Article detail
Review Article | Open Access 2025;2(1):51-61 | https://doi.org/10.58832/ctr.2025.6.6.5
From biomarkers to drug delivery: advances in extracellular vesicles in cancer diagnosis and targeted therapy
Linnea Lindqvist 1 , Felix Andersson 1,* 1Department of Oncology-Pathology, Karolinska Institutet, 171 77 Solna, Sweden * Address for Correspondences:
Felix Andersson
Department of Oncology-Pathology, Karolinska Institutet, 171 77 Solna, Sweden
E-mail: felixandersson046@gmail.com
Running Title: Extracellular vesicles for cancer theranostics
Received 20 April 2025
Revised 26 May 2025
Accepted 05 June 2025
Published Online 15 June 2025
Keywords: Extracellular vesicle; Cancer; Biomarkers; Tumor progression; Targeted therapy
Abstract

Extracellular vesicles (EVs), lipid-bilayer nanoparticles secreted by diverse cell types, have emerged as pivotal mediators in cancer progression through intercellular communication. These vesicles encapsulate oncogenic biomolecules—including proteins, nucleic acids, and metabolites—enabling the reprogramming of recipient cells to foster tumor initiation, angiogenesis, immune evasion, and metastatic niche formation. Tumor-derived EVs (TDEVs) reflect parental cell molecular profiles, offering non-invasive biomarkers for cancer diagnosis. Their stability in bodily fluids and enrichment of tumor-specific markers, such as mutant DNA, oncogenic miRNAs, and immunosuppressive ligands, surpass conventional circulating biomarkers in sensitivity and specificity, positioning EVs as promising tools for liquid biopsy. In addition to their diagnostic potential, EVs have dual roles as therapeutic targets and drug delivery vehicles. TDEVs promote chemoresistance by transferring drug efflux pumps, pro-survival RNAs, and immunosuppressive factors, but their inherent biocompatibility and targeting capabilities make them ideal nanocarriers. In preclinical models, engineered EVs loaded with chemotherapeutics, siRNA, or CRISPR-Cas9 components exhibit enhanced tumor accumulation and reduced off-target toxicity. Surface modifications, such as peptide conjugation or antibody integration, further improve their homing specificity. This review synthesizes advances in EV biology, diagnostic applications, and therapeutic innovations, highlighting their translational potential in oncology. By combining mechanistic insights with translational opportunities, we highlight the evolving promise of EVs as versatile platforms for cancer management.

Reference

1. Waqas MY, Javid MA, Nazir MM, Niaz N, Nisar MF, Manzoor Z, et al. Extracellular vesicles and exosome: insight from physiological regulatory perspectives. Journal of physiology and biochemistry. 2022;78(3):573-80.

2. Malkin EZ, Bratman SV. Bioactive DNA from extracellular vesicles and particles. Cell death & disease. 2020;11(7):584.

3. Desai SA, Patel VP, Bhosle KP, Nagare SD, Thombare KC. The tumor microenvironment: shaping cancer progression and treatment response. Journal of Chemotherapy. 2025;37(1):15-44.

4. Zhang C, Qin C, Dewanjee S, Bhattacharya H, Chakraborty P, Jha NK, et al. Tumor-derived small extracellular vesicles in cancer invasion and metastasis: Molecular mechanisms, and clinical significance. Molecular Cancer. 2024;23(1):18.

5. Mun J-Y, Leem S-H, Lee JH, Kim HS. Dual relationship between stromal cells and immune cells in the tumor microenvironment. Frontiers in immunology. 2022;13:864739.

6. Zakari S, Niels NK, Olagunju GV, Nnaji PC, Ogunniyi O, Tebamifor M, et al. Emerging biomarkers for non-invasive diagnosis and treatment of cancer: a systematic review. Frontiers in oncology. 2024;14:1405267.

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

8. Claridge B, Lozano J, Poh QH, Greening DW. Development of extracellular vesicle therapeutics: Challenges, considerations, and opportunities. Frontiers in Cell and Developmental Biology. 2021;9:734720.

9. Li Z, Wang X, Wang X, Yi X, Wong YK, Wu J, et al. Research progress on the role of extracellular vesicles in neurodegenerative diseases. Translational Neurodegeneration. 2023;12(1):43.

10. Xu M, Ji J, Jin D, Wu Y, Wu T, Lin R, et al. The biogenesis and secretion of exosomes and multivesicular bodies (MVBs): Intercellular shuttles and implications in human diseases. Genes & Diseases. 2023;10(5):1894-907.

11. Clancy JW, Schmidtmann M, D’Souza‐Schorey C. The ins and outs of microvesicles. FASEB BioAdvances. 2021;3(6):399-406.

12. Kumar A, Kumar P, Sharma M, Kim S, Singh S, Kridel SJ, et al. Role of extracellular vesicles secretion in paclitaxel resistance of prostate cancer cells. Cancer Drug Resistance. 2022;5(3):612.

13. Kumar MA, Baba SK, Sadida HQ, Marzooqi SA, Jerobin J, Altemani FH, et al. Extracellular vesicles as tools and targets in therapy for diseases. Signal transduction and targeted therapy. 2024;9(1):27.

14. Jahan S, Mukherjee S, Ali S, Bhardwaj U, Choudhary RK, Balakrishnan S, et al. Pioneer role of extracellular vesicles as modulators of cancer initiation in progression, drug therapy, and vaccine prospects. Cells. 2022;11(3):490.

15. Ghadami S, Dellinger K. The lipid composition of extracellular vesicles: applications in diagnostics and therapeutic delivery. Frontiers in Molecular Biosciences. 2023;10:1198044.

16. Encarnação CC, Faria GM, Franco VA, Botelho LGX, Moraes JA, Renovato-Martins M. Interconnections within the tumor microenvironment: extracellular vesicles as critical players of metabolic reprogramming in tumor cells. Journal of Cancer Metastasis and Treatment. 2024;10:N/A-N/A.

17. Lucotti S, Kenific CM, Zhang H, Lyden D. Extracellular vesicles and particles impact the systemic landscape of cancer. The EMBO journal. 2022;41(18):e109288.

18. Muttiah B, Muhammad Fuad ND, Jaafar F, Abdullah NAH. Extracellular Vesicles in Ovarian Cancer: From Chemoresistance Mediators to Therapeutic Vectors. Biomedicines. 2024;12(8):1806.

19. Kotelevets L, Chastre E. Extracellular vesicles in colorectal cancer: from tumor growth and metastasis to biomarkers and nanomedications. Cancers. 2023;15(4):1107.

20. Bister N, Pistono C, Huremagic B, Jolkkonen J, Giugno R, Malm T. Hypoxia and extracellular vesicles: A review on methods, vesicular cargo and functions. Journal of extracellular vesicles. 2020;10(1):e12002.

21. Arneth B. Tumor microenvironment. Medicina. 2019;56(1):15.

22. Tian J-W, Zhang H-J, Li S-Y, Guo Y-L, Chen G, Yu Z-L. Tumor cell-derived extracellular vesicles in modulating phenotypes and immune functions of macrophages: mechanisms and therapeutic applications. Journal of Cancer. 2023;14(8):1321.

23. Brena D, Huang M-B, Bond V. Extracellular vesicle-mediated transport: Reprogramming a tumor microenvironment conducive with breast cancer progression and metastasis. Translational oncology. 2022;15(1):101286.

24. Lin T, Pu X, Zhou S, Huang Z, Chen Q, Zhang Y, et al. Identification of exosomal miR-484 role in reprogramming mitochondrial metabolism in pancreatic cancer through Wnt/MAPK axis control. Pharmacological Research. 2023;197:106980.

25. Fletcher S, Prochownik EV. Small-molecule inhibitors of the Myc oncoprotein. Biochimica Et Biophysica Acta (BBA)-Gene Regulatory Mechanisms. 2015;1849(5):525-43.

26. Nweke E, Ntwasa M, Brand M, Devar J, Smith M, Candy G. Increased expression of plakoglobin is associated with upregulated MAPK and PI3K/AKT signalling pathways in early resectable pancreatic ductal adenocarcinoma. Oncology letters. 2020;19(6):4133-41.

27. Nawaz M, Shah N, Zanetti BR, Maugeri M, Silvestre RN, Fatima F, et al. Extracellular vesicles and matrix remodeling enzymes: the emerging roles in extracellular matrix remodeling, progression of diseases and tissue repair. Cells. 2018;7(10):167.

28. Tognoli ML, Vlahov N, Steenbeek S, Grawenda AM, Eyres M, Cano‐Rodriguez D, et al. RASSF1C oncogene elicits amoeboid invasion, cancer stemness, and extracellular vesicle release via a SRC/Rho axis. The EMBO journal. 2021;40(20):e107680.

29. Nanru P. Immunomodulatory effects of immune cell-derived extracellular vesicles in melanoma. Frontiers in Immunology. 2024;15:1442573.

30. Ji Q, Zhou L, Sui H, Yang L, Wu X, Song Q, et al. Primary tumors release ITGBL1-rich extracellular vesicles to promote distal metastatic tumor growth through fibroblast-niche formation. Nature communications. 2020;11(1):1211.

31. Pontis F. Role of Plasma Extracellular Vesicles in Lung Cancer Pre-Metastatic Niche Formation: The Open University; 2025.

32. Musiu C. Role of tumor-derived exosomes on reprogramming myeloid cells. 2021.

33. Li C, Yang C, Yang Q. The role of miRNAs in the extracellular vesicle-mediated interplay between breast tumor cells and cancer-associated fibroblasts. Journal of Cancer Metastasis and Treatment. 2024;10:N/A-N/A.

34. Duan X, Yu X, Gan J. Extracellular vesicle-packaged miR-4253 secreted by cancer-associated fibroblasts facilitates cell proliferation in gastric cancer by inducing macrophage M2 polarization. Cancer Biology & Therapy. 2024;25(1):2424490.

35. Xia W, Tan Y, Liu Y, Xie N, Zhu H. Prospect of extracellular vesicles in tumor immunotherapy. Frontiers in Immunology. 2025;16:1525052.

36. Patras L, Paul D, Matei IR. Weaving the nest: extracellular matrix roles in pre-metastatic niche formation. Frontiers in Oncology. 2023;13:1163786.

37. Su T, Zhang P, Zhao F, Zhang S. Exosomal MicroRNAs mediating crosstalk between cancer cells with cancer-associated fibroblasts and tumor-associated macrophages in the tumor microenvironment. Frontiers in oncology. 2021;11:631703.

38. Yuan Q, Jia L, Yang J, Li W. The role of macrophages in liver metastasis: mechanisms and therapeutic prospects. Frontiers in Immunology. 2025;16:1542197.

39. Rodrigues-Junior DM, Tan SS, Lim SK, Leong HS, Melendez ME, Ramos CRN, et al. Circulating extracellular vesicle-associated TGFβ3 modulates response to cytotoxic therapy in head and neck squamous cell carcinoma. Carcinogenesis. 2019;40(12):1452-61.

40. Alfì E, Thairi C, Femmino S, Alloatti G, Moccia F, Brizzi MF, et al. Extracellular vesicles (EVs) in ischemic conditioning and angiogenesis: Focus on endothelial derived EVs. Vascular Pharmacology. 2021;140:106873.

41. Wang X, Wang L, Lin H, Zhu Y, Huang D, Lai M, et al. Research progress of CTC, ctDNA, and EVs in cancer liquid biopsy. Frontiers in Oncology. 2024;14:1303335.

42. Savoia P, Azzimonti B, Rolla R, Zavattaro E. Role of the microbiota in skin neoplasms: new therapeutic horizons. Microorganisms. 2023;11(10):2386.

43. Xu D, Chen W-Q, Liang M-X, Chen X, Liu Z, Fei Y-J, et al. Tumor-derived small extracellular vesicles promote breast cancer progression by upregulating PD-L1 expression in macrophages. Cancer Cell International. 2023;23(1):137.

44. Filipska M, Rosell R. Mutated circulating tumor DNA as a liquid biopsy in lung cancer detection and treatment. Molecular Oncology. 2021;15(6):1667-82.

45. Del Re M, Crucitta S, Gianfilippo G, Passaro A, Petrini I, Restante G, et al. Understanding the mechanisms of resistance in EGFR-positive NSCLC: from tissue to liquid biopsy to guide treatment strategy. International journal of molecular sciences. 2019;20(16):3951.

46. Taylor ML, Giacalone AG, Amrhein KD, Wilson Jr RE, Wang Y, Huang X. Nanomaterials for molecular detection and analysis of extracellular vesicles. Nanomaterials. 2023;13(3):524.

47. Forte D, Pellegrino RM, Falvo P, Garcia-Gonzalez P, Alabed HB, Maltoni F, et al. Parallel single-cell metabolic analysis and extracellular vesicle profiling reveal vulnerabilities with prognostic significance in acute myeloid leukemia. Nature Communications. 2024;15(1):10878.

48. Schubert A, Boutros M. Extracellular vesicles and oncogenic signaling. Molecular oncology. 2021;15(1):3-26.

49. Yang A, Sun H, Wang X. Intercellular transfer of multidrug resistance mediated by extracellular vesicles. Cancer Drug Resistance. 2024;7:36.

50. Simón L, Sanhueza S, Gaete-Ramírez B, Varas-Godoy M, Quest AF. Role of the pro-inflammatory tumor microenvironment in extracellular vesicle-mediated transfer of therapy resistance. Frontiers in Oncology. 2022;12:897205.

51. Han Y, Jones TW, Dutta S, Zhu Y, Wang X, Narayanan SP, et al. Overview and update on methods for cargo loading into extracellular vesicles. Processes. 2021;9(2):356.

52. Uslu D, Abas BI, Demirbolat GM, Cevik O. Effect of platelet exosomes loaded with doxorubicin as a targeted therapy on triple-negative breast cancer cells. Molecular Diversity. 2024;28(2):449-60.

53. Rezaie J, Akbari A, Rahbarghazi R. Inhibition of extracellular vesicle biogenesis in tumor cells: A possible way to reduce tumorigenesis. Cell Biochemistry and Function. 2022;40(3):248-62.

54. Zocchi MR, Tosetti F, Benelli R, Poggi A. Cancer nanomedicine special issue review anticancer drug delivery with nanoparticles: Extracellular vesicles or synthetic nanobeads as therapeutic tools for conventional treatment or immunotherapy. Cancers. 2020;12(7):1886.

55. Namba Y, Sogawa C, Okusha Y, Kawai H, Itagaki M, Ono K, et al. Depletion of lipid efflux pump ABCG1 triggers the intracellular accumulation of extracellular vesicles and reduces aggregation and tumorigenesis of metastatic cancer cells. Frontiers in Oncology. 2018;8:376.

56. De Rubis G, Bebawy M. Extracellular vesicles in chemoresistance. New Frontiers: Extracellular Vesicles. 2021:211-45.

57. Zhang X, Wang S-w. MiR-21-5p delivered by exosomes of placental mesenchymal stem cells targets the PTEN/AKT/mTOR axis to inhibit ovarian granulosa cell apoptosis. 2023.

58. Słomka A, Mocan T, Wang B, Nenu I, Urban SK, Gonzalez-Carmona MA, et al. EVs as potential new therapeutic tool/target in gastrointestinal cancer and HCC. Cancers. 2020;12(10):3019.

59. Richards KE, Xiao W, Hill R, Team UPR. Cancer-associated fibroblasts confer gemcitabine resistance to pancreatic cancer cells through PTEN-targeting miRNAs in exosomes. Cancers. 2022;14(11):2812.

60. Pandya P, Al-Qasrawi DS, Klinge S, Justilien V. Extracellular vesicles in non-small cell lung cancer stemness and clinical applications. Frontiers in immunology. 2024;15:1369356.

61. Whitehead CA, Kaye AH, Drummond KJ, Widodo SS, Mantamadiotis T, Vella LJ, et al. Extracellular vesicles and their role in glioblastoma. Critical Reviews in Clinical Laboratory Sciences. 2020;57(4):227-52.

62. Al-Dossary AA, Tawfik EA, Isichei AC, Sun X, Li J, Alshehri AA, et al. Engineered EV-mimetic nanoparticles as therapeutic delivery vehicles for high-grade serous ovarian cancer. Cancers. 2021;13(12):3075.

63. Panzarini E, Tacconi S, Carata E, Mariano S, Tata AM, Dini L. Molecular characterization of temozolomide-treated and non temozolomide-treated glioblastoma cells released extracellular vesicles and their role in the macrophage response. International Journal of Molecular Sciences. 2020;21(21):8353.

64. Ketabat F, Pundir M, Mohabatpour F, Lobanova L, Koutsopoulos S, Hadjiiski L, et al. Controlled drug delivery systems for oral cancer treatment—current status and future perspectives. Pharmaceutics. 2019;11(7):302.

65. Villata S, Canta M, Cauda V. EVs and bioengineering: from cellular products to engineered nanomachines. International journal of molecular sciences. 2020;21(17):6048.

66. Al-Jipouri A, Almurisi SH, Al-Japairai K, Bakar LM, Doolaanea AA. Liposomes or extracellular vesicles: a comprehensive comparison of both lipid bilayer vesicles for pulmonary drug delivery. Polymers. 2023;15(2):318.

67. Kostyushev D, Kostyusheva A, Brezgin S, Smirnov V, Volchkova E, Lukashev A, et al. Gene editing by extracellular vesicles. International Journal of Molecular Sciences. 2020;21(19):7362.

Date 2025.6