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

An update review of smart nanotherapeutics and liver cancer: opportunities and challenges

    Farid Mostafaei

    Faculty of Pharmacy, Tabriz University of Medical Science, Tabriz, Iran

    ,
    Somayeh Mahdinloo

    Faculty of Pharmacy, Tabriz University of Medical Science, Tabriz, Iran

    ,
    Hadi Valizadeh

    Drug Applied Research Center & Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran

    ,
    Salar Hemmati

    Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran

    ,
    Mahdieh Abdi

    Faculty of Pharmacy, Tabriz University of Medical Science, Tabriz, Iran

    ,
    Muhammad Sarfraz

    College of Pharmacy, Al Ain University, Al Ain, United Arab Emirates

    ,
    Behzad Baradaran

    Immunology Research Center & Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran

    &
    Parvin Zakeri-Milani

    *Author for correspondence:

    E-mail Address: Parvinzm@gmail.com

    Liver & Gastrointestinal Diseases Research Center & Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran

    Published Online:https://doi.org/10.2217/nnm-2023-0196

    Hepatocellular carcinoma (HCC) is the most common form of primary liver cancer, typically diagnosed in advanced stages. Chemotherapy is necessary for treating advanced liver cancer; however, several challenges affect its effectiveness. These challenges include low specificity, high dosage requirements, high systemic toxicity and severe side effects, which significantly limit the efficacy of chemotherapy. These limitations can hinder the treatment of HCC. This review focuses on the prevalence of HCC, different types of liver cancer and the staging of the disease, along with available treatment methods. Additionally, explores recent and relevant studies on smart drug- and gene-delivery systems specifically designed for HCC. These systems include targeted endogenous and exogenous stimuli-responsive platforms.

    Plain language summary

    Liver cancer is the third leading cause of cancer deaths in the world that is usually diagnosed in the last stages. Chemotherapy is commonly used to treat advanced liver cancer, but it faces several challenges that reduce its effectiveness. These challenges include low specificity (not targeting cancer cells specifically), high dosage requirements and side effects that can affect anywhere in the body. As a result, the efficacy of chemotherapy is significantly limited, making it difficult to treat liver cancer. This review discusses the prevalence of liver cancer, different types of liver cancer and how the disease is staged. It also explores various treatment methods available for liver cancer. Furthermore, the article explores recent and relevant studies on smart drug- and gene-delivery systems that are specifically designed to target liver cancer. These systems include platforms that respond to targeted and internal or external stimuli. They aim to improve the effectiveness of treatment for liver cancer.

    Graphical abstract

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

    References

    • 1. Mahmoud K, Swidan S, El-Nabarawi M, Teaima M. Lipid based nanoparticles as a novel treatment modality for hepatocellular carcinoma: a comprehensive review on targeting and recent advances. J. Nanobiotechnol. 20(1), 1–42 (2022).
    • 2. International Agency for research on cancer. Global cancer observatory (1965). www.gco.iarc.fr/today/online-analysis-pie (Accessed 19 January 2023).
    • 3. American Cancer Society. About liver cancer (1913). www.cancer.org/cancer/liver-cancer/about/what-is-key-statistics (Accessed 19 January 2023).
    • 4. Cancer Research UK. About cancer (2002). www.cancerresearchuk.org/about-cancer/liver-cancer/about-liver-cancer (Accessed 19 January 2023).
    • 5. Mohamed NK, Hamad MA, Hafez MZE, Wooley KL, Elsabahy M. Nanomedicine in management of hepatocellular carcinoma: challenges and opportunities. Int. J. Cancer 140(7), 1475–1484 (2017).
    • 6. Elnaggar MH, Abushouk AI, Hassan AHE et al. Nanomedicine as a putative approach for active targeting of hepatocellular carcinoma. Semin. Cancer Biol. 69, 91–99 (2019). •• This review article offers valuable insights into the treatment approaches for hepatocellular carcinoma (HCC), as well as the receptors linked to this particular cancer type.
    • 7. American Cancer Society. Treating liver cancer (1913). www.cancer.org/cancer/liver-cancer/treating/chemotherapy (Accessed 19 January 2023).
    • 8. American Cancer Society. Treating liver cancer (1913). www.cancer.org/cancer/liver-cancer/treating/targeted-therapy (Accessed 19 January 2023).
    • 9. Fathi M, Dalir Abdolahinia E, Barar J, Omidi Y. Smart stimuli-responsive biopolymeric nanomedicines for targeted therapy of solid tumors. Nanomedicine 15(22), 2171–2200 (2020).
    • 10. van der Meel R, Sulheim E, Shi Y, Kiessling F, Mulder WJM. Smart cancer nanomedicine. Nat. Nanotechnol. 14(11), 1007–1017 (2019). •• In this article, various aspects of nano-smart therapy have been well discussed.
    • 11. Mossenta M, Busato D, Bo MD, Macor P. Novel nanotechnology approaches to overcome drug resistance in the treatment of hepatocellular carcinoma: glypican 3 as a useful target for innovative therapies. Int. J. Mol. Sci. 23(17), 10038 (2022).
    • 12. Zhou Q, Sun X, Zeng L, Liu J, Zhang Z. A randomized multicenter phase II clinical trial of mitoxantrone-loaded nanoparticles in the treatment of 108 patients with unresected hepatocellular carcinoma. Nanomed. Nanotechnol. Biol. Med. 5(4), 419–423 (2009).
    • 13. U.S. National Institutes of Health. U.S. National Library of Medicine (1887). https://clinicaltrials.gov/ct2/results/LiverCancer/NCT01655693 (Accessed 19 January 2023).
    • 14. ONXEO (1997). www.onxeo.com/onxeo-completes-enrollment-phase-iii-study-livatag-treatment-hepatocellular-carcinoma/ (Accessed 19 January 2023).
    • 15. D'Souza AA, Devarajan PV. Asialoglycoprotein receptor mediated hepatocyte targeting - strategies and applications. J. Control. Rel. 203, 126–139 (2015).
    • 16. Alonso S. Exploiting the bioengineering versatility of lactobionic acid in targeted nanosystems and biomaterials. J. Control. Rel. 287, 216–234 (2018).
    • 17. Lee J, Choi M, Song I. Recent advances in doxorubicin formulation to enhance pharmacokinetics and tumor targeting. Pharmaceuticals 16(6), 802 (2023).
    • 18. Li F, Qin Y, Lee J et al. Stimuli-responsive nano-assemblies for remotely controlled drug delivery. J. Control. Rel. 322, 566–592 (2020).
    • 19. Mura S, Nicolas J, Couvreur P. Stimuli-responsive nanocarriers for drug delivery. Nat. Mater. 12(11), 991–1003 (2013).
    • 20. Alsawaftah NM, Awad NS, Pitt WG. pH-responsive nanocarriers in cancer therapy. Polymers 14(5), 936 (2022).
    • 21. Kumari R, Sunil D, Ningthoujam RS. Hypoxia-responsive nanoparticle based drug delivery systems in cancer therapy: an up-to-date review. J. Control. Rel. 319, 135–156 (2020).
    • 22. Li M, Zhao G, Su W, Shuai Q. Enzyme-responsive nanoparticles for anti-tumor drug delivery. Front. Chem. 8, 647 (2020).
    • 23. Liu Z, Jiang W, Nam J, Moon JJ, Kim BYS. Immuno-modulating nanomedicine for cancer therapy. Nano Lett. 18(11), 6655–6659 (2018).
    • 24. Thakur N, Thakur S, Chatterjee S, Das J. Nanoparticles as smart carriers for enhanced cancer immunotherapy. Front. Chem. 8, 597806 (2020).
    • 25. Gindy ME, Prud'homme RK. Multifunctional nanoparticles for imaging, delivery and targeting in cancer therapy. Expert. Opin. Drug Deliv. 6(8), 865–878 (2009).
    • 26. Wang J, Zhang Z, Ai Y, Liu F, Chen MM, Liu D. Lactobionic acid-modified thymine-chitosan nanoparticles as potential carriers for methotrexate delivery. Carbohydr. Res. 501, 108275 (2021).
    • 27. Li T, Yu P, Chen Y et al. N-acetylgalactosamine-decorated nanoliposomes for targeted delivery of paclitaxel to hepatocellular carcinoma. Eur. J. Med. Chem. 222, 113605 (2021).
    • 28. Ganguly S, Dewanjee S, Sen R, Chattopadhyay D. Apigenin-loaded galactose tailored PLGA nanoparticles: a possible strategy for liver targeting to treat hepatocellular carcinoma. Colloids Surf. B. 204, 111778 (2021).
    • 29. Cai Y, Xu Y, Chan HF, Fang X, He C, Chen M. Glycyrrhetinic acid mediated drug delivery carriers for hepatocellular carcinoma therapy. Mol. Pharm. 13(3), 699–709 (2016).
    • 30. Chen J, Jiang H, Wu Y, Li Y, Gao Y. A novel glycyrrhetinic acid-modified oxaliplatin liposome for liver-targeting and in vitro/vivo evaluation. Drug Des. Devel. Ther. 9, 2265–2275 (2015).
    • 31. Yan G, Chen Q, Xu L, Wei H, Ma C, Sun Y. Preparation and evaluation of liver-targeting micelles loaded with oxaliplatin. Artif. Cells Nanomed. Biotechnol. 44(2), 491–496 (2016).
    • 32. Lv Y, Li J, Chen H, Bai Y, Zhang L. Glycyrrhetinic acid-functionalized mesoporous silica nanoparticles as hepatocellular carcinoma-targeted drug carrier. Int. J. Nanomed. 12, 4361–4370 (2017).
    • 33. Hu J, Zheng Y, Wen Z et al. Construction of redox-sensitive liposomes modified by glycyrrhetinic acid and evaluation of anti-hepatocellular carcinoma activity. Chem. Phys. Lipids 252, 105292 (2023). •• An example of dual targeted and stimuli responsive drug-delivery system against hepatocellular carcinoma.
    • 34. Kong L, Sui G, Guo R et al. A multi-strategy liposome targeting hepatocellular carcinoma cells and stem cells enhances the chemotherapy effect of doxorubicin in hepatocellular carcinoma. J. Drug Deliv. Sci. Technol. 81, 104188 (2023).
    • 35. Nisha R, Kumar P, Kumar U et al. Assessment of hyaluronic acid-modified imatinib mesylate cubosomes through CD44 targeted drug delivery in NDEA-induced hepatic carcinoma. Int. J. Pharm. 622, 121848 (2022).
    • 36. Saadat M, Mostafaei F, Mahdinloo S et al. Drug delivery of pH-sensitive nanoparticles into the liver cancer cells. J. Drug Deliv. Sci. Technol. 63, 102557 (2021).
    • 37. Bian Y, Guo D. Targeted therapy for hepatocellular carcinoma: co-delivery of sorafenib and curcumin using lactosylated ph-responsive nanoparticles. Drug Des. Devel. Ther. 14, 647–659 (2020).
    • 38. Li XX, Chen J, Shen JM et al. pH-sensitive nanoparticles as smart carriers for selective intracellular drug delivery to tumor. Int. J. Pharm. 545(1–2), 274–285 (2018).
    • 39. Zhang YQ, Shen Y, Liao MM et al. Galactosylated chitosan triptolide nanoparticles for overcoming hepatocellular carcinoma: enhanced therapeutic efficacy, low toxicity, and validated network regulatory mechanisms. Nanomed.: Nanotechnol. Biol. Med. 15(1), 86–97 (2019).
    • 40. Zha Q, Wang X, Cheng X et al. Acid–degradable carboxymethyl chitosan nanogels via an ortho ester linkage mediated improved penetration and growth inhibition of 3D tumor spheroids in vitro. Mater. Sci. Eng. C 78, 246–257 (2017).
    • 41. Mojarad-Jabali S, Farshbaf M, Walker PR et al. An update on actively targeted liposomes in advanced drug delivery to glioma. Int. J. Pharm. 602, 120645 (2021).
    • 42. Zahednezhad F, Saadat M, Valizadeh H, Zakeri-Milani P, Baradaran B. Liposome and immune system interplay: challenges and potentials. J. Control. Rel. 305, 194–209 (2019).
    • 43. Wang Y, Ding R, Zhang Z, Zhong C, Wang J, Wang M. Curcumin-loaded liposomes with the hepatic and lysosomal dual-targeted effects for therapy of hepatocellular carcinoma. Int. J. Pharm. 602, 120628 (2021).
    • 44. Hu X, Zhang J, Deng L, Hu H, Hu J, Zheng G. Galactose-modified pH-sensitive niosomes for controlled release and hepatocellular carcinoma target delivery of Tanshinone IIA. AAPS PharmSciTech 22(3), 1–14 (2021).
    • 45. Mahdinloo S, Hemmati S, Valizadeh H et al. Synthesis and preparation of vitamin A coupled butein-loaded solid lipid nanoparticles for liver fibrosis therapy in rats. Int. J. Pharm. 625, 122063 (2022).
    • 46. Mahmoudian M, Valizadeh H, Löbenberg R, Zakeri-Milani P. Bortezomib-loaded lipidic-nano drug delivery systems; formulation, therapeutic efficacy, and pharmacokinetics. J. Microencapsul. 38(3), 192–202 (2021).
    • 47. Chuang CH, Wu PC, Tsai TH et al. Development of pH-sensitive cationic PEGylated solid lipid nanoparticles for selective cancer-targeted therapy. J. Biomed. Nanotechnol. 13(2), 192–203 (2017).
    • 48. Li Y, Miao Y, Chen M et al. Stepwise targeting and responsive lipid-coated nanoparticles for enhanced tumor cell sensitivity and hepatocellular carcinoma therapy. Theranostics 10(8), 3722–3736 (2020).
    • 49. Fouladi F, Steffen KJ, Mallik S. Enzyme-responsive liposomes for the delivery of anticancer drugs. Bioconjug. Chem. 28(4), 857–868 (2017).
    • 50. Kuruvilla SP, Tiruchinapally G, Crouch AC, ElSayed MEH, Greve JM. Dendrimer-doxorubicin conjugates exhibit improved anticancer activity and reduce doxorubicin-induced cardiotoxicity in a murine hepatocellular carcinoma model. PLOS ONE 12(8), e0181944 (2017).
    • 51. Liu Y, Ding X, Li J et al. Enzyme responsive drug delivery system based on mesoporous silica nanoparticles for tumor therapy in vivo. Nanotechnology 26(14), 145102 (2015).
    • 52. Ruan J, Zheng H, Rong X et al. Over-expression of cathepsin B in hepatocellular carcinomas predicts poor prognosis of HCC patients. Mol. Cancer 15(1), 1–13 (2016).
    • 53. Lee S, Song SJ, Lee J, Ha TH, Choi JS. Cathepsin b-responsive liposomes for controlled anticancer drug delivery in Hep G2 cells. Pharmaceutics 12(9), 1–12 (2020).
    • 54. Gilbert HF. Thiol/disulfide exchange equilibria and disulfidebond stability. Methods Enzymol. 251, 8–28 (1995).
    • 55. Mirhadi E, Mashreghi M, Faal Maleki M et al. Redox-sensitive nanoscale drug delivery systems for cancer treatment. Int. J. Pharm. 589, 119882 (2020).
    • 56. Li J, Huo M, Wang J et al. Redox-sensitive micelles self-assembled from amphiphilic hyaluronic acid-deoxycholic acid conjugates for targeted intracellular delivery of paclitaxel. Biomaterials 33(7), 2310–2320 (2012).
    • 57. Kuppusamy P, Li H, Ilangovan G et al. Noninvasive imaging of tumor redox status and its modification by tissue glutathione levels. Cancer Res. 62(1), 307–312 (2002).
    • 58. Torchilin VP. Multifunctional, stimuli-sensitive nanoparticulate systems for drug delivery. Nat. Rev. Drug Discov. 13(11), 813–827 (2014).
    • 59. Cai M, Li B, Lin L et al. A reduction and pH dual-sensitive nanodrug for targeted theranostics in hepatocellular carcinoma. Biomater. Sci. 8(12), 3485–3499 (2020). • An example of reduction and pH dual-sensitive nanotheranostics drug-delivery system modified with anti GPC3 Ab.
    • 60. Alsawaftah N, Pitt WG, Husseini GA. Dual-targeting and stimuli-triggered liposomal drug delivery in cancer treatment. ACS Pharmacol. Transl. Sci. 4(3), 1028–1049 (2021).
    • 61. Depalo N, Iacobazzi RM, Valente G et al. Sorafenib delivery nanoplatform based on superparamagnetic iron oxide nanoparticles magnetically targets hepatocellular carcinoma. Nano Res. 10(7), 2431–2448 (2017).
    • 62. Liao S, Liu C, Lin F, Wu KC. Functionalized magnetic iron oxide / alginate core-shell nanoparticles for targeting hyperthermia. Int. J. Nanomed. 3315–3328 (2015).
    • 63. Yang S, Cai C, Wang H et al. Drug delivery strategy in hepatocellular carcinoma therapy. Cell Commun. Signal. 20(1), 1–14 (2022).
    • 64. Boissenot T, Bordat A, Fattal E, Tsapis N. Ultrasound-triggered drug delivery for cancer treatment using drug delivery systems: from theoretical considerations to practical applications. J. Control. Rel. 241, 144–163 (2016).
    • 65. Wang J, Xia Y, Liu H et al. Poly(lactobionamidoethyl methacrylate)-based amphiphiles with ultrasound-labile components in manufacture of drug delivery nanoparticulates for augmented cytotoxic efficacy to hepatocellular carcinoma. J. Colloid Interface Sci. 551, 1–9 (2019).
    • 66. Xu J, Cheng X, Tan L et al. Microwave responsive nanoplatform via p-selectin mediated drug delivery for treatment of hepatocellular carcinoma with distant metastasis. Nano Lett. 19(5), 2914–2927 (2019).
    • 67. Liu L, Zong Z, Liu Q et al. A novel galactose-PEG-conjugated biodegradable copolymer is an efficient gene delivery vector for immunotherapy of hepatocellular carcinoma. Biomaterials 184, 20–30 (2018).
    • 68. Böttger R, Chao P, Al Fayez N, Hohenwarter L, Li S. Lipid-based nanoparticle technologies for liver targeting. Adv. Drug Deliv. Rev. 154, 79–101 (2020). •• This review article thoroughly discusses lipid nanoparticles for targeting different liver diseases and reviews the formulations that are currently in clinical trials for treating liver diseases, including hepatocellular carcinoma.
    • 69. Rana A, Adhikary M, Singh PK, Das BC, Bhatnagar S. Smart drug delivery: a window to future of translational medicine. Front. Chem. 10, 1095598 (2023).
    • 70. Liu C, Yang M, Zhang D, Chen M, Zhu D. Clinical cancer immunotherapy: current progress and prospects. Front. Immunol. 13, 961805 (2022).
    • 71. Xia Y, Guo M, Xu T et al. siRNA-loaded selenium nanoparticle modified with hyaluronic acid for enhanced hepatocellular carcinoma therapy. Int. J. Nanomed. 1539–1552 (2018).
    • 72. Perrone F, Craparo EF, Cemazar M et al. Targeted delivery of siRNAs against hepatocellular carcinoma-related genes by a galactosylated polyaspartamide copolymer. J. Control. Rel. 330, 1132–1151 (2021). • In this article, a new copolymer called galactosylated polyaspartamide was used as a nonviral vector for gene therapy against hepatocellular carcinoma.
    • 73. Journal AI, Yao Y, Wang T, Liu Y, Zhang N. Co-delivery of sorafenib and VEGF-siRNA via pH-sensitive liposomes for the synergistic treatment of hepatocellular carcinoma. Artif. Cells. Nanomed. Biotechnol. 47(1), 1374–1383 (2019). • In this article, the codelivery of drug and gene by pH-sensitive liposomes was investigated to increase the therapeutic effect of sorafenib against hepatocellular carcinoma.
    • 74. Dika IE, Lim HY, Yong WP et al. An open-label, multicenter, Phase I, dose escalation study with Phase II expansion cohort to determine the safety, pharmacokinetics, and preliminary antitumor activity of intravenous TKM-080301 in subjects with advanced hepatocellular carcinoma. Oncologist 24(6), 747–e218 (2019).
    • 75. Dou Y, Hynynen K, Allen C. To heat or not to heat: challenges with clinical translation of thermosensitive liposomes. J. Control. Rel. 249, 63–73 (2017).
    • 76. Tak WY, Lin S, Wang Y et al. Phase III heat study adding lyso-thermosensitive liposomal doxorubicin to radiofrequency ablation in patients with unresectable hepatocellular carcinoma lesions. Clin. Cancer Res. 24(1), 73–83 (2018).
    • 77. Wood BJ, Poon RT, Locklin JK et al. Phase I study of heat-deployed liposomal doxorubicin during radiofrequency ablation for hepatic malignancies. J. Vasc. Interv. Radiol. 23(2), 248–255 (2012).
    • 78. U.S. National Institutes of Health. U.S. National Library of Medicine (1877). https://classic.clinicaltrials.gov/ct2/show/record/NCT04331743 (Accessed 6 September 2023).
    • 79. U.S. National Institutes of Health. U.S. National Library of Medicine (1887). https://classic.clinicaltrials.gov/ct2/show/NCT05497453 (Accessed 6 September 2023).