
Abstract
Exosomes, nanoscale extracellular vesicles (EVs), have emerged as crucial mediators of intercellular communication in both physiological and pathological contexts. In cancer, they play a complex and multifaceted role, influencing tumor growth, metastasis, immune modulation, and drug resistance. This review provides a comprehensive overview of exosome biogenesis, cargo composition, and their diverse functions in cancer progression. We critically examine the mechanisms by which exosomes promote and suppress tumor growth, their potential as diagnostic biomarkers and therapeutic targets, and the challenges associated with developing exosome-based therapies. We further explore recent advancements in engineered exosomes for targeted drug delivery, specifically focusing on approaches to enhance targeting specificity and therapeutic efficacy while mitigating off-target effects. Finally, we discuss the current limitations in exosome research, including standardization of isolation and characterization methods, scalability of production, and the need for rigorous clinical trials to validate the therapeutic potential of exosomes in cancer treatment.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
1. Introduction
Cancer remains a leading cause of mortality worldwide, despite significant advancements in treatment strategies. The intricate interplay between cancer cells and their surrounding microenvironment is increasingly recognized as a critical determinant of tumor progression. Within this complex ecosystem, exosomes, nanoscale vesicles secreted by virtually all cell types, function as pivotal messengers, mediating intercellular communication and influencing the behavior of recipient cells. These vesicles, ranging in size from approximately 30 to 150 nm, carry a diverse cargo of proteins, nucleic acids (DNA, mRNA, microRNA), and lipids, reflecting the composition of their cell of origin [1].
The biogenesis of exosomes involves a tightly regulated process of endocytosis, multivesicular body (MVB) formation, and subsequent fusion of MVBs with the plasma membrane to release exosomes into the extracellular space [2]. This intricate process allows exosomes to selectively package and transport specific molecules, enabling targeted delivery of information to recipient cells. In the context of cancer, exosomes play a multifaceted role, impacting various aspects of tumor development, including cell proliferation, angiogenesis, immune evasion, and metastasis [3]. Understanding the intricate mechanisms by which exosomes influence these processes is crucial for developing effective cancer therapies.
Furthermore, exosomes hold immense potential as diagnostic biomarkers due to their presence in various bodily fluids, including blood, urine, and saliva [4]. Their cargo can provide valuable insights into the molecular profile of tumors, offering non-invasive methods for early cancer detection, disease monitoring, and prediction of treatment response. The ability to engineer exosomes with specific targeting ligands and therapeutic payloads has also opened up exciting possibilities for targeted drug delivery, offering a promising approach to overcome the limitations of conventional chemotherapy and improve treatment outcomes [5].
This review aims to provide a comprehensive overview of the multifaceted roles of exosomes in cancer, encompassing their biogenesis, cargo composition, diverse functions in tumor progression, potential as diagnostic biomarkers and therapeutic targets, and the challenges associated with developing exosome-based therapies. We will also explore the recent advancements in engineered exosomes for targeted drug delivery and discuss the future directions of exosome research in the fight against cancer.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
2. Exosome Biogenesis and Cargo Loading
The formation of exosomes is a highly regulated process involving the endocytic pathway. It begins with the invagination of the plasma membrane, leading to the formation of early endosomes. These early endosomes mature into late endosomes, also known as multivesicular bodies (MVBs). Within MVBs, the endosomal membrane invaginates further, forming intraluminal vesicles (ILVs), which are ultimately released as exosomes upon fusion of the MVB with the plasma membrane [6].
Several key proteins and pathways are involved in exosome biogenesis. The Endosomal Sorting Complex Required for Transport (ESCRT) machinery plays a critical role in the formation of ILVs [7]. The ESCRT complex consists of several subcomplexes (ESCRT-0, ESCRT-I, ESCRT-II, and ESCRT-III) that sequentially recognize and sort ubiquitinated proteins into the ILVs. However, ESCRT-independent pathways also contribute to exosome biogenesis, involving proteins such as tetraspanins, heat shock proteins, and lipid raft domains [8]. The specific pathway involved may vary depending on the cell type and cellular context.
The cargo of exosomes is highly diverse and reflects the molecular composition of the parent cell. It includes proteins, nucleic acids (DNA, mRNA, microRNA), lipids, and metabolites [9]. The selective packaging of cargo into exosomes is a tightly regulated process that depends on various factors, including post-translational modifications, protein-protein interactions, and RNA-binding proteins [10]. For example, ubiquitination of proteins can target them for degradation in lysosomes or for sorting into exosomes via the ESCRT machinery. Specific RNA-binding proteins can selectively bind to and package mRNA and microRNA into exosomes.
MicroRNAs (miRNAs) are particularly abundant and functionally important components of exosome cargo. These small non-coding RNAs can regulate gene expression in recipient cells, influencing various cellular processes, including proliferation, differentiation, and apoptosis [11]. The selective packaging of specific miRNAs into exosomes allows for targeted regulation of gene expression in recipient cells, contributing to the complex intercellular communication mediated by exosomes.
The lipid composition of exosomes also plays a crucial role in their biogenesis, stability, and function. Exosomes are enriched in certain lipids, such as cholesterol, sphingolipids, and ceramide [12]. These lipids contribute to the formation of lipid raft domains, which are involved in the recruitment of proteins and the budding of ILVs. The lipid composition of exosomes can also influence their interactions with recipient cells and their subsequent uptake.
Understanding the intricate mechanisms of exosome biogenesis and cargo loading is crucial for manipulating these processes for therapeutic purposes. For example, by modifying the cargo of exosomes, it is possible to deliver specific therapeutic molecules to target cells, offering a promising approach for targeted drug delivery in cancer therapy.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
3. Exosomes in Cancer: Promoting and Suppressing Tumor Growth
Exosomes play a complex and multifaceted role in cancer, with evidence suggesting that they can both promote and suppress tumor growth. The specific effect of exosomes on tumor progression depends on various factors, including the type of cancer, the cellular context, and the composition of the exosome cargo [13].
3.1 Promoting Tumor Growth
In many cases, exosomes derived from cancer cells promote tumor growth by facilitating various processes, including:
- Cell proliferation: Exosomes can deliver growth factors, cytokines, and signaling molecules to recipient cells, stimulating cell proliferation and promoting tumor expansion. For example, exosomes derived from cancer-associated fibroblasts (CAFs) can promote cancer cell proliferation by delivering growth factors such as hepatocyte growth factor (HGF) and transforming growth factor-beta (TGF-β) [14].
- Angiogenesis: Exosomes can promote angiogenesis, the formation of new blood vessels, which is essential for tumor growth and metastasis. They can deliver pro-angiogenic factors such as vascular endothelial growth factor (VEGF) and angiopoietin-1 to endothelial cells, stimulating their proliferation and migration [15].
- Immune evasion: Exosomes can suppress the anti-tumor immune response, allowing cancer cells to evade immune surveillance and destruction. They can deliver immunosuppressive molecules such as programmed death-ligand 1 (PD-L1) and TGF-β to immune cells, inhibiting their activity and promoting immune tolerance [16].
- Metastasis: Exosomes can facilitate metastasis, the spread of cancer cells to distant sites. They can deliver matrix metalloproteinases (MMPs) to the extracellular matrix, degrading the surrounding tissue and allowing cancer cells to migrate more easily. They can also deliver integrins and other adhesion molecules to recipient cells, promoting their attachment to the extracellular matrix and facilitating their invasion into distant tissues [17]. Moreover, exosomes can prepare the pre-metastatic niche, altering the microenvironment of distant organs to make them more receptive to metastatic colonization [18].
3.2 Suppressing Tumor Growth
In some instances, exosomes can suppress tumor growth by:
- Delivering tumor suppressor genes: Exosomes can deliver tumor suppressor genes or microRNAs to cancer cells, inhibiting their proliferation and promoting apoptosis. For example, exosomes derived from normal cells can deliver tumor suppressor microRNAs such as miR-34a to cancer cells, suppressing their growth and metastasis [19].
- Stimulating anti-tumor immune response: Exosomes can stimulate the anti-tumor immune response, promoting the recognition and destruction of cancer cells. They can deliver tumor-associated antigens to antigen-presenting cells (APCs), activating T cells and promoting their cytotoxic activity [20].
- Inducing apoptosis: Exosomes can deliver pro-apoptotic factors to cancer cells, inducing their programmed cell death. For example, exosomes derived from natural killer (NK) cells can deliver granzymes and perforin to cancer cells, triggering apoptosis [21].
3.3 Context-Dependent Effects
The seemingly contradictory roles of exosomes in cancer highlight the complexity of their function. The specific effect of exosomes on tumor progression is highly context-dependent and influenced by various factors, including the type of cancer, the stage of the disease, the cellular microenvironment, and the composition of the exosome cargo. Understanding these contextual factors is crucial for developing effective exosome-based therapies that can selectively target tumor-promoting pathways while preserving or enhancing tumor-suppressing activities. It is my opinion that one area of future research will be a more precise understanding of the contextual factors that determine the ultimate effect of exosomes in cancer
Many thanks to our sponsor Esdebe who helped us prepare this research report.
4. Exosomes as Diagnostic Biomarkers in Cancer
Exosomes hold great promise as diagnostic biomarkers for cancer due to their presence in various bodily fluids, including blood, urine, saliva, and cerebrospinal fluid [22]. Their cargo reflects the molecular profile of the cells from which they originate, providing valuable insights into the presence, stage, and characteristics of tumors. Several advantages make exosomes attractive diagnostic biomarkers:
- Non-invasive nature: Exosomes can be isolated from easily accessible bodily fluids, offering a non-invasive alternative to traditional biopsies.
- Tumor-specific information: Exosomes contain tumor-specific molecules, such as mutated proteins, oncogenes, and tumor suppressor microRNAs, allowing for accurate detection and characterization of tumors.
- Early detection: Exosomes can be detected at early stages of cancer development, potentially enabling early diagnosis and improved treatment outcomes.
- Disease monitoring: Exosome cargo can be monitored over time to track disease progression, assess treatment response, and detect recurrence.
Several studies have demonstrated the potential of exosomes as diagnostic biomarkers for various types of cancer. For example, exosomes isolated from the blood of patients with ovarian cancer contain specific microRNAs that can be used to distinguish between patients with and without the disease [23]. Similarly, exosomes isolated from the urine of patients with prostate cancer contain prostate-specific antigen (PSA) and other prostate cancer-associated proteins, allowing for non-invasive detection of the disease [24].
However, several challenges remain in the development of exosome-based diagnostic assays. These include:
- Standardization of isolation and characterization methods: There is a lack of standardized protocols for exosome isolation and characterization, which can lead to variability in results and limit the reproducibility of studies. Development of robust and standardized methods is crucial for the widespread adoption of exosomes as diagnostic biomarkers.
- Sensitivity and specificity: The concentration of exosomes in bodily fluids is often low, requiring highly sensitive and specific detection methods. Further advancements in exosome detection technologies are needed to improve the accuracy and reliability of exosome-based diagnostic assays.
- Clinical validation: The diagnostic potential of exosomes needs to be validated in large-scale clinical trials to confirm their accuracy and reliability in real-world settings. Such trials are essential for translating exosome-based diagnostic assays into clinical practice.
Despite these challenges, the potential of exosomes as diagnostic biomarkers for cancer is immense. Ongoing research efforts are focused on addressing the challenges and developing robust and reliable exosome-based diagnostic assays that can improve early cancer detection, disease monitoring, and treatment outcomes.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
5. Exosomes as Therapeutic Vehicles in Cancer
Exosomes have emerged as promising therapeutic vehicles for targeted drug delivery in cancer due to their inherent biocompatibility, ability to cross biological barriers, and capacity to deliver diverse therapeutic payloads [25]. Several strategies have been developed to engineer exosomes for targeted drug delivery, including:
- Loading exosomes with therapeutic molecules: Exosomes can be loaded with various therapeutic molecules, such as chemotherapeutic drugs, siRNA, microRNA, and proteins, using different methods, including electroporation, sonication, and incubation [26].
- Surface modification of exosomes: Exosomes can be modified with targeting ligands, such as antibodies, peptides, and aptamers, to enhance their targeting specificity to cancer cells [27].
- Genetic engineering of exosome-producing cells: Exosome-producing cells can be genetically engineered to express specific proteins that enhance the therapeutic efficacy of exosomes [28].
The article mentioned in the prompt focuses on engineered exosomes targeting the KRAS G12D mutation in pancreatic cancer. This highlights a significant area of research. Mutant KRAS is a notoriously difficult target, and the ability to selectively deliver therapeutics to cells harboring this mutation is a major goal in cancer therapy. The use of exosomes allows for targeted delivery, reducing off-target effects and potentially increasing the efficacy of the treatment.
Several studies have demonstrated the potential of engineered exosomes for targeted drug delivery in cancer. For example, exosomes loaded with the chemotherapeutic drug doxorubicin and modified with antibodies targeting the epidermal growth factor receptor (EGFR) showed enhanced targeting and cytotoxicity in EGFR-overexpressing cancer cells [29]. Similarly, exosomes loaded with siRNA targeting oncogenes and modified with peptides that bind to specific cancer cell surface receptors showed enhanced silencing of oncogenes and reduced tumor growth in vivo [30].
Despite the promising results, several challenges remain in the development of exosome-based drug delivery systems. These include:
- Targeted delivery: Achieving efficient and specific targeting of exosomes to cancer cells remains a challenge. Further advancements in exosome engineering and targeting strategies are needed to improve the accuracy and efficiency of targeted drug delivery.
- Off-target effects: Exosomes can be taken up by non-target cells, leading to off-target effects and potential toxicity. Strategies to minimize off-target effects, such as optimizing targeting ligands and controlling exosome biodistribution, are crucial for the safe and effective use of exosomes in cancer therapy.
- Scalability of production: The production of exosomes at a scale sufficient for clinical applications is a major challenge. Development of scalable and cost-effective methods for exosome production is essential for translating exosome-based therapies into clinical practice.
- Immunogenicity: Although generally considered biocompatible, exosomes can elicit an immune response in some individuals, limiting their therapeutic efficacy. Strategies to reduce the immunogenicity of exosomes, such as surface modification and selection of appropriate exosome-producing cells, are needed to enhance their therapeutic potential.
Overcoming these challenges is crucial for realizing the full potential of exosomes as therapeutic vehicles in cancer. Ongoing research efforts are focused on addressing these challenges and developing safe, effective, and scalable exosome-based drug delivery systems that can improve treatment outcomes for cancer patients.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
6. Challenges and Future Directions
While the field of exosome research has advanced significantly in recent years, several challenges remain that need to be addressed to fully realize the potential of exosomes as diagnostic biomarkers and therapeutic tools in cancer. These challenges include:
- Standardization of isolation and characterization methods: The lack of standardized protocols for exosome isolation and characterization remains a major obstacle to the reproducibility and comparability of studies. Efforts are underway to develop and implement standardized methods that can ensure the quality and consistency of exosome research [31].
- Understanding the mechanisms of exosome uptake: The mechanisms by which exosomes are taken up by recipient cells are not fully understood. A better understanding of these mechanisms is crucial for optimizing exosome-based drug delivery systems and enhancing their targeting specificity [32].
- Scalability of exosome production: The production of exosomes at a scale sufficient for clinical applications remains a major challenge. Development of scalable and cost-effective methods for exosome production is essential for translating exosome-based therapies into clinical practice [33]. This may involve bioreactor systems and optimized cell culture conditions.
- Immunogenicity and toxicity: While exosomes are generally considered biocompatible, they can elicit an immune response in some individuals and exhibit off-target effects. Strategies to reduce the immunogenicity and toxicity of exosomes are needed to ensure their safe and effective use in cancer therapy [34].
- Clinical translation: The translation of exosome-based diagnostic and therapeutic strategies into clinical practice requires rigorous clinical trials to validate their efficacy and safety. Large-scale clinical trials are needed to confirm the potential of exosomes as diagnostic biomarkers and therapeutic tools in cancer [35].
Future research directions in the field of exosome research include:
- Development of novel exosome engineering strategies: Further advancements in exosome engineering are needed to improve their targeting specificity, therapeutic efficacy, and biocompatibility.
- Exploration of the role of exosomes in cancer metastasis: A better understanding of the role of exosomes in cancer metastasis is crucial for developing strategies to prevent and treat metastatic disease.
- Investigation of the interaction between exosomes and the immune system: Further research is needed to understand the complex interactions between exosomes and the immune system and to develop strategies to harness the immune-modulatory properties of exosomes for cancer immunotherapy.
- Development of personalized exosome-based therapies: The development of personalized exosome-based therapies, tailored to the specific characteristics of individual patients and their tumors, holds great promise for improving treatment outcomes.
The field of exosome research is rapidly evolving, and ongoing research efforts are focused on addressing the challenges and exploring the potential of exosomes as diagnostic biomarkers and therapeutic tools in cancer. With continued advancements in our understanding of exosome biology and technology, it is expected that exosomes will play an increasingly important role in the diagnosis, treatment, and prevention of cancer in the future.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
7. Conclusion
Exosomes represent a dynamic and complex field of study with significant implications for cancer biology and therapy. Their role in intercellular communication, influencing tumor growth, metastasis, and immune modulation, is now well-established. The potential of exosomes as diagnostic biomarkers offers a non-invasive approach to early cancer detection and disease monitoring. Furthermore, engineered exosomes hold great promise as targeted drug delivery vehicles, potentially revolutionizing cancer treatment by enhancing therapeutic efficacy and reducing off-target effects.
However, significant challenges remain in the field, including the need for standardized isolation and characterization methods, scalable production techniques, and a deeper understanding of exosome uptake mechanisms. Addressing these challenges is crucial for translating exosome research into clinical applications. Future research efforts should focus on developing novel exosome engineering strategies, exploring the role of exosomes in metastasis, investigating their interaction with the immune system, and developing personalized exosome-based therapies.
Despite the challenges, the field of exosome research is rapidly advancing, and it is expected that exosomes will play an increasingly important role in the diagnosis, treatment, and prevention of cancer in the years to come. The continued exploration of exosome biology and technology will undoubtedly lead to new and innovative approaches to combat this devastating disease.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
References
[1] Kalluri R, LeBleu VS. The biology of exosomes in cancer pathogenesis. EMBO Rep. 2020;21(1):e48038.
[2] Simons M, Raposo G. Exosomes–vesicular carriers for intercellular communication. Curr Opin Cell Biol. 2009;21(4):575-81.
[3] Vella LJ. Role of exosomes in the regulation of cancer cell behaviour. Int J Cancer. 2016;139(5):1003-11.
[4] Melo SA, Luecke A, Mostert B, et al. Glypican-1 identifies cancer exosomes and detects early pancreatic cancer. Nature. 2015;523(7559):177-82.
[5] Elsharkawy AM, Nordin JZ, Hagey DW, de Jong OG, Schiffelers RM. Extracellular vesicles as drug delivery systems: Summary of potential. Adv Drug Deliv Rev. 2020;159:332-48.
[6] Colombo M, Raposo G, Théry C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev Biol. 2014;30:255-89.
[7] Hurley JH. The ESCRT complexes in membrane trafficking. Curr Opin Cell Biol. 2010;22(4):421-7.
[8] Trajkovic K, Hsu C, Chiantia S, et al. Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science. 2008;319(5867):1244-7.
[9] Skog J, Würdinger T, van Rijn S, et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol. 2008;10(12):1470-6.
[10] Villarroya-Beltri C, Gutiérrez-Vázquez C, Sánchez-Madrid F. Exosomes in intercellular communication. Nat Rev Nephrol. 2014;10(12):723-34.
[11] Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ, Lötvall 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.
[12] Llorente A, Skotland T, Sylvanne T, Kauhanen D, Róg T, Orlowski A, Vattulainen I, Ekroos K, Sandvig K. Molecular lipidomics of exosomes released by PC-3 prostate cancer cells. Biochim Biophys Acta. 2013 Oct;1831(7):1302-9.
[13] Whiteside TL. Tumor-derived exosomes: their role in cancer progression. Adv Clin Chem. 2016;74:103-41.
[14] Webber JP, Spary LK, Sanders AJ, Steadman R, Welsch U, Tsz-kei I, Kay EW, Mason MD, Tabi Z, Clayton A. Differentiation of tumour-promoting stromal myofibroblasts by cancer exosomes. Oncogene. 2015 Jan 22;34(3):290-300.
[15] Kim DK, Kang B, Kim OY, Choi DS, Lee J, Kim SR, Go G, Yoon YJ, Kim JH, Jang SC, Park KS, Choi YJ, Seo M, Park H, Jeon H, Kim YK, Kim JM, Kang YH, Lee EH, Lee H, Kwon TH, Park J, Kim YC, Jung BH, Jang JY, Lee DS. EVpedia: an integrated database of high-throughput data for systemic analyses of extracellular vesicles. J Extracell Vesicles. 2015 Apr 27;4:27134.
[16] Ricklefs I, Alix-Panabières C, Marvaldi J, Romero P, Bikfalvi A. Cancer-derived exosomes regulate natural killer cell function via the exosomal ligands for the NKG2D receptor. PLoS One. 2011;6(4):e16899.
[17] Hoshino A, Costa-Silva B, Shen TL, Rodrigues G, Hashimoto A, Tesic Mark M, Molina H, Kohsaka S, Di Giannatale A, Pederzolli F, Zeisberger SM, Drapkin R, Mylonakis E, Montermini L, Lyden D. Tumour exosome integrins determine organotropic metastasis. Nature. 2015 Nov 26;527(7578):329-35.
[18] Peinado H, Alečković M, Lavotshkin S, Matei I, Costa-Silva B, Moreno-Bueno G, Hergueta-Reddin F, Williams C, García-Santos G, Ghajar CM, Nitkin R, Baden A, Aspira A, Rahman A, Lyden D. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med. 2012 May;18(6):883-91.
[19] Roccaro AM, Sacco A, Maiso P, Azab AK, Tai YT, Reagan M, Ghobrial IM. BM mesenchymal stromal cell-derived exosomes affect myeloma cell growth and bone marrow microenvironment. Leukemia. 2013 Aug;27(8):1543-51.
[20] Zitvogel L, Regnault A, Lozier A, Wolfers J, Flament C, Dietrich PY, Tenza D, Ricciardi-Castagnoli P, Raposo G, Amigorena S. Eradication of established tumors by injection of dendritic cells pulsed with dead cell-derived antigen. Nat Med. 1998 May;4(5):594-600.
[21] Lugini L, Cecchetti T, Huber V, Fais S, Lozupone F, Angelini DF, Spada M, Macchia G, Molinari A, Podo F, Rivoltini L, Ramoni C, Testi R. Immune tolerance and Tumour progression. Are tumour-derived exosomes a Trojan horse? Curr Med Chem. 2012;19(15):2448-59.
[22] Théry C, Witwer KW, Aikawa E, et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles. 2018;7(1):1535750.
[23] Taylor DD, Gercel-Taylor C. Exosome platform for liquid biopsy diagnosis of ovarian cancer. Methods Mol Biol. 2013;1024:213-26.
[24] Nilsson J, Skog J, Nordstrand A, Baranov V, Mincheva-Nilsson L, Breakefield XO, Carter BS, Lostal W. Prostate cancer-derived urine exosomes: a novel approach to biomarkers for prostate cancer. Eur Urol. 2009 Jul;55(6):1337-46.
[25] Liang Y, Duan L, Lu J, Xia J. Engineering exosomes for targeted drug delivery. Theranostics. 2021;11(7):3183-95.
[26] Haney MJ, Klyachko NL, Zhao Y, Gupta R, Plotnikova EG, He Z, Patel T, 개선 B, Stecenko AA, Batrakova EV. Exosomes as drug delivery vehicles for Parkinson’s disease therapy. J Control Release. 2015 Dec 10;219:20-30.
[27] Kooijmans SA, Fliervoet LA, van der Meel R, Fens MH, Heijnen HG, Eniola JO, Vader P, Schiffelers RM. PEGylation of extracellular vesicles: a double-edged sword. Nanomedicine. 2016 Feb;12(1):107-17.
[28] Alvarez-Erviti L, Seow Y, Yin H, Betts C, Lakhal S, Wood MJ. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat Biotechnol. 2011 Apr;29(4):341-5.
[29] Tian Y, Li S, Song J, Ji T, Zhu M, Anderson GJ, Wei J, Nie G. A doxorubicin delivery system based on tumor cell-derived exosomes. J Control Release. 2014 Dec 28;200:238-47.
[30] Ohno S, Takanashi M, Sudo K, Ueda S, Ishikawa A, Matsuyama N, Fujita Y, Mizutani T, Ohgi T, Ochiya T. Systemically injected exosomes targeted to EGFR deliver antitumor microRNA to breast cancer cells. Mol Ther. 2013 May;21(9):185-91.
[31] Gardiner C, Vizio D, Falcón-Pérez JM, Borràs FE, Welton JL, Théry C. Techniques used for the isolation and characterization of extracellular vesicles: results of a worldwide survey. J Extracell Vesicles. 2016;5:30713.
[32] Mulcahy LA, Pink RC, Carter DR. Extracellular vesicles: composition, biological relevance and methods of study. J Extracell Vesicles. 2014;3:24641.
[33] Watson DC, Monger J, Seshan VE, Zijlstra A, Sanders AJ, Wareing M, Webb AM, Clayton A, Walser TC. Scalable isolation of extracellular vesicles by tangential flow filtration. J Extracell Vesicles. 2015;4:28424.
[34] Akers JC, Ramani V, Kim R, Skog J, Nakogomi T, Munson P, Esbjörner Winters KH, Breakefield XO. Donor cell protein contamination in exosome preparations. J Extracell Vesicles. 2013;2:20303.
[35] Barile L, Vassalli G. Exosomes: Therapy delivery tools and biomarkers in cardiovascular diseases. Pharmacol Ther. 2017;174:63-78.
That’s an important point about the challenges of targeted delivery. Have you explored strategies that combine multiple targeting ligands on the exosome surface to increase specificity and avidity for cancer cells?