Targeted Liposomal Drug Delivery: Overview of the Current Applications and Challenges
Abstract
:1. Introduction
2. Passive Targeted Liposomes
2.1. Enhanced Permeation and Retention Effect
2.2. Liposomes Conjugated with Polyethylene Glycol
3. Active Targeted Liposomes
3.1. Antibodies
3.2. Peptides
3.3. Folate
3.4. Aptamer
4. pH-Sensitive Liposomes
5. Temperature Sensitive Liposomes
6. Future Perspectives
7. Conclusions
Funding
Conflicts of Interest
References
- Aghila Rani, K.G.; Hamad, M.A.; Zaher, D.M.; Sieburth, S.M.; Madani, N.; Al-Tel, T.H. Drug development post COVID-19 pandemic: Toward a better system to meet current and future global health challenges. Expert. Opin. Drug Discov. 2021, 16, 365–371. [Google Scholar] [CrossRef] [PubMed]
- Sun, D.; Gao, W.; Hu, H.; Zhou, S. Why 90% of clinical drug development fails and how to improve it? Acta Pharm. Sin. B 2022, 12, 3049–3062. [Google Scholar] [CrossRef] [PubMed]
- Harrison, R.K. Phase II and phase III failures: 2013–2015. Nat. Rev. Drug Discov. 2016, 15, 817–818. [Google Scholar] [CrossRef]
- Gavas, S.; Quazi, S.; Karpiński, T.M. Nanoparticles for Cancer Therapy: Current Progress and Challenges. Nanoscale Res. Lett. 2021, 16, 173. [Google Scholar] [CrossRef] [PubMed]
- William, B.; Noémie, P.; Brigitte, E.; Géraldine, P. Supercritical fluid methods: An alternative to conventional methods to prepare liposomes. Chem. Eng. J. 2020, 383, 123106. [Google Scholar] [CrossRef]
- Giordani, S.; Marassi, V.; Zattoni, A.; Roda, B.; Reschiglian, P. Liposomes characterization for market approval as pharmaceutical products: Analytical methods, guidelines and standardized protocols. J. Pharm. Biomed. Anal. 2023, 236, 115751. [Google Scholar] [CrossRef]
- FDA. Drugs@FDA: FDA-Approved Drugs; FDA: White Oak, MD, USA, 2024. [Google Scholar]
- Large, D.E.; Abdelmessih, R.G.; Fink, E.A.; Auguste, D.T. Liposome composition in drug delivery design, synthesis, characterization, and clinical application. Adv. Drug Deliv. Rev. 2021, 176, 113851. [Google Scholar] [CrossRef]
- Akbarzadeh, A.; Rezaei-Sadabady, R.; Davaran, S.; Joo, S.W.; Zarghami, N.; Hanifehpour, Y.; Samiei, M.; Kouhi, M.; Nejati-Koshki, K. Liposome: Classification, preparation, and applications. Nanoscale Res. Lett. 2013, 8, 102. [Google Scholar] [CrossRef]
- Nsairat, H.; Khater, D.; Sayed, U.; Odeh, F.; Al Bawab, A.; Alshaer, W. Liposomes: Structure, composition, types, and clinical applications. Heliyon 2022, 8, e09394. [Google Scholar] [CrossRef]
- Jahn, A.; Vreeland, W.N.; DeVoe, D.L.; Locascio, L.E.; Gaitan, M. Microfluidic Directed Formation of Liposomes of Controlled Size. Langmuir 2007, 23, 6289–6293. [Google Scholar] [CrossRef]
- Mortazavi, S.M.; Mohammadabadi, M.R.; Khosravi-Darani, K.; Mozafari, M.R. Preparation of liposomal gene therapy vectors by a scalable method without using volatile solvents or detergents. J. Biotechnol. 2007, 129, 604–613. [Google Scholar] [CrossRef]
- Alavi, M.; Mozafari, M.R.; Hamblin, M.R.; Hamidi, M.; Hajimolaali, M.; Katouzian, I. Industrial-scale methods for the manufacture of liposomes and nanoliposomes: Pharmaceutical, cosmetic, and nutraceutical aspects. Micro. Nano Bio. Asp. 2022, 1, 26–35. [Google Scholar]
- Briuglia, M.-L.; Rotella, C.; McFarlane, A.; Lamprou, D.A. Influence of cholesterol on liposome stability and on in vitro drug release. Drug Deliv. Transl. Res. 2015, 5, 231–242. [Google Scholar] [CrossRef]
- Nsairat, H.; Ibrahim, A.A.; Jaber, A.M.; Abdelghany, S.; Atwan, R.; Shalan, N.; Abdelnabi, H.; Odeh, F.; El-Tanani, M.; Alshaer, W. Liposome bilayer stability: Emphasis on cholesterol and its alternatives. J. Liposome Res. 2024, 34, 178–202. [Google Scholar] [CrossRef] [PubMed]
- Sun, J.; Zhang, L.; Wang, J.; Feng, Q.; Liu, D.; Yin, Q.; Xu, D.; Wei, Y.; Ding, B.; Shi, X.; et al. Tunable rigidity of (polymeric core)-(lipid shell) nanoparticles for regulated cellular uptake. Adv. Mater. 2015, 27, 1402–1407. [Google Scholar] [CrossRef]
- Dua, J.; Rana, A.; Bhandari, A. Liposome: Methods of preparation and applications. Int. J. Pharm. Stud. Res. 2012, 3, 14–20. [Google Scholar]
- Bnyan, R.; Khan, I.; Ehtezazi, T.; Saleem, I.; Gordon, S.; O’Neill, F.; Roberts, M. Surfactant Effects on Lipid-Based Vesicles Properties. J. Pharm. Sci. 2018, 107, 1237–1246. [Google Scholar] [CrossRef]
- Torchilin, V.P. Recent advances with liposomes as pharmaceutical carriers. Nat. Rev. Drug Discov. 2005, 4, 145–160. [Google Scholar] [CrossRef] [PubMed]
- Rafiyath, S.M.; Rasul, M.; Lee, B.; Wei, G.; Lamba, G.; Liu, D. Comparison of safety and toxicity of liposomal doxorubicin vs. conventional anthracyclines: A meta-analysis. Exp. Hematol. Oncol. 2012, 1, 10. [Google Scholar] [CrossRef]
- O’Brien, M.E.R.; Wigler, N.W.; Inbar, M.; Rosso, R.; Grischke, E.; Santoro, A.; Catane, R.; Kieback, D.G.; Tomczak, P.; Ackland, S.P.; et al. Reduced cardiotoxicity and comparable efficacy in a phase IIItrial of pegylated liposomal doxorubicin HCl(CAELYX™/Doxil®) versus conventional doxorubicin forfirst-line treatment of metastatic breast cancer. Ann. Oncol. 2004, 15, 440–449. [Google Scholar] [CrossRef]
- Najahi-Missaoui, W.; Arnold, R.D.; Cummings, B.S. Safe Nanoparticles: Are We There Yet? Int. J. Mol. Sci. 2021, 22, 385. [Google Scholar] [CrossRef] [PubMed]
- Johnston, M.J.W.; Semple, S.C.; Klimuk, S.K.; Ansell, S.; Maurer, N.; Cullis, P.R. Characterization of the drug retention and pharmacokinetic properties of liposomal nanoparticles containing dihydrosphingomyelin. Biochim. Et Biophys. Acta (BBA)—Biomembr. 2007, 1768, 1121–1127. [Google Scholar] [CrossRef] [PubMed]
- Fulton, M.D.; Najahi-Missaoui, W. Liposomes in Cancer Therapy: How Did We Start and Where Are We Now. Int. J. Mol. Sci. 2023, 24, 6615. [Google Scholar] [CrossRef] [PubMed]
- Al-Azayzih, A.; Missaoui, W.N.; Cummings, B.S.; Somanath, P.R. Liposome-mediated delivery of the p21 activated kinase-1 (PAK-1) inhibitor IPA-3 limits prostate tumor growth in vivo. Nanomed. Nanotechnol. Biol. Med. 2016, 12, 1231–1239. [Google Scholar] [CrossRef] [PubMed]
- Konerding, M.A.; Fait, E.; Gaumann, A. 3D microvascular architecture of pre-cancerous lesions and invasive carcinomas of the colon. Br. J. Cancer 2001, 84, 1354–1362. [Google Scholar] [CrossRef] [PubMed]
- Leu, A.J.; Berk, D.A.; Lymboussaki, A.; Alitalo, K.; Jain, R.K. Absence of Functional Lymphatics within a Murine Sarcoma: A Molecular and Functional Evaluation. Cancer Res. 2000, 60, 4324–4327. [Google Scholar]
- Brown, J.M. Tumor Hypoxia in Cancer Therapy. In Methods in Enzymology; Academic Press: Cambridge, MA, USA, 2007; pp. 295–321. [Google Scholar]
- Vaupel, P.; Fortmeyer, H.P.; Runkel, S.; Kallinowski, F. Blood Flow, Oxygen Consumption, and Tissue Oxygenation of Human Breast Cancer Xenografts in Nude Rats. Cancer Res. 1987, 47, 3496–3503. [Google Scholar]
- Koch, S.; Mayer, F.; Honecker, F.; Schittenhelm, M.; Bokemeyer, C. Efficacy of cytotoxic agents used in the treatment of testicular germ cell tumours under normoxic and hypoxic conditions in vitro. Br. J. Cancer 2003, 89, 2133–2139. [Google Scholar] [CrossRef]
- Wu, J. The Enhanced Permeability and Retention (EPR) Effect: The Significance of the Concept and Methods to Enhance Its Application. J. Pers. Med. 2021, 11, 771. [Google Scholar] [CrossRef]
- Tahara, Y.; Yoshikawa, T.; Sato, H.; Mori, Y.; Zahangir, M.H.; Kishimura, A.; Mori, T.; Katamaya, Y. Encapsulation of a nitric oxide donor into a liposome to boost the enhanced permeation and retention (EPR) effect. MedChemComm 2017, 8, 415–421. [Google Scholar] [CrossRef]
- Mitsunaga, M.; Ogawa, M.; Kosaka, N.; Rosenblum, L.T.; Choyke, P.L.; Kobayashi, H. Cancer cell–selective in vivo near infrared photoimmunotherapy targeting specific membrane molecules. Nat. Med. 2011, 17, 1685–1691. [Google Scholar] [CrossRef] [PubMed]
- Sano, K.; Nakajima, T.; Choyke, P.L.; Kobayashi, H. Markedly Enhanced Permeability and Retention Effects Induced by Photo-immunotherapy of Tumors. ACS Nano 2013, 7, 717–724. [Google Scholar] [CrossRef] [PubMed]
- Cattel, L.; Ceruti, M.; Dosio, F. From Conventional to Stealth Liposomes: A New Frontier in Cancer Chemotherapy. J. Chemother. 2004, 16 (Suppl. S4), 94–97. [Google Scholar] [CrossRef]
- Juliano, R.L.; Stamp, D. The effect of particle size and charge on the clearance rates of liposomes and liposome encapsulated drugs. Biochem. Biophys. Res. Commun. 1975, 63, 651–658. [Google Scholar] [CrossRef] [PubMed]
- Gabizon, A.A. Stealth Liposomes and Tumor Targeting: One Step Further in the Quest for the Magic Bullet. Clin. Cancer Res. 2001, 7, 223–225. [Google Scholar] [PubMed]
- Yang, T.; Cui, F.-D.; Choi, M.-K.; Cho, J.-W.; Chung, S.-J.; Shim, C.-K.; Kim, D.-D. Enhanced solubility and stability of PEGylated liposomal paclitaxel: In vitro and in vivo evaluation. Int. J. Pharm. 2007, 338, 317–326. [Google Scholar] [CrossRef] [PubMed]
- Ghaferi, M.; Asadollahzadeh, M.J.; Akbarzadeh, A.; Shahmabadi, H.E.; Alavi, S.E. Enhanced Efficacy of PEGylated Liposomal Cisplatin: In Vitro and In Vivo Evaluation. Int. J. Mol. Sci. 2020, 21, 559. [Google Scholar] [CrossRef]
- Singh, A.; Neupane, Y.R.; Shafi, S.; Mangla, B.; Kohli, K. PEGylated liposomes as an emerging therapeutic platform for oral nanomedicine in cancer therapy: In vitro and in vivo assessment. J. Mol. Liq. 2020, 303, 112649. [Google Scholar] [CrossRef]
- Tang, L.; Winkeljann, B.; Feng, S.; Song, J.; Liu, Y. Recent advances in superlubricity of liposomes for biomedical applications. Colloids Surf. B Biointerfaces 2022, 218, 112764. [Google Scholar] [CrossRef]
- Allen, T.M.; Hansen, C.; Martin, F.; Redemann, C.; Yau-Young, A. Liposomes containing synthetic lipid derivatives of poly(ethylene glycol) show prolonged circulation half-lives in vivo. Biochim. Biophys. Acta (BBA)—Biomembr. 1991, 1066, 29–36. [Google Scholar] [CrossRef]
- Nag, O.K.; Awasthi, V. Surface Engineering of Liposomes for Stealth Behavior. Pharmaceutics 2013, 5, 542–569. [Google Scholar] [CrossRef] [PubMed]
- Nosova, A.S.; Koloskova, O.O.; Nikonova, A.A.; Simonova, V.A.; Smirnov, V.V.; Kudlay, D.; Khaitov, M.R. Diversity of PEGylation methods of liposomes and their influence on RNA delivery. MedChemComm 2019, 10, 369–377. [Google Scholar] [CrossRef] [PubMed]
- Uster, P.S.; Allen, T.M.; Daniel, B.E.; Mendez, C.J.; Newman, M.S.; Zhu, G.Z. Insertion of poly(ethylene glycol) derivatized phospholipid into pre-formed liposomes results in prolonged in vivo circulation time. FEBS Lett. 1996, 386, 243–246. [Google Scholar] [CrossRef] [PubMed]
- Awasthi, V.; Garcia, D.; Klipper, R.; Goins, B.A.; Phillips, W.T. Neutral and anionic liposome-encapsulated hemoglobin: Effect of postinserted poly (ethylene glycol)-distearoylphosphatidylethanolamine on distribution and circulation kinetics. J. Pharmacol. Exp. Ther. 2004, 309, 241–248. [Google Scholar] [CrossRef] [PubMed]
- Guo, C.; Yuan, H.; Wang, Y.; Feng, Y.; Zhang, Y.; Yin, T.; He, H.; Gou, J.; Tang, X. The interplay between PEGylated nanoparticles and blood immune system. Adv. Drug Deliv. Rev. 2023, 200, 115044. [Google Scholar] [CrossRef] [PubMed]
- Abu Lila, A.S.; Kiwada, H.; Ishida, T. The accelerated blood clearance (ABC) phenomenon: Clinical challenge and approaches to manage. J. Control. Release 2013, 172, 38–47. [Google Scholar] [CrossRef] [PubMed]
- Ishida, T.; Ichikawa, T.; Ichihara, M.; Sadzuka, Y.; Kiwada, H. Effect of the physicochemical properties of initially injected liposomes on the clearance of subsequently injected PEGylated liposomes in mice. J. Control. Release 2004, 95, 403–412. [Google Scholar] [CrossRef] [PubMed]
- Dams, E.T.; Laverman, P.; Oyen, W.J.; Storm, G.; Scherphof, G.L.; van Der Meer, J.W.; Corstens, F.H.; Boerman, O.C. Accelerated blood clearance and altered biodistribution of repeated injections of sterically stabilized liposomes. J. Pharmacol. Exp. Ther. 2000, 292, 1071–1079. [Google Scholar] [PubMed]
- Ishida, T.; Ichihara, M.; Wang, X.; Yamamoto, K.; Kimura, J.; Majima, E.; Kiwada, H. Injection of PEGylated liposomes in rats elicits PEG-specific IgM, which is responsible for rapid elimination of a second dose of PEGylated liposomes. J. Control. Release 2006, 112, 15–25. [Google Scholar] [CrossRef] [PubMed]
- Wang, F.; Ye, X.; Wu, Y.; Wang, H.; Sheng, C.; Peng, D.; Chen, W. Time Interval of Two Injections and First-Dose Dependent of Accelerated Blood Clearance Phenomenon Induced by PEGylated Liposomal Gambogenic Acid: The Contribution of PEG-Specific IgM. J. Pharm. Sci. 2019, 108, 641–651. [Google Scholar] [CrossRef]
- Ishida, T.; Masuda, K.; Ichikawa, T.; Ichihara, M.; Irimura, K.; Kiwada, H. Accelerated clearance of a second injection of PEGylated liposomes in mice. Int. J. Pharm. 2003, 255, 167–174. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.Y.; Ishida, T.; Ichihara, M.; Kiwada, H. Influence of the physicochemical properties of liposomes on the accelerated blood clearance phenomenon in rats. J. Control. Release 2005, 104, 91–102. [Google Scholar] [CrossRef] [PubMed]
- Charrois, G.J.; Allen, T.M. Multiple injections of pegylated liposomal Doxorubicin: Pharmacokinetics and therapeutic activity. J. Pharmacol. Exp. Ther. 2003, 306, 1058–1067. [Google Scholar] [CrossRef] [PubMed]
- Ishida, T.; Kiwada, H. Accelerated blood clearance (ABC) phenomenon upon repeated injection of PEGylated liposomes. Int. J. Pharm. 2008, 354, 56–62. [Google Scholar] [CrossRef] [PubMed]
- Suzuki, T.; Ichihara, M.; Hyodo, K.; Yamamoto, E.; Ishida, T.; Kiwada, H.; Kikuchi, H.; Ishihara, H. Influence of dose and animal species on accelerated blood clearance of PEGylated liposomal doxorubicin. Int. J. Pharm. 2014, 476, 205–212. [Google Scholar] [CrossRef] [PubMed]
- Vollmers, H.P.; Brändlein, S. Natural antibodies and cancer. New Biotechnol. 2009, 25, 294–298. [Google Scholar] [CrossRef] [PubMed]
- Stanfield, R.L.; Wilson, I.A. Antibody Structure. In Antibodies for Infectious Diseases; Wiley: Hoboken, NJ, USA, 2015; pp. 49–62. [Google Scholar]
- Van Wagoner, C.M.; Rivera-Escalera, F.; Jaimes-Delgadillo, N.C.; Chu, C.C.; Zent, C.S.; Elliot, M.R. Antibody-mediated phagocytosis in cancer immunotherapy. Immunol. Rev. 2023, 319, 128–141. [Google Scholar] [CrossRef] [PubMed]
- Zahavi, D.; Weiner, L. Monoclonal Antibodies in Cancer Therapy. Antibodies 2020, 9, 34. [Google Scholar] [CrossRef] [PubMed]
- Mullard, A. 2023 FDA approvals. Nat. Rev. Drug Discov. 2024, 23, 88–95. [Google Scholar] [CrossRef]
- ElBayoumi, T.A.; Torchilin, V.P. Tumor-Specific Anti-Nucleosome Antibody Improves Therapeutic Efficacy of Doxorubicin-Loaded Long-Circulating Liposomes against Primary and Metastatic Tumor in Mice. Mol. Pharm. 2009, 6, 246–254. [Google Scholar] [CrossRef]
- Arabi, L.; Badiee, A.; Mosaffa, F.; Jaafari, M.R. Targeting CD44 expressing cancer cells with anti-CD44 monoclonal antibody improves cellular uptake and antitumor efficacy of liposomal doxorubicin. J. Control. Release 2015, 220, 275–286. [Google Scholar] [CrossRef] [PubMed]
- Lukyanov, A.N.; Elbayoumi, T.A.; Chakilam, A.R.; Torchilin, V.P. Tumor-targeted liposomes: Doxorubicin-loaded long-circulating liposomes modified with anti-cancer antibody. J. Control. Release 2004, 100, 135–144. [Google Scholar] [CrossRef] [PubMed]
- Kirpotin, D.B.; Drummond, D.C.; Shao, Y.; Shalaby, M.R.; Hong, K.; Nielsen, U.B.; Marks, J.D.; Benz, C.C.; Park, J.W. Antibody Targeting of Long-Circulating Lipidic Nanoparticles Does Not Increase Tumor Localization but Does Increase Internalization in Animal Models. Cancer Res. 2006, 66, 6732–6740. [Google Scholar] [CrossRef] [PubMed]
- Moase, E.H.; Qi, W.; Ishida, T.; Gabos, Z.; Longenecker, B.M.; Zimmerman, G.L.; Ding, L.; Krantz, M.; Allen, T.M. Anti-MUC-1 immunoliposomal doxorubicin in the treatment of murine models of metastatic breast cancer. Biochim. Biophys. Acta (BBA)—Biomembr. 2001, 1510, 43–55. [Google Scholar] [CrossRef]
- Vingerhoeds, M.H.; Steerenberg, P.A.; Hendriks, J.J.; Dekker, L.C.; Van Hoesel, Q.G.; Crommelin, D.J.; Storm, G. Immunoliposome-mediated targeting of doxorubicin to human ovarian carcinoma in vitro and in vivo. Br. J. Cancer 1996, 74, 1023–1029. [Google Scholar] [CrossRef] [PubMed]
- Sawant, R.R.; Torchilin, V.P. Challenges in Development of Targeted Liposomal Therapeutics. AAPS J. 2012, 14, 303–315. [Google Scholar] [CrossRef] [PubMed]
- Eloy, J.O.; Petrilli, R.; Trevizan, L.N.F.; Chorilli, M. Immunoliposomes: A review on functionalization strategies and targets for drug delivery. Colloids Surf. B Biointerfaces 2017, 159, 454–467. [Google Scholar] [CrossRef] [PubMed]
- Fidan, Y.; Muçaj, S.; Timur, S.S.; Gürsoy, R.N. Recent advances in liposome-based targeted cancer therapy. J. Liposome Res. 2023. online ahead of print. [Google Scholar] [CrossRef]
- Ohradanova-Repic, A.; Nogueria, E.; Hartl, I.; Gomes, A.C.; Preto, A.; Steinhuber, E.; Mühlgrabner, V.; Repic, M.; Kuttke, M.; Zwirzitz, A.; et al. Fab antibody fragment-functionalized liposomes for specific targeting of antigen-positive cells. Nanomed. Nanotechnol. Biol. Med. 2018, 14, 123–130. [Google Scholar] [CrossRef] [PubMed]
- Zalba, S.; Contreras, A.M.; Haeri, A.; Ten Hagen, T.L.M.; Navarro, I.; Konig, G.; Garrido, M.J. Cetuximab-oxaliplatin-liposomes for epidermal growth factor receptor targeted chemotherapy of colorectal cancer. J. Control. Release 2015, 210, 26–38. [Google Scholar] [CrossRef]
- Maruyama, K.; Takahashi, N.; Tagawa, T.; Nagaike, K.; Iwatsuru, M. Immunoliposomes bearing polyethyleneglycol-coupled Fab′ fragment show prolonged circulation time and high extravasation into targeted solid tumors in vivo. FEBS Lett. 1997, 413, 177–180. [Google Scholar] [CrossRef]
- Steichen, S.D.; Caldorera-Moore, M.; Peppas, N.A. A review of current nanoparticle and targeting moieties for the delivery of cancer therapeutics. Eur. J. Pharm. Sci. 2013, 48, 416–427. [Google Scholar] [CrossRef]
- Riaz, M.K.; Riaz, M.A.; Zhang, X.; Lin, C.; Wong, K.H.; Chen, X.; Zhang, G.; Lu, A.; Yang, Z. Surface Functionalization and Targeting Strategies of Liposomes in Solid Tumor Therapy: A Review. Int. J. Mol. Sci. 2018, 19, 195. [Google Scholar] [CrossRef]
- Feng, L.; Mumper, R.J. A critical review of lipid-based nanoparticles for taxane delivery. Cancer Lett. 2013, 334, 157–175. [Google Scholar] [CrossRef]
- Nevola, L.; Giralt, E. Modulating protein–protein interactions: The potential of peptides. Chem. Commun. 2015, 51, 3302–3315. [Google Scholar] [CrossRef] [PubMed]
- Erckes, V.; Steuer, C. A story of peptides, lipophilicity and chromatography—Back and forth in time. RSC Med. Chem. 2022, 13, 676–687. [Google Scholar] [CrossRef]
- Wang, L.; Wang, N.; Zhang, W.; Cheng, X.; Yan, Z.; Shao, G.; Wang, X.; Wang, R.; Fu, C. Therapeutic peptides: Current applications and future directions. Signal Transduct. Target. Ther. 2022, 7, 48. [Google Scholar] [CrossRef] [PubMed]
- Baig, M.H.; Ahmad, K.; Saeed, M.; Alharbi, A.M.; Barreto, G.E.; Ashraf, G.M.; Choi, I. Peptide based therapeutics and their use for the treatment of neurodegenerative and other diseases. Biomed. Pharmacother. 2018, 103, 574–581. [Google Scholar] [CrossRef]
- Sawyer, W.; Manning, M. Synthetic analogs of oxytocin and the vasopressins. Annu. Rev. Pharmacol. 1973, 13, 5–17. [Google Scholar] [CrossRef]
- Muttenthaler, M.; King, G.F.; Adams, D.J.; Alewood, P.F. Trends in peptide drug discovery. Nat. Rev. Drug Discov. 2021, 20, 309–325. [Google Scholar] [CrossRef] [PubMed]
- Eliopoulos, G.M.; Willey, S.; Reiszner, E.; Spitzer, P.G.; Caputo, G.; Moellering, R.C., Jr. In vitro and in vivo activity of LY 146032, a new cyclic lipopeptide antibiotic. Antimicrob. Agents Chemother. 1986, 30, 532–535. [Google Scholar] [CrossRef]
- Chen, A.; Zervos, M.; Vazquez, J.A. Dalbavancin: A novel antimicrobial. Int. J. Clin. Pract. 2007, 61, 853–863. [Google Scholar] [CrossRef]
- Rosenthal, S.; Decano, A.G.; Bandali, A.; Lai, D.; Malat, G.E.; Bias, T.E. Oritavancin (Orbactiv): A new-generation lipoglycopeptide for the treatment of acute bacterial skin and skin structure infections. Pharm. Ther. 2018, 43, 143. [Google Scholar]
- Okuyama, R.; Aruga, A.; Hatori, T.; Takeda, K.; Yamamoto, M. Immunological responses to a multi-peptide vaccine targeting cancer-testis antigens and VEGFRs in advanced pancreatic cancer patients. Oncoimmunology 2013, 2, e27010. [Google Scholar] [CrossRef]
- Abdelmageed, M.I.; Abdelmoneim, A.H.; Mustafa, M.I.; Elfadol, N.M.; Murshed, N.S.; Shantier, S.W.; Makhawi, A.M. Design of a Multiepitope-Based Peptide Vaccine against the E Protein of Human COVID-19: An Immunoinformatics Approach. BioMed Res. Int. 2020, 2020, 2683286. [Google Scholar] [CrossRef]
- Kaumaya, P.T. B-cell epitope peptide cancer vaccines: A new paradigm for combination immunotherapies with novel checkpoint peptide vaccine. Future Oncol. 2020, 16, 1767–1791. [Google Scholar] [CrossRef] [PubMed]
- Wang, S.-C.; Hung, M.-C. HER2 overexpression and cancer targeting. Semin. Oncol. 2001, 28, 115–124. [Google Scholar] [CrossRef]
- Najahi-Missaoui, W.; Quach, N.D.; Somanath, P.R.; Cummings, B.S. Liposomes Targeting P21 Activated Kinase-1 (PAK-1) and Selective for Secretory Phospholipase A2 (sPLA2) Decrease Cell Viability and Induce Apoptosis in Metastatic Triple-Negative Breast Cancer Cells. Int. J. Mol. Sci. 2020, 21, 9396. [Google Scholar] [CrossRef] [PubMed]
- Curnis, F.; Arrigoni, G.; Sacchi, A.; Fischetti, L.; Arap, W.; Pasqualini, R.; Corti, A. Differential Binding of Drugs Containing the NGR Motif to CD13 Isoforms in Tumor Vessels, Epithelia, and Myeloid Cells1. Cancer Res. 2002, 62, 867–874. [Google Scholar]
- Wang, F.; Chen, L.; Zhang, R.; Chen, Z.; Zhu, L. RGD peptide conjugated liposomal drug delivery system for enhance therapeutic efficacy in treating bone metastasis from prostate cancer. J. Control. Release 2014, 196, 222–233. [Google Scholar] [CrossRef]
- Vakhshiteh, F.; Khabazian, E.; Atyabi, F.; Ostad, S.N.; Madjd, Z.; Dinarvand, R. Peptide-conjugated liposomes for targeted miR-34a delivery to suppress breast cancer and cancer stem-like population. J. Drug Deliv. Sci. Technol. 2020, 57, 101687. [Google Scholar] [CrossRef]
- Liu, X.; Li, Z.; Wang, X.; Chen, Y.; Wu, F.; Men, K.; Xu, T.; Luo, Y.; Yang, L. Novel antimicrobial peptide–modified azithromycin-loaded liposomes against methicillin-resistant Staphylococcus aureus. Int. J. Nanomed. 2016, 11, 6781–6794. [Google Scholar] [CrossRef]
- Sonju, J.J.; Dahal, A.; Singh, S.S.; Jois, S.D. Peptide-functionalized liposomes as therapeutic and diagnostic tools for cancer treatment. J. Control. Release 2021, 329, 624–644. [Google Scholar] [CrossRef]
- Belfiore, L.; Saunders, D.N.; Ranson, M.; Thurecht, K.J.; Storm, G.; Vine, K.L. Towards clinical translation of ligand-functionalized liposomes in targeted cancer therapy: Challenges and opportunities. J. Control. Release 2018, 277, 1–13. [Google Scholar] [CrossRef]
- Guimarães, D.; Cavaco-Paulo, A.; Nogueira, E. Design of liposomes as drug delivery system for therapeutic applications. Int. J. Pharm. 2021, 601, 120571. [Google Scholar] [CrossRef] [PubMed]
- Choi, S.-W.; Mason, J.B. Folate and carcinogenesis: An integrated scheme. J. Nutr. 2000, 130, 129–132. [Google Scholar] [CrossRef]
- Kim, Y.I. Folate, colorectal carcinogenesis, and DNA methylation: Lessons from animal studies. Environ. Mol. Mutagen. 2004, 44, 10–25. [Google Scholar] [CrossRef]
- Giovannucci, E. Epidemiologic studies of folate and colorectal neoplasia: A review. J. Nutr. 2002, 132, 2350S–2355S. [Google Scholar] [CrossRef] [PubMed]
- Kelemen, L.E. The role of folate receptor α in cancer development, progression and treatment: Cause, consequence or innocent bystander? Int. J. Cancer 2006, 119, 243–250. [Google Scholar] [CrossRef] [PubMed]
- Vergote, I.B.; Marth, C.; Coleman, R.L. Role of the folate receptor in ovarian cancer treatment: Evidence, mechanism, and clinical implications. Cancer Metastasis Rev. 2015, 34, 41–52. [Google Scholar] [CrossRef]
- Kabilova, T.O.; Shmendel, E.V.; Gadkikh, D.V.; Chernolovskaya, E.L.; Markov, O.V.; Morozova, N.G.; Maslov, M.A.; Zenkova, M.A. Targeted delivery of nucleic acids into xenograft tumors mediated by novel folate-equipped liposomes. Eur. J. Pharm. Biopharm. 2018, 123, 59–70. [Google Scholar] [CrossRef]
- Patil, Y.; Amitay, Y.; Ohana, P.; Shmeeda, H.; Gabizon, A. Targeting of pegylated liposomal mitomycin-C prodrug to the folate receptor of cancer cells: Intracellular activation and enhanced cytotoxicity. J. Control. Release 2016, 225, 87–95. [Google Scholar] [CrossRef] [PubMed]
- Wang, L.; Evans, J.C.; Ahmed, L.; Allen, C. Folate receptor targeted nanoparticles containing niraparib and doxorubicin as a potential candidate for the treatment of high grade serous ovarian cancer. Sci. Rep. 2023, 13, 3226. [Google Scholar] [CrossRef] [PubMed]
- Leamon, C.P.; Cooper, S.R.; Hardee, G.E. Folate-Liposome-Mediated Antisense Oligodeoxynucleotide Targeting to Cancer Cells: Evaluation in Vitro and in Vivo. Bioconjugate Chem. 2003, 14, 738–747. [Google Scholar] [CrossRef] [PubMed]
- Gabizon, A.; Horowitz, A.T.; Goren, D.; Tzemach, D.; Shmeeda, H.; Zalipsky, S. In Vivo Fate of Folate-Targeted Polyethylene-Glycol Liposomes in Tumor-Bearing Mice. Clin. Cancer Res. 2003, 9, 6551–6559. [Google Scholar] [PubMed]
- Xia, W.; Low, P.S. Folate-Targeted Therapies for Cancer. J. Med. Chem. 2010, 53, 6811–6824. [Google Scholar] [CrossRef]
- Park, Y.I.; Kwon, S.-H.; Lee, G.; Motoyama, K.; Kim, M.W.; Lin, M.; Niidome, T.; Choi, J.H.; Lee, R. pH-sensitive multi-drug liposomes targeting folate receptor β for efficient treatment of non-small cell lung cancer. J. Control. Release 2021, 330, 1–14. [Google Scholar] [CrossRef] [PubMed]
- Tombelli, S.; Minunni, M.; Mascini, M. Analytical applications of aptamers. Biosens. Bioelectron. 2005, 20, 2424–2434. [Google Scholar] [CrossRef]
- Bunka, D.H.J.; Platonova, O.; Stockley, P.G. Development of aptamer therapeutics. Curr. Opin. Pharmacol. 2010, 10, 557–562. [Google Scholar] [CrossRef] [PubMed]
- Mullard, A. FDA approves second RNA aptamer. Nat. Rev. Drug Discov. 2023, 22, 774. [Google Scholar] [CrossRef]
- Keefe, A.D.; Pai, S.; Ellington, A. Aptamers as therapeutics. Nat. Rev. Drug Discov. 2010, 9, 537–550. [Google Scholar] [CrossRef]
- Wu, Y.X.; Kwon, Y.J. Aptamers: The “evolution” of SELEX. Methods 2016, 106, 21–28. [Google Scholar] [CrossRef] [PubMed]
- Agnello, L.; Camorani, S.; Fedele, M.; Cerchia, L. Aptamers and antibodies: Rivals or allies in cancer targeted therapy? Explor. Target. Antitumor Ther. 2021, 2, 107–121. [Google Scholar] [CrossRef] [PubMed]
- Wong, K.-Y.; Wong, M.-S.; Liu, J. Aptamer-functionalized liposomes for drug delivery. Biomed. J. 2023. [Google Scholar] [CrossRef]
- Mashreghi, M.; Zamani, P.; Karimi, M.; Mehrabian, A.; Arabsalmani, M.; Zarqi, J.; Moosavian, S.A.; Jaafari, M.R. Anti-epithelial cell adhesion molecule RNA aptamer-conjugated liposomal doxorubicin as an efficient targeted therapy in mice bearing colon carcinoma tumor model. Biotechnol. Prog. 2021, 37, e3116. [Google Scholar] [CrossRef] [PubMed]
- Shakib, Z.; Mahmoudi, A.; Moosavian, S.A.; Malaekeh-Nikouei, B. PEGylated solid lipid nanoparticles functionalized by aptamer for targeted delivery of docetaxel in mice bearing C26 tumor. Drug Dev. Ind. Pharm. 2022, 48, 69–78. [Google Scholar] [CrossRef] [PubMed]
- Kim, M.; Lee, J.S.; Kim, W.; Lee, J.H.; Jun, B.H.; Kim, K.S.; Kim, D.-E. Aptamer-conjugated nano-liposome for immunogenic chemotherapy with reversal of immunosuppression. J. Control. Release 2022, 348, 893–910. [Google Scholar] [CrossRef] [PubMed]
- Ding, Z.; Wang, D.; Shi, W.; Yang, X.; Duan, S.; Mo, F.; Hou, X.; Liu, A.; Lu, X. In vivo Targeting of Liver Cancer with Tissue- and Nuclei-Specific Mesoporous Silica Nanoparticle-Based Nanocarriers in mice. Int. J. Nanomed. 2020, 15, 8383–8400. [Google Scholar] [CrossRef]
- Wong, K.-Y.; Liu, Y.; Zhou, L.; Wong, M.-S.; Liu, J. Mucin-targeting-aptamer functionalized liposomes for delivery of cyclosporin A for dry eye diseases. J. Mater. Chem. B 2023, 11, 4684–4694. [Google Scholar] [CrossRef] [PubMed]
- Sun, S.; Liu, H.; Hu, Y.; Wang, Y.; Zhao, M.; Yuan, Y.; Han, Y.; Jing, Y.; Cui, J.; Ren, X.; et al. Selection and identification of a novel ssDNA aptamer targeting human skeletal muscle. Bioact. Mater. 2023, 20, 166–178. [Google Scholar] [CrossRef] [PubMed]
- He, Y.; Huang, Y.; Xu, H.; Yang, X.; Liu, N.; Xu, Y.; Ma, R.; Zhai, J.; Ma, Y.; Guan, S. Aptamer-modified M cell targeting liposomes for oral delivery of macromolecules. Colloids Surf. B Biointerfaces 2023, 222, 113109. [Google Scholar] [CrossRef]
- Bauer, M.; Strom, M.; Hammond, D.S.; Shigdar, S. Anything You Can Do, I Can Do Better: Can Aptamers Replace Antibodies in Clinical Diagnostic Applications? Molecules 2019, 24, 4377. [Google Scholar] [CrossRef] [PubMed]
- Fan, Y.; Chen, C.; Huang, Y.; Zhang, F.; Lin, G. Study of the pH-sensitive mechanism of tumor-targeting liposomes. Colloids Surf. B Biointerfaces 2017, 151, 19–25. [Google Scholar] [CrossRef] [PubMed]
- Abri Aghdam, M.; Bagheri, R.; Mosafer, J.; Baradaran, B.; Hashemzaei, M.; Baghbanzadeh, A.; de la Guardia, M.; Mokhtarzadah, A. Recent advances on thermosensitive and pH-sensitive liposomes employed in controlled release. J. Control. Release 2019, 315, 1–22. [Google Scholar] [CrossRef] [PubMed]
- Alrbyawi, H.; Poudel, I.; Annaji, M.; Boddu, S.H.S.; Arnold, R.D.; Tiwari, A.K.; Babu, R.J. pH-Sensitive Liposomes for Enhanced Cellular Uptake and Cytotoxicity of Daunorubicin in Melanoma (B16-BL6) Cell Lines. Pharmaceutics 2022, 14, 1128. [Google Scholar] [CrossRef] [PubMed]
- Dos Reis, S.B.; de Oliveira Silva, J.; Garcia-Fossa, F.; Leite, E.A.; Malachias, A.; Pound-Lana, G.; Mosqueira, V.C.F.; Oliveira, M.C.; de Barros, A.L.B.; de Jesus, M.B. Mechanistic insights into the intracellular release of doxorubicin from pH-sensitive liposomes. Biomed. Pharmacother. 2021, 134, 110952. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.; Liu, H.; Li, N.; Li, J.; Wang, M.; Ren, X. A (Traditional Chinese Medicine) TCM-Inspired Doxorubicin Resistance Reversing Strategy: Preparation, Characterization, and Application of a Co-loaded pH-Sensitive Liposome. AAPS PharmSciTech 2023, 24, 181. [Google Scholar] [CrossRef] [PubMed]
- Men, W.; Zhu, P.; Dong, S.; Liu, W.; Zhou, K.; Bai, Y.; Liu, X.; Gong, S.; Zhang, S. Layer-by-layer pH-sensitive nanoparticles for drug delivery and controlled release with improved therapeutic efficacy in vivo. Drug Deliv. 2020, 27, 180–190. [Google Scholar] [CrossRef]
- Gupta, S.; De Mel, J.U.; Schneider, G.J. Dynamics of liposomes in the fluid phase. Curr. Opin. Colloid. Interface Sci. 2019, 42, 121–136. [Google Scholar] [CrossRef]
- Yatvin, M.B.; Weinstein, J.N.; Dennis, W.H.; Blumenthal, R. Design of Liposomes for Enhanced Local Release of Drugs by Hyperthermia. Science 1978, 202, 1290–1293. [Google Scholar] [CrossRef]
- Ta, T.; Porter, T.M. Thermosensitive liposomes for localized delivery and triggered release of chemotherapy. J. Control. Release 2013, 169, 112–125. [Google Scholar] [CrossRef]
- Winter, N.D.; Murphy, R.K.J.; O’Halloran, T.V.; Schatz, G.C. Development and modeling of arsenic-trioxide–loaded thermosensitive liposomes for anticancer drug delivery. J. Liposome Res. 2011, 21, 106–115. [Google Scholar] [CrossRef]
- Needham, D.; Anyarambhatla, G.; Kong, G.; Dewhirst, M.W. A New Temperature-sensitive Liposome for Use with Mild Hyperthermia: Characterization and Testing in a Human Tumor Xenograft Model. Cancer Res. 2000, 60, 1197–1201. [Google Scholar] [PubMed]
- Kong, G.; Anyarambhatla, G.; Petros, W.P.; Braun, R.D.; Colvin, O.M.; Needham, D.; Dewhirst, M.W. Efficacy of liposomes and hyperthermia in a human tumor xenograft model: Importance of triggered drug release. Cancer Res. 2000, 60, 6950–6957. [Google Scholar]
- Ta, T.; Convertine, A.J.; Reyes, C.R.; Stayton, P.S.; Porter, T.M. Thermosensitive Liposomes Modified with Poly(N-isopropylacrylamide-co-propylacrylic acid) Copolymers for Triggered Release of Doxorubicin. Biomacromolecules 2010, 11, 1915–1920. [Google Scholar] [CrossRef] [PubMed]
- Xi, L.; Li, C.; Wang, Y.; Gong, Y.; Su, F.; Li, S. Novel Thermosensitive Polymer-Modified Liposomes as Nano-Carrier of Hydrophobic Antitumor Drugs. J. Pharm. Sci. 2020, 109, 2544–2552. [Google Scholar] [CrossRef] [PubMed]
- Mazzotta, E.; Tavano, L.; Muzzalupo, R. Thermo-sensitive vesicles in controlled drug delivery for chemotherapy. Pharmaceutics 2018, 10, 150. [Google Scholar] [CrossRef]
- Jin, Y.; Liang, X.; An, Y.; Dai, Z. Microwave-triggered smart drug release from liposomes co-encapsulating doxorubicin and salt for local combined hyperthermia and chemotherapy of cancer. Bioconjugate Chem. 2016, 27, 2931–2942. [Google Scholar] [CrossRef]
- Chaudhry, M.; Lyon, P.; Coussios, C.; Carlisle, R. Thermosensitive liposomes: A promising step toward localised chemotherapy. Expert. Opin. Drug Deliv. 2022, 19, 899–912. [Google Scholar] [CrossRef]
- Astani, S.A.; Brown, M.L.; Steusloff, K. Comparison of procedure costs of various percutaneous tumor ablation modalities. Radiol. Manag. 2014, 36, 12–17. [Google Scholar]
- Swenson, C.E.; Haemmerich, D.; Maul, D.H.; Knox, B.; Ehrhart, N.; Reed, R.A. Increased duration of heating boosts local drug deposition during radiofrequency ablation in combination with thermally sensitive liposomes (ThermoDox) in a porcine model. PLoS ONE 2015, 10, e0139752. [Google Scholar] [CrossRef]
- Wang, M.; Liu, Y.; Zhang, X.; Luo, L.; Li, L.; Xing, S.; He, Y.; Cao, W.; Zhu, R.; Gao, D. Gold nanoshell coated thermo-pH dual responsive liposomes for resveratrol delivery and chemo-photothermal synergistic cancer therapy. J. Mater. Chem. B 2017, 5, 2161–2171. [Google Scholar] [CrossRef]
- Liu, Y.; Zhang, X.; Liu, Z.; Wang, L.; Luo, L.; Wang, M.; Wang, Q.; Gao, D. Gold nanoshell-based betulinic acid liposomes for synergistic chemo-photothermal therapy. Nanomed. Nanotechnol. Biol. Med. 2017, 13, 1891–1900. [Google Scholar] [CrossRef]
- Henderson, T.A.; Morries, L.D. Near-infrared photonic energy penetration: Can infrared phototherapy effectively reach the human brain? Neuropsychiatr. Dis. Treat. 2015, 11, 2191–2208. [Google Scholar] [CrossRef] [PubMed]
- Dromi, S.; Frenkel, V.; Luk, A.; Traughber, B.; Angstadt, M.; Bur, M.; Poff, J.; Xie, J.; Libutti, S.K.; Li, K.C.P.; et al. Pulsed-high intensity focused ultrasound and low temperature–sensitive liposomes for enhanced targeted drug delivery and antitumor effect. Clin. Cancer Res. 2007, 13, 2722–2727. [Google Scholar] [CrossRef]
- Sebeke, L.C.; Gómez, J.D.C.; Hiejman, E.; Rademann, P.; Simon, A.C.; Ekdawi, S.; Vlachakis, S.; Toker, D.; Mink, B.L.; Schubert-Quecke, C.; et al. Hyperthermia-induced doxorubicin delivery from thermosensitive liposomes via MR-HIFU in a pig model. J. Control. Release 2022, 343, 798–812. [Google Scholar] [CrossRef]
- Namakshenas, P.; Mojra, A. Efficient drug delivery to hypoxic tumors using thermosensitive liposomes with encapsulated anti-cancer drug under high intensity pulsed ultrasound. Int. J. Mech. Sci. 2023, 237, 107818. [Google Scholar] [CrossRef]
- Pande, S. Liposomes for drug delivery: Review of vesicular composition, factors affecting drug release and drug loading in liposomes. Artif. Cells Nanomed. Biotechnol. 2023, 51, 428–440. [Google Scholar] [CrossRef]
- Sawaftah, N.A.; Paul, V.; Awad, N.; Husseini, G.A. Modeling of Anti-Cancer Drug Release Kinetics From Liposomes and Micelles: A Review. IEEE Trans. NanoBiosci. 2021, 20, 565–576. [Google Scholar] [CrossRef]
- Degobert, G.; Aydin, D. Lyophilization of Nanocapsules: Instability Sources, Formulation and Process Parameters. Pharmaceutics 2021, 13, 1112. [Google Scholar] [CrossRef]
- Lobato, K.B.d.S.; Paese, K.; Forgearini, J.C.; Guterres, S.S.; Jablonski, A.; de Oliveira Rios, A. Characterisation and stability evaluation of bixin nanocapsules. Food Chem. 2013, 141, 3906–3912. [Google Scholar] [CrossRef]
- Fonte, P.; Reis, S.; Sarmento, B. Facts and evidences on the lyophilization of polymeric nanoparticles for drug delivery. J. Control. Release 2016, 225, 75–86. [Google Scholar] [CrossRef] [PubMed]
- Gatto, M.S.; Najahi-Missaoui, W. Lyophilization of Nanoparticles, Does It Really Work? Overview of the Current Status and Challenges. Int. J. Mol. Sci. 2023, 24, 14041. [Google Scholar] [CrossRef] [PubMed]
- Guimarães, D.; Noro, J.; Silva, C.; Cavaco-Paulo, A.; Nogueira, E. Protective Effect of Saccharides on Freeze-Dried Liposomes Encapsulating Drugs. Front. Bioeng. Biotechnol. 2019, 7, 424. [Google Scholar] [CrossRef] [PubMed]
- Crowe, J.H.; Crowe, L.M. Factors affecting the stability of dry liposomes. Biochim. Biophys. Acta 1988, 939, 327–334. [Google Scholar] [CrossRef]
- Zylberberg, C.; Matosevic, S. Pharmaceutical liposomal drug delivery: A review of new delivery systems and a look at the regulatory landscape. Drug Deliv. 2016, 23, 3319–3329. [Google Scholar] [CrossRef]
- Liposome Drug Products: Chemistry, Manufacturing, and Controls; Human Pharmacokinetics and Bioavailability; and Labeling Documentation. Available online: https://www.fda.gov/regulatory-information/search-fda-guidance-documents/liposome-drug-products-chemistry-manufacturing-and-controls-human-pharmacokinetics-and (accessed on 24 March 2024).
- Mashreghi, M.; Zamani, P.; Moosavian, S.A.; Jaafari, M.R. Anti-Epcam aptamer (Syl3c)-functionalized liposome for targeted delivery of doxorubicin: In vitro and in vivo antitumor studies in mice bearing C26 colon carcinoma. Nanoscale Res. Lett. 2020, 15, 101. [Google Scholar] [CrossRef]
- Swami, R.; Kumar, Y.; Chaudhari, D.; Katiyar, S.S.; Kuche, K.; Katare, P.B.; Banerjee, S.K.; Jain, S. pH sensitive liposomes assisted specific and improved breast cancer therapy using co-delivery of SIRT1 shRNA and Docetaxel. Mater. Sci. Eng. C 2021, 120, 111664. [Google Scholar] [CrossRef] [PubMed]
- Xu, H.; Li, Y.; Paxton, J.W.; Wu, Z. Co-Delivery Using pH-Sensitive Liposomes to Pancreatic Cancer Cells: The Effects of Curcumin on Cellular Concentration and Pharmacokinetics of Gemcitabine. Pharm. Res. 2021, 38, 1209–1219. [Google Scholar] [CrossRef]
- de Oliveira Silva, J.; Fernandes, R.S.; Oda, C.M.R.; Ferreira, T.H.; Botelho, A.F.M.; Melo, M.M.; de Miranda, M.C.; Gomes, D.A.; Cassali, G.D.; Townsend, D.M.; et al. Folate-coated, long-circulating and pH-sensitive liposomes enhance doxorubicin antitumor effect in a breast cancer animal model. Biomed. Pharmacother. 2019, 118, 109323. [Google Scholar] [CrossRef]
- Tie, Y.; Zheng, H.; He, Z.; Yang, J.; Shao, B.; Liu, L.; Luo, L.; Yuan, X.; Liu, Y.; Zhang, X.; et al. Targeting folate receptor β positive tumor-associated macrophages in lung cancer with a folate-modified liposomal complex. Signal Transduct. Target. Ther. 2020, 5, 6. [Google Scholar] [CrossRef]
- Kato, N.; Sato, T.; Fuchigami, Y.; Suga, T.; Geng, L.; Tsurumaru, M.; Hagimori, M.; Mukai, H.; Kawakami, S. Synthesis and evaluation of a novel adapter lipid derivative for preparation of cyclic peptide-modified PEGylated liposomes: Application of cyclic RGD peptide. Eur. J. Pharm. Sci. 2022, 176, 106239. [Google Scholar] [CrossRef] [PubMed]
- Laginha, K.M.; Verwoert, S.; Charrios, G.J.R.; Allen, T.M. Determination of Doxorubicin Levels in Whole Tumor and Tumor Nuclei in Murine Breast Cancer Tumors. Clin. Cancer Res. 2005, 11, 6944–6949. [Google Scholar] [CrossRef] [PubMed]
- d’Avanzo, N.; Torrieri, G.; Figueiredo, P.; Celia, C.; Paolino, D.; Correia, A.; Moslova, K.; Teesalu, T.; Fresta, M.; Santos, H.A. LinTT1 peptide-functionalized liposomes for targeted breast cancer therapy. Int. J. Pharm. 2021, 597, 120346. [Google Scholar] [CrossRef] [PubMed]
- Suelmann, B. Image-Guided Targeted Doxorubicin Delivery with Hyperthermia to Optimize Loco-regional Control in Breast Cancer (i-GO). Available online: https://clinicaltrials.gov/study/NCT03749850 (accessed on 25 April 2024).
- ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ (accessed on 25 April 2024).
- Thomas, V.; O’Halloran, H.C.; Mazar, A. Nanoparticle Arsenic-Platinum Compositions; Northwestern University: Evanston, IL, USA, 2021. [Google Scholar]
- Daryl, C.; Drummond, W.K. Treating Ephrin Receptor a2 (epha2) Positive Cancer with Targeted Docetaxel-Generating Nano-liposome Compositions. U.S. Patent Application 16/085,508, 16 March 2017. [Google Scholar]
- Meers, P.R.; Pak, C.; Ali, S.; Janoff, A.S.; Franklin, J.C.; Erukulla, R.K.; Cabral-Lilly, D.; Ahl, P.L. Peptide-Lipid Conjugates, Liposomes and Lipsomal Drug Delivery. U.S. Patent 6,339,069, 15 January 2002. [Google Scholar]
- Micklus, M.; Greig, N.; Rapoport, S. Targeting of Liposomes to the Blood-Brain Barrier. U.S. Patent Application 09/794,719, 28 February 2002. [Google Scholar]
- San Kim, I.; Lee, B.H.; Lu, M.J.M.; Liang, H.-F.; Ko, Y.-J.; Lo, Y.-C.; Chang, L.-W.; Wei, M.-C. Target-Aiming Drug Delivery System for Diagnosis and Treatment of Cancer Containing Liposome Labeled with Peptides Which Specifically Targets Interleukin-4 Receptors, and Manufacturing Method Thereof. U.S. Patent 9,833,464, 12 May 2017. [Google Scholar]
- Jaspers, S.R.; Presnell, S.R. Methods of Treating Inflammation Using IL-17A and IL-17F Cross-Reactive Monoclonal Antibodies. U.S. Patent 10,023,656, 17 July 2018. [Google Scholar]
- Fang, J.; Nakamura, H.; Maeda, H. The EPR effect: Unique features of tumor blood vessels for drug delivery, factors involved, and limitations and augmentation of the effect. Adv. Drug Deliv. Rev. 2011, 63, 136–151. [Google Scholar] [CrossRef]
- Liu, X.; Huang, G. Formation strategies, mechanism of intracellular delivery and potential clinical applications of pH-sensitive liposomes. Asian J. Pharm. Sci. 2013, 8, 319–328. [Google Scholar] [CrossRef]
Marketed Name | API | Approval Year | Indication | Reference |
---|---|---|---|---|
Doxil® | Doxorubicin | 1995 | Ovarian cancer, Kaposi’s sarcoma, multiple myeloma | [7] |
DaunoXome® | Daunorubicin | 1996 | Kaposi’s sarcoma | [7] |
Onivyde® | Irinotecan hydrochloride trihydrate | 1996 | Pancreatic adenocarcinoma | [7] |
AmBisome® | Amphotericin B | 1997 | Breast cancer | [7] |
DepoCyt® | Cytarabine | 1999 | Lymphomatous meningitis | [7] |
Visudyne® | Verteporphin | 2000 | Age-related macular degeneration | [7] |
DepoDur® | Morphine sulfate | 2004 | Pain management | [7] |
Exparel® | Bupivacaine | 2011 | Anesthesia | [7] |
Marqibo® | Vincristine | 2012 | Leukemia | [7] |
Shingrix® | Recombinant varicella zoster virus glycoprotein E | 2017 | Shingles | [7] |
Vyxeos® | Daunorubicin + cytarabine | 2017 | Leukemia | [7] |
Arikayce® | Amikacin | 2018 | Lung infection | [7] |
Onpattro® | Patisiran | 2018 | Hereditary transthyretin-mediated amyloidosis | [7] |
Comirnaty® | mRNA | 2021 | COVID-19 | [7] |
Spikevax® | mRNA | 2022 | COVID-19 | [7] |
Clinical Phase | Targeting Mechanism | Intervention | Indication | Name or Clinical Trial ID | Reference |
---|---|---|---|---|---|
Preclinical | Aptamer and PEG | Doxorubicin | Cancer | Syl3c-PLD | [161] |
Aptamer and PEG | Doxorubicin | Cancer | ER-lip | [118] | |
pH Sensitive | Docetaxel, SIRT1 shRNA | Breast cancer | DTX-lipoplex | [162] | |
pH Sensitive and PEG | Gemcitabine Curcumin | Pancreatic cancer | PSL | [163] | |
pH Sensitive, Folate, and PEG | Doxorubicin | Breast Cancer | SpHL-DOX-Fol | [164] | |
Folate and PEG | BIM-S plasmid | Non-small lung cancer | F-PLP/pBIM | [165] | |
Peptide (cRGD) and PEG | Doxorubicin | Cancer | cRGD-KK-SG/Dox-PEGylated liposomes | [166] | |
Antibody (anti-CD19 and anti-CD20 monoclonal antibodies) and PEG | Doxorubicin | Breast cancer | DOPC-PLD | [167] | |
Peptide (LinTT1) and PEG | Doxorubicin and sorafenib | Breast cancer | LinTT1 Liposome | [168] | |
Phase 1 | Thermosensitive and PEG | Doxorubicin with HIFU | Breast cancer | NCT03749850 | [169] |
Thermosensitive and PEG | Doxorubicin with HIFU | Liver tumor | NCT02181075 | [170] | |
Thermosensitive and PEG | Doxorubicin with HIFU | Pediatric solid tumors | NCT02536183 | [170] | |
Phase 2 | Thermosensitive and PEG | Doxorubicin with microwave hypothermia | Breast cancer | NCT00826085 | [170] |
Thermosensitiveand PEG | Doxorubicin with radiofrequency ablation | Colon cancer and liver metastasis | NCT01464593 | [170] | |
Antibody (Cetuximab Fab fragment) | Doxorubicin | Breast cancer | NCT02833766 | [170] | |
Phase 3 | Thermosensitive and PEG | Doxorubicin with radiofrequency ablation | Hepatocellular carcinoma | NCT02112656 | [170] |
Patent Number | Title | Description | Reference |
---|---|---|---|
US201916592263A | Nanoparticle arsenic–platinum compositions | This patent covers the co-encapsulation of active forms of arsenic and platinum drugs into liposomes and methods of using these liposomes for the diagnosis and treatment of cancer. Specifically, these liposomes are stable at physiological temperatures and release the encapsulated drug at low pH. | [171] |
US2017022629W | Treating ephrin receptor a2 (Epha2) positive cancer with targeted docetaxel-generating nano liposome compositions | This patent describes docetaxel-carrying liposomes conjugated with both PEG and an antibody for EphA2. They are documented to release docetaxel at physiological pH | [172] |
US6339069B1 | Peptide–lipid conjugates, liposomes and liposomal drug delivery | This patent describes a liposome formulated with peptide-conjugated lipids that upon contact with the corresponding peptidase will destabilize and release the encapsulated drug | [173] |
US20020025313A1 | Targeting of liposomes to the blood–brain barrier | This patent covers targeted liposomes for the delivery of drugs to the brain using conjugated antibody fragments. The listed targets include transferrin receptor, insulin receptor, insulin-like growth factor-1, insulin-like growth factor-2, and more. | [174] |
US9833464B2 | Target-aiming drug delivery system for diagnosis and treatment of cancer containing liposome labeled with peptides which specifically targets interleukin-4 receptors, and manufacturing method thereof | This patent covers the liposomal drug delivery for diagnosis and treatment of cancer using conjugated peptides that target interleukin-4 receptors | [175] |
US10023656B2 | Methods of treating inflammation using IL-17A and IL-17F cross-reactive monoclonal antibodies | This patent describes the use of antibodies for blocking, reducing, antagonizing, or neutralizing the activity of interleukin-17A and iterleukin-17F. The patent includes methods of using these antibodies, including liposomal vehicles. | [176] |
Targeting Strategy | Mechanism | Advantages | Limitations | References |
---|---|---|---|---|
Passive Targeting | EPR Effect | Natural phenomenon Can be further enhanced or combined with other therapies (e.g., phototherapy) | Limited specificity Magnitude of effect varies by characteristics of tumor | [33,177] |
PEGylation | Prolonged circulation Less drug leakage and instabilities | May induce ABC phenomenon | [38,40,47] | |
Active Targeting | Antibodies | High specificity Fab portions can be used for possibly better pharmacokinetics | Possible immunogenicity Mixed in vivo results | [63,67,72,75] |
Peptides | Versatile applications Low immunogenicity | Stability and storage concerns | [36,82,84,86] | |
Folate | High affinity for folate receptors in many cancers | Not applicable to all tumors or diseases Pharmacokinetic concerns | [102,104,105] | |
Aptamers | Wide range of targets Low immunogenicity | Stability concerns Unfamiliar to the industry | [111,113,114] | |
External Cues | pH Sensitive | Low off-target toxicity Intracellular delivery | Premature drug release | [127,178] |
Temperature-Sensitive | Effective drug unloading Manual release | Limited applicability | [134,142] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Gatto, M.S.; Johnson, M.P.; Najahi-Missaoui, W. Targeted Liposomal Drug Delivery: Overview of the Current Applications and Challenges. Life 2024, 14, 672. https://doi.org/10.3390/life14060672
Gatto MS, Johnson MP, Najahi-Missaoui W. Targeted Liposomal Drug Delivery: Overview of the Current Applications and Challenges. Life. 2024; 14(6):672. https://doi.org/10.3390/life14060672
Chicago/Turabian StyleGatto, Matthew S., McNeely P. Johnson, and Wided Najahi-Missaoui. 2024. "Targeted Liposomal Drug Delivery: Overview of the Current Applications and Challenges" Life 14, no. 6: 672. https://doi.org/10.3390/life14060672
APA StyleGatto, M. S., Johnson, M. P., & Najahi-Missaoui, W. (2024). Targeted Liposomal Drug Delivery: Overview of the Current Applications and Challenges. Life, 14(6), 672. https://doi.org/10.3390/life14060672