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

Liposomal form of 2-alkylthioimidazolone-based copper complexes for combined cancer therapy

    Tamara M Iakimova

    Faculty of Materials Science, Lomonosov Moscow State University, Moscow, 119991, Russia

    School of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia

    ,
    Anna A Bubley

    School of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia

    ,
    Olga P Boychenko

    School of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia

    ,
    Dmitry A Guk

    School of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia

    ,
    Alexander N Vaneev

    School of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia

    Research Laboratory of Biophysics, National University of Science & Technology, Moscow, 119049, Russia

    ,
    Andrey N Prusov

    AN Belozersky Research Institute, Moscow, 119991, Russia

    ,
    Alexander S Erofeev

    Research Laboratory of Biophysics, National University of Science & Technology, Moscow, 119049, Russia

    Research Laboratory of Scanning Probe Microscopy, Moscow Polytechnical University, Moscow, 107023, Russia

    ,
    Petr V Gorelkin

    Research Laboratory of Biophysics, National University of Science & Technology, Moscow, 119049, Russia

    ,
    Olga O Krasnovskaya

    School of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia

    ,
    Natalia L Klyachko

    *Author for correspondence:

    E-mail Address: klyachko@enzyme.chem.msu.ru

    School of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia

    &
    Kseniia Yu Vlasova

    **Author for correspondence:

    E-mail Address: vlasovakseniiayurevna@gmail.com

    School of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia

    Published Online:https://doi.org/10.2217/nnm-2023-0210

    Aim: To develop an optimized approach for encapsulating a 2-alkylthioimidazolone-based copper coordination compound within liposomes, which could offer treatment of cancer and bacterial infections by reactive oxygen species generation toxicity mechanisms. Materials & methods: For drug-loaded liposome preparation, lipids and drug mixture in organic solvents was injected into copper salt solution, forming a coordination compound simultaneously embedded in the lipid bilayer. In vitro tests were performed on MCF7 and MDA-MB-231 breast cancer cells. Results: Liposomes had a loading capacity of up to 1.75% (molar drug-to-lipid ratio). In vitro tests showed increased viability and accumulation of the liposomal formulation compared with free drug as well as lack of cytotoxicity in hepatocytes. Conclusion: This optimized technique for encapsulating large copper complexes in liposomes could be used to improve their delivery and better treat cancer and bacterial infections.

    Plain language summary

    This work introduces a new technique for copper-containing drugs encapsulation in a drug-delivery system. The drug, a promising copper compound, is embedded in lipid nanovesicles – tiny fat particles – for intravenous injection. In addition to chemical characterization of the obtained drug form, tests on cancer cells showed a noticeable effect, whereas healthy cell types were not harmed. Copper possesses not only anticancer effects but also antimicrobial properties, which are also shown by the drug form, and a test of combined suppression of cancer cell lines and bacteria was successful. Hence, the obtained drug form has the potential for dual treatment of cancer and bacterial infections.

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

    References

    • 1. Verma M, Sheoran P, Chaudhury A. Application of nanotechnology for cancer treatment. In: Gahlawat SDuhan JSalar RSiwach PKumar SKaur P (Eds). Advances in Animal Biotechnology and Its Applications. Springer, Singapore, 161–178 (2018).
    • 2. Anders C, Carey LA. Understanding and treating triple-negative breast cancer. Oncology (Williston Park) 22(11), 1233–1239 (2008).
    • 3. Guan X, Ma F, Fan Y, Zhu W, Hong R, Xu B. Platinum-based chemotherapy in triple-negative breast cancer: a systematic review and meta-analysis of randomized-controlled trials. Anticancer Drugs 26(8), 894–901 (2015).
    • 4. Isakoff SJ, Mayer EL, He L et al. TBCRC009: a multicenter phase II clinical trial of platinum monotherapy with biomarker assessment in metastatic triple-negative breast cancer. J. Clin. Oncol. 33(17), 1902–1909 (2015).
    • 5. Liedtke C, Mazouni C, Hess KR et al. Response to neoadjuvant therapy and long-term survival in patients with triple-negative breast cancer. J. Clin. Oncol. 26(8), 1275–1281 (2008).
    • 6. Geenen JJJ, Linn SC, Beijnen JH, Schellens JHM. PARP inhibitors in the treatment of triple-negative breast cancer. Clin. Pharmacokinet. 57(4), 427–437 (2018).
    • 7. Greenberg S, Rugo HS. Triple-negative breast cancer: role of antiangiogenic agents. Cancer J. 16(1), 33–38 (2010).
    • 8. Nakai K, Hung MC, Yamaguchi H. A perspective on anti-EGFR therapies targeting triple-negative breast cancer. Am. J. Cancer Res. 6(8), 1609–1623 (2016).
    • 9. Fang X, Cao J, Shen A. Advances in anti-breast cancer drugs and the application of nano-drug delivery systems in breast cancer therapy. J. Drug Deliv. Sci. Technol. 57, 101662 (2020).
    • 10. Alvarez N, Viña D, Leite CM et al. Synthesis and structural characterization of a series of ternary copper(II)-L-dipeptide-neocuproine complexes. Study of their cytotoxicity against cancer cells including MDA-MB-231, triple negative breast cancer cells. J. Inorg. Biochem. 203, 110930 (2020).
    • 11. Balsa LM, Ruiz MC, Santa Maria de la Parra L, Baran EJ, León IE. Anticancer and antimetastatic activity of copper(II)–tropolone complex against human breast cancer cells, breast multicellular spheroids and mammospheres. J. Inorg. Biochem. 204, 110975 (2020). •• Demonstrates the effect of copper complexes in triple-negative cancer treatment.
    • 12. Siddik ZH. Cisplatin: mode of cytotoxic action and molecular basis of resistance. Oncogene 22(47), 7265–7279 (2003).
    • 13. Carcelli M, Tegoni M, Bartoli J et al. In vitro and in vivo anticancer activity of tridentate thiosemicarbazone copper complexes: unravelling an unexplored pharmacological target. Eur. J. Med. Chem. 194, 112266 (2020).
    • 14. Majouga AG, Zvereva MI, Rubtsova MP et al. Mixed valence copper(I,II) binuclear complexes with unexpected structure: synthesis, biological properties and anticancer activity. J. Med. Chem. 57(14), 6252–6258 (2014). • The main reference on the copper compound Cu2Im, presented in this paper as the main model drug for liposome encapsulation.
    • 15. Qin QP, Meng T, Tan MX et al. Synthesis, crystal structure and biological evaluation of a new dasatinib copper(II) complex as telomerase inhibitor. Eur. J. Med. Chem. 143, 1597–1603 (2018).
    • 16. Beeton ML, Aldrich-Wright JR, Bolhuis A. The antimicrobial and antibiofilm activities of copper(II) complexes. J. Inorg. Biochem. 140, 167–172 (2014).
    • 17. Rolston KVI. New antimicrobial agents for the treatment of bacterial infections in cancer patients. Hematol. Oncol. 27(3), 107–114 (2009).
    • 18. Liu H, Han Y, Fu H et al. Construction and expression of sTRAIL–melittin combining enhanced anticancer activity with antibacterial activity in Escherichia coli. Appl. Microbiol. Biotechnol. 97, 2877–2884 (2013).
    • 19. Kis B, Pavel IZ, Avram S et al. Antimicrobial activity, in vitro anticancer effect (MCF-7 breast cancer cell line), antiangiogenic and immunomodulatory potentials of Populus nigra L. buds extract. BMC Complement. Med. Ther. 22(7), 4 (2022).
    • 20. Ruiz-Azuara L, Bravo-Gómez ME. Copper compounds in cancer chemotherapy. Curr. Med. Chem. 17(31), 3606–3615 (2010).
    • 21. Tiwari G, Tiwari R, Bannerjee S et al. Drug delivery systems: an updated review. Int. J. Pharm. Investig. 2(1), 2–11 (2012).
    • 22. Larrañeta E, Stewart S, Ervine M, Al-Kasasbeh R, Donnelly RF. Hydrogels for hydrophobic drug delivery. Classification, synthesis and applications. J. Funct. Biomater. 9(1), 13 (2018).
    • 23. Saman RA, Iqbal M. Nanotechnology-based drug delivery systems: past, present and future. In: Nanotechnology: Applications in Energy, Drug and Food. Siddiquee SMelvin GRahman M (Eds). Springer, Cham, 175–185 (2019).
    • 24. Saraf S, Jain A, Tiwari A, Verma A, Panda PK, Jain SK. Advances in liposomal drug delivery to cancer: an overview. J. Drug Deliv. Sci. Technol. 56(Part A), 101549 (2020).
    • 25. Alavi M, Karimi N, Safaei M. Application of various types of liposomes in drug delivery systems. Adv. Pharm. Bull. 7(1), 3–9 (2017).
    • 26. Olusanya TOB, Ahmad RRH, Ibegbu DM, Smith JR, Elkordy AA. Liposomal drug delivery systems and anticancer drugs. Molecules 23(4), 1–17 (2018).
    • 27. Gubernator J. Active methods of drug loading into liposomes: recent strategies for stable drug entrapment and increased in vivo activity. Expert Opin. Drug Deliv. 8(5), 565–580 (2011).
    • 28. Lu G, Gao X, Zhang H et al. Near-infrared light (NIR)-responsive nanoliposomes combining photodynamic therapy and chemotherapy for breast tumor control. Chin. Chem. Lett. 33(4), 1923–1926 (2022).
    • 29. Li Z, Xu Q, Lin X et al. Integrating of lipophilic platinum(IV) prodrug into liposomes for cancer therapy on patient-derived xenograft model. Chin. Chem. Lett. 33(4), 1875–1879 (2022).
    • 30. Leung AWY, Chen KTJ, Ryan GM et al. DMPC/Chol liposomal copper CX5461 is therapeutically superior to a DSPC/Chol formulation. J. Control. Rel. 345, 75–90 (2022).
    • 31. Paun RA, Dumut DC, Centorame A et al. One-step synthesis of nanoliposomal copper diethyldithiocarbamate and its assessment for cancer therapy. Pharmaceutics 14(3), 640 (2022).
    • 32. Liu H, Kong Y, Liu Z et al. Sphingomyelin-based PEGylation Cu (DDC)2 liposomes prepared via the dual function of Cu2+ for cancer therapy: facilitating DDC loading and exerting synergistic antitumor effects. Int. J. Pharm. 621, 121788 (2022).
    • 33. Wehbe M. Copper-based therapeutics: creating a formulation platform to facilitate development of an emerging drug class. [PhD thesis] University of British Columbia, Vancouver, Canada (2017).
    • 34. Leung AWY, Anantha M, Dragowska WH, Wehbe M, Bally MB. Copper-CX-5461: a novel liposomal formulation for a small molecule rRNA synthesis inhibitor. J. Control. Rel. 286, 1–9 (2018).
    • 35. Marengo A, Forciniti S, Dando I et al. Pancreatic cancer stem cell proliferation is strongly inhibited by diethyldithiocarbamate–copper complex loaded into hyaluronic acid decorated liposomes. Biochim. Biophys. Acta Gen. Subj. 1863(1), 61–72 (2019).
    • 36. Charles J, Stewart M. Colorirnetric determination of phospholipids with ammonium ferrothiocyanate. Anal. Biochem. 14, 10–14 (1979).
    • 37. Brittes J, Lúcio M, Nunes C, Lima JLFC, Reis S. Effects of resveratrol on membrane biophysical properties: relevance for its pharmacological effects. Chem. Phys. Lipids 163(8), 747–754 (2010). • Description of derivative spectrophotometry technique, which was applied for analysis of copper complex concentration changes in liposomal suspension.
    • 38. Di Giulio A, Maurizi G, Odoardi P, Saletti MA, Amicosante G, Oratore A. Encapsulation of ampicillin in reverse-phase evaporation liposomes: a direct evaluation by derivative spectrophotometry. Int. J. Pharm. 74(2–3), 183–188 (1991).
    • 39. Vaneev AN, Gorelkin PV, Garanina AS et al. In vitro and in vivo electrochemical measurement of reactive oxygen species after treatment with anticancer drugs. Anal. Chem. 92(12), 8010–8014 (2020).
    • 40. Nishimura A, Morita M, Nishimura Y, Sugino Y. A rapid and highly efficient method for preparation of competent Escherichia coli cells. Nucleic Acids Res. 18(20), 6169 (1990).
    • 41. Singh R, Patil S, Singh N, Gupta S. Dual functionality nanobioconjugates targeting intracellular bacteria in cancer cells with enhanced antimicrobial activity. Sci. Rep. 7(1), 1–10 (2017). • Reference for an approach of mammalian cells infection by bacteria internalization.
    • 42. Anderson JC, Clarke EJ, Arkin AP, Voigt CA. Environmentally controlled invasion of cancer cells by engineered bacteria. J. Mol. Biol. 355(4), 619–627 (2006).
    • 43. Mathuber M, Hager S, Keppler BK, Heffeter P, Kowol CR. Liposomal formulations of anticancer copper(II) thiosemicarbazone complexes. Dalton Trans. 50(44), 16053–16066 (2021).
    • 44. Jaafar-Maalej C, Diab R, Andrieu V, Elaissari A, Fessi H. Ethanol injection method for hydrophilic and lipophilic drug-loaded liposome preparation. J. Liposome Res. 20(3), 228–243 (2010). •• Description of a traditional liposome preparation technique – ethanol injection method, on base of which the optimized drug-encapsulation approach presented in this work was developed.
    • 45. Norkus E, Vaškelis A, Žakaite I. Influence of ionic strength and OH- ion concentration on the Cu(II) complex formation with EDTA in alkaline solutions. Talanta 43(3), 465–470 (1996).
    • 46. Briuglia ML, Rotella C, McFarlane A, Lamprou DA. Influence of cholesterol on liposome stability and on in vitro drug release. Drug Deliv. Transl. Res. 5(3), 231–242 (2015).
    • 47. Deniz A, Sade A, Severcan F, Keskin D, Tezcaner A, Banerjee S. Celecoxib-loaded liposomes: effect of cholesterol on encapsulation and in vitro release characteristics. Biosci. Rep. 30(5), 365–373 (2010). •• Description of cholesterol influence on mechanical characteristics of liposomal bilayer and encapsulation capability.
    • 48. Paradkar MM, Irudayara J. Determination of cholesterol in dairy products by infrared techniques: 2. FT-NIR method. Int. J. Dairy Technol. 55(3), 133–138 (2002).
    • 49. Gupta U, Singh VK, Kumar V, Khajuria Y. Spectroscopic studies of cholesterol: Fourier transform infra-red and vibrational frequency analysis. Mater. Focus 3(3), 211–217 (2014). •• The research on spectroscopic studies of cholesterol, applied as a reference for conducted FTIR investigation of cholesterol-Im interaction in lipid bilayer.
    • 50. Zhang Y, Yuan S, Zhou W, Xu J, Li Y. Spectroscopic evidence and molecular simulation investigation of the π–π interaction between pyrene molecules and carbon nanotubes. J. Nanosci. Nanotechnol. 7(7), 2366–2375 (2007).
    • 51. Deka MJ, Chowdhury D. Tuning electrical properties of graphene with different π-stacking organic molecules. J. Phys. Chem. C 120(7), 4121–4129 (2016).
    • 52. Lúcio M, Ferreira H, Lima JL, Reis S. Interactions between oxicams and membrane bilayers: an explanation for their different COX selectivity. Med. Chem. 2(5), 447–456 (2006). • Description of derivative spectrophotometry technique, utilized for analysis of copper complex concentration change in liposomal suspension.
    • 53. Taylor KMG, Morris RM. Thermal analysis of phase transition behaviour in liposomes. Thermochim. Acta. 248(C), 289–301 (1995).
    • 54. Beloglazkina EK, Krasnovskaya OO, Guk DA et al. Synthesis, characterization, and cytotoxicity of binuclear copper(II) complexes with tetradentate nitrogen-containing ligands bis-5-(2-pyridylmethylidene)-3,5-dihydro-4H-imidazol-4-ones. Polyhedron 148, 129–137 (2018). • A research on binuclear copper complexes, demonstrating a cytotoxicity mechanism similar to the compounds presented in research.
    • 55. Shanbhag VC, Gudekar N, Jasmer K, Papageorgiou C, Singh K, Petris MJ. Copper metabolism as a unique vulnerability in cancer. Biochim. Biophys. Acta Mol. Cell Res. 1868(2), 118893 (2021).
    • 56. Kong R, Sun G. Targeting copper metabolism: a promising strategy for cancer treatment. Front. Pharmacol. 14, 1203447 (2023).
    • 57. Yang M, Wu X, Hu J et al. COMMD10 inhibits HIF1α/CP loop to enhance ferroptosis and radiosensitivity by disrupting Cu-Fe balance in hepatocellular carcinoma. J. Hepatol. 76(5), 1138–1150 (2022).
    • 58. Samuelsson E, Shen H, Blanco E, Ferrari M, Wolfram J. Contribution of Kupffer cells to liposome accumulation in the liver. Colloids Surf. B Biointerfaces 158, 356–362 (2017).
    • 59. Inglut CT, Sorrin AJ, Kuruppu T et al. Immunological and toxicological considerations for the design of liposomes. Nanomaterials (Basel) 10(2), 1–25 (2020).
    • 60. Pagano RE, Weinstein JN. Interactions of liposomes with mammalian cells. Ann. Rev. Biophys. Bioeng. 7, 435–468 (1978).
    • 61. Hao Y, Chen Y, He X et al. Polymeric nanoparticles with ROS-responsive prodrug and platinum nanozyme for enhanced chemophotodynamic therapy of colon cancer. Adv. Sci. (Weinh.) 7(20), 1–14 (2020).
    • 62. Hao Y, Chen Y, He X et al. RGD peptide modified platinum nanozyme co-loaded glutathione-responsive prodrug nanoparticles for enhanced chemo-photodynamic bladder cancer therapy. Biomaterials 293, 121975 (2023).
    • 63. Actis P, Tokar S, Clausmeyer J et al. Electrochemical nanoprobes for single-cell analysis. ACS Nano 8(1), 875–884 (2014).
    • 64. Yamansarov EY, Lopatukhina EV, Evteev SA et al. Discovery of bivalent GalNAc-conjugated betulin as a potent ASGPR-directed agent against hepatocellular carcinoma. Bioconjug. Chem. 32(4), 763–781 (2021).
    • 65. Krasnovskaya OO, Guk DA, Naumov AE et al. Novel copper-containing cytotoxic agents based on 2-thioxoimidazolones. J. Med. Chem. 63(21), 13031–13063 (2020). •• The reference on synthesis and characterization of copper complexes 61k and 72k, applied for liposome encapsulation in this work.
    • 66. Harikumar PS. Antibacterial activity of copper nanoparticles and copper nanocomposites against Escherichia coli bacteria. Int. J. Sci. 2(02), 83–90 (2016).
    • 67. Li Y, Zhang W, Niu J, Chen Y. Mechanism of photogenerated reactive oxygen species and correlation with the antibacterial properties of engineered metal-oxide nanoparticles. ACS Nano 6(6), 5164–5173 (2012).