Abstract
Intercellular communication can be mediated by direct cell-to-cell contact and indirect interactions through secretion of soluble chemokines, cytokines, and growth factors. Extracellular vesicles (EVs) have emerged as important mediators of cell-to-cell and cell-to-environment communications. EVs from tumor cells, immune cells, and stromal cells can remodel the tumor microenvironment and promote cancer cell survival, proliferation, metastasis, immune evasion, and therapeutic resistance. Most importantly, EVs as natural nanoparticles can be manipulated to serve as a potent delivery system for targeted cancer therapy. EVs can be engineered or modified to improve their ability to target tumors and deliver therapeutic substances, such as chemotherapeutic drugs, nucleic acids, and proteins, for the treatment of cancer. This review provides an overview of the biogenesis and recycling of EVs, discusses their roles in cancer development, and highlights their potential as a delivery system for targeted cancer therapy.
Keywords: exosome, tumor microenvironment, immune response, metastasis, cancer therapy
Introduction
Intercellular communication is critical for cancer development. Such activity can be facilitated by the exchange of extracellular vesicles (EVs), which contain nucleic acids, proteins, and metabolites, from donor cells to recipient cells. The secretion of EVs was initially considered a waste disposal pathway that eliminates unneeded cellular components. It was later realized that EVs are more than waste carriers and play essential roles in transferring components between cells (Couch et al., 2021). EV-mediated cell-to-cell communication is associated with cancer initiation, progression, and immune response (van Niel et al., 2018; Dixson et al., 2023). Several oncogenic proteins, such as MYC, AURKB, and mutant p53, have been found to educate a tumor-promoting microenvironment through the secretion of EVs (Cooks et al., 2018; Novo et al., 2018; Kilinc et al., 2021), whereas tumor suppressors foster a tumor-suppressive microenvironment via EVs (Schuldner et al., 2019; Zhang et al., 2020b). Because of the biocompatibility of EVs and their ability to deliver cargos, EVs have been exploited to develop new targeted therapeutics for cancer. In this review, we summarize recent progress on the functions and mechanisms of EVs in cancer development and, particularly, emphasize the role of EVs in the development of novel anticancer therapy.
Life cycle of EVs
EVs generally fall into two major categories, exosomes and ectosomes (or microvesicles), based on their distinct biogenesis pathways (Kalluri and LeBleu, 2020). Of note, the term ‘exosome’ has also been widely used to describe the RNA-processing machinery (Houseley et al., 2006). While ectosomes in the size range of 50–1000 nm in diameter are generated by direct outward budding at the plasma membrane, exosomes that are generally in a size range of 30–150 nm in diameter arise from double invagination of the plasma membrane (Figure 1). Endocytosis, the first invagination of the plasma membrane, leads to the formation of early-sorting endosome containing extracellular constituents and cell surface proteins. Early-sorting endosomes can mature into late-sorting endosomes, followed by a second invagination. The inward budding of the endosomal membrane gives rise to multivesicular bodies (MVBs) that contain intraluminal vesicles. MVBs can be transported to the plasma membrane through the cytoskeletal and microtubule network. Intraluminal vesicles are ultimately released to the extracellular space as exosomes through exocytosis. Alternatively, MVBs can also be fused with autophagosomes or lysosomes for degradation. Numerous molecules have been found to be critical for the biogenesis of EVs, such as the endosomal sorting complexes required for transport (ESCRT) family proteins (Hurley, 2008; Colombo et al., 2013), Ras-related protein GTPase Rab (Hsu et al., 2010; Wang et al., 2014), ADP-ribosylation factor 6 (Muralidharan-Chari et al., 2009), apoptosis-linked gene 2-interacting protein X (Baietti et al., 2012), soluble N-ethylmaleimide-sensitive factor attachment protein receptor complex proteins (Jahn and Scheller, 2006; Fader et al., 2009), etc. However, the detailed mechanisms for the biogenesis and secretion of EVs are still being revealed.
Figure 1.

Biogenesis and release of exosomes. Unlike ectosomes, exosomes arise from double invagination of the plasma membrane.
Once secreted into the extracellular space, EVs can reach and bind to recipient cell surface. The bound EVs may remain at the plasma membrane to trigger various intracellular signaling pathways or can be internalized through different manners (Figure 2; Mulcahy et al., 2014). The internalization can be executed through receptor-mediated endocytosis (Nechaev et al., 2013; Gonda et al., 2019), macropinocytosis (Tian et al., 2014a; Li et al., 2020), phagocytosis (Feng et al., 2010), clathrin-coated pits (Tian et al., 2014a; Li et al., 2020), lipid rafts (Sapon et al., 2023), or caveolae (Le Saux et al., 2020), resulting in the entry of intact EVs, which are contained in MVBs. These EV-containing MVBs are usually targeted to lysosomes, which leads to the degradation of their carried proteins and lipids, thereby providing a relevant source of metabolites to the recipient cells. In some cases, these internalized EVs may escape from lysosomal degradation via back-fusion with the MVB membrane, releasing their contents into the cytoplasm, or via re-secretion into the extracellular space. In addition, EVs can directly fuse with the plasma membrane to deposit their contents into the cytoplasm of recipient cells, which bypasses lysosomal surveillance and is essential for cell uptake of therapeutic cargoes that are delivered by EVs.
Figure 2.
Cellular uptake of EVs. EVs bind to recipient cell surface to trigger various intracellular signaling pathways or be internalized through different mechanisms.
Roles of EVs in cancer
The crosstalk between cancer cells and the tumor microenvironment is critical for tumor growth, metastasis, and immune evasion. The tumor microenvironment includes a large variety of cell types, such as endothelial cells, fibroblasts, lymphocytes, and macrophages, as well as the extracellular matrix (ECM). EVs that carry various nucleic acids, proteins, lipids, and other bioactive molecules have been found to be involved in multiple aspects of cancer development and therapeutic resistance by remodeling the tumor microenvironment (Figure 3; Kalluri and McAndrews, 2023; Sohal and Kasinski, 2023).
Figure 3.
Roles of EVs in cancer. EVs mediate cancer predisposition, proliferation, migration and metastasis, immune response, and therapeutic resistance.
The rapid growth of tumors requires the activation of proliferative signals and inhibition of growth suppressors (Hanahan and Weinberg, 2011). EV-mediated signal or cargo transfer plays an important role in these biological processes. For instance, EVs derived from fibroblasts, endothelial cells, and granulocytes contain c-Myc, miR-126, and miR-320, respectively, foster a tumor-favoring microenvironment, thereby increasing the predisposition to lung cancer (Pontis et al., 2021). In addition, cancer cell-derived EVs carrying c-Myc, ZIP4, or an active form of epidermal growth factor receptor (EGFR) promote their own proliferation in an autocrine manner (Al-Nedawi et al., 2008; Jin et al., 2018; Borzi et al., 2019). EVs also regulate the migration and distant metastasis of tumor cells (Becker et al., 2016). EVs containing proteolytic enzymes, such as matrix metalloproteinases and ADAM (a disintegrin and metalloprotease domain) family proteins, change the ECM compositions (Shimoda and Khokha, 2017). Moreover, cancer-associated fibroblasts, which are the most abundant stromal cells in the tumor microenvironment, secret EVs that can remodel the ECM to facilitate the spread of tumor cells (Li et al., 2021). Importantly, EVs can facilitate pre-metastatic niche formation and promote the metastasis of tumor cells to distant organs. The seminal study by Hoshino et al. (2015) reported that EVs from lung-, liver-, and brain-tropic tumor cells are preferentially taken up by lung fibroblasts and epithelial cells, liver Kupffer cells, and brain endothelial cells, respectively. They also showed that organotropic tumor exosomes create pre-metastatic environments for the metastasis of tumor cells that are not typically expected to colonize those areas (Hoshino et al., 2015). One of the mechanisms is through the transfer of exosomal integrins to resident cells. These integrins induce Src phosphorylation and pro-inflammatory S100 gene expression, which is required for pre-metastatic niche formation (Hoshino et al., 2015). Additional cargoes that mediate EV-induced cancer cell metastasis include multiple miRNAs, lipids, and proteins as reviewed elsewhere (Becker et al., 2016; Hu et al., 2023; Kalluri and McAndrews, 2023; Sohal and Kasinski, 2023).
EVs also regulate immune activation or suppression to control cancer initiation and progression (Marar et al., 2021). In 1996, EVs were discovered to act as immunomodulators. Raposo et al. (1996) found that both human and murine B lymphocytes release EVs that are enriched with major histocompatibility complex (MHC) class II to trigger T cell response. A later study reported that mature dendritic cells (DCs) release antigen-presenting EVs that contain MHC class I, MHC class II, and T cell costimulatory molecules, which activate cytotoxic T cell response to eradicate tumor cells (Zitvogel et al., 1998). As a major type of antigen-presenting cells, DCs produce EVs that can induce maturation of various types of immune cells (Wei et al., 2017; Schierer et al., 2018), orchestrate differentiation of type 1 helper T (Th1), Th2, and regulatory T cells (Cai et al., 2012; Tkach et al., 2017), and promote activation of CD4+ and CD8+ T cells (Lindenbergh et al., 2019; Cui et al., 2020; Hagerbrand et al., 2022). Conversely, activated T cells transfer EVs that contain genomic and mitochondrial DNAs to DCs, thereby inducing antiviral response and conferring protection to DCs (Torralba et al., 2018). Myeloid-derived suppressor cells can transfer GPR84 via EVs to induce senescence of CD8+ T cells by activating the p53 pathway (Liu et al., 2023). Tumor cells also release EVs to modulate immune response to influence their own growth. For instance, tumor EVs that carry PD-L1 on the membrane can directly activate PD-1/PD-L1 immune checkpoint, thus impairing the proliferation of CD8+ T cells, inhibiting interleukin 2 (IL-2) and interferon gamma (IFNγ) production, and increasing overall tumor burden (Chen et al., 2018; Ricklefs et al., 2018; Kim et al., 2019; Daassi et al., 2020). Depletion of EV-associated PD-L1 suppresses tumor growth and overcomes tumor resistance to anti-PD-L1 antibody therapy (Poggio et al., 2019). Besides, tumor EVs can deliver oncogenic mutant p53 to induce a tumor-promoting microenvironment and suppress CD4+ T lymphocytes (Dong et al., 2023). Interestingly, cancer cell-derived EVs can also stimulate antitumor immune response. They deliver tumor-associated antigens and peptide–MHC complexes for antigen presentation (Dionisi et al., 2018; Lin et al., 2020) or DNA fragments to induce maturation in DCs via the cyclic GMP–AMP synthase–stimulator of IFN genes (STING) pathway (Kitai et al., 2017).
Moreover, EVs can induce tumor resistance to therapies (Yang et al., 2022). EVs from tumor cells transfer functional cargoes that mediate resistance from resistant cells to sensitive cells, thereby promoting drug export (Ifergan et al., 2005; Safaei et al., 2005) or modulating cell survival signals that negate the cytotoxic effect of drugs (Maacha et al., 2019). For instance, cisplatin-resistant gastric cancer cells can release EVs that contain miR-769-5p, leading to the inactivation of p53 and cisplatin resistance in sensitive cells (Jing et al., 2022). Also, tumor-supporting cells, like cancer-associated fibroblasts, release EVs containing Annexin A6, miR-21, or lncRNA H19, which contributes to the development of stem-like properties and resistance to chemotherapy in tumor cells (Au Yeung et al., 2016; Ren et al., 2018; Uchihara et al., 2020).
Surface engineering for enhanced cancer targetability
As natural lipid nanoparticles, EVs have advantages over conventional lipid nanoparticles. EVs display higher biocompatibility and lower immunogenicity, as they do not carry exogenous factors that are recognized and targeted by the immune system. For the same reason, they themselves are less cytotoxic as drug carriers. Another merit of EVs is that they can cross the blood–brain barrier (BBB), thus providing an excellent delivery strategy to treat brain tumors, such as glioma (Rufino-Ramos et al., 2017; Macedo-Pereira et al., 2023). In addition, EVs may exhibit specificity for targeting tumors. For example, mesenchymal stem cell (MSC)-derived EVs can be recruited to primary and metastatic tumors (Weng et al., 2021; Hu et al., 2023; Meng et al., 2023), while tumor cell-derived EVs show some targeting specificity to the parental tumors (Kalluri and McAndrews, 2023). Furthermore, EVs from MSCs or DCs can modulate the tumor microenvironment by transferring immunomodulatory factors (Nail et al., 2023; Rebelo et al., 2023). In recent years, an increasing number of clinical trials have been conducted to evaluate the safety and effectiveness of EV-based cancer therapies (Table 1).
Table 1.
Clinical trials of EV-based cancer therapies.
| Name | Institute | Cancer | Stage | NCT number |
|---|---|---|---|---|
| iExosomes | M.D. Anderson Cancer Center | Pancreatic cancer with KRAS-G12D mutation | Phase 1 | NCT03608631 |
| exoSTING | Codiak BioSciences | Advanced solid tumors | Phase 1/2 | NCT04592484 |
| exoASO-STAT6 | Codiak BioSciences | Advanced HCC and liver metastases | Phase 1 | NCT05375604 |
| DC-derived exosomes | Gustave Roussy, Cancer Campus | Non-small cell lung cancer | Phase 2 | NCT01159288 |
| Chimeric exosomal tumor vaccines | Fudan University Pudong Hospital | Recurrent or metastatic bladder cancer | Phase 1 | NCT05559177 |
| Plant exosomes | University of Louisville | Colon cancer | Phase 1 | NCT01294072 |
| Plant exosomes | University of Louisville | Head and neck cancer with oral mucositis | Phase 1 | NCT01668849 |
| UCMSC-Exo | Wuhan Union Hospital | Myelosuppression in acute myeloid leukemia | Phase 1 | NCT06245746 |
EVs can be engineered or modified to improve their targeting specificity for tumors (Liang et al., 2021). Generally, the strategies include genetic and chemical modifications (Figure 4). Genetic engineering involves the fusion of tumor-targeting or tumor-penetrating peptides with transmembrane proteins, including LAMP2B, PDGFR, PTGFRN, CD63, CD9, and CD81, on EVs and the expression of the fused protein in donor cells, which produce EVs with the functional peptides on the surface. The LAMP2 gene encodes three protein isoforms, LAMP2A, LAMP2B, and LAMP2C. While LAMP2A is required for chaperone-mediated autophagy and LAMP2C may play a role in the degradation of RNA and DNA (Eskelinen et al., 2002, 2006), LAMP2B is enriched on the surface of EVs and thus considered a bridge connecting EVs to peptides with different properties. LAMP2B comprises an N-terminal 29-amino acid signal peptide, a large extracellular domain, and a C-terminal transmembrane domain, followed by a very short cytoplasmic domain. Thus, tumor-targeting peptides, such as RGD (Ruoslahti, 1996), RVG (El-Andaloussi et al., 2012), and tLyp-1 (Bai et al., 2020), are usually fused to the N-terminus of LAMP2B. For example, EVs expressing LAMP2B fused with the αv integrin-specific RGD peptide, which homes to tumors by binding to αv integrins (Ruoslahti, 1996), can specifically deliver anticancer drugs to tumors. Hepatocellular carcinoma (HCC)-derived EVs that express RGD peptides on the surface were found to have profound potential in the treatment of HCC (Wu et al., 2021). The modification of EVs using RGD also enhances the therapeutic effect of EV-delivered paclitaxel in the treatment of pancreatic cancer (Al Faruque et al., 2022). RGD-modified EVs loaded with siRNAs against PD-L1, which recruits tumor-associated myeloid cells, can coordinate with radiation to suppress the growth of glioblastoma (Tian et al., 2022). Moreover, RGD-modified EVs have shown prominent tumor targetability in the treatment of ovarian cancer, gastric cancer, osteosarcoma, etc. (Du et al., 2022; Guo et al., 2022; Zhao et al., 2022). Internalizing RGD (iRGD), an improved RGD peptide, was found to home to tumors by binding to both αv integrins and neuropilin-1 (Sugahara et al., 2009). It was reported that iRGD-modified EVs produced by mouse immature DCs and loaded with doxorubicin via electroporation can specifically inhibit αv integrin-positive breast cancer cell growth in vitro and in vivo (Tian et al., 2014b). iRGD-modified EVs from HEK-293T cells were also shown to deliver doxorubicin combined with radioactive 131I to target αv integrin-overexpressing anaplastic thyroid carcinoma (Wang et al., 2022). In addition, iRGD-tagged EVs containing microRNA antagonists efficiently suppress the angiogenesis and growth of nasopharyngeal carcinoma by upregulating the tumor suppressor SPRY3 (Wang et al., 2020). Furthermore, iRGD can facilitate the infiltration of lymphocytes by binding to neuropilin-1, which overcomes the endothelial barrier, to improve immunotherapy (Ding et al., 2019), thus making this tumor-penetrating peptide more fascinating in EV-based therapies.
Figure 4.
Engineering EVs for cancer therapy. EVs can be engineered with chemicals, nucleic acids, peptides, and proteins.
Besides, targeting proteins or antibodies can be genetically displayed on the surface of EVs (Liang et al., 2021). Human epidermal growth factor receptor 2 (HER2) can activate multiple intracellular pathways to support tumor cell growth and proliferation. The HER2-encoding gene ERBB2 has been found to be amplified or overexpressed in various cancers, including breast, gastric, colorectal, esophageal, lung, bladder, endometrial, and ovarian cancers (Galogre et al., 2023), which makes this membrane-bound protein an effective target for cancer therapy (Baselga and Swain, 2009). Recently, EVs decorated with the HER2–LAMP2B fusion proteins were reported to efficiently home to HER2-positive colon cancer (Liang et al., 2020). In addition, a humanized anti-HER2 single-chain variable fragment (husFvs) that can specifically recognize HER2 epitopes was constructed (Ou-Yang et al., 2018). The husFvs fused with LAMP2B was displayed on the surface of EVs, which efficiently deliver GSDMD-N mRNAs to HER2-positive breast cancer cells, consequently inducing tumor pyroptosis and immune response (Xing et al., 2023).
EV surface can be chemically modified to increase cancer targetability. Chemical reactions are responsible for the conjugation of peptides or antibodies to EV surface (Liang et al., 2021). For example, the neuropilin-1-targeted peptide RGE was conjugated to EVs by a cycloaddition reaction of sulfonyl azide (Jia et al., 2018). The peptide c (RGDyK) was conjugated to EV surface using bio-orthogonal copper-free azide alkyne cyclo-addition (Tian et al., 2018). Both modifications increased the ability of EVs to cross the BBB. Modification with a polyethylene glycol (PEG) corona is another decoration to increase the compatibility and solubility while reducing the immunogenicity of nanoparticles, such as EVs. EVs, which were decorated with ligands specific for EGFR, conjugated to phospholipid (DMPE)-PEG were reported to target EGFR-overexpressing tumor cells with increased cell specificity and prolonged circulation time (Kooijmans et al., 2016). This is consistent with another study showing that incorporation of EVs with aminoethyl anisamide-PEG improves the circulation time in the blood and the targetability to pulmonary metastases (Kim et al., 2018).
Cargo loading for cancer therapy
EVs are suitable for the delivery of various antitumor drugs, as hydrophobic molecules can be inserted in their lipid bilayer and hydrophilic molecules can be loaded in their lumen (Figure 4; Choi et al., 2022; Liu et al., 2022). The therapeutic cargoes can be divided into three categories: chemotherapeutic drugs, nucleic acids, and proteins. Chemotherapeutic drugs include a broad spectrum of molecules that can eradicate cancer cells by perturbing cell cycle, impairing microtubule stability, triggering DNA damage, or targeting the growth-promoting signaling pathways. Engineered EVs have been shown to efficiently deliver paclitaxel (Saari et al., 2015; Al Faruque et al., 2022), 5-fluorouracil (Liang et al., 2020), doxorubicin (Jang et al., 2013; Tian et al., 2014b), celastrol (Song et al., 2021), curcumin (Zhuang et al., 2011), sorafenib (Zhang et al., 2020a), imatinib (Bellavia et al., 2017), and senaparib (Chen et al., 2022).
Nucleic acids, such as mRNAs, miRNAs, and siRNAs, which modulate gene expression, can be delivered by EVs for cancer therapy (Zhang et al., 2022). The first attempt to deliver therapeutic mRNAs for cancer treatment was made in 2018. Wang et al. (2018) reported that EV-based HchrR6 mRNA delivery leads to the activation of the prodrug CNOB, efficiently inhibiting the growth of HER2-positive breast cancer cells. GSDMD-N mRNA, as aforementioned, was delivered by EVs to recipient cells to induce pyroptosis and immune response (Xing et al., 2023). In another study, human red blood cell-derived EVs were shown to transport Cas9 mRNAs to edit the genomes in both human cells and xenograft mouse models (Usman et al., 2018). Moreover, EVs may serve as a vector for the development of a variety of mRNA vaccines (Tsai et al., 2021; Popowski et al., 2022; Zhang et al., 2022).
Small inhibitory RNAs, such as miRNAs, siRNAs, and antisense oligonucleotides (ASOs), are a group of promising drugs for targeted cancer therapy. For example, GE11 peptide-modified EVs deliver let-7a miRNA to EGFR-expressing xenograft breast tumors, inhibiting the expression of HMGA2 mRNA and thus the tumor development in vivo (Ohno et al., 2013). Apo-A1-modified EVs loaded with miR-26a suppress the growth of HepG2 cells by regulating the expression of cell cycle-associated genes (Liang et al., 2018). miRNA inhibitor-containing EVs were also described to overcome chemoresistance. For instance, miR-21 was shown to trigger chemoresistance in multiple cancers (Giovannetti et al., 2010; Bourguignon et al., 2012; Naro et al., 2018). EVs harboring the miR-21 inhibitor significantly increase tumor sensitivity to doxorubicin (Zhan et al., 2020) and 5-fluorouracil (Liang et al., 2020). Engineered EVs that harbor siRNAs become a powerful strategy for cancer therapy, as this approach can efficiently target previously ‘undruggable’ genes (Friedrich and Aigner, 2022; Zhang and Zhang, 2023). EVs derived from mesenchymal cells were engineered to deliver siRNAs against oncogenic KrasG12D, a driving gene mutation in multiple cancers, leading to the repression of pancreatic cancer in several mouse models (Kamerkar et al., 2017). Han et al. (2021) also reported that engineered EVs loaded with siRNAs specific for SIRT6 suppress the growth and metastasis of SIRT6-driven prostate cancer. Survivin siRNA-containing EVs were shown to suppress the growth of prostate, breast, and colorectal cancers, using different mouse models (Pi et al., 2018). Recently, EVs loaded with siRNAs were shown to boost immunotherapy by targeting PD-L1, CD38, or YTHDF1 in glioblastoma (Tian et al., 2022), HCC (Deng and Ke, 2023), or gastric cancer (You et al., 2023), respectively.
EVs can also be engineered to deliver peptide or protein drugs to malignant sites. It was reported that SIRPα can be displayed on EV surface by fusing with PDGFR, and the engineered EVs block CD47, a ‘don't eat me’ signal, to promote the engulfment of tumor cells by macrophages (Koh et al., 2017). IL-12 was expressed on EV surface by conjugating with PTGFRN, and these EVs were shown to suppress tumor growth by increasing tumor antigen-specific CD8+ T cells (Lewis et al., 2021). EVs can also directly deliver antigens to induce T cell response. For instance, the OVA antigen was fused with the EV surface protein CD63. These engineered EVs, as a type of therapeutic vaccine, primed naive mice to induce OVA-specific CD4+ and CD8+ T cells, resulting in the regression of xenograft tumors (Kanuma et al., 2017).
Concluding remarks
EVs are a group of important mediators between cell-to-cell communications. By remodeling the tumor microenvironment, EVs from tumor cells, immune cells, and stromal cells can promote cancer cell survival, proliferation, metastasis, immune evasion, and therapeutic resistance. Most importantly, EVs as natural nanoparticles can be manipulated to serve as a potent delivery system for targeted cancer therapy. While tremendous progress has been made in the understanding of the biological functions and potential clinical uses of EVs, numerous unanswered questions remain after decades of research in this field. Although several molecules and complexes, such as the ESCRT and Rab family proteins, have been characterized as essential for EV biogenesis, specific mechanisms underlying EV formation and, particularly, the internalization and lysosomal degradation of EVs in different cell types are still unclear. The internalization of EVs mediated by receptor-mediated endocytosis, macropinocytosis, phagocytosis, clathrin-coated pits, lipid rafts, and caveolae can lead to the formation of MVBs that usually undergo degradation through lysosomes. This may severely impair the uptake and functions of therapeutic cargoes delivered by engineered EVs. Enhancing the ability of EVs to directly fuse with the plasma membrane or preventing lysosome-mediated degradation of MVBs would significantly improve the effectiveness of cargo delivery. In addition, the diverse origins and complex composition of EVs may lead to concerns about their biosafety when used in clinical settings. Finally, the most alluring question is how we can train EVs to specifically target cancer cells in a more precise manner. Conventional approaches like phage display, when coupled with artificial intelligence-driven screening, could expedite the discovery of molecules and peptides that specifically target tumors.
Contributor Information
Tao Han, Institutes of Health Central Plains, Xinxiang Key Laboratory for Molecular Oncology, Xinxiang Medical University, Xinxiang 453003, China.
Qian Hao, Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
Tengfei Chao, Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
Qinggang Sun, Institutes of Health Central Plains, Xinxiang Key Laboratory for Molecular Oncology, Xinxiang Medical University, Xinxiang 453003, China; School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China.
Yitian Chen, Institutes of Health Central Plains, Xinxiang Key Laboratory for Molecular Oncology, Xinxiang Medical University, Xinxiang 453003, China; School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China.
Bo Gao, Umibio Co. Ltd, Shanghai 201210, China.
Liping Guan, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China.
Wenjie Ren, Institutes of Health Central Plains, Xinxiang Key Laboratory for Molecular Oncology, Xinxiang Medical University, Xinxiang 453003, China.
Xiang Zhou, Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China.
Funding
This work was supported by grants from the National Natural Science Foundation of China (82173022 to Q.H., 82172891 and 82002731 to T.H., and 82072879 to X.Z.), the Innovative Research Team of High-Level Local University in Shanghai, the Doctoral Foundation of Xinxiang Medical University (XYBSKYZZ202001 to T.H.), the Key Research and Development Program of Henan Province (221111310100 to W.R.), and the open project of the Third Affiliated Hospital of Xinxiang Medical University (2022KFKTYB03).
Conflict of interest: none declared.
Author contributions: T.H., Q.H., and T.C. drafted the paper; Q.S. and Y.C. collected materials; B.G. provided important instructions; L.G. drew the figures under the supervision of T.H. and X.Z.; and W.R. and X.Z. supervised the project and revised the paper.
References
- Al Faruque H., Choi E.S., Kim J.H. et al. (2022). Enhanced effect of autologous EVs delivering paclitaxel in pancreatic cancer. J. Control Release 347, 330–346. [DOI] [PubMed] [Google Scholar]
- Al-Nedawi K., Meehan B., Micallef J. et al. (2008). Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles derived from tumour cells. Nat. Cell Biol. 10, 619–624. [DOI] [PubMed] [Google Scholar]
- Au Yeung C.L., Co N.N., Tsuruga T. et al. (2016). Exosomal transfer of stroma-derived miR21 confers paclitaxel resistance in ovarian cancer cells through targeting APAF1. Nat. Commun. 7, 11150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bai J., Duan J., Liu R. et al. (2020). Engineered targeting tLyp-1 exosomes as gene therapy vectors for efficient delivery of siRNA into lung cancer cells. Asian J. Pharm. Sci. 15, 461–471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baietti M.F., Zhang Z., Mortier E. et al. (2012). Syndecan–syntenin–ALIX regulates the biogenesis of exosomes. Nat. Cell Biol. 14, 677–685. [DOI] [PubMed] [Google Scholar]
- Baselga J., Swain S.M. (2009). Novel anticancer targets: revisiting ERBB2 and discovering ERBB3. Nat. Rev. Cancer 9, 463–475. [DOI] [PubMed] [Google Scholar]
- Becker A., Thakur B.K., Weiss J.M. et al. (2016). Extracellular vesicles in cancer: cell-to-cell mediators of metastasis. Cancer Cell 30, 836–848. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bellavia D., Raimondo S., Calabrese G. et al. (2017). Interleukin 3-receptor targeted exosomes inhibit in vitro and in vivo chronic myelogenous leukemia cell growth. Theranostics 7, 1333–1345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Borzi C., Calzolari L., Ferretti A.M. et al. (2019). c-Myc shuttled by tumour-derived extracellular vesicles promotes lung bronchial cell proliferation through miR-19b and miR-92a. Cell Death Dis. 10, 759. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bourguignon L.Y., Earle C., Wong G. et al. (2012). Stem cell marker (Nanog) and Stat-3 signaling promote microRNA-21 expression and chemoresistance in hyaluronan/CD44-activated head and neck squamous cell carcinoma cells. Oncogene 31, 149–160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cai Z., Zhang W., Yang F. et al. (2012). Immunosuppressive exosomes from TGF-β1 gene-modified dendritic cells attenuate Th17-mediated inflammatory autoimmune disease by inducing regulatory T cells. Cell Res. 22, 607–610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen G., Huang A.C., Zhang W. et al. (2018). Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature 560, 382–386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen Y., Wang L., Zheng M. et al. (2022). Engineered extracellular vesicles for concurrent anti-PDL1 immunotherapy and chemotherapy. Bioact. Mater. 9, 251–265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Choi H., Yim H., Park C. et al. (2022). Targeted delivery of exosomes armed with anti-cancer therapeutics. Membranes 12, 85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Colombo M., Moita C., van Niel G. et al. (2013). Analysis of ESCRT functions in exosome biogenesis, composition and secretion highlights the heterogeneity of extracellular vesicles. J. Cell Sci. 126, 5553–5565. [DOI] [PubMed] [Google Scholar]
- Cooks T., Pateras I.S., Jenkins L.M. et al. (2018). Mutant p53 cancers reprogram macrophages to tumor supporting macrophages via exosomal miR-1246. Nat. Commun. 9, 771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Couch Y., Buzas E.I., Di Vizio D. et al. (2021). A brief history of nearly EV-erything—the rise and rise of extracellular vesicles. J. Extracell. Vesicles 10, e12144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cui X., Wang S., Zhao N. et al. (2020). Thyrocyte-derived exosome-targeted dendritic cells stimulate strong CD4+ T lymphocyte responses. Mol. Cell. Endocrinol. 506, 110756. [DOI] [PubMed] [Google Scholar]
- Daassi D., Mahoney K.M., Freeman G.J. (2020). The importance of exosomal PDL1 in tumour immune evasion. Nat. Rev. Immunol. 20, 209–215. [DOI] [PubMed] [Google Scholar]
- Deng J., Ke H. (2023). Overcoming the resistance of hepatocellular carcinoma to PD-1/PD-L1 inhibitor and the resultant immunosuppression by CD38 siRNA-loaded extracellular vesicles. Oncoimmunology 12, 2152635. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ding N., Zou Z., Sha H. et al. (2019). iRGD synergizes with PD-1 knockout immunotherapy by enhancing lymphocyte infiltration in gastric cancer. Nat. Commun. 10, 1336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dionisi M., De Archangelis C., Battisti F. et al. (2018). Tumor-derived microvesicles enhance cross-processing ability of clinical grade dendritic cells. Front. Immunol. 9, 2481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dixson A.C., Dawson T.R., Di Vizio D. et al. (2023). Context-specific regulation of extracellular vesicle biogenesis and cargo selection. Nat. Rev. Mol. Cell Biol. 24, 454–476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dong X., Li C., Deng C. et al. (2023). Regulated secretion of mutant p53 negatively affects T lymphocytes in the tumor microenvironment. Oncogene 43, 92–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Du M., Gu J., Liu C. et al. (2022). Genome-wide CRISPR screen identified Rad18 as a determinant of doxorubicin sensitivity in osteosarcoma. J. Exp. Clin. Cancer Res. 41, 154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- El-Andaloussi S., Lee Y., Lakhal-Littleton S. et al. (2012). Exosome-mediated delivery of siRNA in vitro and in vivo. Nat. Protoc. 7, 2112–2126. [DOI] [PubMed] [Google Scholar]
- Eskelinen E.L. (2006). Roles of LAMP-1 and LAMP-2 in lysosome biogenesis and autophagy. Mol. Aspects Med. 27, 495–502. [DOI] [PubMed] [Google Scholar]
- Eskelinen E.L., Illert A.L., Tanaka Y. et al. (2002). Role of LAMP-2 in lysosome biogenesis and autophagy. Mol. Biol. Cell 13, 3355–3368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fader C.M., Sanchez D.G., Mestre M.B. et al. (2009). TI-VAMP/VAMP7 and VAMP3/cellubrevin: two v-SNARE proteins involved in specific steps of the autophagy/multivesicular body pathways. Biochim. Biophys. Acta 1793, 1901–1916. [DOI] [PubMed] [Google Scholar]
- Feng D., Zhao W.L., Ye Y.Y. et al. (2010). Cellular internalization of exosomes occurs through phagocytosis. Traffic 11, 675–687. [DOI] [PubMed] [Google Scholar]
- Friedrich M., Aigner A. (2022). Therapeutic siRNA: state-of-the-art and future perspectives. BioDrugs 36, 549–571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Galogre M., Rodin D., Pyatnitskiy M. et al. (2023). A review of HER2 overexpression and somatic mutations in cancers. Crit. Rev. Oncol. Hematol. 186, 103997. [DOI] [PubMed] [Google Scholar]
- Giovannetti E., Funel N., Peters G.J. et al. (2010). MicroRNA-21 in pancreatic cancer: correlation with clinical outcome and pharmacologic aspects underlying its role in the modulation of gemcitabine activity. Cancer Res. 70, 4528–4538. [DOI] [PubMed] [Google Scholar]
- Gonda A., Kabagwira J., Senthil G.N. et al. (2019). Internalization of exosomes through receptor-mediated endocytosis. Mol. Cancer Res. 17, 337–347. [DOI] [PubMed] [Google Scholar]
- Guo Z., Zhang Y., Xu W. et al. (2022). Engineered exosome-mediated delivery of circDIDO1 inhibits gastric cancer progression via regulation of miR-1307-3p/SOCS2 axis. J. Transl. Med. 20, 326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hagerbrand K., Varas L., Deronic A. et al. (2022). Bispecific antibodies targeting CD40 and tumor-associated antigens promote cross-priming of T cells resulting in an antitumor response superior to monospecific antibodies. J. Immunother. Cancer 10, e005018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Han Q., Xie Q.R., Li F. et al. (2021). Targeted inhibition of SIRT6 via engineered exosomes impairs tumorigenesis and metastasis in prostate cancer. Theranostics 11, 6526–6541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hanahan D., Weinberg R.A. (2011). Hallmarks of cancer: the next generation. Cell 144, 646–674. [DOI] [PubMed] [Google Scholar]
- Hoshino A., Costa-Silva B., Shen T.L. et al. (2015). Tumour exosome integrins determine organotropic metastasis. Nature 527, 329–335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Houseley J., LaCava J., Tollervey D. (2006). RNA-quality control by the exosome. Nat. Rev. Mol. Cell Biol. 7, 529–539. [DOI] [PubMed] [Google Scholar]
- Hsu C., Morohashi Y., Yoshimura S. et al. (2010). Regulation of exosome secretion by Rab35 and its GTPase-activating proteins TBC1D10A-C. J. Cell Biol. 189, 223–232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hu M., Kenific C.M., Boudreau N. et al. (2023). Tumor-derived nanoseeds condition the soil for metastatic organotropism. Semin. Cancer Biol. 93, 70–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hurley J.H. (2008). ESCRT complexes and the biogenesis of multivesicular bodies. Curr. Opin. Cell Biol. 20, 4–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ifergan I., Scheffer G.L., Assaraf Y.G. (2005). Novel extracellular vesicles mediate an ABCG2-dependent anticancer drug sequestration and resistance. Cancer Res. 65, 10952–10958. [DOI] [PubMed] [Google Scholar]
- Jahn R., Scheller R.H. (2006). SNAREs—engines for membrane fusion. Nat. Rev. Mol. Cell Biol. 7, 631–643. [DOI] [PubMed] [Google Scholar]
- Jang S.C., Kim O.Y., Yoon C.M. et al. (2013). Bioinspired exosome-mimetic nanovesicles for targeted delivery of chemotherapeutics to malignant tumors. ACS Nano 7, 7698–7710. [DOI] [PubMed] [Google Scholar]
- Jia G., Han Y., An Y. et al. (2018). NRP-1 targeted and cargo-loaded exosomes facilitate simultaneous imaging and therapy of glioma in vitro and in vivo. Biomaterials 178, 302–316. [DOI] [PubMed] [Google Scholar]
- Jin H., Liu P., Wu Y. et al. (2018). Exosomal zinc transporter ZIP4 promotes cancer growth and is a novel diagnostic biomarker for pancreatic cancer. Cancer Sci. 109, 2946–2956. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jing X., Xie M., Ding K. et al. (2022). Exosome-transmitted miR-769-5p confers cisplatin resistance and progression in gastric cancer by targeting CASP9 and promoting the ubiquitination degradation of p53. Clin. Transl. Med. 12, e780. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kalluri R., LeBleu V.S. (2020). The biology, function, and biomedical applications of exosomes. Science 367, eaau6977. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kalluri R., McAndrews K.M. (2023). The role of extracellular vesicles in cancer. Cell 186, 1610–1626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kamerkar S., LeBleu V.S., Sugimoto H. et al. (2017). Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer. Nature 546, 498–503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kanuma T., Yamamoto T., Kobiyama K. et al. (2017). CD63-mediated antigen delivery into extracellular vesicles via DNA vaccination results in robust CD8+ T cell responses. J. Immunol. 198, 4707–4715. [DOI] [PubMed] [Google Scholar]
- Kilinc S., Paisner R., Camarda R. et al. (2021). Oncogene-regulated release of extracellular vesicles. Dev. Cell 56, 1989–2006. e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim D.H., Kim H., Choi Y.J. et al. (2019). Exosomal PD-L1 promotes tumor growth through immune escape in non-small cell lung cancer. Exp. Mol. Med. 51, 1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim M.S., Haney M.J., Zhao Y. et al. (2018). Engineering macrophage-derived exosomes for targeted paclitaxel delivery to pulmonary metastases: in vitro and in vivo evaluations. Nanomedicine 14, 195–204. [DOI] [PubMed] [Google Scholar]
- Kitai Y., Kawasaki T., Sueyoshi T. et al. (2017). DNA-containing exosomes derived from cancer cells treated with topotecan activate a STING-dependent pathway and reinforce antitumor immunity. J. Immunol. 198, 1649–1659. [DOI] [PubMed] [Google Scholar]
- Koh E., Lee E.J., Nam G.H. et al. (2017). Exosome-SIRPα, a CD47 blockade increases cancer cell phagocytosis. Biomaterials 121, 121–129. [DOI] [PubMed] [Google Scholar]
- Kooijmans S.A.A., Fliervoet L.A.L., van der Meel R. et al. (2016). PEGylated and targeted extracellular vesicles display enhanced cell specificity and circulation time. J. Control Release 224, 77–85. [DOI] [PubMed] [Google Scholar]
- Le Saux S., Aarrass H., Lai-Kee-Him J. et al. (2020). Post-production modifications of murine mesenchymal stem cell (mMSC) derived extracellular vesicles (EVs) and impact on their cellular interaction. Biomaterials 231, 119675. [DOI] [PubMed] [Google Scholar]
- Lewis N.D., Sia C.L., Kirwin K. et al. (2021). Exosome surface display of IL12 results in tumor-retained pharmacology with superior potency and limited systemic exposure compared with recombinant IL12. Mol. Cancer Ther. 20, 523–534. [DOI] [PubMed] [Google Scholar]
- Li C., Teixeira A.F., Zhu H.J. et al. (2021). Cancer associated-fibroblast-derived exosomes in cancer progression. Mol. Cancer 20, 154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li H., Pinilla-Macua I., Ouyang Y. et al. (2020). Internalization of trophoblastic small extracellular vesicles and detection of their miRNA cargo in P-bodies. J. Extracell. Vesicles 9, 1812261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liang G., Kan S., Zhu Y. et al. (2018). Engineered exosome-mediated delivery of functionally active miR-26a and its enhanced suppression effect in HepG2 cells. Int. J. Nanomedicine 13, 585–599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liang G., Zhu Y., Ali D.J. et al. (2020). Engineered exosomes for targeted co-delivery of miR-21 inhibitor and chemotherapeutics to reverse drug resistance in colon cancer. J. Nanobiotechnol. 18, 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liang Y., Duan L., Lu J. et al. (2021). Engineering exosomes for targeted drug delivery. Theranostics 11, 3183–3195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin W., Xu Y., Chen X. et al. (2020). Radiation-induced small extracellular vesicles as ‘carriages’ promote tumor antigen release and trigger antitumor immunity. Theranostics 10, 4871–4884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lindenbergh M.F.S., Koerhuis D.G.J., Borg E.G.F. et al. (2019). Bystander T-cells support clonal T-cell activation by controlling the release of dendritic cell-derived immune-stimulatory extracellular vesicles. Front. Immunol. 10, 448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu J., Liu J., Qin G. et al. (2023). MDSCs-derived GPR84 induces CD8+ T-cell senescence via p53 activation to suppress the antitumor response. J. Immunother. Cancer 11, e007802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu S., Wu X., Chandra S. et al. (2022). Extracellular vesicles: emerging tools as therapeutic agent carriers. Acta Pharm. Sin. B 12, 3822–3842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maacha S., Bhat A.A., Jimenez L. et al. (2019). Extracellular vesicles-mediated intercellular communication: roles in the tumor microenvironment and anti-cancer drug resistance. Mol. Cancer 18, 55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Macedo-Pereira A., Martins C., Lima J. et al. (2023). Digging the intercellular crosstalk via extracellular vesicles: may exosomes be the drug delivery solution for target glioblastoma? J. Control Release 358, 98–115. [DOI] [PubMed] [Google Scholar]
- Marar C., Starich B., Wirtz D. (2021). Extracellular vesicles in immunomodulation and tumor progression. Nat. Immunol. 22, 560–570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meng W., Wang L., Du X. et al. (2023). Engineered mesenchymal stem cell-derived extracellular vesicles constitute a versatile platform for targeted drug delivery. J. Control Release 363, 235–252. [DOI] [PubMed] [Google Scholar]
- Mulcahy L.A., Pink R.C., Carter D.R. (2014). Routes and mechanisms of extracellular vesicle uptake. J. Extracell. Vesicles 3, 24641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Muralidharan-Chari V., Clancy J., Plou C. et al. (2009). ARF6-regulated shedding of tumor cell-derived plasma membrane microvesicles. Curr. Biol. 19, 1875–1885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nail H.M., Chiu C.C., Leung C.H. et al. (2023). Exosomal miRNA-mediated intercellular communications and immunomodulatory effects in tumor microenvironments. J. Biomed. Sci. 30, 69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Naro Y., Ankenbruck N., Thomas M. et al. (2018). Small molecule inhibition of microRNA miR-21 rescues chemosensitivity of renal-cell carcinoma to topotecan. J. Med. Chem. 61, 5900–5909. [DOI] [PubMed] [Google Scholar]
- Nechaev S., Gao C., Moreira D. et al. (2013). Intracellular processing of immunostimulatory CpG-siRNA: toll-like receptor 9 facilitates siRNA dicing and endosomal escape. J. Control Release 170, 307–315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Novo D., Heath N., Mitchell L. et al. (2018). Mutant p53s generate pro-invasive niches by influencing exosome podocalyxin levels. Nat. Commun. 9, 5069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ohno S., Takanashi M., Sudo K. et al. (2013). Systemically injected exosomes targeted to EGFR deliver antitumor microRNA to breast cancer cells. Mol. Ther. 21, 185–191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ou-Yang Q., Yan B., Li A. et al. (2018). Construction of humanized anti-HER2 single-chain variable fragments (husFvs) and achievement of potent tumor suppression with the reconstituted husFv-Fdt-tBid immunoapoptotin. Biomaterials 178, 170–182. [DOI] [PubMed] [Google Scholar]
- Pi F., Binzel D.W., Lee T.J. et al. (2018). Nanoparticle orientation to control RNA loading and ligand display on extracellular vesicles for cancer regression. Nat. Nanotechnol. 13, 82–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Poggio M., Hu T., Pai C.C. et al. (2019). Suppression of exosomal PD-L1 induces systemic anti-tumor immunity and memory. Cell 177, 414–427.e13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pontis F., Roz L., Mensah M. et al. (2021). Circulating extracellular vesicles from individuals at high-risk of lung cancer induce pro-tumorigenic conversion of stromal cells through transfer of miR-126 and miR-320. J. Exp. Clin. Cancer Res. 40, 237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Popowski K.D., Moatti A., Scull G. et al. (2022). Inhalable dry powder mRNA vaccines based on extracellular vesicles. Matter 5, 2960–2974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raposo G., Nijman H.W., Stoorvogel W. et al. (1996). B lymphocytes secrete antigen-presenting vesicles. J. Exp. Med. 183, 1161–1172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rebelo R., Xavier C.P.R., Giovannetti E. et al. (2023). Fibroblasts in pancreatic cancer: molecular and clinical perspectives. Trends Mol. Med. 29, 439–453. [DOI] [PubMed] [Google Scholar]
- Ren J., Ding L., Zhang D. et al. (2018). Carcinoma-associated fibroblasts promote the stemness and chemoresistance of colorectal cancer by transferring exosomal lncRNA H19. Theranostics 8, 3932–3948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ricklefs F.L., Alayo Q., Krenzlin H. et al. (2018). Immune evasion mediated by PD-L1 on glioblastoma-derived extracellular vesicles. Sci. Adv. 4, eaar2766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rufino-Ramos D., Albuquerque P.R., Carmona V. et al. (2017). Extracellular vesicles: novel promising delivery systems for therapy of brain diseases. J. Control Release 262, 247–258. [DOI] [PubMed] [Google Scholar]
- Ruoslahti E. (1996). RGD and other recognition sequences for integrins. Annu. Rev. Cell Dev. Biol. 12, 697–715. [DOI] [PubMed] [Google Scholar]
- Saari H., Lazaro-Ibanez E., Viitala T. et al. (2015). Microvesicle- and exosome-mediated drug delivery enhances the cytotoxicity of paclitaxel in autologous prostate cancer cells. J. Control Release 220, 727–737. [DOI] [PubMed] [Google Scholar]
- Safaei R., Larson B.J., Cheng T.C. et al. (2005). Abnormal lysosomal trafficking and enhanced exosomal export of cisplatin in drug-resistant human ovarian carcinoma cells. Mol. Cancer Ther. 4, 1595–1604. [DOI] [PubMed] [Google Scholar]
- Sapon K., Manka R., Janas T. et al. (2023). The role of lipid rafts in vesicle formation. J. Cell Sci. 136, jcs260887. [DOI] [PubMed] [Google Scholar]
- Schierer S., Ostalecki C., Zinser E. et al. (2018). Extracellular vesicles from mature dendritic cells (DC) differentiate monocytes into immature DC. Life Sci. Alliance 1, e201800093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schuldner M., Dorsam B., Shatnyeva O. et al. (2019). Exosome-dependent immune surveillance at the metastatic niche requires BAG6 and CBP/p300-dependent acetylation of p53. Theranostics 9, 6047–6062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shimoda M., Khokha R. (2017). Metalloproteinases in extracellular vesicles. Biochim. Biophys. Acta Mol. Cell Res. 1864, 1989–2000. [DOI] [PubMed] [Google Scholar]
- Sohal I.S., Kasinski A.L. (2023). Emerging diversity in extracellular vesicles and their roles in cancer. Front. Oncol. 13, 1167717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Song H., Liu B., Dong B. et al. (2021). Exosome-based delivery of natural products in cancer therapy. Front. Cell Dev. Biol. 9, 650426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sugahara K.N., Teesalu T., Karmali P.P. et al. (2009). Tissue-penetrating delivery of compounds and nanoparticles into tumors. Cancer Cell 16, 510–520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tian T., Liang R., Erel-Akbaba G. et al. (2022). Immune checkpoint inhibition in GBM primed with radiation by engineered extracellular vesicles. ACS Nano 16, 1940–1953. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tian T., Zhang H.X., He C.P. et al. (2018). Surface functionalized exosomes as targeted drug delivery vehicles for cerebral ischemia therapy. Biomaterials 150, 137–149. [DOI] [PubMed] [Google Scholar]
- Tian T., Zhu Y.L., Zhou Y.Y. et al. (2014a). Exosome uptake through clathrin-mediated endocytosis and macropinocytosis and mediating miR-21 delivery. J. Biol. Chem. 289, 22258–22267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tian Y., Li S., Song J. et al. (2014b). A doxorubicin delivery platform using engineered natural membrane vesicle exosomes for targeted tumor therapy. Biomaterials 35, 2383–2390. [DOI] [PubMed] [Google Scholar]
- Tkach M., Kowal J., Zucchetti A.E. et al. (2017). Qualitative differences in T-cell activation by dendritic cell-derived extracellular vesicle subtypes. EMBO J. 36, 3012–3028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Torralba D., Baixauli F., Villarroya-Beltri C. et al. (2018). Priming of dendritic cells by DNA-containing extracellular vesicles from activated T cells through antigen-driven contacts. Nat. Commun. 9, 2658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tsai S.J., Atai N.A., Cacciottolo M. et al. (2021). Exosome-mediated mRNA delivery in vivo is safe and can be used to induce SARS-CoV-2 immunity. J. Biol. Chem. 297, 101266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Uchihara T., Miyake K., Yonemura A. et al. (2020). Extracellular vesicles from cancer-associated fibroblasts containing Annexin A6 induces FAK–YAP activation by stabilizing β1 integrin, enhancing drug resistance. Cancer Res. 80, 3222–3235. [DOI] [PubMed] [Google Scholar]
- Usman W.M., Pham T.C., Kwok Y.Y. et al. (2018). Efficient RNA drug delivery using red blood cell extracellular vesicles. Nat. Commun. 9, 2359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van Niel G., D'Angelo G., Raposo G. (2018). Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Biol. 19, 213–228. [DOI] [PubMed] [Google Scholar]
- Wang C., Li N., Li Y. et al. (2022). Engineering a HEK-293T exosome-based delivery platform for efficient tumor-targeting chemotherapy/internal irradiation combination therapy. J. Nanobiotechnol. 20, 247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang J., Jiang Q., Faleti O.D. et al. (2020). Exosomal delivery of antagomirs targeting viral and cellular microRNAs synergistically inhibits cancer angiogenesis. Mol. Ther. Nucleic Acids 22, 153–165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang J.H., Forterre A.V., Zhao J. et al. (2018). Anti-HER2 scFv-directed extracellular vesicle-mediated mRNA-based gene delivery inhibits growth of HER2-positive human breast tumor xenografts by prodrug activation. Mol. Cancer Ther. 17, 1133–1142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang T., Gilkes D.M., Takano N. et al. (2014). Hypoxia-inducible factors and RAB22A mediate formation of microvesicles that stimulate breast cancer invasion and metastasis. Proc. Natl Acad. Sci. USA 111, E3234–E3242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wei G., Jie Y., Haibo L. et al. (2017). Dendritic cells derived exosomes migration to spleen and induction of inflammation are regulated by CCR7. Sci. Rep. 7, 42996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weng Z., Zhang B., Wu C. et al. (2021). Therapeutic roles of mesenchymal stem cell-derived extracellular vesicles in cancer. J. Hematol. Oncol. 14, 136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu H., Xing H., Wu M.C. et al. (2021). Extracellular-vesicles delivered tumor-specific sequential nanocatalysts can be used for MRI-informed nanocatalytic therapy of hepatocellular carcinoma. Theranostics 11, 64–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xing Y., Zhang F., Ji P. et al. (2023). Efficient delivery of GSDMD-N mRNA by engineered extracellular vesicles induces pyroptosis for enhanced immunotherapy. Small 19, e2204031. [DOI] [PubMed] [Google Scholar]
- Yang Q., Xu J., Gu J. et al. (2022). Extracellular vesicles in cancer drug resistance: roles, mechanisms, and implications. Adv. Sci. 9, e2201609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- You Q., Wang F., Du R. et al. (2023). m6A reader YTHDF1-targeting engineered small extracellular vesicles for gastric cancer therapy via epigenetic and immune regulation. Adv. Mater. 35, e2204910. [DOI] [PubMed] [Google Scholar]
- Zhan Q., Yi K., Qi H. et al. (2020). Engineering blood exosomes for tumor-targeting efficient gene/chemo combination therapy. Theranostics 10, 7889–7905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang C., Zhang B. (2023). RNA therapeutics: updates and future potential. Sci. China Life Sci. 66, 12–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang G., Huang X., Xiu H. et al. (2020a). Extracellular vesicles: natural liver-accumulating drug delivery vehicles for the treatment of liver diseases. J. Extracell. Vesicles 10, e12030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang S., Wang C., Ma B. et al. (2020b). Mutant p53 drives cancer metastasis via RCP-mediated Hsp90α secretion. Cell Rep. 32, 107879. [DOI] [PubMed] [Google Scholar]
- Zhang Y., Liu Q., Zhang X. et al. (2022). Recent advances in exosome-mediated nucleic acid delivery for cancer therapy. J. Nanobiotechnol. 20, 279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao Z., Shuang T., Gao Y. et al. (2022). Targeted delivery of exosomal miR-484 reprograms tumor vasculature for chemotherapy sensitization. Cancer Lett. 530, 45–58. [DOI] [PubMed] [Google Scholar]
- Zhuang X., Xiang X., Grizzle W. et al. (2011). Treatment of brain inflammatory diseases by delivering exosome encapsulated anti-inflammatory drugs from the nasal region to the brain. Mol. Ther. 19, 1769–1779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zitvogel L., Regnault A., Lozier A. et al. (1998). Eradication of established murine tumors using a novel cell-free vaccine: dendritic cell-derived exosomes. Nat. Med. 4, 594–600. [DOI] [PubMed] [Google Scholar]



