We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Research Article

Engineering extracellular vesicles mimetics for targeted chemotherapy of drug-resistant ovary cancer

    Xiaoguang Liu‡

    Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity & Child Health Care Hospital, Nanjing, 210001, China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Guangquan Liu‡

    Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity & Child Health Care Hospital, Nanjing, 210001, China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Yinghua Mao‡

    Centre for Diseases Prevention & Control of Eastern Theater, Nanjing, 210002, China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Jie Luo

    Department of Healthcare, General Hospital of Eastern Theater Command, Nanjing, 210002, China

    ,
    Yongping Cao

    Centre for Diseases Prevention & Control of Eastern Theater, Nanjing, 210002, China

    ,
    Weilong Tan

    Centre for Diseases Prevention & Control of Eastern Theater, Nanjing, 210002, China

    ,
    Wenhao Li

    Centre for Diseases Prevention & Control of Eastern Theater, Nanjing, 210002, China

    ,
    Huanhuan Yu

    Department of Clinical Pharmacy, General Hospital of Eastern Theater Command, Nanjing, 210002, China

    ,
    Xuemei Jia

    **Author for correspondence:

    E-mail Address: xmjia@njmu.edu.cn

    Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity & Child Health Care Hospital, Nanjing, 210001, China

    &
    Hong Li

    *Author for correspondence: Tel.: +86 199 5176 6830;

    E-mail Address: lihong0708@sina.com

    Centre for Diseases Prevention & Control of Eastern Theater, Nanjing, 210002, China

    Published Online:https://doi.org/10.2217/nnm-2023-0289

    Aim: To develop nanocarriers for targeting the delivery of chemotherapeutics to overcome multidrug-resistant ovarian cancer. Materials & methods: Doxorubicin-loaded nanovesicles were obtained through serial extrusion, followed by loading of P-glycoprotein siRNA and folic acid. The targeting ability and anticancer efficacy of the nanovesicles were evaluated. Results: The doxorubicin-loaded nanovesicles showed a high production yield. The presence of P-glycoprotein siRNA and folic acid resulted in reversed drug resistance and tumor targeting. This nanoplatform tremendously inhibited the viability of multidrug-resistant ovarian cancer cells, which was able to target tumor tissue and suppress tumor growth without adverse effects. Conclusion: These bioengineered nanovesicles could serve as novel extracellular vesicles mimetics for chemotherapeutics delivery to overcome multidrug resistance.

    Plain language summary

    When treating cancer affecting the ovaries, which is an organ in the female reproductive system, two challenges that arise are the inefficient delivery of chemotherapeutic drugs and the development of drug resistance inside the tumor. In this study, very small nano-scale particles called nanovesicles, which contain a chemotherapeutic drug called doxorubicin, were developed in an attempt to overcome both of these concerns. These nanovesicles were secreted by a healthy cell from an ovary, isolated and loaded with doxorubicin. These nanovesicles were also loaded with siRNA, which, in this case, prevents the synthesis of a protein in ovarian tumor cells called P-glycoprotein. This protein is responsible for pumping chemotherapy drugs back out of tumor cells, so preventing its synthesis was intended to counter chemotherapeutic resistance. The targeting ability of the nanovesicle was also enhanced with folic acid, as folic acid receptors are present on the surface of these tumor cells in higher numbers. These nanovesicles were readily and specifically taken up by ovarian tumor cells in mice with induced ovarian cancer. This reversed drug resistance and enhanced the toxic effects of doxorubicin on the tumor cells, which, in turn, increased tumor cell death and prevented tumor cell migration. No obvious adverse effect was found in mice treated with the nanovesicle system compared with the free chemotherapy drug with critical systematic toxicity. This research provides new avenues for ovarian cancer treatment, with combined therapies of siRNAs and chemotherapeutic drugs, targeted to tumor cells specifically, within nanovesicles.

    Tweetable abstract

    Doxorubicin-loaded extracellular vesicles-mimetic nanovesicles were developed by the extrusion method, followed by siRNA loading and targeted modification, resulting in effective delivery of chemotherapeutics to overcome drug resistance of ovarian cancer.

    Graphical abstract

    Papers of special note have been highlighted as: • of interest

    References

    • 1. Morice P, Gouy S, Leary A. Mucinous ovarian carcinoma. N. Engl. J. Med. 380(13), 1256–1266 (2019).
    • 2. Lheureux S, Braunstein M, Oza AM. Epithelial ovarian cancer: evolution of management in the era of precision medicine. CA Cancer J. Clin. 69(4), 280–304 (2019).
    • 3. Wang Z, Guo B, Yue S, Zhao S, Meng F, Zhong Z. HER-2-mediated nano-delivery of molecular targeted drug potently suppresses orthotopic epithelial ovarian cancer and metastasis. Int. J. Pharm. 625, 122126 (2022).
    • 4. Kuroki L, Guntupalli SR. Treatment of epithelial ovarian cancer. BMJ 371, m3773 (2020).
    • 5. Song M, Fang Z, Wang J, Liu K. A nano-targeted co-delivery system based on gene regulation and molecular blocking strategy for synergistic enhancement of platinum chemotherapy sensitivity in ovarian cancer. Int. J. Pharm. 640, 123022 (2023).
    • 6. Gupta S, Pathak Y, Gupta MK, Vyas SP. Nanoscale drug delivery strategies for therapy of ovarian cancer: conventional vs targeted. Artif. Cells Nanomed. Biotechnol. 47(1), 4066–4088 (2019).
    • 7. Gokduman K. Sensitization of cisplatin-resistant ovarian cancer cells by magnetite iron oxide nanoparticles: an in vitro study. Nanomedicine 14(24), 3177–3191 (2019).
    • 8. Mittal D, Biswas L, Verma AK. Redox resetting of cisplatin-resistant ovarian cancer cells by cisplatin-encapsulated nanostructured lipid carriers. Nanomedicine 16(12), 979–995 (2021).
    • 9. Fletcher JI, Williams RT, Henderson MJ, Norris MD, Haber M. ABC transporters as mediators of drug resistance and contributors to cancer cell biology. Drug Resist. Updat. 26, 1–9 (2016).
    • 10. Yang X, Wang Y, Chen S, Zhang S, Cui C. Cetuximab-modified human serum albumin nanoparticles co-loaded with doxorubicin and MDR1 siRNA for the treatment of drug-resistant breast tumors. Int. J. Nanomed. 16, 7051–7069 (2021).
    • 11. Chen M, Wang L, Wang F et al. Quick synthesis of a novel combinatorial delivery system of siRNA and doxorubicin for a synergistic anticancer effect. Int. J. Nanomed. 14, 3557–3569 (2019).
    • 12. Tran NH, Nguyen DD, Nguyen NM et al. Dual-targeting exosomes for improved drug delivery in breast cancer. Nanomedicine 18(7), 599–611 (2023). • Illustrates a novel targeting extracellular vesicle-based drug-delivery system and its antitumor effect.
    • 13. Kang JY, Mun D, Chun Y et al. Engineered small extracellular vesicle-mediated NOX4 siRNA delivery for targeted therapy of cardiac hypertrophy. J. Extracell. Vesicles 12(10), e12371 (2023). • An interesting engineering design for extracellular vesicles for siRNA delivering, enhancing the current study.
    • 14. Bie N, Yong T, Wei Z, Gan L, Yang X. Extracellular vesicles for improved tumor accumulation and penetration. Adv. Drug Deliv. Rev. 188, 114450 (2022).
    • 15. Cao XH, Liang MX, Wu Y, Yang K, Tang JH, Zhang W. Extracellular vesicles as drug vectors for precise cancer treatment. Nanomedicine 16(17), 1519–1537 (2021).
    • 16. Zhang W, Ngo L, Tsao SC, Liu D, Wang Y. Engineered cancer-derived small extracellular vesicle-liposome hybrid delivery system for targeted treatment of breast cancer. ACS Appl. Mater. Interfaces 15(13), 16420–16433 (2023).
    • 17. Liu W, Takahashi Y, Morishita M, Nishikawa M, Takakura Y. Development of CD40L-modified tumor small extracellular vesicles for effective induction of antitumor immune response. Nanomedicine 15(17), 1641–1652 (2020).
    • 18. Wu Q, Zhou L, Lv D, Zhu X, Tang H. Exosome-mediated communication in the tumor microenvironment contributes to hepatocellular carcinoma development and progression. J. Hematol. Oncol. 12(1), 53 (2019).
    • 19. Du Y, Wang H, Yang Y et al. Extracellular vesicle mimetics: preparation from top-down approaches and biological functions. Adv. Healthc. Mater. 11(19), e2200142 (2022).
    • 20. Nelson BC, Maragh S, Ghiran IC et al. Measurement and standardization challenges for extracellular vesicle therapeutic delivery vectors. Nanomedicine 15(22), 2149–2170 (2020).
    • 21. Evers MJW, Van De Wakker SI, De Groot EM et al. Functional siRNA delivery by extracellular vesicle-liposome hybrid nanoparticles. Adv. Healthc. Mater. 11(5), e2101202 (2022).
    • 22. Zhang F, Isak AN, Yang S et al. Smartly responsive DNA-miRNA hybrids packaged in exosomes for synergistic enhancement of cancer cell apoptosis. Nanoscale 14(17), 6612–6619 (2022).
    • 23. Wu JY, Li YJ, Wang J et al. Multifunctional exosome-mimetics for targeted anti-glioblastoma therapy by manipulating protein corona. J. Nanobiotechnol. 19(1), 405 (2021).
    • 24. Kim HY, Kumar H, Jo MJ et al. Therapeutic efficacy-potentiated and diseased organ-targeting nanovesicles derived from mesenchymal stem cells for spinal cord injury treatment. Nano. Lett. 18(8), 4965–4975 (2018). • Designs targeting nanovesicles derived from mesenchymal stem cells.
    • 25. Bandeira E, Jang SC, Lässer C, Johansson K, Rådinger M, Park KS. Effects of mesenchymal stem cell-derived nanovesicles in experimental allergic airway inflammation. Respir. Res. 24(1), 3 (2023).
    • 26. Oh K, Kim SR, Kim DK et al. In vivo differentiation of therapeutic insulin-producing cells from bone marrow cells via extracellular vesicle-mimetic nanovesicles. ACS Nano 9(12), 11718–11727 (2015).
    • 27. Gu Y, Du Y, Jiang L et al. αvβ3 integrin-specific exosomes engineered with cyclopeptide for targeted delivery of triptolide against malignant melanoma. J. Nanobiotechnol. 20(1), 384 (2022). • Designs engineered targeting nanovesicles for cancer therapy.
    • 28. Lee CS, Fan J, Hwang HS et al. Bone-targeting exosome mimetics engineered by bioorthogonal surface functionalization for bone tissue engineering. Nano Lett. 23(4), 1202–1210 (2023).
    • 29. Taylor DD. Isolation and molecular characterization of extracellular vesicles. Methods 87, 1–2 (2015).
    • 30. Yan J, Zhang N, Zhang Z et al. Redox-responsive polyethyleneimine/tetrahedron DNA/doxorubicin nanocomplexes for deep cell/tissue penetration to overcome multidrug resistance. J. Control. Rel. 329, 36–49 (2021).
    • 31. Wang AT, Liang DS, Liu YJ, Qi XR. Roles of ligand and TPGS of micelles in regulating internalization, penetration and accumulation against sensitive or resistant tumor and therapy for multidrug resistant tumors. Biomaterials 53, 160–172 (2015).
    • 32. Carron PM, Crowley A, O'Shea D et al. Targeting the folate receptor: improving efficacy in inorganic medicinal chemistry. Curr. Med. Chem. 25(23), 2675–2708 (2018).
    • 33. Szebényi K, Füredi A, Bajtai E et al. Effective targeting of breast cancer by the inhibition of P-glycoprotein mediated removal of toxic lipid peroxidation byproducts from drug tolerant persister cells. Drug Resist. Updat. 71, 101007 (2023).
    • 34. Li SP, Lin ZX, Jiang XY, Yu XY. Exosomal cargo-loading and synthetic exosome-mimics as potential therapeutic tools. Acta Pharmacol. Sin. 39(4), 542–551 (2018).
    • 35. Yong T, Wei Z, Gan L, Yang X. Extracellular-vesicle-based drug delivery systems for enhanced antitumor therapies through modulating the cancer-immunity cycle. Adv. Mater. 34(52), e2201054 (2022).
    • 36. Wang T, Luo Y, Lv H, Wang J, Zhang Y, Pei R. Aptamer-based erythrocyte-derived mimic vesicles loaded with siRNA and doxorubicin for the targeted treatment of multidrug-resistant tumors. ACS Appl. Mater. Interfaces 11(49), 45455–45466 (2019). • Similar engineering design for extracellular vesicles as the current study.
    • 37. Pooresmaeil M, Namazi H. Folic acid-modified photoluminescent dialdehyde carboxymethyl cellulose crosslinked bionanogels for pH-controlled and tumor-targeted co-drug delivery. Int. J. Biol. Macromol. 200, 247–262 (2022).
    • 38. Wei H, Chen J, Wang S et al. A nanodrug consisting of doxorubicin and exosome derived from mesenchymal stem cells for osteosarcoma treatment in vitro. Int. J. Nanomed. 14, 8603–8610 (2019).
    • 39. Lennaárd AJ, Mamand DR, Wiklander RJ, El Andaloussi S, Wiklander OPB. Optimised electroporation for loading of extracellular vesicles with doxorubicin. Pharmaceutics 14(1), 38 (2021).
    • 40. Thakur A, Sidu RK, Zou H, Alam MK, Yang M, Lee Y. Inhibition of glioma cells' proliferation by doxorubicin-loaded exosomes via microfluidics. Int. J. Nanomed. 15, 8331–8343 (2020).
    • 41. Bar A, Kryukov O, Etzion S, Cohen S. Engineered extracellular vesicle-mediated delivery of miR-199a-3p increases the viability of 3D-printed cardiac patches. Int. J. Biol. Macromol. 9(2), 670 (2023).
    • 42. Abbasi M, Lavasanifar A, Uludag H. Recent attempts at RNAi-mediated P-glycoprotein downregulation for reversal of multidrug resistance in cancer. Med. Res. Rev. 33(1), 33–53 (2013).
    • 43. Reynolds A, Leake D, Boese Q, Scaringe S, Marshall WS, Khvorova A. Rational siRNA design for RNA interference. Nat. Biotechnol. 22(3), 326–330 (2004).
    • 44. Bajracharya R, Song JG, Patil BR et al. Functional ligands for improving anticancer drug therapy: current status and applications to drug delivery systems. Drug Deliv. 29(1), 1959–1970 (2022).
    • 45. Fahmy SA, Preis E, Dayyih AA et al. Thermosensitive liposomes encapsulating nedaplatin and picoplatin demonstrate enhanced cytotoxicity against breast cancer cells. ACS Omega 7(46), 42115–42125 (2022).
    • 46. Veronese FM, Caliceti P, Schiavon O, Sergi M. Polyethylene glycol-superoxide dismutase, a conjugate in search of exploitation. Adv. Drug Deliv. Rev. 54(4), 587–606 (2002).
    • 47. Gangadaran P, Ahn BC. Extracellular vesicle- and extracellular vesicle mimetics-based drug delivery systems: new perspectives, challenges, and clinical developments. Pharmaceutics 12(5), 442 (2020).
    • 48. Chitti SV, Nedeva C, Manickam R, Fonseka P, Mathivanan S. Extracellular vesicles as drug targets and delivery vehicles for cancer therapy. Pharmaceutics 14(12), 2822 (2022).
    • 49. Li Y, Wang J, Wientjes MG, Au JL. Delivery of nanomedicines to extracellular and intracellular compartments of a solid tumor. Adv. Drug Deliv. Rev. 64(1), 29–39 (2012).
    • 50. Parolini I, Federici C, Raggi C et al. Microenvironmental pH is a key factor for exosome traffic in tumor cells. J. Biol. Chem. 284(49), 34211–34222 (2009).
    • 51. Aloss K, Hamar P. Recent preclinical and clinical progress in liposomal doxorubicin. Pharmaceutics 15(3), 893 (2023).
    • 52. Figueiredo VC, Mccarthy JJ. Targeting cancer via ribosome biogenesis: the cachexia perspective. Cell Mol. Life Sci. 78(15), 5775–5787 (2021).
    • 53. Daly LE, Éb NB, Power DG, Cushen SJ, James K, Ryan AM. Loss of skeletal muscle during systemic chemotherapy is prognostic of poor survival in patients with foregut cancer. J. Cachexia Sarcopenia Muscle 9(2), 315–325 (2018).
    • 54. Figueiredo VC, Mccarthy JJ. Regulation of ribosome biogenesis in skeletal muscle hypertrophy. Physiology 34(1), 30–42 (2019).
    • 55. Zhou S, Starkov A, Froberg MK, Leino RL, Wallace KB. Cumulative and irreversible cardiac mitochondrial dysfunction induced by doxorubicin. Cancer Res. 61(2), 771–777 (2001).
    • 56. Akhtar N, Mohammed HA, Yusuf M, Al-Subaiyel A, Sulaiman GM, Khan RA. SPIONs conjugate supported anticancer drug doxorubicin's delivery: current status, challenges, and prospects. Nanomaterials 12(20), 3686 (2022).