Next Article in Journal
Paediatric Partial-Thickness Burn Therapy: A Meta-Analysis and Systematic Review of Randomised Controlled Trials
Next Article in Special Issue
Impact of Storage Conditions on EV Integrity/Surface Markers and Cargos
Previous Article in Journal
Total Wrist Arthroplasty for Posttraumatic Wrist Osteoarthritis: A Cohort Study Comparing Three Indications
Previous Article in Special Issue
Non-Exosomal and Exosome-Derived miRNAs as Promising Biomarkers in Canine Mammary Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Extracellular Vesicles and Cell Pathways Involved in Cancer Chemoresistance

by
Lara Console
* and
Mariafrancesca Scalise
*
Department of Biology, Ecology and Earth Sciences (DiBEST), Laboratory of Biochemistry, Molecular Biotechnology and Molecular Biology, University of Calabria, Via P. Bucci 4c, 87036 Arcavacata di Rende, Italy
*
Authors to whom correspondence should be addressed.
Life 2022, 12(5), 618; https://doi.org/10.3390/life12050618
Submission received: 20 March 2022 / Revised: 15 April 2022 / Accepted: 16 April 2022 / Published: 21 April 2022
(This article belongs to the Special Issue Exosomes: Biogenesis, Biologic Function and Clinical Potential)

Abstract

:
Chemoresistance is a pharmacological condition that allows transformed cells to maintain their proliferative phenotype in the presence of administered anticancer drugs. Recently, extracellular vesicles, including exosomes, have been identified as additional players responsible for the chemoresistance of cancer cells. These are nanovesicles that are released by almost all cell types in both physiological and pathological conditions and contain proteins and nucleic acids as molecular cargo. Extracellular vesicles released in the bloodstream reach recipient cells and confer them novel metabolic properties. Exosomes can foster chemoresistance by promoting prosurvival and antiapoptotic pathways, affecting cancer stem cells and immunotherapies, and stimulating drug efflux. In this context, a crucial role is played by membrane transporters belonging to ABC, SLC, and P-type pump families. These proteins are fundamental in cell metabolism and drug transport in either physiological or pathological conditions. In this review, different roles of extracellular vesicles in drug resistance of cancer cells will be explored.

1. Introduction

Cancer is the second leading cause of death globally, representing one of the most significant public health problems [1]. Although novel therapeutic strategies have been developed, chemotherapy remains the leading approach for tumor treatment [2]. Despite significant advances in increasing the efficacy of chemotherapeutics, chemoresistance remains one of the major obstacles in cancer treatment and represents a significant risk to the survival of patients [3,4,5]. Two main classes of chemoresistance can be distinguished: intrinsic and acquired [6]. The intrinsic resistance is associated with factors, such as stem cells in the tumor mass, present before any drug exposure. The acquired resistance is a stepwise process involving various mechanisms, including increased DNA repair, altered expression of oncogenes or tumor suppressor genes, altered drug targets, increased drug efflux, and increased autophagy [7,8]. A growing body of evidence suggests that extracellular vesicles can play a role in mediating drug resistance of cancer cells [9]. Indeed, these vesicles, released from almost all cells, carry different cargo types, including mRNA, microRNA, long noncoding RNA, lipids, and proteins, that may influence the phenotype and metabolisms of recipient cells even they are located far from the donors [7,10].
The cargo of extracellular vesicles changes accordingly with the physio-pathological conditions of the donor cells [10,11]. In the chemoresistance scenario, extracellular vesicles from drug-resistant cancer cells can provide drug-sensitive cells with nucleic acids and proteins that confer a resistance phenotype (Figure 1 and Table 1). Moreover, some evidence demonstrates that extracellular vesicles can be directly involved in drug extrusion from cancer cells [12]. The understanding of vesicle-mediated drug resistance is required for finding new targets of chemoresistance, as well as to identify suitable biomarkers devoted to managing treatments at the earliest sign of drug resistance. Since vesicles can be collected from body fluids such as blood, urine, and saliva, they represent an easily accessible source of biomarkers for monitoring the effectiveness of therapy [10]. Besides other advantages and novelties, detecting biomarkers from body fluids via noninvasive methods also avoids the harm and discomfort of solid tissue biopsy [13].
Extracellular vesicles (EVs) can be classified into different subtypes. Using the nomenclature suggested by Minimal Information for Studies of Extracellular Vesicles 2018 (MISEV2018) [69], it is possible to distinguish three different subtypes of vesicles: large EVs (>1 µm), medium EVs (>150 nm and <1 µm), and small EVs (<150 nm) [69,70]. Another possible classification is based on the cell origin of the extracellular vesicles. A first class includes apoptotic bodies, which bud directly from the plasma membrane during late apoptosis and contain disaggregated cell components and organelles. The medium and small size extracellular vesicles are also, respectively, described as microvesicles that directly bud from the cell membrane and exosomes which originate from the exocytic fusion of multivesicular bodies with the plasma membrane (Figure 1).
However, due to the heterogeneity in terms of size and the overlap of some biochemical characteristics (markers) between exosomes and microvesicles, it is difficult to unequivocally distinguish these two subgroups of vesicles [71,72]. As a consequence, most studies on the extracellular vesicles are carried out on a mixture of exosomes and microvesicles. For this reason, we will use the term EVs instead of exosomes or microvesicles throughout the review.
Another important issue to consider about extracellular vesicles is the possible coisolation of particles such as ribonucleoprotein aggregates, lipoproteins, and exomers [69]. For instance, miRNAs bound with high-density lipoproteins may be co-isolated as a contaminant of miRNAs embedded into extracellular vesicles [73]. Thus, contaminants can affect the correct interpretation of the experimental results. Further analyses of the isolated EVs to determine the presence and the proportion of such contaminants are highly desirable [69]. However, in certain cases such as when only small amounts of working materials are available, these additional investigations cannot be performed. In this case, as suggested by MISEV2018, the results will be examined as action mediated by EV-enriched preparations, rather than EV-specific activity [69].

2. Extracellular Vesicles, Drug Efflux, and Membrane Transporters

Drug efflux is a common mechanism underlying the chemoresistance of cancer cells. In this respect, vesicles have been shown to play some key roles. One of the most common cancer features is the presence of an acidic microenvironment primarily due to altered glycolysis and hypoxia that causes the production of lactic acid and release of protons to the extracellular milieu [74,75]. Recent studies demonstrated a link between microenvironment acidity and increased secretion of vesicles [76,77]. In particular, there is evidence that drug-resistant cancer cells secrete more exosomes than sensitive cells and that these vesicles contain anticancer drugs. Accordingly, Federici et al. showed that human melanoma cells could develop resistance against cisplatin, exporting the drug via vesicles whose release is increased in the presence of an acidic environment [78]. The pretreatment of melanoma cells with the proton pump inhibitor lansoprazole reduces exosome secretion and improves cisplatin efficacy [78]. Accordingly, in vitro and in vivo experiments showed that inhibition of vesicle biogenesis enhances the accumulation of doxorubicin and pixantrone in B-cell lymphoma [79]. The treatment of breast and ovarian cancer cells with doxorubicin and pixantrone leads to increased secretion of vesicles containing considerable amounts of these drugs, compared to untreated cells that in turn induce resistance [80].
Another mechanism by which vesicle secretion can be increased is based on annexin A3 action. Upregulation of annexin A3, belonging to the phospholipid-binding protein family, induces biogenesis and release of exosomes [81]. According to that, it was found that this protein is enriched in drug-resistant ovarian cancer cells and its upregulation correlated with the reduction in the intracellular Pt-drug concentration, which in turn prevents apoptosis [7,82]. Moreover, annexin A3 was also directly found in extracellular vesicles, suggesting that the transfer of this protein may induce chemoresistance in recipient cells [81].
Altogether, these findings correlate with a well-assessed role of vesicles in removing toxic compounds from cells, representing a first biological function ascribed to exosome secretion.
In the context of vesicle-mediated drug resistance caused by the efflux or improper accumulation of therapeutic agents in cancer cells, membrane transporters can be involved as primary players.
Membrane transporters are classified into two major groups, according to the source of energy used for the transport reaction, namely ABC and SLC super families. ATP-binding cassettes (ABCs) are primary active transporters since they use ATP hydrolysis as energy for driving transport [83,84], whereas solute carriers (SLCs) are secondary active transporters that exploit the gradients of ions or other substrates across the cell membranes [85,86]. Another class of proteins, called P-type pumps, involved in many cell processes and present in virtually all living organisms, have to be added to the list of transporters [87]. These proteins share with the ABCs the source of energy, namely ATP hydrolysis.
The following sections will deal with the link between exosome-mediated drug resistance and transporters belonging to ABC, SLC, and P-type pump families.

2.1. ABC Transporters and Exosomes in Drug Efflux

ABCs originated early during evolution and are conserved across all kingdoms of life, sharing some structural features: regardless of being prokaryotic or eukaryotic proteins, ABCs are constituted by two transmembrane domains (TMDs) and two nucleotide-binding domains (NBDs) which are involved in the binding and hydrolysis of ATP [88,89]. ABC are classified into seven families, including more than 50 members in humans [88] (https://www.genenames.org/data/genegroup/#!/group/417, accessed on 19 March 2022). An eminent example of ABC transporters involved in drug efflux is given by the P-glycoprotein (P-gp), also known as ABCB1 or MDR1 [90,91,92]. Among the ABC transporters, P-gp is the best studied due to the discovery of its role in cancer development and progression [90,91,92]. Indeed, P-gp is famous as a drug transporter mainly involved in the efflux of chemotherapy agents and is, hence, one of the most relevant players in chemoresistance. Notwithstanding the large variety and different structures, a common feature among the substrates recognized by P-gp is the hydrophobic nature of the molecules [90,91,92]. This is probably due to the peculiar transport mechanism of P-gp that consists of anchoring the substrate to the membrane before the actual translocation through P-gp, coupled to the hydrolysis of ATP [93].
Given the role of P-gp in mediating drug efflux, strong efforts have been made to identify good and specific inhibitors [94]. However, an intriguing paradox exists: indeed, a pharmacological molecule, even though tested as an inhibitor, may behave as a substrate of P-gp. Indeed, a sizable number of drugs failed to be true inhibitors of P-gp, being rather substrates of the protein [94]. These results made it difficult to overcome the chemoresistance linked to P-gp overexpression in human cancers.
This scenario is made even more complex considering that several SNPs have been identified and annotated in P-gp, further influencing the substrate specificity [95]. Moving from these premises, it is not surprising that alteration of the P-gp expression profoundly influences the disposition and pharmacodynamics of many anticancer drugs [96].
Several studies reported the presence of P-gp in extracellular vesicles that can induce chemoresistance in sensitive recipient cancer cells [53,62,97]. In particular, P-gp is inserted in the membrane of vesicles as in the native membrane, i.e., with the transmembrane-spanning domain crossing the vesicle bilayer [97]. For instance, vesicles derived from docetaxel-resistant prostate cancer cells (PC3) transfer the drug resistance to sensitive cells by delivering P-gp as a cargo [62]. Similarly, extracellular vesicles released from doxorubicin-resistant osteosarcoma cells induce resistance in recipient cells by delivering P-gp mRNA and protein [53] (Figure 1 and Table 2). Another study demonstrated that the P-gp level in blood vesicles from patients with docetaxel-resistant prostate cancer was relatively higher than in patients that did not receive therapy [62].
The transient receptor potential channel 5 (TrpC5) was also found in extracellular vesicles deriving from adriamycin-resistant Michigan Cancer Foundation (MCF)-7 cells (Table 2) [56,98]. The transfer of TrpC5 through the vesicular membrane confers the phenotype of resistance in drug-sensitive cells. The acquisition of exosomal TrpC5 leads to Ca2+ influx in breast cancer recipient cells. The increase in Ca2+ influx was specifically ascribed to TrpC5, as confirmed by using specific TrpC5 inhibitors as well as blockers of other cell Ca2+ channels. The increase in intracellular Ca2+ concentration of recipient cells caused the nuclear translocation of the transcription factor Nuclear Factor of Activated T-cells Cytoplasmic 3 (NFATC3), which induces P-gp overexpression [56]. Similarly, the adriamycin-resistant breast cancer cells secreted vesicles containing UCH-L1 (Table 2) [54]. It is a member of the ubiquitin carboxyterminal hydrolase (UCH) family, which upregulates P-gp expression through the MAPK/ERK signaling pathway [105]. The uptake of UCH-L1-containing vesicles into adriamycin-sensitive breast cancer cells transferred the chemoresistance phenotype [54]. Furthermore, it has been reported that vesicles released by docetaxel-resistant breast cancer cell line (MCF-7) rendered recipient-sensitive MCF-7 cells resistant to the same drug by transferring the P-gp protein [55].
Besides proteins, extracellular vesicles can deliver other bioactive molecules, such as nucleic acids (Figure 2 and Table 2). As an example, extracellular vesicles derived from cancer-associated fibroblasts (CAFs) carry a specific long noncoding RNA (lncRNA), named LINC00355. This lncRNA is responsible for buffering the miRNA miR-34b-5p, known as a modulator of the P-gp protein [47]. The absorption of CAF vesicles, carrying LINC00355, by bladder cancer cells triggers the overexpression of P-gp with the consequent acquirement of a cisplatin-resistance phenotype (Table 2) [47].
A mechanism based on miRNA has also been described for ovarian cancer; in this case, the resistance of cancer cells to paclitaxel is mediated by exosomes containing miR-1246, which can target caveolin 1, i.e., a regulator of P-gp expression (Table 2). Therefore, miR-1246 released into recipient cancer cells by extracellular vesicles reduces the expression of caveolin 1, which triggers the overexpression of P-gp1, causing resistance to paclitaxel [34].
Another protein involved in drug resistance is multidrug resistance protein 1 (MRP1), also known as ABCC1 [106]. MRP1 is mainly localized at the basolateral surface of epithelial and the apical surface of brain capillaries [107]. This localization allows the efflux towards the blood of the MRP1 substrates contributing to drug and xenobiotic disposition in normal and cancer cells [106,107]. Like P-gp, ABCC1 recognizes a large variety of antineoplastic drugs, antivirals, and toxicants [106,107]. Interestingly, vesicles released from non-small-cell lung cancer contain the circular RNA phosphatidylinositol-4-phosphate 5-kinase type 1 alpha (circ_PIP5K1A) [49]. This molecule has miR-101 as a primary target; the disruption of miR-101 causes the overexpression of ABCC1 given that ABCC1 protein levels are negatively regulated by miR-101 [49]. Therefore, the miR-101/ABCC1 axis is disrupted in recipient cells, and these acquire a cisplatin-resistant phenotype (Table 2). Conversely, the knockdown of exosomal circ_PIP5K1A promoted cisplatin sensitivity in recipient lung cancer cells [49].
Another member of the ABC superfamily with a well-acknowledged role in cancer resistance is ABCG2, better known as breast cancer resistance protein (BCRP). As indicated by the name, this protein was first identified in breast cancer cells (MCF-7) as highly resistant to doxorubicin. Then, it was discovered that this protein is mainly expressed in the placental syncytiotrophoblasts, in the canalicular membrane of the hepatocytes, and on the luminal side of epithelial cells in the small and large intestine [108]. ABCG2 recognizes as a substrate a broad spectrum of anticancer drugs, sulfate and glucuronide conjugates of xenobiotics, natural compounds, and toxins [108]. In the frame of chemoresistance related to extracellular vesicles, some reports indicated regulation of ABCG2 expression by long noncoding RNAs and circular RNA present in exosomes released by cancer cells (Figure 2 and Table 2). For example, the long noncoding RNA known as linc-VLDLR is an upstream positive regulator of ABCG2 [40]. The linc-VLDLR is highly present in hepatocellular cancer (HCC) cells resistant to sorafenib, camptothecin, and doxorubicin [99]. Interestingly, vesicles released by the resistant cells contain linc-VLDLR as a cargo. The exposure of sensitive HCC cells to extracellular vesicles carrying linc-VLDLR increased ABCG2 expression, with a consequent reduction in cancer cell death [99]. The same mechanism has also been described for the linc-VLDLR/ABCG2 axis in esophageal cancer cells [40].
Finally, another link between extracellular vesicles, ABC transporters, and chemoresistance has been proposed: indeed, besides other mechanisms, the biogenesis of vesicles in hematological neoplasm is dependent on the lipid transporter ABCA3 (Table 2). ABCA3 levels in malignant lymphoma and myeloma are augmented with a consequent increase in exosome secretions. This phenomenon is linked to the resistance to anticancer drugs such as adriamycin, picoxenone [109], and rituximab [67].

2.2. P-Type ATPases and Extracellular Vesicles

The P-type ATPase family is formed by five groups (indicated by I–V symbols), each including different classes able to recognize a specific ion as a substrate [87]. These proteins are constituted by an even number of membrane-spanning domains with molecular masses ranging from 70 to 150 kDa [87]. As suggested by the name, P-type ATPases can hydrolyze ATP, allowing for ion translocation through the cell membranes [87]. The overall architecture of these proteins is well conserved within the family members and consists of four domains indicated as P-domain, N-domain, A-domain, and M-domain [87]. The P-domain is the phosphorylation site, being the core of the catalytic mechanism, and includes the N-domain, i.e., the ATP-binding domain. The A-domain is located at the N-terminus of the pumps and is responsible for the conformational changes occurring during the transport cycle; it is plausible that the A-domain plays a regulatory role [87]. Finally, the M-domain is the transmembrane portion of the pump responsible for the ion-path formation [87]. Two P-type pumps have been identified as involved in the resistance to Pt-based drugs: the copper efflux transporters ATP7A and ATP7B (Table 2) [61]. These participate in heavy metal detoxification, thus being crucial for cell life [61,110]. Therefore, it is not surprising that alterations of human ATP7A and ATP7B are associated with Wilson’s disease, which is characterized by abnormal copper accumulation in the liver and the brain [110]. In contrast to Wilson’s phenotype, the upregulation of ATP7A and ATP7B has been associated with Alzheimer’s disease and chemoresistance. Interestingly, the authors of [111] found that vesicles released by ovarian cancer cells resistant to Pt-based drugs harbor higher amounts of ATP7A and ATP7B than exosomes derived from drug-sensitive cells. The same authors also described augmented exosome secretion from resistant cells compared to that from their sensitive counterparts [61].

2.3. Solute Carrier (SLC) Transporters and Extracellular Vesicles

SLC transporters are classified into 60 families, including more than 500 members. In humans, indeed, SLCs are involved in the absorption of several nutrients and cofactors and their distribution in intracellular organelles [112]. Moreover, SLCs are responsible for the excretion of catabolites and reabsorption phenomena occurring in the kidney [112]. The role of SLCs in mediating cancer chemoresistance is well acknowledged due to their ability to mediate drug transport, besides their physiological substrates [113,114,115,116]. Several SLCs can be found in the dedicated database of exosome cargos, named Exocarta [117], which also collects the rough proteomic data about exosomes released by cancer cells; however, only little information is currently available on the function of these proteins as exosome cargo(s) [35,100,101,118].
However, some information that links SLCs to drug resistance induced by extracellular vesicles is available (Figure 2). An eminent example is the case of the equilibrative nucleoside transporter 2 (ENT2) [35,119]. It belongs to the SLC29 family and is responsible for the uptake of purine and pyrimidine nucleosides [120]. ENT2 is a ubiquitous protein mainly localized at the plasma membrane but has also been detected in nuclear membranes. Besides nucleobases, ENT2 is involved in the transport of nucleoside-based drugs [120]. The ability to recognize several pharmacological compounds as substrates made this protein a hot pharmacological target for several diseases, including cancer and acquired immunodeficiency syndrome (AIDS) [121]. In the frame of chemoresistance mediated by extracellular vesicles, the role of ENT2 has been linked to exosomes released from cancer-associated fibroblasts (CAFs) to lymphoma [35,119]. Indeed, CAFs support lymphoma cell growth and chemoresistance to gemcitabine and cytarabine by downregulating ENT2 expression (Figure 1) [122]. CAF-derived vesicles were found to contain the miRNA-4717-5p that targets a deubiquitinase, triggering a cascade event: the disruption of deubiquitinase causes higher ubiquitination of ENT2 with following faster ENT2 degradation (Table 2). Ultimately, increased resistance to anticancer drugs is observed [35].
Another SLC member involved in chemoresistance is SL9A1, known as NHE1 [123]. This protein belongs to the SLC9 family, a subgroup of the eukaryotic and prokaryotic monovalent cation proton antiporter (CPA) superfamily (Transport Protein Database http://tcdb.ucsd.edu/tcdb/, accessed on 19 March 2022) [124,125]. NHE1 is a glycosylated plasma membrane protein with a large hydrophilic C-terminus responsible for regulating its activity [126]. NHE1 is ubiquitously expressed in mammalian cells and is responsible for the exchange of Na+ and H+ with the consequent alkalization of the cell in defense of H+ derived from metabolism or electrically driven H+-accumulation [124,125]. NHE1 plays a crucial role in cell migration, proliferation, and death, and it has been linked to the sensitivity of breast cancer cells to cisplatin [125]. In good agreement, silencing of SCL9A1 was responsible for the inhibition of cell migration, inhibition, and matrix metalloproteinase production in breast cancer cells [127].
Interestingly, extracellular vesicles derived from adipose mesenchymal stem cells (AdMSC-Exos) were shown to have an antiproliferative effect in recipient breast cancer cells, ameliorating their drug resistance [100].
Indeed, these vesicles carry the miRNA miR-1236, which is an upstream negative regulator of NHE1 in breast cancer cells where a link between NHE and the Wingless-related integration site (Wnt)–catenin pathway has been proposed [100]. NHE1 activates β-catenin which, in turn, induces cancer cell proliferation and resistance to DPP. Therefore, when vesicles containing miR-1236 enter breast cancer cells, the degradation of NHE1 mRNA is responsible for the above-described antiproliferative effects [100].
Another SLC protein linked to exosomes and chemoresistance is SLC7A11, known as xCT [128]. This protein forms a functional heterodimer with the glycoprotein CD98 (SLC3A2) via a disulfide bond between two conserved cysteine residues, whose 3D structure has been recently determined by CryoEM [129]. The protein xCT is well acknowledged as a crucial player in cancer development and progression [130], being overexpressed in virtually all human cancers, despite the narrower expression in nonpathological conditions [131]. xCT is a plasma membrane transporter responsible for exchanging cystine with glutamate with a 1:1 stoichiometry; the cystine taken up from the extracellular milieu is reduced to cysteine for GSH synthesis [132]. This transport mechanism underlies the role of xCT in the redox homeostasis of cells and, then, in the oxidative stress response. Indeed, xCT has been described as the mediator of a recently discovered cell death pathway called ferroptosis, characterized by an iron-dependent abnormal peroxidation [133]. In particular, xCT impairment causes cystine depletion and consequent accumulation of lipid hydroperoxides [134]. The involvement in human cancers prompted the research of drugs with inhibitory effects on the xCT transport activity, even though a link with chemoresistance and poor survival of glioblastoma patients has been proposed [135].
In the context of anticancer research, an inhibitor of xCT that has been approved by FDA is sulfasalazine (SAS). SAS upregulates ROS via xCT inhibition leading to cell death [136]. In the exosome scenario, a link between xCT and immune checkpoint blockade (ICB) therapy has been proposed in melanoma patients [101]. The study proposed that the inhibition of xCT by SAS is strategic for treating melanoma patients in line with the altered levels of glutamate found in cancer cells [101]. However, in the same study, it has been shown that extracellular vesicles, released from melanoma cells treated with SAS, reduced the efficacy of the ICB therapy [101]. Indeed, ICB therapy aims at enhancing antitumoral immunity by acting on specific immune system components [137]. Therefore, the authors concluded that melanoma patients affected by inflammatory diseases may not be eligible for the combination of SAS and immunotherapy [101].

3. Extracellular-Vesicle-Induced Drug Resistance Promoting Prosurvival and Antiapoptotic Pathways

A growing body of evidence has demonstrated that exosomes can mediate drug resistance by modulating several cell processes (Figure 3) [3,7,138]. One of these is apoptosis, which is disturbed in both acquired and intrinsic resistance to chemotherapies [7,138]. Indeed, drug resistance frequently makes tumor cells able to inhibit apoptosis, resulting in cancer cell survival and unfavorable outcomes [139]. Extracellular vesicles can decrease proapoptotic signaling in the donor cells or increase antiapoptotic signaling in the recipient cells (Figure 3 and Table 2) [140].
In the first case, vesicles embed and remove proapoptotic proteins such as caspase-3 from the donor cells. The decrease in the intracellular concentration of caspase-3 promotes the shift towards an antiapoptotic state of the donor cells [141]. As demonstrated by Boing et al., inhibition of the release of vesicles carrying caspase-3 rescued the normal level of apoptosis in endothelial donor cells [141]. Another example of this mechanism occurs in colorectal cancer, where the tumor-suppressive miRNAs miR-145 and miR-34 are removed from cells through extracellular vesicles. This triggers a decrease in apoptosis level and an increase in 5-fluorouracil resistance of these cells (Table 2) [30].
In recipient cells, extracellular vesicles can promote tumor survival by supplying nucleic acids and proteins that directly interfere with antiapoptotic pathways [39,43] (Figure 3). As an example, vesicles released by platelets carry the membrane receptor CD41 (integrin a-IIb), which interacts with the extracellular matrix of cancer cells such as bone marrow myeloma cells [140]. The interaction with CD41-related exosomes reduces the phosphorylation of the c-Jun-N terminal kinase (JNK), with a consequent increase in antiapoptotic proteins such as Bcl-2 and reduced cleavage of caspase-3 [102]. For instance, vesicles can promote cell survival by delivering the lncRNA small nucleolar RNA host gene 14 (SNHG14) to recipient cells [43]. Indeed, the upregulation of SNHG14 has been associated with trastuzumab-resistance of HER2+ breast cell lines by targeting the apoptosis regulator Bcl-2/Bax signaling pathway. This lncRNA is highly expressed in vesicles derived from resistant cells [43]. The incubation of sensitive cells with vesicles containing SNHG14 induces trastuzumab resistance (Table 2) [43]. Following that, higher vesicular levels of lncRNA SNHG14 were detected in the serum of patients who exhibited resistance compared to responsive patients [43].
The Bcl-2/Bax pathway was also found to be modulated by another lncRNA named PART1, secreted into exosomes that disseminate drug resistance to sensitive cells (Figure 1 and Table 2) [39]. Indeed, in esophageal squamous cell carcinoma, PART1 promotes resistance to gefitinib because it is able to competitively bind miR-129 which in turn facilitates the expression of the antiapoptotic Bcl-2 protein [39] (Figure 3 and Table 2). Moreover, extracellular PART1, embedded into vesicles, can transfer the gefitinib resistance to recipient cells with the same molecular mechanism. In good agreement, high serum levels of PART1 were associated with poor response to patient treatment [39].
Furthermore, vesicles can confer resistance by delivering miRNAs into chemosensitive tumor cells, altering cell cycle control and inducing antiapoptotic programs (Figure 3) [23]. This is the case of vesicular miR-21 that induced cisplatin resistance in oral squamous cell carcinoma by targeting phosphatase and tensin homolog (PTEN) and programmed cell death protein 4 (PDCD4), which are tumor suppressors involved in apoptosis, cell transformation, invasion, and tumor progression [27]. PTEN is also suppressed through the phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) pathway activation induced by vesicular miR-32-5p (Table 2). Indeed, in hepatocellular carcinoma, drug-resistant cells deliver miR-32-5p-containing exosomes inducing resistance into sensitive counterparts by modulating angiogenesis and epithelial–mesenchymal transition [29]. miRNAs also target TP53 [142], another crucial player in cell cycle arrest and apoptosis [36]. Alterations of its expression and function cause resistance to canonical anticancer drugs. In prostate cancer cells, vesicles induce resistance to cisplatin, docetaxel, and doxorubicin through the action of miR-27a, which targets p53 mRNA (Table 2) [36].
Another cell process enhanced in resistant cancer cells is autophagy [143]. The activation of autophagy in response to environmental stress, including that deriving from chemotherapeutic agents, helps cell survival and is implicated in the development of drug resistance [144]. For instance, vesicles derived from hepatitis B virus (HBV)-associated liver cancer cells induce oxaliplatin resistance by activating chaperone-mediated autophagy [103]. In the case of non-small-cell lung cancer, vesicles can mediate chemoresistance by delivering miR-425-3p, whose transcription is upregulated by cisplatin-induced c-Myc. Vesicular miR-425-3p confers chemoresistance through the activation of autophagy targeting AKT1 [14] (Table 2). Moreover, glioblastoma-derived stem cells secrete exosomes that enhance chemoresistance; indeed, these vesicles contain programmed death-ligand 1 (PD-L1), which activates the AMP-activated protein kinase (AMPK)/Unc-51-like kinase 1 (ULK1) pathway mediating autophagy activation that, in turn, results in the increased TMZ-resistance in glioblastoma cells (Table 2) [68]. Vesicular circulating plasmacytoma variant translocation 1 (PVT1) was found to facilitate drug resistance in gastric cancer cells; it induces autophagy activation by modulating the miR-30a-5p, whose target is YAP1, a factor involved in the transcriptional modulation of several genes related to cell proliferation and apoptosis suppression [50].
Finally, vesicles can promote tumor cell survival by triggering DNA repair [145]. Indeed, many anticancer agents target cancer cells, inducing DNA lesions [146,147]. Vesicles that contain in their lumen a long noncoding RNA named SBF2 antisense RNA1 (lncRNA SBF2-AS1) are secreted by glioblastoma cells resistant to temozolomide (Table 2). SBF2-AS1 acts on miR-151a that normally represses X-ray repair cross-complementing 4 (XRCC4) [42,148]. Consequently, vesicles from temozolomide-resistant glioblastoma cells spread a resistant phenotype, delivering high levels of SBF2-AS1 to sensitive cells, which in turn deregulates XRCC4 and enhances the DNA double-strand break repair process [42,148]. Vesicles from reactive astrocytes spread temozolomide resistance by delivering O6-alkylguanine DNA alkyltransferase (MGMT) mRNA to glioma cells. Indeed, MGMT plays a crucial role in repairing DNA damage induced by temozolomide [51].

4. Cancer Stem Cell Derived Vesicles and Chemoresistance

Tumors consist of a heterogeneous population of stromal cells, immune cells, fibroblasts, and cancer stem cells (CSCs) [149]. These are characterized by self-renewal capacity, upregulation of drug efflux pumps, increased DNA repair, and dormancy [150]. As a consequence of the above-mentioned features, CSCs are resistant to standard therapies such as anticancer drug treatments and radiotherapy [151]. Several studies individuate extracellular vesicles as the carrier through which CSCs transfer active molecules to non-CSCs for drug resistance. It was demonstrated that CSCs isolated from gemcitabine-resistant pancreatic cancer release vesicles that can stimulate the expression of drug-resistance-related proteins, such as P-gp, Y-box binding protein 1, and breast cancer resistance protein, in a drug-sensitive pancreatic cancer cell line [104]. Moreover, these vesicles are enriched in miR-210 [104], which can inhibit drug-induced apoptosis and increase the phosphorylation of ribosomal protein S6 kinase beta-1, a downstream target of mTOR [152]. Another miRNA associated with chemoresistant phenotype is miR-92a-3p (Table 2).
miR-92a-3p, embedded into vesicles secreted by cancer-associated fibroblasts, induces drug resistance in colorectal cancer cells, promoting cell stemness phenotype and epithelial–mesenchymal transition (EMT) [31]. Indeed, miR-92a-3p targets F-box and WD repeat domain-containing 7 (FBXW7) and modulator of apoptosis protein 1 (MOAP1), which are involved in the modulation of mTOR and apoptosis, respectively (Table 2) [31,153]. Cancer-associated fibroblasts exosomes can also transfer IL-6, activin-A, and granulocyte colony-stimulating factor (G-CSF), inducing gene expression changes with consequent activation of stemness-associated pathways and methotrexate resistance in lung carcinoma cells [19]. Similarly, exosome Wnt induces resistance to 5-fluorouracil via reprogramming differentiated colorectal cancer cells [63] (Table 2).

5. Vesicle-Mediated Resistance to Immunotherapies

Cancers have evolved several strategies to evade immune surveillance; one of these consists of modulating the tumor microenvironment by inhibiting immune response or inducing immune suppressor cells via exosome secretion [154]. Cancer-derived exosomes can impair lymphocyte response, inhibit monocyte differentiation, induce apoptosis in activated T lymphocytes, and downmodulate the cytolytic activity of natural killer (NK) cells [138]. Moreover, vesicles also play a role in immunotherapy resistance. Indeed, these vesicles might act as a decoy target for anticancer immunotherapies [3,155,156]. Vesicles derived from certain tumors can directly bind and neutralize, at least partially, antibody-based drugs. Trastuzumab is a humanized monoclonal antibody that is widely used to treat HER2+ breast cancer [157] and targets the extracellular domain of HER2. HER2+ breast cancer cells release vesicles expressing this receptor on the surface and competing with that of cancer cells in the binding of trastuzumab, causing a reduction in its bioavailability and efficacy (Table 2) [58]. Moreover, it was recently suggested that a long noncoding RNA named AFAP1-AS1 plays a critical role in establishing trastuzumab resistance [44]. Vesicles mediated the AFAP1-AS1 transfer from trastuzumab-resistant cells to sensitive cells, disseminating drug resistance [44]. The mechanism by which AFAP1-AS1 induces chemoresistance is related to its ability to upregulate HER-2 protein expression through associating with A+U rich RNA binding factor 1 (AUF1) (Table 2) [44]. Rituximab is another antibody used to treat cancer [158] that directly interacts with a vesicle cargo protein.
This chimeric antibody, used in lymphoma therapy, recognizes the cell surface CD20 antigen and induces apoptosis, cytolysis, and complement-dependent cell cytotoxicity (Table 2) [67]. Vesicles isolated from lymphoma specimens and aggressive B-cell lines harbor, at their membrane, a high amount of CD20 that can bind to rituximab, lowering the drug efficacy [67]. The mechanism by which vesicles reduce the efficacy of rituximab is enhanced by the action of ABCA3, as described in Section 2.1 [67].

6. Extracellular Vesicles as a Tool to Monitor Chemoresistance

The resistance to anticancer agents remains the leading cause of treatment failure for many oncological patients [3,4,5]. Thus, developing a panel of biomarkers is necessary to identify chemotherapy-resistant patients. As described in the above paragraphs, emerging evidence has revealed the correlation between the onset of chemoresistance and some vesicular components. These data are mainly obtained using vesicles isolated from cancer cell cultures. However, in the last year, a growing number of studies also include the analysis of liquid biopsies from patients to obtain a preliminary in vivo validation [159,160].
In this regard, some examples of lung, prostate, and breast cancers will be described. Janpipatkul et al. analyzed the vesicular miRNA profile of eight patients affected by non-small-cell lung cancer (NSCLS) [161]. The patients were diagnosed with advanced NSCLC with EGFR mutations and started treatment with osimertinib. This drug is a third-generation inhibitor targeting the epidermal growth factor receptor tyrosine kinase (EGFR); it is used to treat a subgroup of NSCLS patients harboring the T790M-EGFR mutation [161]. It is reported that resistance could appear after 9–13 months of treatment [162]. The analysis of vesicular miRNAs isolated from blood samples indicated that miR-323-3p, miR-1468-3p, miR-5189-5p, and miR-6513-5p are suitable candidate biomarkers for the discrimination of osimertinib-resistant from osimertinib-sensitive NSCLC patients [161]. In another study, blood sampling was performed before Pt-based chemotherapy administration and repeated after the occurrence of chemoresistance in 19 NSCLC patients [14]. The analysis of vesicular miRNAs showed that miR-425-3p is abundantly present and might represent a potential biomarker for identifying cisplatin resistance in NSCLC patients [14]. The same miR-425-3p was analyzed in another study that involved 170 patients (76 platinum-resistant and 94 platinum-sensitive). In this case, after collecting serum samples of lung cancer, the authors concluded that miR-425-3p is a good candidate for predicting the clinical response to Pt-based chemotherapy [153]. Furthermore, miR-222-3p significantly correlated with the patient response to chemotherapy; indeed, a study involving 50 patients with lung cancer (NSCLC) demonstrated that high levels of miR-222-3p in serum seem to be predictive of a negative response to gemcitabine treatment [16].
The first-line treatment for prostate cancer, the most commonly diagnosed malignancy in men, is androgen deprivation therapy (ADT); however, about 50% of patients become resistant [163]. In the case of resistance, one of the possible therapeutic interventions is docetaxel, but most patients also acquire docetaxel resistance [164]. A recent study demonstrated that a high copy number of the variant isoform CD44v8-10 mRNA in vesicles is correlated with docetaxel resistance; the study was conducted on blood samples from three groups of patients: controls (n = 15), prostate cancer patients that had not previously received docetaxel therapy (n = 50), and patients with docetaxel-resistant prostate cancer (n = 10). On the contrary, the serum exosomal mRNA of the CD44 standard isoform was not significantly different among the three groups [163]. In the same context, the P-gp level could also serve as a marker for docetaxel resistance in prostate cancer; indeed, in blood exosomes from six docetaxel-resistant patients, the P-gp level was higher than that in patients that had not previously received docetaxel therapy [62]. Another possible strategy to treat ADT-resistant patients is the use of androgen signaling-targeted therapies such as abiraterone and enzalutamide [165]. A study enrolling 36 patients demonstrated that abiraterone and enzalutamide resistance can be predicted by measuring the level of androgen receptor splice variant 7 RNA in plasma-derived vesicles [166]. Moreover, the currently adopted criteria for diagnosing prostate cancer include the use of the prostate-specific antigen (PSA), which showed a low specificity for prostate cancer. This results in an increase in unnecessary biopsies as part of surveillance programs [167]. On the other hand, due to the heterogeneity of primary prostate cancer, tumor biopsy may not necessarily detect the true characteristics of a tumor [168,169]. In light of this, extracellular vesicles collected from body fluids such as urine more likely reflect the current state of cancer cells from which they originated [169]. McKiernan et al. designed and validated a predictive urine-based extracellular vesicle test for prostate cancer using a cohort of 503 men. The amount of exosomal mRNA of prostate cancer antigen 3 (PCA3), ETS-related gene (ERG), and SAM pointed domain-containing Ets transcription factor (SPDEF) was determined for each patient. The results highlight that this test is predictive of high-grade prostate cancer and may contribute to reducing unnecessary biopsies [167,170].
While prostate cancer is the most commonly diagnosed cancer in men, breast cancer is the second leading cause of women’s death [171]. The human epidermal growth factor receptor-2 (HER2) is overexpressed in many breast cancer patients and is the target of the trastuzumab, as described in Section 5. However, a consistent number of patients became resistant to this therapy within 1 year of exposure [172,173,174]. A study showed that vesicular miR-1246 and miR-155 were upregulated in four trastuzumab-resistant patients compared with four patients that responded to therapy [172]. Another study analyzed the expression level of serum vesicular lncRNA-SNHG14 from 38 responding patients and 34 non-responding to trastuzumab therapy, revealing that the lncRNA was upregulated in patients with resistance [43]. It has to be highlighted that trastuzumab is not the only treatment available for breast cancer; indeed, anthracycline and taxane are frequently used in breast cancer patients as neoadjuvant chemotherapies to decrease tumor size and prevent metastasis [175].
However, not all patients respond to the treatment due to intrinsic or acquired resistance [176]. Yang et al. analyzed the expression of the glutathione S-transferase P1 (GSTP1) in exosomes from 30 patients treated with anthracycline/taxane-based neoadjuvant chemotherapy. Results suggested that GSTP1 was significantly higher in the lumen of vesicles from the 14 patients who did not respond to the therapy than in responsive patients [175]. Another study on 131 patients with breast cancer demonstrated that the level of transient receptor potential channel TRPC5 in the membrane of vesicles is a promising candidate as a noninvasive chemoresistance marker [98]. Ning et al. analyzed exosomes isolated from the blood of 93 patients with breast cancer and discovered that the vesicular ubiquitin carboxyl-terminal hydrolase-L1 (UCH-L1) is a useful biomarker for detecting chemoresistance in breast cancer [54].
Similar studies have also been conducted on other cancer types, showing that extracellular vesicles can be used to monitor chemotherapy efficacy [41,42,177,178,179]. In the case of colorectal cancer (CRC), serum samples from patients have been collected and analyzed [180]; in particular, extracellular vesicles containing the lncRNA UCA1 have been identified as responsible for cetuximab resistance phenotype through degradation of miR-204-5p [180]. In the case of ovarian cancer, serum samples were found to contain extracellular vesicles carrying the circular RNA circFoxp1 [177]. This molecule is responsible for cisplatin resistance by targeting miRNAs miR-22 and miR-150-3p [177]. The sorafenib resistance of hepatocellular carcinoma (HCC) has been linked to extracellular vesicles found in the serum of patients containing a lower amount of miR-744 [178]. In the case of renal cell carcinoma (RCC), the resistance to sunitinib treatment is ascribed to an lncRNA, named lncRNA activated in RCC with sunitinib resistance (lncARSR), which binds to miR-34/miR-449 [41]. Glioblastoma is one of the most aggressive tumors, and TMZ is the first-line chemotherapy; however, chemoresistance to TMZ is a common occurrence in glioblastoma and is mediated by miR-1587 and miR-151a contained in serum vesicles [179]. Another study conducted on glioblastoma samples highlighted the miR-4315 released by lymphocytes as responsible for resistance to the anti-PD-1 therapy [42]. In the case of chronic myeloid leukemia (CML), the resistance to imatinib is mediated by miR-365 released in extracellular vesicles [181]. In all the mentioned studies, the extracellular vesicles produced by resistant cancer cells can induce the resistant phenotype in sensitive recipient cells [41,42,177,178,179]. Therefore, circulating serum vesicles can be considered a promising tool for identifying the cancer stage and responsiveness to treatment.

7. Conclusions

Extracellular vesicles represent a highly attractive source of biomarkers because these vesicles can be easily collected from different body fluids by liquid biopsy. This is greatly relevant in terms of the life quality of cancer patients because it is possible to monitor cancer treatment response via a noninvasive procedure. Compared to other types of molecules that can be isolated from body fluids, such as circulating miRNA or circulating tumor DNA, extracellular vesicles are highly preferable because their membrane protects cargo(s) from degradation (Figure 1). Moreover, vesicle analysis offers the opportunity to isolate multiple biomarkers simultaneously. Indeed, comparing a panel of markers may give more accurate results or information to predict therapeutic response.
However, still, some limitations exist: an aspect to consider is the type of body fluid used for collecting extracellular vesicles. Urine is a suitable source from which to isolate vesicles for detecting biomarkers of the urinary system such as those signifying prostate, bladder, and kidney cancers. Another body fluid that is used in the detection of biomarkers for detecting oral and lung cancer is saliva [182]. Saliva is the most proximal body fluid in oral cancer and is easily accessible in a noninvasive manner [183]. An example of the potential use of saliva as a source of biomarkers to predict oral cancer is reported by Gai et al. In this study, miR-302b-3p and miR-517b-3p were found selectively enriched in salivary EVs from 16 oral squamous cell carcinoma patients compared to those from 6 healthy controls [184]. Moreover, He et al. showed that the level of miR-24-3p from the salivary vesicles of 45 OSCC patients is significantly higher compared to that of 10 normal controls [185]. Another example is represented by the salivary vesicle GOLM1-NAA35 chimeric RNA, which was proposed as a noninvasive biomarker candidate for reliable assessment of therapeutic response, recurrence, and early detection of esophageal squamous cell carcinoma [186].
However, for other cancer types, blood is the source of choice for vesicle isolation. In this respect, it has to be stressed that blood contains a large number of proteins that can contaminate exosome preparation, making the detection of poorly expressed biomarkers difficult.
Other prerequisites for developing a functional clinical test are cheapness and easy execution. Currently, the exosome isolation methods are time-consuming or expensive. Nevertheless, the feasibility of using extracellular vesicles biomarkers in precision medicine is demonstrated by the fact that in 2019, the EPI test, the first EV-based Clinical Laboratory Improvement Amendments (CLIA)-validated and clinically available test, received the FDA breakthrough design designation for fast-tracked approval process. It is a noninvasive urine exosome-based diagnostic test that measures the RNA of prostate cancer antigen 3 (PCA3), ETS-related gene (ERG), and SAM pointed domain-containing Ets transcription factor (SPDEF). This test can guide physicians in determining the need for a prostate biopsy in patients 50 years of age or older with a prostate-specific antigen (PSA) in the grey zone [187].
Moreover, as extracellular vesicles naturally carry bioactive molecules between cells, some studies suggest that these may serve to deliver drugs or RNAs in the context of cancer therapy or to reverse drug resistance [140,188]. Indeed, extracellular vesicles may offer some advantages as delivery carriers, such as biocompatibility, non-cytotoxicity, and low immunogenicity [140,189,190]. In this light, Liang et al. showed that the administration of engineered extracellular vesicles loaded with the 5-fluorouracil and the miR-21 inhibitor oligonucleotide (miR-21i) to resistant colon cancer cells effectively reverses drug resistance [188,191]. Therefore, even if the way is still long, all the mentioned findings strongly support the development of a therapeutic and diagnostic era based on exosomes.

Author Contributions

Conceptualization, L.C.; methodology, L.C.; software, L.C.; validation, L.C., M.S.; formal analysis, L.C., M.S.; investigation, L.C., M.S.; resources, L.C., M.S.; data curation, L.C., M.S.; writing—original draft preparation, L.C., M.S.; writing—review and editing, L.C., M.S.; visualization, L.C., M.S.; supervision, L.C.; project administration, L.C., M.S.; funding acquisition, L.C., M.S. All authors have read and agreed to the published version of the manuscript.

Funding

The work was supported by University of Calabria, progetti ‘ex 60%’ 2021 to M.S. L.C., post-doctoral fellow, was funded by Project STAR 2, PIR01_0008, Ministero dell’Università e Ricerca/Italian Ministry University and Research, Laboratorio LPCB, STAR IR, Via Tito Flavio, Università della Calabria, 87036 Rende (CS), Italy.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Pucci, C.; Martinelli, C.; Ciofani, G. Innovative approaches for cancer treatment: Current perspectives and new challenges. Ecancermedicalscience 2019, 13, 961. [Google Scholar] [CrossRef] [PubMed]
  3. Mostafazadeh, M.; Samadi, N.; Kahroba, H.; Baradaran, B.; Haiaty, S.; Nouri, M. Potential roles and prognostic significance of exosomes in cancer drug resistance. Cell Biosci. 2021, 11, 1–15. [Google Scholar] [CrossRef] [PubMed]
  4. Vasan, N.; Baselga, J.; Hyman, D.M. A view on drug resistance in cancer. Nature 2019, 575, 299–309. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Shomar, A.; Barak, O.; Brenner, N. Cancer progression as a learning process. iScience 2022, 25, 103924. [Google Scholar] [CrossRef] [PubMed]
  6. Kelderman, S.; Schumacher, T.N.; Haanen, J.B. Acquired and intrinsic resistance in cancer immunotherapy. Mol. Oncol. 2014, 8, 1132–1139. [Google Scholar] [CrossRef] [PubMed]
  7. Li, J.; Gao, N.; Gao, Z.; Liu, W.; Pang, B.; Dong, X.; Li, Y.; Fan, T. The Emerging Role of Exosomes in Cancer Chemoresistance. Front. Cell Dev. Biol. 2021, 9, 737962. [Google Scholar] [CrossRef]
  8. Bukowski, K.; Kciuk, M.; Kontek, R. Mechanisms of Multidrug Resistance in Cancer Chemotherapy. Int. J. Mol. Sci. 2020, 21, 3233. [Google Scholar] [CrossRef]
  9. Maleki, S.; Jabalee, J.; Garnis, C. The Role of Extracellular Vesicles in Mediating Resistance to Anticancer Therapies. Int. J. Mol. Sci. 2021, 22, 4166. [Google Scholar] [CrossRef]
  10. Console, L.; Scalise, M.; Indiveri, C. Exosomes in inflammation and role as biomarkers. Clin. Chim. Acta 2019, 488, 165–171. [Google Scholar] [CrossRef]
  11. Zhou, B.; Xu, K.; Zheng, X.; Chen, T.; Wang, J.; Song, Y.; Shao, Y.; Zheng, S. Application of exosomes as liquid biopsy in clinical diagnosis. Signal Transduct. Target. Ther. 2020, 5, 144. [Google Scholar] [CrossRef] [PubMed]
  12. Shedden, K.; Xie, X.T.; Chandaroy, P.; Chang, Y.T.; Rosania, G.R. Expulsion of small molecules in vesicles shed by cancer cells: Association with gene expression and chemosensitivity profiles. Cancer Res. 2003, 63, 4331–4337. [Google Scholar] [PubMed]
  13. Tai, Y.L.; Chen, K.C.; Hsieh, J.T.; Shen, T.L. Exosomes in cancer development and clinical applications. Cancer Sci. 2018, 109, 2364–2374. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Ma, Y.; Yuwen, D.; Chen, J.; Zheng, B.; Gao, J.; Fan, M.; Xue, W.; Wang, Y.; Li, W.; Shu, Y.; et al. Exosomal Transfer Of Cisplatin-Induced miR-425-3p Confers Cisplatin Resistance In NSCLC Through Activating Autophagy. Int. J. Nanomed. 2019, 14, 8121–8132. [Google Scholar] [CrossRef] [Green Version]
  15. Qin, X.; Yu, S.; Zhou, L.; Shi, M.; Hu, Y.; Xu, X.; Shen, B.; Liu, S.; Yan, D.; Feng, J. Cisplatin-resistant lung cancer cell-derived exosomes increase cisplatin resistance of recipient cells in exosomal miR-100-5p-dependent manner. Int. J. Nanomed. 2017, 12, 3721–3733. [Google Scholar] [CrossRef] [Green Version]
  16. Wei, F.; Ma, C.; Zhou, T.; Dong, X.; Luo, Q.; Geng, L.; Ding, L.; Zhang, Y.; Zhang, L.; Li, N.; et al. Exosomes derived from gemcitabine-resistant cells transfer malignant phenotypic traits via delivery of miRNA-222-3p. Mol. Cancer 2017, 16, 132. [Google Scholar] [CrossRef] [Green Version]
  17. Liu, X.; Jiang, T.; Li, X.; Zhao, C.; Li, J.; Zhou, F.; Zhang, L.; Zhao, S.; Jia, Y.; Shi, J.; et al. Exosomes transmit T790M mutation-induced resistance in EGFR-mutant NSCLC by activating PI3K/AKT signalling pathway. J. Cell. Mol. Med. 2020, 24, 1529–1540. [Google Scholar] [CrossRef] [Green Version]
  18. Zhang, Y.; Li, M.; Hu, C. Exosomal transfer of miR-214 mediates gefitinib resistance in non-small cell lung cancer. Biochem. Biophys. Res. Commun. 2018, 507, 457–464. [Google Scholar] [CrossRef]
  19. Rodrigues, C.F.D.; Serrano, E.; Patricio, M.I.; Val, M.M.; Albuquerque, P.; Fonseca, J.; Gomes, C.M.F.; Abrunhosa, A.J.; Paiva, A.; Carvalho, L.; et al. Stroma-derived IL-6, G-CSF and Activin-A mediated dedifferentiation of lung carcinoma cells into cancer stem cells. Sci. Rep. 2018, 8, 11573. [Google Scholar] [CrossRef]
  20. 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] [Green Version]
  21. Liu, X.; Lu, Y.; Xu, Y.; Hou, S.; Huang, J.; Wang, B.; Zhao, J.; Xia, S.; Fan, S.; Yu, X.; et al. Exosomal transfer of miR-501 confers doxorubicin resistance and tumorigenesis via targeting of BLID in gastric cancer. Cancer Lett. 2019, 459, 122–134. [Google Scholar] [CrossRef] [PubMed]
  22. Santos, J.C.; Lima, N.D.S.; Sarian, L.O.; Matheu, A.; Ribeiro, M.L.; Derchain, S.F.M. Exosome-mediated breast cancer chemoresistance via miR-155 transfer. Sci. Rep. 2018, 8, 829. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Chen, W.X.; Cai, Y.Q.; Lv, M.M.; Chen, L.; Zhong, S.L.; Ma, T.F.; Zhao, J.H.; Tang, J.H. Exosomes from docetaxel-resistant breast cancer cells alter chemosensitivity by delivering microRNAs. Tumour Biol. 2014, 35, 9649–9659. [Google Scholar] [CrossRef]
  24. Li, X.J.; Ren, Z.J.; Tang, J.H.; Yu, Q. Exosomal MicroRNA MiR-1246 Promotes Cell Proliferation, Invasion and Drug Resistance by Targeting CCNG2 in Breast Cancer. Cell Physiol. Biochem. 2017, 44, 1741–1748. [Google Scholar] [CrossRef]
  25. Cornell, L.; Wander, S.A.; Visal, T.; Wagle, N.; Shapiro, G.I. MicroRNA-Mediated Suppression of the TGF-beta Pathway Confers Transmissible and Reversible CDK4/6 Inhibitor Resistance. Cell Rep. 2019, 26, 2667–2680.e7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Qin, X.; Guo, H.; Wang, X.; Zhu, X.; Yan, M.; Wang, X.; Xu, Q.; Shi, J.; Lu, E.; Chen, W.; et al. Exosomal miR-196a derived from cancer-associated fibroblasts confers cisplatin resistance in head and neck cancer through targeting CDKN1B and ING5. Genome Biol. 2019, 20, 12. [Google Scholar] [CrossRef] [PubMed]
  27. Liu, T.; Chen, G.; Sun, D.; Lei, M.; Li, Y.; Zhou, C.; Li, X.; Xue, W.; Wang, H.; Liu, C.; et al. Exosomes containing miR-21 transfer the characteristic of cisplatin resistance by targeting PTEN and PDCD4 in oral squamous cell carcinoma. Acta Biochim. Biophys. Sin. 2017, 49, 808–816. [Google Scholar] [CrossRef] [Green Version]
  28. Binenbaum, Y.; Fridman, E.; Yaari, Z.; Milman, N.; Schroeder, A.; Ben David, G.; Shlomi, T.; Gil, Z. Transfer of miRNA in Macrophage-Derived Exosomes Induces Drug Resistance in Pancreatic Adenocarcinoma. Cancer Res. 2018, 78, 5287–5299. [Google Scholar] [CrossRef] [Green Version]
  29. Fu, X.; Liu, M.; Qu, S.; Ma, J.; Zhang, Y.; Shi, T.; Wen, H.; Yang, Y.; Wang, S.; Wang, J.; et al. Exosomal microRNA-32-5p induces multidrug resistance in hepatocellular carcinoma via the PI3K/Akt pathway. J. Exp. Clin. Cancer Res. 2018, 37, 52. [Google Scholar] [CrossRef] [Green Version]
  30. Akao, Y.; Khoo, F.; Kumazaki, M.; Shinohara, H.; Miki, K.; Yamada, N. Extracellular disposal of tumor-suppressor miRs-145 and -34a via microvesicles and 5-FU resistance of human colon cancer cells. Int. J. Mol. Sci. 2014, 15, 1392–1401. [Google Scholar] [CrossRef] [Green Version]
  31. Hu, J.L.; Wang, W.; Lan, X.L.; Zeng, Z.C.; Liang, Y.S.; Yan, Y.R.; Song, F.Y.; Wang, F.F.; Zhu, X.H.; Liao, W.J.; et al. CAFs secreted exosomes promote metastasis and chemotherapy resistance by enhancing cell stemness and epithelial-mesenchymal transition in colorectal cancer. Mol. Cancer 2019, 18, 91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Zhu, X.; Shen, H.; Yin, X.; Yang, M.; Wei, H.; Chen, Q.; Feng, F.; Liu, Y.; Xu, W.; Li, Y. Macrophages derived exosomes deliver miR-223 to epithelial ovarian cancer cells to elicit a chemoresistant phenotype. J. Exp. Clin. Cancer Res. 2019, 38, 81. [Google Scholar] [CrossRef] [PubMed]
  33. Weiner-Gorzel, K.; Dempsey, E.; Milewska, M.; McGoldrick, A.; Toh, V.; Walsh, A.; Lindsay, S.; Gubbins, L.; Cannon, A.; Sharpe, D.; et al. Overexpression of the microRNA miR-433 promotes resistance to paclitaxel through the induction of cellular senescence in ovarian cancer cells. Cancer Med. 2015, 4, 745–758. [Google Scholar] [CrossRef] [PubMed]
  34. Kanlikilicer, P.; Bayraktar, R.; Denizli, M.; Rashed, M.H.; Ivan, C.; Aslan, B.; Mitra, R.; Karagoz, K.; Bayraktar, E.; Zhang, X.; et al. Corrigendum to ‘Exosomal miRNA confers chemo resistance via targeting Cav1/p-gp/M2-type macrophage axis in ovarian cancer’ [EBioMedicine 38 (2018) 100–112]. EBioMedicine 2020, 52, 102630. [Google Scholar] [CrossRef]
  35. Kunou, S.; Shimada, K.; Takai, M.; Sakamoto, A.; Aoki, T.; Hikita, T.; Kagaya, Y.; Iwamoto, E.; Sanada, M.; Shimada, S.; et al. Exosomes secreted from cancer-associated fibroblasts elicit anti-pyrimidine drug resistance through modulation of its transporter in malignant lymphoma. Oncogene 2021, 40, 3989–4003. [Google Scholar] [CrossRef]
  36. Cao, Z.; Xu, L.; Zhao, S. Exosome-derived miR-27a produced by PSC-27cells contributes to prostate cancer chemoresistance through p53. Biochem. Biophys. Res. Commun. 2019, 515, 345–351. [Google Scholar] [CrossRef]
  37. Chen, X.; Wang, Z.; Tong, F.; Dong, X.; Wu, G.; Zhang, R. lncRNA UCA1 Promotes Gefitinib Resistance as a ceRNA to Target FOSL2 by Sponging miR-143 in Non-small Cell Lung Cancer. Mol. Ther. Nucleic Acids 2020, 19, 643–653. [Google Scholar] [CrossRef]
  38. Li, Z.; Niu, H.; Qin, Q.; Yang, S.; Wang, Q.; Yu, C.; Wei, Z.; Jin, Z.; Wang, X.; Yang, A.; et al. lncRNA UCA1 Mediates Resistance to Cisplatin by Regulating the miR-143/FOSL2-Signaling Pathway in Ovarian Cancer. Mol. Ther. Nucleic Acids 2019, 17, 92–101. [Google Scholar] [CrossRef] [Green Version]
  39. Kang, M.; Ren, M.; Li, Y.; Fu, Y.; Deng, M.; Li, C. Exosome-mediated transfer of lncRNA PART1 induces gefitinib resistance in esophageal squamous cell carcinoma via functioning as a competing endogenous RNA. J. Exp. Clin. Cancer Res. 2018, 37, 171. [Google Scholar] [CrossRef]
  40. Chen, Y.; Liu, L.; Li, J.; Du, Y.; Wang, J.; Liu, J. Effects of long noncoding RNA (linc-VLDLR) existing in extracellular vesicles on the occurrence and multidrug resistance of esophageal cancer cells. Pathol. Res. Pract. 2019, 215, 470–477. [Google Scholar] [CrossRef]
  41. Qu, L.; Ding, J.; Chen, C.; Wu, Z.J.; Liu, B.; Gao, Y.; Chen, W.; Liu, F.; Sun, W.; Li, X.F.; et al. Exosome-Transmitted lncARSR Promotes Sunitinib Resistance in Renal Cancer by Acting as a Competing Endogenous RNA. Cancer Cell 2016, 29, 653–668. [Google Scholar] [CrossRef] [PubMed]
  42. Zhang, Z.; Yin, J.; Lu, C.; Wei, Y.; Zeng, A.; You, Y. Exosomal transfer of long non-coding RNA SBF2-AS1 enhances chemoresistance to temozolomide in glioblastoma. J. Exp. Clin. Cancer Res. 2019, 38, 166. [Google Scholar] [CrossRef] [PubMed]
  43. Dong, H.; Wang, W.; Chen, R.; Zhang, Y.; Zou, K.; Ye, M.; He, X.; Zhang, F.; Han, J. Exosome-mediated transfer of lncRNASNHG14 promotes trastuzumab chemoresistance in breast cancer. Int. J. Oncol. 2018, 53, 1013–1026. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Han, M.; Gu, Y.; Lu, P.; Li, J.; Cao, H.; Li, X.; Qian, X.; Yu, C.; Yang, Y.; Yang, X.; et al. Exosome-mediated lncRNA AFAP1-AS1 promotes trastuzumab resistance through binding with AUF1 and activating ERBB2 translation. Mol. Cancer 2020, 19, 26. [Google Scholar] [CrossRef] [Green Version]
  45. Ren, J.; Ding, L.; Zhang, D.; Shi, G.; Xu, Q.; Shen, S.; Wang, Y.; Wang, T.; Hou, Y. Carcinoma-associated fibroblasts promote the stemness and chemoresistance of colorectal cancer by transferring exosomal lncRNA H19. Theranostics 2018, 8, 3932–3948. [Google Scholar] [CrossRef]
  46. Deng, X.; Ruan, H.; Zhang, X.; Xu, X.; Zhu, Y.; Peng, H.; Zhang, X.; Kong, F.; Guan, M. Long noncoding RNA CCAL transferred from fibroblasts by exosomes promotes chemoresistance of colorectal cancer cells. Int. J. Cancer 2020, 146, 1700–1716. [Google Scholar] [CrossRef]
  47. Luo, G.; Zhang, Y.; Wu, Z.; Zhang, L.; Liang, C.; Chen, X. Exosomal LINC00355 derived from cancer-associated fibroblasts promotes bladder cancer cell resistance to cisplatin by regulating miR-34b-5p/ABCB1 axis. Acta Biochim. Biophys. Sin. 2021, 53, 558–566. [Google Scholar] [CrossRef]
  48. Wang, X.; Zhang, H.; Yang, H.; Bai, M.; Ning, T.; Deng, T.; Liu, R.; Fan, Q.; Zhu, K.; Li, J.; et al. Exosome-delivered circRNA promotes glycolysis to induce chemoresistance through the miR-122-PKM2 axis in colorectal cancer. Mol. Oncol. 2020, 14, 539–555. [Google Scholar] [CrossRef]
  49. Shao, N.; Song, L.; Sun, X. Exosomal circ_PIP5K1A regulates the progression of non-small cell lung cancer and cisplatin sensitivity by miR-101/ABCC1 axis. Mol. Cell. Biochem. 2021, 476, 2253–2267. [Google Scholar] [CrossRef]
  50. Yao, W.; Guo, P.; Mu, Q.; Wang, Y. Exosome-Derived Circ-PVT1 Contributes to Cisplatin Resistance by Regulating Autophagy, Invasion, and Apoptosis Via miR-30a-5p/YAP1 Axis in Gastric Cancer Cells. Cancer Biother. Radiopharm. 2021, 36, 347–359. [Google Scholar] [CrossRef]
  51. Yu, T.; Wang, X.; Zhi, T.; Zhang, J.; Wang, Y.; Nie, E.; Zhou, F.; You, Y.; Liu, N. Delivery of MGMT mRNA to glioma cells by reactive astrocyte-derived exosomes confers a temozolomide resistance phenotype. Cancer Lett. 2018, 433, 210–220. [Google Scholar] [CrossRef] [PubMed]
  52. Lobb, R.J.; van Amerongen, R.; Wiegmans, A.; Ham, S.; Larsen, J.E.; Moller, A. Exosomes derived from mesenchymal non-small cell lung cancer cells promote chemoresistance. Int. J. Cancer 2017, 141, 614–620. [Google Scholar] [CrossRef] [PubMed]
  53. Torreggiani, E.; Roncuzzi, L.; Perut, F.; Zini, N.; Baldini, N. Multimodal transfer of MDR by exosomes in human osteosarcoma. Int. J. Oncol. 2016, 49, 189–196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Ning, K.; Wang, T.; Sun, X.; Zhang, P.; Chen, Y.; Jin, J.; Hua, D. UCH-L1-containing exosomes mediate chemotherapeutic resistance transfer in breast cancer. J. Surg. Oncol. 2017, 115, 932–940. [Google Scholar] [CrossRef]
  55. Lv, M.M.; Zhu, X.Y.; Chen, W.X.; Zhong, S.L.; Hu, Q.; Ma, T.F.; Zhang, J.; Chen, L.; Tang, J.H.; Zhao, J.H. Exosomes mediate drug resistance transfer in MCF-7 breast cancer cells and a probable mechanism is delivery of P-glycoprotein. Tumour Biol. 2014, 35, 10773–10779. [Google Scholar] [CrossRef]
  56. Ma, X.; Chen, Z.; Hua, D.; He, D.; Wang, L.; Zhang, P.; Wang, J.; Cai, Y.; Gao, C.; Zhang, X.; et al. Essential role for TrpC5-containing extracellular vesicles in breast cancer with chemotherapeutic resistance. Proc. Natl. Acad. Sci. USA 2014, 111, 6389–6394. [Google Scholar] [CrossRef] [Green Version]
  57. Kreger, B.T.; Johansen, E.R.; Cerione, R.A.; Antonyak, M.A. The Enrichment of Survivin in Exosomes from Breast Cancer Cells Treated with Paclitaxel Promotes Cell Survival and Chemoresistance. Cancers 2016, 8, 111. [Google Scholar] [CrossRef] [Green Version]
  58. Ciravolo, V.; Huber, V.; Ghedini, G.C.; Venturelli, E.; Bianchi, F.; Campiglio, M.; Morelli, D.; Villa, A.; Della Mina, P.; Menard, S.; et al. Potential role of HER2-overexpressing exosomes in countering trastuzumab-based therapy. J. Cell. Physiol. 2012, 227, 658–667. [Google Scholar] [CrossRef]
  59. Cao, Y.L.; Zhuang, T.; Xing, B.H.; Li, N.; Li, Q. Exosomal DNMT1 mediates cisplatin resistance in ovarian cancer. Cell Biochem. Funct. 2017, 35, 296–303. [Google Scholar] [CrossRef]
  60. Zhang, F.F.; Zhu, Y.F.; Zhao, Q.N.; Yang, D.T.; Dong, Y.P.; Jiang, L.; Xing, W.X.; Li, X.Y.; Xing, H.; Shi, M.; et al. Microvesicles mediate transfer of P-glycoprotein to paclitaxel-sensitive A2780 human ovarian cancer cells, conferring paclitaxel-resistance. Eur. J. Pharmacol. 2014, 738, 83–90. [Google Scholar] [CrossRef]
  61. 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] [Green Version]
  62. Kato, T.; Mizutani, K.; Kameyama, K.; Kawakami, K.; Fujita, Y.; Nakane, K.; Kanimoto, Y.; Ehara, H.; Ito, H.; Seishima, M.; et al. Serum exosomal P-glycoprotein is a potential marker to diagnose docetaxel resistance and select a taxoid for patients with prostate cancer. Urol. Oncol. 2015, 33, 385.e15. [Google Scholar] [CrossRef] [PubMed]
  63. Hu, Y.B.; Yan, C.; Mu, L.; Mi, Y.L.; Zhao, H.; Hu, H.; Li, X.L.; Tao, D.D.; Wu, Y.Q.; Gong, J.P.; et al. Exosomal Wnt-induced dedifferentiation of colorectal cancer cells contributes to chemotherapy resistance. Oncogene 2019, 38, 1951–1965. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Zhang, Q.; Liu, R.X.; Chan, K.W.; Hu, J.; Zhang, J.; Wei, L.; Tan, H.; Yang, X.; Liu, H. Exosomal transfer of p-STAT3 promotes acquired 5-FU resistance in colorectal cancer cells. J. Exp. Clin. Cancer Res. 2019, 38, 320. [Google Scholar] [CrossRef]
  65. Fan, J.; Wei, Q.; Koay, E.J.; Liu, Y.; Ning, B.; Bernard, P.W.; Zhang, N.; Han, H.; Katz, M.H.; Zhao, Z.; et al. Chemoresistance Transmission via Exosome-Mediated EphA2 Transfer in Pancreatic Cancer. Theranostics 2018, 8, 5986–5994. [Google Scholar] [CrossRef]
  66. Feng, Y.; Zhong, M.; Tang, Y.; Liu, X.; Liu, Y.; Wang, L.; Zhou, H. The Role and Underlying Mechanism of Exosomal CA1 in Chemotherapy Resistance in Diffuse Large B Cell Lymphoma. Mol. Ther. Nucleic Acids 2020, 21, 452–463. [Google Scholar] [CrossRef]
  67. Aung, T.; Chapuy, B.; Vogel, D.; Wenzel, D.; Oppermann, M.; Lahmann, M.; Weinhage, T.; Menck, K.; Hupfeld, T.; Koch, R.; et al. Exosomal evasion of humoral immunotherapy in aggressive B-cell lymphoma modulated by ATP-binding cassette transporter A3. Proc. Natl. Acad. Sci. USA 2011, 108, 15336–15341. [Google Scholar] [CrossRef] [Green Version]
  68. Zheng, Y.; Liu, L.; Wang, Y.; Xiao, S.; Mai, R.; Zhu, Z.; Cao, Y. Glioblastoma stem cell (GSC)-derived PD-L1-containing exosomes activates AMPK/ULK1 pathway mediated autophagy to increase temozolomide-resistance in glioblastoma. Cell Biosci. 2021, 11, 63. [Google Scholar] [CrossRef]
  69. Thery, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef] [Green Version]
  70. Doyle, L.M.; Wang, M.Z. Overview of Extracellular Vesicles, Their Origin, Composition, Purpose, and Methods for Exosome Isolation and Analysis. Cells 2019, 8, 727. [Google Scholar] [CrossRef] [Green Version]
  71. Lai, J.J.; Chau, Z.L.; Chen, S.Y.; Hill, J.J.; Korpany, K.V.; Liang, N.W.; Lin, L.H.; Lin, Y.H.; Liu, J.K.; Liu, Y.C.; et al. Exosome Processing and Characterization Approaches for Research and Technology Development. Adv. Sci. 2022, e2103222. [Google Scholar] [CrossRef] [PubMed]
  72. Ender, F.; Von Bubnoff, N.; Gieseler, F. Extracellular Vesicles: Subcellular Organelles With the Potential to Spread Cancer Resistance. Anticancer Res. 2019, 39, 3395–3404. [Google Scholar] [CrossRef] [PubMed]
  73. Mikaelian, I.; Scicchitano, M.; Mendes, O.; Thomas, R.A.; Leroy, B.E. Frontiers in preclinical safety biomarkers: MicroRNAs and messenger RNAs. Toxicol. Pathol. 2013, 41, 18–31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Gillies, R.J.; Pilot, C.; Marunaka, Y.; Fais, S. Targeting acidity in cancer and diabetes. Biochim. Biophys. Acta Rev. Cancer 2019, 1871, 273–280. [Google Scholar] [CrossRef] [PubMed]
  75. Pavlova, N.N.; Thompson, C.B. The Emerging Hallmarks of Cancer Metabolism. Cell Metab. 2016, 23, 27–47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Logozzi, M.; Spugnini, E.; Mizzoni, D.; Di Raimo, R.; Fais, S. Extracellular acidity and increased exosome release as key phenotypes of malignant tumors. Cancer Metastasis Rev. 2019, 38, 93–101. [Google Scholar] [CrossRef]
  77. 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] [Green Version]
  78. 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] [Green Version]
  79. Koch, R.; Aung, T.; Vogel, D.; Chapuy, B.; Wenzel, D.; Becker, S.; Sinzig, U.; Venkataramani, V.; von Mach, T.; Jacob, R.; et al. Nuclear Trapping through Inhibition of Exosomal Export by Indomethacin Increases Cytostatic Efficacy of Doxorubicin and Pixantrone. Clin. Cancer Res. 2016, 22, 395–404. [Google Scholar] [CrossRef] [Green Version]
  80. Wang, J.; Yeung, B.Z.; Cui, M.; Peer, C.J.; Lu, Z.; Figg, W.D.; Guillaume Wientjes, M.; Woo, S.; Au, J.L. Exosome is a mechanism of intercellular drug transfer: Application of quantitative pharmacology. J. Control. Release 2017, 268, 147–158. [Google Scholar] [CrossRef]
  81. Yin, J.; Yan, X.; Yao, X.; Zhang, Y.; Shan, Y.; Mao, N.; Yang, Y.; Pan, L. Secretion of annexin A3 from ovarian cancer cells and its association with platinum resistance in ovarian cancer patients. J. Cell. Mol. Med. 2012, 16, 337–348. [Google Scholar] [CrossRef]
  82. Yan, X.; Yin, J.; Yao, H.; Mao, N.; Yang, Y.; Pan, L. Increased expression of annexin A3 is a mechanism of platinum resistance in ovarian cancer. Cancer Res. 2010, 70, 1616–1624. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Srikant, S. Evolutionary history of ATP-binding cassette proteins. FEBS Lett. 2020, 594, 3882–3897. [Google Scholar] [CrossRef] [PubMed]
  84. Furuta, T. Structural dynamics of ABC transporters: Molecular simulation studies. Biochem. Soc. Trans. 2021, 49, 405–414. [Google Scholar] [CrossRef] [PubMed]
  85. Liu, X. SLC Family Transporters. Adv. Exp. Med. Biol. 2019, 1141, 101–202. [Google Scholar] [CrossRef]
  86. Cesar-Razquin, A.; Snijder, B.; Frappier-Brinton, T.; Isserlin, R.; Gyimesi, G.; Bai, X.; Reithmeier, R.A.; Hepworth, D.; Hediger, M.A.; Edwards, A.M.; et al. A Call for Systematic Research on Solute Carriers. Cell 2015, 162, 478–487. [Google Scholar] [CrossRef] [Green Version]
  87. Kuhlbrandt, W. Biology, structure and mechanism of P-type ATPases. Nat. Rev. Mol. Cell Biol. 2004, 5, 282–295. [Google Scholar] [CrossRef]
  88. Dean, M.; Annilo, T. Evolution of the ATP-binding cassette (ABC) transporter superfamily in vertebrates. Annu. Rev. Genom. Hum. Genet. 2005, 6, 123–142. [Google Scholar] [CrossRef]
  89. Holland, I.B. Rise and rise of the ABC transporter families. Res. Microbiol. 2019, 170, 304–320. [Google Scholar] [CrossRef]
  90. DeGorter, M.K.; Xia, C.Q.; Yang, J.J.; Kim, R.B. Drug transporters in drug efficacy and toxicity. Annu. Rev. Pharmacol. Toxicol. 2012, 52, 249–273. [Google Scholar] [CrossRef]
  91. Dean, M.; Rzhetsky, A.; Allikmets, R. The human ATP-binding cassette (ABC) transporter superfamily. Genome Res. 2001, 11, 1156–1166. [Google Scholar] [CrossRef] [PubMed]
  92. Ambudkar, S.V.; Kimchi-Sarfaty, C.; Sauna, Z.E.; Gottesman, M.M. P-glycoprotein: From genomics to mechanism. Oncogene 2003, 22, 7468–7485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Gutmann, D.A.; Ward, A.; Urbatsch, I.L.; Chang, G.; van Veen, H.W. Understanding polyspecificity of multidrug ABC transporters: Closing in on the gaps in ABCB1. Trends Biochem. Sci. 2010, 35, 36–42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Varma, M.V.; Ashokraj, Y.; Dey, C.S.; Panchagnula, R. P-glycoprotein inhibitors and their screening: A perspective from bioavailability enhancement. Pharmacol. Res. 2003, 48, 347–359. [Google Scholar] [CrossRef]
  95. Brambila-Tapia, A.J. MDR1 (ABCB1) polymorphisms: Functional effects and clinical implications. Rev. Investig. Clin. 2013, 65, 445–454. [Google Scholar]
  96. Fu, D.; Arias, I.M. Intracellular trafficking of P-glycoprotein. Int. J. Biochem. Cell Biol. 2012, 44, 461–464. [Google Scholar] [CrossRef] [Green Version]
  97. 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]
  98. Wang, T.; Ning, K.; Lu, T.X.; Sun, X.; Jin, L.; Qi, X.; Jin, J.; Hua, D. Increasing circulating exosomes-carrying TRPC5 predicts chemoresistance in metastatic breast cancer patients. Cancer Sci. 2017, 108, 448–454. [Google Scholar] [CrossRef] [Green Version]
  99. Takahashi, K.; Yan, I.K.; Wood, J.; Haga, H.; Patel, T. Involvement of extracellular vesicle long noncoding RNA (linc-VLDLR) in tumor cell responses to chemotherapy. Mol. Cancer Res. 2014, 12, 1377–1387. [Google Scholar] [CrossRef] [Green Version]
  100. Jia, Z.; Zhu, H.; Sun, H.; Hua, Y.; Zhang, G.; Jiang, J.; Wang, X. Adipose Mesenchymal Stem Cell-Derived Exosomal microRNA-1236 Reduces Resistance of Breast Cancer Cells to Cisplatin by Suppressing SLC9A1 and the Wnt/beta-Catenin Signaling. Cancer Manag. Res. 2020, 12, 8733–8744. [Google Scholar] [CrossRef]
  101. Liu, N.; Zhang, J.; Yin, M.; Liu, H.; Zhang, X.; Li, J.; Yan, B.; Guo, Y.; Zhou, J.; Tao, J.; et al. Inhibition of xCT suppresses the efficacy of anti-PD-1/L1 melanoma treatment through exosomal PD-L1-induced macrophage M2 polarization. Mol. Ther. 2021, 29, 2321–2334. [Google Scholar] [CrossRef] [PubMed]
  102. Wang, J.; Hendrix, A.; Hernot, S.; Lemaire, M.; De Bruyne, E.; Van Valckenborgh, E.; Lahoutte, T.; De Wever, O.; Vanderkerken, K.; Menu, E. Bone marrow stromal cell-derived exosomes as communicators in drug resistance in multiple myeloma cells. Blood 2014, 124, 555–566. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Liu, D.X.; Li, P.P.; Guo, J.P.; Li, L.L.; Guo, B.; Jiao, H.B.; Wu, J.H.; Chen, J.M. Exosomes derived from HBV-associated liver cancer promote chemoresistance by upregulating chaperone-mediated autophagy. Oncol. Lett. 2019, 17, 323–331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Yang, Z.; Zhao, N.; Cui, J.; Wu, H.; Xiong, J.; Peng, T. Exosomes derived from cancer stem cells of gemcitabine-resistant pancreatic cancer cells enhance drug resistance by delivering miR-210. Cell Oncol. 2020, 43, 123–136. [Google Scholar] [CrossRef] [Green Version]
  105. Rong, C.; Zhou, R.; Wan, S.; Su, D.; Wang, S.L.; Hess, J. Ubiquitin Carboxyl-Terminal Hydrolases and Human Malignancies: The Novel Prognostic and Therapeutic Implications for Head and Neck Cancer. Front. Oncol. 2020, 10, 592501. [Google Scholar] [CrossRef]
  106. Leslie, E.M.; Deeley, R.G.; Cole, S.P. Toxicological relevance of the multidrug resistance protein 1, MRP1 (ABCC1) and related transporters. Toxicology 2001, 167, 3–23. [Google Scholar] [CrossRef]
  107. Zhou, S.F.; Wang, L.L.; Di, Y.M.; Xue, C.C.; Duan, W.; Li, C.G.; Li, Y. Substrates and inhibitors of human multidrug resistance associated proteins and the implications in drug development. Curr. Med. Chem. 2008, 15, 1981–2039. [Google Scholar] [CrossRef]
  108. Kusuhara, H.; Sugiyama, Y. ATP-binding cassette, subfamily G (ABCG family). Pflug. Arch. 2007, 453, 735–744. [Google Scholar] [CrossRef] [Green Version]
  109. 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]
  110. Bitter, R.M.; Oh, S.; Deng, Z.; Rahman, S.; Hite, R.K.; Yuan, P. Structure of the Wilson disease copper transporter ATP7B. Sci. Adv. 2022, 8, eabl5508. [Google Scholar] [CrossRef]
  111. Lukanovic, D.; Herzog, M.; Kobal, B.; Cerne, K. The contribution of copper efflux transporters ATP7A and ATP7B to chemoresistance and personalized medicine in ovarian cancer. Biomed. Pharmacother. 2020, 129, 110401. [Google Scholar] [CrossRef] [PubMed]
  112. Hediger, M.A.; Clemencon, B.; Burrier, R.E.; Bruford, E.A. The ABCs of membrane transporters in health and disease (SLC series): Introduction. Mol. Asp. Med. 2013, 34, 95–107. [Google Scholar] [CrossRef] [PubMed]
  113. Lin, L.; Yee, S.W.; Kim, R.B.; Giacomini, K.M. SLC transporters as therapeutic targets: Emerging opportunities. Nat. Rev. Drug Discov. 2015, 14, 543–560. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Geier, E.G.; Chen, E.C.; Webb, A.; Papp, A.C.; Yee, S.W.; Sadee, W.; Giacomini, K.M. Profiling solute carrier transporters in the human blood-brain barrier. Clin. Pharmacol. Ther. 2013, 94, 636–639. [Google Scholar] [CrossRef]
  115. Schlessinger, A.; Yee, S.W.; Sali, A.; Giacomini, K.M. SLC classification: An update. Clin. Pharmacol. Ther. 2013, 94, 19–23. [Google Scholar] [CrossRef] [Green Version]
  116. Giacomini, K.M.; Huang, S.M. Transporters in drug development and clinical pharmacology. Clin. Pharmacol. Ther. 2013, 94, 3–9. [Google Scholar] [CrossRef]
  117. Keerthikumar, S.; Chisanga, D.; Ariyaratne, D.; Al Saffar, H.; Anand, S.; Zhao, K.; Samuel, M.; Pathan, M.; Jois, M.; Chilamkurti, N.; et al. ExoCarta: A Web-Based Compendium of Exosomal Cargo. J. Mol. Biol. 2016, 428, 688–692. [Google Scholar] [CrossRef] [Green Version]
  118. Console, L.; Scalise, M.; Tonazzi, A.; Giangregorio, N.; Indiveri, C. Characterization of Exosomal SLC22A5 (OCTN2) carnitine transporter. Sci. Rep. 2018, 8, 3758. [Google Scholar] [CrossRef]
  119. Naes, S.M.; Ab-Rahim, S.; Mazlan, M.; Abdul Rahman, A. Equilibrative Nucleoside Transporter 2: Properties and Physiological Roles. Biomed. Res. Int. 2020, 2020, 5197626. [Google Scholar] [CrossRef]
  120. Young, J.D. The SLC28 (CNT) and SLC29 (ENT) nucleoside transporter families: A 30-year collaborative odyssey. Biochem. Soc. Trans. 2016, 44, 869–876. [Google Scholar] [CrossRef]
  121. Karimian Pour, N.; Piquette-Miller, M. Endotoxin Modulates the Expression of Renal Drug Transporters in HIV-1 Transgenic Rats. J. Pharm. Pharm. Sci. 2018, 21, 117s–129s. [Google Scholar] [CrossRef] [PubMed]
  122. Marce, S.; Molina-Arcas, M.; Villamor, N.; Casado, F.J.; Campo, E.; Pastor-Anglada, M.; Colomer, D. Expression of human equilibrative nucleoside transporter 1 (hENT1) and its correlation with gemcitabine uptake and cytotoxicity in mantle cell lymphoma. Haematologica 2006, 91, 895–902. [Google Scholar] [PubMed]
  123. Ma, D.; Fang, Q.; Wang, P.; Gao, R.; Wu, W.; Lu, T.; Cao, L.; Hu, X.; Wang, J. Induction of heme oxygenase-1 by Na+-H+ exchanger 1 protein plays a crucial role in imatinib-resistant chronic myeloid leukemia cells. J. Biol. Chem. 2015, 290, 12558–12571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Brett, C.L.; Tukaye, D.N.; Mukherjee, S.; Rao, R. The yeast endosomal Na+K+/H+ exchanger Nhx1 regulates cellular pH to control vesicle trafficking. Mol. Biol. Cell 2005, 16, 1396–1405. [Google Scholar] [CrossRef] [Green Version]
  125. Xu, H.; Ghishan, F.K.; Kiela, P.R. SLC9 Gene Family: Function, Expression, and Regulation. Compr. Physiol. 2018, 8, 555–583. [Google Scholar] [CrossRef]
  126. Fliegel, L. Role of Genetic Mutations of the Na(+)/H(+) Exchanger Isoform 1, in Human Disease and Protein Targeting and Activity. Mol. Cell. Biochem. 2021, 476, 1221–1232. [Google Scholar] [CrossRef]
  127. Li, X.; Fliegel, L. Permissive role of Na(+)/H(+) exchanger isoform 1 in migration and invasion of triple-negative basal-like breast cancer cells. Mol. Cell. Biochem. 2022, 477, 1207–1216. [Google Scholar] [CrossRef]
  128. Bhutia, Y.D.; Babu, E.; Ramachandran, S.; Ganapathy, V. Amino Acid transporters in cancer and their relevance to “glutamine addiction”: Novel targets for the design of a new class of anticancer drugs. Cancer Res. 2015, 75, 1782–1788. [Google Scholar] [CrossRef] [Green Version]
  129. Parker, J.L.; Deme, J.C.; Kolokouris, D.; Kuteyi, G.; Biggin, P.C.; Lea, S.M.; Newstead, S. Molecular basis for redox control by the human cystine/glutamate antiporter system xc(). Nat. Commun. 2021, 12, 7147. [Google Scholar] [CrossRef]
  130. Lewerenz, J.; Hewett, S.J.; Huang, Y.; Lambros, M.; Gout, P.W.; Kalivas, P.W.; Massie, A.; Smolders, I.; Methner, A.; Pergande, M.; et al. The cystine/glutamate antiporter system x(c)(-) in health and disease: From molecular mechanisms to novel therapeutic opportunities. Antioxid. Redox. Signal. 2013, 18, 522–555. [Google Scholar] [CrossRef] [Green Version]
  131. Fotiadis, D.; Kanai, Y.; Palacin, M. The SLC3 and SLC7 families of amino acid transporters. Mol. Asp. Med. 2013, 34, 139–158. [Google Scholar] [CrossRef]
  132. Sato, H.; Tamba, M.; Ishii, T.; Bannai, S. Cloning and expression of a plasma membrane cystine/glutamate exchange transporter composed of two distinct proteins. J. Biol. Chem. 1999, 274, 11455–11458. [Google Scholar] [CrossRef] [Green Version]
  133. Jiang, L.; Kon, N.; Li, T.; Wang, S.J.; Su, T.; Hibshoosh, H.; Baer, R.; Gu, W. Ferroptosis as a p53-mediated activity during tumour suppression. Nature 2015, 520, 57–62. [Google Scholar] [CrossRef] [Green Version]
  134. Stockwell, B.R.; Friedmann Angeli, J.P.; Bayir, H.; Bush, A.I.; Conrad, M.; Dixon, S.J.; Fulda, S.; Gascon, S.; Hatzios, S.K.; Kagan, V.E.; et al. Ferroptosis: A Regulated Cell Death Nexus Linking Metabolism, Redox Biology, and Disease. Cell 2017, 171, 273–285. [Google Scholar] [CrossRef] [Green Version]
  135. Polewski, M.D.; Reveron-Thornton, R.F.; Cherryholmes, G.A.; Marinov, G.K.; Aboody, K.S. SLC7A11 Overexpression in Glioblastoma Is Associated with Increased Cancer Stem Cell-Like Properties. Stem Cells Dev. 2017, 26, 1236–1246. [Google Scholar] [CrossRef]
  136. Okamoto, K.; Saito, Y.; Ueda, H.; Narumi, K.; Furugen, A.; Kobayashi, M. Kinetic analysis of cystine uptake and inhibition pattern of sulfasalazine in A549 cells. Biopharm. Drug Dispos. 2021, 42, 389–392. [Google Scholar] [CrossRef]
  137. Lei, Y.; Li, X.; Huang, Q.; Zheng, X.; Liu, M. Progress and Challenges of Predictive Biomarkers for Immune Checkpoint Blockade. Front. Oncol. 2021, 11, 617335. [Google Scholar] [CrossRef]
  138. Zhong, Y.; Li, H.; Li, P.; Chen, Y.; Zhang, M.; Yuan, Z.; Zhang, Y.; Xu, Z.; Luo, G.; Fang, Y.; et al. Exosomes: A New Pathway for Cancer Drug Resistance. Front. Oncol. 2021, 11, 743556. [Google Scholar] [CrossRef]
  139. Neophytou, C.M.; Trougakos, I.P.; Erin, N.; Papageorgis, P. Apoptosis Deregulation and the Development of Cancer Multi-Drug Resistance. Cancers 2021, 13, 4363. [Google Scholar] [CrossRef]
  140. Steinbichler, T.B.; Dudas, J.; Skvortsov, S.; Ganswindt, U.; Riechelmann, H.; Skvortsova, I.I. Therapy resistance mediated by exosomes. Mol. Cancer 2019, 18, 58. [Google Scholar] [CrossRef]
  141. Boing, A.N.; Stap, J.; Hau, C.M.; Afink, G.B.; Ris-Stalpers, C.; Reits, E.A.; Sturk, A.; van Noorden, C.J.; Nieuwland, R. Active caspase-3 is removed from cells by release of caspase-3-enriched vesicles. Biochim. Biophys. Acta 2013, 1833, 1844–1852. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Chen, S.; Thorne, R.F.; Zhang, X.D.; Wu, M.; Liu, L. Non-coding RNAs, guardians of the p53 galaxy. Semin. Cancer Biol. 2021, 75, 72–83. [Google Scholar] [CrossRef] [PubMed]
  143. Li, X.; Zhou, Y.; Li, Y.; Yang, L.; Ma, Y.; Peng, X.; Yang, S.; Liu, J.; Li, H. Autophagy: A novel mechanism of chemoresistance in cancers. Biomed. Pharmacother. 2019, 119, 109415. [Google Scholar] [CrossRef] [PubMed]
  144. Huang, Z.; Zhou, L.; Chen, Z.; Nice, E.C.; Huang, C. Stress management by autophagy: Implications for chemoresistance. Int. J. Cancer 2016, 139, 23–32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Montay-Gruel, P.; Zhu, Y.; Petit, B.; Leavitt, R.; Warn, M.; Giedzinski, E.; Ollivier, J.; Sinclair, D.A.; Vozenin, M.C.; Limoli, C.L. Extracellular Vesicles for the Treatment of Radiation-Induced Normal Tissue Toxicity in the Lung. Front. Oncol. 2020, 10, 602763. [Google Scholar] [CrossRef]
  146. Fernandes, S.G.; Shah, P.; Khattar, E. Recent Advances in Therapeutic Application of DNA Damage Response Inhibitors against Cancer. Anticancer Agents Med. Chem. 2022, 22, 469–484. [Google Scholar] [CrossRef]
  147. Reuvers, T.G.A.; Kanaar, R.; Nonnekens, J. DNA Damage-Inducing Anticancer Therapies: From Global to Precision Damage. Cancers 2020, 12, 2098. [Google Scholar] [CrossRef]
  148. Zeng, A.; Wei, Z.; Yan, W.; Yin, J.; Huang, X.; Zhou, X.; Li, R.; Shen, F.; Wu, W.; Wang, X.; et al. Exosomal transfer of miR-151a enhances chemosensitivity to temozolomide in drug-resistant glioblastoma. Cancer Lett. 2018, 436, 10–21. [Google Scholar] [CrossRef]
  149. Bussard, K.M.; Mutkus, L.; Stumpf, K.; Gomez-Manzano, C.; Marini, F.C. Tumor-associated stromal cells as key contributors to the tumor microenvironment. Breast Cancer Res. 2016, 18, 84. [Google Scholar] [CrossRef] [Green Version]
  150. Su, C.; Zhang, J.; Yarden, Y.; Fu, L. The key roles of cancer stem cell-derived extracellular vesicles. Signal Transduct. Target. Ther. 2021, 6, 109. [Google Scholar] [CrossRef]
  151. Phi, L.T.H.; Sari, I.N.; Yang, Y.G.; Lee, S.H.; Jun, N.; Kim, K.S.; Lee, Y.K.; Kwon, H.Y. Cancer Stem Cells (CSCs) in Drug Resistance and their Therapeutic Implications in Cancer Treatment. Stem Cells Int. 2018, 2018, 5416923. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Scioli, M.G.; Terriaca, S.; Fiorelli, E.; Storti, G.; Fabbri, G.; Cervelli, V.; Orlandi, A. Extracellular Vesicles and Cancer Stem Cells in Tumor Progression: New Therapeutic Perspectives. Int. J. Mol. Sci. 2021, 22, 10572. [Google Scholar] [CrossRef] [PubMed]
  153. Yuwen, D.; Ma, Y.; Wang, D.; Gao, J.; Li, X.; Xue, W.; Fan, M.; Xu, Q.; Shen, Y.; Shu, Y. Prognostic Role of Circulating Exosomal miR-425-3p for the Response of NSCLC to Platinum-Based Chemotherapy. Cancer Epidemiol. Biomark. Prev. 2019, 28, 163–173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Liu, J.; Wu, S.; Zheng, X.; Zheng, P.; Fu, Y.; Wu, C.; Lu, B.; Ju, J.; Jiang, J. Immune suppressed tumor microenvironment by exosomes derived from gastric cancer cells via modulating immune functions. Sci. Rep. 2020, 10, 14749. [Google Scholar] [CrossRef] [PubMed]
  155. Battke, C.; Ruiss, R.; Welsch, U.; Wimberger, P.; Lang, S.; Jochum, S.; Zeidler, R. Tumour exosomes inhibit binding of tumour-reactive antibodies to tumour cells and reduce ADCC. Cancer Immunol. Immunother. 2011, 60, 639–648. [Google Scholar] [CrossRef] [PubMed]
  156. Mashouri, L.; Yousefi, H.; Aref, A.R.; Ahadi, A.M.; Molaei, F.; Alahari, S.K. Exosomes: Composition, biogenesis, and mechanisms in cancer metastasis and drug resistance. Mol. Cancer 2019, 18, 75. [Google Scholar] [CrossRef]
  157. Tagliabue, E.; Balsari, A.; Campiglio, M.; Pupa, S.M. HER2 as a target for breast cancer therapy. Expert Opin. Biol. Ther. 2010, 10, 711–724. [Google Scholar] [CrossRef]
  158. Tobinai, K. Rituximab and other emerging monoclonal antibody therapies for lymphoma. Expert Opin. Emerg. Drugs 2002, 7, 289–302. [Google Scholar] [CrossRef]
  159. Gao, Z.; Pang, B.; Li, J.; Gao, N.; Fan, T.; Li, Y. Emerging Role of Exosomes in Liquid Biopsy for Monitoring Prostate Cancer Invasion and Metastasis. Front. Cell Dev. Biol. 2021, 9, 679527. [Google Scholar] [CrossRef]
  160. Yu, D.; Li, Y.; Wang, M.; Gu, J.; Xu, W.; Cai, H.; Fang, X.; Zhang, X. Exosomes as a new frontier of cancer liquid biopsy. Mol. Cancer 2022, 21, 56. [Google Scholar] [CrossRef]
  161. Janpipatkul, K.; Trachu, N.; Watcharenwong, P.; Panvongsa, W.; Worakitchanon, W.; Metheetrairut, C.; Oranratnachai, S.; Reungwetwattana, T.; Chairoungdua, A. Exosomal microRNAs as potential biomarkers for osimertinib resistance of non-small cell lung cancer patients. Cancer Biomark. 2021, 31, 281–294. [Google Scholar] [CrossRef] [PubMed]
  162. Wang, S.; Song, Y.; Yan, F.; Liu, D. Mechanisms of resistance to third-generation EGFR tyrosine kinase inhibitors. Front. Med. 2016, 10, 383–388. [Google Scholar] [CrossRef] [PubMed]
  163. Kato, T.; Mizutani, K.; Kawakami, K.; Fujita, Y.; Ehara, H.; Ito, M. CD44v8-10 mRNA contained in serum exosomes as a diagnostic marker for docetaxel resistance in prostate cancer patients. Heliyon 2020, 6, e04138. [Google Scholar] [CrossRef] [PubMed]
  164. Liu, J.; Huang, Y.; Zhu, D.; Dai, Y.; Liu, D.; Zhai, Y.; Liang, X.; Wu, L.; Zhao, Q. Establishment and characterization of a docetaxel-resistant human prostate cancer cell line. Oncol. Lett. 2020, 20, 230. [Google Scholar] [CrossRef] [PubMed]
  165. Rice, M.A.; Malhotra, S.V.; Stoyanova, T. Second-Generation Antiandrogens: From Discovery to Standard of Care in Castration Resistant Prostate Cancer. Front. Oncol. 2019, 9, 801. [Google Scholar] [CrossRef]
  166. Del Re, M.; Biasco, E.; Crucitta, S.; Derosa, L.; Rofi, E.; Orlandini, C.; Miccoli, M.; Galli, L.; Falcone, A.; Jenster, G.W.; et al. The Detection of Androgen Receptor Splice Variant 7 in Plasma-derived Exosomal RNA Strongly Predicts Resistance to Hormonal Therapy in Metastatic Prostate Cancer Patients. Eur. Urol. 2017, 71, 680–687. [Google Scholar] [CrossRef]
  167. McKiernan, J.; Donovan, M.J.; Margolis, E.; Partin, A.; Carter, B.; Brown, G.; Torkler, P.; Noerholm, M.; Skog, J.; Shore, N.; et al. A Prospective Adaptive Utility Trial to Validate Performance of a Novel Urine Exosome Gene Expression Assay to Predict High-grade Prostate Cancer in Patients with Prostate-specific Antigen 2-10ng/mL at Initial Biopsy. Eur. Urol. 2018, 74, 731–738. [Google Scholar] [CrossRef] [Green Version]
  168. Wei, L.; Wang, J.; Lampert, E.; Schlanger, S.; DePriest, A.D.; Hu, Q.; Gomez, E.C.; Murakam, M.; Glenn, S.T.; Conroy, J.; et al. Intratumoral and Intertumoral Genomic Heterogeneity of Multifocal Localized Prostate Cancer Impacts Molecular Classifications and Genomic Prognosticators. Eur. Urol. 2017, 71, 183–192. [Google Scholar] [CrossRef] [Green Version]
  169. Urabe, F.; Ochiya, T.; Egawa, S. Re: A Prospective Adaptive Utility Trial to Validate Performance of a Novel Urine Exosome Gene Expression Assay to Predict High-grade Prostate Cancer in Patients with Prostate-specific Antigen 2-10ng/mL at Initial Biopsy. Eur. Urol. 2019, 76, 254–255. [Google Scholar] [CrossRef]
  170. McKiernan, J.; Donovan, M.J.; O’Neill, V.; Bentink, S.; Noerholm, M.; Belzer, S.; Skog, J.; Kattan, M.W.; Partin, A.; Andriole, G.; et al. A Novel Urine Exosome Gene Expression Assay to Predict High-grade Prostate Cancer at Initial Biopsy. JAMA Oncol. 2016, 2, 882–889. [Google Scholar] [CrossRef] [Green Version]
  171. Scott, L.C.; Mobley, L.R.; Kuo, T.M.; Il’yasova, D. Update on triple-negative breast cancer disparities for the United States: A population-based study from the United States Cancer Statistics database, 2010 through 2014. Cancer 2019, 125, 3412–3417. [Google Scholar] [CrossRef] [PubMed]
  172. Zhang, Z.; Zhang, L.; Yu, G.; Sun, Z.; Wang, T.; Tian, X.; Duan, X.; Zhang, C. Exosomal miR-1246 and miR-155 as predictive and prognostic biomarkers for trastuzumab-based therapy resistance in HER2-positive breast cancer. Cancer Chemother. Pharmacol. 2020, 86, 761–772. [Google Scholar] [CrossRef] [PubMed]
  173. Watanabe, S.; Yonesaka, K.; Tanizaki, J.; Nonagase, Y.; Takegawa, N.; Haratani, K.; Kawakami, H.; Hayashi, H.; Takeda, M.; Tsurutani, J.; et al. Targeting of the HER2/HER3 signaling axis overcomes ligand-mediated resistance to trastuzumab in HER2-positive breast cancer. Cancer Med. 2019, 8, 1258–1268. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Masuda, N.; Ohtani, S.; Takano, T.; Inoue, K.; Suzuki, E.; Nakamura, R.; Bando, H.; Ito, Y.; Ishida, K.; Yamanaka, T.; et al. A randomized, 3-arm, neoadjuvant, phase 2 study comparing docetaxel + carboplatin + trastuzumab + pertuzumab (TCbHP), TCbHP followed by trastuzumab emtansine and pertuzumab (T-DM1+P), and T-DM1+P in HER2-positive primary breast cancer. Breast Cancer Res. Treat. 2020, 180, 135–146. [Google Scholar] [CrossRef] [Green Version]
  175. Yang, S.J.; Wang, D.D.; Li, J.; Xu, H.Z.; Shen, H.Y.; Chen, X.; Zhou, S.Y.; Zhong, S.L.; Zhao, J.H.; Tang, J.H. Predictive role of GSTP1-containing exosomes in chemotherapy-resistant breast cancer. Gene 2017, 623, 5–14. [Google Scholar] [CrossRef]
  176. Rexer, B.N.; Arteaga, C.L. Intrinsic and acquired resistance to HER2-targeted therapies in HER2 gene-amplified breast cancer: Mechanisms and clinical implications. Crit. Rev. Oncog. 2012, 17, 1–16. [Google Scholar] [CrossRef] [Green Version]
  177. Luo, Y.; Gui, R. Circulating exosomal circFoxp1 confers cisplatin resistance in epithelial ovarian cancer cells. J. Gynecol. Oncol. 2020, 31, e75. [Google Scholar] [CrossRef]
  178. Wang, G.; Zhao, W.; Wang, H.; Qiu, G.; Jiang, Z.; Wei, G.; Li, X. Exosomal MiR-744 Inhibits Proliferation and Sorafenib Chemoresistance in Hepatocellular Carcinoma by Targeting PAX2. Med. Sci. Monit. 2019, 25, 7209–7217. [Google Scholar] [CrossRef]
  179. Guyon, N.; Garnier, D.; Briand, J.; Nadaradjane, A.; Bougras-Cartron, G.; Raimbourg, J.; Campone, M.; Heymann, D.; Vallette, F.M.; Frenel, J.S.; et al. Anti-PD1 therapy induces lymphocyte-derived exosomal miRNA-4315 release inhibiting Bim-mediated apoptosis of tumor cells. Cell Death Dis. 2020, 11, 1048. [Google Scholar] [CrossRef]
  180. Yang, Y.N.; Zhang, R.; Du, J.W.; Yuan, H.H.; Li, Y.J.; Wei, X.L.; Du, X.X.; Jiang, S.L.; Han, Y. Predictive role of UCA1-containing exosomes in cetuximab-resistant colorectal cancer. Cancer Cell Int. 2018, 18, 164. [Google Scholar] [CrossRef] [Green Version]
  181. Li, M.Y.; Zhao, C.; Chen, L.; Yao, F.Y.; Zhong, F.M.; Chen, Y.; Xu, S.; Jiang, J.Y.; Yang, Y.L.; Min, Q.H.; et al. Quantitative Proteomic Analysis of Plasma Exosomes to Identify the Candidate Biomarker of Imatinib Resistance in Chronic Myeloid Leukemia Patients. Front. Oncol. 2021, 11, 779567. [Google Scholar] [CrossRef] [PubMed]
  182. Sun, Y.; Huo, C.; Qiao, Z.; Shang, Z.; Uzzaman, A.; Liu, S.; Jiang, X.; Fan, L.Y.; Ji, L.; Guan, X.; et al. Comparative Proteomic Analysis of Exosomes and Microvesicles in Human Saliva for Lung Cancer. J. Proteome Res. 2018, 17, 1101–1107. [Google Scholar] [CrossRef] [PubMed]
  183. Panvongsa, W.; Pegtel, D.M.; Voortman, J. More than a Bubble: Extracellular Vesicle microRNAs in Head and Neck Squamous Cell Carcinoma. Cancers 2022, 14, 1160. [Google Scholar] [CrossRef] [PubMed]
  184. Gai, C.; Camussi, F.; Broccoletti, R.; Gambino, A.; Cabras, M.; Molinaro, L.; Carossa, S.; Camussi, G.; Arduino, P.G. Salivary extracellular vesicle-associated miRNAs as potential biomarkers in oral squamous cell carcinoma. BMC Cancer 2018, 18, 439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. He, L.; Ping, F.; Fan, Z.; Zhang, C.; Deng, M.; Cheng, B.; Xia, J. Salivary exosomal miR-24-3p serves as a potential detective biomarker for oral squamous cell carcinoma screening. Biomed. Pharmacother. 2020, 121, 109553. [Google Scholar] [CrossRef] [PubMed]
  186. Lin, Y.; Dong, H.; Deng, W.; Lin, W.; Li, K.; Xiong, X.; Guo, Y.; Zhou, F.; Ma, C.; Chen, Y.; et al. Evaluation of Salivary Exosomal Chimeric GOLM1-NAA35 RNA as a Potential Biomarker in Esophageal Carcinoma. Clin. Cancer Res. 2019, 25, 3035–3045. [Google Scholar] [CrossRef]
  187. Yekula, A.; Muralidharan, K.; Kang, K.M.; Wang, L.; Balaj, L.; Carter, B.S. From laboratory to clinic: Translation of extracellular vesicle based cancer biomarkers. Methods 2020, 177, 58–66. [Google Scholar] [CrossRef]
  188. Li, S.; Yi, M.; Dong, B.; Jiao, Y.; Luo, S.; Wu, K. The roles of exosomes in cancer drug resistance and its therapeutic application. Clin. Transl. Med. 2020, 10, e257. [Google Scholar] [CrossRef]
  189. Wang, J.; Zheng, Y.; Zhao, M. Exosome-Based Cancer Therapy: Implication for Targeting Cancer Stem Cells. Front. Pharmacol. 2016, 7, 533. [Google Scholar] [CrossRef] [Green Version]
  190. Srivastava, A.; Babu, A.; Filant, J.; Moxley, K.M.; Ruskin, R.; Dhanasekaran, D.; Sood, A.K.; McMeekin, S.; Ramesh, R. Exploitation of Exosomes as Nanocarriers for Gene-, Chemo-, and Immune-Therapy of Cancer. J. Biomed. Nanotechnol. 2016, 12, 1159–1173. [Google Scholar] [CrossRef]
  191. Liang, G.; Zhu, Y.; Ali, D.J.; Tian, T.; Xu, H.; Si, K.; Sun, B.; Chen, B.; Xiao, Z. Engineered exosomes for targeted co-delivery of miR-21 inhibitor and chemotherapeutics to reverse drug resistance in colon cancer. J. Nanobiotechnol. 2020, 18, 10. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Representation of the biogenesis of vesicles. From a generic donor cell, budding of different vesicles: exosomes/small extracellular vesicles (size: less than 0.15 µm); microvesicles/medium extracellular vesicles (size: 0.15–1 µm); apoptotic bodies/large extracellular vesicles (size: greater than 1 µm). MVB, multivesicular body; EV, extracellular vesicle. Intracellular organelles, RNAs (red), and proteins (green and light blue) are indicated.
Figure 1. Representation of the biogenesis of vesicles. From a generic donor cell, budding of different vesicles: exosomes/small extracellular vesicles (size: less than 0.15 µm); microvesicles/medium extracellular vesicles (size: 0.15–1 µm); apoptotic bodies/large extracellular vesicles (size: greater than 1 µm). MVB, multivesicular body; EV, extracellular vesicle. Intracellular organelles, RNAs (red), and proteins (green and light blue) are indicated.
Life 12 00618 g001
Figure 2. Representation of pathways regulating membrane transporters in chemoresistance. Small extracellular vesicles carry RNAs (in red, mRNA, miRNA, lncRNA), regulatory proteins (green), and membrane transporters (blue). In the donor cells, mRNAs induce de novo transporter synthesis, miRNAs reduce the expression of negative regulators of transporter function/expression, lncRNAs induce miRNA degradation with consequent transporter overexpression, proteins (green) regulate transporter function, and transporter(s) carried by small EVs are inserted directly in the membrane.
Figure 2. Representation of pathways regulating membrane transporters in chemoresistance. Small extracellular vesicles carry RNAs (in red, mRNA, miRNA, lncRNA), regulatory proteins (green), and membrane transporters (blue). In the donor cells, mRNAs induce de novo transporter synthesis, miRNAs reduce the expression of negative regulators of transporter function/expression, lncRNAs induce miRNA degradation with consequent transporter overexpression, proteins (green) regulate transporter function, and transporter(s) carried by small EVs are inserted directly in the membrane.
Life 12 00618 g002
Figure 3. Representation of pathways regulating chemoresistance. Small extracellular vesicles carry RNAs (in red, mRNA, miRNA, lncRNA), regulatory proteins (green), and membrane transporters (blue) released by chemoresistant cancer cells (donor-red membrane). In the donor cells, RNAs (in red, circRNAs, miRNAs, and lncRNAs) and proteins (green) regulate cell cycle control, proapototic and antiapoptic programs, and autophagy. The donor cells release antiapoptotic proteins (blue) to activate prosurvival pathways.
Figure 3. Representation of pathways regulating chemoresistance. Small extracellular vesicles carry RNAs (in red, mRNA, miRNA, lncRNA), regulatory proteins (green), and membrane transporters (blue) released by chemoresistant cancer cells (donor-red membrane). In the donor cells, RNAs (in red, circRNAs, miRNAs, and lncRNAs) and proteins (green) regulate cell cycle control, proapototic and antiapoptic programs, and autophagy. The donor cells release antiapoptotic proteins (blue) to activate prosurvival pathways.
Life 12 00618 g003
Table 1. Chemoresistance associated with extracellular vesicles.
Table 1. Chemoresistance associated with extracellular vesicles.
EV Cargo TypeCancer TypeDrug ResistanceRef.
miRNALung cancerCisplatin[14,15]
Gemcitabine[16]
Gefitinib[17,18]
Methotrexate[3,19]
Gastric cancerPaclitaxel[20]
Cisplatin[20]
Doxorubicin [21]
Breast cancerPaclitaxel [22]
Doxorubicin[22]
Docetaxel[23,24]
Gemcitabine[24]
CDK4/6 inhibitor[25]
Epirubicin[24]
Head and neck cancerCisplatin[26,27]
Pancreatic cancerGemcitabine[28]
Hepatic carcinoma5-Fluorouracil[29]
Oxaliplatin
Gemcitabine
Sorafenib
Colorectal cancer5-Fluorouracil[30,31]
Oxaliplatin[31]
Ovarian cancerCisplatin[32]
Paclitaxel[33,34]
LymphomaGemcitabine[35]
Prostate cancerCisplatin[36]
Doxorubicin
Docetaxel
lncRNALung cancerGefitinib[37]
Ovarian cancerCisplatin[38]
Esophageal cancerGefitinib[39]
Sorafenib [40]
Camptothecin [40]
Doxorubicin[40]
Renal cell carcinomaSunitinib[41]
GlioblastomaTemozolomide[42]
Breast cancerTrastuzumab[43,44]
Colorectal cancerOxaliplatin[45,46]
Bladder cancerCisplatin[47]
circRNAColorectal cancerOxaliplatin[48]
Lung cancerCisplatin[49]
Gastric cancerCisplatin[50]
mRNAGliomaTemozolomide[51]
Lung cancerCisplatin[52]
Gemcitabine[52]
OsteosarcomaDoxorubicin[53]
ProteinBreast cancerAdriamycin[54]
Docetaxel[55]
Doxorubicin[56]
Paclitaxel[57]
Trastuzumab[58]
Ovarian cancerCisplatin [59]
Paclitaxel [60]
Platinum-based therapy[60]
Cisplatin [61]
OsteosarcomaDoxorubicin[53]
Prostate cancerDocetaxel[62]
Colorectal cancer5-Fluorouracil [63,64]
Pancreatic cancerGemcitabine [65]
B-cell lymphomaR-CHOP [66]
Rituximab[67]
GlioblastomaTemozolomide[68]
Table 2. Exosomal bioactive molecules involved in the induction of drug resistance.
Table 2. Exosomal bioactive molecules involved in the induction of drug resistance.
SectionEffectorsMechanismsCancer TypeDrug ResistanceMarkerRef.
2Acidic environmentIncreased EV secretionMelanomaCisplatinRab-5b[78]
CD63
ABCA3 depletionDecreased EV secretion ameliorates drug sensitivenessB-cell lymphomasDoxorubicin PixantroneCD9[79]
CD63
ADAM10
Annexin A3Increased EV secretionOvarian cancerPt-drugsTEM[81]
Hsp70
P-gpDelivering through EVsProstate cancerDocetaxelCD9[62]
Doxorubicin
MDR-1 mRNA/P-gpDelivering through EVsOsteosarcomaDoxorubicinCD63[53]
P-gpDelivering through EVsBreast cancerDocetaxelTSG101[55]
TrpC5Induction of P-gp expressionBreast cancerAdriamycinFlotillin-2[56]
TrpC5Delivering through EVsBreast cancerAnthracycline TaxaneCD63[98]
Flotillin-1
UCH-L1Induction of P-gp expressionBreast cancerAdriamycinCD63[54]
Flotillin-1
LINC00355Induction of P-gp expressionBladder cancerCisplatinCD9[47]
CD63
miR-1246Downregulation of caveolin1,Ovarian cancerPaclitaxelCD63[34]
upregulation of P-gp
circ_PIP5K1ADownregulation ofLung cancerCisplatinCD81[49]
miR-101CD63
linc-VLDLRUpregulation of ABCG2Hepatocellular cancerSorafenib, camptothecinNTA[99]
Doxorubicin
ABCA3Increased exosome secretionsMalignant lymphomaRituximabFlotillin-2[67]
Alix
CD9
CD63
ATP7AIncreased Pt-drug excretionOvarian cancerPt-based drugsMicroscopy of labeled vesicles[61]
ATP7B
miR-4717-5pDownregulating of ENT2LymphomaGemcitabineCD9[35]
CytarabineCD63
miR-1236SLC9A1 downregulation ameliorates drug sensitivenessBreast cancerCisplatinCD9[100]
CD63
CD81
HSP70
Sulfasalazine (SAS)EVs from cells treatedMelanomaImmune checkpoint blockade (ICB) therapyTEM[101]
with xCT inhibitor SAS reduced ICB therapy efficacy
3miR-145/−34aLower apoptosis levelColon cancer5-FluorouracilNTA[30]
Bone marrow stromal cell derived EVsIncrease in antiapoptotic proteinsMultiple myelomaBortezomibHsp90[102]
Hsp70
CD63
Flotillin-1
SNHG14Modulation of Bcl-2/BaxBreast cancerTrastuzumabCD9[43]
CD63
CD81
Alix
PART1Modulation of miR-129Esophageal squamous cell carcinomaGefitinibCD63[39]
CD81
miR-21Modulation of PTEN/PDCD4Squamous cell carcinomaCisplatinCD81[27]
CD68
miR-32-5pPI3K/Akt pathway activationHepatocellular carcinomaMultidrugCD63[29]
resistanceTSG-101
Flotillin-1
miR-27aTP53Prostate cancerCisplatin, docetaxel, doxorubicinCD63[36]
CD9
EVs from HBV-associatedChaperone-mediated autophagyLiver cancerOxaliplatinCD63[103]
liver cancer cellsCD9
miR-425-3pModulation of AKT1Lung cancerCisplatinTEM[14]
NTA
PD-L1Activation ofGlioblastomaTemozolomideTSG101[68]
AMPK/ULK1
pathway
circ-PVT1ModulatingGastric cancerCisplatinCD63[50]
miR-30a-5pCD9
SBF2-AS1Modulation of miR-151aGlioblastomaTemozolomideCD63[42]
CD81
MGMTDelivering through EVsGliomaTemozolomideCD63[51]
CD81
4miR-210Inhibition of apoptosisPancreatic cancerGemcitabineCD63[104]
CD81
GM130
miR-92a-3pFBXW7, MOAP1Colorectal cancer5-FluorouracilCD63[31]
OxaliplatinCD81
TSG101
G-CSF, IL-6Gene expression changesLung carcinomaMethotrexateEV communication in[19]
Activin-Acocultured cells was blocked by xyloside
WntReprogramming of differentiated cancer cellsColorectal cancer5-FluorouracilCD81[63]
5HER2Antibody bindingBreast cancersTrastuzumabCD63[58]
Flotillin-1
AFAP1-AS1Upregulation HER-2Breast cancersTrastuzumabTSG101[44]
CD81
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Console, L.; Scalise, M. Extracellular Vesicles and Cell Pathways Involved in Cancer Chemoresistance. Life 2022, 12, 618. https://doi.org/10.3390/life12050618

AMA Style

Console L, Scalise M. Extracellular Vesicles and Cell Pathways Involved in Cancer Chemoresistance. Life. 2022; 12(5):618. https://doi.org/10.3390/life12050618

Chicago/Turabian Style

Console, Lara, and Mariafrancesca Scalise. 2022. "Extracellular Vesicles and Cell Pathways Involved in Cancer Chemoresistance" Life 12, no. 5: 618. https://doi.org/10.3390/life12050618

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