Skip to main content

Exosomes: a review of biologic function, diagnostic and targeted therapy applications, and clinical trials

Abstract

Exosomes are extracellular vesicles generated by all cells and they carry nucleic acids, proteins, lipids, and metabolites. They mediate the exchange of substances between cells,thereby affecting biological properties and activities of recipient cells. In this review, we briefly discuss the composition of exocomes and exosome isolation. We also review the clinical applications of exosomes in cancer biology as well as strategies in exosome-mediated targeted drug delivery systems. Finally, the application of exosomes in the context of cancer therapeutics both in practice and literature are discussed.

Introduction

Exosomes are classified as small (30–150 nm), phospholipid bilayer extracellular vehicles released by both prokaryotic and eukaryotic cells for intercellular communication and signaling [1]. Historically, exosomes were considered extracellular vesicles that excrete unwanted cellular waste. However, further research has demonstrated that exosomes are important molecular mediators for cellular communication for transporting proteins, metabolites, and various nucleic acids throughout the body [2, 3] (Fig. 1). Exosomes are also secreted by a wide variety of cells, including immune cells [4], cancer cells [4], and stem cells [5]. Due to their multiple functions in transmitting information between cells, exosomes play a critical role in physiological regulation, disease progression, immune response, and disease development [6].

Fig. 1
figure 1

Biosynthesis and diversity of exosomes. In their early stage of formation in living cells, exosomes are produced through an endosomal pathway and released into the extracellular space. Many cellular contents, such as proteins, lipids, metabolites, small molecules, DNA, RNA, and cell membrane surface proteins, are enclosed in exosomes during this process and fuse with the recipient cells via endocytosis and plasma membrane invagination. Exosomes released from donor cells can be internalized through endocytosis or membrane fusion and can also trigger a biological response in the recipient cell through interaction with cell surface proteins or receptors

Exosomes are produced naturally within the human body and are not prone to immunogenicity, which would otherwise elicit a host response [7]. Clinically, they have been regarded as ideal candidates for applications related to biomarker development and early disease detection [8]. Furthermore, exosomes have garnered recent attention for their potential as drug delivery vehicle, which could improve factors like bioavailability of cargo load, side effect profiles, off-target effect, and pharmacokinetics for drug molecules [9, 10].

Due to their ability to transport cellular material systemically as well as reflect the physiological state of donor cells, exosomes can influence various processes related to inflammation, central nervous system communication, immune response, and different types of tissue repair [11]. Their phospholipid bilayer membrane contains lipid structures like ceramide and cholesterol, which assist in sorting, secretion, and signaling between host cell and extracellular environment [12]. The abundance of RNA species within exosomes plays a critical role in various biological processes, especially in different disease states and progression factors. For example, exosomes are involved in the transfer of pathogens such as virus particles [13] and nucleic acids such as circular RNAs (circRNAs) associated with tumor progression [14]. Some studies have shown that exosomes can serve as potential biomarkers for early diagnosis and monitoring of diseases like cancer [15,16,17]. Accordingly, exosomes may be considered an important therapeutic tool for treatment and early disease detection.

In this review, we provide an overview of the exosome field. We review the unique properties and diverse content of exosomes and current methods to isolate them. In addition, we discuss clinical applications of exosomes in the context of cancer biology. Exosome-mediated targeted drug delivery as well as a current case study also discussed. Finally, we provide an overview of exosomes currently under review in clinical trials and provide insight on future challenges.

The complex architecture of exosomes and their isolation methods

Many constitutive elements have been identified in exosomes from different cell types— including approximately 4400 proteins, 194 lipids, 1639 mRNAs, and 764 miRNAs—illustrating their complexity and potential functional diversity [18]. Typically, exosomes are highly enriched proteins with multiple functions, such as transmembrane proteins (CD9, CD81, CD82), which take part in cell penetration, invasion, and fusion; heat shock proteins (HSP70, HSP90) for stress response in antigen binding and presentation; MVB formation proteins (TSG101 Alix) for exosome release; and proteins for membrane transport and fusion (annexin and Rab) [19, 20]. A list of protein components commonly found in exosomes is shown in Table 1 [21,22,23].

Table 1 Common protein components of exosomes

In addition to select proteins, exosomes also contain multiple patterns of RNAs that can be incorporated into recipient cells. RNA sequencing analysis has demonstrated that microRNAs (miRs) are most abundant in human plasma-derived exosomal RNA species, making up over 42% of all raw reads and 76% of all mappable reads [27]. Other RNA species include ribosomal RNA, long non-coding RNA, piwi-interacting RNA, transfer RNA, small nuclear RNA, and small nucleolar RNA. Once miRs are packed into exosomes, they undergo unidirectional transfer between cells, resulting in an intercellular trafficking network, eliciting transient or persistent phenotypic changes of recipient cells [28]. Besides miRs, other proteins like long RNA species (e.g., long non-coding RNAs and circular RNAs) have also been identified in exosomes that impact various biological processes, including cancer development [29].

Exosomes are are not only classified according to protein and nucleic acids composition but also by their lipid components. Generally, exosomes are enriched in phosphatidyl-serine (PS), phosphatidic acid, cholesterol, sphingomyelin, arachidonic acid and related fatty acids, prostaglandins, and leukotrienes – all of which account for their stability and structural integrity (Table 2) [10, 30,31,32,33,34]. Moreover, exosomes also have some functional lipolytic enzymes, which produce bioactive lipids autonomously. These exosomal bioactive lipids may be internalized into recipient cells and concentrate lipid mediators within the endosomes. Recent studies have revealed that the accumulation of prostaglandins and fatty acids brought by exosomes can trigger the activation of cell-to-cell phospholipases and lipid mediators [33]. Because exosomes have been found to be commonly released and taken up by target cells to modulate cell lipid metabolism, studies have examined their role in lipid-related pathologies (atherosclerosis) as well as their potential role as a molecular signal for disease diagnosis and prognosis [35].

Table 2 Lipid-related enzymes and bioactive lipids in exosomes

Traditional exosome isolation and purification methods mainly involve density gradient centrifugation [42, 43], size exclusion chromatography, and immunoaffinity purification [44, 45]. Ultracentrifugation is one of the most commonly used methods for exosome isolation [42, 46, 47]. This method is dependent on the fact that exosomes are smaller and denser than cells and can be separated by centrifugation at different speeds. Density gradient centrifugation is a separation method based on density differences of exosomes [42, 43]. In this method, fractionating macromolecules like sucrose, glucose, and proteins are poured into ultracentrifuge tubes. Exosomes localize and can be isolated -in a centrifuge tube according to different-sized macromolecules and their known density levels.

Size-exclusion chromatography is another separation method based on exosome size differences [48,49,50]. This method involves adding the sample to a size-exclusion chromatography column and using the filler to perform molecular screening so that larger cell fragments, proteins, and other molecules are screened while exosomes are discharged through the bottom.

Immunoaffinity purification is a separation method based on the binding affinity between exosome surface marker molecules (such as CD63, CD9, etc.) and specific antibodies [42, 51, 52]. This method mixes an affinity column containing specific antibodies with the sample and uses the binding force between the antibodies and labeled molecules to capture exosomes in a column, followed by elution and purification. This method has the advantages of solid selectivity and a good purification effect.

Exosomes and cancer biology

Exosomes promote tumor cell proliferation

Recent studies demonstrate cell-derived exosomes play an important role in tumor growth and progression [53]. For example, recombinant epidermal growth factor (EGF) treatment can promote the uptake of oral squamous cell carcinoma (OSCC) cell-derived exosomes into the OSCC cells. In contrast, EGF receptor (EGFR) knockdown or EGFR inhibitors, including erlotinib and cetuximab, abolishes exosome uptake by OSCC cells [53]. Cell-derived exosomes have also been shown to induce proliferation, migration, invasion, stemness, and chemoresistance of OSCC cells [53]. These biomolecules can activate signaling pathways and promote tumor cell proliferation by binding to receptors on the surface of tumor cells.

Exosomes promote tumor cell metastasis

Extensive research on the tumor microenvironment has shown that cancer cell-derived exosomes are involved in crucial elements of cancer progression, including angiogenesis, premetastatic niche, extracellular matrix formation, epithelial mesenchyme transformation (EMT), cancer stem cell growth, and treatment resistance [54]. One study confirmed that cancer-associated fibroblast (CAF)-derived exosomes are a key driver in ovarian cancer (OVCA) tumor progression [55]. This study revealed specific circRNA (hsa_circIFNGR2) molecule can be used to elucidate the novel function of CAF-derived exosome circIFNGR2 on OVCA cell growth and metastasis. Metastatic organ tropism remains one of the greatest mysteries of cancer etiology since the "seed and soil" hypothesis. Scientists found that EGFR in exosomes secreted by gastric cancer cells can be delivered to the liver and integrated into the plasma membrane of liver stromal cells. Translocated EGFR has been shown to effectively activate hepatocyte growth factor (HGF) by inhibiting the expression of miR-26a/b. In addition, the upregulated paracrine HGF binds to the c-MET receptor on migrating cancer cells, providing fertile "soil" for "seeds" and promoting the landing and proliferation of metastatic cancer cells [36].

Exosomes and the immune host response

During tumor growth, exosome components released by tumor cells can inhibit the normal function of the immune system, thereby avoiding attack and clearance by the host immune system. This inhibitory effect is mainly achieved by regulating the activity of immune cells, promoting immune cell apoptosis, and reducing the number of immune cells. The precise mechanism by which exosomes are involved in tumor progression remains elusive, multifaceted, and a double-edged sword, thus requiring further clarification. Exosomes can facilitate communication between innate immune cells and tumor cells to support or inhibit tumor progression. A past review focused on exosome-mediated intercellular communication between tumor cells and macrophages, neutrophils, mast cells, monocytes, dendritic cells, and natural killer cells [4].

Exosome subtypes and tumor regulation

miRNA

microRNA can be used as a biomarker for the diagnosis of cancer. For example, pancreatic cancer is one of the leading causes of cancer death worldwide. Plasma microRNA is a promising player due to its non-invasive and practical use in tumor diagnosis and prognosis. Recent studies have shown that compared with healthy controls, microRNAs, namely miR-125b-3p, miR-122-5p and miR-205-5p, in plasma exosomes of pancreatic cancer patients are potential plasma exosome-derived of non-invasive biomarkers [56]. In addition to being used as a molecular marker for clinical diagnosis, microRNA in exosomes can also affect the growth of tumor cells by transferring into cells and changing their gene expression. For example, miR-142-3p is one of the miRs upregulated in various breast cancers. It activates the canonical Wnt signaling pathway and transactivates the expression of miR-150, leading to excessive proliferation of cancer cells in vitro and in vivo [57]. The miR in exosomes can also affect the tumor microenvironment. Communication between endothelial cells and tumor cells in the tumor microenvironment is required for cancer metastasis. Previous studies have confirmed that miR-29a expression is upregulated in colorectal cancer (CRC) tissues, and EMT-CRC cells may transport exosomal miR-29a to endothelial cells in the tumor microenvironment to increase vascular permeability, thus promoting the development and metastasis of CRC [58]. This process makes exosomal miR-29a a potential predictive marker for tumor metastasis and a viable therapeutic target for CRC. The miR in exosomes has an essential impact on tumor growth; therefore, future research requires further exploration on the mechanism of miR in exosomes on tumor growth and metastasis and develop new treatment strategies and markers.

Proteins

Proteins (such as TTN-AS1, Rab27b) in exosomes can affect tumor growth through multiple pathways. First, they can directly act on cancer cells to promote proliferation and metastasis [59, 60]. Secondly, exosome proteins can affect the tumor microenvironment, indirectly affecting tumor growth [61]. For example, exosome proteins (SYT7) can promote tumor angiogenesis and provide an adequate supply of nutrients and oxygen, thereby promoting tumor growth and metastasis [62]. Third, some exosome proteins can regulate tumor immune response and inhibit the immune system's reaction to tumors, thereby promoting tumor evasion. Many proteins in exosomes related to tumors have been discovered [63], and these proteins have been confirmed as targets for cancer therapy.

Lipids

Recent studies have shown that exosome lipids are important factors in tumor growth. These lipids mainly include phospholipids, cholesterol, and triacylglycerol. All these types of exosomes regulate tumor growth and metastasis through various mechanisms. First, exosome phospholipids can affect tumor cell growth by changing the composition and structure of cell membranes, thereby affecting signaling and membrane permeability. Enteropathogenic bacterial secretory particles were recently found to stimulate the intestinal epithelium to produce IDEN (intestinal mucosa-derived exosome-like nanoparticles), which contain elevated levels of sphingosine-1-phosphate, CCL20, and prostaglandins E2 (PGE2). As we know, gut-related inflammation plays a crucial role in the progression of colon cancer. In tumor immunity, it has also been found that the recruitment and proliferation of Th17 cells require CCL20 and PGE2, respectively. Demonstrated biological effects of sphingosine-1-phosphate contained in IDEN facilitate tumor growth in spontaneous and transplanted colon cancer mouse models [64]. In addition, phospholipids can serve as signaling molecules to participate in processes such as apoptosis and autophagy, thereby regulating tumor growth and metastasis. Triglycerides in exosomes can also affect tumor growth, since they are essential substances in lipid metabolism and can provide energy and nutrients for cell growth. Studies have shown that triglyceride synthesis and metabolic pathways in tumor cells are abnormally active, which may be related to their proliferation and metastasis capabilities.

Emerging applications of exosomes in cancer diagnosis and therapy

Exosomes as diagnostic tools

As previously mentioned, exosomes are present in many biofluids, which presents an opportunity to monitor cell status under normal or pathological conditions. In biopsies (liquid, plasma, serum, urine, saliva), exosomes have been used to analyze and determine a patient diagnosis, prognosis, progress, and chemoresistance status [65, 66]. Furthermore, up-regulated exosome secretion has been found in several complex conditions including cancer. Under pathological conditions, cellular changes can be monitored based on exosomes released by cells. Discrepancies in levels of specific molecules can be identified and analyzed by transcriptomics, proteomics, and lipidomic investigations [67]. The use of exosomes for disease diagnosis has been investigated in early diagnosis and prognosis of cancer [15,16,17] and cardiovascular diseases [68, 69].

Exosomes in blood samples for cancer detection has gained recent attention. In a retrospective study [16], researchers combined artificial intelligence and surface-enhanced Raman spectroscopy technology to successfully detect exosomes in early-stage cancers (lung, breast, and colon, liver, pancreatic, and gastric cancer). The study also includes a classification model to identify plasma exosome signaling patterns to identify their presence and tissue of origin. The final comprehensive decision model showed a sensitivity of 90.2% and a specificity of 94.4%, while predicting tumor organelles in 72% of positive patients. In colorectal cancer, exosomes have emerged as promising biomarkers for diagnosis due to their rich biological fingerprints and high stability. Recent research highlights the construction of an exosome enrichment platform on a 3D porous sponge microfluidic chip [15], whereby the exosome capture efficiency of the chip is approximately 90%. Deep mass spectrometry analysis and multilevel expression screening reveal a CRC-specific exosomal membrane protein (SORL1). A specific quantum dot-labeled SORL1 detection method was further designed. The systems demonstrated similar diagnostic performance in treating patients with early-stage CRC, young CRC, and CEA-negative CRC.

Compared with traditional biomarker detection methods, using exosomes for disease diagnosis has the following advantages: (1) High sensitivity: Exosomes can carry many biomarkers and, therefore, have high sensitivity. (2) High specificity: Exosomes can carry cell-specific biomarkers and therefore have high specificity. (3) Non-invasive: Exosome collection can be completed through conventional body fluid methods without invasive operations such as tissue sectioning or puncture. (4) Easy to store: The collected exosomes can be stored for a long time through cryopreservation and other methods to facilitate subsequent analysis. Although using exosomes for disease diagnosis has many advantages, several challenges still exist: (1) Standardization issues: Due to the current lack of unified exosome collection, processing, and detection standards, results may vary between different laboratories. (2) Technical issues: The current detection technology for exosomes is not mature enough, and new detection technologies need to be further developed. (3) Sample quantity issue: Since the current understanding of exosomes is not deep enough, many samples are needed for verification and confirmation.

Exosomes in disease treatment

The application of exosomes in cancer treatment mainly depends on two strategies: (1): use as drug carriers to encapsulate drugs [70]; (2) use as bioactive molecules (e.g. miRs) to regulate the tumor microenvironment and inhibit tumor growth and metastasis. Demonstration studies have shown that using exosomes as drug carriers can improve drug inhibition and bioavailability and reduce drug side effects [71, 72]. This kind of RNA nanoparticles can improve the targeting ability and deliver therapeutic drugs to specific cancer cells. It has been reported that RNA nanoparticles can be used to display chemical ligands (such as folic acid [72, 73]), chemical drugs, or RNA aptamers on the surface of exosomes. These combinations can deliver small-molecule chemical drugs to a target via chemical ligands or RNA aptamers. By binding RNA nanoparticles to specific tumor cells, the dose and side effects of the drug can be reduced to achieve the purpose of targeted cancer treatment [74]. In addition, RNA nanoparticles modified on exosomes can also combine with siRNA[75, 76], miR [77,78,79,80], RNA aptamers, or ligands to specifically inhibit cancer cells.

Exosomes in targeted drug delivery

Compared to liposomes and nanoparticle delivery systems which are synthesized in vitro, exosomes originate from the body and offer better biocompatibility and lower immunogenicity than their counterparts [81]. Furthermore, due to the heterogeneity of exosomes, they carry various proteins on the surface, which enter cells through a variety of ways after initial cell contact. Among them, receptor-mediated endocytosis is one main way information and drug transport can be achieved in target tissues [82].

Exosomes have been explored for their inherent ability to be loaded with both small and large molecules in support of their therapeutic use for treatment of various diseases [83]. The construction of engineered exosomes loaded with drugs can provide an efficient, less toxic, and more targeted drug delivery strategy. In addition, exosomes carry different peptides to target specific cells, which provides a strategy to construct more specific drug delivery mechanism. For example, Cui et al. engineered exosomes to deliver siShn3 for anti-osteoporosis functions in mouse models and demonstrated bone targeting capability in vitro and in vivo to achieve osteogenesis [84].

There are two main methods to encapsulate drugs in exosomes: natural and artificial. Natural packaging refers to using the biological properties of exosomes to package drugs inside, while artificial packaging refers to the application of chemical or physical methods to guide drugs into exosomes [85]. Both methods are described in detail below.

Natural drug encapsulation

Exosomes have natural packaging properties and can encapsulate biomolecules inside and release them outside of cells. This natural wrapping mechanism is mainly achieved through modifying the cell membrane's protein, glycosylation, and lipid composition. Drugs can be encapsulated inside and released outside the cells by interacting with these biomolecules on the exosome membrane. The process of encapsulating drugs in exosomes is mainly achieved through the intracellular secretory pathway. Cells wrap drugs or other biomolecules in a membrane to form exosomes. These exosomes can be released by cells, enter the blood circulation system, and be transported to areas that require treatment [86]. The advantage of exosome-encapsulated drugs is that they can cross biological barriers, such as the blood–brain barrier, allowing drugs to reach hard-to-reach sites. In addition, exosomes can protect drugs from enzymatic degradation and immune system attack [87]. Exosomes can also achieve targeted delivery by modulating molecules on their surface. For example, in some diseases, molecules on the cell surface change, and these changes can be recognized by exosomes. Therefore, scientists can change the specific molecules, including drugs, on the surface of exosomes so they can be delivered to specific cells or tissues [88, 89]. In addition, exosomes can also achieve different delivery effects by changing their size and shape [24]. For example, smaller exosomes can pass through biological barriers more efficiently, while spherical exosomes can be more easily engulfed by cells. However, there are still some challenges and problems with exosome-encapsulated drugs. Controlling the speed and location of drug release from exosomes remains a complex issue, and the yield and purity of exosomes also need further improvement [25].

Artificial drug encapsulation

Artificial encapsulation refers to guiding drugs into exosomes through chemical or physical means. This method can effectively improve drug packaging efficiency and control drug release speed. At present, there are mainly the following methods for manual packaging. (1) Electroporation method: Electroporation refers to mixing exosomes and drugs and using an electric field to cause the drugs to penetrate the exosome membrane and enter the interior. This method can achieve efficient drug encapsulation and release but requires high technical requirements and equipment support [26]; (2) Chemical cross-linking method: The chemical cross-linking method refers to using cross-linking agents to cross-link drugs with exosome membranes to achieve drug encapsulation and release. This method can control the drug release speed and time and realize the simultaneous encapsulation of multiple drugs [37]; Thermal shock method: The thermal shock method refers to mixing exosomes with drugs and then changing them through high or low-temperature treatment to achieve the encapsulation and release of drugs. This method is simple to operate but requires precise temperature and time control [38].

Recent studies have examined the therapeutic viability of loaded exosomes and whether cargo targeted delivery to the intended site of action works [90]. One group utilized drug-resistant A2780/DDP cells to test effectiveness of cisplatin-loaded exosomes against cisplatin alone to compare and determine therapeutic viability [91]. The study found that by using cisplatin-loaded exosomes, the cytotoxicity of cisplatin increased by factor of three in drug resistant cells and a factor of 1.4 in drug sensitive cells versus cisplatin alone. Another study tested therapeutic viability through loading blood-derived exosomes with dopamine for treatment of Parkinson’s Disease [92]. The authors found there was greater than 15-fold increase in bioavailability of dopamine in the brain through use of exosome delivery platform. As previous research has shown, the delivery of loaded exosomes can prove to be efficient in drug therapy that potentially avoids challenges that other platforms have encountered including first pass metabolism, limited bioavailability, and blood brain barriers.

Strategies in exosome cargo loading

Due to their high biocompatibility, stability, and low immunogenicity, exosomes have been considered favorable carriers for drug delivery. The lipid bilayer membrane of exosomes also offers protection against cargo degradation, making them a desirable candidate for drug delivery. Several cargo delivery strategies exist involving small-molecule, nucleic acid, and protein drugs (Fig. 2). Recent studies have focused on modifying various aspects of exosome to substantially enhance their targeting ability.

Fig. 2
figure 2

Cargo loading methods for exosome therapeutics. Endogenous cargo delivery with plasmids or viruses involves constructing transgenes and expressing them in vector cells, and then collecting endogenous cargo-packaged exosomes from the cell culture supernatant. These exosomes can express special antigenicity. Endogenous cargo loading often involves miRNAs and proteins with therapeutic efficacy. In exogenous cargo loading, delivery of drugs or therapeutic small molecules involves electroporation, co-incubation, or ultrasound to carry exogenous cargo

Phospholipid modification: Since exosomes mainly comprise phospholipids, phospholipid modification has become a common strategy. Changing the type and content of phospholipids on the surface of exosomes can affect their stability, targeting, and immunogenicity [39]. For example, the blood–brain barrier (BBB) precludes the entrance of most pharmaceutical drug compounds into the brain. Clinically, delivery across BBB tight junctions is achieved through various endogenous transport mechanisms. Therefore, receptor-mediated transcytosis (RMT) through exosomal cell membrane components and BBB cells is one of the most widely studied and used methods to allow delivery of drugs into the brain. A variety of strategies exist involving use of exosomes to can express specific proteins through plasmids or via mechanical coating on exosomes Receptors that mediate transcytosis bind specific ligands to hijack RMT, such as transferrin receptor (TfR), low-density lipoprotein receptor (LDLR), and insulin receptor (INSR). Cell-penetrating peptides and viral components derived from neurotropic viruses can also be used for effective BBB crossing of therapeutic drugs [40].

Protein modification: Proteins on the surface of exosomes can be modified through glycosylation, acetylation, phosphorylation, etc. These modifications can affect exosome targeting, stability, and immunogenicity properties. Previous study have demonstrated modifying exosome surface proteins can affect their interaction with the liver microenvironment, thereby promoting gastric cancer metastasis to the liver [36]. In other studies, targeted delivery of exosomes to the brain can be achieved by labeling various targeting moieties on the surface of exosomes. Therapeutic exosomes can be engineered through chemical modifications (e.g., click chemistry) to express various targeting moieties or genetic modification of exosome-producing cells to express targets fused to exosome membrane-associated components toward peptides such as Lamp2b and tetraspanins (Fig. 3) [41].

Fig. 3
figure 3

Chemical and protein modifications on exosomes to improve targeting efficacy. A Synthetic chemicals (such as folic acid) are combined with exosomes through chemical modification (click chemistry). The drug-carrying exosomes can be targeted to the receptors of cancer cells (such as folate receptors) through folic acid. Targeting peptides can also be fused to express (such as Lamp2b and tetraspanins) through genetic modification of exosome-producing cells (as described in Fig. 2). These synthetic short peptides can cross the blood–brain barrier. B Receptor mediated transcytosis (RMT) is a widely used method to allow exosomes to pass through blood brain barrer tight junctions. Surface-modified exosomes possessing brain targeting have shown enhanced CNS delivery

Chemical decoration: The properties of the exosome surface bilayer can be modified by cholesterol-binding RNA nanoparticles. These RNA nanoparticles can be used to display chemical ligands (such as folic acid), chemical drugs, or RNA adapters on the exosome surface. body. Folate receptors are abundantly expressed on many human tumor cells. Therefore, folic acid modified on the surface of exosomes can improve the targeting ability of RNA nanomedicines and deliver therapeutic drugs in exosomes to specific cancer cells [72, 73].

Case study: exosomes and siRNA nanotechnology

Ribonucleotide reductase M2 subunit (RRM2) is a dimer containing a tyrosine free radical and a non-heme iron, essential for enzyme activity for DNA synthesis and damage repair [93, 94]. When DNA replication occurs, RRM2 is expressed during late G1/early S phase. Overexpression of RRM2 enhances the metastatic and invasive capacity of human cancer cells [95,96,97] and leads to poor prognosis in patients [98, 99]. RRM2 also promotes tumor angiogenesis through regulating expression of proangiogenic factor VEGF and antiangiogenic factor TSP-1 [100]. Previous reports indicated that the antisense molecules to RRM2 effectively decreases RRM2 expression, inhibits enzyme activity and reduces growth of cancer cells in vitro and in vivo [101,102,103]. Here, to leverage the targeting effect and therapeutic potential of folate RNA aptamer for tumor treatment, we designed an arrow-tail RNA nanoparticle to display folate RNA aptamer on the surface of exosomes to deliver M2 siRNA folate RNA Apt/Exo/siM2 for cancer treatment.

In the following case study, we employed RNA nanotechnology to display RNA ligand on exosomes to serve as a carrier for siRNA. An RNA ligand that was anchored on exosomes by cholesterol targets folate receptors to recognize cancer cells [104, 105]. Once entry into the cancer cells was achieved, siRNA silenced the anti-apoptotic factor M2 subunit of ribonucleotide reductase (RRM2). In comparison with other delivery systems with the cyclodextrin-containing polycation nanoparticles to protects siRNA [106], a human transferrin protein (hTf) ligand was displayed on the nanoparticle to target Tf receptors (hTfR) on the cancer cells [107]. The siRNA carried by the nanoparticle also silences RRM2 [108]. Based on in vivo results, the exosome-delivered siRNA silencing RRM2 demonstrated noticeable melanoma growth regression, suggesting the RNA-targeted exosome delivery system with RRM2siRNA could attenuate tumor growth.

Suppression of melanoma and colorectal carcinoma growth in vivo by FOLATEApt/Exo/siM2

We chose melanoma cell lines, A375, for investigation because of the high RRM2 mRNA expression level. A375 cells were incubated with 50 nM Alexa647 labeled-RNA loaded exosome (Fig. 4A). Approximately 86.04% ~ 92.47% of fluorescence positive cells were observed (Fig. 4B). Flow cytometry of A375 cells treated with 3200nM of FOLATEApt/Exo/siM2 demonstrated the increase of the dead cell population in the treated cell line in contrast to the control (Fig. 4C) while the apoptotic cell populations did not differ between the treated and the control (Fig. 4C). Thus, FOLATEApt/Exo/siM2 causes cancer cell death in vitro.

Fig. 4
figure 4

Exo/siM2 fused with cells when A375 cells were incubated with Alexa647 labeled-RNA for 1 h. A The construction of exosomes that display the RNA/folate complex with Alexa647. B Flow cytometry result. After A375 cells incubated with 50 nM Alexa647 labeled-RNA loaded exosome, high percentage of fluorescence positive cells were detected. C A375 cells were incubated with 3200nM Exo/siM2 and D exhibited a significant increase of dead cells and apoptotic cells. The results are presented as the mean ± SD. *P < 0.05

Fluorescent microscopy of A375 cells showed the red fluorescence inside the cells, indicating FOLATEApt/Exo/siM2 entered the cells (Fig. 5A). The in vivo melanoma targeting and anti-tumor efficacy of FOLATEApt/Exo/siM2 were investigated in a tumor xenograft NSG mouse model. The results indicates that the fully packaged exosomes with folate display and siRNA load reduced tumor volume by 100%, compared to siRNA by 77% and folate by 64%. The inhibition was FOLATEApt/Exo/siM2 packaging-dependent (Fig. 5B-D). Therefore, the packaging-dependent tumor suppression by FOLATEApt/Exo/siM2 demonstrates that complete packaging of exosomes with folate, RNA aptamer, and siRNA reduced cancer cell growth.

Fig. 5
figure 5

Tumor growth and inhibition rate (TGI) of A375 tumor in NSG mice. A Cell fluorescence image. (1) and (4), the control group, were the A375 cells staining with Alexa Fluor® 488 phalloidin for cytoskeleton (green) and DAPI for nucleus (blue). (2)(3)(5)(6) were the A375 cells cocultured with Alexa647 labeled-RNA loaded exosome (red) for 1 h and stained with Alexa Fluor® 488 phalloidin for cytoskeleton (green) and DAPI for nucleus (blue). B Tumor growth curve of different treated groups. C Tumor growth inhibition rate (TGI) of different treated groups. D Grow images of A375 tumors after treatment for 49 days. The results are presented as the mean ± SD. *P < 0.05, **P < 0.01

With FOLATEApt/Exo/siM2 packaging-dependent suppression of melanoma established, FOLATEApt/Exo/siM2 suppressed other cancer cell lines as well. Fluoresce microscopic images of malignant colorectal carcinoma cells incubated with FOLATEApt/Exo/siM2 suggest that the exosomes appeared localized in nuclei (Fig. 6A) where transcription occurs. Growth curve of the tumor derived from the colorectal carcinoma cell line HCT-116 treated with FOLATEApt/Exo/siM2 and the packaging controls exhibited FOLATEApt/Exo/siM2 packaging-dependent inhibition (Fig. 6B-D). The results show that the wholly packaged exosomes with folate display and siRNA load decreased tumor volume by 60%, compared with siRNA by 27% and folate by 48% (Fig. 6C).

Fig. 6
figure 6

The growth and suppression rate of HCT-116 tumor in NSG mice. A (1)(2)(3) were colorectal tumor (HCT-116). (1)(4), the control groups, were cells staining with Alexa Fluor® 488 phalloidin for cytoskeleton (green) and DAPI for nucleus (blue). (2)(3) were the cells cocultured with Alexa647 labeled-RNA loaded exosome (red) for 1 h and stained with Alexa Fluor® 488 phalloidin for cytoskeleton (green) and DAPI for nucleus (blue). B Tumor growth curve of different treated groups. C Tumor growth inhibition rate (TGI) of different treated groups. D The end of experiment, the HCT-116 tumors were harvested for gross-view observation. The results are presented as the mean ± SD. *P < 0.05

Clinical applications and current clinical trials involving exosomes

Current clinical trials in exosomes

In recent years, exosomes have been investigated in clinical trials in a variety of contexts and applications. A survey of the current clinical trial landscape from GlobalData database (www.globaldata.com) reveals the biggest areas that exosomes are being explored for clinical therapies are oncology (54%), central nervous system (13%), infectious disease (13%), and immunology (8%) (Table 3). Of the 420 ongoing exosomal drugs in clinical development, more than 65% of the products remain in early phase (Preclinical to Discovery) and not yet filed for independent new drug review by U.S. Food and Drug Administration, while only 6.4%, 7.3%, and 2.3% of them are in Phase 1, 2, and 3 clinical trial, respectively. To date only two products have been marketed for clinical use. First, Patisiran is indicated for treating polyneuropathy associated with hereditary transthyretin-mediated amyloidosis (hATTR) in adults. Patisiran is a first-of-its kind RNA interference (RNAi)-based therapy designed to reduce levels of both WT and mutant transthyretin (TTR) for the treatment of patients with hereditary transthyretin-mediated amyloidosis, a congenital neurological and cardiac disorder [109]. Ibudilast is another commercially available exosome-related product. It is a relatively nonselective phosphodiesterase inhibitor which has been marketed for almost 20 years in Japan for treating asthma [110]. More recently it has been found to have anti-inflammatory activity in both the peripheral immune system and in the CNS via glial cell attenuation in relapsing–remitting (RR) and/or secondary progressive (SP) multiple sclerosis [111].

Table 3 Number of exosome drugs according to indication/therapeutic area

The application of exosomes in the context of treating neurological diseases and cardiovascular disease has attracted recent attention. While there is currently no effective treatment for neurological diseases such as Alzheimer's disease and Parkinson's disease [112], certain bioactive molecules in exosomes can promote the growth and repair of neurons and are expected to become a new way to treat neurological diseases. For example, researchers have found that neurotrophic factors contained in exosomes can promote the growth and repair of neurons [113]. They injected exosomes into the brains of mice and found that exosomes could promote the growth and repair of neurons and improve the cognitive function of the mice. In cardiovacular research, pathologies like such as coronary heart disease and myocardial infarction are some of the leading causes of death worldwide. Bioactive molecules (such as microRNA-132), which are delivered by mesenchymal stem cell-derived exosomes, promote angiogenesis in myocardial infarction [114]. Researchers have found that miR-126 contained in exosomes can promote endothelial cell proliferation and angiogenesis and reduce vascular permeability. They encapsulated miR-126 in exosomes and injected it into the myocardial infarction site of mice. They found that exosomes could promote the proliferation and repair of cardiomyocytes too improve the heart function of mice [115].

Regardless of their clinical indication, exosomes and their usage need to comply with good manufacturing practice (GMP). A GMP grade exosome production process comprises the highest quality of materials, cells, culture environment, manufacturing technologies, and skilled workforce for highly controlled and strictly monitored conditions. Furthermore, purification and quality control release assays are essentials after production to ensure final product meet highest standards prior to administration to patients for intended use. The European Medicines Agency and US Food and Drug Administration have convened workshops and acknowledge new scientific progress in cellular and molecular biotechnology have led to the development of advanced therapies. Both agencies have also released recommendations and classifications for advanced therapy production. As the nascent field of exosomes continue to expand in the realms of therapy and diagnostics, further recommendations, guidelines and criteria for researchers and manufactures are warranted to keep advanced therapies safe for patient use.

Future development of exosomes in therapeutics

Precision modification technology

With the development of precision gene editing technology, precise modification of exosomes can be achieved through genetic engineering in the future, thereby achieving more precise regulation [112]. Previous study introduces the use of precision gene editing technology to precisely modify exosomes to achieve therapeutic effects on Parkinson's disease. Genetic engineering allows therapeutic vectors to be loaded into exosomes and treated by implanting them into cells. The article discusses the application prospects and challenges of exosomes in the field of neurology.

Cell engineering technology

Cell engineering technology can control the cells from which exosomes are derived, thereby affecting their secretion quantity and quality [116]. Another study introduces cell engineering technology to control the cells from which exosomes are derived, thereby affecting exosome secretion quantity and quality. Scientist explores the application of human umbilical cord mesenchymal stem cell exosomes to promote angiogenesis through the Wnt4/β-catenin pathway and introduces the application prospects of cell engineering technology in controlling exosome secretion.

Nanotechnology applications

Nanotechnology offers enormous potential to further explore and impact cell physiology and pathology [117]. Recent studies describes the use of nanotechnology to control the size, shape, and surface properties of exosomes to influence their effects on physiology and pathology. The article explores the potential of using nanotechnology to change the properties of exosomes and apply them to cancer treatment [117].

In summary, exosomes are extracellular vesicles with great potential and play essential roles in physiology and pathology. By modifying it in different ways, its biological activity and stability can be controlled, enabling in-depth research and application of its function and mechanism of action. With the continuous development of technology, exosome modification technology will be more widely used.

Conclusion

Due to their clinical potential and unique biological functions, exosome offer potential breakthroughs in drug delivery, noninvasive disease diagnosis, treatment, and other fields. Compared to liposomes, nanoparticles, microspheres, microemulsions, and other synthetic drug loading systems, exosomes pose natural and unique advantages as potential biomarkers for prognosis and diagnosing disease, drug delivery carriers, cell free therapy and cancer vaccine.

Considering the promise of exosomes in diagnosing and treating various conditions, challenges remain for its translation from the lab to the clinic. First, there are limitations surrounding vesicle isolation and determining whether the isolated exosomes are viable candidate for clinical use. This can be due to multiple factors including inconsistencies related to the isolated particle number, morphology, methodology, conditions, and source of isolated exosomes. Another prominent limitation is variability in exosomal yield and purity related to isolation techniques. While some isolation techniques can provide higher yield and purity than others, inconsistencies in methodology, equipment, and human error prove it is difficult to quantify which method is most appropriate to use. Alongside this are inconsistencies in cargo loading efficiency such as electroporation, sonication, and extrusion.

While exosome research remains in its nascent stages of development, further in-depth understanding of subcellular components and mechanisms involved in exosome formation and specific cell targeting will bring light on their physiological function and clinical potential. Undoubtedly, exosomes represent a promising tool in the field of medicine and may provide a solution to a variety of medical challenges we face today.

Availability of data and materials

Not applicable.

References

  1. Thery C. Exosomes: secreted vesicles and intercellular communications. F1000 Biol Rep. 2011;3:15.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science. 2020;367(6478):eaau6977.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Statello L, Maugeri M, Garre E, Nawaz M, Wahlgren J, Papadimitriou A, et al. Identification of RNA-binding proteins in exosomes capable of interacting with different types of RNA: RBP-facilitated transport of RNAs into exosomes. PLoS ONE. 2018;13(4):e0195969.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Kwantwi LB. Exosome-mediated crosstalk between tumor cells and innate immune cells: implications for cancer progression and therapeutic strategies. J Cancer Res Clin Oncol. 2023;149(11):9487–503.

    Article  PubMed  Google Scholar 

  5. Zhang K, Cheng K. Stem cell-derived exosome versus stem cell therapy. Nat Rev Bioeng. 2023;12:1–2.

    Google Scholar 

  6. Agarwal P, Anees A, Harsiddharay RK, Kumar P, Tripathi PK. A Comprehensive Review on Exosome: Recent Progress and Outlook. Pharm Nanotechnol. 2023.

  7. Mimeault M, Batra SK. Molecular biomarkers of cancer stem/progenitor cells associated with progression, metastases, and treatment resistance of aggressive cancers. Cancer Epidemiol Biomarkers Prev. 2014;23(2):234–54.

    Article  CAS  PubMed  Google Scholar 

  8. Colombo M, Moita C, van Niel G, Kowal J, Vigneron J, Benaroch P, et al. Analysis of ESCRT functions in exosome biogenesis, composition and secretion highlights the heterogeneity of extracellular vesicles. J Cell Sci. 2013;126(Pt 24):5553–65.

    CAS  PubMed  Google Scholar 

  9. Haney MJ, Klyachko NL, Zhao Y, Gupta R, Plotnikova EG, He Z, et al. Exosomes as drug delivery vehicles for Parkinson’s disease therapy. J Control Release. 2015;10(207):18–30.

    Article  Google Scholar 

  10. Valadi H, Ekstrom K, Bossios A, Sjostrand M, Lee JJ, Lotvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007;9(6):654–9.

    Article  CAS  PubMed  Google Scholar 

  11. Colombo M, Raposo G, Thery C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev Biol. 2014;30:255–89.

    Article  CAS  PubMed  Google Scholar 

  12. Del Pozo-Acebo L, Hazas MLL, Tome-Carneiro J, Gil-Cabrerizo P, San-Cristobal R, Busto R et al. Bovine Milk-Derived Exosomes as a Drug Delivery Vehicle for miRNA-Based Therapy. Int J Mol Sci. 2021;22(3).

  13. Peng Y, Yang Y, Li Y, Shi T, Luan Y, Yin C. Exosome and virus infection. Front Immunol. 2023;14:1154217.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Hu Z, Chen G, Zhao Y, Gao H, Li L, Yin Y, et al. Exosome-derived circCCAR1 promotes CD8 + T-cell dysfunction and anti-PD1 resistance in hepatocellular carcinoma. Mol Cancer. 2023;22(1):55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Li P, Chen J, Chen Y, Song S, Huang X, Yang Y, et al. Construction of Exosome SORL1 Detection Platform Based on 3D Porous Microfluidic Chip and its Application in Early Diagnosis of Colorectal Cancer. Small. 2023;19(20):e2207381.

    Article  PubMed  Google Scholar 

  16. Shin H, Choi BH, Shim O, Kim J, Park Y, Cho SK, et al. Single test-based diagnosis of multiple cancer types using Exosome-SERS-AI for early stage cancers. Nat Commun. 2023;14(1):1644.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Yu Z, Yang Y, Fang W, Hu P, Liu Y, Shi J. Dual Tumor Exosome Biomarker Co-recognitions Based Nanoliquid Biopsy for the Accurate Early Diagnosis of Pancreatic Cancer. ACS Nano. 2023;17(12):11384–95.

    Article  CAS  PubMed  Google Scholar 

  18. Xie F, Zhou X, Fang M, Li H, Su P, Tu Y, et al. Extracellular Vesicles in Cancer Immune Microenvironment and Cancer Immunotherapy. Adv Sci (Weinh). 2019;6(24):1901779.

    Article  CAS  PubMed  Google Scholar 

  19. Huang X, Yuan T, Tschannen M, Sun Z, Jacob H, Du M, et al. Characterization of human plasma-derived exosomal RNAs by deep sequencing. BMC Genomics. 2013;10(14):319.

    Article  Google Scholar 

  20. Vlassov AV, Magdaleno S, Setterquist R, Conrad R. Exosomes: current knowledge of their composition, biological functions, and diagnostic and therapeutic potentials. Biochim Biophys Acta. 2012;1820(7):940–8.

    Article  CAS  PubMed  Google Scholar 

  21. Natasha G, Gundogan B, Tan A, Farhatnia Y, Wu W, Rajadas J, et al. Exosomes as immunotheranostic nanoparticles. Clin Ther. 2014;36(6):820–9.

    Article  CAS  Google Scholar 

  22. Simpson RJ, Jensen SS, Lim JW. Proteomic profiling of exosomes: current perspectives. Proteomics. 2008;8(19):4083–99.

    Article  CAS  PubMed  Google Scholar 

  23. Yellon DM, Davidson SM. Exosomes: nanoparticles involved in cardioprotection? Circ Res. 2014;114(2):325–32.

    Article  CAS  PubMed  Google Scholar 

  24. Decuzzi P, Godin B, Tanaka T, Lee SY, Chiappini C, Liu X, et al. Size and shape effects in the biodistribution of intravascularly injected particles. J Control Release. 2010;141(3):320–7.

    Article  CAS  PubMed  Google Scholar 

  25. Wang J, Zheng Y, Zhao M. Exosome-Based Cancer Therapy: Implication for Targeting Cancer Stem Cells. Front Pharmacol. 2016;7:533.

    PubMed  Google Scholar 

  26. Shao H, Chung J, Lee K, Balaj L, Min C, Carter BS, et al. Chip-based analysis of exosomal mRNA mediating drug resistance in glioblastoma. Nat Commun. 2015;11(6):6999.

    Article  Google Scholar 

  27. Hewson C, Capraro D, Burdach J, Whitaker N, Morris KV. Extracellular vesicle associated long non-coding RNAs functionally enhance cell viability. Noncoding RNA Res. 2016;1(1):3–11.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Mittelbrunn M, Gutierrez-Vazquez C, Villarroya-Beltri C, Gonzalez S, Sanchez-Cabo F, Gonzalez MA, et al. Unidirectional transfer of microRNA-loaded exosomes from T cells to antigen-presenting cells. Nat Commun. 2011;2:282.

    Article  PubMed  Google Scholar 

  29. Conigliaro A, Costa V, Lo Dico A, Saieva L, Buccheri S, Dieli F, et al. CD90+ liver cancer cells modulate endothelial cell phenotype through the release of exosomes containing H19 lncRNA. Mol Cancer. 2015;14(14):155.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Esser J, Gehrmann U, D’Alexandri FL, Hidalgo-Estevez AM, Wheelock CE, Scheynius A, et al. Exosomes from human macrophages and dendritic cells contain enzymes for leukotriene biosynthesis and promote granulocyte migration. J Allergy Clin Immunol. 2010;126(5):1032-40 e1–4.

    Article  CAS  PubMed  Google Scholar 

  31. Falguieres T, Castle D, Gruenberg J. Regulation of the MVB pathway by SCAMP3. Traffic. 2012;13(1):131–42.

    Article  CAS  PubMed  Google Scholar 

  32. Laulagnier K, Grand D, Dujardin A, Hamdi S, Vincent-Schneider H, Lankar D, et al. PLD2 is enriched on exosomes and its activity is correlated to the release of exosomes. FEBS Lett. 2004;572(1–3):11–4.

    Article  CAS  PubMed  Google Scholar 

  33. Subra C, Grand D, Laulagnier K, Stella A, Lambeau G, Paillasse M, et al. Exosomes account for vesicle-mediated transcellular transport of activatable phospholipases and prostaglandins. J Lipid Res. 2010;51(8):2105–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Trajkovic K, Hsu C, Chiantia S, Rajendran L, Wenzel D, Wieland F, et al. Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science. 2008;319(5867):1244–7.

    Article  CAS  PubMed  Google Scholar 

  35. Tan NS, Shaw NS, Vinckenbosch N, Liu P, Yasmin R, Desvergne B, et al. Selective cooperation between fatty acid binding proteins and peroxisome proliferator-activated receptors in regulating transcription. Mol Cell Biol. 2002;22(14):5114–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Zhang H, Deng T, Liu R, Bai M, Zhou L, Wang X, et al. Exosome-delivered EGFR regulates liver microenvironment to promote gastric cancer liver metastasis. Nat Commun. 2017;10(8):15016.

    Article  Google Scholar 

  37. Xiang Y, Shen Z, Shi Y. Chemical Cross-Linking and Mass Spectrometric Analysis of the Endogenous Yeast Exosome Complexes. Methods Mol Biol. 2020;2062:383–400.

    Article  CAS  PubMed  Google Scholar 

  38. Xi XM, Xia SJ, Lu R. Drug loading techniques for exosome-based drug delivery systems. Pharmazie. 2021;76(2):61–7.

    CAS  PubMed  Google Scholar 

  39. Guo ZY, Tang Y, Cheng YC. Exosomes as Targeted Delivery Drug System: Advances in Exosome Loading, Surface Functionalization and Potential for Clinical Application. Curr Drug Deliv. 2022.

  40. Chen S, Sun F, Qian H, Xu W, Jiang J. Preconditioning and Engineering Strategies for Improving the Efficacy of Mesenchymal Stem Cell-Derived Exosomes in Cell-Free Therapy. Stem Cells Int. 2022;2022:1779346.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Choi H, Choi K, Kim DH, Oh BK, Yim H, Jo S et al. Strategies for Targeted Delivery of Exosomes to the Brain: Advantages and Challenges. Pharmaceutics. 2022;14(3).

  42. Greening DW, Xu R, Ji H, Tauro BJ, Simpson RJ. A protocol for exosome isolation and characterization: evaluation of ultracentrifugation, density-gradient separation, and immunoaffinity capture methods. Methods Mol Biol. 2015;1295:179–209.

    Article  CAS  PubMed  Google Scholar 

  43. Lobb RJ, Becker M, Wen SW, Wong CS, Wiegmans AP, Leimgruber A, et al. Optimized exosome isolation protocol for cell culture supernatant and human plasma. J Extracell Vesicles. 2015;4:27031.

    Article  PubMed  Google Scholar 

  44. Seo K, Yoo JH, Kim J, Min SJ, Heo DN, Kwon IK, et al. Ginseng-derived exosome-like nanovesicles extracted by sucrose gradient ultracentrifugation to inhibit osteoclast differentiation. Nanoscale. 2023;15(12):5798–808.

    Article  CAS  PubMed  Google Scholar 

  45. Zarovni N, Corrado A, Guazzi P, Zocco D, Lari E, Radano G, et al. Integrated isolation and quantitative analysis of exosome shuttled proteins and nucleic acids using immunocapture approaches. Methods. 2015;1(87):46–58.

    Article  Google Scholar 

  46. Bobrie A, Colombo M, Krumeich S, Raposo G, Thery C. Diverse subpopulations of vesicles secreted by different intracellular mechanisms are present in exosome preparations obtained by differential ultracentrifugation. J Extracell Vesicles. 2012;1.

  47. Kenigsberg S, Wyse BA, Librach CL, da Silveira JC. Protocol for Exosome Isolation from Small Volume of Ovarian Follicular Fluid: Evaluation of Ultracentrifugation and Commercial Kits. Methods Mol Biol. 2017;1660:321–41.

    Article  CAS  PubMed  Google Scholar 

  48. Lobb R, Moller A. Size Exclusion Chromatography: A Simple and Reliable Method for Exosome Purification. Methods Mol Biol. 2017;1660:105–10.

    Article  CAS  PubMed  Google Scholar 

  49. Ludwig N, Razzo BM, Yerneni SS, Whiteside TL. Optimization of cell culture conditions for exosome isolation using mini-size exclusion chromatography (mini-SEC). Exp Cell Res. 2019;378(2):149–57.

    Article  CAS  PubMed  Google Scholar 

  50. Shu S, Allen CL, Benjamin-Davalos S, Koroleva M, MacFarland D, Minderman H, et al. A Rapid Exosome Isolation Using Ultrafiltration and Size Exclusion Chromatography (REIUS) Method for Exosome Isolation from Melanoma Cell Lines. Methods Mol Biol. 2021;2265:289–304.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Song Z, Mao J, Barrero RA, Wang P, Zhang F, Wang T. Development of a CD63 Aptamer for Efficient Cancer Immunochemistry and Immunoaffinity-Based Exosome Isolation. Molecules. 2020;25(23).

  52. Wang YT, Cai MD, Sun LL, Hua RN. A Rapid and Facile Separation-Detection Integrated Strategy for Exosome Profiling Based on Boronic Acid-Directed Coupling Immunoaffinity. Anal Chem. 2021;93(48):16059–67.

    Article  CAS  PubMed  Google Scholar 

  53. Sasabe E, Tomomura A, Liu H, Sento S, Kitamura N, Yamamoto T. Epidermal growth factor/epidermal growth factor receptor signaling blockage inhibits tumor cell-derived exosome uptake by oral squamous cell carcinoma through macropinocytosis. Cancer Sci. 2022;113(2):609–21.

    Article  CAS  PubMed  Google Scholar 

  54. Nag S, Bhattacharya B, Dutta S, Mandal D, Mukherjee S, Anand K, et al. Clinical Theranostics Trademark of Exosome in Glioblastoma Metastasis. ACS Biomater Sci Eng. 2023;9(9):5205–21.

    Article  CAS  PubMed  Google Scholar 

  55. Chen X, Ren X, E J, Zhou Y, Bian R. Exosome-transmitted circIFNGR2 Modulates Ovarian Cancer Metastasis via miR-378/ST5 Axis. Mol Cell Biol. 2023;43(1):22–42.

  56. Marin AM, Mattar SB, Amatuzzi RF, Chammas R, Uno M, Zanette DL et al. Plasma Exosome-Derived microRNAs as Potential Diagnostic and Prognostic Biomarkers in Brazilian Pancreatic Cancer Patients. Biomolecules. 2022;12(6).

  57. Naseri Z, Oskuee RK, Jaafari MR, Forouzandeh MM. Exosome-mediated delivery of functionally active miRNA-142-3p inhibitor reduces tumorigenicity of breast cancer in vitro and in vivo. Int J Nanomedicine. 2018;13:7727–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Liu K, Dou R, Yang C, Di Z, Shi D, Zhang C, et al. Exosome-transmitted miR-29a induces colorectal cancer metastasis by destroying the vascular endothelial barrier. Carcinogenesis. 2023;44(4):356–67.

    Article  CAS  PubMed  Google Scholar 

  59. Nambara S, Masuda T, Hirose K, Hu Q, Tobo T, Ozato Y, et al. Rab27b, a Regulator of Exosome Secretion, Is Associated With Peritoneal Metastases in Gastric Cancer. Cancer Genomics Proteomics. 2023;20(1):30–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Wang Y, Jiang R, Zhao H, Li F, Li Y, Zhu M. TTN-AS1 delivered by gastric cancer cell-derived exosome induces gastric cancer progression through in vivo and in vitro studies. Cell Biol Toxicol. 2023;39(2):557–71.

    Article  CAS  PubMed  Google Scholar 

  61. Yi Q, Xu Z, Thakur A, Zhang K, Liang Q, Liu Y, et al. Current understanding of plant-derived exosome-like nanoparticles in regulating the inflammatory response and immune system microenvironment. Pharmacol Res. 2023;190:106733.

    Article  CAS  PubMed  Google Scholar 

  62. Liu X, Li R, Chen X, Yao J, Wang Q, Zhang J, et al. SYT7 is a key player in increasing exosome secretion and promoting angiogenesis in non-small-cell lung cancer. Cancer Lett. 2023;27:216400.

    Article  Google Scholar 

  63. Ma Z, Xu H, Ye BC. Recent progress in quantitative technologies for the analysis of cancer-related exosome proteins. Analyst. 2023;148(20):4954–66.

    Article  CAS  PubMed  Google Scholar 

  64. Deng Z, Mu J, Tseng M, Wattenberg B, Zhuang X, Egilmez NK, et al. Enterobacteria-secreted particles induce production of exosome-like S1P-containing particles by intestinal epithelium to drive Th17-mediated tumorigenesis. Nat Commun. 2015;24(6):6956.

    Article  Google Scholar 

  65. McKiernan J, Donovan MJ, O’Neill V, Bentink S, Noerholm M, Belzer S, et al. A Novel Urine Exosome Gene Expression Assay to Predict High-grade Prostate Cancer at Initial Biopsy. JAMA Oncol. 2016;2(7):882–9.

    Article  PubMed  Google Scholar 

  66. Street JM, Koritzinsky EH, Glispie DM, Yuen PST. Urine Exosome Isolation and Characterization. Methods Mol Biol. 2017;1641:413–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Belhaj MR, Lawler NG, Hoffman NJ. Metabolomics and Lipidomics: Expanding the Molecular Landscape of Exercise Biology. Metabolites. 2021;11(3).

  68. Wang L, Jia Q, Xinnong C, Xie Y, Yang Y, Zhang A, et al. Role of cardiac progenitor cell-derived exosome-mediated microRNA-210 in cardiovascular disease. J Cell Mol Med. 2019;23(11):7124–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Zhang TR, Huang WQ. Angiogenic Exosome-Derived microRNAs: Emerging Roles in Cardiovascular Disease. J Cardiovasc Transl Res. 2021;14(5):824–40.

    Article  PubMed  Google Scholar 

  70. Yong T, Zhang X, Bie N, Zhang H, Zhang X, Li F, et al. Tumor exosome-based nanoparticles are efficient drug carriers for chemotherapy. Nat Commun. 2019;10(1):3838.

    Article  PubMed  PubMed Central  Google Scholar 

  71. Uddin N, Binzel DW, Shu D, Fu TM, Guo P. Targeted delivery of RNAi to cancer cells using RNA-ligand displaying exosome. Acta Pharm Sin B. 2023;13(4):1383–99.

    Article  CAS  PubMed  Google Scholar 

  72. Zheng Z, Li Z, Xu C, Guo B, Guo P. Folate-displaying exosome mediated cytosolic delivery of siRNA avoiding endosome trapping. J Control Release. 2019;311–312:43–9.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Guo S, Huang F, Guo P. Construction of folate-conjugated pRNA of bacteriophage phi29 DNA packaging motor for delivery of chimeric siRNA to nasopharyngeal carcinoma cells. Gene Ther. 2006;13(10):814–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Pi F, Binzel DW, Lee TJ, Li Z, Sun M, Rychahou P, et al. Nanoparticle orientation to control RNA loading and ligand display on extracellular vesicles for cancer regression. Nat Nanotechnol. 2018;13(1):82–9.

    Article  CAS  PubMed  Google Scholar 

  75. Shu D, Li H, Shu Y, Xiong G, Carson WE 3rd, Haque F, et al. Systemic Delivery of Anti-miRNA for Suppression of Triple Negative Breast Cancer Utilizing RNA Nanotechnology. ACS Nano. 2015;9(10):9731–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Yin H, Wang H, Li Z, Shu D, Guo P. RNA Micelles for the Systemic Delivery of Anti-miRNA for Cancer Targeting and Inhibition without Ligand. ACS Nano. 2019;13(1):706–17.

    Article  CAS  PubMed  Google Scholar 

  77. Guo P. Rolling Circle Transcription of Tandem siRNA to Generate Spherulitic RNA Nanoparticles for Cell Entry. Mol Ther Nucleic Acids. 2012;1(8):e36.

    Article  PubMed  PubMed Central  Google Scholar 

  78. Shu Y, Cinier M, Fox SR, Ben-Johnathan N, Guo P. Assembly of therapeutic pRNA-siRNA nanoparticles using bipartite approach. Mol Ther. 2011;19(7):1304–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Shu Y, Cinier M, Shu D, Guo P. Assembly of multifunctional phi29 pRNA nanoparticles for specific delivery of siRNA and other therapeutics to targeted cells. Methods. 2011;54(2):204–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Tarapore P, Shu Y, Guo P, Ho SM. Application of phi29 motor pRNA for targeted therapeutic delivery of siRNA silencing metallothionein-IIA and survivin in ovarian cancers. Mol Ther. 2011;19(2):386–94.

    Article  CAS  PubMed  Google Scholar 

  81. Herrmann IK, Wood MJA, Fuhrmann G. Extracellular vesicles as a next-generation drug delivery platform. Nat Nanotechnol. 2021;16(7):748–59.

    Article  CAS  PubMed  Google Scholar 

  82. Gonda A, Kabagwira J, Senthil GN, Wall NR. Internalization of Exosomes through Receptor-Mediated Endocytosis. Mol Cancer Res. 2019;17(2):337–47.

    Article  CAS  PubMed  Google Scholar 

  83. Chen H, Wang L, Zeng X, Schwarz H, Nanda HS, Peng X, et al. Exosomes, a New Star for Targeted Delivery. Front Cell Dev Biol. 2021;9:751079.

    Article  PubMed  PubMed Central  Google Scholar 

  84. Cui Y, Guo Y, Kong L, Shi J, Liu P, Li R, et al. A bone-targeted engineered exosome platform delivering siRNA to treat osteoporosis. Bioact Mater. 2022;10:207–21.

    CAS  PubMed  Google Scholar 

  85. Fukuta T, Ikeda-Imafuku M, Kodama S, Kuse J, Matsui K, Iwao Y. One-Step Pharmaceutical Preparation of PEG-Modified Exosomes Encapsulating Anti-Cancer Drugs by a High-Pressure Homogenization Technique. Pharmaceuticals (Basel). 2023;16(1).

  86. Kooijmans SA, Vader P, van Dommelen SM, van Solinge WW, Schiffelers RM. Exosome mimetics: a novel class of drug delivery systems. Int J Nanomedicine. 2012;7:1525–41.

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Yang T, Martin P, Fogarty B, Brown A, Schurman K, Phipps R, et al. Exosome delivered anticancer drugs across the blood-brain barrier for brain cancer therapy in Danio rerio. Pharm Res. 2015;32(6):2003–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Cao H, Chen M, Cui X, Liu Y, Liu Y, Deng S, et al. Cell-Free Osteoarthritis Treatment with Sustained-Release of Chondrocyte-Targeting Exosomes from Umbilical Cord-Derived Mesenchymal Stem Cells to Rejuvenate Aging Chondrocytes. ACS Nano. 2023;17(14):13358–76.

    Article  CAS  PubMed  Google Scholar 

  89. Tian Y, Li S, Song J, Ji T, Zhu M, Anderson GJ, et al. A doxorubicin delivery platform using engineered natural membrane vesicle exosomes for targeted tumor therapy. Biomaterials. 2014;35(7):2383–90.

    Article  CAS  PubMed  Google Scholar 

  90. Dimik M, Abeysinghe P, Logan J, Mitchell M. The exosome: a review of current therapeutic roles and capabilities in human reproduction. Drug Deliv Transl Res. 2023;13(2):473–502.

    Article  PubMed  Google Scholar 

  91. 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(7):1150–62.

    Article  CAS  PubMed  Google Scholar 

  92. Pinnell JR, Cui M, Tieu K. Exosomes in Parkinson disease. J Neurochem. 2021;157(3):413–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Engstrom Y, Eriksson S, Jildevik I, Skog S, Thelander L, Tribukait B. Cell cycle-dependent expression of mammalian ribonucleotide reductase. Differential regulation of the two subunits. J Biol Chem. 1985;260(16):9114–6.

    Article  CAS  PubMed  Google Scholar 

  94. Nordlund P, Reichard P. Ribonucleotide reductases. Annu Rev Biochem. 2006;75:681–706.

    Article  CAS  PubMed  Google Scholar 

  95. Grolmusz VK, Karaszi K, Micsik T, Toth EA, Meszaros K, Karvaly G, et al. Cell cycle dependent RRM2 may serve as proliferation marker and pharmaceutical target in adrenocortical cancer. Am J Cancer Res. 2016;6(9):2041–53.

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Grossi F, Dal Bello MG, Salvi S, Puzone R, Pfeffer U, Fontana V, et al. Expression of Ribonucleotide Reductase Subunit-2 and Thymidylate Synthase Correlates with Poor Prognosis in Patients with Resected Stages I-III Non-Small Cell Lung Cancer. Dis Markers. 2015;2015:302649.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Lu AG, Feng H, Wang PX, Han DP, Chen XH, Zheng MH. Emerging roles of the ribonucleotide reductase M2 in colorectal cancer and ultraviolet-induced DNA damage repair. World J Gastroenterol. 2012;18(34):4704–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Jin CY, Du L, Nuerlan AH, Wang XL, Yang YW, Guo R. High expression of RRM2 as an independent predictive factor of poor prognosis in patients with lung adenocarcinoma. Aging (Albany NY). 2020;13(3):3518–35.

    Article  PubMed  Google Scholar 

  99. Zhang H, Liu X, Warden CD, Huang Y, Loera S, Xue L, et al. Prognostic and therapeutic significance of ribonucleotide reductase small subunit M2 in estrogen-negative breast cancers. BMC Cancer. 2014;11(14):664.

    Article  Google Scholar 

  100. Zhang K, Hu S, Wu J, Chen L, Lu J, Wang X, et al. Overexpression of RRM2 decreases thrombspondin-1 and increases VEGF production in human cancer cells in vitro and in vivo: implication of RRM2 in angiogenesis. Mol Cancer. 2009;28(8):11.

    Article  Google Scholar 

  101. Chen S, Zhou B, He F, Yen Y. Inhibition of human cancer cell growth by inducible expression of human ribonucleotide reductase antisense cDNA. Antisense Nucleic Acid Drug Dev. 2000;10(2):111–6.

    Article  CAS  PubMed  Google Scholar 

  102. Lee Y, Vassilakos A, Feng N, Lam V, Xie H, Wang M, et al. GTI-2040, an antisense agent targeting the small subunit component (R2) of human ribonucleotide reductase, shows potent antitumor activity against a variety of tumors. Cancer Res. 2003;63(11):2802–11.

    CAS  PubMed  Google Scholar 

  103. Rahman MA, Amin AR, Wang X, Zuckerman JE, Choi CH, Zhou B, et al. Systemic delivery of siRNA nanoparticles targeting RRM2 suppresses head and neck tumor growth. J Control Release. 2012;159(3):384–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Li Z, Yang L, Wang H, Binzel DW, Williams TM, Guo P. Non-Small-Cell Lung Cancer Regression by siRNA Delivered Through Exosomes That Display EGFR RNA Aptamer. Nucleic Acid Ther. 2021;31(5):364–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. O’Brien K, Breyne K, Ughetto S, Laurent LC, Breakefield XO. RNA delivery by extracellular vesicles in mammalian cells and its applications. Nat Rev Mol Cell Biol. 2020;21(10):585–606.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Davis ME, Zuckerman JE, Choi CH, Seligson D, Tolcher A, Alabi CA, et al. Evidence of RNAi in humans from systemically administered siRNA via targeted nanoparticles. Nature. 2010;464(7291):1067–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Davis ME. The first targeted delivery of siRNA in humans via a self-assembling, cyclodextrin polymer-based nanoparticle: from concept to clinic. Mol Pharm. 2009;6(3):659–68.

    Article  CAS  PubMed  Google Scholar 

  108. Heidel JD, Liu JY, Yen Y, Zhou B, Heale BS, Rossi JJ, et al. Potent siRNA inhibitors of ribonucleotide reductase subunit RRM2 reduce cell proliferation in vitro and in vivo. Clin Cancer Res. 2007;13(7):2207–15.

    Article  CAS  PubMed  Google Scholar 

  109. Kalola UK, Pellegrini MV. Patisiran. StatPearls. Treasure Island (FL) ineligible companies. Disclosure: Mark Pellegrini declares no relevant financial relationships with ineligible companies. 2023.

  110. Fujimoto T, Sakoda S, Fujimura H, Yanagihara T. Ibudilast, a phosphodiesterase inhibitor, ameliorates experimental autoimmune encephalomyelitis in Dark August rats. J Neuroimmunol. 1999;95(1–2):35–42.

    Article  CAS  PubMed  Google Scholar 

  111. Rolan P, Hutchinson M, Johnson K. Ibudilast: a review of its pharmacology, efficacy and safety in respiratory and neurological disease. Expert Opin Pharmacother. 2009;10(17):2897–904.

    Article  CAS  PubMed  Google Scholar 

  112. Kojima R, Bojar D, Rizzi G, Hamri GC, El-Baba MD, Saxena P, et al. Designer exosomes produced by implanted cells intracerebrally deliver therapeutic cargo for Parkinson’s disease treatment. Nat Commun. 2018;9(1):1305.

    Article  PubMed  PubMed Central  Google Scholar 

  113. Sun G, Li G, Li D, Huang W, Zhang R, Zhang H, et al. hucMSC derived exosomes promote functional recovery in spinal cord injury mice via attenuating inflammation. Mater Sci Eng C Mater Biol Appl. 2018;1(89):194–204.

    Article  Google Scholar 

  114. Ma T, Chen Y, Chen Y, Meng Q, Sun J, Shao L, et al. MicroRNA-132, Delivered by Mesenchymal Stem Cell-Derived Exosomes, Promote Angiogenesis in Myocardial Infarction. Stem Cells Int. 2018;2018:3290372.

    Article  PubMed  PubMed Central  Google Scholar 

  115. Duan S, Wang C, Xu X, Zhang X, Su G, Li Y, et al. Peripheral Serum Exosomes Isolated from Patients with Acute Myocardial Infarction Promote Endothelial Cell Angiogenesis via the miR-126-3p/TSC1/mTORC1/HIF-1alpha Pathway. Int J Nanomedicine. 2022;17:1577–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Zhang B, Wu X, Zhang X, Sun Y, Yan Y, Shi H, et al. Human umbilical cord mesenchymal stem cell exosomes enhance angiogenesis through the Wnt4/beta-catenin pathway. Stem Cells Transl Med. 2015;4(5):513–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Li S, Xu J, Qian J, Gao X. Engineering extracellular vesicles for cancer therapy: recent advances and challenges in clinical translation. Biomater Sci. 2020;8(24):6978–91.

    Article  CAS  PubMed  Google Scholar 

Download references

Funding

This work was supported by Ministry of Health and Welfare (Health and welfare surcharge of tobacco products grant MOHW112-TDU-B-222–014013 and MOHW113-TDU-B-222–134013 to Y. Y.). This work was supported by grants from the Ministry of Science and Technology (MOST 110–2320-B-038–070 to Y.-F.C.) and the National Science and Technology Council (NSTC 112–2320-B-038–020 to Y.-F.C.). We are grateful for the T-star Cancer Center grant (NSTC 113-2634-F-039-001) awarded to Y.-S. H..

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, Y.-F.C. and Y. Y.; writing—original draft preparation, Y.-F.C. and F. L.; writing—final manuscript, Y.-S. H. and Y. Y. Project administration, Y.-S. H. and Y. Y. All authors have read and agreed to the published version of the manuscript.

Corresponding authors

Correspondence to Yuan-Soon Ho or Yun Yen.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, YF., Luh, F., Ho, YS. et al. Exosomes: a review of biologic function, diagnostic and targeted therapy applications, and clinical trials. J Biomed Sci 31, 67 (2024). https://doi.org/10.1186/s12929-024-01055-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12929-024-01055-0

Keywords