Generic placeholder image

Current Pharmaceutical Design

Editor-in-Chief

ISSN (Print): 1381-6128
ISSN (Online): 1873-4286

General Review Article

Therapeutic Advancements in the Management of Diabetes Mellitus with Special Reference to Nanotechnology

Author(s): Pallavi Singh Chauhan, Dhananjay Yadav, Shivam Tayal and Jun-O Jin*

Volume 26, Issue 38, 2020

Page: [4909 - 4916] Pages: 8

DOI: 10.2174/1381612826666200826135401

Price: $65

Abstract

For improvisation of diabetic’s quality of life, nanotechnology is facilitating the development of advanced glucose sensors as well as efficient insulin delivery systems. Our prime focus of the review is to highlight the advancement in diabetic research with special reference to nanotechnology at its interface. Recent studies are more focused on enhancing sensitivity, accuracy, and response by employing metal as well as nanoparticles based glucose sensors. Moreover, the review focuses on nanoscale based approaches i.e. closed-loop insulin delivery systems, which detect any fluctuation in blood glucose levels and allow controlled release of a drug, thus are also called self-regulating insulin release system. Additionally, this review summarizes the role of nanotechnology in the diagnosis and treatment of diabetic complications through little advancement in the existing techniques. To improve health, as well as the quality of life in diabetic’s new sensing systems for blood glucose level evaluation and controlled administration of drugs through efficient drug delivery systems should be explored.

Keywords: Diabetes mellitus, nanotechnology, blood glucose sensors, nano drug therapy, diabetic complications, sensitivity.

[1]
Noh J, Han K-D, Ko S-H, Ko KS, Park C-Y. Trends in the pervasiveness of type 2 diabetes, impaired fasting glucose and co-morbidities during an 8-year-follow-up of nationwide Korean population. Sci Rep 2017; 7(1): 46656.
[http://dx.doi.org/10.1038/srep46656]
[2]
Tabish SA. Is diabetes becoming the biggest epidemic of the twenty-first century? Int J Health Sci (Qassim) 2007; 1(2): V-VIII.
[3]
Marín-Peñalver JJ, Martín-Timón I, Sevillano-Collantes C. del Cañizo-Gómez FJJWjod. Update on the treatment of type 2 diabetes mellitus. World J Diabetes 2016; 15; 7(17): 354-95.
[http://dx.doi.org/10.4239/wjd.v7.i17.354]
[4]
Cade WT. Diabetes-related microvascular and macrovascular diseases in the physical therapy setting. Phys Ther 2008; 88(11): 1322-35.
[http://dx.doi.org/10.2522/ptj.20080008]
[5]
Chawla A, Chawla R, Jaggi S. Microvasular and macrovascular complications in diabetes mellitus: Distinct or continuum? Indian J Endocrinol Metab 2016; 20(4): 546-51.
[http://dx.doi.org/10.4103/2230-8210.183480]
[6]
Saeedi P, Petersohn I, Salpea P, et al. Global and regional diabetes prevalence estimates for 2019 and projections for 2030 and 2045: Results from the International Diabetes Federation Diabetes Atlas, 9(th) edition. Diabetes Res Clin Pract 2019; 157.
[7]
Sherwani SI, Khan HA, Ekhzaimy A, Masood A, Sakharkar MK. Significance of HbA1c test in diagnosis and prognosis of diabetic patients. Biomark Insights 2016; 11: 95-104.
[http://dx.doi.org/10.4137/BMI.S38440]
[8]
American Diabetes Association Economic Costs of Diabetes in the U.S. in 2017. Diabetes Care 2018; 41(5): 917-28.
[http://dx.doi.org/10.2337/dci18-0007]
[9]
Siwach R, Pandey P, Chawla V, Dureja H. Role of nanotechnology in diabetic management. Recent Pat Nanotechnol 2019; 13(1): 28-37.
[http://dx.doi.org/10.2174/1872210513666190104122032]
[10]
Samadder A. Khuda-Bukhsh ARJWJoTM. Nanotechnological approaches in diabetes treatment: A new horizon. World J Transl Med 2014; 3(2): 84.
[http://dx.doi.org/10.5528/wjtm.v3.i2.84]
[11]
Patra JK, Das G, Fraceto LF, et al. Nano based drug delivery systems: recent developments and future prospects. J Nanobiotechnology 2018; 16(1): 71-1.
[http://dx.doi.org/10.1186/s12951-018-0392-8]
[12]
Blair JC, McKay A, Ridyard C, et al. Continuous subcutaneous insulin infusion versus multiple daily injection regimens in children and young people at diagnosis of type 1 diabetes: pragmatic randomised controlled trial and economic evaluation. BMJ 2019; 365: 1226.
[http://dx.doi.org/10.1136/bmj.l1226]
[13]
Ellis K, Mulnier H, Forbes A. Perceptions of insulin use in type 2 diabetes in primary care: a thematic synthesis. BMC Fam Pract 2018; 19(1): 70.
[http://dx.doi.org/10.1186/s12875-018-0753-2]
[14]
Krzymien J, Ladyzynski P. Insulin in type 1 and type 2 diabetes-should the dose of insulin before a meal be based on glycemia or meal content? Nutrients 2019; 11(3): 607.
[http://dx.doi.org/10.3390/nu11030607]
[15]
Silver B, Ramaiya K, Andrew SB, et al. EADSG guidelines: Insulin therapy in diabetes. Diabetes Ther 2018; 9(2): 449-92.
[http://dx.doi.org/10.1007/s13300-018-0384-6]
[16]
Mbanya JC, Sandow J, Landgraf W, Owens DR. Recombinant human insulin in global diabetes management - focus on clinical efficacy. Eur Endocrinol 2017; 13(1): 21-5.
[http://dx.doi.org/10.17925/EE.2017.13.01.21]
[17]
Tandon N, Kalra S, Balhara YPS, et al. Forum for injection technique and therapy expert recommendations, India: the Indian recommendations for best practice in insulin injection technique. Indian J Endocrinol Metab 2017; 21(4): 600-17.
[http://dx.doi.org/10.4103/ijem.IJEM_97_17]
[18]
Bahendeka S, Kaushik R, Swai AB, et al. EADSG guidelines: insulin storage and optimisation of injection technique in diabetes management. Diabetes Ther 2019; 10(2): 341-66.
[http://dx.doi.org/10.1007/s13300-019-0574-x]
[19]
Church TJ, Haines ST. Treatment approach to patients with severe insulin resistance. Clin Diabetes 2016; 34(2): 97-104.
[http://dx.doi.org/10.2337/diaclin.34.2.97]
[20]
Weaver KW, Hirsch IB. The hybrid closed-loop system: evolution and practical applications. Diabetes Technol Ther 2018; 20(S2): S216-23.
[http://dx.doi.org/10.1089/dia.2018.0091]
[21]
Pfeiffer AF, Klein HH. The treatment of type 2 diabetes. Dtsch Arztebl Int 2014; 111(5): 69-81.
[22]
Ng LC, Gupta M. Transdermal drug delivery systems in diabetes management: A review. Asian J Pharm Sci 2019; 15(1): 13-35.
[23]
Feng Q, Liu Y, Huang J, Chen K, Huang J, Xiao K. Uptake, distribution, clearance, and toxicity of iron oxide nanoparticles with different sizes and coatings. Sci Rep 2018; 8(1): 2082.
[http://dx.doi.org/10.1038/s41598-018-19628-z]
[24]
Wang GS, Hoyte C. Review of Biguanide (Metformin) Toxicity. J Intensive Care Med 2018; 34(11-12): 863-76.
[25]
Harsch IA, Kaestner RH, Konturek PC. Hypoglycemic side effects of sulfonylureas and repaglinide in ageing patients - knowledge and self-management. J Physiol Pharmacol 2018; 69(4)
[26]
Han Y, Gao Z, Chen L, et al. Multifunctional oral delivery systems for enhanced bioavailability of therapeutic peptides/proteins. Acta Pharm Sin B 2019; 9(5): 902-22.
[http://dx.doi.org/10.1016/j.apsb.2019.01.004]
[27]
Maideen NMP. Thiazolidinediones and their Drug Interactions involving CYP enzymes American J Physiol Biochem Pharmacol 2018; 8(2)
[http://dx.doi.org/10.5455/ajpbp.20181022083057]
[28]
Bradley C. The glitazones: a new treatment for type 2 diabetes mellitus. Intensive Crit Care Nurs 2002; 18(3): 189-91.
[http://dx.doi.org/10.1016/S0964-3397(02)00010-1]
[29]
Marc G, Stana A, Oniga SD, Pirnau A, Vlase L, Oniga O. New phenolic derivatives of thiazolidine-2,4-dione with antioxidant and antiradical properties: synthesis, characterization, in vitro evaluation, and quantum Studies. Molecules 2019; 24(11)
[http://dx.doi.org/10.3390/molecules24112060]
[30]
Shahinozzaman M, Taira N, Ishii T, Halim MA, Hossain MA, Tawata S. Anti-inflammatory, anti-diabetic, and anti-alzheimer’s effects of prenylated flavonoids from okinawa propolis: an investigation by experimental and computational studies. Molecules 2018; 23(10): 2479.
[http://dx.doi.org/10.3390/molecules23102479]
[31]
Hsia DS, Grove O, Cefalu WT. An update on sodium-glucose co-transporter-2 inhibitors for the treatment of diabetes mellitus. Curr Opin Endocrinol Diabetes Obes 2017; 24(1): 73-9.
[32]
Beitelshees AL, Leslie BR, Taylor SI. Sodium-glucose cotransporter 2 inhibitors: a case study in translational research. Diabetes 2019; 68(6): 1109-20.
[http://dx.doi.org/10.2337/dbi18-0006]
[33]
Hinnen D. Glucagon-like peptide 1 receptor agonists for type 2 diabetes. Diabetes Spectr 2017; 30(3): 202-10.
[http://dx.doi.org/10.2337/ds16-0026.]
[34]
Wright LA, Hirsch IB. Non-insulin treatments for Type 1 diabetes: critical appraisal of the available evidence and insight into future directions. Diabet Med 2019; 36(6): 665-78.
[http://dx.doi.org/10.1111/dme.13941]
[35]
Mendes RG, Wróbel PS, Bachmatiuk A, et al. Carbon Nanostructures as a Multi-Functional Platform for Sensing Applications. Chemosensors 2018; 6(4): 60.
[36]
Chauhan PS, Shrivastava V, Tomar RS. Bio fabrication of copper nanoparticles: a next generation antibacterial agent against wound associated pathogens. Turk J Pharm Sci 2018; 15(3): 238-47.
[http://dx.doi.org/10.4274/tjps.52724]
[37]
Shrivastava V, Chauhan PS. Tomar RSJWJoP, Sciences P. Nanobiotechnology: A potential tool for biomedics. World J Pharmacy Pharm Sci 2015; 4(5): 1929-43.
[38]
Shrivastava V, Chauhan PS, Tomar RS. A biomimetic approach for synthesis of silver nanoparticles using Murraya paniculata leaf extract with reference to antimicrobial activity. J Pharm Sci Res 2016; 8(4): 247.
[39]
Rizvi SA, Saleh AMJSPJ. Applications of nanoparticle systems in drug delivery technology. Saudi Pharm J 2018; 26(1): 64-70.
[http://dx.doi.org/10.1016/j.jsps.2017.10.012]
[40]
Artigues M, Abellà J, Colominas SJS. Analytical parameters of an amperometric glucose biosensor for fast analysis in food samples. Sensors (Basel) 2017; 17(11): 2620.
[41]
Chen C, Zhao X-L, Li Z-H, Zhu Z-G, Qian S-H, Flewitt AJS. Current and emerging technology for continuous glucose monitoring. Sensors (Basel) 2017; 17(1): 182.
[http://dx.doi.org/10.3390/s17010182]
[42]
Zhou Y, Fang Y, Ramasamy RPJS. Non-covalent functionalization of carbon nanotubes for electrochemical biosensor development. Sensors (Basel) 2019; 19(2): 392.
[http://dx.doi.org/10.3390/s19020392]
[43]
Wang H-C, Lee A-R. Recent developments in blood glucose sensors. J Food Drug Analysis 2015; 23(2): 191-200.
[44]
Bihar E, Wustoni S, Pappa AM, Salama KN, Baran D. Inal SJnFE. A fully inkjet-printed disposable glucose sensor on paper. Npj Flex Elec 2018; 2(1): 1-8.
[http://dx.doi.org/10.1038/s41528-018-0044-y]
[45]
Kim J, Campbell AS, Wang JJT. Wearable non-invasive epidermal glucose sensors. A review. Talanta 2018; 177: 163-70.
[46]
Javid B, Fotouhi-Ghazvini F, Zakeri FS. Noninvasive optical diagnostic techniques for mobile blood glucose and bilirubin monitoring. J Med Signals Sens 2018; 8(3): 125-39.
[http://dx.doi.org/10.4103/jmss.JMSS_8_18]]
[47]
Chen Y, Lu S, Zhang S, et al. Skin-like biosensor system via electrochemical channels for noninvasive blood glucose monitoring. Sci Adv 2017; 3(12): e1701629.
[http://dx.doi.org/10.1126/sciadv.1701629]
[48]
Loiseau A, Asila V, Boitel-Aullen G, Lam M, Salmain M, Boujday SJB. Silver-based plasmonic nanoparticles for and their use in biosensing. Biosensors 2019; 9(2): 78.
[49]
Haxha S, Jhoja JJIPJ. Optical based noninvasive glucose monitoring sensor prototype. IEEE Photonics J 2016; 8(6): 1-11.
[http://dx.doi.org/10.1109/JPHOT.2016.2616491]
[50]
Villena Gonzales W, Mobashsher AT, Abbosh A. The progress of glucose monitoring-a review of invasive to minimally and non-invasive techniques, devices and sensors. Sensors (Basel) 2019; 19(4): 800.
[http://dx.doi.org/10.3390/s19040800]
[51]
Ghanbari R, Safaiee R, Sheikhi MH, Golshan MM. Horastani ZKJAm. Interfaces. Graphene decorated with silver nanoparticles as a low-temperature methane gas sensor. ACS Applied Mater Interfaces 2019; 11(24): 21795-806.
[52]
Huang H, Su S, Wu N, et al. Graphene-Based Sensors for Human Health Monitoring. Front Chem 2019; 7: 399.
[http://dx.doi.org/10.3389/fchem.2019.00399]
[53]
Shanbhag VKL, Prasad KS. Graphene based sensors in the detection of glucose in saliva - a promising emerging modality to diagnose diabetes mellitus. Anal Methods 2016; 8(33): 6255-9.
[http://dx.doi.org/10.1039/C6AY01023G]
[54]
Nasir S, Hussein MZ, Zainal Z, Yusof NA. Carbon-based nanomaterials/allotropes: a glimpse of their synthesis, properties and some applications. Materials (Basel) 2018; 11(2)
[http://dx.doi.org/10.3390/ma11020295]
[55]
Dungan K, Verma N. Monitoring technologies-continuous glucose monitoring, mobile technology, biomarkers of glycemic control. 2018. In: Endotext. MDText. com, Inc. 2018.
[56]
Hamouda RA, Hussein MH, Abo-elmagd RA, Bawazir SS. Synthesis and biological characterization of silver nanoparticles derived from the cyanobacterium Oscillatoria limnetica. Sci Rep 2019; 9(1): 13071.
[http://dx.doi.org/10.1038/s41598-019-49444-y]
[57]
Heo YJ, Kim S-H. Toward long-term implantable glucose biosensors for clinical use. Applied Sciences 2019; 9(10): 2158.
[58]
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]
[59]
Wakaskar RR. General overview of lipid-polymer hybrid nanoparticles, dendrimers, micelles, liposomes, spongosomes and cubosomes. J Drug Target 2018; 26(4): 311-8.
[http://dx.doi.org/10.1080/1061186X.2017.1367006]]
[60]
Gharbavi M, Amani J, Kheiri-Manjili H, Danafar H, Sharafi A. Niosome: A Promising Nanocarrier for Natural Drug Delivery through Blood-Brain Barrier. Advance in Pharmacological Sciences 2018; 2018: 6847971.
[http://dx.doi.org/10.1155/2018/6847971]
[61]
Sharma G, Sharma AR, Nam J-S, Doss GPC, Lee S-S, Chakraborty C. Nanoparticle based insulin delivery system: the next generation efficient therapy for Type 1 diabetes. J Nanobiotechnology 2015; 13(1): 74.
[http://dx.doi.org/10.1186/s12951-015-0136-y]
[62]
Mansoor S, Kondiah PPD, Choonara YE, Pillay V. Polymer-based nanoparticle strategies for insulin delivery. Polymers (Basel) 2019; 11(9): 1380.
[http://dx.doi.org/10.3390/polym11091380]
[63]
Wu ZM, Ling L, Zhou LY, et al. Novel preparation of PLGA/HP55 nanoparticles for oral insulin delivery. Nanoscale Res Lett 2012; 7(1): 299-9.
[http://dx.doi.org/10.1186/1556-276X-7-299]
[64]
Sarmento B, Ribeiro A, Veiga F, Sampaio P, Neufeld R, Ferreira D. Alginate/chitosan nanoparticles are effective for oral insulin delivery. Pharm Res 2007; 24(12): 2198-206.
[http://dx.doi.org/10.1007/s11095-007-9367-4]
[65]
Lushchak O, Zayachkivska A, Vaiserman A. Metallic nanoantioxidants as potential therapeutics for type 2 diabetes: a hypothetical background and translational perspectives. Oxid Med Cell Longev 2018; 2018: 9.
[http://dx.doi.org/10.1155/2018/3407375]
[66]
Chowdhury A, Kunjiappan S, Panneerselvam T, Somasundaram B, Bhattacharjee CJINL. Nanotechnology and nanocarrier-based approaches on treatment of degenerative diseases. Int Nano Lett 2017; 7: 91-122.
[http://dx.doi.org/10.1007/s40089-017-0208-0]
[67]
Zhao L, Xiao C, Wang L, Gai G, Ding J. Glucose-sensitive polymer nanoparticles for self-regulated drug delivery. Chem Commun (Camb) 2016; 52(49): 7633-52.
[http://dx.doi.org/10.1039/C6CC02202B]
[68]
Subramani K, Pathak S, Hosseinkhani H. Recent trends in diabetes treatment using nanotechnology. Digest J Nanomaterial Biostructures 2012; 7(1): 85-95.
[69]
Hu S, De Vos P. Polymeric approaches to reduce tissue responses against devices applied for islet-cell encapsulation. Front Bioeng Biotechnol 2019; 7: 134.
[70]
Tomar RS, Chauhan PS, Shrivastava VJW. A critical review on nanoparticle synthesis: physicochemical v/s biological approach. 2014; 4(1): 595-620.
[71]
Zhi Z-L, Khan F, Pickup JC. Multilayer nanoencapsulation: A nanomedicine technology for diabetes research and management. Diab Res Clin Pract 2013; 100(2): 162-9.
[http://dx.doi.org/10.1016/j.diabres.2012.11.027]
[72]
Wang J, Yu J, Zhang Y, et al. Charge-switchable polymeric complex for glucose-responsive insulin delivery in mice and pigs. Sci Adv 2019; 5(7): eaaw4357.
[http://dx.doi.org/10.1126/sciadv.aaw4357]
[73]
Raliya R, Saha D, Chadha TS, Raman B. Biswas PJSr. Non-invasive aerosol delivery and transport of gold nanoparticles to the brain. Sci Reports 2017; 7: 44718.
[http://dx.doi.org/10.1038/srep44718]
[74]
Bruen D, Delaney C, Florea L, Diamond DJS. Glucose sensing for diabetes monitoring: recent developments. Sensors 2017; 17(8): 1866.
[http://dx.doi.org/10.3390/s17081866]
[75]
Gu Z, Aimetti AA, Wang Q, et al. Injectable nano-network for glucose-mediated insulin delivery. ACS Nano 2013; 7(5): 4194-201.
[http://dx.doi.org/10.1021/nn400630x]
[76]
Yellanki SK, Singh J, Nerella NK, Deb SK, Goranti SJRJP. Technology. Nanotechnology for Poorly Soluble Drugs. Res J Pharm Technol 2010; 3(3): 688-93.
[77]
Cavalli E, Mammana S, Nicoletti F, Bramanti P, Mazzon E. The neuropathic pain: An overview of the current treatment and future therapeutic approaches. Int J Immunopathol Pharmacol 2019; 33: 2058738419838383.
[78]
Park J, Ramanathan R, Pham L, Woodrow KA. Chitosan enhances nanoparticle delivery from the reproductive tract to target draining lymphoid organs. Nanomedicine 2017; 13(6): 2015-25.
[http://dx.doi.org/10.1016/j.nano.2017.04.012]
[79]
Upadhyay RK. Drug delivery systems, CNS protection, and the blood brain barrier. BioMed Res Int 2014; 2014: 869269-9.
[http://dx.doi.org/10.1155/2014/869269]
[80]
Wufuer M, Lee G, Hur W, et al. Skin-on-a-chip model simulating inflammation, edema and drug-based treatment. Sci Rep 2016; 6(1): 37471.
[http://dx.doi.org/10.1038/srep37471]
[81]
Zhang Q, Sito L, Mao M, He J, Zhang YS, Zhao XJMS. Current advances in skin-on-a-chip models for drug testing Microphysiol Sys 2018; 2.
[http://dx.doi.org/10.21037/mps.2018.08.01]
[82]
Gallego-Perez D, Pal D, Ghatak S, et al. Topical tissue nano-transfection mediates non-viral stroma reprogramming and rescue. Nat Nanotechnol 2017; 12(10): 974-9.
[http://dx.doi.org/10.1038/nnano.2017.134]
[83]
Nemati S, Kim S-j, Shin YM, Shin H. Current progress in application of polymeric nanofibers to tissue engineering. Nano Converg 2019; 6(1): 36.
[http://dx.doi.org/10.1186/s40580-019-0209-y]
[84]
Vasita R, Katti DS. Nanofibers and their applications in tissue engineering. Int J Nanomedicine 2006; 1(1): 15-30.
[http://dx.doi.org/10.2147/nano.2006.1.1.15]
[85]
Brownlee M. The pathobiology of diabetic complications: a unifying mechanism. Diabetes 2005; 54(6): 1615-25.
[http://dx.doi.org/10.2337/diabetes.54.6.1615]
[86]
Chakkarwar VA. Smoking in diabetic nephropathy: sparks in the fuel tank? World J Diabetes 2012; 3(12): 186-95.
[http://dx.doi.org/10.4239/wjd.v3.i12.186]
[87]
Kamaly N, He JC, Ausiello DA, Farokhzad OC. Nanomedicines for renal disease: current status and future applications. Nat Rev Nephrol 2016; 12(12): 738-53.
[http://dx.doi.org/10.1038/nrneph.2016.156]
[88]
Alomari G, Al-Trad B, Hamdan S, et al. Gold nanoparticles attenuate albuminuria by inhibiting podocyte injury in a rat model of diabetic nephropathy. Drug Deliv Transl Res 2019; 10(1): 216-26.
[89]
Jahani M, Shokrzadeh M, Vafaei-Pour Z, Zamani E. Shaki FJAJoA, Advances V. Potential role of cerium oxide nanoparticles for attenuation of diabetic nephropathy by inhibition of oxidative damage. Asian J Animal Veteinary Adv 2016; 11(4): 226-34.
[90]
Jahangirian H, Lemraski EG, Webster TJ, Rafiee-Moghaddam R, Abdollahi Y. A review of drug delivery systems based on nanotechnology and green chemistry: green nanomedicine. Int J Nanomedicine 2017; 12: 2957-78.
[http://dx.doi.org/10.2147/IJN.S127683]
[91]
Brede C, Labhasetwar V. Applications of nanoparticles in the detection and treatment of kidney diseases. Adv Chronic Kidney Dis 2013; 20(6): 454-65.
[http://dx.doi.org/10.1053/j.ackd.2013.07.006]
[92]
Fangueiro JF, Silva AM, Garcia ML, Souto EB. Current nanotechnology approaches for the treatment and management of diabetic retinopathy Eur J Biopharm 2015; 95(Pt B): 307-22.
[http://dx.doi.org/10.1016/j.ejpb.2014.12.023]
[93]
Feng W, Shi R, Zhang C, Liu S, Yu T, Zhu D. Visualization of skin microvascular dysfunction of type 1 diabetic mice using in vivo skin optical clearing method. J Biomed Optics 2018; 24(3): 031003.
[94]
Kim JH, Kim MH, Jo DH, Yu YS, Lee TG, Kim JH. The inhibition of retinal neovascularization by gold nanoparticles via suppression of VEGFR-2 activation. Biomaterials 2011; 32(7): 1865-71.
[http://dx.doi.org/10.1016/j.biomaterials.2010.11.030]
[95]
Saeed BA, Lim V, Yusof NA, Khor KZ, Rahman HS, Abdul Samad N. Antiangiogenic properties of nanoparticles: a systematic review. Int J Nanomedicine 2019; 14: 5135-46.
[http://dx.doi.org/10.2147/IJN.S199974]
[96]
Zhu S, Gong L, Li Y, Xu H, Gu Z, Zhao Y. Safety assessment of nanomaterials to eyes: an important but neglected issue. Adv Sci (Weinh) 2019; 6(16): 1802289-9.
[http://dx.doi.org/10.1002/advs.201802289]
[97]
Lombardo D, Kiselev MA, Caccamo MT. Smart nanoparticles for drug delivery application: development of versatile nanocarrier platforms in biotechnology and nanomedicine. J Nanomater 2019; 2019: 26.
[http://dx.doi.org/10.1155/2019/3702518]
[98]
Tsai CH, Wang PY, Lin IC, Huang H, Liu GS, Tseng CL. Ocular drug delivery: role of degradable polymeric nanocarriers for ophthalmic application. Int J Mol Sci 2018; 19(9)
[http://dx.doi.org/10.3390/ijms19092830]

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