571
Views
25
CrossRef citations to date
0
Altmetric
Review

Potential Therapeutic Usage of Nanomedicine for Glaucoma Treatment

, , ORCID Icon & ORCID Icon
Pages 5745-5765 | Published online: 06 Aug 2020

References

  • Quigley HA, Broman AT. The number of people with glaucoma worldwide in 2010 and 2020. Br J Ophthalmol. 2006;90(3):262–267. doi:10.1136/bjo.2005.08122416488940
  • Weinreb RN, Aung T, Medeiros FAJJ. The pathophysiology and treatment of glaucoma: a review. Jama. 2014;311(18):1901–1911.24825645
  • Richter GM, Coleman AL. Minimally invasive glaucoma surgery: current status and future prospects. Clin Ophthalmol. 2016;10:189–206. doi:10.2147/OPTH.S8049026869753
  • Schlunck G, Meyer-ter-Vehn T, Klink T, Grehn F. Conjunctival fibrosis following filtering glaucoma surgery. Exp Eye Res. 2016;142:76–82. doi:10.1016/j.exer.2015.03.02126675404
  • Bengtsson B, Heijl A. Lack of visual field improvement after initiation of intraocular pressure reducing treatment in the early manifest glaucoma trial. Invest Ophthalmol Vis Sci. 2016;57(13):5611–5615. doi:10.1167/iovs.16-1938927768797
  • Song YK, Lee CK, Kim J, Hong S, Kim CY, Seong GJ. Instability of 24-hour intraocular pressure fluctuation in healthy young subjects: a prospective, cross-sectional study. BMC Ophthalmol. 2014;14(1):127. doi:10.1186/1471-2415-14-12725370361
  • Grant WM, Burke JF Jr. Why do some people go blind from glaucoma? Ophthalmology. 1982;89(9):991–998. doi:10.1016/S0161-6420(82)34675-87177577
  • Susanna R, De Moraes CG, Cioffi GA. Ritch RJTvs, technology. Why do people (still) go blind from glaucoma? Trans vision sci technol. 2015;4(2):1.
  • Asiedu K, Dzasimatu SK, Kyei SJCL, Eye A. Clinical subtypes of dry eye in youthful clinical sample in Ghana. Contact Lens Anterior Eye. 2019;42(2):206–211.30337142
  • Smith DM, Simon JK, Baker Jr JR Jr. Applications of nanotechnology for immunology. Nat Rev Immunol. 2013;13(8):592–605. doi:10.1038/nri348823883969
  • Chow EK, Ho D. Cancer nanomedicine: from drug delivery to imaging. Sci Transl Med. 2013;5(216):216rv214. doi:10.1126/scitranslmed.3005872
  • Butcher NJ, Mortimer GM, Minchin RF. Drug delivery: unravelling the stealth effect. Nat Nanotechnol. 2016;11(4):310–311. doi:10.1038/nnano.2016.626878145
  • Bayford R, Rademacher T, Roitt I, Wang SX. Emerging applications of nanotechnology for diagnosis and therapy of disease: a review. Physiol Meas. 2017;38(8):R183–R203. doi:10.1088/1361-6579/aa718228480874
  • Johari B, Kadivar M, Lak S, et al. Osteoblast-seeded bioglass/gelatin nanocomposite: a promising bone substitute in critical-size calvarial defect repair in rat. Int J Artif Organs. 2016;39(10):524–533. doi:10.5301/ijao.500053327901555
  • Samadikuchaksaraei A, Gholipourmalekabadi M, Erfani Ezadyar E, et al. Fabrication and in vivo evaluation of an osteoblast-conditioned nano-hydroxyapatite/gelatin composite scaffold for bone tissue regeneration. J Biomed Mater Res A. 2016;104(8):2001–2010. doi:10.1002/jbm.a.3573127027855
  • Kargozar S, Mozafari M, Hashemian SJ, et al. Osteogenic potential of stem cells-seeded bioactive nanocomposite scaffolds: A comparative study between human mesenchymal stem cells derived from bone, umbilical cord Wharton’s jelly, and adipose tissue. J Biomed Mater Res B Appl Biomater. 2018;106(1):61–72. doi:10.1002/jbm.b.3381427862947
  • Weinreb RN, Leung CK, Crowston JG, et al. Primary open-angle glaucoma. Nat Rev Dis Primers. 2016;2(1):16067. doi:10.1038/nrdp.2016.6727654570
  • Lee EJ, Han JC, Park DY, Kee C. A neuroglia-based interpretation of glaucomatous neuroretinal rim thinning in the optic nerve head. Prog Retin Eye Res. 2020;100840. doi:10.1016/j.preteyeres.2020.10084031982595
  • Walsh JT, Robbins SL, Savino PJ. Visual Field Loss in a Patient With Optic Disc Cupping. JAMA Ophthalmol. 2019;137(7):844–845. doi:10.1001/jamaophthalmol.2019.117331120527
  • Levkovitch-Verbin H. Optic nerve cupping represents neuronal loss. Ophthalmology. 2020;127(4S):S43–S44. doi:10.1016/j.ophtha.2020.01.02432200825
  • Chen SJ, Lu P, Zhang WF, Lu JH. High myopia as a risk factor in primary open angle glaucoma. Int J Ophthalmol. 2012;5(6):750–753. doi:10.3980/j.issn.2222-3959.2012.06.1823275912
  • Choquet H, Thai KK, Yin J, et al. A large multi-ethnic genome-wide association study identifies novel genetic loci for intraocular pressure. Nat Commun. 2017;8(1):2108. doi:10.1038/s41467-017-01913-629235454
  • Johnson M, McLaren JW, Overby DR. Unconventional aqueous humor outflow: A review. Exp Eye Res. 2017;158:94–111. doi:10.1016/j.exer.2016.01.01726850315
  • Ho LTY, Osterwald A, Ruf I, et al. Role of the autotaxin-lysophosphatidic acid axis in glaucoma, aqueous humor drainage and fibrogenic activity. Biochim Biophys Acta Mol Basis Dis. 2020;1866(1):165560. doi:10.1016/j.bbadis.2019.16556031648019
  • Goel M, Picciani RG, Lee RK, Bhattacharya SK. Aqueous humor dynamics: a review. Open Ophthalmol J. 2010;4(1):52–59. doi:10.2174/187436410100401005221293732
  • Yan X, Li M, Song Y, et al. Influence of exercise on intraocular pressure, schlemm’s canal, and the trabecular meshwork. Invest Ophthalmol Vis Sci. 2016;57(11):4733–4739. doi:10.1167/iovs.16-1947527607419
  • Vrabec JP, Levin LA. The neurobiology of cell death in glaucoma. Eye (Lond). 2007;21(Suppl 1):S11–S14. doi:10.1038/sj.eye.670288018157171
  • Weinreb RN, Khaw PT. Primary open-angle glaucoma. Lancet. 2004;363(9422):1711–1720. doi:10.1016/S0140-6736(04)16257-015158634
  • Bessero AC, Clarke PG. Neuroprotection for optic nerve disorders. Curr Opin Neurol. 2010;23(1):10–15. doi:10.1097/WCO.0b013e328334446119915465
  • Fang JH, Wang XH, Xu ZR, Jiang FG. Neuroprotective effects of bis(7)-tacrine against glutamate-induced retinal ganglion cells damage. BMC Neurosci. 2010;11(1):31. doi:10.1186/1471-2202-11-3120199668
  • Tezel G. Oxidative stress in glaucomatous neurodegeneration: mechanisms and consequences. Prog Retin Eye Res. 2006;25(5):490–513. doi:10.1016/j.preteyeres.2006.07.00316962364
  • Kong GY, Van Bergen NJ, Trounce IA, Crowston JG. Mitochondrial dysfunction and glaucoma. J Glaucoma. 2009;18(2):93–100. doi:10.1097/IJG.0b013e318181284f19225343
  • Group, C.N.T.G.S. Comparison of glaucomatous progression between untreated patients with normal-tension glaucoma and patients with therapeutically reduced intraocular pressures. Collaborative Normal-Tension Glaucoma Study Group. Am J Ophthalmol. 1998;126(4):487–497. doi:10.1016/S0002-9394(98)00223-29780093
  • Fedorchak MV, Conner IP, Schuman JS, Cugini A, Little SR. Long term glaucoma drug delivery using a topically retained gel/microsphere eye drop. Sci Rep. 2017;7(1):8639. doi:10.1038/s41598-017-09379-828819134
  • Sambhara D, Aref AA. Glaucoma management: relative value and place in therapy of available drug treatments. Ther Adv Chronic Dis. 2014;5(1):30–43. doi:10.1177/204062231351128624381726
  • Hejkal TW, Camras CB. Prostaglandin analogs in the treatment of glaucoma. Semin Ophthalmol. 1999;14(3):114–123. doi:10.3109/0882053990906146410790575
  • Camras C, Toris C. Advances in glaucoma management: risk factors, diagnostic tools, therapies and the role of prostaglandin analogs. Foreword. Surv Ophthalmol. 2008;53(Suppl1):S1–S2. doi:10.1016/j.survophthal.2008.08.01319038617
  • Alizadeh R, Akil H, Tan J, Law SK, Caprioli J. Trabeculectomy outcomes after glaucoma drainage device surgery. J Glaucoma. 2018;27(2):133–139. doi:10.1097/IJG.000000000000084929239864
  • Gurwitz JH, Glynn RJ, Monane M, et al. Treatment for glaucoma: adherence by the elderly. Am J Public Health. 1993;83(5):711–716. doi:10.2105/AJPH.83.5.7118484454
  • Loch C, Zakelj S, Kristl A, et al. Determination of permeability coefficients of ophthalmic drugs through different layers of porcine, rabbit and bovine eyes. Eur J Pharm Sci. 2012;47(1):131–138. doi:10.1016/j.ejps.2012.05.00722659372
  • Scruggs J, Wallace T, Hanna C. Route of absorption of drug and ointment after application to the eye. Ann Ophthalmol. 1978;10(3):267–271.655537
  • Prausnitz MR, Noonan JS. Permeability of cornea, sclera, and conjunctiva: a literature analysis for drug delivery to the eye. J Pharm Sci. 1998;87(12):1479–1488. doi:10.1021/js980259410189253
  • Urtti A. Challenges and obstacles of ocular pharmacokinetics and drug delivery. Adv Drug Deliv Rev. 2006;58(11):1131–1135. doi:10.1016/j.addr.2006.07.02717097758
  • Bito LZ, Baroody RA. The penetration of exogenous prostaglandin and arachidonic acid into, and their distribution within, the mammalian eye. Curr Eye Res. 1981;1(11):659–669. doi:10.3109/027136881090018707346237
  • Wei G, Xu H, Ding PT, SM L, Zheng JM. Thermosetting gels with modulated gelation temperature for ophthalmic use: the rheological and gamma scintigraphic studies. J Control Release. 2002;83(1):65–74. doi:10.1016/S0168-3659(02)00175-X12220839
  • Hamalainen KM, Kananen K, Auriola S, Kontturi K, Urtti A. Characterization of paracellular and aqueous penetration routes in cornea, conjunctiva, and sclera. Invest Ophthalmol Vis Sci. 1997;38(3):627–634.9071216
  • Lavik E, Kuehn MH, Kwon YH. Novel drug delivery systems for glaucoma. Eye (Lond). 2011;25(5):578–586. doi:10.1038/eye.2011.8221475311
  • Patel A, Cholkar K, Agrahari V, Mitra AK. Ocular drug delivery systems: an overview. World J Pharmacol. 2013;2(2):47–64. doi:10.5497/wjp.v2.i2.4725590022
  • Gaudana R, Ananthula HK, Parenky A, Mitra AK. Ocular drug delivery. AAPS J. 2010;12(3):348–360. doi:10.1208/s12248-010-9183-320437123
  • Wei G, Ding PT, Zheng JM, Lu WY. Pharmacokinetics of timolol in aqueous humor sampled by microdialysis after topical administration of thermosetting gels. Biomed Chromatogr. 2006;20(1):67–71. doi:10.1002/bmc.52915954162
  • Cardigos J, Ferreira Q, Crisostomo S, et al. Nanotechnology-ocular devices for glaucoma treatment: a literature review. Curr Eye Res. 2019;44(2):111–117. doi:10.1080/02713683.2018.153621830309248
  • Patel S, Pasquale LR. Glaucoma drainage devices: a review of the past, present, and future. Semin Ophthalmol. 2010;25(5–6):265–270. doi:10.3109/08820538.2010.51884021091010
  • Alvarado JA, Hollander DA, Juster RP, Lee LC. Ahmed valve implantation with adjunctive mitomycin C and 5-fluorouracil: long-term outcomes. Am J Ophthalmol. 2008;146(2):276–284. doi:10.1016/j.ajo.2008.04.00818538300
  • Ponnusamy T, Yu H, John VT, Ayyala RS, Blake DA. A novel antiproliferative drug coating for glaucoma drainage devices. J Glaucoma. 2014;23(8):526–534. doi:10.1097/IJG.0b013e318294869b23632409
  • Sommer A. Collaborative normal-tension glaucoma study. Am J Ophthalmol. 1999;128(6):776–777. doi:10.1016/S0002-9394(99)00369-410612525
  • Krupin T, Liebmann JM, Greenfield DS, Ritch R, Gardiner S. Low-pressure glaucoma study g. a randomized trial of brimonidine versus timolol in preserving visual function: results from the low-pressure glaucoma treatment study. Am J Ophthalmol. 2011;151(4):671–681. doi:10.1016/j.ajo.2010.09.02621257146
  • Schnyder A, Huwyler J. Drug transport to brain with targeted liposomes. NeuroRx. 2005;2(1):99–107. doi:10.1602/neurorx.2.1.9915717061
  • Li X, Hu D, Dang Y, et al. Nano Mapper: an Internet knowledge mapping system for nanotechnology development. J Nanopart Res. 2009;11(3):529–552. doi:10.1007/s11051-008-9491-z21170121
  • Wagner V, Dullaart A, Bock AK, Zweck A. The emerging nanomedicine landscape. Nat Biotechnol. 2006;24(10):1211–1217. doi:10.1038/nbt1006-121117033654
  • Zarbin MA, Montemagno C, Leary JF, Ritch R. Nanotechnology in ophthalmology. Can J Ophthalmol. 2010;45(5):457–476. doi:10.3129/i10-09020871642
  • Bucolo C, Drago F, Salomone S. Ocular drug delivery: a clue from nanotechnology. Front Pharmacol. 2012;3:188. doi:10.3389/fphar.2012.0018823125835
  • Zarbin MA, Montemagno C, Leary JF, Ritch R. Nanomedicine in ophthalmology: the new frontier. Am J Ophthalmol. 2010;150(2):144–162. (). doi:10.1016/j.ajo.2010.03.01920670739
  • Sahoo SK, Dilnawaz F, Krishnakumar S. Nanotechnology in ocular drug delivery. Drug Discov Today. 2008;13(3–4):144–151. doi:10.1016/j.drudis.2007.10.02118275912
  • Lu H, Wang J, Wang T, Zhong J, Bao Y, Hao H. Recent Progress on nanostructures for drug delivery applications. J Nanomater. 2016;2016:5762431. doi:10.1155/2016/5762431
  • Kim HJ, Zhang K, Moore L, Ho D. Diamond nanogel-embedded contact lenses mediate lysozyme-dependent therapeutic release. ACS Nano. 2014;8(3):2998–3005. doi:10.1021/nn500296824506583
  • Goyal G, Garg T, Rath G, Goyal AK. Current nanotechnological strategies for treating glaucoma. Crit Rev Ther Drug Carrier Syst. 2014;31(5):365–405. doi:10.1615/CritRevTherDrugCarrierSyst.201401012325271557
  • Pita-Thomas DW, Goldberg JL. Nanotechnology and glaucoma: little particles for a big disease. Curr Opin Ophthalmol. 2013;24(2):130–135. doi:10.1097/ICU.0b013e32835cfe9223287105
  • Gaafar PM, Abdallah OY, Farid RM, Abdelkader H. Preparation, characterization and evaluation of novel elastic nano-sized niosomes (ethoniosomes) for ocular delivery of prednisolone. J Liposome Res. 2014;24(3):204–215. doi:10.3109/08982104.2014.88185024484536
  • Janagam DR, Wu L, Lowe TL. Nanoparticles for drug delivery to the anterior segment of the eye. Adv Drug Deliv Rev. 2017;122:31–64. doi:10.1016/j.addr.2017.04.00128392306
  • Puglia C, Offerta A, Tirendi GG, et al. Design of solid lipid nanoparticles for caffeine topical administration. Drug Deliv. 2016;23(1):36–40. doi:10.3109/10717544.2014.90301124735249
  • Kim M, Park JH, Jeong H, et al. An Evaluation of the in vivo Safety of Nonporous Silica Nanoparticles: ocular Topical Administration versus Oral Administration. Sci Rep. 2017;7(1):8238. doi:10.1038/s41598-017-08843-928811672
  • Natarajan JV, Darwitan A, Barathi VA, et al. Sustained drug release in nanomedicine: a long-acting nanocarrier-based formulation for glaucoma. ACS Nano. 2014;8(1):419–429. doi:10.1021/nn404602424392729
  • Salatin S, Yari Khosroushahi A. Overviews on the cellular uptake mechanism of polysaccharide colloidal nanoparticles. J Cell Mol Med. 2017;21(9):1668–1686. doi:10.1111/jcmm.1311028244656
  • Nagai N, Ogata F, Otake H, Nakazawa Y, Kawasaki N. Energy-dependent endocytosis is responsible for drug transcorneal penetration following the instillation of ophthalmic formulations containing indomethacin nanoparticles. Int J Nanomedicine. 2019;14:1213–1227. doi:10.2147/IJN.S19668130863055
  • 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. 2019;6(16):1802289. doi:10.1002/advs.201802289
  • Prow TW. Toxicity of nanomaterials to the eye. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2010;2(4):317–333. doi:10.1002/wnan.6520077524
  • Kim KT, Zaikova T, Hutchison JE, Tanguay RL. Gold nanoparticles disrupt zebrafish eye development and pigmentation. Toxicol Sci. 2013;133(2):275–288. doi:10.1093/toxsci/kft08123549158
  • Immordino ML, Dosio F, Cattel L. Stealth liposomes: review of the basic science, rationale, and clinical applications, existing and potential. Int J Nanomedicine. 2006;1(3):297–315.17717971
  • Mastrobattista E, Koning GA, van Bloois L, Filipe AC, Jiskoot W, Storm G. Functional characterization of an endosome-disruptive peptide and its application in cytosolic delivery of immunoliposome-entrapped proteins. J Biol Chem. 2002;277(30):27135–27143. doi:10.1074/jbc.M20042920012021269
  • Monem AS, Ali FM, Ismail MW. Prolonged effect of liposomes encapsulating pilocarpine HCl in normal and glaucomatous rabbits. Int J Pharm. 2000;198(1):29–38. doi:10.1016/S0378-5173(99)00348-810722948
  • Puri A, Loomis K, Smith B, et al. Lipid-based nanoparticles as pharmaceutical drug carriers: from concepts to clinic. Crit Rev Ther Drug Carrier Syst. 2009;26(6):523–580. doi:10.1615/CritRevTherDrugCarrierSyst.v26.i6.1020402623
  • Jain RL, Shastri JP. Study of ocular drug delivery system using drug-loaded liposomes. Int J Pharm Investig. 2011;1(1):35–41. doi:10.4103/2230-973X.76727
  • Yu S, Wang QM, Wang X, et al. Liposome incorporated ion sensitive in situ gels for opthalmic delivery of timolol maleate. Int J Pharm. 2015;480(1–2):128–136. doi:10.1016/j.ijpharm.2015.01.03225615987
  • Law SL, Huang KJ, Chiang CH. Acyclovir-containing liposomes for potential ocular delivery. Corneal penetration and absorption. J Control Release. 2000;63(1–2):135–140. doi:10.1016/S0168-3659(99)00192-310640587
  • Hathout RM, Mansour S, Mortada ND, Guinedi AS. Liposomes as an ocular delivery system for acetazolamide: in vitro and in vivo studies. AAPS PharmSciTech. 2007;8(1):1. doi:10.1208/pt080100117408209
  • Velpandian T, Gupta SK, Gupta YK, Biswas NR, Agarwal HC. Ocular drug targeting by liposomes and their corneal interactions. J Microencapsul. 1999;16(2):243–250. doi:10.1080/02652049928921110080117
  • Mehanna MM, Elmaradny HA, Samaha MW. Mucoadhesive liposomes as ocular delivery system: physical, microbiological, and in vivo assessment. Drug Dev Ind Pharm. 2010;36(1):108–118. doi:10.3109/0363904090309975119656004
  • Sasaki H, Karasawa K, Hironaka K, Tahara K, Tozuka Y, Takeuchi H. Retinal drug delivery using eyedrop preparations of poly-L-lysine-modified liposomes. Eur J Pharm Biopharm. 2013;83(3):364–369. doi:10.1016/j.ejpb.2012.10.01423153668
  • Ruel-Gariepy E, Leclair G, Hildgen P, Gupta A, Leroux JC. Thermosensitive chitosan-based hydrogel containing liposomes for the delivery of hydrophilic molecules. J Control Release. 2002;82(2–3):373–383. doi:10.1016/S0168-3659(02)00146-312175750
  • Tan G, Yu S, Pan H, et al. Bioadhesive chitosan-loaded liposomes: A more efficient and higher permeable ocular delivery platform for timolol maleate. Int J Biol Macromol. 2017;94(Pt A):355–363. doi:10.1016/j.ijbiomac.2016.10.03527760378
  • Natarajan JV, Ang M, Darwitan A, Chattopadhyay S, Wong TT, Venkatraman SS. Nanomedicine for glaucoma: liposomes provide sustained release of latanoprost in the eye. Int J Nanomedicine. 2012;7:123–131. doi:10.2147/IJN.S2546822275828
  • Li T, Tachibana K, Kuroki M, Kuroki M. Gene transfer with echo-enhanced contrast agents: comparison between Albunex, Optison, and Levovist in mice–initial results. Radiology. 2003;229(2):423–428. doi:10.1148/radiol.229202050014512507
  • Taniyama Y, Tachibana K, Hiraoka K, et al. Development of safe and efficient novel nonviral gene transfer using ultrasound: enhancement of transfection efficiency of naked plasmid DNA in skeletal muscle. Gene Ther. 2002;9(6):372–380. doi:10.1038/sj.gt.330167811960313
  • Lindner JR. Microbubbles in medical imaging: current applications and future directions. Nat Rev Drug Discov. 2004;3(6):527–532. doi:10.1038/nrd141715173842
  • Mizoue T, Horibe T, Maruyama K, et al. Targetability and intracellular delivery of anti-BCG antibody-modified, pH-sensitive fusogenic immunoliposomes to tumor cells. Int J Pharm. 2002;237(1–2):129–137. doi:10.1016/S0378-5173(02)00044-311955811
  • Sugano M, Negishi Y, Endo-Takahashi Y, et al. Gene delivery system involving Bubble liposomes and ultrasound for the efficient in vivo delivery of genes into mouse tongue tissue. Int J Pharm. 2012;422(1–2):332–337. doi:10.1016/j.ijpharm.2011.11.00122100513
  • Kouchak M, Bahmandar R, Bavarsad N, Farrahi F. Ocular Dorzolamide Nanoliposomes for Prolonged IOP Reduction: in-Vitro and in-vivo Evaluation in Rabbits. Iran J Pharm Res. 2016;15(1):205–212.
  • Kouchak M, Malekahmadi M, Bavarsad N, Saki Malehi A, Andishmand L. Dorzolamide nanoliposome as a long action ophthalmic delivery system in open angle glaucoma and ocular hypertension patients. Drug Dev Ind Pharm. 2018;44(8):1239–1242. doi:10.1080/03639045.2017.138619628956449
  • Fahmy HM, Saad E, Sabra NM, El-Gohary AA, Mohamed FF, Gaber MH. Treatment merits of Latanoprost/Thymoquinone - Encapsulated liposome for glaucomatus rabbits. Int J Pharm. 2018;548(1):597–608. doi:10.1016/j.ijpharm.2018.07.01229997042
  • Wong TT, Novack GD, Natarajan JV, Ho CL, Htoon HM, Venkatraman SS. Nanomedicine for glaucoma: sustained release latanoprost offers a new therapeutic option with substantial benefits over eyedrops. Drug Deliv Transl Res. 2014;4(4):303–309. doi:10.1007/s13346-014-0196-925787063
  • Mahale NB, Thakkar PD, Mali RG, Walunj DR, Chaudhari SR. Niosomes: novel sustained release nonionic stable vesicular systems–an overview. Adv Colloid Interface Sci. 2012;183–184:46–54. doi:10.1016/j.cis.2012.08.002
  • Kaur IP, Kanwar M. Ocular preparations: the formulation approach. Drug Dev Ind Pharm. 2002;28(5):473–493. doi:10.1081/DDC-12000344512098838
  • Kaur IP, Aggarwal D, Singh H, Kakkar S. Improved ocular absorption kinetics of timolol maleate loaded into a bioadhesive niosomal delivery system. Graefes Arch Clin Exp Ophthalmol. 2010;248(10):1467–1472. doi:10.1007/s00417-010-1383-020437246
  • Aggarwal D, Kaur IP. Improved pharmacodynamics of timolol maleate from a mucoadhesive niosomal ophthalmic drug delivery system. Int J Pharm. 2005;290(1–2):155–159. doi:10.1016/j.ijpharm.2004.10.02615664141
  • Aggarwal D, Pal D, Mitra AK, Kaur IP. Study of the extent of ocular absorption of acetazolamide from a developed niosomal formulation, by microdialysis sampling of aqueous humor. Int J Pharm. 2007;338(1–2):21–26. doi:10.1016/j.ijpharm.2007.01.01917300885
  • Kaur IP, Garg A, Singla AK, Aggarwal D. Vesicular systems in ocular drug delivery: an overview. Int J Pharm. 2004;269(1):1–14. doi:10.1016/j.ijpharm.2003.09.01614698571
  • Liu G, Molas M, Grossmann GA, et al. Biological properties of poly-L-lysine-DNA complexes generated by cooperative binding of the polycation. J Biol Chem. 2001;276(37):34379–34387. doi:10.1074/jbc.M10525020011438546
  • Nagarwal RC, Kant S, Singh PN, Maiti P, Pandit JK. Polymeric nanoparticulate system: a potential approach for ocular drug delivery. J Control Release. 2009;136(1):2–13. doi:10.1016/j.jconrel.2008.12.01819331856
  • Jung HJ, Abou-Jaoude M, Carbia BE, Plummer C, Chauhan A. Glaucoma therapy by extended release of timolol from nanoparticle loaded silicone-hydrogel contact lenses. J Control Release. 2013;165(1):82–89. doi:10.1016/j.jconrel.2012.10.01023123188
  • Harmia T, Speiser P, Kreuter J. A solid colloidal drug delivery system for the eye: encapsulation of pilocarpin in nanoparticles. J Microencapsul. 1986;3(1):3–12. doi:10.3109/026520486090495803508173
  • Rathod LV, Kapadia R, Sawant KK. A novel nanoparticles impregnated ocular insert for enhanced bioavailability to posterior segment of eye: in vitro, in vivo and stability studies. Mater Sci Eng C Mater Biol Appl. 2017;71:529–540. doi:10.1016/j.msec.2016.10.01727987741
  • Singh J, Chhabra G, Pathak K. Development of acetazolamide-loaded, pH-triggered polymeric nanoparticulate in situ gel for sustained ocular delivery: in vitro. Ex vivo evaluation and pharmacodynamic study. Drug Dev Ind Pharm. 2014;40(9):1223–1232. doi:10.3109/03639045.2013.81406123837522
  • Nanjwade BK, Bechra HM, Derkar GK, Manvi FV, Nanjwade VK. Dendrimers: emerging polymers for drug-delivery systems. Eur J Pharm Sci. 2009;38(3):185–196. doi:10.1016/j.ejps.2009.07.00819646528
  • Spataro G, Malecaze F, Turrin CO, et al. Designing dendrimers for ocular drug delivery. Eur J Med Chem. 2010;45(1):326–334. doi:10.1016/j.ejmech.2009.10.01719889480
  • Vandamme TF, Brobeck L. Poly(amidoamine) dendrimers as ophthalmic vehicles for ocular delivery of pilocarpine nitrate and tropicamide. J Control Release. 2005;102(1):23–38. doi:10.1016/j.jconrel.2004.09.01515653131
  • Yang H, Leffler CT. Hybrid dendrimer hydrogel/poly(lactic-co-glycolic acid) nanoparticle platform: an advanced vehicle for topical delivery of antiglaucoma drugs and a likely solution to improving compliance and adherence in glaucoma management. J Ocul Pharmacol Ther. 2013;29(2):166–172. doi:10.1089/jop.2012.019723249385
  • Bravo-Osuna I, Vicario-de-la-Torre M, Andres-Guerrero V, et al. Novel Water-Soluble Mucoadhesive Carbosilane Dendrimers for Ocular Administration. Mol Pharm. 2016;13(9):2966–2976. doi:10.1021/acs.molpharmaceut.6b0018227149661
  • Chen MC, Gupta M, Cheung KC. Alginate-based microfluidic system for tumor spheroid formation and anticancer agent screening. Biomed Microdevices. 2010;12(4):647–654. doi:10.1007/s10544-010-9417-220237849
  • Weinreb RN, Jani R. A novel formulation of an ophthalmic beta-adrenoceptor antagonist. J Parenter Sci Technol. 1992;46(2):51–53.1588458
  • Weinreb RN, Caldwell DR, Goode SM, et al. A double-masked three-month comparison between 0.25% betaxolol suspension and 0.5% betaxolol ophthalmic solution. Am J Ophthalmol. 1990;110(2):189–192. doi:10.1016/S0002-9394(14)76990-92198812
  • Ranpise NS, Kulkarni NS, Mair PD, Ranade AN. Improvement of water solubility and in vitro dissolution rate of aceclofenac by complexation with beta-cyclodextrin and hydroxypropyl-beta-cyclodextrin. Pharm Dev Technol. 2010;15(1):64–70. doi:10.3109/1083745090300216519505225
  • Ludwig A. The use of mucoadhesive polymers in ocular drug delivery. Adv Drug Deliv Rev. 2005;57(11):1595–1639. doi:10.1016/j.addr.2005.07.00516198021
  • Laza-Knoerr AL, Gref R, Couvreur P. Cyclodextrins for drug delivery. J Drug Target. 2010;18(9):645–656. doi:10.3109/1061186100362255220497090
  • Granero GE, Longhi MR. Promising complexes of acetazolamide for topical ocular administration. Expert Opin Drug Deliv. 2010;7(8):943–953. doi:10.1517/17425247.2010.49753620565335
  • Jansook P, Stefansson E, Thorsteinsdottir M, et al. Cyclodextrin solubilization of carbonic anhydrase inhibitor drugs: formulation of dorzolamide eye drop microparticle suspension. Eur J Pharm Biopharm. 2010;76(2):208–214. doi:10.1016/j.ejpb.2010.07.00520637867
  • Wang F, Bao X, Fang A, et al. Nanoliposome-Encapsulated Brinzolamide-hydropropyl-beta-cyclodextrin Inclusion Complex: A Potential Therapeutic Ocular Drug-Delivery System. Front Pharmacol. 2018;9:91. doi:10.3389/fphar.2018.0009129487529
  • Rodriguez-Aller M, Guinchard S, Guillarme D, et al. New prostaglandin analog formulation for glaucoma treatment containing cyclodextrins for improved stability, solubility and ocular tolerance. Eur J Pharm Biopharm. 2015;95(Pt B):203–214. doi:10.1016/j.ejpb.2015.04.03225960331
  • Junghanns JU, Muller RH. Nanocrystal technology, drug delivery and clinical applications. Int J Nanomedicine. 2008;3(3):295–309. doi:10.2147/ijn.s59518990939
  • Wu W, Li J, Wu L, et al. Ophthalmic delivery of brinzolamide by liquid crystalline nanoparticles: in vitro and in vivo evaluation. AAPS PharmSciTech. 2013;14(3):1063–1071. doi:10.1208/s12249-013-9997-223813437
  • Tuomela A, Liu P, Puranen J, et al. Brinzolamide nanocrystal formulations for ophthalmic delivery: reduction of elevated intraocular pressure in vivo. Int J Pharm. 2014;467(1–2):34–41. doi:10.1016/j.ijpharm.2014.03.04824680962
  • Orasugh JT, Sarkar G, Saha NR, et al. Effect of cellulose nanocrystals on the performance of drug loaded in situ gelling thermo-responsive ophthalmic formulations. Int J Biol Macromol. 2019;124:235–245. doi:10.1016/j.ijbiomac.2018.11.21730481535
  • Ikuta Y, Aoyagi S, Tanaka Y, et al. Creation of nano eye-drops and effective drug delivery to the interior of the eye. Sci Rep. 2017;7(1):44229. doi:10.1038/srep4422928290486
  • Mochalin VN, Shenderova O, Ho D, Gogotsi Y. The properties and applications of nanodiamonds. Nat Nanotechnol. 2011;7(1):11–23. doi:10.1038/nnano.2011.20922179567
  • Lloyd AW, Faragher RG, Denyer SP. Ocular biomaterials and implants. Biomaterials. 2001;22(8):769–785. doi:10.1016/S0142-9612(00)00237-411246945
  • Cocero MJMÁ, Mattea F, Varona S, Varona S. Encapsulation and co-precipitation processes with supercritical fluids: fundamentals and applications. J Supercrit Fluid. 2009;47(3):546–555. doi:10.1016/j.supflu.2008.08.015
  • Choi SW, Kim J. Therapeutic Contact Lenses with Polymeric Vehicles for Ocular Drug Delivery: A Review. Materials (Basel). 2018;11(7):1125. doi:10.3390/ma11071125
  • Peng CC, Burke MT, Carbia BE, Plummer C, Chauhan A. Extended drug delivery by contact lenses for glaucoma therapy. J Control Release. 2012;162(1):152–158. doi:10.1016/j.jconrel.2012.06.01722721817
  • Jung HJ, Chauhan A. Temperature sensitive contact lenses for triggered ophthalmic drug delivery. Biomaterials. 2012;33(7):2289–2300. doi:10.1016/j.biomaterials.2011.10.07622182750
  • Wadhwa S, Paliwal R, Paliwal SR, Vyas SP. Hyaluronic acid modified chitosan nanoparticles for effective management of glaucoma: development, characterization, and evaluation. J Drug Target. 2010;18(4):292–302. doi:10.3109/1061186090345002319943753
  • Zhang K, Zhang L, Weinreb RN. Ophthalmic drug discovery: novel targets and mechanisms for retinal diseases and glaucoma. Nat Rev Drug Discov. 2012;11(7):541–559. doi:10.1038/nrd374522699774
  • Liang SY, Lee GA, Shields D. Self-tonometry in glaucoma management–past, present and future. Surv Ophthalmol. 2009;54(4):450–462. doi:10.1016/j.survophthal.2009.04.00619539833
  • Kim J, Kim M, Lee MS, et al. Wearable smart sensor systems integrated on soft contact lenses for wireless ocular diagnostics. Nat Commun. 2017;8(1):14997. doi:10.1038/ncomms1499728447604
  • Leonardi M, Pitchon EM, Bertsch A, Renaud P, Mermoud A. Wireless contact lens sensor for intraocular pressure monitoring: assessment on enucleated pig eyes. Acta Ophthalmol. 2009;87(4):433–437. doi:10.1111/j.1755-3768.2008.01404.x19016660
  • Dunbar GE, Shen BY, Aref AA. The Sensimed Triggerfish contact lens sensor: efficacy, safety, and patient perspectives. Clin Ophthalmol. 2017;11:875–882. doi:10.2147/OPTH.S10970828507427
  • Enders P, Cursiefen C. Device profile of the EYEMATE-IO system for intraocular pressure monitoring: overview of its safety and efficacy. Expert Rev Med Devices. 2020;1–7. doi:10.1080/17434440.2020.1761788
  • Salim S, Du H, Wan J. Comparison of intraocular pressure measurements and assessment of intraobserver and interobserver reproducibility with the portable ICare rebound tonometer and Goldmann applanation tonometer in glaucoma patients. J Glaucoma. 2013;22(4):325–329. doi:10.1097/IJG.0b013e318237caa223542696
  • Peng HT, Shek PN. Novel wound sealants: biomaterials and applications. Expert Rev Med Devices. 2010;7(5):639–659. doi:10.1586/erd.10.4020822387
  • Spierer O, O’Brien TP. Endothelial keratoplasty combined with cataract surgery or alone using polyethylene glycol hydrogel sealant for closure of corneal incisions. J Cataract Refract Surg. 2015;41(3):492–496. doi:10.1016/j.jcrs.2015.02.00225804578
  • Masket S, Hovanesian JA, Levenson J, et al. Hydrogel sealant versus sutures to prevent fluid egress after cataract surgery. J Cataract Refract Surg. 2014;40(12):2057–2066. doi:10.1016/j.jcrs.2014.03.03425465686
  • Barliya T, Sandalon S, Ofri R, Livnat T, Weinberger D. Transcleral approach for closing retinal tears using DuraSeal hydrogel sealant. Indian J Ophthalmol. 2018;66(2):238–243. doi:10.4103/ijo.IJO_758_1729380766
  • Preethi G, Prashanth K. Design and evaluation of controlled-release ocular inserts of brimonidine-tartrate and timolol maleate. Int J Pharm Pharm Sci. 2017;9(1):79–82.
  • Perera SA, Ting DS, Nongpiur ME, et al. Feasibility study of sustained-release travoprost punctum plug for intraocular pressure reduction in an Asian population. Clin Ophthalmol. 2016;10:757–764. doi:10.2147/OPTH.S10218127175058
  • A Phase 2 study evaluating safety and efficacy of the latanoprost punctal plug delivery system (L-PPDS) in subjects with ocular hypertension (OH) or open-angle glaucoma (OAG). 2019 https://iovs.arvojournals.org/article.aspx?articleid=2358808. Accessed 611, 2020.
  • Park AJ, Eliassi-Rad B, Desai MA. Ptosis after glaucoma surgery. Clin Ophthalmol. 2017;11:1483–1489. doi:10.2147/OPTH.S13456228860697
  • Pan T, Brown JD, Ziaie B. An artificial nano-drainage implant (ANDI) for glaucoma treatment. Conf Proc IEEE Eng Med Biol Soc. 2006;1:3174–3177.
  • Paschalis EI, Chodosh J, Sperling RA, Salvador-Culla B, Dohlman C. A novel implantable glaucoma valve using ferrofluid. PLoS One. 2013;8(6):e67404. doi:10.1371/journal.pone.006740423840691
  • Maleki T, Chitnis G, Park JH, Cantor LB, Ziaie B. Biodegradable microfabricated plug-filters for glaucoma drainage devices. IEEE Trans Biomed Eng. 2012;59(6):1507–1513. doi:10.1109/TBME.2011.217903122186928
  • Harake RS, Ding Y, Brown JD, Design PT. Fabrication, and In Vitro Testing of an Anti-biofouling Glaucoma Micro-shunt. Ann Biomed Eng. 2015;43(10):2394–2405. doi:10.1007/s10439-015-1309-425821113
  • Abu-Hassan DW, Acott TS, Kelley MJ. The Trabecular Meshwork: A Basic Review of Form and Function. J Ocul Biol. 2014;2(1).
  • Gong H, Swain D. The histopathological changes in the trabecular outflow pathway and their possible effects on aqueous outflow in eyes with primary open-angle glaucoma. Glaucoma res clin adv. 2016;17–40.
  • Castro A, Du Y. Trabecular Meshwork Regeneration - A Potential Treatment for Glaucoma. Curr Ophthalmol Rep. 2019;7(2):80–88. doi:10.1007/s40135-019-00203-231316866
  • Hu Y, Tan HB, Wang XM, Rong H, Cui HP, Cui H. Bone marrow mesenchymal stem cells protect against retinal ganglion cell loss in aged rats with glaucoma. Clin Interv Aging. 2013;8:1467–1470. doi:10.2147/CIA.S4735024204132
  • Mobaraki M, Abbasi R, Omidian Vandchali S, Ghaffari M, Moztarzadeh F, Mozafari M. Corneal Repair and Regeneration: current Concepts and Future Directions. Front Bioeng Biotechnol. 2019;7:135. doi:10.3389/fbioe.2019.0013531245365
  • Alvarado J, Murphy C, Polansky J, Juster R. Age-related changes in trabecular meshwork cellularity. Invest Ophthalmol Vis Sci. 1981;21(5):714–727.7298275
  • Grierson I, Howes RC. Age-related depletion of the cell population in the human trabecular meshwork. Eye (Lond). 1987;1(Pt 2):204–210.3653434
  • Du Y, Yun H, Yang E, Schuman J. Science v. stem cells from trabecular meshwork home to TM tissue in vivo. Investophthalmol visual sci. 2013;54(2):1450–1459.
  • Snider EJ, Kubelick KP, Tweed K, et al. Improving stem cell delivery to the trabecular meshwork using magnetic nanoparticles. Sci Rep. 2018;8(1):12251. doi:10.1038/s41598-018-30834-730115953
  • Dillinger AE, Guter M, Froemel F, et al. Intracameral delivery of layer‐by‐layer coated siRNA nanoparticles for glaucoma therapy.Small. 2018;14(50):1803239.
  • Dautriche CN, Szymanski D, Kerr M, et al. A biomimetic Schlemm’s canal inner wall: A model to study outflow physiology, glaucoma pathology and high-throughput drug screening. Biomaterials. 2015;65:86–92.26142779
  • Dautriche CN, Xie Y, Sharfstein S. Walking through trabecular meshwork biology: toward engineering design of outflow physiology. Biotechnol adv. 2014;32(5):971–983.24806891
  • Torrejon KY, Papke EL, Halman JR, et al. Bioengineered glaucomatous 3D human trabecular meshwork as an in vitro disease model. Biotechnol bioeng. 2016;113(6):1357–1368.26615056
  • Waduthanthri KD, He Y, Montemagno C, Cetinel S. An injectable peptide hydrogel for reconstruction of the human trabecular meshwork. Acta biomater. 2019;100:244–254.31557533
  • Ward M, Khoobehi A, Lavik E, Langer R, Young M. Neuroprotection of retinal ganglion cells in DBA/2J mice with GDNF-loaded biodegradable microspheres. J pharm sci. 2007;96(3):558–568.17177208
  • Jiang C, Moore MJ, Zhang X, Klassen H, Langer R, Young M. Intravitreal injections of GDNF-loaded biodegradable microspheres are neuroprotective in a rat model of glaucoma. 2007.
  • Checa-Casalengua P, Jiang C, Bravo-Osuna I, et al. Retinal ganglion cells survival in a glaucoma model by GDNF/Vit E PLGA microspheres prepared according to a novel microencapsulation procedure. J Control Rel. 2011;156(1):92–100.
  • García-Caballero C, Prieto-Calvo E, Checa-Casalengua P, et al. Six month delivery of GDNF from PLGA/vitamin E biodegradable microspheres after intravitreal injection in rabbits0. Eur J Pharm Sci. 2017;103:19–26.28259830
  • Nkansah MK, Tzeng SY, Holdt AM, Lavik EBJB. bioengineering. Poly (lactic‐co‐glycolic acid) nanospheres and microspheres for short‐and long‐term delivery of bioactive ciliary neurotrophic factor. Biotechnol bioeng. 2008;100(5):1010–1019.18431801
  • Eastlake K, Wang W, Jayaram H, et al. Phenotypic and functional characterization of muller glia isolated from induced pluripotent stem cell-derived retinal organoids: improvement of retinal ganglion cell function upon transplantation. Stem Cells Transl Med. 2019;8(8):775–784. doi:10.1002/sctm.18-026331037833
  • Ooto S, Akagi T, Kageyama R, et al. Potential for neural regeneration after neurotoxic injury in the adult mammalian retina. Proc Natl Acad Sci U S A. 2004;101(37):13654–13659. doi:10.1073/pnas.040212910115353594
  • Lawrence JM, Singhal S, Bhatia B, et al. MIO-M1 cells and similar muller glial cell lines derived from adult human retina exhibit neural stem cell characteristics. Stem Cells. 2007;25(8):2033–2043. doi:10.1634/stemcells.2006-072417525239