo, A.M.; Rotundo, S.; Cione, E.; Nisticò, N.; Aloisio, A.; Maisano, D.; Tolomeo, A.M.; Dattilo, V.; Lionello, R.; et al. SARS-CoV-2 spike protein-guided exosome isolation facilitates detection of potential miRNA biomarkers in COVID-19 infections. Clin. Chem. Lab. Med. 2023, 61, 1518–1524. [Google Scholar] [CrossRef]
  42. Miranda, J.; Paules, C.; Nair, S.; Lai, A.; Palma, C.; Scholz-Romero, K.; Rice, G.E.; Gratacos, E.; Crispi, F.; Salomon, C. Placental exosomes profile in maternal and fetal circulation in intrauterine growth restriction—Liquid biopsies to monitoring fetal growth. Placenta 2018, 64, 34–43. [Google Scholar] [CrossRef] [PubMed]
  43. Dozio, V.; Sanchez, J.-C. Characterisation of extracellular vesicle-subsets derived from brain endothelial cells and analysis of their protein cargo modulation after TNF exposure. J. Extracell. Vesicles 2017, 6, 1302705. [Google Scholar] [CrossRef]
  44. Jahangiri, B.; Saei, A.K.; Obi, P.O.; Asghari, N.; Lorzadeh, S.; Hekmatirad, S.; Rahmati, M.; Velayatipour, F.; Asghari, M.H.; Saleem, A.; et al. Exosomes, autophagy and ER stress pathways in human diseases: Cross-regulation and therapeutic approaches. Biochim. Biophys. Acta (BBA) Mol. Basis Dis. 2022, 1868, 166484. [Google Scholar] [CrossRef] [PubMed]
  45. Kalluri, R.; LeBleu, V.S. The biology, function, and biomedical applications of exosomes. Science 2020, 367, eaau6977. [Google Scholar] [CrossRef] [PubMed]
  46. Willms, E.; Johansson, H.J.; Mäger, I.; Lee, Y.; Blomberg, K.E.M.; Sadik, M.; Alaarg, A.; Smith, C.E.; Lehtiö, J.; EL Andaloussi, S.; et al. Cells release subpopulations of exosomes with distinct molecular and biological properties. Sci. Rep. 2016, 6, 22519. [Google Scholar] [CrossRef]
  47. Théry, C.; Boussac, M.; Véron, P.; Ricciardi-Castagnoli, P.; Raposo, G.; Garin, J.; Amigorena, S. Proteomic Analysis of Dendritic Cell-Derived Exosomes: A Secreted Subcellular Compartment Distinct from Apoptotic Vesicles. Perspect. Surg. 2001, 166, 7309–7318. [Google Scholar] [CrossRef]
  48. Valadi, H.; Ekström, K.; Bossios, A.; Sjöstrand, M.; Lee, J.J.; Lötvall, J.O. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 2007, 9, 654–659. [Google Scholar] [CrossRef]
  49. Hoen, E.N.; Cremer, T.; Gallo, R.C.; Margolis, L.B. Extracellular vesicles and viruses: Are they close relatives? Proc. Natl. Acad. Sci. USA 2016, 113, 9155–9161. [Google Scholar] [CrossRef]
  50. Hoshino, D.; Kirkbride, K.C.; Costello, K.; Clark, E.S.; Sinha, S.; Grega-Larson, N.; Tyska, M.J.; Weaver, A.M. Exosome Secretion Is Enhanced by Invadopodia and Drives Invasive Behavior. Cell Rep. 2013, 5, 1159–1168. [Google Scholar] [CrossRef]
  51. Shrivastava, S.; Morris, K.V. The Multifunctionality of Exosomes; from the Garbage Bin of the Cell to a Next Generation Gene and Cellular Therapy. Genes 2021, 12, 173. [Google Scholar] [CrossRef]
  52. Ludwig, N.; Yerneni, S.S.; Razzo, B.M.; Whiteside, T.L. Exosomes from HNSCC Promote Angiogenesis through Reprogramming of Endothelial Cells. Mol. Cancer Res. 2018, 16, 1798–1808. [Google Scholar] [CrossRef] [PubMed]
  53. Chowdhury, R.; Webber, J.P.; Gurney, M.; Mason, M.D.; Tabi, Z.; Clayton, A. Cancer exosomes trigger mesenchymal stem cell differentiation into pro-angiogenic and pro-invasive myofibroblasts. Oncotarget 2014, 6, 715–731. [Google Scholar] [CrossRef] [PubMed]
  54. 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]
  55. Peinado, H.; Alečković, M.; Lavotshkin, S.; Matei, I.; Costa-Silva, B.; Moreno-Bueno, G.; Hergueta-Redondo, M.; Williams, C.; García-Santos, G.; Ghajar, C.M.; et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat. Med. 2012, 18, 883–891. [Google Scholar] [CrossRef]
  56. Hoshino, A.; Costa-Silva, B.; Shen, T.-L.; Rodrigues, G.; Hashimoto, A.; Mark, M.T.; Molina, H.; Kohsaka, S.; Di Giannatale, A.; Ceder, S.; et al. Tumour exosome integrins determine organotropic metastasis. Nature 2015, 527, 329–335. [Google Scholar] [CrossRef] [PubMed]
  57. Taylor, S.; Spugnini, E.P.; Assaraf, Y.G.; Azzarito, T.; Rauch, C.; Fais, S. Microenvironment acidity as a major determinant of tumor chemoresistance: Proton pump inhibitors (PPIs) as a novel therapeutic approach. Drug Resist. Updat. 2015, 23, 69–78. [Google Scholar] [CrossRef] [PubMed]
  58. Senthebane, D.A.; Rowe, A.; Thomford, N.E.; Shipanga, H.; Munro, D.; Al Mazeedi, M.A.M.; Almazyadi, H.A.M.; Kallmeyer, K.; Dandara, C.; Pepper, M.S.; et al. The Role of Tumor Microenvironment in Chemoresistance: To Survive, Keep Your Enemies Closer. Int. J. Mol. Sci. 2017, 18, 1586. [Google Scholar] [CrossRef]
  59. Federici, C.; Petrucci, F.; Caimi, S.; Cesolini, A.; Logozzi, M.; Borghi, M.; D’Ilio, S.; Lugini, L.; Violante, N.; Azzarito, T.; et al. Exosome release and low pH belong to a framework of resistance of human melanoma cells to cisplatin. PLoS ONE 2014, 9, e88193. [Google Scholar] [CrossRef] [PubMed]
  60. Wang, L.; Fan, Z.; Zhang, J.; Changyi, Y.; Huang, C.; Gu, Y.; Xu, Z.; Tang, Z.; Lu, W.; Wei, X.; et al. Evaluating tumor metastatic potential by imaging intratumoral acidosis via pH-activatable near-infrared fluorescent probe. Int. J. Cancer 2015, 136, E107–E116. [Google Scholar] [CrossRef]
  61. Tan, Y.; Luo, X.; Lv, W.; Hu, W.; Zhao, C.; Xiong, M.; Yi, Y.; Wang, D.; Wang, Y.; Wang, H.; et al. Tumor-derived exosomal components: The multifaceted roles and mechanisms in breast cancer metastasis. Cell Death Dis. 2021, 12, 547. [Google Scholar] [CrossRef]
  62. 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]
  63. Logozzi, M.; Mizzoni, D.; Angelini, D.F.; Di Raimo, R.; Falchi, M.; Battistini, L.; Fais, S. Microenvironmental pH and Exosome Levels Interplay in Human Cancer Cell Lines of Different Histotypes. Cancers 2018, 10, 370. [Google Scholar] [CrossRef] [PubMed]
  64. Maia, J.; Caja, S.; Moraes, M.C.S.; Couto, N.; Costa-Silva, B. Exosome-Based Cell-Cell Communication in the Tumor Microenvironment. Front. Cell Dev. Biol. 2018, 6, 18. [Google Scholar] [CrossRef] [PubMed]
  65. Yin, K.; Wang, S.; Zhao, R.C. Exosomes from mesenchymal stem/stromal cells: A new therapeutic paradigm. Biomark. Res. 2019, 7, 8. [Google Scholar] [CrossRef]
  66. 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] [PubMed]
  67. Wang, L.; Yang, G.; Zhao, D.; Wang, J.; Bai, Y.; Peng, Q.; Wang, H.; Fang, R.; Chen, G.; Wang, Z.; et al. CD103-positive CSC exosome promotes EMT of clear cell renal cell carcinoma: Role of remote MiR-19b-3p. Mol. Cancer 2019, 18, 86, Erratum in Mol. Cancer 2020, 19, 144. [Google Scholar] [CrossRef]
  68. Liu, Y.; Gu, Y.; Han, Y.; Zhang, Q.; Jiang, Z.; Zhang, X.; Huang, B.; Xu, X.; Zheng, J.; Cao, X. Tumor Exosomal RNAs Promote Lung Pre-metastatic Niche Formation by Activating Alveolar Epithelial TLR3 to Recruit Neutrophils. Cancer Cell 2016, 30, 243–256. [Google Scholar] [CrossRef]
  69. LeBleu, V.S.; Kalluri, R. A peek into cancer-associated fibroblasts: Origins, functions and translational impact. Dis. Model. Mech. 2018, 11, dmm029447. [Google Scholar] [CrossRef]
  70. Manoukian, P.; Bijlsma, M.; van Laarhoven, H. The Cellular Origins of Cancer-Associated Fibroblasts and Their Opposing Contributions to Pancreatic Cancer Growth. Front. Cell Dev. Biol. 2021, 9, 743907. [Google Scholar] [CrossRef]
  71. Wright, K.; Ly, T.; Kriet, M.; Czirok, A.; Thomas, S.M. Cancer-Associated Fibroblasts: Master Tumor Microenvironment Modifiers. Cancers 2023, 15, 1899. [Google Scholar] [CrossRef]
  72. Sahai, E.; Astsaturov, I.; Cukierman, E.; DeNardo, D.G.; Egeblad, M.; Evans, R.M.; Fearon, D.; Greten, F.R.; Hingorani, S.R.; Hunter, T.; et al. A framework for advancing our understanding of cancer-associated fibroblasts. Nat. Rev. Cancer 2020, 20, 174–186. [Google Scholar] [CrossRef] [PubMed]
  73. Dvorak, H.F. Tumors: Wounds That Do Not Heal—Redux. Cancer Immunol. Res. 2015, 3, 1–11. [Google Scholar] [CrossRef] [PubMed]
  74. Hua, Y.; Bergers, G. Tumors vs. Chronic Wounds: An Immune Cell's Perspective. Front. Immunol. 2019, 10, 2178. [Google Scholar] [CrossRef] [PubMed]
  75. Deyell, M.; Garris, C.S.; Laughney, A.M. Cancer metastasis as a non-healing wound. Br. J. Cancer 2021, 124, 1491–1502. [Google Scholar] [CrossRef]
  76. Öhlund, D.; Elyada, E.; Tuveson, D. Fibroblast heterogeneity in the cancer wound. J. Exp. Med. 2014, 211, 1503–1523. [Google Scholar] [CrossRef]
  77. Ziani, L.; Chouaib, S.; Thiery, J. Alteration of the Antitumor Immune Response by Cancer-Associated Fibroblasts. Front. Immunol. 2018, 9, 414. [Google Scholar] [CrossRef]
  78. Li, C.; Teixeira, A.F.; Zhu, H.-J.; Dijke, P.T. Cancer associated-fibroblast-derived exosomes in cancer progression. Mol. Cancer 2021, 20, 154. [Google Scholar] [CrossRef]
  79. Peng, Z.; Tong, Z.; Ren, Z.; Ye, M.; Hu, K. Cancer-associated fibroblasts and its derived exosomes: A new perspective for reshaping the tumor microenvironment. Mol. Med. 2023, 29, 66. [Google Scholar] [CrossRef]
  80. Gu, J.; Qian, H.; Shen, L.; Zhang, X.; Zhu, W.; Huang, L.; Yan, Y.; Mao, F.; Zhao, C.; Shi, Y.; et al. Gastric Cancer Exosomes Trigger Differentiation of Umbilical Cord Derived Mesenchymal Stem Cells to Carcinoma-Associated Fibroblasts through TGF-β/Smad Pathway. PLoS ONE 2012, 7, e52465. [Google Scholar] [CrossRef]
  81. Webber, J.; Steadman, R.; Mason, M.D.; Tabi, Z.; Clayton, A. Cancer Exosomes Trigger Fibroblast to Myofibroblast Differentiation. Cancer Res. 2010, 70, 9621–9630. [Google Scholar] [CrossRef]
  82. Cho, J.A.; Park, H.; Lim, E.H.; Kim, K.H.; Choi, J.S.; Lee, J.H.; Shin, J.W.; Lee, K.W. Exosomes from ovarian cancer cells induce adipose tissue-derived mesenchymal stem cells to acquire the physical and functional characteristics of tumor-supporting myofibroblasts. Gynecol. Oncol. 2011, 123, 379–386. [Google Scholar] [CrossRef] [PubMed]
  83. Cho, J.A.; Park, H.; Lim, E.H.; Lee, K.W. Exosomes from breast cancer cells can convert adipose tissue-derived mesenchymal stem cells into myofibroblast-like cells. Int. J. Oncol. 2011, 40, 130–138. [Google Scholar] [CrossRef]
  84. Catteau, X.; Simon, P.; Noёl, J.-C. Stromal expression of matrix metalloproteinase 2 in cancer-associated fibrobasts is strongly related to human epidermal growth factor receptor 2 status in invasive breast carcinoma. Mol. Clin. Oncol. 2016, 4, 375–378. [Google Scholar] [CrossRef] [PubMed]
  85. Luga, V.; Wrana, J.L. Tumor–Stroma Interaction: Revealing Fibroblast-Secreted Exosomes as Potent Regulators of Wnt-Planar Cell Polarity Signaling in Cancer Metastasis. Cancer Res. 2013, 73, 6843–6847. [Google Scholar] [CrossRef]
  86. Otranto, M.; Sarrazy, V.; Bonté, F.; Hinz, B.; Gabbiani, G.; Desmouliere, A. The role of the myofibroblast in tumor stroma remodeling. Cell Adhes. Migr. 2012, 6, 203–219. [Google Scholar] [CrossRef] [PubMed]
  87. Luga, V.; Zhang, L.; Viloria-Petit, A.M.; Ogunjimi, A.A.; Inanlou, M.R.; Chiu, E.; Buchanan, M.; Hosein, A.N.; Basik, M.; Wrana, J.L. Exosomes Mediate Stromal Mobilization of Autocrine Wnt-PCP Signaling in Breast Cancer Cell Migration. Cell 2012, 151, 1542–1556. [Google Scholar] [CrossRef] [PubMed]
  88. Sidhu, S.S.; Mengistab, A.T.; Tauscher, A.N.; LaVail, J.; Basbaum, C. The microvesicle as a vehicle for EMMPRIN in tumor–stromal interactions. Oncogene 2004, 23, 956–963. [Google Scholar] [CrossRef]
  89. Ramteke, A.; Ting, H.; Agarwal, C.; Mateen, S.; Somasagara, R.; Hussain, A.; Graner, M.; Frederick, B.; Agarwal, R.; Deep, G. Exosomes secreted under hypoxia enhance invasiveness and stemness of prostate cancer cells by targeting adherens junction molecules. Mol. Carcinog. 2013, 54, 554–565. [Google Scholar] [CrossRef]
  90. Krzyszczyk, P.; Schloss, R.; Palmer, A.; Berthiaume, F. The Role of Macrophages in Acute and Chronic Wound Healing and Interventions to Promote Pro-wound Healing Phenotypes. Front. Physiol. 2018, 9, 419. [Google Scholar] [CrossRef]
  91. Logozzi, M.; Mizzoni, D.; Capasso, C.; Del Prete, S.; Di Raimo, R.; Falchi, M.; Angelini, D.F.; Sciarra, A.; Maggi, M.; Supuran, C.T.; et al. Plasmatic exosomes from prostate cancer patients show increased carbonic anhydrase IX expression and activity and low pH. J. Enzym. Inhib. Med. Chem. 2019, 35, 280–288. [Google Scholar] [CrossRef]
  92. Henze, A.-T.; Mazzone, M. The impact of hypoxia on tumor-associated macrophages. J. Clin. Investig. 2016, 126, 3672–3679. [Google Scholar] [CrossRef] [PubMed]
  93. Pollard, J.W. Macrophages define the invasive microenvironment in breast cancer. J. Leukoc. Biol. 2008, 84, 623–630. [Google Scholar] [CrossRef] [PubMed]
  94. Grivennikov, S.I.; Wang, K.; Mucida, D.; Stewart, C.A.; Schnabl, B.; Jauch, D.; Taniguchi, K.; Yu, G.Y.; Osterreicher, C.H.; Hung, K.E.; et al. Adenoma-linked barrier defects and microbial products drive IL-23/IL-17-mediated tumour growth. Nature 2012, 491, 254–258. [Google Scholar] [CrossRef] [PubMed]
  95. Kong, L.; Zhou, Y.; Bu, H.; Lv, T.; Shi, Y.; Yang, J. Deletion of interleukin-6 in monocytes/macrophages suppresses the initiation of hepatocellular carcinoma in mice. J. Exp. Clin. Cancer Res. 2016, 35, 131. [Google Scholar] [CrossRef] [PubMed]
  96. Biswas, S.K.; Mantovani, A. Macrophage plasticity and interaction with lymphocyte subsets: Cancer as a paradigm. Nat. Immunol. 2010, 11, 889–896. [Google Scholar] [CrossRef]
  97. Orecchioni, M.; Ghosheh, Y.; Pramod, A.B.; Ley, K. Macrophage Polarization: Different Gene Signatures in M1(LPS+) vs. Classically and M2(LPS–) vs. Alternatively Activated Macrophages. Front. Immunol. 2019, 10, 1084, Erratum in Front. Immunol. 2020, 11, 234. [Google Scholar] [CrossRef]
  98. Mould, K.J.; Jackson, N.D.; Henson, P.M.; Seibold, M.; Janssen, W.J. Single cell RNA sequencing identifies unique inflammatory airspace macrophage subsets. JCI Insight 2019, 4, e126556. [Google Scholar] [CrossRef]
  99. Qian, B.-Z.; Pollard, J.W. Macrophage Diversity Enhances Tumor Progression and Metastasis. Cell 2010, 141, 39–51. [Google Scholar] [CrossRef]
  100. Chen, Q.; Li, Y.; Gao, W.; Chen, L.; Xu, W.; Zhu, X. Exosome-Mediated Crosstalk Between Tumor and Tumor-Associated Macrophages. Front. Mol. Biosci. 2021, 8, 764222. [Google Scholar] [CrossRef]
  101. Seo, N.; Shirakura, Y.; Tahara, Y.; Momose, F.; Harada, N.; Ikeda, H.; Akiyoshi, K.; Shiku, H. Activated CD8+ T cell extracellular vesicles prevent tumour progression by targeting of lesional mesenchymal cells. Nat. Commun. 2018, 9, 435. [Google Scholar] [CrossRef]
  102. Baig, M.S.; Roy, A.; Rajpoot, S.; Liu, D.; Savai, R.; Banerjee, S.; Kawada, M.; Faisal, S.M.; Saluja, R.; Saqib, U.; et al. Tumor-derived exosomes in the regulation of macrophage polarization. Inflamm. Res. 2020, 69, 435–451. [Google Scholar] [CrossRef]
  103. Marleau, A.M.; Chen, C.-S.; Joyce, J.A.; Tullis, R.H. Exosome removal as a therapeutic adjuvant in cancer. J. Transl. Med. 2012, 10, 134. [Google Scholar] [CrossRef]
  104. LeBleu, V.S.; Kalluri, R. Exosomes Exercise Inhibition of Anti-Tumor Immunity during Chemotherapy. Immunity 2019, 50, 547–549. [Google Scholar] [CrossRef]
  105. Yang, Q.; Xu, J.; Gu, J.; Shi, H.; Zhang, J.; Zhang, J.; Chen, Z.; Fang, X.; Zhu, T.; Zhang, X. Extracellular Vesicles in Cancer Drug Resistance: Roles, Mechanisms, and Implications. Adv. Sci. 2022, 9, e2201609. [Google Scholar] [CrossRef]
  106. Zhou, X.; Liu, Q.; Wang, X.; Yao, X.; Zhang, B.; Wu, J.; Sun, C. Exosomal ncRNAs facilitate interactive ‘dialogue’ between tumor cells and tumor-associated macrophages. Cancer Lett. 2022, 552, 215975. [Google Scholar] [CrossRef]
  107. Zhou, J.; Li, X.; Wu, X.; Zhang, T.; Zhu, Q.; Wang, X.; Wang, H.; Wang, K.; Lin, Y.; Wang, X. Exosomes Released from Tumor-Associated Macrophages Transfer miRNAs That Induce a Treg/Th17 Cell Imbalance in Epithelial Ovarian Cancer. Cancer Immunol. Res. 2018, 6, 1578–1592. [Google Scholar] [CrossRef]
  108. Zhao, S.; Mi, Y.; Guan, B.; Zheng, B.; Wei, P.; Gu, Y.; Zhang, Z.; Cai, S.; Xu, Y.; Li, X.; et al. Tumor-derived exosomal miR-934 induces macrophage M2 polarization to promote liver metastasis of colorectal cancer. J. Hematol. Oncol. 2020, 13, 156, Erratum in J. Hematol. Oncol. 2021, 14, 33. [Google Scholar] [CrossRef]
  109. Takano, Y.; Masuda, T.; Iinuma, H.; Yamaguchi, R.; Sato, K.; Tobo, T.; Hirata, H.; Kuroda, Y.; Nambara, S.; Hayashi, N.; et al. Circulating exosomal microRNA-203 is associated with metastasis possibly via inducing tumor-associated macrophages in colorectal cancer. Oncotarget 2017, 8, 78598–78613. [Google Scholar] [CrossRef]
  110. Shinohara, H.; Kuranaga, Y.; Kumazaki, M.; Sugito, N.; Yoshikawa, Y.; Takai, T.; Taniguchi, K.; Ito, Y.; Akao, Y. Regulated Polarization of Tumor-Associated Macrophages by miR-145 via Colorectal Cancer–Derived Extracellular Vesicles. J. Immunol. 2017, 199, 1505–1515. [Google Scholar] [CrossRef]
  111. Cai, J.; Qiao, B.; Gao, N.; Lin, N.; He, W. Oral squamous cell carcinoma-derived exosomes promote M2 subtype macrophage polarization mediated by exosome-enclosed miR-29a-3p. Am. J. Physiol. Cell. Physiol. 2019, 316, C731–C740. [Google Scholar] [CrossRef]
  112. Chen, X.; Ying, X.; Wang, X.; Wu, X.; Zhu, Q.; Wang, X. Exosomes derived from hypoxic epithelial ovarian cancer deliver microRNA-940 to induce macrophage M2 polarization. Oncol. Rep. 2017, 38, 522–528. [Google Scholar] [CrossRef] [PubMed]
  113. Chen, X.; Zhou, J.; Li, X.; Wang, X.; Lin, Y.; Wang, X. Exosomes derived from hypoxic epithelial ovarian cancer cells deliver microRNAs to macrophages and elicit a tumor-promoted phenotype. Cancer Lett. 2018, 435, 80–91. [Google Scholar] [CrossRef] [PubMed]
  114. Wang, X.; Luo, G.; Zhang, K.; Cao, J.; Huang, C.; Jiang, T.; Liu, B.; Su, L.; Qiu, Z. Hypoxic Tumor-Derived Exosomal miR-301a Mediates M2 Macrophage Polarization via PTEN/PI3Kγ to Promote Pancreatic Cancer Metastasis. Cancer Res. 2018, 78, 4586–4598. [Google Scholar] [CrossRef] [PubMed]
  115. McDonald, D.M.; Choyke, P.L. Imaging of angiogenesis: From microscope to clinic. Nat. Med. 2003, 9, 713–725. [Google Scholar] [CrossRef]
  116. di Tomaso, E.; Capen, D.; Haskell, A.; Hart, J.; Logie, J.J.; Jain, R.K.; McDonald, D.M.; Jones, R.; Munn, L.L. Mosaic Tumor Vessels: Cellular Basis and Ultrastructure of Focal Regions Lacking Endothelial Cell Markers. Cancer Res. 2005, 65, 5740–5749. [Google Scholar] [CrossRef]
  117. McDonald, D.M.; Baluk, P. Significance of blood vessel leakiness in cancer. Cancer Res. 2002, 62, 5381–5385. [Google Scholar]
  118. Boucher, Y.; Jain, R.K. Microvascular pressure is the principal driving force for interstitial hypertension in solid tumors: Implications for vascular collapse. Cancer Res. 1992, 52, 5110–5114. [Google Scholar]
  119. Seaman, S.; Stevens, J.; Yang, M.Y.; Logsdon, D.; Graff-Cherry, C.; Croix, B.S. Genes that Distinguish Physiological and Pathological Angiogenesis. Cancer Cell 2007, 11, 539–554. [Google Scholar] [CrossRef]
  120. Goh, P.P.; Sze, D.M.; Roufogalis, B.D. Molecular and Cellular Regulators of Cancer Angiogenesis. Curr. Cancer Drug Targets 2007, 7, 743–758. [Google Scholar] [CrossRef]
  121. Hida, K.; Klagsbrun, M. A New Perspective on Tumor Endothelial Cells: Unexpected Chromosome and Centrosome Abnormalities. Cancer Res. 2005, 65, 2507–2510. [Google Scholar] [CrossRef]
  122. Hida, K.; Maishi, N.; Annan, D.A.; Hida, Y. Contribution of Tumor Endothelial Cells in Cancer Progression. Int. J. Mol. Sci. 2018, 19, 1272. [Google Scholar] [CrossRef] [PubMed]
  123. Amin, D.N.; Hida, K.; Bielenberg, D.R.; Klagsbrun, M. Tumor Endothelial Cells Express Epidermal Growth Factor Receptor (EGFR) but not ErbB3 and Are Responsive to EGF and to EGFR Kinase Inhibitors. Cancer Res. 2006, 66, 2173–2180. [Google Scholar] [CrossRef] [PubMed]
  124. Tsuchiya, K.; Hida, K.; Hida, Y.; Muraki, C.; Ohga, N.; Akino, T.; Kondo, T.; Miseki, T.; Nakagawa, K.; Shindoh, M.; et al. Adrenomedullin antagonist suppresses tumor formation in renal cell carcinoma through inhibitory effects on tumor endothelial cells and endothelial progenitor mobilization. Int. J. Oncol. 2010, 36, 1379–1386. [Google Scholar] [CrossRef] [PubMed]
  125. Matsuda, K.; Ohga, N.; Hida, Y.; Muraki, C.; Tsuchiya, K.; Kurosu, T.; Akino, T.; Shih, S.-C.; Totsuka, Y.; Klagsbrun, M.; et al. Isolated tumor endothelial cells maintain specific character during long-term culture. Biochem. Biophys. Res. Commun. 2010, 394, 947–954. [Google Scholar] [CrossRef]
  126. Heldin, C.-H.; Rubin, K.; Pietras, K.; Östman, A. High interstitial fluid pressure—An obstacle in cancer therapy. Nat. Rev. Cancer 2004, 4, 806–813. [Google Scholar] [CrossRef]
  127. Butler, J.M.; Kobayashi, H.; Rafii, S. Instructive role of the vascular niche in promoting tumour growth and tissue repair by angiocrine factors. Nat. Rev. Cancer 2010, 10, 138–146. [Google Scholar] [CrossRef]
  128. Cao, Z.; Ding, B.-S.; Guo, P.; Lee, S.B.; Butler, J.M.; Casey, S.C.; Simons, M.; Tam, W.; Felsher, D.W.; Shido, K.; et al. Angiocrine Factors Deployed by Tumor Vascular Niche Induce B Cell Lymphoma Invasiveness and Chemoresistance. Cancer Cell 2014, 25, 350–365. [Google Scholar] [CrossRef]
  129. Cao, Z.; Scandura, J.M.; Inghirami, G.G.; Shido, K.; Ding, B.-S.; Rafii, S. Molecular Checkpoint Decisions Made by Subverted Vascular Niche Transform Indolent Tumor Cells into Chemoresistant Cancer Stem Cells. Cancer Cell 2016, 31, 110–126. [Google Scholar] [CrossRef]
  130. Mao, Y.; Wang, Y.; Dong, L.; Zhang, Y.; Zhang, Y.; Wang, C.; Zhang, Q.; Yang, S.; Cao, L.; Zhang, X.; et al. Hypoxic exosomes facilitate angiogenesis and metastasis in esophageal squamous cell carcinoma through altering the phenotype and transcriptome of endothelial cells. J. Exp. Clin. Cancer Res. 2019, 38, 389. [Google Scholar] [CrossRef]
  131. Chen, C.; Liu, Y.; Liu, L.; Si, C.; Xu, Y.; Wu, X.; Wang, C.; Sun, Z.; Kang, Q. Exosomal circTUBGCP4 promotes vascular endothelial cell tipping and colorectal cancer metastasis by activating Akt signaling pathway. J. Exp. Clin. Cancer Res. 2023, 42, 46. [Google Scholar] [CrossRef]
  132. Biagioni, A.; Laurenzana, A.; Menicacci, B.; Peppicelli, S.; Andreucci, E.; Bianchini, F.; Guasti, D.; Paoli, P.; Serratì, S.; Mocali, A.; et al. uPAR-expressing melanoma exosomes promote angiogenesis by VE-Cadherin, EGFR and uPAR overexpression and rise of ERK1,2 signaling in endothelial cells. Cell. Mol. Life Sci. 2021, 78, 3057–3072. [Google Scholar] [CrossRef] [PubMed]
  133. Zeng, Z.; Li, Y.; Pan, Y.; Lan, X.; Song, F.; Sun, J.; Zhou, K.; Liu, X.; Ren, X.; Wang, F.; et al. Cancer-derived exosomal miR-25-3p promotes pre-metastatic niche formation by inducing vascular permeability and angiogenesis. Nat. Commun. 2018, 9, 5395. [Google Scholar] [CrossRef] [PubMed]
  134. Pruneri, G.; Vingiani, A.; Denkert, C. Tumor infiltrating lymphocytes in early breast cancer. Breast 2017, 37, 207–214. [Google Scholar] [CrossRef] [PubMed]
  135. Li, R.; Cao, L. The role of tumor-infiltrating lymphocytes in triple-negative breast cancer and the research progress of adoptive cell therapy. Front. Immunol. 2023, 14, 1194020. [Google Scholar] [CrossRef]
  136. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef]
  137. Kitamura, T.; Qian, B.-Z.; Pollard, J.W. Immune cell promotion of metastasis. Nat. Rev. Immunol. 2015, 15, 73–86. [Google Scholar] [CrossRef]
  138. Zhang, Z.; Liu, S.; Zhang, B.; Qiao, L.; Zhang, Y. T Cell Dysfunction and Exhaustion in Cancer. Front. Cell Dev. Biol. 2020, 8, 17. [Google Scholar] [CrossRef]
  139. Anderson, N.M.; Simon, M.C. The tumor microenvironment. Curr. Biol. 2020, 30, R921–R925. [Google Scholar] [CrossRef]
  140. Whiteside, T.L. Tumor-Infiltrating Lymphocytes and Their Role in Solid Tumor Progression. Exp. Suppl. 2022, 113, 89–106. [Google Scholar] [CrossRef]
  141. Dieci, M.V.; Mathieu, M.C.; Guarneri, V.; Conte, P.; Delaloge, S.; Andre, F.; Goubar, A. Prognostic and predictive value of tumor-infiltrating lymphocytes in two phase III randomized adjuvant breast cancer trials. Ann. Oncol. 2015, 26, 1698–1704. [Google Scholar] [CrossRef]
  142. Zhao, Y.; Ge, X.; He, J.; Cheng, Y.; Wang, Z.; Wang, J.; Sun, L. The prognostic value of tumor-infiltrating lymphocytes in colorectal cancer differs by anatomical subsite: A systematic review and meta-analysis. World J. Surg. Oncol. 2019, 17, 85. [Google Scholar] [CrossRef] [PubMed]
  143. Hashemi, S.; Fransen, M.; Niemeijer, A.; Ben Taleb, N.; Houda, I.; Veltman, J.; Commissaris, A.B.; Daniels, H.; Crombag, L.; Radonic, T.; et al. Surprising impact of stromal TIL’s on immunotherapy efficacy in a real-world lung cancer study. Lung Cancer 2021, 153, 81–89. [Google Scholar] [CrossRef] [PubMed]
  144. Haj-Shomaly, J.; Vorontsova, A.; Barenholz-Cohen, T.; Levi-Galibov, O.; Devarasetty, M.; Timaner, M.; Raviv, Z.; Cooper, T.J.; Soker, S.; Hasson, P.; et al. T Cells Promote Metastasis by Regulating Extracellular Matrix Remodeling following Chemotherapy. Cancer Res. 2022, 82, 278–291. [Google Scholar] [CrossRef] [PubMed]
  145. Hall, M.; Liu, H.; Malafa, M.; Centeno, B.; Hodul, P.J.; Pimiento, J.; Pilon-Thomas, S.; Sarnaik, A.A. Expansion of tumor-infiltrating lymphocytes (TIL) from human pancreatic tumors. J. Immunother. Cancer 2016, 4, 61. [Google Scholar] [CrossRef]
  146. Baghaei, K.; Tokhanbigli, S.; Asadzadeh, H.; Nmaki, S.; Zali, M.R.; Hashemi, S.M. Exosomes as a novel cell-free therapeutic approach in gastrointestinal diseases. J. Cell. Physiol. 2019, 234, 9910–9926. [Google Scholar] [CrossRef]
  147. Kishton, R.J.; Vodnala, S.K.; Vizcardo, R.; Restifo, N.P. Next generation immunotherapy: Enhancing stemness of polyclonal T cells to improve anti-tumor activity. Curr. Opin. Immunol. 2022, 74, 39–45. [Google Scholar] [CrossRef]
  148. Olejarz, W.; Dominiak, A.; Żołnierzak, A.; Kubiak-Tomaszewska, G.; Lorenc, T. Tumor-Derived Exosomes in Immunosuppression and Immunotherapy. J. Immunol. Res. 2020, 2020, 6272498. [Google Scholar] [CrossRef]
  149. Whiteside, T.L. Immune modulation of T-cell and NK (natural killer) cell activities by TEXs (tumour-derived exosomes). Biochem. Soc. Trans. 2013, 41, 245–251. [Google Scholar] [CrossRef]
  150. Whiteside, T.L. Exosomes carrying immunoinhibitory proteins and their role in cancer. Clin. Exp. Immunol. 2017, 189, 259–267. [Google Scholar] [CrossRef]
  151. Nakazawa, Y.; Nishiyama, N.; Koizumi, H.; Kanemaru, K.; Nakahashi-Oda, C.; Shibuya, A. Tumor-derived extracellular vesicles regulate tumor-infiltrating regulatory T cells via the inhibitory immunoreceptor CD300a. eLife 2021, 10, e61999. [Google Scholar] [CrossRef]
  152. Wieckowski, E.U.; Visus, C.; Szajnik, M.; Szczepanski, M.J.; Storkus, W.J.; Whiteside, T.L. Tumor-Derived Microvesicles Promote Regulatory T Cell Expansion and Induce Apoptosis in Tumor-Reactive Activated CD8+ T Lymphocytes. J. Immunol. 2009, 183, 3720–3730. [Google Scholar] [CrossRef] [PubMed]
  153. Wieckowski, E.; Whiteside, T.L. Human Tumor-Derived vs Dendritic Cell-Derived Exosomes Have Distinct Biologic Roles and Molecular Profiles. Immunol. Res. 2006, 36, 247–254. [Google Scholar] [CrossRef] [PubMed]
  154. Montes, C.L.; Chapoval, A.I.; Nelson, J.; Orhue, V.; Zhang, X.; Schulze, D.H.; Strome, S.E.; Gastman, B.R. Tumor-Induced Senescent T Cells with Suppressor Function: A Potential Form of Tumor Immune Evasion. Cancer Res. 2008, 68, 870–879. [Google Scholar] [CrossRef] [PubMed]
  155. Zhang, Y.; Pfannenstiel, L.W.; Bolesta, E.; Montes, C.L.; Zhang, X.; Chapoval, A.I.; Gartenhaus, R.B.; Strome, S.E.; Gastman, B.R. Interleukin-7 Inhibits Tumor-Induced CD27−CD28− Suppressor T Cells: Implications for Cancer Immunotherapy. Clin. Cancer Res. 2011, 17, 4975–4986. [Google Scholar] [CrossRef]
  156. Maybruck, B.T.; Pfannenstiel, L.W.; Diaz-Montero, M.; Gastman, B.R. Tumor-derived exosomes induce CD8+ T cell suppressors. J. Immunother. Cancer 2017, 5, 65. [Google Scholar] [CrossRef]
  157. Kim, J.W.; Wieckowski, E.; Taylor, D.D.; Reichert, T.E.; Watkins, S.; Whiteside, T.L. Fas ligand-positive membranous vesicles isolated from sera of patients with oral cancer induce apoptosis of activated T lymphocytes. Clin. Cancer Res. 2005, 11, 1010–1020. [Google Scholar] [CrossRef]
  158. Abusamra, A.J.; Zhong, Z.; Zheng, X.; Li, M.; Ichim, T.E.; Chin, J.L.; Min, W.-P. Tumor exosomes expressing Fas ligand mediate CD8+ T-cell apoptosis. Blood Cells Mol. Dis. 2005, 35, 169–173. [Google Scholar] [CrossRef]
  159. Salama, P.; Phillips, M.; Grieu, F.; Morris, M.; Zeps, N.; Joseph, D.; Platell, C.; Iacopetta, B. Tumor-Infiltrating FOXP3+ T Regulatory Cells Show Strong Prognostic Significance in Colorectal Cancer. J. Clin. Oncol. 2009, 27, 186–192. [Google Scholar] [CrossRef]
  160. Agarwal, A.; Fanelli, G.; Letizia, M.; Tung, S.L.; Boardman, D.; Lechler, R.; Lombardi, G.; Smyth, L.A. Regulatory T Cell-Derived Exosomes: Possible Therapeutic and Diagnostic Tools in Transplantation. Front. Immunol. 2014, 5, 555. [Google Scholar] [CrossRef]
  161. Greening, D.W.; Gopal, S.K.; Xu, R.; Simpson, R.J.; Chen, W. Exosomes and their roles in immune regulation and cancer. Semin. Cell Dev. Biol. 2015, 40, 72–81. [Google Scholar] [CrossRef]
  162. Seo, N.; Akiyoshi, K.; Shiku, H. Exosome-mediated regulation of tumor immunology. Cancer Sci. 2018, 109, 2998–3004. [Google Scholar] [CrossRef] [PubMed]
  163. Schwarzenbach, H.; Gahan, P.B. Exosomes in Immune Regulation. Noncoding RNA 2021, 7, 4. [Google Scholar] [CrossRef] [PubMed]
  164. Li, J.; Huang, S.; Zhou, Z.; Lin, W.; Chen, S.; Chen, M.; Ye, Y. Exosomes derived from rAAV/AFP-transfected dendritic cells elicit specific T cell-mediated immune responses against hepatocellular carcinoma. Cancer Manag. Res. 2018, 10, 4945–4957. [Google Scholar] [CrossRef] [PubMed]
  165. Lu, Z.; Zuo, B.; Jing, R.; Gao, X.; Rao, Q.; Liu, Z.; Qi, H.; Guo, H.; Yin, H. Dendritic cell-derived exosomes elicit tumor regression in autochthonous hepatocellular carcinoma mouse models. J. Hepatol. 2017, 67, 739–748. [Google Scholar] [CrossRef] [PubMed]
  166. Kaplan, R.N.; Riba, R.D.; Zacharoulis, S.; Bramley, A.H.; Vincent, L.; Costa, C.; MacDonald, D.D.; Jin, D.K.; Shido, K.; Kerns, S.A.; et al. VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature 2005, 438, 820–827. [Google Scholar] [CrossRef] [PubMed]
  167. Dong, Q.; Liu, X.; Cheng, K.; Sheng, J.; Kong, J.; Liu, T. Pre-metastatic Niche Formation in Different Organs Induced by Tumor Extracellular Vesicles. Front. Cell Dev. Biol. 2021, 9, 733627. [Google Scholar] [CrossRef]
  168. Hiratsuka, S.; Nakamura, K.; Iwai, S.; Murakami, M.; Itoh, T.; Kijima, H.; Shipley, J.; Senior, R.M.; Shibuya, M. MMP9 induction by vascular endothelial growth factor receptor-1 is involved in lung-specific metastasis. Cancer Cell 2002, 2, 289–300. [Google Scholar] [CrossRef]
  169. Hiratsuka, S.; Watanabe, A.; Sakurai, Y.; Akashi-Takamura, S.; Ishibashi, S.; Miyake, K.; Shibuya, M.; Akira, S.; Aburatani, H.; Maru, Y. The S100A8–serum amyloid A3–TLR4 paracrine cascade establishes a pre-metastatic phase. Nat. Cell Biol. 2008, 10, 1349–1355. [Google Scholar] [CrossRef]
  170. Sceneay, J.; Chow, M.T.; Chen, A.; Halse, H.M.; Wong, C.S.; Andrews, D.M.; Sloan, E.K.; Parker, B.S.; Bowtell, D.D.; Smyth, M.J.; et al. Primary Tumor Hypoxia Recruits CD11b+/Ly6Cmed/Ly6G+ Immune Suppressor Cells and Compromises NK Cell Cytotoxicity in the Premetastatic Niche. Cancer Res. 2012, 72, 3906–3911. [Google Scholar] [CrossRef]
  171. Peinado, H.; Lavotshkin, S.; Lyden, D. The secreted factors responsible for pre-metastatic niche formation: Old sayings and new thoughts. Semin. Cancer Biol. 2011, 21, 139–146. [Google Scholar] [CrossRef]
  172. Li, R.; Wen, A.; Lin, J. Pro-Inflammatory Cytokines in the Formation of the Pre-Metastatic Niche. Cancers 2020, 12, 3752. [Google Scholar] [CrossRef] [PubMed]
  173. Adekoya, T.O.; Richardson, R.M. Cytokines and Chemokines as Mediators of Prostate Cancer Metastasis. Int. J. Mol. Sci. 2020, 21, 4449. [Google Scholar] [CrossRef] [PubMed]
  174. de Lope, L.R.; Sánchez-Herrero, E.; Serna-Blasco, R.; Provencio, M.; Romero, A. Cancer as an infective disease: The role of EVs in tumorigenesis. Mol. Oncol. 2022, 17, 390–406. [Google Scholar] [CrossRef]
  175. Sánchez, G.B.; 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]
  176. Gao, Y.; Bado, I.; Wang, H.; Zhang, W.; Rosen, J.M.; Zhang, X.H.-F. Metastasis Organotropism: Redefining the Congenial Soil. Dev. Cell 2019, 49, 375–391. [Google Scholar] [CrossRef] [PubMed]
  177. Zhou, W.; Fong, M.Y.; Min, Y.; Somlo, G.; Liu, L.; Palomares, M.R.; Yu, Y.; Chow, A.; O’Connor, S.T.F.; Chin, A.R.; et al. Cancer-secreted miR-105 destroys vascular endothelial barriers to promote metastasis. Cancer Cell 2014, 25, 501–515. [Google Scholar] [CrossRef] [PubMed]
  178. Lin, B.; Liu, C.; Shi, E.; Jin, Q.; Zhao, W.; Wang, J.; Ji, R. MiR-105-3p acts as an oncogene to promote the proliferation and metastasis of breast cancer cells by targeting GOLIM4. BMC Cancer 2021, 21, 275. [Google Scholar] [CrossRef] [PubMed]
  179. Hannafon, B.N.; Carpenter, K.J.; Berry, W.L.; Janknecht, R.; Dooley, W.C.; Ding, W.-Q. Exosome-mediated microRNA signaling from breast cancer cells is altered by the anti-angiogenesis agent docosahexaenoic acid (DHA). Mol. Cancer 2015, 14, 133. [Google Scholar] [CrossRef]
  180. 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]
  181. Kosaka, N.; Iguchi, H.; Hagiwara, K.; Yoshioka, Y.; Takeshita, F.; Ochiya, T. Neutral Sphingomyelinase 2 (nSMase2)-dependent Exosomal Transfer of Angiogenic MicroRNAs Regulate Cancer Cell Metastasis. J. Biol. Chem. 2013, 288, 10849–10859. [Google Scholar] [CrossRef]
  182. Grange, C.; Tapparo, M.; Collino, F.; Vitillo, L.; Damasco, C.; Deregibus, M.C.; Tetta, C.; Bussolati, B.; Camussi, G. Microvesicles Released from Human Renal Cancer Stem Cells Stimulate Angiogenesis and Formation of Lung Premetastatic Niche. Cancer Res. 2011, 71, 5346–5356. [Google Scholar] [CrossRef] [PubMed]
  183. Labi, V.; Erlacher, M. How cell death shapes cancer. Cell Death Dis. 2015, 6, e1675. [Google Scholar] [CrossRef] [PubMed]
  184. Cox, J.; Weinman, S. Mechanisms of doxorubicin resistance in hepatocellular carcinoma. Hepatic Oncol. 2016, 3, 57–59. [Google Scholar] [CrossRef] [PubMed]
  185. Safaei, R.; Larson, B.J.; Cheng, T.C.; Gibson, M.A.; Otani, S.; Naerdemann, W.; Howell, S.B. Abnormal lysosomal trafficking and enhanced exosomal export of cisplatin in drug-resistant human ovarian carcinoma cells. Mol. Cancer Ther. 2005, 4, 1595–1604. [Google Scholar] [CrossRef] [PubMed]
  186. Dong, X.; Bai, X.; Ni, J.; Zhang, H.; Duan, W.; Graham, P.; Li, Y. Exosomes and breast cancer drug resistance. Cell Death Dis. 2020, 11, 987. [Google Scholar] [CrossRef]
  187. Song, H.; Liu, B.; Dong, B.; Xu, J.; Zhou, H.; Na, S.; Liu, Y.; Pan, Y.; Chen, F.; Li, L.; et al. Exosome-Based Delivery of Natural Products in Cancer Therapy. Front. Cell Dev. Biol. 2021, 9, 650426. [Google Scholar] [CrossRef]
  188. Xue, D.; Han, J.; Liang, Z.; Jia, L.; Liu, Y.; Tuo, H.; Peng, Y. Current Perspectives on the Unique Roles of Exosomes in Drug Resistance of Hepatocellular Carcinoma. J. Hepatocell. Carcinoma 2022, 9, 99–112. [Google Scholar] [CrossRef]
  189. Zheng, P.; Chen, L.; Yuan, X.; Luo, Q.; Liu, Y.; Xie, G.; Ma, Y.; Shen, L. Exosomal transfer of tumor-associated macrophage-derived miR-21 confers cisplatin resistance in gastric cancer cells. J. Exp. Clin. Cancer Res. 2017, 36, 53. [Google Scholar] [CrossRef]
  190. Ji, R.; Zhang, B.; Zhang, X.; Xue, J.; Yuan, X.; Yan, Y.; Wang, M.; Zhu, W.; Qian, H.; Xu, W. Exosomes derived from human mesenchymal stem cells confer drug resistance in gastric cancer. Cell Cycle 2015, 14, 2473–2483. [Google Scholar] [CrossRef]
  191. Zhao, H.; Achreja, A.; Iessi, E.; Logozzi, M.; Mizzoni, D.; Di Raimo, R.; Nagrath, D.; Fais, S. The key role of extracellular vesicles in the metastatic process. Biochim. Biophys. Acta (BBA) Rev. Cancer 2017, 1869, 64–77. [Google Scholar] [CrossRef]
  192. Spugnini, E.P.; Logozzi, M.; Di Raimo, R.; Mizzoni, D.; Fais, S. A Role of Tumor-Released Exosomes in Paracrine Dissemination and Metastasis. Int. J. Mol. Sci. 2018, 19, 3968. [Google Scholar] [CrossRef] [PubMed]
  193. Boelens, M.C.; Wu, T.J.; Nabet, B.Y.; Xu, B.; Qiu, Y.; Yoon, T.; Azzam, D.J.; Victor, C.T.-S.; Wiemann, B.Z.; Ishwaran, H.; et al. Exosome Transfer from Stromal to Breast Cancer Cells Regulates Therapy Resistance Pathways. Cell 2014, 159, 499–513. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Extracellular vesicle production. Extracellular vesicles, and in particular exosomes, originate from multivesicular bodies (MVBs) which are formed during the inward budding of the plasma membrane. The subsequent inward budding of the MVB membrane encapsulates and packages cytosolic components resulting in the formation of EVs. EV cargo comprises selectively packaged proteins (e.g., tetraspanins, cytoplasmic proteins, and enzymes), nucleic acids (e.g., DNA, RNA, and miRNAs) and lipids, which have been shown to be cell-type dependent reflecting the metabolic status of their cells of origin. The subsequent fusion of MVBs with the plasma membrane results in the release of EVs into the tumor microenvironment.
Figure 1. Extracellular vesicle production. Extracellular vesicles, and in particular exosomes, originate from multivesicular bodies (MVBs) which are formed during the inward budding of the plasma membrane. The subsequent inward budding of the MVB membrane encapsulates and packages cytosolic components resulting in the formation of EVs. EV cargo comprises selectively packaged proteins (e.g., tetraspanins, cytoplasmic proteins, and enzymes), nucleic acids (e.g., DNA, RNA, and miRNAs) and lipids, which have been shown to be cell-type dependent reflecting the metabolic status of their cells of origin. The subsequent fusion of MVBs with the plasma membrane results in the release of EVs into the tumor microenvironment.
Biomedicines 11 02534 g001
Figure 2. The “ecosystem” of the tumor microenvironment (TME). The tumor microenvironment of solid tumors is composed of extracellular matrix (ECM) components and a multitude of different stromal cells, including macrophages, dendritic cells, neutrophils, and myeloid-derived suppressor cells, adipocytes, fibroblasts, and endothelial cells, in addition to both non-cancerous and cancerous epithelial cells.
Figure 2. The “ecosystem” of the tumor microenvironment (TME). The tumor microenvironment of solid tumors is composed of extracellular matrix (ECM) components and a multitude of different stromal cells, including macrophages, dendritic cells, neutrophils, and myeloid-derived suppressor cells, adipocytes, fibroblasts, and endothelial cells, in addition to both non-cancerous and cancerous epithelial cells.
Biomedicines 11 02534 g002
Figure 3. Tumor-derived EVs are critical mediators in tumor-cell metastasis. Extracellular secretion by tumor cells act as nanosized intracellular messages between cancer cells and surrounding stromal cells within the tumor microenvironment. Their cargo, which is composed of proteins, lipids, DNA, mRNAs, miRNAs, and other non-coding RNAs, is tailored by their cells of origin to specifically and directly promote cancer progression and metastasis by affecting epithelial-to-mesenchymal transition, tumor cell intravasation, dissemination, extravasation, and most critically, pre-metastatic niche formation.
Figure 3. Tumor-derived EVs are critical mediators in tumor-cell metastasis. Extracellular secretion by tumor cells act as nanosized intracellular messages between cancer cells and surrounding stromal cells within the tumor microenvironment. Their cargo, which is composed of proteins, lipids, DNA, mRNAs, miRNAs, and other non-coding RNAs, is tailored by their cells of origin to specifically and directly promote cancer progression and metastasis by affecting epithelial-to-mesenchymal transition, tumor cell intravasation, dissemination, extravasation, and most critically, pre-metastatic niche formation.
Biomedicines 11 02534 g003
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

Mitchell, M.I.; Loudig, O. Communicator Extraordinaire: Extracellular Vesicles in the Tumor Microenvironment Are Essential Local and Long-Distance Mediators of Cancer Metastasis. Biomedicines 2023, 11, 2534. https://doi.org/10.3390/biomedicines11092534

AMA Style

Mitchell MI, Loudig O. Communicator Extraordinaire: Extracellular Vesicles in the Tumor Microenvironment Are Essential Local and Long-Distance Mediators of Cancer Metastasis. Biomedicines. 2023; 11(9):2534. https://doi.org/10.3390/biomedicines11092534

Chicago/Turabian Style

Mitchell, Megan I., and Olivier Loudig. 2023. "Communicator Extraordinaire: Extracellular Vesicles in the Tumor Microenvironment Are Essential Local and Long-Distance Mediators of Cancer Metastasis" Biomedicines 11, no. 9: 2534. https://doi.org/10.3390/biomedicines11092534

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