Generic placeholder image

Recent Patents on Nanotechnology

Editor-in-Chief

ISSN (Print): 1872-2105
ISSN (Online): 2212-4020

Review Article

Recent Developments in Nano-Formulations Against Diabetes

Author(s): Swaralipi Choudhury and Prasun Patra*

Volume 17, Issue 4, 2023

Published on: 29 August, 2022

Page: [340 - 358] Pages: 19

DOI: 10.2174/1872210516666220622114505

Price: $65

Abstract

Diabetes mellitus (DM) is a life-threatening metabolic syndrome, but patient compliance is poor due to the pain and inconvenience caused by the subcutaneous injection of insulin and other macromolecular diabetic therapies. Current challenges in DM management are to optimize the use of available therapies and reduce complications. For clinical improvements, future therapies need to be easier to use, achieving tighter glycemic control, better safety profiles, and reduced manufacturing costs. The medical applications of nanotechnology are enormous and have been proven to be the best approach to improve compliance and clinical efficacy by overturning biopharmaceutical obstacles. Nanoformulations enhance the properties of conventional drugs and are specific to the targeted delivery site. The aim of the present review is to provide an overview of the application of nano-formulations in diabetes management. We analyze the current state of most of the available approaches which are in various stages of research and development. Herein, we review the developing role of nanotechnology in diabetes management and focus on the technologies that we feel are most likely to have an impact.

Keywords: Diabetes mellitus (DM), nano-formulations, nanoparticles, nano-medicine, insulin delivery, nanodevices.

Graphical Abstract
[1]
Okur ME, Karantas ID, Siafaka PI. Diabetes mellitus: A review on pathophysiology, current status of oral medications and future perspectives. Acta Pharma Sci 2017; 55(1): 61.
[http://dx.doi.org/10.23893/1307-2080.APS.0555]
[2]
Tang KS. The current and future perspectives of zinc oxide nanoparticles in the treatment of diabetes mellitus. Life Sci 2019; 239: 117011.
[http://dx.doi.org/10.1016/j.lfs.2019.117011] [PMID: 31669241]
[3]
Jeevanandam J, Danquah MK, Debnath S, Meka VS, Chan YS. Opportunities for nano-formulations in type 2 diabetes mellitus treatments. Curr Pharm Biotechnol 2015; 16(10): 853-70.
[http://dx.doi.org/10.2174/1389201016666150727120618] [PMID: 26212563]
[4]
Harsoliya MS, Patel VM, Modasiya M. Recent advances and applications of nanotechnology in diabetes. Int J Pharm Biol Arch 2012; 3: 255.
[5]
Simos YV, Spyrou K, Patila M, et al. Trends of nanotechnology in type 2 diabetes mellitus treatment. Science Direct 2021; 16(1): 62-76.
[http://dx.doi.org/10.1016/j.ajps.2020.05.001] [PMID: 33613730]
[6]
Rahiman S, Tantry BA. Nanomedicine current trends in diabetes management. J Nanomed Nanotechnol 2012; 3(5): 2-7.
[7]
Jeevanandam J, Chan YS, Danquah MK. Nano-formulations of drugs: Recent developments, impact and challenges. Biochimie 2016; 128-129: 99-112.
[http://dx.doi.org/10.1016/j.biochi.2016.07.008] [PMID: 27436182]
[8]
Zhao X, Hao Y, Yuan L, et al. Nano-formulations for transdermal drug delivery: A review. Chin Chem Lett 2018; 29: 1713.
[http://dx.doi.org/10.1016/j.cclet.2018.10.037]
[9]
Khogta S, Patel J, Barve K, Londhe V. Herbal nano-formulations for topical delivery. J Herb Med 2020; 20: 100300.
[http://dx.doi.org/10.1016/j.hermed.2019.100300]
[10]
Dewanjee S, Chakraborty P, Mukherjee B, De Feo V. Plant-based antidiabetic nanoformulations: The emerging paradigm for effective therapy. Int J Mol Sci 2020; 21(6): 2217.
[http://dx.doi.org/10.3390/ijms21062217] [PMID: 32210082]
[11]
George A, Shah PA, Shrivastav PS. Natural biodegradable polymers based nano-formulations for drug delivery: A review. Int J Pharm 2019; 561: 244-64.
[http://dx.doi.org/10.1016/j.ijpharm.2019.03.011] [PMID: 30851391]
[12]
Wong CY. Recent advancements in oral administration of insulin- loaded liposomal drug delivery systems for diabetes mellitus. Int J Pharm 2018; 549(1-2): 201-17.
[13]
Singh AP, Biswas A, Shukla A, Maiti P. Targeted therapy in chronic diseases using nanomaterial-based drug delivery vehicles. Signal Transduct Target Ther 2019; 4: 33.
[http://dx.doi.org/10.1038/s41392-019-0068-3] [PMID: 31637012]
[14]
Kesharwani P, Gorain B, Low SY, et al. Nanotechnology based approaches for anti-diabetic drugs delivery. Diabetes Res Clin Pract 2018; 136: 52-77.
[http://dx.doi.org/10.1016/j.diabres.2017.11.018] [PMID: 29196152]
[15]
Nie X, Chen Z, Pang L, et al. Oral nano drug delivery systems for the treatment of type 2 diabetes mellitus: An available administration strategy for antidiabetic phytocompounds. Int J Nanomedicine 2020; 15: 10215-40.
[http://dx.doi.org/10.2147/IJN.S285134] [PMID: 33364755]
[16]
Yücel Ç, Karatoprak GS. Aktaş Y. Nanoliposomal resveratrol as a novel approach to treatment of diabetes mellitus. J Nanosci Nanotechnol 2018; 18(6): 3856-64.
[http://dx.doi.org/10.1166/jnn.2018.15247] [PMID: 29442719]
[17]
Arafat M, Kirchhoefer C, Mikov M, Sarfraz M, Löbenberg R. Nanosized liposomes containing bile salt: A vesicular nanocarrier for enhancing oral bioavailability of BCS class III drug. J Pharm Pharm Sci 2017; 20(0): 305-18.
[http://dx.doi.org/10.18433/J3CK88] [PMID: 28885915]
[18]
Jacobsen AC, Jensen SM, Fricker G, Brandl M, Treusch AH. Archaeal lipids in oral delivery of therapeutic peptides. Eur J Pharm Sci 2017; 108: 101-10.
[http://dx.doi.org/10.1016/j.ejps.2016.12.036] [PMID: 28108360]
[19]
Bhat MI. Niosomes a controlled and novel drug delivery system: A brief review. World J Pharm Res 2019; 8: 481.
[20]
Desai SV, Joshi B, Upadhyay V. An overview on niosomes as novel drug delivery system. Res J Pharm Dos Forms Technol 2020; 12: 271.
[http://dx.doi.org/10.5958/0975-4377.2020.00045.2]
[21]
Samed N, Sharma V, Sundaramurthy A. Hydrogen bonded niosomes for encapsulation and release of hydrophilic and hydrophobic anti-diabetic drugs: An efficient system for oral anti-diabetic formulation. Appl Surf Sci 2018; 449: 567-73.
[http://dx.doi.org/10.1016/j.apsusc.2017.11.055]
[22]
Bini KB, Akhilesh D, Prabhakara P, Kamath JV. Development and characterization of non-ionic surfactant vesicles (niosomes) for oral delivery. Inter J Drug Develop Res 2012; 4: 147-54.
[23]
Sharma PK, Saxena P, Jaswanth A, Chalamaiah M, Balasubramanian A. Anti-diabetic activity of lycopene niosomes: Experimental observation. J Pharm Drug Develop 2017; 4: 103.
[24]
Ge X, Wei M, He S, Yuan WE. Advances of non-ionic surfactant vesicles (Niosomes) and their application in drug delivery. Pharmaceutics 2019; 11(2): 55.
[http://dx.doi.org/10.3390/pharmaceutics11020055] [PMID: 30700021]
[25]
Khoee S, Yaghoobian M. Niosomes: A novel approach in modern drug delivery systems. Nanostruct Drug Deliv 2017; 6: 207-37.
[http://dx.doi.org/10.1016/B978-0-323-46143-6.00006-3]
[26]
Zielińska A, Carreiró F, Oliveira AM, et al. Polymeric nanoparticles: Production, characterization, toxicology and ecotoxicology. Molecules 2020; 25(16): 3731.
[http://dx.doi.org/10.3390/molecules25163731] [PMID: 32824172]
[27]
El- Say KM. Sawy HS. Polymeric nanoparticles: Promising platform for drug delivery. Int J Pharm 2017; 528(1-2): 675-91.
[28]
Jarai BM, Kolewe EL, Stillman ZS, Raman N, Fromen CA. Polymeric nanoparticles. In: Eun JC, Lorraine L, Carlos R, Eds. Nanoparticles for Biomedical Applications. London: Elsevier 2020; Vol. 18: pp. 303-24.
[http://dx.doi.org/10.1016/B978-0-12-816662-8.00018-7]
[29]
Kong M, Peng X, Cui H, Liu P, Pang B, Zhang K. pH- responsive polymeric nanoparticles with tunable sizes for targeted drug delivery. Royal Society Chem 2020; 10: 4860-8.
[http://dx.doi.org/10.1039/C9RA10280A]
[30]
Wang JL, Du XJ, Yang JX, et al. The effect of surface poly(ethylene glycol) length on in vivo drug delivery behaviors of polymeric nanoparticles. Biomaterials 2018; 182: 104-13.
[http://dx.doi.org/10.1016/j.biomaterials.2018.08.022] [PMID: 30114562]
[31]
Begines B, Ortiz T, Pérez-Aranda M, et al. Polymeric nanoparticles for drug delivery: Recent developments and future prospects. Nanomaterials (Basel) 2020; 10(7): 1403.
[http://dx.doi.org/10.3390/nano10071403] [PMID: 32707641]
[32]
Zhang P, Zhang Y, Liu CG. Polymeric nanoparticles based on carboxymethyl chitosan in combination with painless microneedle therapy systems for enhancing transdermal insulin delivery. RSC Advances 2020; 10: 24319-29.
[http://dx.doi.org/10.1039/D0RA04460A]
[33]
Sgorla D, Lechanteur A, Almeida A, et al. Development and characterization of lipid-polymeric nanoparticles for oral insulin delivery. Expert Opin Drug Deliv 2018; 15(3): 213-22.
[http://dx.doi.org/10.1080/17425247.2018.1420050] [PMID: 29257904]
[34]
Crucho CIC, Barros MT. Polymeric nanoparticles: A study on the preparation variables and characterization methods. Mater Sci Eng C 2017; 80: 771-84.
[http://dx.doi.org/10.1016/j.msec.2017.06.004] [PMID: 28866227]
[35]
Chaudhari H, Chaudhari HS, Popat R, Adhao VS, Shrikharde VN. Dendrimers: Novel carriers for drug delivery. J Appl Pharm Res 2016; 4(1): 1-19.
[36]
Mishra V, Yadav N, Saraogi GK, Tambuwala MM, Giri N. Dendrimer based nanoarchitectures in diabetes management: An overview. Curr Pharm Des 2019; 25(23): 2569-83.
[http://dx.doi.org/10.2174/1381612825666190716125332] [PMID: 31333099]
[37]
Chavda VP. Nanobased nanodrug delivery: A comprehensive review. In: Shyam SM, Shivendu R, Nandita D, Raghvendra KM, Sabu T, Eds. Applications of Targeted Nano Drugs and Delivery Systems-Nanoscience and Nanotechnology in Drug Delivery. London: Elsevier 2019; pp. 69-92.
[38]
Pan J, Attia SA, Filipczak N, Torchilin VP. Dendrimers for drug delivery purposes. In: Masoud M, Ed. Nanoengineered Biomaterials for Advanced Drug Delivery. London: Elsevier 2020; pp. 201-42.
[http://dx.doi.org/10.1016/B978-0-08-102985-5.00010-3]
[39]
Dias AP, da Silva Santos S, da Silva JV, et al. Dendrimers in the context of nanomedicine. Int J Pharm 2020; 573: 118814.
[http://dx.doi.org/10.1016/j.ijpharm.2019.118814] [PMID: 31759101]
[40]
Ghosh S, Ghosh S, Sil PC. Role of nanostructures in improvising oral medicine. Toxicol Rep 2019; 6: 358-68.
[http://dx.doi.org/10.1016/j.toxrep.2019.04.004] [PMID: 31080743]
[41]
Huang D, Wu D. Biodegradable dendrimers for drug delivery. Mater Sci Eng C 2018; 90: 713-27.
[http://dx.doi.org/10.1016/j.msec.2018.03.002] [PMID: 29853143]
[42]
Sherje AP, Jadhav M, Dravyakar BR, Kadam D. Dendrimers: A versatile nanocarrier for drug delivery and targeting. Int J Pharm 2018; 548(1): 707-20.
[http://dx.doi.org/10.1016/j.ijpharm.2018.07.030] [PMID: 30012508]
[43]
Akbarzadeh A, Khalilov R, Mostafavi E, et al. Role of dendrimers in advanced drug delivery and biomedical applications: A review. Exp Oncol 2018; 40(3): 178-83.
[http://dx.doi.org/10.31768/2312-8852.2018.40(3):178-183] [PMID: 30285011]
[44]
Nikzamir M, Hanifehpour Y, Akbarzadeh A, Panahi Y. Applications of dendrimers in nanomedicine and drug delivery: A review. J Inorg Organomet Polym Mater 2021; 31: 2246.
[http://dx.doi.org/10.1007/s10904-021-01925-2]
[45]
Gothwal A, Malik S, Gupta U, Jain NK. Toxicity and biocompatibility aspects of dendrimers. In: Abhay C, Hitesh K, Eds. Pharmaceutical Applications of Dendrimers Micro and Nano Technologies. London: Elsevier 2020; pp. 251-74.
[http://dx.doi.org/10.1016/B978-0-12-814527-2.00011-1]
[46]
Patra JK, Das G, Fraceto LF, et al. Nano based drug delivery systems: Recent developments and future prospects. J Nanobiotechnology 2018; 16(1): 71.
[http://dx.doi.org/10.1186/s12951-018-0392-8] [PMID: 30231877]
[47]
Patel V, Rajani C, Paul D, et al. Dendrimers as novel drug delivery system and its applications. In: Rakesh KT, Ed. Drug Delivery System. London: Academic Press 2020; pp. 333-92.
[48]
Baxi K, Momin M, Sawarkar S. Immunotherapy- A novel Facet of Modern Therapeutics In: Sujata PS, Vandana SN, Shariq S, Eds. (1st ed..). Singapore: Springer 2021; p. 153.
[49]
Pham DT, Chokamonsirikun A, Phattaravorakarn V, Tiyaboonchai W. Polymeric micelles for pulmonary drug delivery: A comprehensive review. J Mater Sci 2021; 56: 2016.
[http://dx.doi.org/10.1007/s10853-020-05361-4]
[50]
Mandal A, Bisht R, Rupenthal ID, Mitra AK. Polymeric micelles for ocular drug delivery: From structural frameworks to recent preclinical studies. J Control Release 2017; 248: 96-116.
[http://dx.doi.org/10.1016/j.jconrel.2017.01.012] [PMID: 28087407]
[51]
Fu Y, Ding Y, Zhang Y, Liu J, Yu P. Polyethylene glycol (PEG) Related controllable and sustainable antidiabetic drug delivery systems. Eur J Med Chem 2021; 217: 113372.
[52]
Lu Y, Yue Z, Xie J, et al. Micelles with ultralow critical micelle concentration as carriers for drug delivery. Nat Biomed Eng 2018; 2(5): 318-25.
[http://dx.doi.org/10.1038/s41551-018-0234-x] [PMID: 30936455]
[53]
Rassu G, Pavan B, Mandracchia D, et al. Polymeric nanomicelles based on insulin D alpha-tocopherol succinate for the treatment pf diabetic retinopathy. J Drug Deliv Sci Technol 2021; 61: 102286.
[http://dx.doi.org/10.1016/j.jddst.2020.102286]
[54]
Azadi R, Mousavi SE, Kazemi NM, Rezayat SM, Jaafari MR. Preparation and characterization of berberine loaded micelle formulations with approach to oral oral drug delivery. Trends Pharmacol Sci 2020; 6: 255.
[55]
Bahman F, Taurin S, Altayeb D, Taha S, Bakhiet M, Greish K. Oral insulin delivery using poly (styrene co-maleic acid) micelles in a diabetic mouse model. Pharmaceutics 2020; 12(11): 1026.
[http://dx.doi.org/10.3390/pharmaceutics12111026] [PMID: 33120872]
[56]
Wen N, Lü S, Xu X, et al. A polysaccharide-based micelle-hydrogel synergistic therapy system for diabetes and vascular diabetes complications treatment. Mater Sci Eng C 2019; 100: 94-103.
[http://dx.doi.org/10.1016/j.msec.2019.02.081] [PMID: 30948130]
[57]
Kamerova K, Haladjova E, Grancharov G, et al. Co-assembly of block copolymers as a tool for developing novel micellar carriers of insulin for controlled drug delivery. Eur Polym J 2018; 104: 1.
[http://dx.doi.org/10.1016/j.eurpolymj.2018.04.039]
[58]
Subramani K, Pathak S, Hosseinkhani H. Recent trends in diabetes treatment using nanotechnology. Dig J Nanomater Biostruct 2012; 7(1): 85-95.
[59]
Kumria R, Goomber G. Emerging trends in insulin delivery: Buccal route. J Diabetol 2011; 2(2): 5.
[60]
Hu C, Jia W. Therapeutic medications against diabetes: What we have and what we expect. Adv Drug Deliv Rev 2019; 139: 3-15.
[http://dx.doi.org/10.1016/j.addr.2018.11.008] [PMID: 30529309]
[61]
Veuillez F, Kalia YN, Jacques Y, Deshusses J, Buri P. Factors and strategies for improving buccal absorption of peptides. Eur J Pharm Biopharm 2001; 51(2): 93-109.
[http://dx.doi.org/10.1016/S0939-6411(00)00144-2] [PMID: 11226816]
[62]
Carino GP, Mathiowitz E. Oral insulin delivery. Adv Drug Deliv Rev 1999; 35(2-3): 249-57.
[http://dx.doi.org/10.1016/S0169-409X(98)00075-1] [PMID: 10837700]
[63]
Arya A, Kumar L, Pokharia DB, Tripathi K. Applications of nanotechnology in diabetes. Dig J Nanomater Biostruct 2008; 3: 221.
[64]
Woldu MA, Lenjisa JL. Nanoparticles and the new area in diabetes management. Int J Basic Clin Pharmacol 2014; 3: 277.
[http://dx.doi.org/10.5455/2319-2003.ijbcp20140405]
[65]
Souto EB, Souto SB, Campos JR, et al. Nanoparticle delivery systems in the treatment of diabetes complications. Molecules 2019; 24(23): 4209.
[http://dx.doi.org/10.3390/molecules24234209] [PMID: 31756981]
[66]
Khan Ghilzai NM. New developments in insulin delivery. Drug Dev Ind Pharm 2003; 29(3): 253-65.
[http://dx.doi.org/10.1081/DDC-120018199] [PMID: 12741607]
[67]
Bhumkar DR, Joshi HM, Sastry M, Pokharkar VB. Chitosan reduced gold nanoparticles as novel carriers for transmucosal delivery of insulin. Pharm Res 2007; 24(8): 1415-26.
[http://dx.doi.org/10.1007/s11095-007-9257-9] [PMID: 17380266]
[68]
van der Lubben IM, Verhoef JC, Borchard G, Junginger HE. Chitosan for mucosal vaccination. Adv Drug Deliv Rev 2001; 52(2): 139-44.
[http://dx.doi.org/10.1016/S0169-409X(01)00197-1] [PMID: 11718937]
[69]
Bariya SH, Gohel MC, Mehta TA, Sharma OP. Microneedles: An emerging transdermal drug delivery system. J Pharm Pharmacol 2012; 64(1): 11-29.
[http://dx.doi.org/10.1111/j.2042-7158.2011.01369.x] [PMID: 22150668]
[70]
Yu W, Jiang G, Zhang Y, Liu D, Xu B, Zhou J. Polymer microneedles fabricated from alginate and hyaluronate for transdermal delivery of insulin. Mater Sci Eng C 2017; 80: 187-96.
[http://dx.doi.org/10.1016/j.msec.2017.05.143] [PMID: 28866156]
[71]
Seong KY, Seo MS, Hwang DY, et al. A self-adherent, bullet-shaped microneedle patch for controlled transdermal delivery of insulin. J Control Release 2017; 265: 48-56.
[http://dx.doi.org/10.1016/j.jconrel.2017.03.041] [PMID: 28344013]
[72]
Kashyap D, Tuli HS, Yerer MB, et al. Natural product-based nanoformulations for cancer therapy: Opportunities and challenges. Semin Cancer Biol 2021; 69: 5-23.
[http://dx.doi.org/10.1016/j.semcancer.2019.08.014] [PMID: 31421264]
[73]
Oh Yoon Sin. Plant derived compounds targeting pancreatic beta cells for the treatment of diabetes. Evid Based Complement Alternat Med 2015; 1: 1.
[74]
Rouse M, Younès A, Egan JM. Resveratrol and curcumin enhance pancreatic β-cell function by inhibiting phosphodiesterase activity. J Endocrinol 2014; 223(2): 107-17.
[http://dx.doi.org/10.1530/JOE-14-0335] [PMID: 25297556]
[75]
Singh AK, Raj V, Keshari AK, et al. Isolated mangiferin and naringenin exert antidiabetic effect via PPARα/GLUT4 dual agonistic action with strong metabolic regulation. Chem Biol Interact 2018; 280: 33-44.
[http://dx.doi.org/10.1016/j.cbi.2017.12.007] [PMID: 29223569]
[76]
Ekar T, Kreft S. Common risks of adulterated and mislabeled herbal preparations. Food Chem Toxicol 2019; 123: 288-97.
[http://dx.doi.org/10.1016/j.fct.2018.10.043] [PMID: 30339960]
[77]
Adeniyi A, Asase A, Ekpe PK, Asitoakor BK, Adu-Gyamfi A, Avekor PY. Ethnobotanical study of medicinal plants from Ghana; confirmation of ethnobotanical uses, and review of biological and toxicological studies on medicinal plants used in Apra Hills Sacred Grove. J Herb Med 2018; 14: 76.
[http://dx.doi.org/10.1016/j.hermed.2018.02.001]
[78]
Modak M, Dixit P, Londhe J, Ghaskadbi S, Devasagayam TPA. Indian herbs and herbal drugs used for the treatment of diabetes. J Clin Biochem Nutr 2007; 40(3): 163-73.
[http://dx.doi.org/10.3164/jcbn.40.163] [PMID: 18398493]
[79]
Anand K, Tiloke C, Naidoo P, Chuturgoon AA. Phytonanotherapy for management of diabetes using green synthesis nanoparticles. J Photochem Photobiol B 2017; 173: 626-39.
[http://dx.doi.org/10.1016/j.jphotobiol.2017.06.028] [PMID: 28709077]
[80]
Khursheed R, Singh SK, Wadhwa S, et al. Treatment strategies against diabetes: Success so far and challenges ahead. Eur J Pharmacol 2019; 862: 172625.
[http://dx.doi.org/10.1016/j.ejphar.2019.172625] [PMID: 31449807]
[81]
Allam AN, Komeil IA, Fouda MA, Abdallah OY. Preparation, characterization and in vivo evaluation of curcumin self-nano phospholipid dispersion as an approach to enhance oral bioavailability. Int J Pharm 2015; 489(1-2): 117-23.
[http://dx.doi.org/10.1016/j.ijpharm.2015.04.067] [PMID: 25936626]
[82]
Gouda W, Hafiz NA, Mageed L, et al. Effects of nano-curcumin on gene expression of insulin and insulin receptor. Bull Natl Res Cent 2019; 43: 128.
[http://dx.doi.org/10.1186/s42269-019-0164-0]
[83]
Wang Y, Tao B, Wan Y, et al. Drug delivery based pharmacological enhancement and current insights of quercetin with therapeutic potential against oral diseases. Biomed Pharmacother 2020; 128: 110372.
[http://dx.doi.org/10.1016/j.biopha.2020.110372] [PMID: 32521458]
[84]
Chitkara D, Nikalaje SK, Mittal A, Chand M, Kumar N. Development of quercetin nanoformulation and in vivo evaluation using streptozotocin induced diabetic rat model. Drug Deliv Transl Res 2012; 2(2): 112-23.
[http://dx.doi.org/10.1007/s13346-012-0063-5] [PMID: 25786720]
[85]
Alshehri SM, Shakeel F, Ibrahim MA, et al. Dissolution and bioavailability improvement of bioactive apigenin using solid dispersions prepared by different techniques. Saudi Pharm J 2019; 27(2): 264-73.
[http://dx.doi.org/10.1016/j.jsps.2018.11.008] [PMID: 30766439]
[86]
Zhang Z, Cui C, Wei F, Lv H. Improved solubility and oral bioavailability of apigenin via Soluplus/Pluronic F127 binary mixed micelles system. Drug Dev Ind Pharm 2017; 43(8): 1276-82.
[http://dx.doi.org/10.1080/03639045.2017.1313857] [PMID: 28358225]
[87]
Ahangarpour A, Oroojan AA, Khorsandi L, Kouchak M, Badavi M. Solid lipid nanoparticles of myricitrin have antioxidant and antidiabetic effects on streptozotocin-nicotinamide-induced diabetic model and myotube cell of male mouse. Oxid Med Cell Longev 2018; 2018: 7496936.
[http://dx.doi.org/10.1155/2018/7496936] [PMID: 30116491]
[88]
Dang H, Hasan M, Zhao H, et al. Luteolin-loaded solid lipid nanoparticles synthesis, characterization, & improvement of bioavailability, pharmacokinetics in vitro and vivo studies. J Nanopart Res 2014; 16: 2347.
[http://dx.doi.org/10.1007/s11051-014-2347-9]
[89]
Samadarsi R, Dutta D. Design and characterization of Mangiferin nanoparticles for oral delivery. J Food Eng 2018; 247: 80-94.
[http://dx.doi.org/10.1016/j.jfoodeng.2018.11.020]
[90]
Gupta L, Sharma AK, Gothwal A, et al. Dendrimer encapsulated and conjugated delivery of berberine: A novel approach mitigating toxicity and improving in vivo pharmacokinetics. Int J Pharm 2017; 528(1-2): 88-99.
[http://dx.doi.org/10.1016/j.ijpharm.2017.04.073] [PMID: 28533175]
[91]
He Y, Al-Mureish A, Wu N. Nanotechnology in the treatment of diabetic complications: A comprehensive narrative review. J Diabetes Res 2021; 2021: 6612063.
[http://dx.doi.org/10.1155/2021/6612063] [PMID: 34007847]
[92]
Veiseh O, Tang BC, Whitehead KA, Anderson DG, Langer R. Managing diabetes with nanomedicine: Challenges and opportunities. Nat Rev Drug Discov 2015; 14(1): 45-57.
[http://dx.doi.org/10.1038/nrd4477] [PMID: 25430866]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy