Download PDF
Review  |  Open Access  |  11 Dec 2024

Overcoming drug resistance through extracellular vesicle-based drug delivery system in cancer treatment

Views: 85 |  Downloads: 13 |  Cited:  0
Cancer Drug Resist 2024;7:50.
10.20517/cdr.2024.107 |  © The Author(s) 2024.
Author Information
Article Notes
Cite This Article

Abstract

Drug resistance is a major challenge in cancer therapy that often leads to treatment failure and disease relapse. Despite advancements in chemotherapeutic agents and targeted therapies, cancers often develop drug resistance, making these treatments ineffective. Extracellular vesicles (EVs) have gained attention for their potential applications in drug delivery because of their natural origin, biocompatibility, and ability to cross biological barriers. Using the unique properties of EVs could enhance drug accumulation at target sites, minimize systemic toxicity, and precisely target specific cells. Here, we discuss the characteristics and functionalization of EVs, the mechanisms of drug resistance, and the applications of engineered EVs to overcome drug resistance. This review provides a comprehensive overview of the advancements in EV-based drug delivery systems and their applications in overcoming cancer drug resistance. We highlight the potential of EV-based drug delivery systems to revolutionize cancer therapy and offer promising strategies for more effective treatment modalities.

Keywords

Extracellular vesicles, drug delivery systems, drug resistance, tumor microenvironment, cancer therapy

INTRODUCTION

Drug resistance remains a formidable obstacle in cancer therapy, often leading to treatment failure and disease relapse[1,2]. Despite the advancements in the development of chemotherapeutic agents and targeted therapies, cancers often develop resistance mechanisms that render these treatments ineffective[3]. The emergence of drug-resistant cancer cells can be attributed to various factors including genetic mutations, altered drug metabolism, efflux pump overexpression, and modifications in cell signaling pathways[4-6]. These complexities necessitate innovative approaches for enhancing the efficacy of cancer treatments.

Extracellular vesicles (EVs), including exosomes, microvesicles, and apoptotic bodies, have garnered considerable attention in recent years because of their potential role in intercellular communication and ability to transfer bioactive molecules between cells[7-11]. EVs are lipid bilayer-enclosed particles released by cells that carry diverse cargoes such as proteins, lipids, nucleic acids, and other small molecules[12-14]. Their natural origin, biocompatibility, and inherent ability to cross biological barriers make them promising candidates for drug delivery[15,16].

Recent studies have highlighted the potential of EV-based drug delivery systems to overcome drug resistance in cancer[7,13,17]. By leveraging their unique properties, EVs can be engineered to directly deliver chemotherapeutic agents, small interfering RNAs (siRNAs), microRNAs (miRNAs), and other therapeutic molecules to cancer cells, thereby enhancing drug accumulation at target sites and minimizing systemic toxicity[18-20]. In addition, EVs can be modified to target specific cell types, thereby providing a level of precision that conventional drug delivery methods lack[21-23].

This review aims to provide a comprehensive overview of the current advancements in EV-based drug delivery systems and their applications in overcoming drug resistance in cancer. We discuss EVs and their functionalization, mechanisms of drug resistance in cancer, and the application of engineered EVs to overcome drug resistance. By exploring these aspects, we hope to shed light on the potential of EV-based drug delivery systems to revolutionize cancer therapy and address the pressing issue of drug resistance in cancer.

EVS AND THEIR FUNCTIONALIZATION

EVs

In 1985, Pan et al. observed the vesicle structures secreted during the maturation of reticulocytes using a microscope[24]. Over the past few decades, in-depth studies have provided a clearer understanding of the biological processes and structure of EVs. The 2023 edition of the International Society for EVs provides a clear definition. The term “Evs” refers to particles that are released from cells, delimited by a lipid bilayer, and cannot replicate on their own (that is, they do not contain a functional nucleus)[8]. EVs are classified into three subtypes based on their size, structure, and biological processes of formation: exosomes, microvesicles, and apoptotic bodies [Figure 1][9]. Exosomes are vesicles formed by the inward budding of the membrane through endocytosis and then secreted outside the cell. Microvesicles are formed by the outward budding and shedding of the cell membrane; apoptotic bodies are produced by the disintegration of cells during programmed cell death[10,11]. EVs act as messengers for intercellular communication, with numerous studies indicating that they play significant roles in tumor formation and development and are closely related to cancer resistance mechanisms[7]. In addition, EVs serve as excellent drug carriers, capable of delivering proteins, DNA, RNA, and other molecules for the treatment of cancer or chronic wounds[12-14,25].

Overcoming drug resistance through extracellular vesicle-based drug delivery system in cancer treatment

Figure 1. Schematic representation of subtypes of EVs. EVs: Extracellular vesicles.

Characterization of EVs

EVs are complex structures containing various proteins and lipids. Common laboratory characterization methods for studying EV structures include nanoparticle tracking analysis (NTA), dynamic light scattering (DLS), transmission electron microscopy (TEM), flow cytometry, and western blotting (WB).

NTA uses laser scattering and the principle of the random Brownian motion of particles to measure the size distribution and concentration of EVs in liquid suspensions, making it one of the most used characterization methods in EV research[26,27]. DLS, similar in principle to NTA, also detects the size of EVs but measures the overall scattering intensity from the entire sample[28,29]. TEM enables direct observation of EV morphological characteristics[30]. Flow cytometry can be used to quantitatively analyze EVs by detecting the signal intensity of fluorescence-labeled antibodies specific to membrane surface markers[31-33]. WB is commonly used to detect proteins within EVs, which are first separated by gel electrophoresis and labeled with antibodies for qualitative or semi-quantitative analysis of the labeled proteins[8,34].

Isolation of EVs

EVs are small vesicles of varying sizes that are secreted by cells and are characterized by low toxicity, low immunogenicity, and excellent biocompatibility, making them ideal drug delivery vehicles. The following sections outline several commonly used methods for isolating EVs from cells. Different EV isolation techniques of EVs are summarized in Table 1.

Table 1

Comparison of different isolation methods of EVs

Isolation methodPrincipleAdvantagesDisadvantagesApplicationRef.
UltracentrifugationIsolation based on size and density through centrifugal force to separate the impurities from samplesHigh productivity;
Simple procedure;
Easy to use
Time-consuming;
Damage to the EVs;
Formation of aggregates and precipitation
Blood, urine, cerebrospinal fluid[35-40]
UltrafiltrationIsolation uses a porous filter of size‐exclusion limits, which removes smaller molecules (non‐EV) by flow‐throughSimple procedure; Suitable for large-volume isolation; Retain EVs functionality and integrityTime-consuming;
Low purity;
Low productivity
Blood, urine, and saliva[41-44]
PrecipitationInduced clumping of EVs in the form of pellet by decreasing the solubilitySimple procedure;
No additional equipment required;
Suitable for large-volume isolation
Low efficiency;
Time-consuming;
High cost;
Low purity with co-precipitation of protein and other molecules
Plasma, urine, saliva[45-47]
Size-exclusion chromatographyIsolation based on single isolation column containing porous beads with radii smaller than EVsHigh purity;
High homogeneity; Retain EVs functionality and integrity
Low productivity;
Not suitable for higher sample volumes
Cell culture supernatants and complex biological fluids like plasma[48-55]
ImmunoaffinityUsing immunoaffinity substances such as proteins, aptamers, and antibodies that can recognize the surface-specific structure of EVs to recognize and capture EVsHigh efficiency;
High specificity and purity;
Retain EVs functionality and integrity
Low productivity;
Only enrichment of EVs with specific proteins;
Not suitable for larger sample volumes;
High cost
Tumor cells, immune cells[56-59]

Ultracentrifugation

Ultracentrifugation separates components in a solution based on size and density differences and is the most used method because of its simplicity[35,38,39]. EVs can be isolated from samples such as human urine, saliva, intestinal digestive fluid, cardiac and alveolar cell culture media, mouse hippocampal interstitial fluid, and milk using ultracentrifugation[60-66]. However, this method has several drawbacks, including time-consuming extraction and the low purity of isolated EVs, making it suitable only for small sample sizes[36,40,67].

Precipitation

Precipitation is a straightforward, instrument-free technique for exosome isolation. It is a polymer-based method in which a sample is mixed with a polymer at a low temperature and the salt concentration is adjusted, inducing EV aggregation and precipitation by reducing solubility[45-47]. For example, Mishra et al. used polyethylene glycol (PEG) to precipitate EVs from the plasma of patients to achieve high purity[68]. Similarly, Pecksen et al. used PEG to isolate monocyte-derived EVs from a co-culture of induced pluripotent stem cells and human monocytes[69]. Matchett and Kornbluth used a modified PEG-acetate reagent to isolate natural killer (NK) cell-derived EVs[70].

Size-exclusion chromatography

Size-exclusion chromatography (SEC) effectively separates EVs according to particle size, using porous beads that are smaller than EVs. It is suitable for isolating EVs from complex tissue or fluid samples and has significant clinical research applications[49,50,52,53,55]. For example, Livkisa et al. used SEC to remove impurities, such as carrier proteins, coagulation factors, and complement proteins, from platelet lysates, thereby achieving high-purity EVs[71].

Immunoaffinity capture

Immunoaffinity capture uses substances such as proteins, aptamers, and antibodies that specifically recognize and capture EVs based on the unique structures on their surfaces. This method exhibits high efficiency, specificity, and purity[56-59]. For instance, Koksal et al. used phycoerythrin-conjugated human CD9, AFP, and GPC3 antibodies to capture exosomes from the sera of patients[72]. Wang et al. modified nanofibers with CD63 antibodies to effectively capture EVs expressing surface CD63 proteins[73].

Ultrafiltration

The ultrafiltration technique uses a porous filter of a certain size with exclusion limits to eliminate smaller molecules (non-EVs) by flow-through[74]. This method is typically combined with other methods. Hanson et al. used ultrafiltration and SEC to isolate EVs from C2C12 myoblasts[75]. Moreover, ultrafiltration combined with differential centrifugation or ultracentrifugation can be used to isolate EVs derived from stem cells and gut bacteria[76-78].

Other methods

Existing methods for separating EVs cannot meet the experimental requirements, and there is an urgent need for new separation methods or improvements to existing ones. Bathini et al. proposed a liquid biopsy chip using the synthetic peptide Vn96 for EV isolation based on magnetic particles[79]. Chattrairat et al. upgraded the conventional well plate assay to an all-in-one nanowire-integrated well plate assay for charge-based EV capture and membrane protein analysis[80]. Zhang et al. developed a rapid EV aggregation induced in-situ miRNA detection technology based on a cationic lipid-polymer hybrid nanoparticle encapsulating cascade system of catalytic hairpin assembly and CRISPR-Cas12a, achieving an enrichment efficiency of > 90% in 30 min, 5 times higher than that of ultracentrifugation[81].

Strategies to functionalize EVs

Methods for functionalizing EVs primarily include surface modification and loading of targeted drug molecules.

Surface modification strategies

Before using natural EVs for drug delivery, EV membranes can be modified with various molecules to increase the specificity for target cells. The surface modification strategies for EVs include genetic modification, chemical modification, metabolic labeling, enzymatic conjugation, membrane fusion with liposomes, and affinity binding[82-85]. Here, we focus on the current EV surface modification strategies, including genetic and chemical modifications [Figure 2].

Overcoming drug resistance through extracellular vesicle-based drug delivery system in cancer treatment

Figure 2. Alteration of EV surface by chemical modification and genetic engineering. EVs: Extracellular vesicles; EGFR: epidermal growth factor receptor; PDGFR: platelet-derived growth factor receptor; GPI: glycosylphosphatidylinositol.

Genetic modifications
This approach enhances EV targeting to specific cellular and tissue environments but is limited to targeting genetically encodable motifs[86]. Genetic modifications involve the fusion of gene sequences of proteins or polypeptides with selected EV membrane protein gene sequences[21] [Table 2].

Table 2

Applications of genetic modifications to EVs

Fusion proteinEVs sourceModification detailsTargetCommentsRef.
Lamp-2bMSCsIL3LSCsEnhanced bone marrow homing and selective targeting of LSCs[22]
Lamp-2bDendritic cellsRVG peptideNeurons, microglia, OligodendrocytesDemonstrates effective siRNA brain delivery[87]
Lamp-2bMouse immature dendritic cellsiRGD peptideαv integrin-positive breast cancer cellsEnhanced targeting ability[88]
Lamp-2bHEKT 293 cellICAM-1Recombinant LFA-1Interacts with T cells, efficiently delivers functional plasmids[89]
PDGFRExpi293F cellsCD3 and EGFR antibodyT cell CD3 and
EGFR
Cross-links T cells and EGFR-positive cancer cells, triggers antitumor immunity[90]
PDGFRExpi293 cellsCD3 and HER2 antibodiesT cell CD3 and breast cancer-associated HER2 receptorsPromising exosome-based HER2 breast cancer immunotherapy[91]
CD63HEK293T cellsApo-A1 complexHepG2 cellsShows effective targeted miRNA delivery for liver cancer therapy[92]

Lamp2b, a member of the lysosome-associated membrane protein family, is highly expressed in EVs derived from dendritic cells (DCs) and is commonly used as a fusion protein for genetic modification of EVs. Fusing the Rabies Virus Glycoprotein-Derived Peptide with the Lamp2b on EV surfaces improves the brain-targeting ability of EVs, making them potential carriers for brain disease therapeutics[87]. The iRGD specifically binds to αv-integrins, and fusing it with Lamp2b on EV surfaces efficiently targets αv-integrin-positive breast cancer cells[88]. EVs overexpressing the fused protein Lamp2b-IL3 effectively target LSCs and are a potential drug carrier for acute myeloid leukemia[22]. Fusing Intercellular adhesion molecule-1 with Lamp2b on EVs can specifically target T cells[89].

The platelet-derived growth factor receptor (PDGFR) is expressed on EV membranes and can be fused with various antibodies. For example, combining anti-CD3 and anti-epidermal growth factor receptor (EGFR) antibodies with the PDGFR region on EV membranes can simultaneously target T cells and EGFR-expressing triple-negative breast cancer cells[90]. Similarly, the fusion of anti-CD3 and anti-HER2 antibodies targets T cells and HER2-expressing breast cancer cells[91].

The CD63, a common tetraspanin on EV surfaces, has been used for genetic modifications. The fusion of CD63 with the Apo-A1 sequence enables the targeting of HepG2 cells[92]. Furthermore, combining the inhibitory domain of the myostatin propeptide with CD63 can enhance the delivery and efficacy of the treatment for Duchenne muscular dystrophy[93].

Chemical modifications
Chemical modifications involve the attachment of specific molecules to the surface of EVs via chemical reactions[94]. These modifying molecules are primarily antibodies, peptides, or other targeting aptamers that selectively bind to receptors on cancer or immune cells to enhance their therapeutic effects[95-97] [Table 3].

Table 3

Applications of chemical modifications to EVs

EVs sourceModification detailsTargetCommentsRef.
MSCsc(RGDyK) peptideThe lesion region of the ischemic brainSuppress the inflammatory response and cellular apoptosis in the lesion region[98]
BM-MSCsCathepsin KSMCsImprove the cell-specific uptake of EVs and may be effective in facilitating AAA-targeted therapy[99]
M1 macrophagesAntibodies of CD47 and SIRPαaCD47 and CD47 on tumor cell surfaceActively target tumors through the specific recognition between aCD47 and CD47 on tumor cell surface[100]
U-87 cellsDSPE-PEG2000-FAU-87 cellsReduce the side effects of TMZ (minimal change in body weight), prolong survival, and inhibit tumor growth in mouse glioma models[101]
Neuro2A cellsNanobody-PEG-lipidsEGFRImprove cell specificity and prolong circulation time, potentially increasing;
EV accumulation in targeted tissues and improving cargo delivery.
[102]
Neuro2A cellsGPI-linked EGFR nanobodiesEGFRGreatly improve EV binding to tumour cells[103]
Primary dendritic cellsAptamer AS1411MDA-MB-231 cellsTumor targeting[104]
Mouse liver proliferative cellsGlycosylation-Promote the accumulation of EVs in the lungs[105]
M2 macrophagesBMSC-specific aptamerBMSCspromote the proliferation, migration, and osteogenic differentiation of BMSC[106]
HeLa cellspH-sensitive fusogenic peptide-The effective cellular uptake and cytosolic release of an artificially encapsulated dextran macromolecule (70 kDa) in exosomes[107]
HEK 293 T cellsCPP-conjugated-lipidsHUVECsImprove the intracellular delivery of exosomes[108]
Raw264.7 cellsNeuropilin-1-targeted peptideGlioma cellscross the BBB, achieve targeted imaging and therapy of glioma[23]

The lipid surfaces of EVs can be functionalized to improve their targeting capabilities by incorporating various small molecules using click chemistry. Using click chemistry, neuropilin-1 targeting peptides (RGERPPR, RGE) can be conjugated to the lipid surface of EVs, facilitating targeted delivery to glioma tissue[23]. There are reports where the parental cells can be metabolically engineered to secrete EVs with surface-modified azides. Click chemistry can be used to modify the azides on the surface of EVs with antibodies or peptides to enhance the targeting ability of EVs[83]. For instance, exosomes modified with c(RGDyK) peptides using click chemistry targeted ischemic brain regions in a mouse model of transient middle cerebral artery occlusion[98]. Cathepsin K (CatK), a cysteine protease overexpressed in the vascular walls of abdominal aortic aneurysms, can be targeted by attaching CatK-targeting peptides to EVs using click chemistry, thereby improving their uptake by tumor cells[99]. Similarly, azide-modified exosomes can be conjugated with dibenzocyclooctyne-modified antibodies of CD47 and SIRPα (aCD47 and aSIRPα) through a pH-sensitive linker. These exosomes can actively target tumors[100].

Chemical modifications of EV membranes can also be achieved using other methods. Functionalized PEG fused with EV membrane can achieve different functional effects. For example, EVs combined with folic acid-modified functional PEG can effectively target glioma cells and deliver drugs[101]. EVs conjugated with EGFR antibody-modified functional PEG can target tumor cells overexpressing EGFR receptors[102]. Glycosylphosphatidylinositol (GPI) anchor proteins, commonly occurring on cell membranes and expressed on EV membranes, can bind to anti- EGFR nanobodies, enabling the targeting of tumor cells overexpressing EGFR[103].

The EVs membrane can also bind to the aptamer AS1411 through hydrophobic interactions, allowing the targeting of breast cancer tissues[104]. In addition, chemical modifications can influence the affinity of EVs for specific tissues. For example, EVs modified with neuraminidase have a strong affinity for the liver, whereas EVs modified with glycosidase have a strong affinity for the lung tissue[105].

Therapeutic cargo loading in engineered EVs

Functionalized EVs not only require surface modifications to enhance targeting capabilities but also need to load drugs into the vesicles. The commonly used methods for drug loading are divided into two categories: endogenous and exogenous loading [Figure 3]. Endogenous loading involves transfection to make donor cells express certain molecules or incubation of the donor cells with drugs, allowing the cells to uptake these drug molecules and subsequently secrete drug-loaded EVs[109]. Endogenous loading does not disrupt the structure and activity of EVs and provides stable loading; however, loading efficiency is relatively low[110]. Exogenous loading involves isolating EVs from cells and then loading drugs into the vesicles through physical or chemical methods such as electroporation, sonication, membrane permeabilization, freeze-thaw cycles, and extrusion[111]. Exogenous loading offers a higher loading efficiency but can affect the structure and activity of EVs to some extent[112,113]. The choice of drug-loading method depends on specific experimental needs, with detailed applications provided in Tables 4 and 5.

Overcoming drug resistance through extracellular vesicle-based drug delivery system in cancer treatment

Figure 3. Therapeutic cargo loading methods in engineered EVs. Endogenous loading involves transfecting donor cells to express specific molecules or incubating them with drugs, enabling the cells to absorb the drugs and release drug-loaded extracellular vesicles. Exogenous loading involves isolating extracellular vesicles from cells and then using physical or chemical methods to load drugs into the vesicles. EVs: Extracellular vesicles.

Table 4

Applications of endogenous loading strategies for EVs

CargoLoading methodIsolation methodLoading efficiencyRef.
OxaliplatinIncubationSEC-[114]
PEG-HGNsIncubation-~ 50%[115]
DSPE-PEG-RGDIncubationUC-[116]
LinezolidIncubationPrecipitation-[117]
microRNA let-7a-5pTransfectionUC, UFThe let-7a-5p copy numbers within 1 × 1010 EVs had 6.46 × 1010 copies[118]
MicroRNA-27aTransfectionUF-[119]
VMP1TransfectionUF, UC-[120]
pDNA/CPPTransfectionPrecipitation, UF, SEC76% to 86%[121]
GDF-15TransfectionUC, UF-[122]
RAGE siRNATransfectionUC-[123]
CD64WT, CD64CK, TP53, and small hairpin KRASG12DTransfectionUC, UF, SECThe colocalization ratios of encapsulated TP53 mRNA or siKRASG12D and CD64CK surface protein within dtEV were ~ 60%[124]
Table 5

Applications of exogenous loading strategies for EVs

CargoLoading methodIsolation methodLoading efficiencyRef.
microRNAExtrusionUC, UF-[125]
ClodronateExtrusionUC-[126]
DapagliflozinExtrusionUC~ 45%[127]
PTXExtrusionUC, UF~ 14.23%[128]
GQDs/Cy5-miRSonicationUC5.6 times higher than the untreated sEVs[129]
Erastin and Rose BengalSonicationUC-[130]
PTXSonicationUC-[131]
Tripeptidyl peptidase-1SonicationUC-[132]
FXSonicationUC, UFThe loading rates of LpEVs-FX and GA-LpEVs-FX were approximately 69% and 86%[133]
miRNA-497,
DOX
Membrane PermeabilizationUC, UF4.3-fold and 7.2-fold greater than traditional methods[134]
miR-195-5pElectroporationUC-[135]
siRNAElectroporationUC-[136]
IL-10ElectroporationUC~ 70% [137]
DDHD1-siRNAElectroporationUC13%[138]
CCL2-siRNAElectroporationUC-[139]
miR-223ElectroporationUCaround 60%[140]
Ca(HCO3)2ElectroporationUC-[141]
DoxElectroporationUC-[142]
HALElectroporationUC~ 25.14%[143]
DoxElectroporationSEC, UF190-fold increased response compared to naked doxorubicin[144]
gRNA: Cas9 RNP complexesElectroporationUC~ 80%[145]
Zinc oxide NanocrystalsFreeze-Thaw
UC-[146]

MECHANISMS OF CANCER DRUG RESISTANCE

Drug resistance is a major challenge for effective cancer treatment. Depending on the treatment method, cancer resistance can be classified as chemotherapy, targeted therapy, immunotherapy, or endocrine therapy resistance. The mechanisms of cancer resistance include increased drug efflux, reduced drug uptake, drug inactivation, target mutations, signaling pathway alterations, apoptotic defects, phenotype switching, and resistance to immunotherapy.

The following sections introduce the specific mechanisms of cancer resistance and the methods to overcome them.

Increased drug efflux

Drug efflux refers to the process by which cells expel drug molecules from the inside to the outside through specific transport proteins on the membrane. These transport proteins primarily belong to the ATP-binding cassette (ABC) transporter family, including P-glycoprotein (P-gp), multidrug resistance-associated proteins (MRPs), breast cancer resistance protein (BCRP), cholesterol transport protein (ABCA1), and bile salt export pump (BSEP). They regulate drug distribution, absorption, and excretion, thereby protecting cells from death caused by high intracellular drug concentrations[1]. During tumor treatment, ABC transporters can reduce the effective concentration of drugs inside tumor cells, resulting in multidrug resistance (MDR).

In most cases, overcoming the resistance caused by increased drug efflux involves increasing drug concentration; however, this often leads to systemic toxicity[1]. The latest strategy involves collateral sensitivity (CS), in which cells exhibiting resistant phenotypes owing to ABC transporter overexpression are highly sensitive to certain compounds. This phenomenon is known as CS and can be considered a lethal weakness in cancer cells overexpressing ABC transporters[147]. Zhang et al. reported that using BAY-1082439 and CRISPR/Cas9 to knock out Pik3ca and Pik3cb can inhibit the activation of PI3K 110α and 110β catalytic subunits, downregulating ABC transporters P-gp/ABCB1 and BCRP/ABCG2, thereby re-establishing drug sensitivity in resistant epidermoid carcinoma and non-small cell lung cancer (NSCLC) cells[148]. They also discovered that CDK6 deletion altered ABCB1 levels, increased drug accumulation in tumors, significantly inhibited tumor growth and metastasis, and improved survival. Thus, the CDK6-PI3K pathway may serve as a new target for reversing ABCB1-mediated cancer resistance[149]. Additionally, computer-aided drug design, high-throughput screening, or structure-based drug design and synthesis can be used to develop chemotherapeutic adjuvant inhibitors of ABC transporters[150-153].

Reduced drug uptake

Similar to increased drug efflux, reduced drug uptake decreases the concentration of drug molecules in the cellular environment, thereby limiting their efficacy. Chemotherapeutic drugs such as methotrexate, 5-fluorouracil, 8-azaguanine, and cisplatin are most affected by this resistance mechanism. They use transport proteins, such as solute carrier (SLC) transporters, to enter cells. Downregulation of SLC transporter expression and function in tumor cells limits drug uptake, leading to reduced drug efficacy[154]. Overcoming this resistance mechanism involves maximizing the passive permeability of anticancer drugs or using alternative active transport pathways[3].

Nonselective upregulation of SLC transporters can lead to increased drug accumulation in normal cells. Therefore, it is crucial to develop drugs that selectively regulate the expression of SLC transporters in cancer cells[155,156]. Huttunen et al. showed that attacking several SLCs to alter the nutrient environment of cancer cells could serve as adjuvant therapy for other chemotherapeutic drugs, providing an alternative to ABC transporter inhibitors[157]. The poor prognosis of advanced metastatic differentiated thyroid cancer (DTC) is primarily due to reduced expression of the sodium/iodide symporter (NIS) or decreased targeting of NIS to the cell membrane, which reduces the efficacy of radioactive iodine therapy[158]. Ullmann et al. reported that dual treatment with BRAF and MEK inhibitors could upregulate NIS expression, increase iodine uptake in tumors, enhance efficacy, and overcome resistance[159].

Drug inactivation

After drugs enter the body and exert their effects, they are metabolized and excreted. Carrier molecules and enzymes responsible for drug metabolism play important roles in resistance to chemotherapy[1]. Glutathione (GSH) is involved in maintaining the cellular redox balance and can detoxify xenobiotics, enhancing MDR in cancer cells[1]. Reducing GSH levels is an effective strategy for increasing the sensitivity of cancer cells to chemotherapy[160]. Some drugs require enzymatic metabolism to become active, and mutations or downregulation of these enzymes can affect drug efficacy. For example, cytarabine (Ara-C) must be metabolized into the active substance Ara-C triphosphate. Mutations or downregulation of enzymes such as deoxycytidine kinase can lead to drug resistance in tumor cells[161].

The cytochrome P450 (CYP) family primarily participates in drug metabolism. Overexpression of these enzymes has been observed in drug-resistant tumor cells. Kawahara et al. discovered that overexpression of the CYP enzymes CYP3A4 and CYP2C8 in malignant breast cells leads to the metabolic inactivation of paclitaxel in liver tissue[162]. Cytochrome P450 1B1 (CYP1B1) is overexpressed in various solid tumors. Hachey et al. used scaffold hopping to explore the chemical space of CYP1B1 inhibitors and found a new lead compound that inhibited CYP1B1 in trace amounts, potentially restoring drug sensitivity in resistant cells[163].

Target mutation

Target mutations can lead to the development of resistance to therapeutic molecules[3]. Target mutations include steric impact on drug binding, impact on cofactor affinity, and conformational change of target.

Steric impact on drug binding

First, the substitution of smaller amino acids with larger ones increases steric hindrance and reduces the affinity of the drug molecules. Second, mutations of polar amino acid side chains to hydrophobic ones, or vice versa, can disrupt polar or nonpolar interactions and increase steric hindrance[3]. Abnormal activation of CDK9 is associated with the super-enhancer-mediated transcription of short-lived proteins required for cancer cell survival. Targeting CDK9 results in potent antitumor activity. Hu et al. showed that tumor cells develop resistance to CDK9 inhibitors due to the CDK9 L156F mutation, which disrupts the binding of the inhibitor to CDK9 through steric hindrance, thereby affecting the stability and catalytic activity of the CDK9 protein[164]. Targeting the proto-oncogene protein RET with small-molecule tyrosine kinase inhibitors (TKIs) is an effective treatment strategy for thyroid cancer. Meng et al. reported that mutations in the gatekeeper residues V804M and V804L of the RET kinase domain create steric hindrance, leading to TKI resistance[165].

Impact on cofactor affinity

If the mechanism of action of a drug involves competition with cofactors or substrates, target mutations that increase affinity for cofactors or substrates can lead to resistance[3]. Conversely, mutations that decrease drug-target affinity can also cause resistance. Neratinib, a HER2 tyrosine kinase inhibitor (TKI), is effective for the treatment of breast cancer. The HER3 E928G kinase domain mutation enhanced the affinity of HER2/HER3, reducing the binding of HER2 to neratinib, thereby causing resistance[166].

Conformational change of target

Mutations can cause resistance by inducing conformational changes that activate kinase mechanisms[3]. Most HER2 exon 20 insertions (ex20ins) are resistant to EGFR or pan-HER TKIs. Zhao et al. showed that ex20ins in HER2-mutant lung cancers lead to ligand-independent kinase activation by altering the conformational landscape of HER2 kinase and restricting its conformation in the active state. This mechanism explains the TKI[167].

Signaling pathway alterations

Tumor cells can achieve resistance to drug treatment by activating certain signaling pathways through specific gene changes[168].

BRAF inhibitors (BRAFis) are targeted drugs used to treat melanoma. Wang et al. showed that treatment with BRAFis increased the expression of IRF9 and STAT2 in melanoma cells. Overexpression of IRF9 or STAT2 slowed BRAFis-induced tumor shrinkage, while knockdown of IRF9 or STAT2 accelerated BRAFis-induced tumor shrinkage. IRF9-STAT2 signaling controls GSDME-dependent pyroptosis by restoring GSDME transcription. Targeting IRF9/STAT2 may prevent BRAFis resistance in melanoma by inducing pyroptosis[169]. Kobayashi et al. summarized the signaling pathways related to cancer immune evasion and immune checkpoint inhibitor (ICI) resistance[170].

Apoptotic defects

The goal of most cancer drugs is to induce tumor-selective cell death. Disruption of apoptotic mechanisms can lead to drug resistance, particularly in cases where chemotherapy resistance is related to defects in cell death mechanisms[3].

The first-line treatment for most hematological malignancies (HMs) relies primarily on cytotoxic agents. However, HM recurrence is often associated with defects in the DNA damage response (DDR) pathways and anti-apoptotic blocks. BH3 mimetics represent a new class of pro-apoptotic anticancer drugs that can directly target the mitochondria, independent of TP53 gene aberrations. These drugs effectively clear non-dividing malignant cells with adverse molecular cytogenetic alterations[171]. Treatment of prostate cancer (PCa) primarily inhibits tumor growth by inducing apoptosis. However, defects in the apoptotic response frequently lead to resistance. Therefore, triggering non-apoptotic cell death may be an alternative strategy for preventing cancer resistance. Montagnani Marelli et al. found that the combination of δ-tocotrienol (δ-TT) and docetaxel (DTX) enhanced DTX cytotoxicity in DU145 cells. In addition, δ-TT induced necrotic apoptosis in DTX-resistant cells[172].

Phenotype switching

Phenotype switching refers to the ability of cells to change among various forms, allowing them to adapt to environmental changes. This switching can serve as a drug resistance mechanism independent of drug-target pathways. The molecular mechanisms underlying cancer cell phenotype remodeling include both endogenous and exogenous factors. Endogenous mechanisms involve changes in epigenetic modifications and the expression of related transcription factors, whereas exogenous mechanisms are primarily related to changes in the tumor microenvironment (TME), such as hypoxia, infiltration of tumor-promoting immune cells, and the release of pro-inflammatory factors[173].

The phenotypic transition from EGFR-mutant lung adenocarcinoma (LUAD) to small cell lung cancer (SCLC) is considered a clinical mechanism of resistance to EGFR inhibitors[3]. It has been reported that EHMT2 participates in SCLC transformation and EGFR-TKI resistance through enzyme activity-dependent mechanisms[174]. EHMT2 affects the accumulation of H3K9me2 in the promoter region of SFRP1, thereby remodeling chromatin structure and inhibiting the expression of the WNT/β-catenin pathway negative regulator SFRP1. This abnormal activation of the WNT/β-catenin pathway in SCLC-transformed cells ultimately drives the neuroendocrine transformation of NSCLC to SCLC.

Immunotherapy resistance

The mechanisms of resistance to cancer immunotherapy involve interactions among tumor cells, the TME, and the host immune system. These resistance mechanisms can be divided into several major categories:

Tumor Cell Evasion

Initially, tumor cells overexpress immune checkpoint molecules such as PD-L1 (programmed death-ligand 1), TIM-3 (T-cell immunoglobulin and mucin-domain containing-3), LAG-3 (lymphocyte-activation gene 3), and VISTA (V-domain Ig suppressor of T cell activation), which inhibit T cell activation by binding to their respective receptors[175]. Concurrently, genetic mutations, like those in the β-2 microglobulin (B2M) gene, reduce MHC-I expression, preventing effective antigen presentation and T cell recognition[176,177]. To ensure survival once recognized, tumor cells upregulate anti-apoptotic proteins such as BCL-2, making them resistant to immune-mediated apoptosis[178]. Additionally, tumor cells leverage epigenetic modifications - such as DNA methylation - to regulate tumor antigens and immune-related gene expression, dynamically adapting to immune pressures[179]. Furthermore, tumor cell plasticity, including epithelial-mesenchymal transition (EMT), allows them to alter their phenotype to evade immune attack, and tumor heterogeneity creates distinct subpopulations, complicating treatment due to differential responses to therapy[180]. Together, these mechanisms form a multi-layered defense strategy that enables tumor cells to escape immune detection, resist treatment, and ultimately contribute to therapeutic failure.

Abnormal Tumor Vasculature

Tumor vessels are often irregular and tortuous, resulting in unstable blood flow and poor perfusion, effectively limiting immune cell infiltration[181]. Additionally, the abnormal structure of tumor vasculature leads to hypoxia, which suppresses effector immune cell activity and drives the production of immunosuppressive factors such as VEGF, IL-10 (interleukin-10), and TGF-β (transforming growth factor-beta), while also promoting PD-L1 upregulation, thereby facilitating immune evasion[182]. Furthermore, plasma leakage from these abnormal vessels creates high interstitial pressure, forming a physical barrier that further hinders immune cell migration and reduces immunotherapy effectiveness[183].

Immunosuppressive TME

The TME often contains a high concentration of immunosuppressive cells, such as regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), and tumor-associated macrophages (TAMs), which secrete inhibitory cytokines (e.g., TGF-β and IL-10)[184]. Moreover, metabolic products like lactate and adenosine accumulate in the TME, further inhibiting immune cell activity, while hypoxia promotes PD-L1 expression, reducing the immune system’s ability to recognize and attack tumor cells[185].

T Cell Dysfunction

T cells can become exhausted after prolonged antigen exposure, characterized by the overexpression of inhibitory receptors and decreased secretion of effector cytokines. Additionally, some tumors inhibit the formation of memory T cells, increasing the likelihood of relapse after initial treatment[186,187].

ENGINEERING EVS AS DRUG CARRIERS TO OVERCOME DRUG RESISTANCE

Drug resistance is closely related to tumor recurrence and mortality, posing the greatest obstacle to clinical cancer treatment. Therefore, developing effective strategies to overcome cancer resistance is crucial. This section focuses on the progress in the use of EVs as drug carriers to overcome resistance to cancer treatments, including chemotherapy, targeted therapy, immunotherapy, and endocrine therapy.

Overcoming chemotherapy resistance

Studies have shown that EVs can improve the efficacy of chemotherapy by delivering chemotherapeutic drugs, RNA, or other biomolecules to regulate cancer chemotherapy resistance [Table 6].

Table 6

EVs in overcoming cancer chemotherapy resistance

EVs sourceCargo carriedDrugMechanismsRef.
Melanoma cellssurvivin-T34AGemcitabineBlock Survivin, induce caspase activation and apoptosis[188]
AMSCsmiR-199aDOXInhibit mTOR activation and the phosphorylation of 4EBP1 and 70S6K[189]
BM-MSCsmiR-1245-FUSuppress cell viability, invasion, and migration, as well as inducing cell apoptosis in pancreatic cancer by regulating EZH2[190]
MSCsmiR-1236DDPSLC9A1 downregulation and Wnt/β-catenin inactivation[191]
hUCMSCsmiR-146aDTX, TAXReduce LAMC2 expression via the PI3K/AKT signaling pathway[192]
BMSCsmiR-193aDDPSuppress the colony formation, invasion, migration, and proliferation, as well as advancing apoptosis by downregulating LRRC1[193]
MSCsanti-miR-9TMZDecrease the expression of the drug transporter gene MDR1[194]
HEK293T cellslncRNA DARS-AS1 siRNADOXSuppress TGF-β/Smad3 signaling pathway-induced autophagy[18]
hMSCsmiR-199aTMZDownregulation of AGAP2[195]
WJ-MSCsmiR-124TMZ-[196]
HEk293T cellssi-c-MetCisplatinSi-c-Met inhibited migration, invasion and promoted apoptosis in vitro[20]
MDA-MA-231, MCF-7, 4T1 cellssiMTA1GemcitabineEnhance the sensitivity of MCF-7 to gemcitabine and weaken the metastasis ability of MCF-7[197]
HCT116, SW480 cellsCPT1A siRNAOxaliplatinSilence CPT1A by siRNA or etomoxir, a specific small-molecule inhibitor of CPT1A[198]
TMZ-resistant cellsmiR-151aTMZRestore miR-151a expression sensitized TMZ-resistant GBM cells via inhibiting XRCC4-mediated DNA repair[199]
HEK293T, HUVEC, OVCAR-3 cellsmiR-484Cisplatininduce vessel normalization[200]
HEK293T cellsanti-miR-214CisplatinDownregulate miR-214 and overexpress possible target proteins[201]
HEK293T cellsmiR-374a-5p inhibitorOxaliplatinInhibit MiR-374a-5p[202]
UPCI-SCC-131 cellsmiR-155 inhibitorCisplatinUpregulate FOXO3a and inhibit drug efflux transporter expression[203]
4T1 cellsanti-miR-21DOXBlock the function of endogenous carcinogenic miR-21[204]
293T cellsPGM5-AS1OxaliplatinPrevent proliferation, migration, and acquired oxaliplatin tolerance[205]
FHCscirc-FBXW7OxaliplatinExosome mediated circ-FBXW7 transfer enhances oxaliplatin sensitivity by binding to miR-18b-5p[206]
BM-MSCsPTX, GEMPPaclitaxel, GemcitabineOvercome the restrictions of pathological ECM and enhance the accumulation of therapeutics in tumor[207]
Mononuclear M1 and M2 macrophagesCisplatinCisplatinChemotherapy drugs preferentially accumulate in cancer cells and M1 macrophages have the effect of killing tumor[208]
SKOV3-CDDP cellsmiR497、TPCisplatinInhibit the PI3K/AKT/mTOR signaling pathway, deplete GSH, elevate intracellular ROS, and regulate macrophage polarization[209]
HEK293T cellsDOXDOXReduce P-gp expression in drug-resistant MCF7/ADR cells[210]
FibroblastsCD47
GM-CSF
DTX
DTXImprove drug delivery efficiency and enhance the immune response in the tumor microenvironment[211]
THLG-293T, LG-293T cells5-FU, miR-21i5-FUInduce cell cycle arrest, reduce tumor proliferation, increase apoptosis, inhibit the tumor cell migration, and suppress PTEN and hMSH2 expressions[212]
RAW 264.7 macrophagesPTXPTXIncrease PTX solubility and overcome P-gp-mediated drug efflux[213]

Overcoming drug efflux-induced resistance

miR-9 is upregulated in temozolomide (TMZ)-resistant Glioblastoma (GBM) cells and is involved in the expression of the drug efflux transporter P-gp. Munoz et al. loaded anti-miR-9 into EVs derived from mesenchymal stem cells (MSCs), thereby reducing the expression of proteins associated with resistance[194]. Liu et al. inserted lipidomimetic chain-conjugated HA (lipHA) into the membranes of EVs[210]. As lipHA-hEVs can reduce P-gp expression in resistant MCF7/ADR cells, loading doxorubicin onto lipHA-hEVs effectively inhibited its efflux, thus overcoming doxorubicin resistance[210]. Treatment of metastatic colorectal cancer (mCRC) is inevitably affected by oxaliplatin resistance. Studies have shown that EV-delivered circular RNA F-box and WD repeat domain-containing 7 (circ-FBXW7) can bind to miR-128-5p and restore oxaliplatin sensitivity in resistant CRC cells by inhibiting drug efflux, thus providing a novel treatment strategy for patients with CRC with oxaliplatin resistance[206].

Overcoming reduced drug uptake-induced resistance

Studies have shown that loading cisplatin onto EVs derived from mononuclear M1 and M2 macrophages results in the preferential accumulation of the chemotherapeutic drug in tumors, significantly increasing its cytotoxicity against resistant A2780/DDP and A2780 cells[208]. Hyperthermic intraperitoneal chemotherapy (HIPEC) is a standard clinical treatment for metastatic peritoneal cancer. However, its efficacy is limited by low drug penetration efficiency and the rapid development of resistance. Lv et al. prepared genetically engineered exosome-thermosensitive liposome hybrid NPs (gETL NPs) to enhance drug delivery efficiency and overcome drug resistance[211]. gETL NPs effectively penetrated tumor tissue after intravenous injection and released DTX under the low-temperature conditions of HIPEC, increasing drug concentration and efficacy in tumors. In addition, CD47 molecules on the surface of gETL NPs bound to the signal regulatory protein alpha receptor (SIRP𝛼) on macrophages, preventing tumor cells from escaping the immune system, thus enhancing macrophage phagocytosis of tumor cells. Furthermore, gETL NPs delivered GM-CSF (granulocyte-macrophage colony-stimulating factor), promoting the polarization of macrophages from the M2 type to the M1 type, further enhancing antitumor capability. By improving drug delivery efficiency and enhancing the immune response in the TME, gETL NPs effectively inhibited tumor growth and reduced resistance.

Overcoming drug inactivation-induced resistance

The primary reasons for gemcitabine resistance in pancreatic ductal adenocarcinoma (PDAC) are decreased activity of gemcitabine metabolism-limiting enzymes and insufficient conversion of gemcitabine phosphate products. Studies have shown that loading PTX, gemcitabine monophosphate (GEMP), and the intermediate products of gemcitabine metabolism onto EVs can overcome chemotherapy resistance[207].

Overcoming signal pathway alteration-induced resistance

Studies have shown that EVs can deliver miR-199a to target the mTOR pathway, inhibiting mTOR activation and subsequent phosphorylation of 4EBP1 and 70S6K in hepatocellular carcinoma (HCC) cells, thus restoring doxorubicin sensitivity in HCC cells[189]. Li et al. fused EVs from SKOV3-CDDP cells with liposomes modified with the tumor-targeting peptide cRGD and loaded them with triptolide (TP) and miR-497 to create miR-497/TP-HENPs[209]. These miR-497/TP-HENPs can overcome cisplatin resistance in tumor cells by inhibiting the PI3K/AKT/mTOR signaling pathway, increasing reactive oxygen species (ROS) production, and regulating macrophage polarization.

Overcoming apoptosis defect-induced resistance

GBM is a common primary malignant brain tumor in adults, and TMZ is the only chemotherapeutic drug with clear efficacy against GBM. However, TMZ resistance severely affects treatment outcomes. GBM resistance to TMZ is associated with the loss of miR-151a. Engineered EVs carrying miR-151a inhibited XRCC4-mediated DNA damage repair and restored TMZ sensitivity in GBM[199]. Defects in apoptosis in NSCLC lead to cisplatin resistance, which is an important cause of clinical treatment failure. Studies have shown that miR-193a promotes apoptosis in cisplatin-resistant NSCLC cells by downregulating LRRC1 and can also inhibit colony formation, invasion, proliferation, and migration. Wu et al. have developed EVs derived from human BM-MSCs loaded with miR-193a to reduce NSCLC resistance to cisplatin treatment[193]. In addition, the sensitivity of gastric cancer cells to cisplatin is closely linked to c-Met siRNA, with resistant cells exhibiting lower c-Met siRNA levels compared to sensitive cells. Researchers have used EVs derived from HEK293T cells to deliver c-Met siRNA, thereby enhancing the sensitivity of cisplatin-resistant gastric cancer cells by promoting apoptosis[20]. The apoptosis inhibitor protein survivin is considered an important factor in resistance to various cancer treatments. Using melanoma-derived EVs to deliver Survivin-T34A significantly induces caspase activation, enhances the cytotoxic effect of gemcitabine, and promotes pancreatic cancer cell apoptosis[188].

Overcoming targeted therapy resistance

Targeted antitumor drugs act specifically on tumor cells, minimizing effects on normal cells. This precision improves efficacy and reduces adverse reactions. However, long-term use of these drugs may lead to resistance. These mechanisms include genetically related resistance due to target mutations and non-genetic resistance due to the phenotypic remodeling of tumor cells under the influence of epigenetic factors or transcriptional regulation. EVs may be a new method and strategy for overcoming resistance to targeted therapy [Table 7].

Table 7

EVs in overcoming resistance to other therapies

TreatmentEVs sourceCargo carriedDrugMechanismsRef.
Targeted therapyHEK293T cells BCR-ABL siRNAIMInhibits BCR-ABL and improves the combination of the imatinib[214]
hUC-MSCs-IMPromotes cell viability inhibition and apoptosis and increases Bax expression and decreases Bcl-2 expression[215]
AMSCsmiR-122SorafenibDownregulates the expression of CCNG1, IGF1R, and ADAM10 genes, and upregulates the expression of Caspase 3 and Bax[19]
BM-MSCssiGRP78SorafenibInhibits the expression of GRP78 in sorafenib-resistant cancer cells[216]
HCC cellssiRNA-1SorafenibPrevents PRP19-mediated YBX1 ubiquitination and degradation in the nucleus[217]
ImmunotherapyB16F10 cellsPOM1, Metformin PD-1Increases the level of pro-inflammatory extracellular ATP to prevent the accumulation of immunosuppressive adenosine and alleviates hypoxia[218]
BM-MSCsGalectin-9 siRNA-Blocks galectin-9/dectin-1 axis to reverse immunosuppression[219]
EndocrinotherapyHUCMSCsmiR-let-7c-Antagonizes androgen receptor expression and activity by downregulating HMGA2, RAS, and Myc[220]

Overcoming target mutation-induced resistance

Imatinib (IM) is a selective Bcr-Abl inhibitor, and mutations in the BCR-ABL gene reduce its binding affinity for IM, resulting in acquired resistance. Bellavia et al. prepared EVs that can deliver BCR-ABL siRNA to CML cells and overcome targeted drug resistance by inhibiting Bcr-Abl[214].

Overcoming pathway alteration-induced resistance

circRNA-SORE is upregulated in sorafenib-resistant HCC cells. This prevents PRP19-mediated YBX1 degradation, affects the expression of downstream YBX1 target genes, and ultimately leads to sorafenib resistance. Tumor-derived exosomes delivering si-circRNA-SORE significantly increased the sensitivity of tumor cells to sorafenib treatment[217]. Sorafenib-resistant cancer cells overexpress GRP78 to a greater extent than sorafenib-sensitive cancer cells, making GRP78 a therapeutic target for HCC[221]. Li et al. prepared exosomes loaded with siGRP78 and observed that they inhibited GRP78 expression in sorafenib-resistant cancer cells. Therefore, the combination of these exosomes with sorafenib enhanced the inhibition of HCC cell growth and metastasis in vivo[216]. Studies have shown that EVs transfected with miR-122 reduce the resistance of HCC cells to sorafenib. This may be because miR-122-loaded EVs reduce the expression of CCNG1, IGF1R, and ADAM10 genes, upregulate the expression of the apoptosis-related genes Caspase 3 and Bcl-2-associated X protein (Bax), and increase HCC cell sensitivity to sorafenib[19].

Overcoming immunotherapy resistance

The advent of immunotherapy has brought cancer treatment into a new era[222]. However, the resulting resistance has also led to most patients with cancer not benefiting clinically. Researchers have used exosomes to overcome resistance to immunotherapy [Table 7].

Wu et al. used EVs derived from B16F10 cells to deliver POM1 (a CD39 antagonist) and metformin (an AMPK agonist) for cancer treatment. They demonstrated that this platform elevated extracellular ATP (eATP) levels, which triggered the activation of the P2X7-NLRP3-inflammasome to drive macrophage pyroptosis and potentiated the maturation and antigen capacity of DCs. This, in turn, enhances the cytotoxic function of T cells and NK cells, leading to a synergistic antitumor immune response that suppresses cancer progression, metastasis, and recurrence while overcoming anti-PD1 resistance[218]. A previous study showed that CD38 is highly expressed in HCC, and EVs/siCD38 inhibit CD38 enzyme activity, decrease adenosine production, and promote macrophage repolarization to the M1 type, thereby inhibiting HCC cell growth and metastasis in vitro and tumor growth in mice and overcoming PD-1/PD-L1 inhibitor resistance[223].

Overcoming endocrine therapy resistance

Endocrine therapy, also known as hormone therapy, is a method for treating cancer by slowing or stopping the growth of certain hormone-dependent cancer tissues. The most commonly targeted pathways are the estrogen and androgen signaling pathways, which play key roles in breast and prostate cancers, respectively[224]. The growth of PCa cells depends on androgens. Therefore, removing or inhibiting androgen activity, known as androgen deprivation therapy (ADT), is the primary approach to treating PCa. However, patients undergoing ADT eventually transition from castration-sensitive to castration-resistant PCa, leading to ADT resistance. Kurniawati et al. used MSCs loaded with lethal 7c (let-7c), which antagonizes androgen receptor (AR) expression and activity by downregulating HMGA2, RAS, and Myc, thereby overcoming ADT resistance[220].

CONCLUSION

EV-based drug delivery systems are promising and multifaceted for overcoming cancer drug resistance. As advancements in EV engineering and functionalization continue, these systems have the potential to enhance the precision and efficacy of cancer therapies by specifically targeting resistant cancer cells, thereby minimizing off-target effects and systemic toxicities. Integrating EV-based delivery with existing therapies can create synergistic effects and simultaneously overcome multiple resistance mechanisms. The unique ability of EVs to cross biological barriers such as the blood-brain barrier further expands their therapeutic potential, particularly for metastatic and hard-to-reach tumors. Continued improvements in EV isolation, characterization, and functionalization will enhance their therapeutic capabilities.

Despite the considerable potential of EV therapy in cancer treatment, its practical application faces significant obstacles. The high heterogeneity of EVs complicates standardization, as EVs from different sources vary widely in size, composition, and function, impairing efficacy assessment and reproducibility. Additionally, the lack of industrial standards for purification and stabilization remains a major barrier, with existing isolation methods being inefficient, costly, and unable to guarantee high purity or consistency. Moreover, verifying the therapeutic efficacy of EVs is still a big challenge due to the complex in vivo environment, which involves biological barriers, making precise control over drug delivery and efficacy evaluation difficult.

Nevertheless, ongoing technological advancements and research efforts present viable opportunities to overcome these challenges in EV therapy. Establishing standardized processes for EV preparation and purification, refining engineering modifications, and gaining deeper insights into in vivo delivery mechanisms are essential steps to improve the efficacy and reproducibility of EV-based treatments. In addition, harnessing the synergistic potential of EVs in combination with established therapies, such as immunotherapy and chemotherapy, may offer promising solutions to overcome cancer resistance. Although EV therapy holds substantial promise as a safe, effective, and personalized cancer treatment, its successful clinical application still needs significant research support and the establishment of appropriate regulatory standards.

DECLARATIONS

Authors’ contributions

Literature mining: EVs and their functionalization: Zheng L, Jia Z

Mechanisms of cancer drug resistance: Liang B, Chang R

Engineering EVs as drug carriers to overcome drug resistance: Wang Y, Zhu Y

Writing the draft: General aspects: Zheng L, Liang B

EVs and their functionalization: Zheng L, Jia Z

Mechanisms of cancer drug resistance: Liang B, Chang R

Engineering EVs as drug carriers to overcome drug resistance: Wang Y, Zhu Y

Formatting and bibliographic aspects: Fan J

Conceiving the structure and revising the manuscript: Zhang Z, Du B, Kong D

Availability of data and materials

Not applicable.

Financial support and sponsorship

This study was supported by grants from the National Natural Science Foundation of China (82204461, 82061148017), the NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine (2023QRECM05), and the State Key Laboratory of Drug Research (SKLDR-2024-KF-04).

Conflicts of interest

Kong D is an Editorial Board member of the Journal Cancer Drug Resistance, while the other authors have declared that they have no conflicts of interest.

Ethical approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Copyright

© The Author(s) 2024.

REFERENCES

1. Bukowski K, Kciuk M, Kontek R. Mechanisms of multidrug resistance in cancer chemotherapy. Int J Mol Sci 2020;21:3233.

2. Gottesman MM, Fojo T, Bates SE. Multidrug resistance in cancer: role of ATP-dependent transporters. Nat Rev Cancer 2002;2:48-58.

3. Ward RA, Fawell S, Floc'h N, Flemington V, McKerrecher D, Smith PD. Challenges and opportunities in cancer drug resistance. Chem Rev 2021;121:3297-351.

4. Roberts AW, Wei AH, Huang DCS. BCL2 and MCL1 inhibitors for hematologic malignancies. Blood 2021;138:1120-36.

5. Vogel A, Segatto O, Stenzinger A, Saborowski A. FGFR2 inhibition in cholangiocarcinoma. Annu Rev Med 2023;74:293-306.

6. Xiang S, Lu X. Selective type II TRK inhibitors overcome xDFG mutation mediated acquired resistance to the second-generation inhibitors selitrectinib and repotrectinib. Acta Pharm Sin B 2024;14:517-32.

7. Xiong L, Wei Y, Jia Q, et al. The application of extracellular vesicles in colorectal cancer metastasis and drug resistance: recent advances and trends. J Nanobiotechnology 2023;21:143.

8. Welsh JA, Goberdhan DCI, O'Driscoll L, et al; MISEV Consortium. Minimal information for studies of extracellular vesicles (MISEV2023): from basic to advanced approaches. J Extracell Vesicles 2024;13:e12404.

9. Li SR, Man QW, Gao X, et al. Tissue-derived extracellular vesicles in cancers and non-cancer diseases: present and future. J Extracell Vesicles 2021;10:e12175.

10. Li M, Fang F, Sun M, Zhang Y, Hu M, Zhang J. Extracellular vesicles as bioactive nanotherapeutics: an emerging paradigm for regenerative medicine. Theranostics 2022;12:4879-903.

11. Du S, Guan Y, Xie A, et al. Extracellular vesicles: a rising star for therapeutics and drug delivery. J Nanobiotechnology 2023;21:231.

12. Dixson AC, Dawson TR, Di Vizio D, Weaver AM. Context-specific regulation of extracellular vesicle biogenesis and cargo selection. Nat Rev Mol Cell Biol 2023;24:454-76.

13. Ming-Kun C, Zi-Xian C, Mao-Ping C, Hong C, Zhuang-Fei C, Shan-Chao Z. Engineered extracellular vesicles: a new approach for targeted therapy of tumors and overcoming drug resistance. Cancer Commun 2024;44:205-25.

14. Rädler J, Gupta D, Zickler A, Andaloussi SE. Exploiting the biogenesis of extracellular vesicles for bioengineering and therapeutic cargo loading. Mol Ther 2023;31:1231-50.

15. Luo R, Liu M, Tan T, et al. Emerging significance and therapeutic potential of extracellular vesicles. Int J Biol Sci 2021;17:2476-86.

16. Yang Q, Xu J, Gu J, et al. Extracellular vesicles in cancer drug resistance: roles, mechanisms, and implications. Adv Sci 2022;9:e2201609.

17. Shan C, Liang Y, Wang K, Li P. Mesenchymal stem cell-derived extracellular vesicles in cancer therapy resistance: from biology to clinical opportunity. Int J Biol Sci 2024;20:347-66.

18. Liu X, Zhang G, Yu T, et al. CL4-modified exosomes deliver lncRNA DARS-AS1 siRNA to suppress triple-negative breast cancer progression and attenuate doxorubicin resistance by inhibiting autophagy. Int J Biol Macromol 2023;250:126147.

19. Lou G, Song X, Yang F, et al. Exosomes derived from miR-122-modified adipose tissue-derived MSCs increase chemosensitivity of hepatocellular carcinoma. J Hematol Oncol 2015;8:122.

20. Zhang Q, Zhang H, Ning T, et al. Exosome-delivered c-met siRNA could reverse chemoresistance to cisplatin in gastric cancer. Int J Nanomedicine 2020;15:2323-35.

21. Liang Y, Duan L, Lu J, Xia J. Engineering exosomes for targeted drug delivery. Theranostics 2021;11:3183-95.

22. Wen J, Chen Y, Liao C, et al. Engineered mesenchymal stem cell exosomes loaded with miR-34c-5p selectively promote eradication of acute myeloid leukemia stem cells. Cancer Lett 2023;575:216407.

23. Jia G, Han Y, An Y, et al. NRP-1 targeted and cargo-loaded exosomes facilitate simultaneous imaging and therapy of glioma in vitro and in vivo. Biomaterials 2018;178:302-16.

24. Pan BT, Teng K, Wu C, Adam M, Johnstone RM. Electron microscopic evidence for externalization of the transferrin receptor in vesicular form in sheep reticulocytes. J Cell Biol 1985;101:942-8.

25. Dai W, Dong Y, Han T, et al. Microenvironmental cue-regulated exosomes as therapeutic strategies for improving chronic wound healing. NPG Asia Mater 2022;14:419.

26. Onukwugha NE, Kang YT, Nagrath S. Emerging micro-nanotechnologies for extracellular vesicles in immuno-oncology: from target specific isolations to immunomodulation. Lab Chip 2022;22:3314-39.

27. Panagopoulou MS, Wark AW, Birch DJS, Gregory CD. Phenotypic analysis of extracellular vesicles: a review on the applications of fluorescence. J Extracell Vesicles 2020;9:1710020.

28. Imanbekova M, Suarasan S, Lu Y, Jurchuk S, Wachsmann-Hogiu S. Recent advances in optical label-free characterization of extracellular vesicles. Nanophotonics 2022;11:2827-63.

29. Kolenc A, Maličev E. Current methods for analysing mesenchymal stem cell-derived extracellular vesicles. Int J Mol Sci 2024;25:3439.

30. Wei H, Chen J, Wang S, et al. A nanodrug consisting of doxorubicin and exosome derived from mesenchymal stem cells for osteosarcoma treatment in vitro. Int J Nanomedicine 2019;14:8603-10.

31. Wu Y, Deng W, Klinke DJ 2nd. Exosomes: improved methods to characterize their morphology, RNA content, and surface protein biomarkers. Analyst 2015;140:6631-42.

32. Welsh JA, Arkesteijn GJA, Bremer M, et al. A compendium of single extracellular vesicle flow cytometry. J Extracell Vesicles 2023;12:e12299.

33. Pospichalova V, Svoboda J, Dave Z, et al. Simplified protocol for flow cytometry analysis of fluorescently labeled exosomes and microvesicles using dedicated flow cytometer. J Extracell Vesicles 2015;4:25530.

34. Bağcı C, Sever-Bahcekapili M, Belder N, Bennett APS, Erdener ŞE, Dalkara T. Overview of extracellular vesicle characterization techniques and introduction to combined reflectance and fluorescence confocal microscopy to distinguish extracellular vesicle subpopulations. Neurophotonics 2022;9:021903.

35. Wang JM, Li YJ, Wu JY, et al. Comparative evaluation of methods for isolating small extracellular vesicles derived from pancreatic cancer cells. Cell Biosci 2021;11:37.

36. Teixeira-Marques A, Monteiro-Reis S, Montezuma D, et al. Improved recovery of urinary small extracellular vesicles by differential ultracentrifugation. Sci Rep 2024;14:12267.

37. Shi G, Yang X, Wang J, et al. Isolation of extracellular outer membrane vesicles (OMVs) from Escherichia coli using EVscore47 beads. Molecules 2024;29:1831.

38. Moccia V, Centelleghe C, Giusti I, et al. Isolation and characterization of cetacean cell-derived extracellular vesicles. Animals 2023;13:3304.

39. Lee Y, Ni J, Wasinger VC, Graham P, Li Y. Comparison study of small extracellular vesicle isolation methods for profiling protein biomarkers in breast cancer liquid biopsies. Int J Mol Sci 2023;24:15462.

40. Ciliberti MG, Santillo A, Sevi A, et al. First insight into extracellular vesicle-miRNA characterization in a sheep in vitro model of inflammation. Front Vet Sci 2023;10:1186989.

41. Vo N, Tran C, Tran NHB, et al. A novel multi-stage enrichment workflow and comprehensive characterization for HEK293F-derived extracellular vesicles. J Extracell Vesicles 2024;13:e12454.

42. Visan KS, Lobb RJ, Ham S, et al. Comparative analysis of tangential flow filtration and ultracentrifugation, both combined with subsequent size exclusion chromatography, for the isolation of small extracellular vesicles. J Extracell Vesicles 2022;11:e12266.

43. Mansur S, Habib S, Hawkins M, Brown SR, Weinman ST, Bao Y. Preparation of nanoparticle-loaded extracellular vesicles using direct flow filtration. Pharmaceutics 2023;15:1551.

44. Li Z, Liu C, Cheng Y, et al. Cascaded microfluidic circuits for pulsatile filtration of extracellular vesicles from whole blood for early cancer diagnosis. Sci Adv 2023;9:eade2819.

45. Sunkara V, Park J, Han J, et al. Exosome precipitation by ionic strength modulation: ExoPRISM. ACS Appl Mater Interfaces 2023;15:56807-19.

46. Morozumi M, Izumi H, Shimizu T, Takeda Y. Comparison of isolation methods using commercially available kits for obtaining extracellular vesicles from cow milk. J Dairy Sci 2021;104:6463-71.

47. Mladenović D, Khamari D, Kittel Á, Koort K, Buzás EI, Zarovni N. Acidification of blood plasma facilitates the separation and analysis of extracellular vesicles. J Thromb Haemost 2023;21:1032-42.

48. Zhang Z, Yu K, You Y, et al. Comprehensive characterization of human brain-derived extracellular vesicles using multiple isolation methods: implications for diagnostic and therapeutic applications. J Extracell Vesicles 2023;12:e12358.

49. Williams S, Jalal AR, Lewis MP, Davies OG. A survey to evaluate parameters governing the selection and application of extracellular vesicle isolation methods. J Tissue Eng 2023;14:20417314231155114.

50. Vychytilova-Faltejskova P, Vilmanova S, Pifkova L, et al. Optimized procedure for high-throughput transcriptome profiling of small extracellular vesicles isolated from low volume serum samples. Clin Chem Lab Med 2024;62:157-67.

51. Tengler L, Tiedtke M, Schütz J, et al. Optimization of extracellular vesicles preparation from saliva of head and neck cancer patients. Sci Rep 2024;14:946.

52. Soupir AC, Tian Y, Stewart PA, et al. Detectable lipidomes and metabolomes by different plasma exosome isolation methods in healthy controls and patients with advanced prostate and lung cancer. Int J Mol Sci 2023;24:1830.

53. Soares M, Pinto MM, Nobre RJ, et al. Isolation of extracellular vesicles from human follicular fluid: size-exclusion chromatography versus ultracentrifugation. Biomolecules 2023;13:278.

54. Northrop-Albrecht EJ, Taylor WR, Huang BQ, Kisiel JB, Lucien F. Assessment of extracellular vesicle isolation methods from human stool supernatant. J Extracell Vesicles 2022;11:e12208.

55. Martínez-Greene JA, Gómez-Chavarín M, Ramos-Godínez MDP, Martínez-Martínez E. Isolation of hepatic and adipose-tissue-derived extracellular vesicles using density gradient separation and size exclusion chromatography. Int J Mol Sci 2023;24:12704.

56. Zhang C, Huo X, Zhu Y, et al. Electrodeposited magnetic nanoporous membrane for high-yield and high-throughput immunocapture of extracellular vesicles and lipoproteins. Commun Biol 2022;5:1358.

57. Yang K, Jia M, Cheddah S, et al. Peptide ligand-SiO2 microspheres with specific affinity for phosphatidylserine as a new strategy to isolate exosomes and application in proteomics to differentiate hepatic cancer. Bioact Mater 2022;15:343-54.

58. Guo X, Hu F, Zhao S, Yong Z, Zhang Z, Peng N. Immunomagnetic separation method integrated with the Strep-tag II system for rapid enrichment and mild release of exosomes. Anal Chem 2023;95:3569-76.

59. Fortunato D, Giannoukakos S, Giménez-Capitán A, Hackenberg M, Molina-Vila MA, Zarovni N. Selective isolation of extracellular vesicles from minimally processed human plasma as a translational strategy for liquid biopsies. Biomark Res 2022;10:57.

60. Colella AP, Prakash A, Miklavcic JJ. Homogenization and thermal processing reduce the concentration of extracellular vesicles in bovine milk. Food Sci Nutr 2024;12:131-40.

61. Hartung F, Haimerl P, Schindela S, et al. Extracellular vesicle miRNAs drive aberrant macrophage responses in NSAID-exacerbated respiratory disease. Allergy 2024;79:1893-907.

62. Yung C, Zhang Y, Kuhn M, et al. Neonatal enteroids absorb extracellular vesicles from human milk-fed infant digestive fluid. J Extracell Vesicles 2024;13:e12422.

63. Fan W, Xu Z, Zhang J, et al. Naringenin regulates cigarette smoke extract-induced extracellular vesicles from alveolar macrophage to attenuate the mouse lung epithelial ferroptosis through activating EV miR-23a-3p/ACSL4 axis. Phytomedicine 2024;124:155256.

64. Hallal SM, Tűzesi Á, Sida LA, et al. Glioblastoma biomarkers in urinary extracellular vesicles reveal the potential for a ‘liquid gold’ biopsy. Br J Cancer 2024;130:836-51.

65. Pait MC, Kaye SD, Su Y, et al. Novel method for collecting hippocampal interstitial fluid extracellular vesicles (EV(ISF) ) reveals sex-dependent changes in microglial EV proteome in response to Aβ pathology. J Extracell Vesicles 2024;13:e12398.

66. Tominaga Y, Kawamura T, Ito E, et al. Pleiotropic effects of extracellular vesicles from induced pluripotent stem cell-derived cardiomyocytes on ischemic cardiomyopathy: a preclinical study. J Heart Lung Transplant 2024;43:85-99.

67. Kawano K, Kuzuma Y, Yoshio K, et al. Extracellular-vesicle catch-and-release isolation system using a net-charge invertible curvature-sensing peptide. Anal Chem 2024;96:3754-62.

68. Mishra A, Zehra S, Bharti PK, et al. Spectroscopic insight into breast cancer: profiling small extracellular vesicles lipids via infrared spectroscopy for diagnostic precision. Sci Rep 2024;14:9347.

69. Pecksen E, Tkachuk S, Schröder C, et al. Monocytes prevent apoptosis of iPSCs and promote differentiation of kidney organoids. Stem Cell Res Ther 2024;15:132.

70. Matchett EC, Kornbluth J. Extracellular vesicles derived from immortalized human natural killer cell line NK3.3 as a novel therapeutic for multiple myeloma. Front Immunol 2023;14:1265101.

71. Livkisa D, Chang TH, Burnouf T, et al. Extracellular vesicles purified from serum-converted human platelet lysates offer strong protection after cardiac ischaemia/reperfusion injury. Biomaterials 2024;306:122502.

72. Koksal AR, Ekmen N, Aydin Y, et al. A single-step immunocapture assay to quantify HCC exosomes using the highly sensitive fluorescence nanoparticle-tracking analysis. J Hepatocell Carcinoma 2023;10:1935-54.

73. Wang Z, Dai J, He H, et al. Cellulose nanofibrils of high immunoaffinity for efficient enrichment of small extracellular vesicles. Small Methods 2024;8:e2400426.

74. Kaur M, Fusco S, Van den Broek B, et al. Most recent advances and applications of extracellular vesicles in tackling neurological challenges. Med Res Rev 2024;44:1923-66.

75. Hanson B, Vorobieva I, Zheng W, et al. EV-mediated promotion of myogenic differentiation is dependent on dose, collection medium, and isolation method. Mol Ther Nucleic Acids 2023;33:511-28.

76. Zheng T, Hao H, Liu Q, et al. Effect of extracelluar vesicles derived from Akkermansia muciniphila on intestinal barrier in colitis mice. Nutrients 2023;15:4722.

77. Yang Z, Liang Z, Rao J, et al. Mesenchymal stem cell-derived extracellular vesicles therapy in traumatic central nervous system diseases: a systematic review and meta-analysis. Neural Regen Res 2023;18:2406-12.

78. de Pedro MÁ, López E, González-Nuño FM, et al. Menstrual blood-derived mesenchymal stromal cells: impact of preconditioning on the cargo of extracellular vesicles as potential therapeutics. Stem Cell Res Ther 2023;14:187.

79. Bathini S, Pakkiriswami S, Ouellette RJ, Ghosh A, Packirisamy M. Magnetic particle based liquid biopsy chip for isolation of extracellular vesicles and characterization by gene amplification. Biosens Bioelectron 2021;194:113585.

80. Chattrairat K, Yasui T, Suzuki S, et al. All-in-one nanowire assay system for capture and analysis of extracellular vesicles from an ex vivo brain tumor model. ACS Nano 2023;17:2235-44.

81. Zhang T, Xie Z, Zheng X, et al. CRISPR-Cas12a powered hybrid nanoparticle for extracellular vesicle aggregation and in-situ microRNA detection. Biosens Bioelectron 2024;245:115856.

82. Zhou X, Miao Y, Wang Y, et al. Tumour-derived extracellular vesicle membrane hybrid lipid nanovesicles enhance siRNA delivery by tumour-homing and intracellular freeway transportation. J Extracell Vesicles 2022;11:e12198.

83. Liu Q, Li D, Pan X, Liang Y. Targeted therapy using engineered extracellular vesicles: principles and strategies for membrane modification. J Nanobiotechnology 2023;21:334.

84. Hao D, Lu L, Song H, et al. Engineered extracellular vesicles with high collagen-binding affinity present superior in situ retention and therapeutic efficacy in tissue repair. Theranostics 2022;12:6021-37.

85. Fei Z, Zheng J, Zheng X, Ren H, Liu G. Engineering extracellular vesicles for diagnosis and therapy. Trends Pharmacol Sci 2024;45:931-40.

86. Ma Y, Brocchini S, Williams GR. Extracellular vesicle-embedded materials. J Control Release 2023;361:280-96.

87. Alvarez-Erviti L, Seow Y, Yin H, Betts C, Lakhal S, Wood MJ. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat Biotechnol 2011;29:341-5.

88. Tian Y, Li S, Song J, et al. A doxorubicin delivery platform using engineered natural membrane vesicle exosomes for targeted tumor therapy. Biomaterials 2014;35:2383-90.

89. Farahani MS, Hosseini-Beheshti E, Moazzeni SM, Moghadam MF. Engineered extracellular vesicles expressing ICAM-1: a promising targeted delivery system for T cell modifications. Biochim Biophys Acta Gen Subj 2024;1868:130541.

90. Cheng Q, Shi X, Han M, Smbatyan G, Lenz HJ, Zhang Y. Reprogramming exosomes as nanoscale controllers of cellular immunity. J Am Chem Soc 2018;140:16413-7.

91. Shi X, Cheng Q, Hou T, et al. Genetically engineered cell-derived nanoparticles for targeted breast cancer immunotherapy. Mol Ther 2020;28:536-47.

92. Liang G, Kan S, Zhu Y, Feng S, Feng W, Gao S. Engineered exosome-mediated delivery of functionally active miR-26a and its enhanced suppression effect in HepG2 cells. Int J Nanomedicine 2018;13:585-99.

93. Ran N, Gao X, Dong X, et al. Effects of exosome-mediated delivery of myostatin propeptide on functional recovery of mdx mice. Biomaterials 2020;236:119826.

94. Nonaka T. Application of engineered extracellular vesicles to overcome drug resistance in cancer. Front Oncol 2022;12:1070479.

95. Johnson V, Vasu S, Kumar US, Kumar M. Surface-engineered extracellular vesicles in cancer immunotherapy. Cancers 2023;15:2838.

96. Mohammadi AH, Ghazvinian Z, Bagheri F, Harada M, Baghaei K. Modification of extracellular vesicle surfaces: an approach for targeted drug delivery. BioDrugs 2023;37:353-74.

97. Wu P, Zhang B, Ocansey DKW, Xu W, Qian H. Extracellular vesicles: a bright star of nanomedicine. Biomaterials 2021;269:120467.

98. Tian T, Zhang HX, He CP, et al. Surface functionalized exosomes as targeted drug delivery vehicles for cerebral ischemia therapy. Biomaterials 2018;150:137-49.

99. S S, Camardo A, Dahal S, Ramamurthi A. Surface-functionalized stem cell-derived extracellular vesicles for vascular elastic matrix regenerative repair. Mol Pharm 2023;20:2801-13.

100. Nie W, Wu G, Zhang J, et al. Responsive exosome nano-bioconjugates for synergistic cancer therapy. Angew Chem Int Ed Engl 2020;59:2018-22.

101. Liu HM, Zhang Y. Folic acid-decorated astrocytes-derived exosomes enhanced the effect of temozolomide against glioma. Kaohsiung J Med Sci 2024;40:435-44.

102. Kooijmans SAA, Fliervoet LAL, van der Meel R, et al. PEGylated and targeted extracellular vesicles display enhanced cell specificity and circulation time. J Control Release 2016;224:77-85.

103. Kooijmans SA, Aleza CG, Roffler SR, van Solinge WW, Vader P, Schiffelers RM. Display of GPI-anchored anti-EGFR nanobodies on extracellular vesicles promotes tumour cell targeting. J Extracell Vesicles 2016;5:31053.

104. Wang Y, Chen X, Tian B, et al. Nucleolin-targeted extracellular vesicles as a versatile platform for biologics delivery to breast cancer. Theranostics 2017;7:1360-72.

105. Royo F, Cossío U, Ruiz de Angulo A, Llop J, Falcon-Perez JM. Modification of the glycosylation of extracellular vesicles alters their biodistribution in mice. Nanoscale 2019;11:1531-7.

106. Shou J, Li S, Shi W, et al. 3WJ RNA nanoparticles-aptamer functionalized exosomes from M2 macrophages target BMSCs to promote the healing of bone fractures. Stem Cells Transl Med 2023;12:758-74.

107. Nakase I, Futaki S. Combined treatment with a pH-sensitive fusogenic peptide and cationic lipids achieves enhanced cytosolic delivery of exosomes. Sci Rep 2015;5:10112.

108. Huang T, Sato Y, Kuramochi A, et al. Surface modulation of extracellular vesicles with cell-penetrating peptide-conjugated lipids for improvement of intracellular delivery to endothelial cells. Regen Ther 2023;22:90-8.

109. Lin SW, Tsai JC, Shyong YJ. Drug delivery of extracellular vesicles: preparation, delivery strategies and applications. Int J Pharm 2023;642:123185.

110. Zheng D, Ruan H, Chen W, et al. Advances in extracellular vesicle functionalization strategies for tissue regeneration. Bioact Mater 2023;25:500-26.

111. Roerig J, Schulz-Siegmund M. Standardization approaches for extracellular vesicle loading with oligonucleotides and biologics. Small 2023;19:e2301763.

112. Chen C, Li Y, Wang Q, Cai N, Wu L, Yan X. Single-particle assessment of six different drug-loading strategies for incorporating doxorubicin into small extracellular vesicles. Anal Bioanal Chem 2023;415:1287-98.

113. Song J, Song B, Yuan L, Yang G. Multiplexed strategies toward clinical translation of extracellular vesicles. Theranostics 2022;12:6740-61.

114. Chandler K, Millar J, Ward G, et al. Imaging of light-enhanced extracellular vesicle-mediated delivery of oxaliplatin to colorectal cancer cells via laser ablation, inductively coupled plasma mass spectrometry. Cells 2023;13:24.

115. Sancho-Albero M, Encabo-Berzosa MDM, Beltrán-Visiedo M, et al. Efficient encapsulation of theranostic nanoparticles in cell-derived exosomes: leveraging the exosomal biogenesis pathway to obtain hollow gold nanoparticle-hybrids. Nanoscale 2019;11:18825-36.

116. Faruque H, Choi ES, Kim JH, Kim E. Enhanced effect of autologous EVs delivering paclitaxel in pancreatic cancer. J Control Release 2022;347:330-46.

117. Yang X, Shi G, Guo J, Wang C, He Y. Exosome-encapsulated antibiotic against intracellular infections of methicillin-resistant Staphylococcus aureus. Int J Nanomedicine 2018;13:8095-104.

118. Chen SY, Chen YL, Li PC, et al. Engineered extracellular vesicles carrying let-7a-5p for alleviating inflammation in acute lung injury. J Biomed Sci 2024;31:30.

119. Zhang Y, Liu Z, Chopp M, et al. Small extracellular vesicles derived from cerebral endothelial cells with elevated microRNA 27a promote ischemic stroke recovery. Neural Regen Res 2025;20:224-33.

120. Qu M, Liu X, Wang X, Li Z, Zhou L, Li H. Palmitoylation of vacuole membrane protein 1 promotes small extracellular vesicle secretion via interaction with ALIX and influences intercellular communication. Cell Commun Signal 2024;22:150.

121. Nebogatova J, Härk HH, Puskar A, et al. A method for using cell-penetrating peptides for loading plasmid DNA into secreted extracellular vesicles. Biomolecules 2023;13:1751.

122. Zou A, Xiao T, Chi B, et al. Engineered exosomes with growth differentiation factor-15 overexpression enhance cardiac repair after myocardial injury. Int J Nanomedicine 2024;19:3295-314.

123. Wu Y, Yao X, Shi X, et al. Myeloma extracellular vesicle-derived RAGE increases inflammatory responses and myotube atrophy in multiple myeloma through activation of the TLR4/NF-κB p65 pathway. Apoptosis 2024;29:849-64.

124. Chiang CL, Ma Y, Hou YC, et al. Dual targeted extracellular vesicles regulate oncogenic genes in advanced pancreatic cancer. Nat Commun 2023;14:6692.

125. Wang X, Hu S, Zhu D, Li J, Cheng K, Liu G. Comparison of extruded cell nanovesicles and exosomes in their molecular cargos and regenerative potentials. Nano Res 2023;16:7248-59.

126. Sun L, Fan M, Huang D, et al. Clodronate-loaded liposomal and fibroblast-derived exosomal hybrid system for enhanced drug delivery to pulmonary fibrosis. Biomaterials 2021;271:120761.

127. Zhang W, Wang L, Guo H, Chen L, Huang X. Dapagliflozin-loaded exosome mimetics facilitate diabetic wound healing by HIF-1α-mediated enhancement of angiogenesis. Adv Healthc Mater 2023;12:e2202751.

128. Zhang L, Fan C, Hao W, et al. NSCs migration promoted and drug delivered exosomes-collagen scaffold via a bio-specific peptide for one-step spinal cord injury repair. Adv Healthc Mater 2021;10:e2001896.

129. Fu P, Guo Y, Luo Y, et al. Visualization of microRNA therapy in cancers delivered by small extracellular vesicles. J Nanobiotechnology 2023;21:457.

130. Du J, Wan Z, Wang C, et al. Designer exosomes for targeted and efficient ferroptosis induction in cancer via chemo-photodynamic therapy. Theranostics 2021;11:8185-96.

131. Wang P, Wang H, Huang Q, et al. Exosomes from M1-polarized macrophages enhance paclitaxel antitumor activity by activating macrophages-mediated inflammation. Theranostics 2019;9:1714-27.

132. Haney MJ, Klyachko NL, Harrison EB, Zhao Y, Kabanov AV, Batrakova EV. TPP1 delivery to lysosomes with extracellular vesicles and their enhanced brain distribution in the animal model of batten disease. Adv Healthc Mater 2019;8:e1801271.

133. Wu C, Xiang S, Wang H, et al. Orally deliverable sequence-targeted fucoxanthin-loaded biomimetic extracellular vesicles for alleviation of nonalcoholic fatty liver disease. ACS Appl Mater Interfaces 2024;16:9854-67.

134. Lee C, Kumar S, Park J, Choi Y, Clarissa EM, Cho YK. Tonicity-induced cargo loading into extracellular vesicles. Lab Chip 2024;24:2069-79.

135. Santos NL, Bustos SO, Reis PP, Chammas R, Andrade LNS. Extracellular vesicle-packaged miR-195-5p sensitizes melanoma to targeted therapy with kinase inhibitors. Cells 2023;12:1317.

136. Draguet F, Dubois N, Bouland C, et al. Extracellular vesicles derived from human umbilical cord mesenchymal stromal cells as an efficient nanocarrier to deliver siRNA or drug to pancreatic cancer cells. Cancers 2023;15:2901.

137. Kammala AK, Mosebarger A, Radnaa E, et al. Extracellular vesicles-mediated recombinant IL-10 protects against ascending infection-associated preterm birth by reducing fetal inflammatory response. Front Immunol 2023;14:1196453.

138. Rabienezhad Ganji N, Urzì O, Tinnirello V, et al. Proof-of-concept study on the use of tangerine-derived nanovesicles as siRNA delivery vehicles toward colorectal cancer cell line SW480. Int J Mol Sci 2023;25:546.

139. Rong Y, Wang Z, Tang P, et al. Engineered extracellular vesicles for delivery of siRNA promoting targeted repair of traumatic spinal cord injury. Bioact Mater 2023;23:328-42.

140. Liu W, Liu A, Li X, et al. Dual-engineered cartilage-targeting extracellular vesicles derived from mesenchymal stem cells enhance osteoarthritis treatment via miR-223/NLRP3/pyroptosis axis: toward a precision therapy. Bioact Mater 2023;30:169-83.

141. Zhang S, Liang Y, Ji P, et al. Truncated PD1 engineered gas-producing extracellular vesicles for ultrasound imaging and subsequent degradation of PDL1 in tumor cells. Adv Sci 2024;11:e2305891.

142. Zhu S, Huang H, Liu D, Wen S, Shen L, Lin Q. Augmented cellular uptake and homologous targeting of exosome-based drug loaded IOL for posterior capsular opacification prevention and biosafety improvement. Bioact Mater 2022;15:469-81.

143. Wu G, Zhang J, Zhao Q, et al. Molecularly engineered macrophage-derived exosomes with inflammation tropism and intrinsic heme biosynthesis for atherosclerosis treatment. Angew Chem Int Ed Engl 2020;59:4068-74.

144. Lennaárd AJ, Mamand DR, Wiklander RJ, El Andaloussi S, Wiklander OPB. Optimised electroporation for loading of extracellular vesicles with doxorubicin. Pharmaceutics 2021;14:38.

145. Pan X, Huang P, Ali SS, et al. CRISPR-Cas9 engineered extracellular vesicles for the treatment of dominant progressive hearing loss. bioRxiv 2023. Available from https://www.biorxiv.org/content/10.1101/2023.09.14.557853v2 [accessed 12 December 2024].

146. Dumontel B, Susa F, Limongi T, et al. Nanotechnological engineering of extracellular vesicles for the development of actively targeted hybrid nanodevices. Cell Biosci 2022;12:61.

147. Zattoni IF, Delabio LC, Dutra JP, et al. Targeting breast cancer resistance protein (BCRP/ABCG2): functional inhibitors and expression modulators. Eur J Med Chem 2022;237:114346.

148. Zhang L, Li Y, Wang Q, et al. The PI3K subunits, P110α and P110β are potential targets for overcoming P-gp and BCRP-mediated MDR in cancer. Mol Cancer 2020;19:10.

149. Zhang L, Li Y, Hu C, et al. CDK6-PI3K signaling axis is an efficient target for attenuating ABCB1/P-gp mediated multi-drug resistance (MDR) in cancer cells. Mol Cancer 2022;21:103.

150. Dakhlaoui I, Vahdati S, Maalej E, et al. Synthesis and biological assessment of new pyrimidopyrimidines as inhibitors of breast cancer resistance protein (ABCG2). Bioorg Chem 2021;116:105326.

151. Goebel J, Chmielewski J, Hrycyna CA. The roles of the human ATP-binding cassette transporters P-glycoprotein and ABCG2 in multidrug resistance in cancer and at endogenous sites: future opportunities for structure-based drug design of inhibitors. Cancer Drug Resist 2021;4:784-804.

152. Zhang H, Xu H, Ashby CR Jr, Assaraf YG, Chen ZS, Liu HM. Chemical molecular-based approach to overcome multidrug resistance in cancer by targeting P-glycoprotein (P-gp). Med Res Rev 2021;41:525-55.

153. da Silva Zanzarini I, Henrique Kita D, Scheiffer G, et al. Magnolol derivatives as specific and noncytotoxic inhibitors of breast cancer resistance protein (BCRP/ABCG2). Bioorg Chem 2024;146:107283.

154. Puris E, Fricker G, Gynther M. The role of solute carrier transporters in efficient anticancer drug delivery and therapy. Pharmaceutics 2023;15:364.

155. Brouwer KLR, Evers R, Hayden E, et al. Regulation of drug transport proteins-from mechanisms to clinical impact: a white paper on behalf of the international transporter consortium. Clin Pharmacol Ther 2022;112:461-84.

156. Zhou S, Shu Y. Transcriptional regulation of solute carrier (SLC) drug transporters. Drug Metab Dispos 2022;50:1238-50.

157. Huttunen J, Tampio J, Järvinen J, Montaser AB, Markowicz-Piasecka M, Huttunen KM. Amino acid derivative of probenecid potentiates apoptosis-inducing effects of vinblastine by increasing oxidative stress in a cancer cell-specific manner. Chem Biol Interact 2024;388:110833.

158. Oh JM, Ahn BC. Molecular mechanisms of radioactive iodine refractoriness in differentiated thyroid cancer: impaired sodium iodide symporter (NIS) expression owing to altered signaling pathway activity and intracellular localization of NIS. Theranostics 2021;11:6251-77.

159. Ullmann TM, Liang H, Moore MD, et al. Dual inhibition of BRAF and MEK increases expression of sodium iodide symporter in patient-derived papillary thyroid cancer cells in vitro. Surgery 2020;167:56-63.

160. Ogiwara H, Takahashi K, Sasaki M, et al. Targeting the vulnerability of glutathione metabolism in ARID1A-deficient cancers. Cancer Cell 2019;35:177-90.e8.

161. Momparler RL. Biochemical pharmacology of cytosine arabinoside. Med Pediatr Oncol 1982;10 Suppl 1:45-8.

162. Kawahara B, Faull KF, Janzen C, Mascharak PK. Carbon monoxide inhibits cytochrome P450 enzymes CYP3A4/2C8 in human breast cancer cells, increasing sensitivity to paclitaxel. J Med Chem 2021;64:8437-46.

163. Hachey AC, Fenton AD, Heidary DK, Glazer EC. Design of cytochrome P450 1B1 inhibitors via a scaffold-hopping approach. J Med Chem 2023;66:398-412.

164. Hu C, Shen L, Zou F, et al. Predicting and overcoming resistance to CDK9 inhibitors for cancer therapy. Acta Pharm Sin B 2023;13:3694-707.

165. Meng S, Wu H, Wang J, Qiu Q. Systematic analysis of tyrosine kinase inhibitor response to RET gatekeeper mutations in thyroid cancer. Mol Inform 2016;35:495-505.

166. Hanker AB, Brown BP, Meiler J, et al. Co-occurring gain-of-function mutations in HER2 and HER3 modulate HER2/HER3 activation, oncogenesis, and HER2 inhibitor sensitivity. Cancer Cell 2021;39:1099-114.e8.

167. Zhao S, Fang W, Pan H, et al. Conformational landscapes of HER2 Exon 20 insertions explain their sensitivity to kinase inhibitors in lung adenocarcinoma. J Thorac Oncol 2020;15:962-72.

168. Wood KC. Mapping the pathways of resistance to targeted therapies. Cancer Res 2015;75:4247-51.

169. Wang D, Fu Z, Gao L, et al. Increased IRF9-STAT2 signaling leads to adaptive resistance toward targeted therapy in melanoma by restraining GSDME-dependent pyroptosis. J Invest Dermatol 2022;142:2476-87.e9.

170. Kobayashi Y, Lim SO, Yamaguchi H. Oncogenic signaling pathways associated with immune evasion and resistance to immune checkpoint inhibitors in cancer. Semin Cancer Biol 2020;65:51-64.

171. Klener P, Sovilj D, Renesova N, Andera L. BH3 mimetics in hematologic malignancies. Int J Mol Sci 2021;22:10157.

172. Marelli M, Beretta G, Moretti RM. Necroptosis induced by delta-tocotrienol overcomes docetaxel chemoresistance in prostate cancer cells. Int J Mol Sci 2023;24:4923.

173. Shi ZD, Pang K, Wu ZX, et al. Tumor cell plasticity in targeted therapy-induced resistance: mechanisms and new strategies. Signal Transduct Target Ther 2023;8:113.

174. Yang C, Ma S, Zhang J, et al. EHMT2-mediated transcriptional reprogramming drives neuroendocrine transformation in non-small cell lung cancer. Proc Natl Acad Sci U S A 2024;121:e2317790121.

175. Qin S, Xu L, Yi M, Yu S, Wu K, Luo S. Novel immune checkpoint targets: moving beyond PD-1 and CTLA-4. Mol Cancer 2019;18:155.

176. Wang H, Liu B, Wei J. Beta2-microglobulin(B2M) in cancer immunotherapies: biological function, resistance and remedy. Cancer Lett 2021;517:96-104.

177. Lin W, Chen L, Zhang H, et al. Tumor-intrinsic YTHDF1 drives immune evasion and resistance to immune checkpoint inhibitors via promoting MHC-I degradation. Nat Commun 2023;14:265.

178. Joung J, Kirchgatterer PC, Singh A, et al. CRISPR activation screen identifies BCL-2 proteins and B3GNT2 as drivers of cancer resistance to T cell-mediated cytotoxicity. Nat Commun 2022;13:1606.

179. Zhong F, Lin Y, Zhao L, Yang C, Ye Y, Shen Z. Reshaping the tumour immune microenvironment in solid tumours via tumour cell and immune cell DNA methylation: from mechanisms to therapeutics. Br J Cancer 2023;129:24-37.

180. Lorenzo-Sanz L, Lopez-Cerda M, da Silva-Diz V, et al. Cancer cell plasticity defines response to immunotherapy in cutaneous squamous cell carcinoma. Nat Commun 2024;15:5352.

181. Guelfi S, Hodivala-Dilke K, Bergers G. Targeting the tumour vasculature: from vessel destruction to promotion. Nat Rev Cancer 2024;24:655-75.

182. Sattiraju A, Kang S, Giotti B, et al. Hypoxic niches attract and sequester tumor-associated macrophages and cytotoxic T cells and reprogram them for immunosuppression. Immunity 2023;56:1825-43.e6.

183. Zhang T, Jia Y, Yu Y, Zhang B, Xu F, Guo H. Targeting the tumor biophysical microenvironment to reduce resistance to immunotherapy. Adv Drug Deliv Rev 2022;186:114319.

184. Goswami S, Anandhan S, Raychaudhuri D, Sharma P. Myeloid cell-targeted therapies for solid tumours. Nat Rev Immunol 2023;23:106-20.

185. Li N, Kang Y, Wang L, et al. ALKBH5 regulates anti-PD-1 therapy response by modulating lactate and suppressive immune cell accumulation in tumor microenvironment. Proc Natl Acad Sci U S A 2020;117:20159-70.

186. Chow A, Perica K, Klebanoff CA, Wolchok JD. Clinical implications of T cell exhaustion for cancer immunotherapy. Nat Rev Clin Oncol 2022;19:775-90.

187. Andrews LP, Butler SC, Cui J, et al. LAG-3 and PD-1 synergize on CD8+ T cells to drive T cell exhaustion and hinder autocrine IFN-γ-dependent anti-tumor immunity. Cell 2024;187:4355-72.e22.

188. Aspe JR, Diaz Osterman CJ, Jutzy JM, Deshields S, Whang S, Wall NR. Enhancement of Gemcitabine sensitivity in pancreatic adenocarcinoma by novel exosome-mediated delivery of the Survivin-T34A mutant. J Extracell Vesicles 2014:3.

189. Lou G, Chen L, Xia C, et al. MiR-199a-modified exosomes from adipose tissue-derived mesenchymal stem cells improve hepatocellular carcinoma chemosensitivity through mTOR pathway. J Exp Clin Cancer Res 2020;39:4.

190. Xu Y, Liu N, Wei Y, et al. Anticancer effects of miR-124 delivered by BM-MSC derived exosomes on cell proliferation, epithelial mesenchymal transition, and chemotherapy sensitivity of pancreatic cancer cells. Aging 2020;12:19660-76.

191. Jia Z, Zhu H, Sun H, et al. Adipose mesenchymal stem cell-derived exosomal microRNA-1236 reduces resistance of breast cancer cells to cisplatin by suppressing SLC9A1 and the Wnt/β-catenin signaling. Cancer Manag Res 2020;12:8733-44.

192. Qiu L, Wang J, Chen M, Chen F, Tu W. Exosomal microRNA-146a derived from mesenchymal stem cells increases the sensitivity of ovarian cancer cells to docetaxel and taxane via a LAMC2-mediated PI3K/Akt axis. Int J Mol Med 2020;46:609-20.

193. Wu H, Mu X, Liu L, et al. Bone marrow mesenchymal stem cells-derived exosomal microRNA-193a reduces cisplatin resistance of non-small cell lung cancer cells via targeting LRRC1. Cell Death Dis 2020;11:801.

194. Munoz JL, Bliss SA, Greco SJ, Ramkissoon SH, Ligon KL, Rameshwar P. Delivery of functional anti-miR-9 by mesenchymal stem cell-derived exosomes to glioblastoma multiforme cells conferred chemosensitivity. Mol Ther Nucleic Acids 2013;2:e126.

195. Yu L, Gui S, Liu Y, et al. Exosomes derived from microRNA-199a-overexpressing mesenchymal stem cells inhibit glioma progression by down-regulating AGAP2. Aging 2019;11:5300-18.

196. Sharif S, Ghahremani MH, Soleimani M. Delivery of exogenous miR-124 to glioblastoma multiform cells by wharton’s jelly mesenchymal stem cells decreases cell proliferation and migration, and confers chemosensitivity. Stem Cell Rev Rep 2018;14:236-46.

197. Li P, Cao G, Huang Y, et al. siMTA1-loaded exosomes enhanced chemotherapeutic effect of gemcitabine in Luminal-b type breast cancer by inhibition of EMT/HIF-α and autophagy pathways. Front Oncol 2020;10:541262.

198. Lin D, Zhang H, Liu R, et al. iRGD-modified exosomes effectively deliver CPT1A siRNA to colon cancer cells, reversing oxaliplatin resistance by regulating fatty acid oxidation. Mol Oncol 2021;15:3430-46.

199. Zeng A, Wei Z, Yan W, et al. Exosomal transfer of miR-151a enhances chemosensitivity to temozolomide in drug-resistant glioblastoma. Cancer Lett 2018;436:10-21.

200. Zhao Z, Shuang T, Gao Y, et al. Targeted delivery of exosomal miR-484 reprograms tumor vasculature for chemotherapy sensitization. Cancer Lett 2022;530:45-58.

201. Wang X, Zhang H, Bai M, et al. Exosomes serve as nanoparticles to deliver anti-miR-214 to reverse chemoresistance to cisplatin in gastric cancer. Mol Ther 2018;26:774-83.

202. Ji R, Zhang X, Gu H, et al. Mir-374a-5p: a new target for diagnosis and drug resistance therapy in gastric cancer. Mol Ther Nucleic Acids 2019;18:320-31.

203. Sayyed AA, Gondaliya P, Mali M, et al. MiR-155 inhibitor-laden exosomes reverse resistance to cisplatin in a 3D tumor spheroid and xenograft model of oral cancer. Mol Pharm 2021;18:3010-25.

204. Bose RJC, Uday Kumar S, Zeng Y, et al. Tumor cell-derived extracellular vesicle-coated nanocarriers: an efficient theranostic platform for the cancer-specific delivery of anti-miR-21 and imaging agents. ACS Nano 2018;12:10817-32.

205. Hui B, Lu C, Wang J, et al. Engineered exosomes for co-delivery of PGM5-AS1 and oxaliplatin to reverse drug resistance in colon cancer. J Cell Physiol 2022;237:911-33.

206. Xu Y, Qiu A, Peng F, Tan X, Wang J, Gong X. Exosomal transfer of circular RNA FBXW7 ameliorates the chemoresistance to oxaliplatin in colorectal cancer by sponging miR-18b-5p. Neoplasma 2021;68:108-18.

207. Zhou Y, Zhou W, Chen X, et al. Bone marrow mesenchymal stem cells-derived exosomes for penetrating and targeted chemotherapy of pancreatic cancer. Acta Pharm Sin B 2020;10:1563-75.

208. Zhang X, Liu L, Tang M, Li H, Guo X, Yang X. The effects of umbilical cord-derived macrophage exosomes loaded with cisplatin on the growth and drug resistance of ovarian cancer cells. Drug Dev Ind Pharm 2020;46:1150-62.

209. Li L, He D, Guo Q, et al. Exosome-liposome hybrid nanoparticle codelivery of TP and miR497 conspicuously overcomes chemoresistant ovarian cancer. J Nanobiotechnology 2022;20:50.

210. Liu J, Ye Z, Xiang M, et al. Functional extracellular vesicles engineered with lipid-grafted hyaluronic acid effectively reverse cancer drug resistance. Biomaterials 2019;223:119475.

211. Lv Q, Cheng L, Lu Y, et al. Thermosensitive exosome-liposome hybrid nanoparticle-mediated chemoimmunotherapy for improved treatment of metastatic peritoneal cancer. Adv Sci 2020;7:2000515.

212. Liang G, Zhu Y, Ali DJ, et al. Engineered exosomes for targeted co-delivery of miR-21 inhibitor and chemotherapeutics to reverse drug resistance in colon cancer. J Nanobiotechnology 2020;18:10.

213. Kim MS, Haney MJ, Zhao Y, et al. Development of exosome-encapsulated paclitaxel to overcome MDR in cancer cells. Nanomedicine 2016;12:655-64.

214. Bellavia D, Raimondo S, Calabrese G, et al. Interleukin 3- receptor targeted exosomes inhibit in vitro and in vivo chronic myelogenous leukemia cell growth. Theranostics 2017;7:1333-45.

215. Liu Y, Song B, Wei Y, et al. Exosomes from mesenchymal stromal cells enhance imatinib-induced apoptosis in human leukemia cells via activation of caspase signaling pathway. Cytotherapy 2018;20:181-8.

216. Li H, Yang C, Shi Y, Zhao L. Exosomes derived from siRNA against GRP78 modified bone-marrow-derived mesenchymal stem cells suppress Sorafenib resistance in hepatocellular carcinoma. J Nanobiotechnology 2018;16:103.

217. Xu J, Ji L, Liang Y, et al. CircRNA-SORE mediates sorafenib resistance in hepatocellular carcinoma by stabilizing YBX1. Signal Transduct Target Ther 2020;5:298.

218. Wu L, Xie W, Li Y, et al. Biomimetic nanocarriers guide extracellular ATP homeostasis to remodel energy metabolism for activating innate and adaptive immunity system. Adv Sci 2022;9:e2105376.

219. Zhou W, Zhou Y, Chen X, et al. Pancreatic cancer-targeting exosomes for enhancing immunotherapy and reprogramming tumor microenvironment. Biomaterials 2021;268:120546.

220. Kurniawati I, Liu MC, Hsieh CL, Do AD, Sung SY. Targeting castration-resistant prostate cancer using mesenchymal stem cell exosomes for therapeutic microRNA-let-7c delivery. Front Biosci 2022;27:256.

221. Dauer P, Sharma NS, Gupta VK, et al. ER stress sensor, glucose regulatory protein 78 (GRP78) regulates redox status in pancreatic cancer thereby maintaining “stemness”. Cell Death Dis 2019;10:132.

222. Kalbasi A, Ribas A. Tumour-intrinsic resistance to immune checkpoint blockade. Nat Rev Immunol 2020;20:25-39.

223. Deng J, Ke H. Overcoming the resistance of hepatocellular carcinoma to PD-1/PD-L1 inhibitor and the resultant immunosuppression by CD38 siRNA-loaded extracellular vesicles. Oncoimmunology 2023;12:2152635.

224. Ranganathan P, Chakrabarty A, Hiscox S, Limaye AM, Vella V. Editorial: resistance to endocrine therapies in cancer. Front Endocrinol 2020;11:196.

Cite This Article

Review
Open Access
Overcoming drug resistance through extracellular vesicle-based drug delivery system in cancer treatment
Long Zheng, ... Dexin Kong

How to Cite

Zheng, L.; Chang, R.; Liang, B.; Wang, Y.; Zhu, Y.; Jia, Z.; Fan, J.; Zhang, Z.; Du, B.; Kong, D. Overcoming drug resistance through extracellular vesicle-based drug delivery system in cancer treatment. Cancer. Drug. Resist. 2024, 7, 50. http://dx.doi.org/10.20517/cdr.2024.107

Download Citation

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Simply select your manager software from the list below and click on download.

Export Citation File:

Type of Import

Tips on Downloading Citation

This feature enables you to download the bibliographic information (also called citation data, header data, or metadata) for the articles on our site.

Citation Manager File Format

Use the radio buttons to choose how to format the bibliographic data you're harvesting. Several citation manager formats are available, including EndNote and BibTex.

Type of Import

If you have citation management software installed on your computer your Web browser should be able to import metadata directly into your reference database.

Direct Import: When the Direct Import option is selected (the default state), a dialogue box will give you the option to Save or Open the downloaded citation data. Choosing Open will either launch your citation manager or give you a choice of applications with which to use the metadata. The Save option saves the file locally for later use.

Indirect Import: When the Indirect Import option is selected, the metadata is displayed and may be copied and pasted as needed.

About This Article

Special Issue

This article belongs to the Special Issue Microvesicle and Cancer Drug Resistance
© The Author(s) 2024. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, sharing, adaptation, distribution and reproduction in any medium or format, for any purpose, even commercially, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Data & Comments

Data

Views
85
Downloads
13
Citations
0
Comments
0
1

Comments

Comments must be written in English. Spam, offensive content, impersonation, and private information will not be permitted. If any comment is reported and identified as inappropriate content by OAE staff, the comment will be removed without notice. If you have any queries or need any help, please contact us at support@oaepublish.com.

0
Download PDF
Share This Article
Scan the QR code for reading!
See Updates
Contents
Figures
Related
Cancer Drug Resistance
ISSN 2578-532X (Online)

Portico

All published articles will preserved here permanently:

https://www.portico.org/publishers/oae/

Portico

All published articles will preserved here permanently:

https://www.portico.org/publishers/oae/