ENZYMOSOMES: A RISING EFFECTUAL TOOL FOR TARGETED DRUG DELIVERY SYSTEM

Authors

  • Shefrin S. Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Amrita University, AIMS Health Science Campus, Kochi, India
  • Sreelaxmi C. S. Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Amrita University, AIMS Health Science Campus, Kochi, India
  • Vishnu Vijayan Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Amrita University, AIMS Health Science Campus, Kochi, India
  • Sreeja C. Nair Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Amrita University, AIMS Health Science Campus, Kochi, India

DOI:

https://doi.org/10.22159/ijap.2017v9i6.22556

Keywords:

Enzymosomes, Phospholipids, Superoxide dismutase, Immuno-enzymosomes, Alkaline enzymosomes, Anti-platelet enzymosomes, Targeted drug delivery

Abstract

The study aims to develop enzymosomes as an emerging novel drug delivery system for site-specific action. Enzymosomes utilises the specific nature of an enzyme, which is binding to a specific substrate at a controlled rate and catalysing product production step. An enzyme is encapsulated by coupling covalently to the surface of liposomes/lipid vesicles to form enzymosomes. Enzymes links through acylation, direct conjugation, physical adsorption, encapsulation methods to prepare enzymosomes with targeted action. Such novel drug delivery systems prove effective drug release and concomitantly reduces undesirable side effects of conventional treatment methods and hence showcase improvement in the long-term therapy of the disease. They are a promising substitute to conventional treatment therapies of gout, antiplatelet activities etc. Enzymosomes are newly designed supramolecular vesicular delivery systems to be useful as a tool in pharmaceutics for the raising of drug targeting and physicochemical properties and hence bioavailability. It shows beneficial effects of drugs with a narrow precision because targeting of these drugs to their site of action improves the drugs overall pharmacodynamics and pharmacokinetic profile. It also minimizes alterations in the normal enzymatic activity, thus enhancing half-life and achieve enzyme activity on targeted sites such as cancerous cells.

Downloads

Download data is not yet available.

References

Kumar R, Kumar S, Jha SS, Jha AK. Vesicular system-carrier for drug delivery. Chem Sinica 2011;2:192-202.

Kunwarpuriya AS, Doke VV, Changedia S, Khutle NM. Sphingosome: a novel vesicular drug delivery system. Eur J Pharm Med Res 2015;2:509-25.

Lade S, Burle S, Kosalge S, Kannao S. Lipid-based drug delivery systems: a comprehensive review. Int J Innov Pharm Sci Res 2014;2:2465-75.

Fathima KM, Nitheesh A, Paul A, Nair SC. Sphingosome vesicular system. Int J Pharm Sci Rev Res 2016;39:208-13.

Zishan M, Kushwaha P, Singh K. An overview of: vesicular drug delivery system. World J Pharm Pharm Sci 2017;6:546-60.

Doke V, Kelan D, Khadse D, Khutle N, Chaudhari Y. Ethosome: a novel vesicular drug delivery. World J Pharm Res 2015;4:1199-210.

Biswas GR, Majee SB. Niosomes in ocular drug delivery. Eur J Pharm Med Res 2017;4:813-9.

Vyas SP, Khar RK. Targeted and controlled drug delivery. 1st ed. New Delhi: CBS Publisher; 2002.

Akiladevi D, Basak S. Ethosomes-a noninvasive approach for transdermal drug delivery. Int J Curr Pharma Res 2010;2:1-4.

Jain S, Jain P. Transferosomes: a novel vesicular carrier for enhanced transdermal delivery: development, characterization and performance evaluation. Drug Dev Ind Pharm 2003;29:1013-26.

Nandure HP, Puranik P, Giram P, Lone V. Ethosome: a novel drug carrier. Int J Pharm Res Allied Sci 2013;2:18-30.

Sreelatha D, Brahma CK. Colon targeted drug delivery–a review of primary and novel approaches. J Global Trends Pharm Sci 2013;4:1174-83.

Touitou E, Godin B, Dayan N, Weiss C. Intracellular delivery mediated by an ethosomal carrier. Biomaterials 2001;22:3053-9.

Bhingare U, Khadabadi SS, Shinde N. Pharmacosomes: a novel drug delivery system. Int J Pharm Res Allied Sci 2014;1:14-20.

Moghimipour E, Handali S. Liposomes as drug delivery systems: properties and applications. Res J Pharm Biol Chem Sci 2013;4:169-85.

Pawar P, Kalamkar R, Jain A, Amberkar S. Ethosomes: a novel tool for herbal drug delivery. Int J Pharm Pharm Sci 2015;3:192-202.

Overholtzer M, Brugge JS. The cell biology of cell-in-cell structures. Nat Rev Mol Cell Biol 2008;9:796-809.

Engel H, Rondeau E, Windhab EJ, Walde P. External surface area determination of lipid vesicles using trinitrobenzenesulfonate and ultraviolet/visible spectrophotometry. Anal Biochem 2013;442:262-71.

Villalonga ML, Diez P, Sanchez A, Gamella M, PingarroÌn JM, Villalonga R. Neoglycoenzymes. Chem Rev 2014;114:4868-917.

Akbarzadeh A, Sadabady RR, Davaran S. Liposome: classification, preparation and applications. Nanoscale Res Lett 2013;8:102-11.

Sharma S, Mishra L, Grover I, Gupta A Kaur. Liposome: vesicular system an overview. Int J Pharm Pharm Sci 2010;2:11-7.

Zylberberg C, Matosevic S. Pharmaceutical liposomal drug delivery: a review of new delivery systems and look at the regulatory landscape. Drug Delivery 2016;23:3319-29.

Manish G, Vimukta S. Targeted drug delivery system: a review. Res J Chem Sci 2011;1:135-8.

Kamal K, Garg G, Harikumar SL, Aggarwal G. Potential role of nanotechnology for skin drug delivery. World J Pharm Pharm Sci 2016;5:428-53.

Pardridge WM. Drug transport across the blood-brain barrier. J Cereb Blood Flow Metab 2012;32:1959-72.

Marianecci C, Rinaldi F, Hanieh PN. Drug delivery in overcoming the blood-brain barrier: role of nasal mucosal grafting. Drug Des Dev Ther 2017;11:325-35.

Salunkhe SS, Bhatia NM, Kawade VS, Bhatia MS. Development of lipid-based nanoparticulate drug delivery systems and drug carrier complexes for delivery to brain. J Appl Pharm Sci 2015;5:110-29.

Cui S, Zhi D, Zhao Y. Cationic liposomes with folic acid as targeting ligand for gene delivery. Bioorg Med Chem Lett 2016;26:4025-9.

Gorle S, Sewbalas A, Ariatti M, Singh M. Ligand-tagged cationic liposome facilitates efficient gene delivery to folate receptors. Curr Sci 2016;3:662-70.

Pattni BS, Chupin VV, Torchilin VP. New developments in liposomal drug delivery. Chem Rev 2015;115:10938-66.

Andhale VA, Patil PR, Dhas AU, Chauhan PD, Desai SV. Liposome: an emerging tool in drug carrier system. Int J Pharm Technol 2016;8:10982-1011.

Popovska O, SimonovskaJ, Kavrakovski Z, Rafailovska V. An overview: methods for preparation and characterization of liposomes as drug delivery systems. Int J Pharmphytopharm Res 2014;3:182-9.

Saroj S, Baby DA, Sabitha M. Current trends in lipid-based delivery systems and its applications in drug delivery. Asian J Pharm Clin Res 2012;5:4-9.

Kavitha AN, Deepthi V. Liposomal drug delivery system-a review. RGUHS J Pharm Sci 2014;4:47-56.

Amidon GL, Shah VP. A theoretical basis for a biopharmaceutical drug classification: the correlation of in vitro drug product dissolution and in vivo bioavailability. AAPS J 2014;16:894–98.

Leelarungrayub J, Manorsoi J, Manorsoi A. Anti-inflammatory activity of niosomes entrapped wit Plai oil (ZingibercassumunarRoxb.) by therapeutic ultrasound in rat model. Int J Nanomed 2017;12:2469-76.

Shrestha H, Bala R, Arora S. Lipid-based drug delivery systems. J Pharm 2014;10:1-10.

Devarajan V, Ravichandran V. Nanoemulsions: as modified drug delivery tool. Pharmacie Globale 2011;4:1-6.

Kumar R, Kumar S, Jha SS, Jha AK. Vesicular system-carrier for drug delivery. Pharm Sinica 2011;2:192-202.

Soni K, Kukereja BK, Kapur M, Kohli K. Lipid nanoparticles: future of oral drug delivery and their current trends and regulatory issues. Int J Curr Pharm Rev Res 2015;7:1-18.

Kakadia PG, Conway BR. Solid-lipid nanoparticles: a potential approach for dermal drug delivery. Am J Pharmacol Sci 2014;2:1-7.

Trombino S, Mellace S, Cassano R. Solid lipid nanoparticles for antifungal drugs delivery for topical applications. Ther Delivery 2016;7:639-47.

Badilli U, Turk CTS, Amasya G, Tarimci N. Novel drug delivery system for dermal uptake of etofenamate: semisolid SLN dispersion. Curr Drug Delivery 2017;14:386-93.

Das S, Chaudhury A. Recent advances in lipid nanoparticle formulations with the solid matrix for oral drug delivery. AAPS PharmSciTech 2011;12:62-76.

Rajitha P, Gopinath D, Biswas R, Sabitha M, Jayakumar R. Chitosan nanoparticles in drug therapy and infectious diseases. Expert Opin Drug Delivery 2016;13:1177-94.

Gaspar MM, Martins MB, Corvo ML, Cruz MEM. Design and characterization of enzymosomes with surface-exposed superoxide dismutase. Biochim Biophys Acta 2003;16:211-7.

Corvo ML, Marinho HS, Marcelino P. Superoxide dismutase enzymosomes: carrier capacity optimization, in vivo behaviour and therapeutic activity. Pharm Res 2015;32:91-102.

Sabitha K, Venugopal B, Rafi M, Ramana KV. Role of antioxidant enzymes in glucose and lipid metabolism in association with obesity and type 2 diabetes. Am J Med Sci 2014;2:21-4.

Kobsa S, Saltzman WM. Bioengineering approaches to controlled protein delivery. Pediatr Res 2008;63:513-9.

Anwekar H, Patel S, Singhai AK. Liposome-as drug carrier. Int J Life Sci Pharma Res 2011;2:945-51.

Solaro R, Chiellini F, Battisti A. Targeted delivery of protein drugs by nanocarriers. Materials 2010;3:1928-80.

Peer D, Karp JM, Hong S, Farokhzad OC, Margalit R, Langer R. Nanocarriers as an emerging platform for cancer therapy. Nat Nanotechnol 2007;2:751-60.

Sivasankar M, Katyayani T. Liposomes: the future of formulations. Int J Res Pharm Chem 2011;1:259-67.

Zhao H, Lin ZY, Yildirimer L, Dhinakar A, Zhao X, Wu J. Polymer-based nanoparticles for protein delivery: design, strategies, and applications. J Med Chem 2016;4:4060-71.

Liu G, Li K, Wang H. Polymeric micelles based on PEGulated chitosan-g lipoic acid carrier for efficient intracellular drug delivery. J Biomater Appl 2016;31:1039-48.

Fanum M. Colloids in drug delivery. 1st ed. USA: CRC Press; 2016.

Venkatesh DN, Rao P. Nanoparticles for cancer treatment-a comprehensive review. World J Pharm Pharm Sci 2016;5:481-99.

Gaspar MM, Boerman OC, Laverman P. Enzymosomes with surface–exposed superoxide dismutase: in vivo behaviour and therapeutic activity in model of adjuvant arthritis. J Controlled Release 2007;117:186-95.

Xu G, McLeod HL. Strategies for enzyme/prodrug cancer therapy. Clin Cancer Res 2001;7:3314-24.

Bisswanger H. Enzyme assays. Perspect Sci 2014;1:51-5.

Chen T, Wang R, Lu T, Liang G, Lu T. Modification with liposomes by the dansyl-labeled heterobifunctional crosslinker. J Biomater Appl 2011;26:117-25.

Yang Y, Wang J, Shigematsu H. Self assembly of size-controlled liposomes on DNA nanoplates. Nat Chem 2016;8:476-83.

Shekhar C. Lean and mean: nanoparticle-based delivery improves the performance of cancer drugs. Chem Biol 2009;16:349-50.

Saeed HM, Abdel-Fattah YR, Gohar YM, Elbaz MA. Purification and characterization of extracellular Pseudomonas aeruginosaurate oxidase enzyme. Pol J Microbiol 2004;53:45–52.

Wu J, Lu S, Zheng Z, Zhu L, Zhan X. Modification with the polysialic acid-PEG copolymer as a new method for improving the therapeutic efficacy of proteins. Prep Biochem Biotechnol 2016;46:788-97.

Szczupak A, Aizik D, Morais S. The electrosome: a surface-displayed enzymatic cascade in a biofuel cell’s anode and a high-density surface-displayed biocathodic enzyme. Nanomaterials 2017;7:1-17.

Secundo F. Conformational changes of enzymes upon immobilisation. Chem Soc Rev 2013;42:6250-61.

Tan Q, Zhang J, Wang N. Uricase from Bacillus fastidious loaded in alkaline enzymosomes: enhanced biochemical and pharmacological characteristics in hypouricemic rats. Eur J Pharm Biopharm 2012;82:43-8.

Sherman MR, Saifer MG, Perez-Ruiz F. PEG-uricase in the management of treatment-resistant gout and hyperuricemia. Adv Drug Delivery Rev 2008;60:59–68.

Tan QY, Wang N, Yang H, Zhang LK, Liu S, Chen L, et al. Characterization, stabilization and activity of uricase loaded in lipid vesicles. Int J Pharm 2010;384:165–72.

Zhao Y, Zhao L, Yang G, Tao J, Bu Y, Liao F. Characterization of a uricase from Bacillus fastidious A. T. C. C. 26904 and its application to serum uric acid assay by a patented kinetic uricase method. Biotechnol Appl Biochem 2006;45:75–80.

Budai M, Chapela P, Gróf P, Zimmer A, Wales ME, Wild JR, et al. Physicochemical characterization of stealth liposomes encapsulating an organophosphate-hydrolysing enzyme. J Liposome Res 2009;19:163–8.

Zhou Y, Zhang M, He D. Uricase alkaline enzymosomes with enhanced stabilities and anti-hyperuricemia effects induced by favourable microenvironmental changes. Sci Rep 2016;7:1-14.

Walde P, Ichikawa S. Enzymes inside lipid vesicles: preparation, reactivity and applications. Biomol Eng 2001;18:143–77.

Tan QY, Wang N, Yang H. Preparation and characterization of lipid vesicles containing uricase. Drug Delivery 2010;17:28–37.

Szczurek P, Mosiichuk N, Woliński J. Oral uricase eliminates blood uric acid in the hyperuricemic pig model. PLoS ONE 2017;12:179-95.

Grassi D, Ferri L, Desideri G. Chronic hyperuricemia, uric acid deposit and cardiovascular risk. Curr Pharm Des 2013;19:2432–8.

Xiong H, Zhou Y, Zhou Q. Nanosomal micro assemblies for highly efficient and safe delivery of therapeutic enzymes. ACS Appl Mater Interfaces 2015;7:20255-63.

Liu X, Zhang Z, Zhang Y. Artificial metalloenzymeâ€based enzyme replacement therapy for the treatment of hyperuricemia. Adv Funct Mater 2016;26:7921-8.

Marcus AJ, Broekman MJ, Drosopoulos JH. Inhibition of platelet recruitment by endothelial cell CD39/ecto-ADPase: significance for occlusive vascular diseases. Ital Heart J 2001;2:824-30.

Chaikof EL, Haller CA, Cui1 W, Wen J, Robson SC. CD39 enzymosomes inhibit platelet activation in vitro and in vivo. ‎J Surg Res 2006;130:234-5.

Nielsen UB, Kirpotin DB, Pickering EM, Drummond DC, Marks JD. A novel assay for monitoring internalization of nanocarrier coupled antibodies. BMC Immunol 2006;7:1-15.

Jing Y, Trefna HD, Persson M, Svedham S. Heat-activated liposome targeting to streptavidin-coated surfaces. Biochim Biophys Acta 2015;1848:1417-23.

Howard M, Zern BJ, Anselmo AC. Vascular targeting of nanocarriers: perplexing aspects of the seemingly straightforward paradigm. ACS Nano 2014;8:4100-32.

Powers AD, Palecek SP. Protein analytical for diagnosing, monitoring, choosing treatment for cancer patients. J Healthcare Eng 2012;3:503-34.

Su Y, Xie Z, Kim GB, Dong C, Yang J. Design strategies and applications of circulating cell-mediated drug delivery systems. ACS Biomater Sci Eng 2015;1:201-17.

Dowhan W. Understanding phospholipid function: why are there so many lipids? J Biol Chem 2017;292:10755-66.

Collins MD, Gordon S. Giant liposome preparation for imaging and patch-clamp electrophysiology. J Visualized Exp 2013;50:1-9.

Kube S, Hersch N, Naumovska E. Fusogenic liposomes as nanocarriers for the delivery of intracellular proteins. Lagmuir 2017;33:1051-9.

Smith E, Collins I. Photoaffinity labeling in target-and binding-site identification. Future Med Chem 2015;7:159-83.

Fogen D, Wu SC, Ng KKS, Wong SL. Engineering streptavidin and streptavidin binding peptide with infinite binding affinity and reversible binding capability: purification of a tagged recombinant protein to high purity via affinity-driven thiol coupling. PLoS One 2015;10:137-9.

Barbet J, Machy P, Leserman LD. Monoclonal antibody covalently coupled to liposomes: specific targeting to cells. J Supramol Struct Cell Biochem 1981;243:16-23.

Picktel E, Niemirowicz K, Watek M. Recent insights in nanotechnology-based drugs and formulations designed for effective anti-cancer therapy. J Nanobiotechnol 2016;14:1-23.

Zhao C, Busch DJ, Vershel CP, Stachowiak JC. Multi-functional transmembrane protein ligands for cell-specific targeting of plasma membrane-derived vesicles. SMALL 2016;12:3387-48.

Jain S, Gautam V, Naseem S. Acute-phase proteins: as a diagnostic tool. J Pharm Bioall Sci 2011;3:118-27.

Cherian AK, Rana AC, Jain SK. Self-assembled carbohydrate-stabilized ceramic nanoparticles for the parenteral delivery of insulin. Drug Dev Ind Pharm 2000;26:459-63.

Rahmanian N, Eskandani M, Barar J, Omidi. Recent trends in targeted therapy of cancer using grapheme oxide-modified multifunctional nanomedicines. J Drug Target 2017;25:202-15.

Korting SM, Mehnert W, Korting HC. Lipid nanoparticles for improved topical application of drugs for skin diseases. Adv Drug Delivery Rev 2007;59:427-43.

Sutariya V, Patel P. Aquasome: a novel carrier for drug delivery. Int J Pharm Sci Res 2012;3:688-91.

Nair SC, Nair AS, Vidhya KM, Saranya TR, Sreelakshmy KR. Emulsomes: a novel liposomal formulation for sustained drug delivery. Int Res J Pharm Appl Sci 2013;3:192-6.

Published

07-11-2017

How to Cite

S., S., S., S. C., Vijayan, V., & Nair, S. C. (2017). ENZYMOSOMES: A RISING EFFECTUAL TOOL FOR TARGETED DRUG DELIVERY SYSTEM. International Journal of Applied Pharmaceutics, 9(6), 1–9. https://doi.org/10.22159/ijap.2017v9i6.22556

Issue

Section

Review Article(s)

Most read articles by the same author(s)