Open Access Article
This Open Access Article is licensed under a Creative Commons Attribution-Non Commercial 3.0 Unported Licence

Hybrid exosomes: a rising horizon for precision cancer therapy

Swarup Sonara, Asmit Dasa, Sidhanti Nyahatkarb, Rajib Dharc, Ketki Kaleled, Vinod R. M. T. Balasubramaniama, Ling Shing Wonge, Vinoth Kumarasamyf and Vetriselvan Subramaniyan*c
aJeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Subang Jaya 47500, Selangor, Malaysia. E-mail: swarup.sonar@monash.edu; asmit.das@monash.edu; vinod.balasubramaniam@monash.edu
bDepartment of Dentistry, VYWS Dental College & Hospital, Amravati, Maharashtra, India. E-mail: sidhantinyahatkar249@gmail.com
cDivision of Pharmacology, Sir Jeffrey Cheah Sunway Medical School, Faculty of Medical and Life Sciences, Sunway University, Bandar Sunway, 47500 Selangor Darul Ehsan, Malaysia. E-mail: rajib.d@imail.sunway.edu.my; vetris@sunway.edu.my
dDepartment of Oncology, Neuron Institute of Applied Research, Amravati, Maharashtra, India. E-mail: drketkikalele@gmail.com
eFaculty of Health and Life Sciences, INTI International University, Nilai, 71800, Malaysia. E-mail: lingshing.wong@newinti.edu.my
fDepartment of Parasitology and Medical Entomology, Faculty of Medicine, University Kebangsaan Malaysia, Jalan Yaacob Latif, 56000 Cheras, Kuala Lumpur, Malaysia. E-mail: vinoth@ukm.edu.my

Received 10th July 2025 , Accepted 15th December 2025

First published on 2nd January 2026


Abstract

Extracellular vesicles (EVs) are nanoscale vesicles, which show significant promise as biomarkers for cancer diagnosis and prognosis, by providing valuable information about cancer progression and treatment response. Their therapeutic potential (including their popular subset: exosomes) is significant, but challenges remain. These limitations with natural exosomes, necessitate innovative engineering strategies. However, current methods for engineering exosomes, such as chimeric and surface modifications, still need to be improved. A prominent issue is drug off-targeting, leading to ineffective treatment and side effects. To address these challenges, “hybrid exosomes” have been engineered by combining the inherent biocompatibility of natural exosomes with the versatility of synthetic nanoparticles. Cutting-edge design strategies for hybrid exosomes, such as bio-hybrid approaches, emphasize their superior drug loading capacity, and targeted delivery to tumor sites, resulting in minimized toxicity profiles. Furthermore, we showcase recent breakthroughs in leveraging hybrid exosomes for the effective delivery and cellular uptake of chemotherapeutic agents and immunotherapies, which offer significantly enhanced therapeutic outcomes in preclinical cancer models, with emerging clinical relevance. This review explores the evolving field of hybrid exosomes, a novel approach to cancer therapeutics and highlights their potential to overcome existing limitations in cancer treatment. Hybrid exosomes offer a transformative approach to cancer treatment, promising affordable and effective precision therapy with a significant impact on cancer therapeutics.


1 Introduction

The global shadow cast by cancer continues to lengthen, with the year 2020 alone witnessing a staggering 19.3 million new diagnoses and 10 million lives lost.1 Projections paint an even grimmer picture, forecasting a surge to 28.4 million cases by 2040, driven by a confluence of population growth and aging.2,3 These stark figures underscore the urgent imperative for a paradigm shift in our approach to cancer diagnosis and treatment.1,3,4 At the heart of cancer's devastating progression lies a complex interplay between malignant cells and their surrounding microenvironment.5,6 This intricate ecosystem, known as the tumor microenvironment,7,8 comprises of cellular and non-cellular components, including immune cells, fibroblasts, blood vessels, and the extracellular matrix.5,7 These components play a pivotal role to influence tumor growth, invasion, and metastasis.6,8,9 Emerging from the depths of this intricate interplay are exosomes,10 nanoscale vesicles secreted by cells, serving as key mediators of intercellular communication.11–13 These nano vesicles carry a diverse cargo of biomolecules, including proteins, lipids, and nucleic acids, deeply influencing the behavior of recipient cells.12,13 Tumor-derived exosomes, in particular, have revealed a darker side, implicated in promoting angiogenesis, suppressing immune surveillance, and facilitating metastasis14,15 key processes that drive cancer advancement.10 Yet, within these seemingly double-edged messengers lies a glimmer of hope. The unique molecular signatures of exosomes offer a source of potential biomarkers for early cancer diagnosis and prognosis16–18 potentially empowering clinicians to intervene at a stage when treatment is most likely to be effective.7,19 Recent advancements in exosome research have spurred the exploration and development of exosome-based therapeutics (a cell-free approach).9,20–22 Their inherent qualities – nanoscale size, biocompatibility, an ability to evade immune detection, and a remarkable capacity to traverse biological barriers – position them as ideal candidates for targeted drug delivery.23,24 However, natural exosomes are not without their limitations. Their drug-carrying capacity is often restricted, and off-target effects remain a concern, hindering their full therapeutic potential.25 Undeterred, scientists embarked on a quest to engineer exosomes. Despite the initial efforts in exosome engineering, including surface modifications and the creation of chimeric exosomes, certain limitations persisted. This spurred the development of “hybrid exosomes,” a new class of engineered exosomes designed to overcome these limitations and achieve enhanced therapeutic properties.26 These engineered vesicles represent a bold leap forward, merging the innate advantages of natural exosomes with the precision engineering of synthetic nanoparticles. This fusion of nature's ingenuity and human innovation aims to amplify drug loading capacity, enhance targeted delivery to malignant cells, and minimize the unwanted side effects of toxicity and immunogenicity.26 This ambitious endeavor is being pursued through a multifaceted approach. Top-down methods focus on meticulously sculpting nanovesicles, while bottom-up strategies center on designing hybrid exosomes with bespoke properties. Bio-hybrid techniques, a testament to the power of synergy, strive to seamlessly merge synthetic nanoparticles with natural exosomes, creating a new breed of therapeutic agents.27 As the global cancer burden continues its relentless ascent, the development of novel diagnostic and therapeutic strategies is no longer a matter of scientific curiosity but a humanitarian imperative. Exosomes have taken center stage, their potential as both biomarkers and therapeutic agents undeniable. Yet, it is in the realm of hybrid exosomes, where nature's ingenuity meets human innovation, that the brightest hope for conquering cancer may lie. These engineered marvels hold the potential to overcome the limitations of their natural counterparts, ushering in a new era of precision medicine where cancer treatment is tailored to the individual, minimizing suffering and maximizing treatment outcomes. This review explore cancer and tumor development interlink, dynamic therapeutic exosomes sources, hybrid exosomes, clinical trials and its future orientation.

2 Exosome biogenesis

Exosome biogenesis (Fig. 1) is a meticulously regulated cascade of pathways, molecular components and associated events in the cellular endomembrane system, leading to the formation of these nanoscale extracellular vesicles (30–120 nm approx.). The Endosomal Sorting Complex Required for Transport (ESCRT) pathway is pivotal in this process, comprising of several complexes: ESCRT-0, ESCRT-I, ESCRT-II, and ESCRT-III, alongside proteins such as VPS4 (the disassembly engine) and ALIX (versatile adaptor protein).28–30 Initially, ESCRT-0 identifies ubiquitinated cargo at the endosomal membrane, initiating the sorting process. This is followed by the sequential recruitment of ESCRT-I, -II, and -III, which facilitate membrane invagination and vesicle scission to produce intraluminal vesicles (ILVs).31,32 VPS4, an ATPase, disassembles ESCRT components, enabling their reuse after the formation of multivesicular bodies (MVBs) containing ILVs.28,31,33 Beyond the ESCRT pathway, ceramide-rich microdomains, potentially via nuclear envelope budding, contribute to the diverse routes of exosome biogenesis.34 Ceramide-dependent mechanisms induce membrane curvature and budding, while tetraspanin-enriched microdomains assist in cargo sorting.34–37 While membrane lipid microdomains, including specific components like SPFH proteins and caveolin, and heat shock proteins (HSP70/90) are implicated in exosome biogenesis, their precise roles require further investigation.35,37,38 Ultimately, MVBs can either fuse with lysosomes for degradation or with the plasma membrane to release ILVs as exosomes into the extracellular milieu.
image file: d5ra04927j-f1.tif
Fig. 1 Exosome biogenesis and its molecular cargos. (Reproduced with permission from ref. 167 Copyright @ 2021 American Chemical Society).

3 Exosome isolation and characterization

Exosome isolation (Fig. 2) hinges on exploiting the biophysical properties of these nanovesicles, leading to a diverse array of techniques.39–41 Differential ultracentrifugation, a cornerstone method, leverages sequential centrifugation steps at increasing gravitational forces (g) to pellet exosomes based on their sedimentation coefficient, effectively separating them from larger vesicles and cellular debris.15,41 Density gradient centrifugation refines this by layering a sucrose or iodixanol gradient, allowing exosomes to band at their buoyant density (1.13–1.18 g mL−1) for enhanced purity.42 Size-exclusion chromatography, on the other hand, separates based on the hydrodynamic radius, with smaller exosomes eluting later due to their restricted access to the porous stationary phase.43,44 Tangential flow filtration, scalable for large volumes, utilizes a membrane with defined pore sizes (typically 50–100 nm) to selectively retain exosomes while smaller molecules pass through.45,46 Ultrafiltration operates similarly but relies on centrifugal force to drive the sample through the membrane, making it ideal for small volumes.40,47 Precipitation-based methods, often commercially available kits, exploit the altered solubility of exosomes in the presence of water-excluding polymers like polyethylene glycol, leading to their precipitation.40,48 Immunoaffinity capture offers high specificity by employing antibodies immobilized on beads or surfaces to selectively bind exosomes expressing specific surface markers.49,50 Microfluidic isolation leverages miniaturized devices integrating various separation mechanisms, including filtration, affinity capture, and acoustic trapping, for high-throughput and automated isolation.47,51,52 Acoustic fluid handling manipulates exosomes based on their acoustic properties, using sound waves to focus and separate them.53 Magnetophoresis utilizes magnetic beads coated with antibodies against exosomal markers, allowing for efficient capture and release of exosomes through magnetic fields.54 Deterministic lateral displacement (DLD) exploits the size and deformability of exosomes, separating them based on their trajectory in microfluidic channels with asymmetrically arranged obstacles.55,56 Field-flow fractionation separates particles in a thin channel under an external field, such as a flow field or electric field, based on their differential migration.57–60 Exosome characterization relies on a diverse toolkit of techniques to elucidate their biophysical and biochemical properties.61 Nanoparticle tracking analysis visualizes and tracks the Brownian motion of individual exosomes, using light scattering to determine their size distribution and concentration.62,63 Dynamic light scattering also exploits light scattering but measures the fluctuations in scattered intensity due to Brownian motion, providing information on hydrodynamic size and polydispersity.60,64 Electron microscopy, encompassing transmission EM and scanning EM, offers high-resolution visualization of exosome morphology, size, and structure.61 Atomic force microscopy provides nanoscale topographical information by scanning a sharp tip over the exosome surface, revealing details about size, shape, and surface features.61,65 Western blotting, a staple technique, detects the presence and relative abundance of specific proteins in exosome lysates, confirming the presence of exosomal markers and target proteins.15,66 Flow cytometry, though challenging due to exosome size, can analyze individual exosomes for size, granularity, and surface marker expression using fluorescently labeled antibodies.48,67 Enzyme-linked immunosorbent assay quantifies specific proteins or other molecules in exosome samples using antibody-based detection, offering high sensitivity and specificity.41,61 Raman spectroscopy analyzes the inelastic scattering of light by molecules in exosomes, providing information about their biochemical composition and structure.68 Mass spectrometry identifies and quantifies proteins, lipids, and metabolites in exosome samples, offering a comprehensive molecular profile.57,69,70 RNA sequencing analyzes the RNA content of exosomes, revealing their mRNA, miRNA, and other RNA species, providing insights into their function and origin.61 Lipidomics focuses on characterizing the lipid composition of exosomes, providing insights into their membrane structure and function.69,71 Finally, Integrated Magnetic-Electrochemical Exosome (iMEX) sensor can be used for exosome characterization, specifically for quantifying exosome surface markers. This method is a rapid and sensitive approach that emphasizes the iMEX's ability to detect varying numbers of EVs spiked into human plasma, demonstrating its effectiveness in quantifying exosomes.61
image file: d5ra04927j-f2.tif
Fig. 2 Exosomes isolation and characterization method. Immunoaffinity enrichment. (a) Schematics of a microfluidic chip, microscopic view of the device: (b) Y-shaped injector, (c) serpentine fluidic mixer for immunomagnetic binding, (d) magnetic aggregates, and (e) bound EVs on immunomagnetic beads. Microfluidic device (f) scanning electron micrographs of magnetic microbeads after immunoaffinity capture, (g) image of the microfluidic iMER (immuno-magnetic exosome RNA analysis) prototype (for (a–g) reproduced with permission from ref. 61 Copyright @ 2018 American Chemical Society).

4 Role of exosomes in cancer

The tumor microenvironment (TME) encompasses an intricate network of cellular and non-cellular components that collectively foster cancer progression and metastasis.11,72,73 Hypoxic, or oxygen-deprived, conditions within the tumor niche are a hallmark feature, characterized by the insufficient blood supply and inadequate oxygen availability.74,75 In response to this hostile, nutrient-depleted environment, cancer cells exhibit remarkable adaptability. Activating various survival pathways and oncogenic signaling cascades, primarily mediated by hypoxia-inducible factors (HIFs).76–78 This allows the malignant cells to proliferate uncontrollably, evade cell death, and divert essential resources, such as oxygen and nutrients, for their rapid growth and expansion.77,79,80 Through this process, the cancer cells effectively hijack the body's natural homeostatic mechanisms, disrupting the delicate balance of the surrounding normal tissue and creating a permissive microenvironment that supports their malignant transformation and metastasis.74,81 Researchers also indicate that hypoxia drives the secretion of exosomes from tumor cells.82,83 These nanoscale vesicles, secreted by tumor cells, act as critical mediators of intercellular communication, which carry molecular signals that further alter the TME, promoting angiogenesis84 and tumorigenesis (Fig. 3).82,83,85
image file: d5ra04927j-f3.tif
Fig. 3 Role of tumor-derived exosomes in TME (reproduced with permission under Creative Commons CC BY 4.0 license from ref. 21 Copyright @ 2020 The Authors).

One crucial aspect of the TME is the process of angiogenesis, where new blood vessels are formed to support the rapidly dividing cancer cells and facilitate metastasis by creating pathways for cancer cells to enter the bloodstream. Interestingly, transformed cancer cells can exhibit characteristics of pericytes, cells that typically surround and support blood vessels. These cancer cells can cluster around blood vessels, influencing tumor development and metastasis. This intricate interplay between angiogenesis, pericyte-like cancer cells, and the TME highlights the complexity of tumor progression.86 Exosomes, nanoscale vesicles, play a pivotal role in mediating intercellular communication within the TME.87 Tumor-derived exosomes (TEXs) can promote angiogenesis by transferring pro-angiogenic factors and microRNAs to endothelial cells, enhancing their proliferation and migration.72,87,88 For instance, exosomes from hypoxic tumor cells have been shown to contain elevated levels of miR-210, which promotes angiogenesis by targeting genes involved in endothelial cell function.89,90 TEXs exhibit a multifaceted influence on the tumor microenvironment, profoundly impacting immune cell behavior and ultimately facilitating immune evasion.91–93 These nanoscale vesicles, orchestrate a complex interplay between tumor cells and various immune cell populations. One of the key mechanisms by which exosomes contribute to tumor progression is by inducing a shift in the balance of immune cells towards an immunosuppressive state.91,92,94 TEXs have been shown to reprogram macrophages to promote cancer development.95,96 For instance, exosomal miR-934 has been shown to induce a shift in macrophages towards the pro-tumorigenic M2 phenotype, contributing to a tumor-supportive microenvironment that facilitates metastasis to the liver.96 This polarization is associated with enhanced angiogenesis, moreover, M2 macrophages suppress anti-tumor immune responses, creating a permissive environment for tumor progression.95,97 Beyond their influence on macrophages, TEXs can also promote the differentiation of monocytes into immunosuppressive myeloid-derived suppressor cells (MDSCs), further inhibiting T cell responses.98 TEXs achieve this by transferring signaling molecules, including proteins like prostaglandin E2 (PGE2) and transforming growth factor beta (TGF-β), and miRNAs such as miR-21, miR-10a, miR-494-3p, and miR-1260a, to recipient monocytes. These molecules activate downstream signaling pathways, including STAT3 and MyD88, leading to the differentiation of monocytes into MDSCs.98–101 Consequently, MDSCs expressing high levels of immunosuppressive molecules like Arg1, IL-6, VEGF, and Cox2 accumulate within the tumor microenvironment.98,99 Tumor-derived exosomes directly suppress the cytotoxic activity of T cells and natural killer cells, contributing to immune evasion. This is achieved through exosomal cargo such as TGF-β, a potent immunosuppressive cytokine, which inhibits T cell activation and promotes regulatory T cell differentiation, and programmed death-ligand 1 (PD-L1), which engages with programmed cell death protein 1 (PD-1) on T cells, leading to T cell exhaustion and suppression of anti-tumor immunity.102 Dendritic cells are pivotal in initiating immune responses. However, exosomes derived from tumors can convert DCs into tolerogenic cells, which fail to activate T cells effectively. This conversion is often mediated by exosomal TGF-β, which enhances the expression of inhibitory receptors on DCs, leading to a diminished anti-tumor response.102 This intricate interplay between tumor cells, exosomes, and immune cells highlights the key mechanisms by which tumors evade immune surveillance, ultimately promoting their growth and facilitating metastasis.91,95,98,102 Research suggests exosomes to be critically involved in the cascade of metastasis, a hallmark of cancer progression, by facilitating epithelial-mesenchymal transition (EMT), a process where epithelial cells lose their polarity and cell-to-cell adhesion, acquiring mesenchymal properties that enhance their migratory and invasive capabilities. TEXs can transfer bioactive molecules, including microRNAs (miRNAs), to recipient cells within the TME, promoting EMT and thereby facilitating metastasis.103 For instance, exosomes released from tumor cells can facilitate colorectal cancer metastasis by influencing the interaction between cancer cells undergoing epithelial-mesenchymal transition and M2-subtype tumor-associated macrophages (TAMs). Specifically, exosomal microRNA-106b-5p has been implicated in activating this cross-talk, promoting a tumor-supportive microenvironment that enhances the metastatic potential of colorectal cancer cells.104 Furthermore, exosomes contribute to organ-specific metastasis, a phenomenon where tumor cells exhibit a predilection for metastasizing to specific organs.105,106 Exosomal surface proteins like tetraspanins and integrins play a crucial role in cancer progression via metastasis and organotropism.105,107 These surface proteins, including integrins α6β4 and αvβ5, acting as adhesion molecules, are selectively packaged into exosomes and mediate the interaction between circulating tumor cells and their target microenvironments.107 This interaction is based on the specific binding affinity of these integrins to ligands expressed on the target cells. For instance, exosomes from lung-tropic tumor cells, enriched with specific integrins, preferentially bind to and fuse with lung fibroblasts and epithelial cells. Similarly, liver-tropic exosomes, carrying a different set of integrins, interact with Kupffer cells in the liver.107 This selective adhesion, guided by the integrin expression patterns on exosomes, not only facilitates the anchoring of tumor cells to the new site but also triggers intracellular signaling cascades.105,107 One such pathway involves the activation of Src kinase, a key regulator of cell survival, proliferation, and migration. The phosphorylation of Src, initiated by integrin engagement, promotes the survival and proliferation of tumor cells within the new environment.105,107,108 This mechanism highlights the crucial role of exosomal integrins in orchestrating the metastatic process and underscores their potential as biomarkers for predicting organ-specific metastasis. Adding to the complexity of cancer progression, exosomes also contribute to therapeutic resistance (Fig. 4), either before treatment begins or emerging over time, a major obstacle in cancer treatment.109,110 This resistance to therapy is a major contributor to the global cancer burden, contributing to staggering numbers of deaths.1,111 Tumor-derived exosomes can transfer drug resistance genes and proteins, such as those conferring resistance to paclitaxel, enabling recipient cells to evade the effects of chemotherapy or targeted therapies. For example, exosomes from cisplatin-resistant lung cancer cells can transfer miRNA-100-5p, which alters mTOR signaling and enhances survival under chemotherapy.110,112 Additionally, exosomal proteins like P-glycoprotein 1 (P-gp), a permeability glycoprotein, contribute to drug efflux mechanisms, where the uptake of P-gp-containing exosomes from resistant cells can induce resistance in sensitive cells.113,114 Exosomes released from breast cancer cells can contribute to resistance against HER2-targeted therapies through two primary mechanisms. First, exosomes overexpressing HER2 can directly bind to and sequester targeted drugs, reducing their efficacy. Second, these exosomes can reprogram the gene expression of recipient HER2-positive breast cancer cells, promoting a shift towards a HER2-independent phenotype that renders them less susceptible to HER2-targeted treatments.114,115 In conclusion, exosomes demonstrate a dual nature in the context of cancer, acting as both drivers of disease progression and potential sources of innovative therapeutic strategies.


image file: d5ra04927j-f4.tif
Fig. 4 Role of exosomes in therapeutic resistance (reproduced with permission under Creative Commons CC BY 4.0 license from ref. 169. Copyright @ 2019 The Authors).

5 Therapeutic exosomes

Exosomes, nanoscale vesicles secreted by cells majorly utilising the ESCRT pathway, are increasingly recognized for their superior therapeutic potential over conventional treatment modalities, demonstrating enhanced targeting efficacy and protected cargo delivery properties.116,117 Exosome-based therapeutic (Fig. 5) strategies represent an effective cancer nanomedicine.118 Naturally derived exosomes from various sources, such as milk, bacteria, mesenchymal stem cells, neurons, and even plants, have demonstrated remarkable potential in areas like cancer immunotherapy, regenerative medicine, and drug delivery.116,119 Simultaneously, the field has witnessed the advent of synthetically engineered173–175 or chimeric exosomes, which have been tailored to enhance targeting capabilities and improve cargo loading for gene therapy and other specialized applications.118,120 Stem cell derived exosomes a promising anti-cancer therapeutic tool.121 Another study demonstrated the potential of engineered macrophage-derived exosomes as a cancer immunotherapy.176 By using gamma irradiation, they engineered these exosomes to carry pro-inflammatory cytokines and tumor antigens. These engineered exosomes effectively repolarized M2 macrophages into the anti-tumor M1 phenotype, resulting in increased inflammatory mediator production and enhanced T cell activation. In vivo, these exosomes significantly inhibited tumor growth and improved survival in a mouse model of colon cancer, highlighting the potential of engineering macrophage-derived exosomes to enhance anti-tumor immune responses.122 Tumor-derived exosomes (TEXs) demonstrate a complex role in cancer immunology.123 They can act as potent messengers, carrying tumor-associated antigens to dendritic cells, key orchestrators of the immune response. This interaction promotes dendritic cell maturation and enhances their ability to activate T cells, leading to a targeted anti-tumor immune response.123 Pancreatic cancer-derived exosomes, abundant in tumor antigens, effectively activate CD8+ T cell responses via dendritic cell antigen presentation, highlighting their potential as an anti-tumor therapeutic strategy.124,125 Red blood cell-derived exosomes are emerging as a promising drug delivery platform. Their high biocompatibility, coupled with the ability to be engineered for targeted delivery, makes them attractive candidates.126 Drugs and therapeutic loaded and their surface markers facilitate binding to endothelial cells via mechanisms like receptor-mediated endocytosis, enhancing drug delivery efficiency.127,128 Interestingly, plant-derived exosomes have demonstrated the ability to mimic the structure and function of mammalian exosomes, opening up new avenues for therapeutic delivery.129 Plant-derived exosomes, specifically from ginger, exhibit anti-inflammatory properties.130 These vesicles, structurally and functionally similar yet safer than mammalian exosomes,131 effectively deliver bioactive compounds that modulate inflammatory pathways, including NF-κB and MAPK. Notably, GDENs inhibit NLRP3 inflammasome activation, highlighting their potential as therapeutic agents for inflammatory diseases.130,132,133 Bacterial EV-based delivery systems are being actively explored for their therapeutic and diagnostic potential in a wide range of diseases. These applications span across various conditions, including hyperammonemia, infections, cancer, and kidney failure.134 Current time milk-derived exosomes are an effective cancer therapeutic approach (due to it's dual nature, these sources require more research).135 Bacterial outer membrane vesicles, particularly from Akkermansia muciniphila, demonstrated potent anti-tumor activity by activating dendritic cells and triggering an interferon-γ-mediated immune response. This results in enhanced T cell cytotoxicity and an altered tumor microenvironment that effectively targets and destroys tumor cells.136 Bovine milk-derived exosomes are gaining recognition as a novel class of therapeutic agents, demonstrating efficacy as nanocarriers for enhanced drug delivery and immunotherapy, particularly in the context of cancer. These naturally secreted nanovesicles exhibit the capacity to encapsulate and stabilize cancer antigens,137 leading to enhanced antigen presentation by dendritic cells and subsequent activation of a cytotoxic CD8+ T cell response against tumor cells.137,138 The field of exosome-based therapeutics has also witnessed the emergence of synthetically engineered or chimeric exosomes. Genetically engineered exosomes, modified with specific ligands or genetic material, have shown enhanced targeting capabilities for cancer therapies.139 Chimeric exosomes, engineered to display both tumor-associated antigens and checkpoint inhibitors, offer a promising approach to enhancing cancer immunotherapy. These exosomes activate dendritic cells, leading to enhanced T cell activation and a more robust anti-tumor immune response. This approach promotes increased production of inflammatory mediators and cytotoxic T lymphocyte proliferation, ultimately boosting the immune system's ability to target and eliminate cancer cells.140,141 Engineered exosomes show promise for efficient gene delivery. A study suggests that these synthetic exosomes effectively deliver plasmid DNA and siRNA into target cells via clathrin-mediated endocytosis. Once inside lung cancer cells (A549), the delivered genetic material successfully modulates gene expression, notably enhancing gene silencing effects, highlighting the potential of engineered exosomes as a valuable tool for gene therapy. Click chemistry is a promising approach for modifying exosomal surfaces, enabling robust functionalization with therapeutic molecules to enhance their therapeutic potential.142 Exosomes can be engineered for enhanced tumor targeting by conjugating azide-modified exosomes with DBCO-modified antibodies, improving their potential for cancer immunotherapy.140,143 Functionalization of exosomes with therapeutic ligands via click-chemistry enhances drug delivery and immune modulation within tumor microenvironments.144 This approach also facilitates the attachment of fluorescent and imaging agents to exosomes, enabling their tracking and monitoring in vivo.144 Hybrid exosomes represent a convergent evolution in drug delivery, combining the advantages of different exosome sources or incorporating synthetic components. Hybrid exosomes with their superior biocompatibility and therapeutic efficacy, minimize the toxicological concerns associated with some existing approaches, potentially surpassing them.
image file: d5ra04927j-f5.tif
Fig. 5 Therapeutic exosomes. (Reproduced with permission under Creative Commons CC BY 4.0 license from ref. 177 Copyright @ 2024 The Authors).

6 Hybrid therapeutic exosomes

After chimeric exosomes and surface-modified exosomes, hybrid exosomes were introduced to cancer therapy with numerous promising characteristics like a high drug loading capacity and targeted cellular uptake, along with low toxicity, high biocompatibility and low immunogenicity to reduce the limitations of exosome-based cancer therapeutics.145,166 Hybrid exosomes (Fig. 6) are being designed by combining with various nanoparticles to increase the efficiency and resolve the challenge associated with exosome-based cancer therapeutics like drug off-targeting.146,170 In recent research, two approaches have been highlighted for designing artificial exosomes, to be specific they are top-down (for developing nanovesicles or NVs) and bottom-up (for designing exosome-mimetic or EM). Additionally, bio-hybrid is another significant technique for combining synthetic nanoparticles (NPs) with natural extracellular vesicles, specially exosomes to design hybrid exosomes.147 Numerous experiments have been conducted to combine synthetic nanoparticles (majorly liposomes) with various cell-derived exosomes with different bio-hybrid approaches to increase the efficacy of clinical applications (Table 1). Exosomes are being combined with liposomes through membrane fusion strategies to develop hybrid exosomes that are utilized for targeted drug delivery in chemotherapy. For instance, Lv et al., have designed thermosensitive exosome–liposome hybrid nanoparticles and used them to co-deliver granulocyte-macrophage colony-stimulating factor (GM-CSF) and docetaxel for treating metastatic peritoneal carcinoma. Their study found that these hybrid exosomes penetrated the tumor microenvironment effectively after intravenous injection, leading significant contribution to tumor suppression and improved chemotherapeutic efficacy.148
image file: d5ra04927j-f6.tif
Fig. 6 Application of hybrid exosomes in the clinical field. (Reproduced with permission under Creative Commons CC BY 4.0 license from ref. 178 Copyright @ 2022 The Authors).
Table 1 Combination of synthetic nanoparticles with various cell-derived exosomes with different bio-hybrid approaches
Synthetic nanoparticles Natural vesicles Biohybrid approach Comparison to natural exosomes Specific advances Significant application Ref.
Liposomes (DOPC, DOTAP, DSPE-PEG2000) Raw264.7 cell-derived exosomes Freeze-thawing Increased size and similar protein markers Membrane surface engineering Exosome modification 151
Liposomes (DSPE-PEG2000) Genetically engineered fibroblast-derived exosomes Freeze-thawing Similar morphology and protein markers Lipid engineering of exosomes Thermo-sensitive chemoimmunotherapy 148
Liposomes (lipofectamine 2000) HEK293FT cell-derived exosomes Simple incubation Increased size, but similar protein markers Efficient encapsulation of large plasmids CRISPR/Cas9 system transfer to MSCs 152
Liposomes (POPC, DOPE) HUVEC-derived EVs Incubation with PEG-mediated fusion Increased size but similar morphology and protein markers Efficient EV cargo loading and delivery Drug loading and delivery 149
Liposomes (L-α-phosphatidylcholine and cholesterol) Mouse macrophage J774A.1 cell-derived sEVs Extruding (400 and 200 nm) Increased size but similar protein markers Colloidal stability drug loading and pH-sensitive sustained drug release Tumor-targeted drug delivery 153
Lipids (DOTAP, POPC, DPPC or POPG) EVs derived from fibroblast 3T3 cells or A549 lung cancer cells Extruding (400, 200 and 100 nm) Similar size and with native EV fractions Mass production (6- to 43-fold vesicles) Efficient siRNA delivery 154


In a similar effort, Piffoux et al. also developed liposome–exosome hybrid nanoparticles with PEG to deliver meta-tetra (hydroxyphenyl)chlorin or mTHPC (an anti-tumor photosensitizer) to cancer cells. They found that the hybrid exosome achieved intracellular delivery that was 3 to 4 times greater than that of the free drug or the drug's liposomal formulation.149 However, recent research suggests exosome–liposome hybrid nanoparticles can improve the carrier stability and drug-loading capability of paclitaxel (PTX) and showed this PTX-loaded nanodrug delivery system improves treatment efficacy in mice with colorectal tumors. Additionally, it alters the tumor immune microenvironment by boosting CD4+ and CD8+ T cell activation, enhancing M1 macrophage polarization, and reducing Treg cell levels. This hybrid system offers significant potential for advancements in exosome engineering and its future applications can significantly contribute to precision cancer therapeutics.150

7 Clinical trial

In recent years, extracellular vesicles (EVs), particularly exosomes, have a significant role in cancer theranostics research. Several preclinical studies have been conducted demonstrating the potential of exosomes in medical research.13,155 Furthermore, numerous clinical trials have been conducted, showcasing their utility as diagnostic and prognostic biomarkers, drug delivery systems, and innovative therapeutic approaches.156 Exosomes serve as robust messengers, carrying vital information from cells into various body fluids. When cells are affected by disease, they release exosomes with distinct molecular payloads (cargos).157 By decoding these signatures, scientists can uncover potential biomarkers for diseases, enabling early detection and monitoring for improved disease management. For instance, the differential expression of several miRNAs is capable of providing essential information regarding cancer progression and validating treatment efficacy.158,159 A huge number of exosome clinical trials (Fig. 7) focus on cancer markers detection compared to other domain.160 On the other hand, exosome-based drug delivery is gaining significant attention in clinical trials as a promising strategy for addressing global health challenges, including cancer.161 Exosomes derived from diverse sources are being explored for their potential to revolutionize cancer treatment. Furthermore, various cell-derived exosomes are emerging as a focal point of modern research in medicine and health sciences, due to their distinctive properties and capacity to augment therapeutic outcomes. Notably, several clinical trials are currently investigating exosome-based cancer vaccine approaches, aiming to enhance treatment efficacy and improve patient outcomes.160,162 Despite the promising potential of hybrid exosome approaches in cancer treatment, a significant gap exists in pre-clinical and clinical trials, hindering further advancements in this innovative field. As a novel and smart strategy, hybrid exosome approaches warrant more extensive investigation to fully realize their therapeutic potential. Therefore, it is imperative to conduct additional clinical trials that focus on bio-hybrid exosome approaches, harnessing their unique properties to develop more effective cancer treatments. By doing so, we can unlock new possibilities for cancer treatment.
image file: d5ra04927j-f7.tif
Fig. 7 Clinical trial of exosomes. (Reproduced with permission under Creative Commons CC BY 4.0 license from ref. 156 Copyright @ 2024 The Authors).

8 Challenges and future perspective

Despite significant advances in exosome research, the field faces notable challenges, including gaps in understanding exosome biogenesis, isolation techniques, and their inherent heterogeneity. The concept of hybrid exosomes offers a promising approach for clinical therapeutics, particularly in oncology, but their practical use, especially in targeted drug delivery, encounters several difficulties. These include issues with large-scale production, purification, modification, drug loading, and storage, as well as the complexity introduced by the heterogeneity168 of extracellular vesicle subpopulations.163,164,171,172 Hybrid exosomes, produced through biohybrid methods, also face challenges such as low yields and the demanding process of combining synthetic liposomes with natural vesicles. Characterization is difficult due to their similarity to liposomes and natural exosomes, complicating purification. Major hurdles include refining preparation protocols, ensuring accurate characterization, and addressing biocompatibility.147 Membrane fusion hybrid exosomes (MFHEs) integrate the benefits of both exosomes and liposomes, showing potential for improved targeted drug delivery. However, enhancing the fusion between liposomes and exosomes while preventing unwanted liposome fusion remains a challenge. Recent use of single-stranded DNA to protect liposomes has improved their fusion with exosomes.165 A combination of exosome biology and advanced nanotechnology becomes a solution for effective exosomes isolation.40,164 Despite these issues, hybrid exosomes offer enhanced delivery efficiency and stability compared to liposomes or exosomes alone. The biohybrid approach offers a significant advantage by combining natural exosome components with other materials, leading to improved delivery efficiency compared to liposomes and enhanced stability compared to exosomes alone. This makes them highly versatile. Furthermore, the fusion technique used in these approaches allows for effective drug loading, accommodating both biological cargoes in liposomes and therapeutic agents in exosomes.149 Additionally, a recent study has shown that genetically engineered exosomes-thermosensitive liposomes hybrid nanoparticles (gETL NPs) improve drug delivery to metastatic peritoneal carcinoma (mPC) tumors, effectively inhibit tumor growth.148 Moreover, the exosome–liposome hybrid approach has shown higher efficacy in delivering CRISPR-Cas9 for cancer treatment. Considering all factors, it is evident that further research could advance hybrid exosome approaches into a cutting-edge therapeutic tool for cancer treatment.

9 Conclusion

In conclusion, engineered exosomes hold immense promise in cancer therapy as versatile vehicles for targeted drug delivery and gene regulation. Through precise targeting of specific genes or proteins, exosomes offer potent therapeutic effects with reduced off-target effects and toxicity. These engineered nanovesicles show potential in overcoming drug resistance, suppressing tumor growth, and enhancing the efficacy of conventional treatments. However, challenges such as standardization of isolation techniques, optimization of cargo loading, and elucidation of in vivo behavior remain to be addressed. With further research and development, engineered exosomes represent a promising avenue for personalized and effective cancer treatment strategies.

Conflicts of interest

The authors of this article declare no conflicts of interest.

Data availability

Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.

References

  1. F. Bray, M. Laversanne, H. Sung, J. Ferlay, R. L. Siegel, I. Soerjomataram and A. Jemal, Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries, Ca-Cancer J. Clin., 2024, 74(3), 229–263 Search PubMed.
  2. R. Atun and F. Cavalli, The global fight against cancer: challenges and opportunities, Lancet, 2018, 391(10119), 412–413 Search PubMed.
  3. H. Sung, J. Ferlay, R. L. Siegel, M. Laversanne, I. Soerjomataram, A. Jemal and F. Bray, Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries, Ca-Cancer J. Clin., 2021, 71(3), 209–249 Search PubMed.
  4. N. L. Syn, L. Wang, E. K. Chow, C. T. Lim and B. C. Goh, Exosomes in Cancer Nanomedicine and Immunotherapy: Prospects and Challenges, Trends Biotechnol., 2017, 35(7), 665–676 Search PubMed.
  5. N. M. Anderson and M. C. Simon, The tumor microenvironment, Curr. Biol., 2020, 30(16), R921–R925 Search PubMed.
  6. M. A. Swartz, N. Iida, E. W. Roberts, S. Sangaletti, M. H. Wong, F. E. Yull, L. M. Coussens and Y. A. DeClerck, Tumor microenvironment complexity: emerging roles in cancer therapy, Cancer Res., 2012, 72(10), 2473–2480 Search PubMed.
  7. Q. Wang, X. Shao, Y. Zhang, M. Zhu, F. X. C. Wang, J. Mu, J. Li, H. Yao and K. Chen, Role of tumor microenvironment in cancer progression and therapeutic strategy, Cancer Med., 2023, 12(10), 11149–11165 Search PubMed.
  8. D. F. Quail and J. A. Joyce, Microenvironmental regulation of tumor progression and metastasis, Nat. Med., 2013, 19(11), 1423–1437 Search PubMed.
  9. L. Guo and N. Guo, Exosomes: Potent regulators of tumor malignancy and potential bio-tools in clinical application, Crit. Rev. Oncol. Hemat., 2015, 95(3), 346–358 Search PubMed.
  10. R. H. Blackwell, K. E. Foreman and G. N. Gupta, The Role of Cancer-Derived Exosomes in Tumorigenicity & Epithelial-to-Mesenchymal Transition, Cancers, 2017, 9(8), 105 Search PubMed.
  11. J. Maia, S. Caja, M. C. Strano Moraes, N. Couto and B. Costa-Silva, Exosome-Based Cell-Cell Communication in the Tumor Microenvironment, Front. Cell Dev. Biol., 2018, 6, 18 Search PubMed.
  12. C. Rajagopal and K. B. Harikumar, The Origin and Functions of Exosomes in Cancer, Front. Oncol., 2018, 8, 66 Search PubMed.
  13. R. Kalluri, The biology and function of exosomes in cancer, J. Clin. Invest., 2016, 126(4), 1208–1215 Search PubMed.
  14. U. H. Weidle, F. Birzele, G. Kollmorgen and R. Rüger, The Multiple Roles of Exosomes in Metastasis, Cancer Genomics Proteomics, 2017, 14(1), 1–15 Search PubMed.
  15. R. J. Lobb, L. G. Lima and A. Möller, Exosomes: Key mediators of metastasis and pre-metastatic niche formation, Semin. Cell Dev. Biol., 2017, 67, 3–10 Search PubMed.
  16. X. Wang, L. Tian, J. Lu and I. O. Ng, Exosomes and cancer - Diagnostic and prognostic biomarkers and therapeutic vehicle, Oncogenesis, 2022, 11(1), 54 Search PubMed.
  17. N. Dilsiz, Role of exosomes and exosomal microRNAs in cancer, Future Sci. OA, 2020, 6(4), FSO465 Search PubMed.
  18. Z. H. Zhang, H. M. Cao and X. Li, Exosomes serve as molecular diagnostic biomarkers and carriers for tissue engineering applications, Mater. Today Commun., 2025, 112978 Search PubMed.
  19. Z. Wang, Q. Wang, F. Qin and J. Chen, Exosomes: a promising avenue for cancer diagnosis beyond treatment, Front. Cell Dev. Biol., 2024, 12, 1344705 Search PubMed.
  20. K. Zhang and K. Cheng, Stem cell-derived exosome versus stem cell therapy, Nat. Rev. Bioeng., 2023, 1–2 Search PubMed.
  21. J. Dai, Y. Su, S. Zhong, L. Cong, B. Liu, J. Yang, Y. Tao, Z. He, C. Chen and Y. Jiang, Exosomes: key players in cancer and potential therapeutic strategy, Signal Transduction Targeted Ther., 2020, 5(1), 145 Search PubMed.
  22. G. H. Nam, Y. Choi, G. B. Kim, S. Kim, S. A. Kim and I. S. Kim, Emerging Prospects of Exosomes for Cancer Treatment: From Conventional Therapy to Immunotherapy, Adv. Mater., 2020, 32(51), e2002440 Search PubMed.
  23. S. M. Patil, S. S. Sawant and N. K. Kunda, Exosomes as drug delivery systems: A brief overview and progress update, Eur. J. Pharm. Biopharm., 2020, 154, 259–269 Search PubMed.
  24. W. Liao, Y. Du, C. Zhang, F. Pan, Y. Yao, T. Zhang and Q. Peng, Exosomes: The next generation of endogenous nanomaterials for advanced drug delivery and therapy, Acta Biomater., 2019, 86, 1–14 Search PubMed.
  25. X. Luan, K. Sansanaphongpricha, I. Myers, H. Chen, H. Yuan and D. Sun, Engineering exosomes as refined biological nanoplatforms for drug delivery, Acta Pharmacol. Sin., 2017, 38(6), 754–763 Search PubMed.
  26. M. H. Chan, Z. X. Chang, C. F. Huang, L. J. Lee, R. S. Liu and M. Hsiao, Integrated therapy platform of exosomal system: hybrid inorganic/organic nanoparticles with exosomes for cancer treatment, Nanoscale Horiz., 2022, 7(4), 352–367 Search PubMed.
  27. J. Mondal, S. Pillarisetti, V. Junnuthula, M. Saha, S. R. Hwang, I. K. Park and Y. K. Lee, Hybrid exosomes, exosome-like nanovesicles and engineered exosomes for therapeutic applications, J. Controlled Release, 2023, 353, 1127–1149 Search PubMed.
  28. Q. F. Han, W. J. Li, K. S. Hu, J. Gao, W. L. Zhai, J. H. Yang and S. J. Zhang, Exosome biogenesis: machinery, regulation, and therapeutic implications in cancer, Mol. Cancer, 2022, 21(1), 207 Search PubMed.
  29. J. Rädler, D. Gupta, A. Zickler and S. E. Andaloussi, Exploiting the biogenesis of extracellular vesicles for bioengineering and therapeutic cargo loading, Mol. Ther., 2023, 31(5), 1231–1250 Search PubMed.
  30. N. P. Hessvik and A. Llorente, Current knowledge on exosome biogenesis and release, Cell. Mol. Life Sci., 2018, 75(2), 193–208 Search PubMed.
  31. L. Christ, C. Raiborg, E. M. Wenzel, C. Campsteijn and H. Stenmark, Cellular Functions and Molecular Mechanisms of the ESCRT Membrane-Scission Machinery, Trends Biochem. Sci., 2017, 42(1), 42–56 Search PubMed.
  32. T. Wollert and J. H. Hurley, Molecular mechanism of multivesicular body biogenesis by ESCRT complexes, Nature, 2010, 464(7290), 864–869 Search PubMed.
  33. A. L. Schuh and A. Audhya, The ESCRT machinery: from the plasma membrane to endosomes and back again, Crit. Rev. Biochem. Mol. Biol., 2014, 49(3), 242–261 Search PubMed.
  34. S. B. Arya, S. Chen, F. Jordan-Javed and C. A. Parent, Ceramide-rich microdomains facilitate nuclear envelope budding for non-conventional exosome formation, Nat. Cell Biol., 2022, 24(7), 1019–1028 Search PubMed.
  35. Y. Zhang, Y. Liu, H. Liu and W. H. Tang, Exosomes: biogenesis, biologic function and clinical potential, Cell Biosci., 2019, 9, 19 Search PubMed.
  36. A. Das, P. Saha, K. Kalele and S. Sonar, Clinical signature of exosomal tetraspanin proteins in cancer, Clin. Transl. Discovery, 2024, 4, e341 Search PubMed.
  37. S. B. Arya, S. P. Collie and C. A. Parent, The ins-and-outs of exosome biogenesis, secretion, and internalization, Trends Cell Biol., 2024, 34(2), 90–108 Search PubMed.
  38. G. O. Skryabin, A. V. Komelkov, E. E. Savelyeva and E. M. Tchevkina, Lipid Rafts in Exosome Biogenesis, Biochemistry, 2020, 85(2), 177–191 Search PubMed.
  39. M. Omrani, H. Beyrampour-Basmenj, R. Jahanban-Esfahlan, M. Talebi, M. Raeisi, Z. A. Serej, N. Akbar-Gharalari, S. Khodakarimi, J. Wu and A. Ebrahimi-Kalan, Global trend in exosome isolation and application: an update concept in management of diseases, Mol. Cell. Biochem., 2024, 479(3), 679–691 Search PubMed.
  40. W. Z. Liu, Z. J. Ma and X. W. Kang, Current status and outlook of advances in exosome isolation, Anal. Bioanal. Chem., 2022, 414(24), 7123–7141 Search PubMed.
  41. C. Gardiner, D. Di Vizio, S. Sahoo, C. Théry, K. W. Witwer, M. Wauben and A. F. Hill, Techniques used for the isolation and characterization of extracellular vesicles: results of a worldwide survey, J. Extracell. Vesicles, 2016, 5, 32945 Search PubMed.
  42. S. Kamerkar, V. S. LeBleu, H. Sugimoto, S. Yang, C. F. Ruivo, S. A. Melo, J. J. Lee and R. Kalluri, Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer, Nature, 2017, 546(7659), 498–503 Search PubMed.
  43. K. Takov, D. M. Yellon and S. M. Davidson, Comparison of small extracellular vesicles isolated from plasma by ultracentrifugation or size-exclusion chromatography: yield, purity and functional potential, J. Extracell. Vesicles, 2018, 8(1), 1560809 Search PubMed.
  44. I. Lozano-Ramos, I. Bancu, A. Oliveira-Tercero, M. P. Armengol, A. Menezes-Neto, H. A. Del Portillo, R. Lauzurica-Valdemoros and F. E. Borràs, Size-exclusion chromatography-based enrichment of extracellular vesicles from urine samples, J. Extracell. Vesicles, 2015, 4, 27369 Search PubMed.
  45. R. Ghosh, Rapid antibody screening by membrane chromatographic immunoassay technique, J. Chromatogr. B:Anal. Technol. Biomed. Life Sci., 2006, 844(1), 163–167 Search PubMed.
  46. B. Lebreton, A. Brown and R. van Reis, Application of high-performance tangential flow filtration (HPTFF) to the purification of a human pharmaceutical antibody fragment expressed in Escherichia coli, Biotechnol. Bioeng., 2008, 100(5), 964–974 Search PubMed.
  47. L. Ding, X. Yang, Z. Gao, C. Y. Effah, X. Zhang, Y. Wu and L. Qu, A Holistic Review of the State-of-the-Art Microfluidics for Exosome Separation: An Overview of the Current Status, Existing Obstacles, and Future Outlook, Small, 2021, 17(29), e2007174 Search PubMed.
  48. Y. Tian, M. Gong, Y. Hu, H. Liu, W. Zhang, M. Zhang, X. Hu, D. Aubert, S. Zhu, L. Wu and X. Yan, Quality and efficiency assessment of six extracellular vesicle isolation methods by nano-flow cytometry, J. Extracell. Vesicles, 2019, 9(1), 1697028 Search PubMed.
  49. G. K. Patel, M. A. Khan, H. Zubair, S. K. Srivastava, M. Khushman, S. Singh and A. P. Singh, Comparative analysis of exosome isolation methods using culture supernatant for optimum yield, purity and downstream applications, Sci. Rep., 2019, 9(1), 5335 Search PubMed.
  50. M. Zhang, K. Jin, L. Gao, Z. Zhang, F. Li, F. Zhou and L. Zhang, Methods and technologies for exosome isolation and characterization, Small Methods, 2018, 2(9), 1800021 Search PubMed.
  51. S. Lin, Z. Yu, D. Chen, Z. Wang, J. Miao, Q. Li, D. Zhang, J. Song and D. Cui, Progress in Microfluidics-Based Exosome Separation and Detection Technologies for Diagnostic Applications, Small, 2020, 16(9), e1903916 Search PubMed.
  52. B. Lin, Y. Lei, J. Wang, L. Zhu, Y. Wu, H. Zhang, L. Wu, P. Zhang and C. Yang, Microfluidic-Based Exosome Analysis for Liquid Biopsy, Small Methods, 2021, 5(3), e2001131 Search PubMed.
  53. Z. Wang, F. Li, J. Rufo, C. Chen, S. Yang, L. Li, J. Zhang, J. Cheng, Y. Kim, M. Wu, E. Abemayor, M. Tu, D. Chia, R. Spruce, N. Batis, H. Mehanna, D. T. W. Wong and T. J. Huang, Acoustofluidic Salivary Exosome Isolation: A Liquid Biopsy Compatible Approach for Human Papillomavirus-Associated Oropharyngeal Cancer Detection, J. Mol. Diagn., 2020, 22(1), 50–59 Search PubMed.
  54. K. Zhang, Y. Yue, S. Wu, W. Liu, J. Shi and Z. Zhang, Rapid Capture and Nondestructive Release of Extracellular Vesicles Using Aptamer-Based Magnetic Isolation, ACS Sens., 2019, 4(5), 1245–1251 Search PubMed.
  55. S. M. Santana, M. A. Antonyak, R. A. Cerione and B. J. Kirby, Microfluidic isolation of cancer-cell-derived microvesicles from hetergeneous extracellular shed vesicle populations, Biomed. Microdevices, 2014, 16(6), 869–877 Search PubMed.
  56. B. H. Wunsch, J. T. Smith, S. M. Gifford, C. Wang, M. Brink, R. L. Bruce, R. H. Austin, G. Stolovitzky and Y. Astier, Nanoscale lateral displacement arrays for the separation of exosomes and colloids down to 20 nm, Nat. Nanotechnol., 2016, 11(11), 936–940 Search PubMed.
  57. D. Kang, S. Oh, S. M. Ahn, B. H. Lee and M. H. Moon, Proteomic analysis of exosomes from human neural stem cells by flow field-flow fractionation and nanoflow liquid chromatography-tandem mass spectrometry, J. Proteome Res., 2008, 7(8), 3475–3480 Search PubMed.
  58. H. Zhang and D. Lyden, Asymmetric-flow field-flow fractionation technology for exomere and small extracellular vesicle separation and characterization, Nat. Protoc., 2019, 14(4), 1027–1053 Search PubMed.
  59. H. Zhang, D. Freitas, H. S. Kim, K. Fabijanic, Z. Li, H. Chen, M. T. Mark, H. Molina, A. B. Martin, L. Bojmar, J. Fang, S. Rampersaud, A. Hoshino, I. Matei, C. M. Kenific, M. Nakajima, A. P. Mutvei, P. Sansone, W. Buehring, H. Wang, J. P. Jimenez, L. Cohen-Gould, N. Paknejad, M. Brendel, K. Manova-Todorova, A. Magalhães, J. A. Ferreira, H. Osório, A. M. Silva, A. Massey, J. R. Cubillos-Ruiz, G. Galletti, P. Giannakakou, A. M. Cuervo, J. Blenis, R. Schwartz, M. S. Brady, H. Peinado, J. Bromberg, H. Matsui, C. A. Reis and D. Lyden, Identification of distinct nanoparticles and subsets of extracellular vesicles by asymmetric flow field-flow fractionation, Nat. Cell Biol., 2018, 20(3), 332–343 Search PubMed.
  60. S. Sitar, A. Kejžar, D. Pahovnik, K. Kogej, M. Tušek-Žnidarič, M. Lenassi and E. Žagar, Size characterization and quantification of exosomes by asymmetrical-flow field-flow fractionation, Anal. Chem., 2015, 87(18), 9225–9233 Search PubMed.
  61. H. Shao, H. Im, C. M. Castro, X. Breakefield, R. Weissleder and H. Lee, New Technologies for Analysis of Extracellular Vesicles, Chem. Rev., 2018, 118(4), 1917–1950 Search PubMed.
  62. V. Sokolova, A. K. Ludwig, S. Hornung, O. Rotan, P. A. Horn, M. Epple and B. Giebel, Characterisation of exosomes derived from human cells by nanoparticle tracking analysis and scanning electron microscopy, Colloids Surf., B, 2011, 87(1), 146–150 Search PubMed.
  63. R. A. Dragovic, C. Gardiner, A. S. Brooks, D. S. Tannetta, D. J. Ferguson, P. Hole, B. Carr, C. W. Redman, A. L. Harris, P. J. Dobson, P. Harrison and I. L. Sargent, Sizing and phenotyping of cellular vesicles using Nanoparticle Tracking Analysis, Nanomedicine, 2011, 7(6), 780–788 Search PubMed.
  64. A. S. Lawrie, A. Albanyan, R. A. Cardigan, I. J. Mackie and P. Harrison, Microparticle sizing by dynamic light scattering in fresh-frozen plasma, Vox Sang., 2009, 96(3), 206–212 Search PubMed.
  65. Y. Yuana, T. H. Oosterkamp, S. Bahatyrova, B. Ashcroft, P. Garcia Rodriguez, R. M. Bertina and S. Osanto, Atomic force microscopy: a novel approach to the detection of nanosized blood microparticles, J. Thromb. Haemostasis, 2010, 8(2), 315–323 Search PubMed.
  66. T. Katsuda, N. Kosaka and T. Ochiya, The roles of extracellular vesicles in cancer biology: toward the development of novel cancer biomarkers, Proteomics, 2014, 14(4–5), 412–425 Search PubMed.
  67. N. Arraud, C. Gounou, D. Turpin and A. R. Brisson, Fluorescence triggering: A general strategy for enumerating and phenotyping extracellular vesicles by flow cytometry, Cytometry, Part A, 2016, 89(2), 184–195 Search PubMed.
  68. I. Tatischeff, E. Larquet, J. M. Falcón-Pérez, P. Y. Turpin and S. G. Kruglik, Fast characterisation of cell-derived extracellular vesicles by nanoparticles tracking analysis, cryo-electron microscopy, and Raman tweezers microspectroscopy, J. Extracell. Vesicles, 2012, 1, 19179 Search PubMed.
  69. S. Kreimer, A. M. Belov, I. Ghiran, S. K. Murthy, D. A. Frank and A. R. Ivanov, Mass-spectrometry-based molecular characterization of extracellular vesicles: lipidomics and proteomics, J. Proteome Res., 2015, 14(6), 2367–2384 Search PubMed.
  70. G. Pocsfalvi, C. Stanly, A. Vilasi, I. Fiume, G. Capasso, L. Turiák, E. I. Buzas and K. Vékey, Mass spectrometry of extracellular vesicles, Mass Spectrom. Rev., 2016, 35(1), 3–21 Search PubMed.
  71. D. S. Choi, D. K. Kim, Y. K. Kim and Y. S. Gho, Proteomics, transcriptomics and lipidomics of exosomes and ectosomes, Proteomics, 2013, 13(10–11), 1554–1571 Search PubMed.
  72. S. Maheshwari, A. K. Singh, R. K. Arya, D. Pandey, A. Singh and D. Datta, Exosomes: Emerging Players of Intercellular Communication in Tumor Microenvironment, Discoveries, 2014, 2(3), e26 Search PubMed.
  73. D. Hanahan and R. A. Weinberg, Hallmarks of cancer: the next generation, Cell, 2011, 144(5), 646–674 Search PubMed.
  74. V. Petrova, M. Annicchiarico-Petruzzelli, G. Melino and I. Amelio, The hypoxic tumour microenvironment, Oncogenesis, 2018, 7(1), 10 Search PubMed.
  75. Z. Chen, F. Han, Y. Du, H. Shi and W. Zhou, Hypoxic microenvironment in cancer: molecular mechanisms and therapeutic interventions, Signal Transduction Targeted Ther., 2023, 8(1), 70 Search PubMed.
  76. G. L. Semenza, Hypoxia-inducible factors: mediators of cancer progression and targets for cancer therapy, Trends Pharmacol. Sci., 2012, 33(4), 207–214 Search PubMed.
  77. P. Vaupel, The role of hypoxia-induced factors in tumor progression, Oncologist, 2004, 9(Suppl 5), 10–17 Search PubMed.
  78. W. Meng, Y. Hao, C. He, L. Li and G. Zhu, Exosome-orchestrated hypoxic tumor microenvironment, Mol. Cancer, 2019, 18(1), 57 Search PubMed.
  79. B. Muz, P. de la Puente, F. Azab and A. K. Azab, The role of hypoxia in cancer progression, angiogenesis, metastasis, and resistance to therapy, Hypoxia, 2015, 3, 83–92 Search PubMed.
  80. J. Zhou, T. Schmid, S. Schnitzer and B. Brüne, Tumor hypoxia and cancer progression, Cancer Lett., 2006, 237(1), 10–21 Search PubMed.
  81. A. C. Chiang and J. Massagué, Molecular basis of metastasis, N. Engl. J. Med., 2008, 359(26), 2814–2823 Search PubMed.
  82. C. Shao, F. Yang, S. Miao, W. Liu, C. Wang, Y. Shu and H. Shen, Role of hypoxia-induced exosomes in tumor biology, Mol. Cancer, 2018, 17(1), 120 Search PubMed.
  83. J. E. Park, H. S. Tan, A. Datta, R. C. Lai, H. Zhang, W. Meng, S. K. Lim and S. K. Sze, Hypoxic tumor cell modulates its microenvironment to enhance angiogenic and metastatic potential by secretion of proteins and exosomes, Mol. Cell. Proteomics, 2010, 9(6), 1085–1099 Search PubMed.
  84. K. Z. Salem, M. Moschetta, A. Sacco, L. Imberti, G. Rossi, I. M. Ghobrial, S. Manier and A. M. Roccaro, Exosomes in Tumor Angiogenesis, Methods Mol. Biol., 2016, 1464, 25–34 Search PubMed.
  85. W. Wang, Y. Han, H. A. Jo, J. Lee and Y. S. Song, Non-coding RNAs shuttled via exosomes reshape the hypoxic tumor microenvironment, J. Hematol. Oncol., 2020, 13(1), 67 Search PubMed.
  86. Z. Jiang, J. Zhou, L. Li, S. Liao, J. He, S. Zhou and Y. Zhou, Pericytes in the tumor microenvironment, Cancer Lett., 2023, 556, 216074 Search PubMed.
  87. C. Roma-Rodrigues, A. R. Fernandes and P. V. Baptista, Exosome in tumour microenvironment: overview of the crosstalk between normal and cancer cells, BioMed Res. Int., 2014, 2014, 179486 Search PubMed.
  88. C. Aslan, S. Maralbashi, F. Salari, H. Kahroba, F. Sigaroodi, T. Kazemi and P. Kharaziha, TEXs: Implication in angiogenesis and antiangiogenesis cancer therapy, J. Cell. Physiol., 2019, 234(10), 16885–16903 Search PubMed.
  89. P. Fasanaro, Y. D'Alessandra, V. Di Stefano, R. Melchionna, S. Romani, G. Pompilio, M. C. Capogrossi and F. Martelli, MicroRNA-210 modulates endothelial cell response to hypoxia and inhibits the receptor tyrosine kinase ligand Ephrin-A3, J. Biol. Chem., 2008, 283(23), 15878–15883 Search PubMed.
  90. S. Landskroner-Eiger, I. Moneke and W. C. Sessa, miRNAs as modulators of angiogenesis, Cold Spring Harbor Perspect. Med., 2013, 3(2), a006643 Search PubMed.
  91. X. Shao, S. Hua, T. Feng, D. K. W. Ocansey and L. Yin, Hypoxia-Regulated TEXs and Tumor Progression: A Focus on Immune Evasion, Int. J. Mol. Sci., 2022, 23(19), 11789 Search PubMed.
  92. W. Guo, T. Qiao, B. Dong, T. Li, Q. Liu and X. Xu, The Effect of Hypoxia-Induced Exosomes on Anti-Tumor Immunity and Its Implication for Immunotherapy, Front. Immunol., 2022, 13, 915985 Search PubMed.
  93. T. L. Whiteside, TEXs and Their Role in Tumor-Induced Immune Suppression, Vaccines, 2016, 4(4), 35 Search PubMed.
  94. L. You, W. Wu, X. Wang, L. Fang, V. Adam, E. Nepovimova, Q. Wu and K. Kuca, The role of hypoxia-inducible factor 1 in tumor immune evasion, Med. Res. Rev., 2021, 41(3), 1622–1643 Search PubMed.
  95. S. M. Morrissey, F. Zhang, C. Ding, D. E. Montoya-Durango, X. Hu, C. Yang, Z. Wang, F. Yuan, M. Fox, H. G. Zhang, H. Guo, D. Tieri, M. Kong, C. T. Watson, R. A. Mitchell, X. Zhang, K. M. McMasters, J. Huang and J. Yan, TEXs drive immunosuppressive macrophages in a pre-metastatic niche through glycolytic dominant metabolic reprogramming, Cell Metab., 2021, 33(10), 2040–2058 Search PubMed.
  96. S. Zhao, Y. Mi, B. Guan, B. Zheng, P. Wei, Y. Gu, Z. Zhang, S. Cai, Y. Xu, X. Li, X. He, X. Zhong, G. Li, Z. Chen and D. Li, Tumor-derived exosomal miR-934 induces macrophage M2 polarization to promote liver metastasis of colorectal cancer, J. Hematol. Oncol., 2020, 13(1), 156 Search PubMed.
  97. M. S. Baig, A. Roy, S. Rajpoot, D. Liu, R. Savai, S. Banerjee, M. Kawada, S. M. Faisal, R. Saluja, U. Saqib, T. Ohishi and K. K. Wary, TEXs in the regulation of macrophage polarization, Inflammation Res., 2020, 69(5), 435–451 Search PubMed.
  98. X. Tian, H. Shen, Z. Li, T. Wang and S. Wang, Tumor-derived exosomes, myeloid-derived suppressor cells, and tumor microenvironment, J. Hematol. Oncol., 2019, 12(1), 84 Search PubMed.
  99. X. Xiang, A. Poliakov, C. Liu, Y. Liu, Z. B. Deng, J. Wang, Z. Cheng, S. V. Shah, G. J. Wang, L. Zhang, W. E. Grizzle, J. Mobley and H. G. Zhang, Induction of myeloid-derived suppressor cells by tumor exosomes, Int. J. Cancer, 2009, 124(11), 2621–2633 Search PubMed.
  100. X. Guo, W. Qiu, Q. Liu, M. Qian, S. Wang, Z. Zhang, X. Gao, Z. Chen, H. Xue and G. Li, Immunosuppressive effects of hypoxia-induced glioma exosomes through myeloid-derived suppressor cells via the miR-10a/Rora and miR-21/Pten Pathways, Oncogene, 2018, 37(31), 4239–4259 Search PubMed.
  101. D. Basso, E. Gnatta, A. Padoan, P. Fogar, S. Furlanello, A. Aita, D. Bozzato, C. F. Zambon, G. Arrigoni, C. Frasson, C. Franchin, S. Moz, T. Brefort, T. Laufer, F. Navaglia, S. Pedrazzoli, G. Basso and M. Plebani, PDAC-derived exosomes enrich the microenvironment in MDSCs in a SMAD4-dependent manner through a new calcium related axis, Oncotarget, 2017, 8(49), 84928–84944 Search PubMed.
  102. R. Hosseini, L. Asef-Kabiri, H. Yousefi, H. Sarvnaz, M. Salehi, M. E. Akbari and N. Eskandari, The roles of tumor-derived exosomes in altered differentiation, maturation and function of dendritic cells, Mol. Cancer, 2021, 20(1), 83 Search PubMed.
  103. J. Jiang, J. Li, X. Zhou, X. Zhao, B. Huang and Y. Qin, Exosomes Regulate the Epithelial-Mesenchymal Transition in Cancer, Front. Oncol., 2022, 12, 864980 Search PubMed.
  104. C. Yang, R. Dou, C. Wei, K. Liu, D. Shi, C. Zhang, Q. Liu, S. Wang and B. Xiong, Tumor-derived exosomal microRNA-106b-5p activates EMT-cancer cell and M2-subtype TAM interaction to facilitate CRC metastasis, Mol. Ther., 2021, 29(6), 2088–2107 Search PubMed.
  105. L. Zhao, X. Ma and J. Yu, Exosomes and organ-specific metastasis, Mol. Ther.--Methods Clin. Dev., 2021, 22, 133–147 Search PubMed.
  106. A. C. Obenauf and J. Massagué, Surviving at a Distance: Organ-Specific Metastasis, Trends Cancer, 2015, 1(1), 76–91 Search PubMed.
  107. A. Hoshino, B. Costa-Silva, T. L. Shen, G. Rodrigues, A. Hashimoto, M. Tesic Mark, H. Molina, S. Kohsaka, A. Di Giannatale, S. Ceder, S. Singh, C. Williams, N. Soplop, K. Uryu, L. Pharmer, T. King, L. Bojmar, A. E. Davies, Y. Ararso, T. Zhang, H. Zhang, J. Hernandez, J. M. Weiss, V. D. Dumont-Cole, K. Kramer, L. H. Wexler, A. Narendran, G. K. Schwartz, J. H. Healey, P. Sandstrom, K. J. Labori, E. H. Kure, P. M. Grandgenett, M. A. Hollingsworth, M. de Sousa, S. Kaur, M. Jain, K. Mallya, S. K. Batra, W. R. Jarnagin, M. S. Brady, O. Fodstad, V. Muller, K. Pantel, A. J. Minn, M. J. Bissell, B. A. Garcia, Y. Kang, V. K. Rajasekhar, C. M. Ghajar, I. Matei, H. Peinado, J. Bromberg and D. Lyden, Tumour exosome integrins determine organotropic metastasis, Nature, 2015, 527(7578), 329–335 Search PubMed.
  108. E. S. Grigoryeva, L. A. Tashireva, O. E. Savelieva, M. V. Zavyalova, N. O. Popova, G. A. Kuznetsov, E. S. Andryuhova and V. M. Perelmuter, The Association of Integrins β3, β4, and αVβ5 on Exosomes, CTCs and Tumor Cells with Localization of Distant Metastasis in Breast Cancer Patients, Int. J. Mol. Sci., 2023, 24(3), 2929 Search PubMed.
  109. M. Wandrey, J. Jablonska, R. H. Stauber and D. Gül, Exosomes in Cancer Progression and Therapy Resistance: Molecular Insights and Therapeutic Opportunities, Life, 2023, 13(10), 2033 Search PubMed.
  110. Y. Zhong, H. Li, P. Li, Y. Chen, M. Zhang, Z. Yuan, Y. Zhang, Z. Xu, G. Luo, Y. Fang and X. Li, Exosomes: A New Pathway for Cancer Drug Resistance, Front. Oncol., 2021, 11, 743556 Search PubMed.
  111. R. L. Siegel, K. D. Miller, N. S. Wagle and A. Jemal, Cancer statistics, 2023, Ca-Cancer J. Clin., 2023, 73(1), 17–48 Search PubMed.
  112. X. Qin, S. Yu, L. Zhou, M. Shi, Y. Hu, X. Xu, B. Shen, S. Liu, D. Yan and J. Feng, Cisplatin-resistant lung cancer cell-derived exosomes increase cisplatin resistance of recipient cells in exosomal miR-100-5p-dependent manner, Int. J. Nanomed., 2017, 12, 3721–3733 Search PubMed.
  113. L. Liu, D. Jiang, S. Bai, X. Zhang and Y. Kang, Research progress of exosomes in drug resistance of breast cancer, Front. Bioeng. Biotechnol., 2024, 11, 1214648 Search PubMed.
  114. X. Dong, X. Bai, J. Ni, H. Zhang, W. Duan, P. Graham and Y. Li, Exosomes and breast cancer drug resistance, Cell Death Dis., 2020, 11(11), 987 Search PubMed.
  115. V. G. Martinez, S. O'Neill, J. Salimu, S. Breslin, A. Clayton, J. Crown and L. O'Driscoll, Resistance to HER2-targeted anti-cancer drugs is associated with immune evasion in cancer cells and their derived extracellular vesicles, Oncoimmunology, 2017, 6(12), e1362530 Search PubMed.
  116. G. Kibria, E. K. Ramos, Y. Wan, D. R. Gius and H. Liu, Exosomes as a Drug Delivery System in Cancer Therapy: Potential and Challenges, Mol. Pharm., 2018, 15(9), 3625–3633 Search PubMed.
  117. S. F. Nassar, K. Raddassi, B. Ubhi, J. Doktorski and A. Abulaban, Precision Medicine: Steps along the Road to Combat Human Cancer, Cells, 2020, 9(9), 2056 Search PubMed.
  118. N. L. Syn, L. Wang, E. K. Chow, C. T. Lim and B. C. Goh, Exosomes in Cancer Nanomedicine and Immunotherapy: Prospects and Challenges, Trends Biotechnol., 2017, 35(7), 665–676 Search PubMed.
  119. C. Liu and C. Su, Design strategies and application progress of therapeutic exosomes, Theranostics, 2019, 9(4), 1015–1028 Search PubMed.
  120. M. E. Marcus and J. N. Leonard, FedExosomes: Engineering Therapeutic Biological Nanoparticles that Truly Deliver, Pharmaceuticals, 2013, 6(5), 659–680 Search PubMed.
  121. F. Tan, X. Li, Z. Wang, J. Li, K. Shahzad and J. Zheng, Clinical applications of stem cell-derived exosomes, Signal Transduction Targeted Ther., 2024, 9(1), 17 Search PubMed.
  122. H. Lee, S. H. Kang, G. H. Jeong, S. S. Lee, B. Y. Chung, G. J. Kim and H. W. Bai, Gamma irradiation-engineered macrophage-derived exosomes as potential immunomodulatory therapeutic agents, PLoS One, 2024, 19(6), e0303434 Search PubMed.
  123. S. Wang, J. Sun, R. M. Dastgheyb and Z. Li, Tumor-derived extracellular vesicles modulate innate immune responses to affect tumor progression, Front. Immunol., 2022, 13, 1045624 Search PubMed.
  124. C. J. Osterman, J. C. Lynch, P. Leaf, A. Gonda, H. R. Ferguson Bennit, D. Griffiths and N. R. Wall, Curcumin Modulates Pancreatic Adenocarcinoma Cell-Derived Exosomal Function, PLoS One, 2015, 10(7), e0132845 Search PubMed.
  125. Q. Li, S. Cai, M. Li, K. I. Salma, X. Zhou, F. Han, J. Chen and T. Huyan, Tumor-Derived Extracellular Vesicles: Their Role in Immune Cells and Immunotherapy, Int. J. Nanomed., 2021, 16, 5395–5409 Search PubMed.
  126. L. Yang, S. Huang, Z. Zhang, Z. Liu and L. Zhang, Roles and Applications of Red Blood Cell-Derived Extracellular Vesicles in Health and Diseases, Int. J. Mol. Sci., 2022, 23(11), 5927 Search PubMed.
  127. W. M. Usman, T. C. Pham, Y. Y. Kwok, L. T. Vu, V. Ma, B. Peng, Y. S. Chan, L. Wei, S. M. Chin, A. Azad, A. B. He, A. Y. H. Leung, M. Yang, N. Shyh-Chang, W. C. Cho, J. Shi and M. T. N. Le, Efficient RNA drug delivery using red blood cell extracellular vesicles, Nat. Commun., 2018, 9(1), 2359 Search PubMed.
  128. S. Biagiotti, F. Abbas, M. Montanari, C. Barattini, L. Rossi, M. Magnani, S. Papa and B. Canonico, Extracellular Vesicles as New Players in Drug Delivery: A Focus on Red Blood Cells-Derived EVs, Pharmaceutics, 2023, 15(2), 365 Search PubMed.
  129. J. Kim, S. Li, S. Zhang and J. Wang, Plant-derived exosome-like nanoparticles and their therapeutic activities, Asian J. Pharm. Sci., 2022, 17(1), 53–69 Search PubMed.
  130. F. Bahri, M. Mansoori, S. Vafaei, S. Fooladi, Y. Mir, M. Mehrabani, Y. Hojabri, M. H. Nematollahi and S. Iravani, A Comprehensive Review on Ginger-Derived Extracellular Nanoparticles: Feasible Therapeutic Nano-Agents Against Diseases, Mater. Adv., 2024, 5, 1846 Search PubMed.
  131. S. Sonar and K. Anand, Plant-derived exosomes: A Green Nanomedicine for Cancer, Clin. Transl. Discovery, 2024, 4, e333 Search PubMed.
  132. Z. Jin, J. Na, X. Lin, R. Jiao, X. Liu and Y. Huang, Plant-derived exosome-like nanovesicles: A novel nanotool for disease therapy, Heliyon, 2024, 10(9), e30630 Search PubMed.
  133. N. Mu, J. Li, L. Zeng, J. You, R. Li, A. Qin, X. Liu, F. Yan and Z. Zhou, Plant-Derived Exosome-Like Nanovesicles: Current Progress and Prospects, Int. J. Nanomed., 2023, 18, 4987–5009 Search PubMed.
  134. Z. Li, Y. Wang, J. Liu, P. Rawding, J. Bu, S. Hong and Q. Hu, Chemically and Biologically Engineered Bacteria-Based Delivery Systems for Emerging Diagnosis and Advanced Therapy, Adv. Mater., 2021, 33(38), e2102580 Search PubMed.
  135. A. Das, S. Sonar, K. Kalele and V. Subramaniyan, Milk exosomes: Harnessingnature's duality for cancer therapy, Clin. Transl. Discovery, 2024, 4, e349 Search PubMed.
  136. O. Y. Kim, H. T. Park, N. T. H. Dinh, S. J. Choi, J. Lee, J. H. Kim, S. W. Lee and Y. S. Gho, Bacterial outer membrane vesicles suppress tumor by interferon-γ-mediated antitumor response, Nat. Commun., 2017, 8(1), 626 Search PubMed.
  137. A. M. Timofeeva, A. P. Paramonik, S. S. Sedykh and G. A. Nevinsky, Milk Exosomes: Next-Generation Agents for Delivery of Anticancer Drugs and Therapeutic Nucleic Acids, Int. J. Mol. Sci., 2023, 24(12), 10194 Search PubMed.
  138. R. Munagala, F. Aqil, J. Jeyabalan and R. C. Gupta, Bovine milk-derived exosomes for drug delivery, Cancer Lett., 2016, 371(1), 48–61 Search PubMed.
  139. X. Luan, K. Sansanaphongpricha, I. Myers, H. Chen, H. Yuan and D. Sun, Engineering exosomes as refined biological nanoplatforms for drug delivery, Acta Pharmacol. Sin., 2017, 38(6), 754–763 Search PubMed.
  140. I. Jung, S. Shin, M. C. Baek and K. Yea, Modification of immune cell-derived exosomes for enhanced cancer immunotherapy: current advances and therapeutic applications, Exp. Mol. Med., 2024, 56(1), 19–31 Search PubMed.
  141. Z. Zhao, Y. Chen, N. M. Francisco, Y. Zhang and M. Wu, The application of CAR-T cell therapy in hematological malignancies: advantages and challenges, Acta Pharm. Sin. B, 2018, 8(4), 539–551 Search PubMed.
  142. T. Smyth, K. Petrova, N. M. Payton, I. Persaud, J. S. Redzic, M. W. Graner, P. Smith-Jones and T. J. Anchordoquy, Surface functionalization of exosomes using click chemistry, Bioconjugate Chem., 2014, 25(10), 1777–1784 Search PubMed.
  143. V. Johnson, S. Vasu, U. S. Kumar and M. Kumar, Surface-Engineered Extracellular Vesicles in Cancer Immunotherapy, Cancers, 2023, 15(10), 2838 Search PubMed.
  144. J. He, W. Ren, W. Wang, W. Han, L. Jiang, D. Zhang and M. Guo, Exosomal targeting and its potential clinical application, Drug Delivery Transl. Res., 2022, 12(10), 2385–2402 Search PubMed.
  145. A. Liu, G. Yang, Y. Liu and T. Liu, Research progress in membrane fusion-based hybrid exosomes for drug delivery systems, Front. Bioeng. Biotechnol., 2022, 10, 939441 Search PubMed.
  146. R. Raguraman, D. Bhavsar, D. Kim, X. Ren, V. Sikavitsas, A. Munshi and R. Ramesh, Tumor-targeted exosomes for delivery of anticancer drugs, Cancer Lett., 2023, 558, 216093 Search PubMed.
  147. Y. J. Li, J. Y. Wu, J. Liu, W. Xu, X. Qiu, S. Huang, X. B. Hu and D. X. Xiang, Artificial exosomes for translational nanomedicine, J. Nanobiotechnol., 2021, 19(1), 242 Search PubMed.
  148. Q. Lv, L. Cheng, Y. Lu, X. Zhang, Y. Wang, J. Deng, J. Zhou, B. Liu and J. Liu, Thermosensitive Exosome-Liposome Hybrid Nanoparticle-Mediated Chemoimmunotherapy for Improved Treatment of Metastatic Peritoneal Cancer, Adv. Sci., 2020, 7(18), 2000515 Search PubMed.
  149. M. Piffoux, A. K. A. Silva, C. Wilhelm, F. Gazeau and D. Tareste, Modification of Extracellular Vesicles by Fusion with Liposomes for the Design of Personalized Biogenic Drug Delivery Systems, ACS Nano, 2018, 12(7), 6830–6842 Search PubMed.
  150. X. Wang, D. Li, G. Li, J. Chen, Y. Yang, L. Bian, J. Zhou, Y. Wu and Y. Chen, Enhanced Therapeutic Potential of Hybrid Exosomes Loaded with Paclitaxel for Cancer Therapy, Int. J. Mol. Sci., 2024, 25(7), 3645 Search PubMed.
  151. Y. T. Sato, K. Umezaki, S. Sawada, S. A. Mukai, Y. Sasaki, N. Harada, H. Shiku and K. Akiyoshi, Engineering hybrid exosomes by membrane fusion with liposomes, Sci. Rep., 2016, 6, 21933 Search PubMed.
  152. Y. Lin, J. Wu, W. Gu, Y. Huang, Z. Tong, L. Huang and J. Tan, Exosome-Liposome Hybrid Nanoparticles Deliver CRISPR/Cas9 System in MSCs, Adv. Sci., 2018, 5(4), 1700611 Search PubMed.
  153. S. Rayamajhi, T. D. T. Nguyen, R. Marasini and S. Aryal, Macrophage-derived exosome-mimetic hybrid vesicles for tumor targeted drug delivery, Acta Biomater., 2019, 94, 482–494 Search PubMed.
  154. Y. Y. Jhan, D. Prasca-Chamorro, G. Palou Zuniga, D. M. Moore, S. Arun Kumar, A. K. Gaharwar and C. J. Bishop, Engineered extracellular vesicles with synthetic lipids via membrane fusion to establish efficient gene delivery, Int. J. Pharm., 2020, 573, 118802 Search PubMed.
  155. M. Mendt, K. Rezvani and E. Shpall, Mesenchymal stem cell-derived exosomes for clinical use, Bone Marrow Transplant., 2019, 54, 789–792 Search PubMed.
  156. S. Sonar, Clinical trial status of exosomes-based cancer theranostics, Clin. Transl. Discovery, 2024, 4, e327 Search PubMed.
  157. B. Zhou, K. Xu, X. Zheng, T. Chen, J. Wang, Y. Song, Y. Shao and S. Zheng, Application of exosomes as liquid biopsy in clinical diagnosis, Signal Transduction Targeted Ther., 2020, 5(1), 144 Search PubMed.
  158. X. Y. Xue, Y. X. Liu, C. Wang, X. J. Gu, Z. Q. Xue, X. L. Zang, X. D. Ma, H. Deng, R. Liu, L. Pan and S. H. Liu, Identification of exosomal miRNAs as diagnostic biomarkers for cholangiocarcinoma and gallbladder carcinoma, Signal Transduction Targeted Ther., 2020, 5(1), 77 Search PubMed.
  159. M. Barceló, M. Castells, L. Bassas, F. Vigués and S. Larriba, Semen miRNAs Contained in Exosomes as Non-Invasive Biomarkers for Prostate Cancer Diagnosis, Sci. Rep., 2019, 9(1), 13772 Search PubMed.
  160. J. Rezaie, M. Feghhi and T. Etemadi, A review on exosomes application in clinical trials: perspective, questions, and challenges, Cell Commun. Signaling, 2022, 20(1), 145 Search PubMed.
  161. E. J. Bunggulawa, W. Wang, T. Yin, N. Wang, C. Durkan, Y. Wang and G. Wang, Recent advancements in the use of exosomes as drug delivery systems, J. Nanobiotechnol., 2018, 16(1), 81 Search PubMed.
  162. Z. Xu, S. Zeng, Z. Gong and Y. Yan, Exosome-based immunotherapy: a promising approach for cancer treatment, Mol. Cancer, 2020, 19(1), 160 Search PubMed.
  163. J. Wang, D. Chen and E. A. Ho, Challenges in the development and establishment of exosome-based drug delivery systems, J. Controlled Release, 2021, 329, 894–906 Search PubMed.
  164. X. Fang, Y. Wang, S. Wang and B. Liu, Nanomaterials assisted exosomes isolation and analysis towards liquid biopsy, Mater. Today Bio, 2022, 16, 100371 Search PubMed.
  165. Y. Yang, Z. Wu, L. Wang, K. Zhou, K. Xia, Q. Xiong, L. Liu, Z. Zhang, E. R. Chapman, Y. Xiong, T. J. Melia, E. Karatekin, H. Gu and C. Lin, Sorting sub-150-nm liposomes of distinct sizes by DNA-brick-assisted centrifugation, Nat. Chem., 2021, 13(4), 335–342 Search PubMed.
  166. B. M. Hussen, G. S. H. Faraj, M. F. Rasul, H. J. Hidayat, A. Salihi, A. Baniahmad, M. Taheri and S. Ghafouri-Frad, Strategies to overcome the main challenges of the use of exosomes as drug carrier for cancer therapy, Cancer Cell Int., 2022, 22(1), 323 Search PubMed.
  167. H. Yan, Y. Li, S. Cheng and Y. Zeng, Advances in Analytical Technologies for Extracellular Vesicles, Anal. Chem., 2021, 93(11), 4739–4774 Search PubMed.
  168. R. T. Morales and J. Ko, Future of Digital Assays to Resolve Clinical Heterogeneity of Single Extracellular Vesicles, ACS Nano, 2022, 16(8), 11619–11645 Search PubMed.
  169. I. Li and B. Y. Nabet, Exosomes in the tumor microenvironment as mediators of cancer therapy resistance, Mol. Cancer, 2019, 18(1), 32 Search PubMed.
  170. L. Li, D. He, Q. Guo, Z. Zhang, D. Ru, L. Wang, K. Gong, F. Liu, Y. Duan and H. Li, Exosome-liposome hybrid nanoparticle codelivery of TP and miR497 conspicuously overcomes chemoresistant ovarian cancer, J. Nanobiotechnol., 2022, 20(1), 50 Search PubMed.
  171. R. Dhar and A. Devi, Engineered cell versus modified exosomes in cancer therapy, Clin. Transl. Discovery, 2024, 4(3), e320 Search PubMed.
  172. R. Dhar, A. Devi, S. Patil and M. R. Tovani-Palone, Exosomes in cancer therapy: Advances and current challenges, Electron. J. Gen. Med., 2023, 20(5), em524 Search PubMed.
  173. M. Lu, H. Xing, X. Zhao, Y. Huang, A. Zheng and X. J. Liang, Engineered extracellular vesicles as a next-generation vaccine platform, Matter, 2024, 7(12), 4180–4205 Search PubMed.
  174. S. Ning, P. Shangguan, X. Zhu, X. Ou, K. Wang, M. Suo, H. Shen, X. Lu, X. Wei, T. Zhang, X. Chen and B. Z. Tang, Pyridinium Rotor Strategy toward a Robust Photothermal Agent for STING Activation and Multimodal Image-Guided Immunotherapy for Triple-Negative Breast Cancer, J. Am. Chem. Soc., 2025, 147(9), 7433–7444 Search PubMed.
  175. S. Ning, X. Zhang, M. Suo, M. Lyu, Y. Pan, Y. Jiang, H. Yang, J. W. Lam, T. Zhang, L. Pan and B. Z. Tang, Platelet-derived exosomes hybrid liposomes facilitate uninterrupted singlet oxygen generation to enhance breast cancer immunotherapy, Cell Rep. Phys. Sci., 2023, 4(7), 101505 Search PubMed.
  176. S. Rayamajhi, T. D. T. Nguyen, R. Marasini and S. Aryal, Macrophage-derived exosome-mimetic hybrid vesicles for tumor targeted drug delivery, Acta Biomater., 2019, 94, 482–494 Search PubMed.
  177. D. Mirgh, S. Sonar, S. Ghosh, M. D. Adhikari, V. Subramaniyan, S. Gorai and K. Anand, Landscape of exosomes to modified exosomes: a state of the art in cancer therapy, RSC Adv., 2024, 14(42), 30807–30829 Search PubMed.
  178. A. Liu, G. Yang, Y. Liu and T. Liu, Research progress in membrane fusion-based hybrid exosomes for drug delivery systems, Front. Bioeng. Biotechnol., 2022, 10, 939441 Search PubMed.

This journal is © The Royal Society of Chemistry 2026
Click here to see how this site uses Cookies. View our privacy policy here.