nanotechnology for medical and surgical glaucoma therapy—a … · 2020-01-18 · tors,...

45
REVIEW Nanotechnology for Medical and Surgical Glaucoma Therapy—A Review Marcelo Luı ´s Occhiutto . Raul C. Maranha ˜o . Vital Paulino Costa . Anastasios G. Konstas Received: August 7, 2019 / Published online: December 10, 2019 Ó The Author(s) 2019 ABSTRACT Glaucoma is the second leading cause of blindness worldwide. Even though significant advances have been made in its management, currently available antiglaucoma therapies suf- fer from considerable drawbacks. Typically, the success and efficacy of glaucoma medications are undermined by their limited bioavailability to target tissues and the inadequate adherence demonstrated by patients with glaucoma. The latter is due to a gradual decrease in tolerability of lifelong topical therapies and the significant burden to patients of prescribed stepwise antiglaucoma regimens with frequent dosing which impact quality of life. On the other hand, glaucoma surgery is restricted by the inability of antifibrotic agents to efficiently control the wound healing process without causing severe collateral damage and long-term complications. Evolution of the treatment paradigm for patients with glaucoma will ideally include prevention of retinal ganglion cell degeneration by the successful delivery of neurotrophic fac- tors, anti-inflammatory drugs, and gene thera- pies. Nanotechnology-based treatments may surpass the limitations of currently available glaucoma therapies through optimized targeted drug delivery, increased bioavailability, and controlled release. This review addresses the recent advances in glaucoma treatment strate- gies employing nanotechnology, including medical and surgical management, neurore- generation, and neuroprotection. Keywords: Glaucoma; Nanomedicine; Nanoparticles; Nanosystems; Nanotechnology; Ocular drug delivery; Ophthamlology Enhanced Digital Features To view enhanced digital features for this article go to https://doi.org/10.6084/ m9.figshare.10265768. M. L. Occhiutto V. P. Costa Department of Ophthalmology, University of Campinas (UNICAMP), Campinas, Brazil R. C. Maranha ˜o Laboratory of Metabolism and Lipids, Heart Institute (InCor), Medical School Hospital, University of Sa ˜o Paulo, Sa ˜o Paulo, Brazil R. C. Maranha ˜o Faculty of Pharmaceutical Sciences, University of Sa ˜o Paulo, Sa ˜o Paulo, Brazil A. G. Konstas (&) 1st and 3rd University Departments of Ophthalmology, Aristotle University of Thessaloniki, Thessaloniki, Greece e-mail: [email protected] Adv Ther (2020) 37:155–199 https://doi.org/10.1007/s12325-019-01163-6

Upload: others

Post on 24-Jul-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

REVIEW

Nanotechnology for Medical and Surgical GlaucomaTherapy—A Review

Marcelo Luıs Occhiutto . Raul C. Maranhao . Vital Paulino Costa .

Anastasios G. Konstas

Received: August 7, 2019 / Published online: December 10, 2019� The Author(s) 2019

ABSTRACT

Glaucoma is the second leading cause ofblindness worldwide. Even though significantadvances have been made in its management,currently available antiglaucoma therapies suf-fer from considerable drawbacks. Typically, thesuccess and efficacy of glaucoma medicationsare undermined by their limited bioavailabilityto target tissues and the inadequate adherencedemonstrated by patients with glaucoma. The

latter is due to a gradual decrease in tolerabilityof lifelong topical therapies and the significantburden to patients of prescribed stepwiseantiglaucoma regimens with frequent dosingwhich impact quality of life. On the other hand,glaucoma surgery is restricted by the inability ofantifibrotic agents to efficiently control thewound healing process without causing severecollateral damage and long-term complications.Evolution of the treatment paradigm forpatients with glaucoma will ideally includeprevention of retinal ganglion cell degenerationby the successful delivery of neurotrophic fac-tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments maysurpass the limitations of currently availableglaucoma therapies through optimized targeteddrug delivery, increased bioavailability, andcontrolled release. This review addresses therecent advances in glaucoma treatment strate-gies employing nanotechnology, includingmedical and surgical management, neurore-generation, and neuroprotection.

Keywords: Glaucoma; Nanomedicine;Nanoparticles; Nanosystems; Nanotechnology;Ocular drug delivery; Ophthamlology

Enhanced Digital Features To view enhanced digitalfeatures for this article go to https://doi.org/10.6084/m9.figshare.10265768.

M. L. Occhiutto � V. P. CostaDepartment of Ophthalmology, University ofCampinas (UNICAMP), Campinas, Brazil

R. C. MaranhaoLaboratory of Metabolism and Lipids, HeartInstitute (InCor), Medical School Hospital,University of Sao Paulo, Sao Paulo, Brazil

R. C. MaranhaoFaculty of Pharmaceutical Sciences, University ofSao Paulo, Sao Paulo, Brazil

A. G. Konstas (&)1st and 3rd University Departments ofOphthalmology, Aristotle University ofThessaloniki, Thessaloniki, Greecee-mail: [email protected]

Adv Ther (2020) 37:155–199

https://doi.org/10.1007/s12325-019-01163-6

Page 2: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

Key Summary Points

This article offers a comprehensive reviewof nanotechnology-based treatments forpatients with glaucoma.

Nanotechnology-based drugs will probablybe incorporated into the arsenal of glaucomaspecialists in the near future, allowingbenefits such as reduced side effects, and lessfrequent dosing, among others.

Toxicity issues related to nanotechnology-based treatments have yet to be addressedto test their safety.

Further human studies in nanomedicine forglaucoma treatment are needed until thispromising pharmacological innovationbecomes available in the ophthalmologist’sdaily therapeutic practice.

INTRODUCTION

Glaucoma is the second leading cause of blind-ness worldwide [1, 2] and the most frequent causeof irreversible blindness. Indeed, the WorldHealth Organization estimates that 5.2 millioncases of blindness are a result of glaucoma (15% ofthe total burden of world blindness) [3]. Glau-coma is often characterized by elevated intraoc-ular pressure (IOP), caused predominantly byblockage of the outflow system leading to theprogressive death of retinal ganglion cells (RGCs)and resulting in optic neuropathy [4].

The main goal of virtually all glaucomatherapies today is to reduce IOP by either sup-pressing aqueous synthesis or by enhancingtrabecular meshwork (TM) and uveoscleraloutflow of aqueous humor [5–7]. Other pro-posed modalities for the treatment of glaucomainclude laser [8], surgery [9], and non-IOP-de-pendent therapies such as neuroprotection [10].

Recently, research has focused on nanopar-ticles and their unique properties, which canprovide a platform to surpass the limitations of

traditional glaucoma therapies. Although somereviews have been published [11–15], we aimedto expand available evidence and provide acomprehensive summary of recent advances innanotechnology-based antiglaucoma therapieswith a focus on the following topics: hypoten-sive drugs, wound healing modulation, neuro-protection, and neuroregeneration.

Figure 1 summarizes the main therapeutictargets in glaucoma treatment according totheir anatomical location. Importantly, all ofthem could benefit from nanotechnology-basedstrategies.

TARGETING INTRAOCULARPRESSURE REDUCTION

Elevated IOP is considered the key risk factor foroptic nerve damage in glaucoma, and mostpatients benefit from IOP-lowering therapies [16].Despite the evidence that factors other than IOPare involved in the pathogenesis of glaucoma[17, 18], current treatment algorithms are basedon decreasing IOP. First-line glaucoma therapytypically starts with eye drops that lower IOP bytwo mechanisms: suppression of aqueous humorproduction (beta-blockers, alpha-agonists, andcarbonic anhydrase inhibitors) or increase ofaqueous humor outflow though the trabecular oruveoscleral pathways (pilocarpine, epinephrine,or prostaglandin analogues) [19]. In patientswhere topical medications are not sufficient tocontroI IOP, laser or surgery can be indicated. Wewill discuss both treatment strategies in the sce-nario of recent advancements in nanomedicine.This article is based on previously conductedstudies and does not contain any studies withhuman participants or animals performed by anyof the authors.

DRUG DELIVERY ROUTES,FEATURES, AND LIMITATIONSOF CURRENT GLAUCOMAMEDICAL THERAPY

Although medical therapy of glaucoma iseffective, it relies on a delivery system that is

156 Adv Ther (2020) 37:155–199

Page 3: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

generally inefficient and presents several draw-backs. First, eye drops have to be administered1–3 times daily, which is frequently associatedwith suboptimal use and lack of adherence [20].This is a more pronounced issue in the elderlypopulation that is predominantly affected byglaucoma [21]. A previous report showed that60% of patients expressed one or more prob-lems with taking their glaucoma medications,which results in a decreased adherence to long-term therapy [22]. Furthermore, followinginstillation, the bioavailability of topical glau-coma medications in the anterior chamber isvery low [23]. Typical aqueous humor bioavail-ability of a drug following the administration of

eye drops ranges from 1% to 10% in animal [24]and human studies [25]. This is due to the var-ious physiological barriers to drug penetrationinto the eye [26].

There are two routes that control and influ-ence the absorption of topically administeredmedication: the corneal and the non-cornealroutes. Small molecules with a hydroxyl grouphave high corneal permeability, favoring thecorneal route [27]. The trajectory of the drugmolecules starts with their passive diffusion viathe corneal epithelium, through the stroma andendothelium into the anterior chamber, wherethe drug will carry out its pharmacologicalaction [28] or will bind to melanin in the ciliary

Fig. 1 Glaucoma therapeutic strategies and nanotechnology-based potential applications

Adv Ther (2020) 37:155–199 157

Page 4: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

body and iris [29], or plasma proteins [30],which prolongs its half-life.

The remaining drug and drug metaboliteswill be removed via the trabecular meshworkthrough Schlemm’s canal into the systemicbloodstream (conventional route), or via theuveoscleral route (unconventional outflow).Both of these pathways lead to the systemicblood circulation [31]. The ‘‘unconventional’’route that includes the ciliary muscle, supracil-iary and suprachoroidal spaces, may drainaqueous humor and/or drugs and theirmetabolites through two possible pathways: auveovortex pathway where they enter thechoroid to drain through the vortex veins, and auveoscleral pathway where they pass across thesclera to be resorbed by orbital vessels [32]. Inaddition to these two routes, there is a thirdroute, the ‘‘uveolymphatic pathway’’, defined byYucel et al. to describe a novel pathway foraqueous humor drainage through lymphaticchannels in the human ciliary body, which canbecome a novel target for glaucoma treatment[33–35]. Drug penetration in the iris and diffu-sion via the aqueous humor flow occurs with asmall portion of the drug (depending on themolecular weight and lipophilicity). Drug con-centrations in the vitreous will be 10–100 timeslower than in the aqueous humor and cornea,respectively [27].

The so-called non-corneal absorption route ispreferred by drugs with low corneal permeabil-ity, such as proteins and large molecules. Theselarge compounds penetrate the eye via theconjunctiva and/or sclera [36, 37]. This routedelivers drugs to the vitreous via passive diffu-sion and allows 20 times lower drug concen-tration in the anterior chamber compared to thecorneal route [38]. However, the conjunctivaand underlying sclera are more permeable andhave a higher surface of absorption for thebloodstream than the cornea [39]. Furthermore,some hypotensive topical drugs, such as timololand betaxolol, have been shown to accumulatein Tenon’s capsule, periocular tissues, and scleraof normal cynomolgus monkeys, and of glau-comatous patients under long-term topicaltherapy [40, 41]. These findings indicate thatperiocular accumulation of certain medicationscould provide differentiated access to the

posterior segment and proximal vasculature ofthe eye.

Systemic absorption of topical medicationscan lead to systemic side effects such ashypotension, bronchospasm, and bradycardia(beta-blockers), or dry mouth and fatigue (al-pha-agonists) [19]. Prostaglandin analogues areprodrugs which have the advantage of beingcleaved to their active forms inside the eye,thereby minimizing side effects in nontargetorgans [42].

There are numerous ongoing studies aimedat offering neuroprotective or neuroregenera-tive treatments [16], but clinical trials have beenunsuccessful in the past at demonstrating theireffectiveness in preventing glaucoma progres-sion [43–45]. One of the main challenges of thisapproach has been how to allow access of thedrug to the back of the eye, where the opticnerve and RGCs reside. There are two pathwaysto allow drugs to reach these posterior struc-tures: crossing the vitreous and the inner lim-iting membrane of the retina, or accessing theretina through the vascular system. Intravitrealinjections allow direct access to the vitreouscavity and bypass this barrier, but increase therisk for serious ocular adverse events, such asendophthalmitis [46]. The intravenous route isaccompanied by systemic side effects and isparticularly problematic since drugs must crossthe blood–retinal barrier to reach the targettissue [47].

To overcome the multiple limitations ofcurrent medical antiglaucoma treatments, thedevelopment of safer and more efficient drugdelivery systems is imperative. These attributesmay be reached through the use of nanomedi-cine, which may improve solubility, reduce tis-sue irritation, prolong shelf life, and providedose accuracy, sustained release, targeteddelivery, and bioavailability [12].

Even though nanoparticulate systems canpotentially improve drug bioavailability of theactive loaded ingredients, they can also beselectively removed more or less quickly fromthe target organ by conventional, uveoscleral,and uveolymphatic routes, depending on thedirect uptake of the nanoparticle, and theirphysicochemical specificities. Therefore, furtherpharmacokinetic studies specific for each

158 Adv Ther (2020) 37:155–199

Page 5: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

nanosystem employed for therapeutic purposeswill be required.

ROLE AND VALUEOF NANOPARTICLES

Nanoparticles (NPs) are tiny structures mostcommonly spherical in shape with sizes in thenanometer range (1–100 nm). As a result oftheir minuscule size, nanoparticles can bypassbiological barriers, providing drug accessdirectly to the target cells [48]. Moreover,smaller nanoparticles have a higher drug-load-ing capacity than the larger ones, owing to theirhigher surface area [49].

NPs are utilized in a wide range of shapessuch as nanoemulsion [50], liposomes [51],dendrimers [52], nanospheres [53], hydrogels[54], nanocrystals [55], cyclodextrins [56], nan-odiamonds [57], microspheres [58], niosomes[59], nanofibers [60], and nanocapsules [61].Figure 2 shows a schematic representation ofvarious nanoparticles mentioned throughoutthis article.

NPs can be designed to enhance activecompound penetration [62] and drug targeting[63] and to promote controlled release [64].Currently, NPs are generally made from sundrymaterials including polymers, lipids, and met-als. Lipid and polymer NPs may be employedsuccessfully to carry drugs [65], isolate theircontents from degradation, and regulate theirrelease [66]. Metallic NPs have specific physic-ochemical properties that serve both diagnostic[67] and therapeutic purposes [68, 69]. Injectednanoparticles accumulate in the liver andspleen and are mostly eliminated by the retic-uloendothelial system soon after administra-tion. It is, however, possible to employ a coatingthat can prevent opsonization and recognitionof NPs by the macrophages. Indeed, NPs may becovered by extra layers to achieve special ther-apeutic properties, to reduce side effects, or toincrease solubility [70]. Engineered NPs may becreated to help the molecule to find its target.For instance, docetaxel and ketoconazole loa-ded in solid lipid nanoparticles had their surfacemodified with folic acid to improve brain tar-geting [71]. Another example is hyaluronic acid-

modified lipid–polymer hybrid NPs, which weredesigned to improve the ocular bioavailabilityof hydrophilic moxifloxacin hydrochloride, inorder to prolong precorneal retention, andenhance its corneal permeability and ocularbioavailability [72].

The selection of the appropriate systemdepends on the target tissue, route of adminis-tration, and the characteristics of the drug to beincorporated into the NPs (size, stability,hydrophobicity, etc.). However, there are com-mon physicochemical properties that allnanosystems must possess, such as biocompat-ibility, biodegradability, absence of toxicity,and stability, which can provide effective andsafe pharmacological action, better than thoseobtained by traditional drug formulations.

EMERGENCE OF NANOMEDICINEFORMULATIONS IN GLAUCOMA

Several drugs traditionally employed in glau-coma treatment, e.g., timolol, carteolol, bri-monidine, pilocarpine, brinzolamide,bimatoprost, travoprost, and latanoprost [73],have been investigated in nanomedicine for-mulations [11, 12, 15]. We emphasize here thatall of these formulations are still under investi-gation and are not currently approved for clin-ical use. In the following pages, we describenanosystems loaded with the most utilizeddrugs for the treatment of glaucoma.

Pilocarpine

Pilocarpine has by and large been avoided byophthalmologists because of several undesirableadverse effects, most of which are due to itsnon-selective ocular and systemic action as amuscarinic receptor agonist: intense miosis,ocular irritation, myopic shift, bronchial mucussecretion, bronchospasm, vasodilation, brady-cardia, excessive salivation, sweating, and diar-rhea. Pilocarpine has been used in a nitrate orhydrochloride pharmaceutical formulation,usually at a 0.5–4% concentration administeredfour times daily because of the short duration ofaction. When administered as a solution, it has

Adv Ther (2020) 37:155–199 159

Page 6: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

Fig. 2 Schematic depiction of some nanoparticles

160 Adv Ther (2020) 37:155–199

Page 7: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

a reasonable water solubility, which shortensthe residence time of aqueous solutions in theeye. Hence, the real obstacle with pilocarpinesolutions has been its very low ocular bioavail-ability (0.1–3%) [74, 75] arising from its poorlipophilicity.

Research efforts have been devoted to opti-mizing the activity and tolerability of pilo-carpine [76]. Many nanosystems have been triedin order to improve pilocarpine’s bioavailabil-ity, solubility, and stability and to extend itspharmacological activity [77]. Table 1 showsexamples of pilocarpine nanoparticulate drugdelivery systems.

Timolol Maleate

Timolol maleate is a non-selective beta-adren-ergic blocker that acts by inhibiting the beta-adrenergic receptors in the ciliary body epithe-lium, promoting the reduction of aqueoushumor production and resulting in decreasedIOP [87]. The hydrophilic nature of timololmaleate leads to slow corneal diffusion and lowbioavailability [88]. Timolol maleate absorptionfrom the eye into the systemic circulation, vianasolacrimal drainage, causes systemic adren-ergic beta-blockage and high incidence of res-piratory and cardiovascular side effects [89]. Toimprove the pharmacological features of timo-lol maleate, it is necessary to enhance its cor-neal penetration and reduce systemicabsorption, which may be attained by the use ofnanocarriers. Table 2 shows examples of timololmaleate-loaded nanoparticulate drug deliverysystems.

Carbonic Anhydrase Inhibitors

Carbonic anhydrase (CA) is a generic denomi-nation to designate several isoenzymes dis-tributed in diverse proportions in the varioustissues responsible for catalyzing the reversiblehydration of carbon dioxide [104]. In the eye,CA is a key enzyme in aqueous humor produc-tion. Nonpigmented ciliary epithelial cells arethe site of aqueous humor secretion, where twoCA isoenzymes (CA-II and CA-IV) are the pre-dominant forms. The inhibition of CA activity

in the ciliary processes promotes decreasedaqueous humor secretion and consequentlyleads to lowering of the IOP [105]. CA-II isconsidered the most relevant intracytoplasmicisoenzyme responsible for aqueous humorsecretion [106]. Therefore, selective pharmaco-logical inhibition of CA-II to achieve an ocularhypotensive effect is the objective in the treat-ment of glaucoma [106].

Three different CA inhibitors (CAI) mole-cules, all of them sulfonamide derivatives, arecurrently used in clinical practice to reduceelevated IOP in glaucoma. Acetazolamide isadministered orally, and dorzolamide andbrinzolamide are both administered topically[107]. Acetazolamide is a compound thatindiscriminately inhibits most of the CA iso-zymes (CA-I, CA-II, CA-IV, CA-VA, CA-VB, CA-VII, CA-XIII, and CA-XIV) present in otherorgans than the eye [108–110], and althoughsystemically administered acetazolamide is veryeffective as an intraocular hypotensive agent,the non-selective inhibition of the enzymeresults in a myriad of undesirable side effects[108–110]. Dorzolamide and brinzolamide arepotent nanomolar inhibitors of human CA-IIisozyme [106, 111–113], with less activityagainst CA-IV, CA-XII, and CA-I [114].

AcetazolamideAcetazolamide (ACZ) is a CAI used systemicallyto obtain a substantial and immediate reductionof IOP in glaucoma management. It is currentlyconsidered one of the most effective availableIOP-lowering agents [115]. Limitations of orallyadministered ACZ include several systemicadverse events (e.g., metabolic acidosis, pares-thesias, fatigue, diuresis, malaise, anorexia, lossof libido, renal function impediment, etc.)[116]. As a rule, large oral doses of ACZ arenecessary to obtain the desired IOP reduction,probably because of its low bioavailability andrelative instability [117]. Topical administrationof ACZ would have been preferred over systemicadministration, but as a result of the pharma-cological limitations of topical ACZ (e.g., poorpermeability via the corneal epithelium, shortresidence time, frequent instillations, loss ofdrug through nasolacrimal drainage, poor pen-etration coefficient (only 1–6% of the active

Adv Ther (2020) 37:155–199 161

Page 8: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

Table1

Experim

entalevidence

withpilocarpine-loaded

nanoparticulatesystem

s

Hypotensive

drug/nanosystem

Pharm

aceuticalform

Stud

ydesign/m

odel

Results

References

Com

plex

ofpilocarpineprodrug,

O,O

0 -dipropionyl-(1,4-

xylylene)bispilocarpate,with

variousb-cyclodextrin

derivatives

Cyclodextrin

Experim

ental/pigm

entedrabbits

Ocularirritation

caused

bypilocarpineprodrugis

elim

inated

withviscousSB

E7-b-CyD

solution

without

comprom

isingtheocular

absorption

of

theprodrug

[78]

Poly(amidoamine)

(PAMAM)

dend

rimers(withprim

ary

amino(G

2,G4),h

ydroxyl

(G2(OH),G4(OH)),and

carboxylate(G

1.5,

G3.5)

surfacegroups

Dendrim

ers

Experim

ental/New

Zealand

albino

rabbits

Intensityof

pilocarpine’spharmacologicalactivity

associated

withG1.5andG4(OH)dend

rimer

solutionswas

reported

tobe

superior

[79]

PilocarpineHClencapsulated

neutralandnegativelycharged

multilamellarvesicles

(MLVs)

Liposom

esExperim

ental/pigm

entedrabbits

NeutralMLVslowered

theIO

Pfrom

20.7

to

15mmHgfor4–

5h

NegativelychargedMLVsdecreasedtheIO

Pfora

shorterperiod

oftime(*

1h),w

hich

was

similar

tothefree

drug

[80]

Pilocarpinenitrate–cyclodextrin

complex

Cyclodextrin

Invitroperm

eabilitystudyusing

isolated

excisedcorneasof

albino

rabbits

HP-b-CD

prom

oted

anincrease

inthecorneal

perm

eabilityof

pilocarpinenitrate(permeability

coefficient

=6.38

910

-5±

2.82

910

-7cm

/s,

comparedto

1.67

910

-5±

2.12

910

-7cm

/s

withneat

pilocarpinenitrate)

[81]

Pilocarpine-loaded

poly(e-

caprolactone)(PCL)

nanocapsules

(PILO-PCL

NCs)andpilocarpine-loaded

PCLnanospheres(PILO-PCL

NSs)

Nanocapsulesand

nanospheres

Invitropilocarpinecumulative

releasestudies,andin

vivo

studies(glaucom

atousrabbit

eyes

intracam

erallyinjected

with

PILO-PCLNSs

orPILO-PCL

NCs)

Pilocarpineloadingcapacity

ofPC

LNCswasnearly

3times

greaterthan

that

inPILO-PCLNSs.

PILO-PCLNC-treated

groupreducedIO

Pforthe

entire

period

ofthestudy(42days),exhibiting

a

sustaineddrug

releaseprofile

[82]

162 Adv Ther (2020) 37:155–199

Page 9: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

Table1

continued

Hypotensive

drug/nanosystem

Pharm

aceuticalform

Stud

ydesign/m

odel

Results

References

Pilocarpine-loaded

hydrogels

(three

differenthydrogels

containing

L-valineresidues,

HVa)

Hydrogels

Invitropilocarpinereleaseandits

effect

oncellproliferation

Cytotoxicityexperimentswith

mouse

immortalized

fibroblast

NIH

3T3cells

Cellviability,after

24hof

incubation

withthenative

and

pilocarpine-loaded

hydrogel

Pilocarpine-loaded

inHVahydrogelsshow

eda3

times

greaterreleasingprofile

[83]

Hydrogelswerenon-toxicto

themouse

fibroblast

NIH

3T3cells

Cellp

roliferationwasincreasedwiththepresence

of

pilocarpine,even

after2days

Pilocarpinenitrate-loaded

liquid

crystalnanoparticles(PN-

loaded

LCNPs)

Liquidcrystal

nanoparticles

Invitroreleaseprofileof

PNMaxim

umdecrease

inIO

Pwas

41.93±

16.79%

withthecommercialdrug

(peakeffect

at2h),

versus

59.21±

7.04%

(peakafter5h)

withPN

-

LCNPs

[84]

Exvivo

penetrationstudy(freshly

excisedalbino

rabbitcornea)

Exvivo

drug

penetrationstudydemonstratedthat

theam

ount

ofPN

penetrationacrossthecornea

at

1hwas

0.54

±0.20

lgwithPN

-LCNPs

and

0.12

±0.06

lgwithPN

solution

Pilocarpinehydrochloride-

loaded

nanocomposite

form

ulations

(cellulose

nanocrystalsandtriblock

poloxamer

copolymer)

Hydrogel/nanocrystals

Invivo

IOPmeasurementsusing

anindentationtonometer

Invitroreleaseof

pilocarpinehydrochloridefrom

thenanocomposite

hydrogelswas

extend

ed

comparedto

thepure

poloxamer

gel

[85]

Invitrodrug

releaseanalysisand

toxicity

assayof

invitrogel

The

greatertheconcentrationof

cellulose

nanocrystals,the

higher

thesustaineddrug

release

property

ofthenanocomposite

hydrogels

Nanocom

posite

form

ulationexhibitednon-toxic

behavior

Adv Ther (2020) 37:155–199 163

Page 10: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

compound actually reaches intraocular tissues)[118], poor biphasic solubility, and poor cornealbioavailability) [119], many researchers havetried to develop optimized ACZ topical drugdelivery systems, some employing nanotech-nology-based approaches [115, 117, 120–124].Table 3 shows examples of ACZ-loadednanoparticulate systems.

DorzolamideDorzolamide (DRZ) is a CAI used extensively inthe treatment of glaucoma. Inhibition of car-bonic anhydrase results in decrease of aqueoushumor production and IOP reduction. The firstcommercially available topical CAI was 2% DRZhydrochloride solution (Trusopt�) [131], avail-able since 1995. When topical 2% DRZ is used asmonotherapy, it reduces IOP by up to 23% frombaseline [132]. However, as a result of its low pHand high viscosity, 2% DRZ causes significantocular irritation, stinging, or burning [133].Moreover, topical 2% DRZ requires 2–3 dailydoses, which hinders long-term adherence totreatment [134]. Therefore, there is a clearclinical demand to enhance the delivery effi-ciency of DRZ, and nano-based drug systemsappear to have the potential to fulfil this goal.Table 4 demonstrates examples of dorzolamide-loaded nanoparticulate systems.

BrinzolamideBrinzolamide is a water-insoluble CAI, whichacts on the ciliary processes and decreasesaqueous humor secretion. This medication isemployed widely in stepwise glaucoma therapymost often in combination with prostaglandinanalogues or in conjunction with brimonidineand beta-blockers [141]. Brinzolamide is for-mulated commercially as a 1% aqueous sus-pension, because lower concentrations resultedin insufficient therapeutic effect [114]. As aresult of the solubilization demand of particleson the ocular surface and because of its highlipophilicity, brinzolamide aqueous suspensionhas a low bioavailability [142], which may elicitblurred vision after administration [143] andforeign body sensation, often leading to dis-comfort after instillation [144]. For these rea-sons, novel topical ocular delivery systems for

Table1

continued

Hypotensive

drug/nanosystem

Pharm

aceuticalform

Stud

ydesign/m

odel

Results

References

Pilocarpine-loaded

chitosan/

Carbopolnanoparticle

ophthalmicForm

ulation

Hydrogel

Invitroreleaseprofile

Invitroanalysisshow

edthat

nanoparticlesdisplayed

thebestsustainedreleaseprofilefor24

hcompared

totheother3form

ulations

[86]

Invivo

(albinorabbits)miotic

effect

ofpilocarpinechitosan/

Carbopolnanoparticleswas

comparedwithvarious

pilocarpineform

ulations

(traditionaleyedrops,gels,

and

liposom

es)

Invivo

studyshow

edextend

edmiosisof

pilocarpine-

loaded

chitosan/C

arbopolnanoparticles,which

wassuperior

tothoseof

liposom

es,pilocarpinegel,

andpilocarpinedrops

164 Adv Ther (2020) 37:155–199

Page 11: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

Table2

Experim

entalevidence

withtimolol

maleate-lo

aded

nanoparticulatesystem

s

Hypotensive

drug/nanosystem

Pharm

aceutical

form

Stud

ydesign/m

odel

Results

References

Tim

olol-lo

aded

gold

nanoparticles(G

NPs)

Gold nanoparticles

Invitroreleasestudy

Maxim

umtimolol

concentrationforblankcontact

lenses

at1hwas

581lg/ml,whilein

theGNP-

ladenCLsitwas

1685

lg/m

l

[90]

Invivo

drug

releaseand

pharmacodynam

icstudies(N

ew

Zealand

AlbinoRabbits)

IntheGNP-ladenCLsgroup,

IOPdecreasedby

4mmHgafter1h,

which

progressivelyincreased

after72

h.In

themarketedeyedrop

group,

maxim

umIO

Pdecrease

was

3mmHgafter3h,

which

returned

tobaselin

eIO

Pafter12

h

Oculartissue

distribution

study

Analysisof

therabbittearfluid

show

edhighertimolol

concentrations

withtheGNP-CLsat

alltime

points

GNPs

wereincorporated

into

contactlenses

(CLs)

After

24hof

GNP-ladenCLwear,atwofoldanda

fourfold

increase

inthetimolol

concentrationin

theconjun

ctivaandin

theiris-ciliarymuscle

occurred,respectively

Tim

olol

maleate/gelatin

nanoparticleconjugate

Hydrogel

Experim

ental/New

Zealand

albino

rabbits

The

average24-h

IOPreductionwas

52%,com

pared

to31%

provided

byregulartimolol

eyedrops

[91]

Tim

olol-lo

aded

nanoparticlesinto

apoly(hydroxyl

ethylmethacrylate)

(p-H

EMA)matrix

Hydrogel

Invitrodrug

releaseexperiments

The

higher

theam

ount

ofnanoparticlesin

the

HEMA,the

highertherateof

drug

release(particle

toHEMAratio25:75provided

atotalreleaseat

95�C

of283.08

±4.48

lg;particleto

HEMA

ratio100:0provided

atotalreleaseat

95�C

of

743.27

±25.40lg,and

also

increasedtherelease

duration)

[92]

Tim

olol-lo

aded

multilamellarvesicles

(MLVs)

Liposom

esExperim

ental/New

Zealand

albino

rabbits

PositivelychargedMLVsof

multilamellarliposom

es

provided

asustainedIO

Preductionformorethan

24h(which

extend

edforabout1week),w

hereas

thefree

drug

lowered

IOPfor4h

[93]

Adv Ther (2020) 37:155–199 165

Page 12: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

Table2

continued

Hypotensive

drug/nanosystem

Pharm

aceutical

form

Stud

ydesign/m

odel

Results

References

Chitosan(REVTMbio1)or

Carbopol

(REVTMbio2

and3)

coated

niosom

altimolol

maleate

(0.25%

)

Niosomes

Invitroreleasepatternof

niosom

al

preparations

Peak

effect

ofIO

Preductionwithmarketed

form

ulationwas

at2h,

comparedto

3hwithall

theREVTMbioform

ulations.H

owever,chitosan-

coated

vesicles

(REVTMbio1)show

edan

effect

sustainedforup

to8h,

comparedto

themarketed

form

ulation,

whose

effect

lasted

for5h.

REVTMbio1

form

ulationcontaining

0.25%

timolol

maleate

show

edsimilareffect

comparedto

0.5%

marketedgelform

ulation

[94]

Tim

olol

maleate

entrappedin

PVA

orPC

L

nanofibers

Biodegradable

polymeric

nanofibers

Invitrodrug

releasestudiesand

invivo

studies(N

ewZealand

albino

rabbits)

Nanofiberswerecapableof

controlleddrug

delivery

forup

to24

h

[95]

Nanofiberform

ulationshow

edasignificant

IOP

reductionfor72

h,whilethemarketedform

ulation

kept

theIO

Preducedfor4h

Chitosan-coated

timolol

maleate

(TM)

mucoadhesivefilm

Hydrogel

Invitrodrug

releasestudiesand

invivo

studies(N

ewZealand

albino

rabbits)

Invitrostudyshow

edthat

85%

oftimolol

was

released

from

themucoadhesivefilmsin

2weeks,

andthetotalcontentwas

released

within4weeks

[96]

IOP-loweringefficacyof

the0.5%

TM

commercial

ophthalmicsolution

was

similarto

thefilms.

How

ever,animalsthat

received

TM-lo

aded

chitosan

filmskept

theirIO

Pat

lower

levelsover

a

10-weekperiod,w

hiletheeffect

ofthetimolol

commercialeyedropsgroupwas

maintainedfor

12days

166 Adv Ther (2020) 37:155–199

Page 13: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

Table2

continued

Hypotensive

drug/nanosystem

Pharm

aceutical

form

Stud

ydesign/m

odel

Results

References

Nanoencapsulation

oftimololin

neatchitosan

(CS)

andN-alkylated

chitosans[chitosanderivatives

withsuccinicanhydride(C

SUC)and

2-carboxybenzaldehyde(C

BCS)]

Hydrogel

Invitrodrug

releasestudy

Inthemajorityof

nanoparticlesform

ulations,tim

olol

isentrappedin

amorphousform

,but

thepresent

study,usingdifferentdiam

etersof

nanoparticles

andCSandtwoN-acylatedderivativesof

CSas

carriers,provedthatdrug

entrapmentefficiencywas

higherin

CBCSderivative.D

ifferenttimololrelease

ratesam

ongthetested

form

ulations

ratifiedthat

they

vary

accordingto

specificcarrier,nanoparticle

size,and

drug

loading

[97]

Tim

olol

maleate

encapsulated

inPL

GA/PLA

microspheres

Microspheres

Invitrodrug

releasestudy

PLGA502H

:PLAblendedmicrospheresreleased

30%

oftheirtimolol

maleate

contentafter1day.

How

ever,the

remaining

drug

was

released

ina

sustainedmannerover

107days

[98]

Tim

olol-lo

aded

chitosan–sodium

alginate

(CS-SA

blend)

nanoparticles

Hydrogel

Invitrodrug

releasestudy

Invitroreleasefrom

theCS-SA

nanoparticlesshow

ed

aburstof

about20%withinthefirsthour

and35%

oftimolol

was

released

afteraperiod

of5h,

demonstrating

that

itmay

bereleased

from

synthesizedparticlesin

asustainedmode

[99]

Tim

olol

maleate

chitosan

coated

liposom

es(TM-

CHL)

Liposom

esIn

vitrodrug

releasestudy

TM-CHLproduced

a3.18-foldincrease

inthe

cornealperm

eation

coefficient

[100]

Invivo

transcornealperm

eation

studies(N

ewZealand

rabbits)

TM-CHLwas

moreeffectivein

loweringIO

Pthan

timolol

eyedrops(final

IOP=19.67±

1.14

mmHgversus

23.80±

1.49

mmHg,respectively)

Tim

olol

liposom

es(TLP),and

TLPwith0.02%

Trancutol

P(TLPG

)

Liposom

al

hydrogel

Invitrotranscornealperm

eation

study

Invivo

pharmacodynam

ics(N

ew

Zealand

andpigm

ented

glaucomatousrabbits)

Tim

olol–liposom

esystem

show

edatranscorneal

penetration1.50-foldhigher

than

that

ofthe

marketedeyedrop,w

hiletheaddition

ofTrancutol

Penhanced

it2.19

times

Tim

ololliposom

algelshowed

superior

IOPreduction

comparedwithTim

ololeyedropsateach

timepoint

[101]

Adv Ther (2020) 37:155–199 167

Page 14: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

Table2

continued

Hypotensive

drug/nanosystem

Pharm

aceutical

form

Stud

ydesign/m

odel

Results

References

Tim

olol

maleate-lo

aded

galactosylated

chitosan

nanoparticles

Hydrogel

Invitroreleasestudy

Tim

olol

maleate-lo

aded

galactosylated

chitosan

nanoparticlesshow

edan

initialburstreleaseof

91%

in8handhadasustainedreleasecomparedwith

thecommercialtimolol

eyedrops

[102]

Invitrotranscornealperm

eation

study

Preocularretentionof

timolol

maleate-lo

aded

galactosylated

chitosan

nanoparticleswas

longer

than

that

oftimolol

eyedrops

Preocularretentionstudy

IOP-loweringeffect

oftimolol

maleate-lo

aded

galactosylated

chitosan

nanoparticlesreached

10.5

±0.51

mmHg4hafterinstillation,

whilethe

peak

effect

ofthecommercialeyedropswas

6.8±

0.35

mmHgat

3h

Invivo

pharmacodynam

icsstudy

(albinorabbits)

IOP-loweringeffect

oftimolol

maleate-lo

aded

galactosylated

chitosan

nanoparticlescontinuedfor

upto

12h,whiletheeffectof

commercialeyedrops

ceased

after8h

Tim

olol-lo

aded

liposom

eincorporated

ion-sensitive

insitu

gels

Liposom

esIn

vitroreleasestudies

Tim

olol-lo

aded

liposom

eform

ulations

show

eda1.93-

fold

increase

inperm

eabilitycoefficients

[103]

Deacetylated

gellangum

Pharmacodynam

icsstudy

(measurementof

intraocular

pressure

innorm

alalbino

rabbits

andin

ocular

hypotensiverabbits)

Tim

olol-lo

aded

liposom

eform

ulations

reducedIO

P

from

30to

300min

afterinstillation(m

inim

um

IOP=11.96±

0.74

mmHgat

1h),w

hilethe

IOPdecreasedfrom

30to

180min

(minim

um

IOP=13.61±

0.95

mmHgat

2h)

withtimolol

eyedrops

168 Adv Ther (2020) 37:155–199

Page 15: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

Table3

Experim

entalevidence

derivedfrom

acetazolam

ide-loaded

nanoparticulatesystem

s

Hypotensive

drug/nanosystem

Pharm

aceuticalform

Stud

ydesign/m

odel

Results

References

Eudragitnanoparticles(N

Ps)of

ACZ

incorporated

into

anocular

insert

Eudragitnanoparticles

Invitrodrug

diffusionstudy

Exvivo

transcornealperm

eabilitystudy(excised

freshgoat

corneas)

Invivo

ocular

tolerabilityandIO

Preduction

study(albinorabbits):anim

alsreceived

drinking

water

(40ml/kg

ofbody

weightof

rabbit)to

increase

IOP

3groups:(1)ACZreferencesolution,(2)

EudragitNPs

dispersion,and

(3)ocular

insert

ofEudragitNPs

Exvivo

transcornealperm

eation

studyshow

ed

thefollowingresults.F

lowacrosscorneal

tissue

(lg/min):drug

suspension.

0.671±

0.020;

NPs

suspension.

2.460±

0.028;

ocular

insert,2.402

±0.032,

which

means

that

ACZ-lo

aded

EudragitNPs

displayedbetter

perm

eabilityandflowacross

cornealtissue

than

thedrug

suspension

Invivo

studieswithoptimized

ACZloaded

EudragitNPs

andocular

insertdemonstrated

substantialIOPloweringandim

proved

ocular

tolerabilitywhencomparedto

ACZ

suspension

[125]

ACZ-lo

aded

nanoparticulatein

situ

gels(N

P-

ISG)

Polymericnanoparticles

withEudragitRL100,

EudragitRS100,o

r

poly(lactide-co-glycolide)

75:25(PLGA)

Exvivo

transcornealperm

eabilitystudy(fresh

goat

cornea)

Exvivo

transcornealperm

eation

studydisplayed

higher

ACZperm

eation

at8hwithNP1

0

(93.5±

2.25

mg/cm

2 )andwithNP-ISG5

(74.50

±2.20

mg/cm

2 )than

withACZeye

drops(20.08

±3.12

mg/cm

2 )andACZ

suspension

(16.03

±2.14

mg/cm

2 )

[126]

Exvivo

cornealtoxicity

study(fresh

goat

cornea)

NP-ISG

didnotdisplayharm

fuleffectson

any

corneallayer

Invivo

pharmacodynam

icactivity

study

(normotensive

rabbit)

1%ACZnanoparticulatein

situ

gelexhibited

greaterIO

P-loweringeffect

1hafter

administration,which

wassustainedforup

to

8h,

while1%

ACZeyedropsonlysustained

itsaction

forapproxim

ately2h

Adv Ther (2020) 37:155–199 169

Page 16: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

Table3

continued

Hypotensive

drug/nanosystem

Pharm

aceuticalform

Stud

ydesign/m

odel

Results

References

ACZ-lo

aded

water-solublemucoadhesive

carbosilane

dend

rimers

C–S

ibackbone

(carbosilane)cationic

dend

rimers

Invitro(cytotoxicityandcellviability)

investigation(using

telomerase-im

mortalized,

human

corneal-lim

balepithelialcelllin

e,

HCLE)

Generations

1and2of

thecationicdend

rimers

andall3generationsof

theanionic

dend

rimerswerewelltoleratedat

10lM,

withhigher

than

80%

cellsurvivalforallof

them

,exceptfortheG3-C

(from

the3rd

generation

ofcarbosilane

cationic

dend

rimers)

[127]

Invivo

(oculartolerability)

study(normotensive

New

Zealand

rabbits):specular

microscopy,

slitlampexam

ination,

andIO

P

measurements

Form

ulationcontaining

ACZ0.07%

(289.4mOsm

;5.6pH

;41.7mN/m

)andG3

cationiccarbosilane

dend

rimers(5

lM)

demonstratedthebestIO

P-loweringeffect.It

obtained

arapid(1

hpost-in

stillation)

and

sustained(upto

7h)

hypotensiveeffect,

reaching

apeak

22.6%

IOPreduction

ACZ-lo

aded

hyperbranched

poly(propylene

imine)

(PPI)dend

rimers

PPIdend

rimers

Invitrodrug

releasestudies

Exvivo

studies(effecton

themorphologyof

human

erythrocytes)

Invivo

studies(normotensive

New

Zealand

rabbits):determ

inationof

ocular

irritation

index,ocular

residencetime,IO

Preduction

(25lL

ofdend

rimer

form

ulationwas

administeredinto

thelower

cul-d

e-sacof

the

eye)

Hem

olytictoxicity

studyshow

edaslightly

higher

hemolysisrate

ofdend

rimer

form

ulations

(D1=4.8%

,D2=5.6%

,

D3=7.2%

)whencomparedto

plaindrug

(3.3%)andplain5.0G

PPIdend

rimer

(7.9%)

PlainACZsolution

produced

IOPreductions

upto

2hafterinstillation,

whereas

the

dend

rimer-based

form

ulationlowered

IOPfor

longer

(4h)

[128]

170 Adv Ther (2020) 37:155–199

Page 17: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

Table3

continued

Hypotensive

drug/nanosystem

Pharm

aceuticalform

Stud

ydesign/m

odel

Results

References

ACZ-lo

aded

ion-activatednanoem

ulsion-based

insitu

gelling

system

susinggellangum

polymer

aloneandin

combination

with

otherpolymers(xanthan

gum,

hydroxym

ethylcellulose,o

rCarbopol)

Nanoemulsion

Gellangum

(invarious

concentrations:0.1%

,

0.2%

,0.3%,0

.4%,0

.5%,

and0.6%

)

Invitroreleasestudies

Optim

ized

form

ulaof

ion-induced

nanoem

ulsion-based

insitu

gelsdemonstrated

asignificantlysustaineddrug

releaseprofile

[129]

Invivo

studies:ocular

irritation

and

pharmacodynam

icstudiesin

adultmaleNew

Zealand

albino

rabbits

Ocularirritation

studyrevealed

nodamageto

theocular

surfaceandotherpartsof

theeye

Areaun

derthecurveof

percentage

decrease

in

IOPversus

timefrom

0to

10h:

nanoem

ulsion

form

ula=189.15

±10.18;

Azopt

�=82.51±

7.53,and

Cidam

ex�=79.77±

7.58

ACZ-lo

aded

EudragitRL100nanoparticle

suspension

(ACZ-E-N

Ps)

Polymericnanoparticles

withEudragitRL100

Invitrodrug

releasestudy

Invivo

studies(albinorabbits):

Group

1received

0.5%

ACZsolution

Group

2received

form

ulationE3(drug

polymer

ratio1:10;organicaqueousphase

ratio1:4;

organicphaseacetone)

Group

3received

form

ulationE8(drug

polymer

ratio1:10;organicaqueousphase

ratio1:3;

organicphaseacetoneandethanol)

Plainsolution

ofACZwas

ableto

lower

the

IOPforabout3hafterinstillation,

whereas

ACZ-E-N

Psolutionsshow

edagreater

efficacy(IOPloweringforup

to8hafter

instillation).T

hepeak

IOPreductionby

plainACZsolution

was

2.98

±0.11

mmHg

at2haftertopicaladministration,

whereas

bestACZ-E-N

Pform

ulations

(E3andE8)

progressivelyreducedIO

P,peakingat

8h

afterinstillation(F3=5.32

±0.07

mmHg;

E8=5.19

±0.06

mmHg;mean±

SD)

[130]

Adv Ther (2020) 37:155–199 171

Page 18: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

Table4

Examples

ofdorzolam

ide-loaded

nanoparticulatesystem

s

Hypotensive

drug/nanosystem

Pharm

aceutical

form

Stud

ydesign/m

odel

Results

References

Chitosan(C

S)andwater-soluble

6-O-carboxymethyl(O

CM-CS)

derivative

ofCSnanoparticles

(NPs)loaded

withDRZ

CSor

OCM-CS

nanoparticles

Invitrostudies(drugreleaseandmucoadhesionof

NPs)

IOPpeak

effect

andduration

ofaction

were

[135]

Invivo

studies(innorm

otensive

albino

rabbits):three

groups

(OCM-CSNPs,C

SNPs,and

marketedeyedrops)

received

asingledose

followed

byIO

Pmeasurements

(after

30min

ofdrug

administrationandthen

every1h

over

aperiod

of8h)

Withthecommercialform

ulation:

2.9mmHg2hafter

instillation,

andtheeffect

disappearedafter4h

WiththeDRZ-lo

aded

OCM-CSNPs:p

eakeffectwasseen

at4h,withan

IOPreductionof

2.2mmHg,andtheeffect

was

sustainedfor8h

WithDRZ-lo

aded

CSNPs:peak

effect

seen

at3h;

IOP

reductionwas

1.9mmHg,andtheeffect

was

sustainedfor

6h

DRZ/c-cyclodextrin(c-CD)(18%

w/v)complexes

stabilizedby

hydroxypropylmethylcellulose

(HPM

C)(0.5%

w/v)

Cyclodextrins

Invitromucoadhesiveandperm

eation

studies

DRZdistribution

24haftertopicalinstillation

indicatedthat

itpenetrates

effectivelyinto

theeye,includingthe

posteriorsegm

ent,whileserum

DRZconcentrations

were

very

low

[136]

Invivo

(using

pigm

entedrabbits):DRZmicroparticle

suspension

was

administeredto

both

eyes.A

fter

administrationtheanim

alswereanesthetized,and

a

sampleof

aqueoushumor

was

collected

once.B

lood

sampleswerealso

collected

ateach

timepoint(2,4,8,and

24h)

DRZ-c-CD

suspension

provided

sustaineddeliveryof

DRZ

intheaqueoushumor

forup

to24

h.Com

mercialDRZ

ledto

concentrations

intheaqueoushumor

near

zero

8h

aftertopicaladministration.

The

DRZconcentrationin

theaqueoushumor

ofeyes

treatedwith3%

(w/v)

dorzolam

idewas

45-foldhigher

than

thoseobtained

after

instillationof

theotherthreeform

ulations

DRZ-lo

aded

microparticles

Microparticles

Invitrodrug

releasestudies

Invivo

studies(employingnorm

otensive

Dutch

belted

rabbits)

Subconjunctivalinjectionof

DRZ-PEG3-PS

Amicroparticles

lowered

IOPby

upto

4.0±

1.5mmHg,whencompared

toun

treatedfelloweyes

for35

days

[137]

DRZ-lo

aded

microparticlesor

blankmicroparticleswere

administeredinto

thesubconjunctivalspaceof

the

superotemporalregionof

each

eyeusinga27-gauge

needle

Fluorescein-labeledPE

G3-PS

Amicroparticlesweredetected

severaldaysaftertheinjection(atleast42

days),indicating

avery

long

invivo

nanoparticledegradationperiod

IOPmeasurementsweremadeaftertheinjection

2%DRZeyedropsreducedIO

Pfor\

6h

172 Adv Ther (2020) 37:155–199

Page 19: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

Table4

continued

Hypotensive

drug/nanosystem

Pharm

aceutical

form

Stud

ydesign/m

odel

Results

References

DRZ-lo

aded

chitosan

nanoparticles

Insitu

gelling

polymeric

(chitosan)

nanoparticles

Invitroreleasestudy

Exvivo

transcornealperm

eabilitystudies(onfreshlyexcised

goat

cornea)

Mucoadhesionstudyof

drug

loaded

insitu

gel

Invivo

studies(using

albino

rabbits):ocular

tolerancetest,

andgammascintigraphy

studyto

accessprecorneal

retentiontime

Optim

ized

form

ulation(C

2S4)

demonstrated,in

theex

vivo

perm

eabilitystudy,adrug

perm

eation

of35.80%

within

2h,

comparedto

75.30%

withthecommercial

form

ulation(D

RZ2%

ophthalmicsolution)

Optim

ized

form

ulationC2S4show

edappropriategelling

featurewith98.1%

entrapmentefficiency

Gam

mascintigraphy

studyshow

edlong

retentiontimeof

the

proposed

form

ulationin

theeye,indicating

anim

proved

bioavailabilityof

DRZ

[138]

DRZ-lo

aded

nanoliposom

eLiposom

esDRZ-lo

aded

nanoliposom

ewas

administeredto

20patients

withocular

hypertension

orprim

aryopen-angleglaucoma

(inboth

eyes)andwerefollowed

upfor28

days

IOP(days0,

14,and

28)was

comparedbetweenthegroup

that

received

DRZ-lo

aded

nanoliposom

eandthegroup

that

received

themarketedDRZsolution

IOPreductionin

theDRZ-lo

aded

nanoliposom

egroupwas

significantlygreaterthan

that

seen

withthecommercially

availableDRZform

ulationgroup(p\

0.05)

IOPloweringrecorded

at2weeks

was

23.26±

9.24%,and

9.25

±5.76%

fortheDRZ-lo

aded

andthecontrolgroup,

respectively

IOPloweringat

4weeks

was

32.60±

7.90%

and

17.48±

7.62%fortheDRZ-lo

aded

andthecontrolgroup,

respectively

[139]

DRZc-cyclodextrin

(c-CD)

nanoparticle

Cyclodextrins

Self-aggregatingc-CD

nanoparticleeyedropscontaining

3%

DRZwereinstilled

once

adayin

human

eyes,com

pared

withthemarketedDRZinstilled

threetimes

aday,in

a

prospectiverand

omized

single-m

askedcrossovertrial

DRZnanoparticleeyedropsonce

adayandcommercial

form

ulationof

dorzolam

ide2%

threetimes

adaydidnot

show

statistically

significant

differencesin

term

sof

efficacy

atalltimepoints.N

anoparticleeyedropscaused

less

burningsensationthan

themarketedsolution

[140]

Adv Ther (2020) 37:155–199 173

Page 20: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

Table5

Examples

ofbrinzolamide-loaded

nanoparticulatesystem

s

Hypotensive

drug/nanosystem

Pharm

aceutical

form

Stud

ydesign/m

odel

Results

References

Brinzolam

ide(BZL)-loaded

liquid

crystalline

nanoparticles(BZLLCNPs)

Liquid

crystalline

nanoparticles

Invitroreleasestudyto

measure

the

releaseof

BZLfrom

LCNPs

Exvivo

cornealpenetrationstudy

(employingNew

Zealand

rabbits)

Efficacy

study(instillation

ofonedrop

of

marketedBZL,1

%BZLsolution,and

BZLLCNPs)

Exvivo

perm

eabilitycoefficient

ofBZL

LCNPs

show

eda3.47-foldincrease

comparedwithcommercialBZL

Twohoursafterinstillation,

peak

IOP

decrease

was

47.67±

3.58%

byBZL

LCNPs,and

33.75±

4.35%

by

commercialBZL

[145]

BZLnanocrystalsuspensions

(BZL-N

psa)

Nanocrystal

Cellulartoxicity

assayusinghuman

cornealepithelialcells

(stand

ardcell

viability

method)

BZL-N

psapH

4.5lowered

IOPafter

60min

(71.4±

5.0%

)moreefficiently

than

BZL-N

psapH

7.4Po

lysorbate80

form

ulation(51.0±

26.3%),and

commercialBZL(49.6±

16.5%)

[146]

Invivo

studies:to

verify

IOPreduction

followingBZL-N

psain

glaucomatous

Wistarrats

Allthetested

form

ulations

and

commercialBZLshow

edmild

orno

toxicity

tothehuman

cornealepithelial

cells

Trimethyllock

(TMLo)

BZLprodrug

nanoparticles

Nanocrystals

Instillationof

nano

eyedropsof

BZL

prodrugin

ocular

norm

otensive

Sprague–Daw

leyrats

TMLoBZLprodrugnano

eyedrops

show

edsimilarefficacyas

commercial

BZLat

1/5themolar

concentration

(5.67mM

TMLoBZLprodrugnano

eyedropswereas

effectiveas

26.1mM

Azopt

TM),withno

toxiceffectsto

the

cornea

[147]

174 Adv Ther (2020) 37:155–199

Page 21: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

Table5

continued

Hypotensive

drug/nanosystem

Pharm

aceutical

form

Stud

ydesign/m

odel

Results

References

BZLnanoem

ulsions(BZLNEs)

Sevenprim

aryBZLNEcombinations

wereused

[variationsof

four

nonionic

surfactants(Tyloxapol,L

abrasol,

Cremophor(RH40),andBrij35),two

oils(C

apryol

90andtriacetin),and

one

co-surfactant(TranscutolP

)]

The

amount

ofBZLwas

0.4%

inall

form

ulations

Nanoemulsion

sIn

vitrodrug

releasestudies

Invivo

therapeuticstudies(ocular

norm

otensive

New

Zealand

albino

rabbits):BZLNEswereinstilled,

followed

byIO

Pmeasurementsat

30,

60,1

20,1

80,2

40,3

00,and

360min

afterinstillation

BZLNEsdisplayedasustainedrelease

profilewithproper

physicochemical

characteristics,facilitatingBZL

penetrationinto

thecornealtissuewith

lower

drug

concentrations

(0.4%

vs1%

withcommercialBZL)

[148]

BZL-hydroxypropyl-cyclodextrin(BZL-

HP-b-CD)inclusioncomplex

Cyclodextrins

Invitrocornealperm

eabilityandrelease

studies

Invivo

study(N

ewZealand

norm

otensive

rabbits):G

roup

A:B

ZL-H

P-b-CD0.2%

inclusioncomplex;Group

B:BZL-H

P-

b-CD

0.5%

inclusioncomplex;Group

C:commercialBZL(1%)

IOPmeasured30,60,120,150,180,240,

and300min

afterinstillation

Invitrocornealaccumulationand

perm

eabilityof

theBZL-H

P-b-CD

inclusioncomplex

was

increased2.91-

fold

comparedto

commercialBZL

Solubilityof

BZLincreased10-foldwith

the(BZL-H

P-b-CD)inclusioncomplex

(BZL-H

P-b-CD)inclusioncomplex

(0.5%

BZL)show

edIO

P-lowering

efficacycomparableto

commercialBZL

invivo

[56]

Adv Ther (2020) 37:155–199 175

Page 22: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

Table5

continued

Hypotensive

drug/nanosystem

Pharm

aceutical

form

Stud

ydesign/m

odel

Results

References

Brinzolam

ide(BZL)-hydropropyl-b

-

cyclodextrin

(HP-b-CD)inclusion

complex

(HP-b-CD/BZL)into

nanoliposom

es

‘‘HP-b-CD/BZL-lo

aded

nanoliposom

es’’

(BCL)

Liposom

es

Cyclodextrins

InvitroBZLreleasestudy

Transcornealperm

eabilitystudy

Invivo

IOPmeasurement

BZLshow

edamoderatesustainedrelease

period

of9h(1–1

0h)

BCLshow

eda9.36-foldincrease

inthe

perm

eabilitycoefficient

comparedwith

commerciallyavailableBZL

BCLform

ulationreducedIO

Pin

less

than

1h,

reachedpeak

efficacy

(-32.3%)at

2handshow

eda

sustainedeffect

for12

h

BZLsuspension

lowered

IOPat

30min

andreacheditspeak

efficacyat

1h.

From

2to

12hafterinstillation,

BCL

resultedin

significantlylower

IOPs

comparedwithBZLsuspension

[149]

Brinzolam

ide(BZL)-loaded

PLGA

nanoparticles

Poly(lactic-co-

glycolicacid)

(PLGA)

nanoparticles

Asinglesubconjunctivalinjectionof

BZL-PLGA

nanoparticles

Invitrodrug

releasestudies

Invivo

IOPmeasurements(on

norm

otensive

albino

rabbits)

Twoform

ulations

ofBZL-lo

aded

PLGA

nanoparticlesdisplayedexcellent

release

efficiencyvalues:A19

released

about

70%

ofthedrug

in6months,while

B11

released

about90%

ofthedrug

in

6months

Aftersubconjunctivaladm

inistrationpeak

IOPloweringwere78.4

±3.4%

for

A19

BZLnanoparticles;71.6

±2.0%

forB11

BZLnanoparticles;and

56.8

±6.3%

forBZLaqueous

suspension

[150]

176 Adv Ther (2020) 37:155–199

Page 23: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

Table6

Examples

ofbrim

onidine-loaded

nanoparticulatesystem

s

Hypotensive

drug/nanosystem

Pharm

aceutical

form

Stud

ydesign/m

odel

Results

References

Nanovesiclesof

BRD

Liposom

esand

niosom

es

Invitroandex

invitrodrug

releasestudies

Invivo

IOP-loweringactivity

inalbino

rabbits

Group

1:marketedform

ulation(BRD

0.02%)

Group

2:liposom

es

Group

3:niosom

es

Invitroandex

invitrodrug

releaseprofilesof

allthe

nanovesiculeform

ulations

show

edamoreextend

ed

drug

releasecomparedto

thecurrently

available

commercialsolution

Efficacy

was

greatercomparedto

thecommercial

product,whose

activity

was

notsustainedbeyond

60min

[154]

Eudragit-basedbrim

onidine

tartrate

nanoparticle

form

ulations

(BRD-lo

aded

ERS–

ERLnanoparticles)

Eudragit

nanoparticles

Invitrodrug

releasestudies

Invivo

pharmacodynam

icstudies(employing

glaucomatousNew

Zealand

rabbits):IO

P-lowering

efficacystudiesperformed

byinstillationof

aqueous

dispersion

ofnanoparticlesandconventionaleyedrop

solution

Alltheselected

BRD-lo

aded

ERS–

ERLnanoparticles

show

edan

extensionof

thedrug

releasein

vitro

Resultsof

invivo

pharmacodynam

icefficacystudiesof

selected

BRD-lo

aded

ERS–

ERLnanoparticle

form

ulations

andmarketedBRD

eyedropsshow

ed

asimilarpeak

IOPreduction,

butprolongedIO

P

efficacy(m

arketedeyedropsduration

of6h

comparedto

36–7

2hwiththenanoparticles

form

ulations)

[155]

BRD-lo

aded

microspheresusing

poly(lacticacid)(PLA)

Microspheres

Invitroreleasekinetics

ofBRD

Invivo

experimentaltreatmentgroups

(employingNew

Zealand

rabbits):

Singlemicroneedleinjectionof

BRD-lo

aded

microspheresinto

thesupraciliaryspace

BRD

(0.15%

)commercialeyedrops(adm

inistered

threetimes

adayto

theupperconjun

ctivalsac,fora

week)

After

topicaldeliveryof

BRD,a

consistent

IOP

reductionof

2–4mmHgwasdetected,but

theIO

P

quicklyreturned

tobaselin

eaftertheinterruption

ofthedrops

BRD-lo

aded

microsphereshigh

dose

group(30%

BRD-lo

aded

microspheres)show

edIO

Preduction

ofthetreatedeyefor33

days,afterasingleinjection

into

thesupraciliaryspace

[156]

Adv Ther (2020) 37:155–199 177

Page 24: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

Table6

continued

Hypotensive

drug/nanosystem

Pharm

aceutical

form

Stud

ydesign/m

odel

Results

References

Optim

ized

BRD-lo

aded

chitosan

(CS)

andsodium

alginate

(ALG)nanoparticles

CSandALG

nanoparticles

Invitrostudies(drugreleaseandcytotoxicity

assays)

Invivo

studies(m

icestrainsBXD29

andBXD96):a

singledose

ofthetestform

ulations

orcommercial

BRD

eyedrops(BRD

0.15%)wereinstilled

into

the

inferior

conjun

ctivalsac

Invitrotoxicity

studiesdidnotshow

significant

differencesbetweennano-based

form

ulations

and

commercialBRD

eyedrops

Allnano-based

form

ulations

show

edagreater

sustainedIO

P-loweringeffect

comparedto

the

commercialBRD.T

imerequired

forIO

Pto

return

tobaselin

eranged

from

17.2

to25.2hfornano-

basedform

ulations,com

paredto

7–7.4hfor

commercialBRD

[157]

BRD-lo

aded

nanostructured

lipid

carriers

Nanostructured

(NLC)and

solid

(SLN)

lipid

nanoparticles

Invitrodrug

releasestudy

Exvivo

perm

eabilitystudy

Invivo

studies(a

singledose

(50lL

)of

lipid

nanoparticleswasinstilled

into

thelower

conjun

ctival

sacof

anorm

otensive

albino

rabbit).Oculartolerance

analysis,IOPmeasurements,and

ocularhistologywere

performed

BothNLCsandSL

Nsshow

edabiphasicrelease

pattern.SL

Nsshowed

61.74±

2.56%drug

released

after2hand74.34±

0.14%after6h,whileNLCs

show

ed66.89±

3.4%

drug

released

after2hand

95.8

±2.31%

after6h.

Com

mercialBRD

show

ed

aun

ique

burstreleaseof

88.76±

1.78%withinthe

firsthour

NLCsdemonstratedaperm

eabilitycoefficient

1.23-

fold

higher

than

that

ofSL

Ns

Peak

IOPloweringwithNLCs,SL

Ns,and

commercialBRD

eyedropswas

13.14±

1.28,

10.03±

0.32,and

7.84

±1.04

mmHg,respectively

The

peak

IOPeffect

occurred

after6hforNLCs,

after4hforcommercialBRD,and

after2hfor

SLNs

NLCsandSL

Nswerefoun

din

theanterior

cham

ber

oftreatedeyes,ind

icatingthat

lipid

nanoparticles

penetratethroughthecornea

[158]

178 Adv Ther (2020) 37:155–199

Page 25: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

this drug must surpass current problems.Table 5 shows examples of brinzolamide-loadednanoparticulate systems.

Brimonidine

Brimonidine (BRD) is an a2-adrenergic agonistwhich acts through two mechanisms: it pro-motes an acute reduction in aqueous produc-tion, associated with an increase in uveoscleraloutflow [151]. Nevertheless, BRD exhibits lowbioavailability through the corneal stroma(1–7%) [152] and shows a short duration ofaction that leads to the necessity of threeinstillations during the day [153]. Conse-quently, BRD demonstrates a relatively reducedadherence, as evidenced by the rate of adher-ence, which ranges from 31% to 67% at12 months [20]. The reluctance of patients toinstill their eye drops many times a day on aregular basis justifies the need to develop newBRD formulations. Table 6 shows examples ofBRD-loaded nanoparticulate systems.

Prostaglandin Analogues

Topically instilled prostaglandin F2a (PGF2a)analogues (latanoprost, travoprost, bimato-prost, and tafluprost) have been the fist-linechoice in the medical management of glaucomaover the last decade, owing to their favorablesafety profile, efficacy, and patient compliance[160, 161]. All PGF2a analogues lead to commonside effects, such as eyelash growth, darkeningof the iris and periocular pigmentation, con-junctival hyperemia, and rare adverse reactions(e.g., damage to the blood–aqueous barrier,cystoid macular edema, and prostaglandin-as-sociated periorbitopathy) [162]. These sideeffects reduce patient compliance. To overcomethe side effects, to reduce dosing, to optimizethe efficacy and safety of this class of drugs, andto get a reliable dosing technology deliveredcontinuously over a prolonged time (weeks ormore), novel nanoformulations of PGF2a ana-logues may prove greatly advantageous. Table 7shows examples of prostaglandin analogue-loa-ded nanoparticulate systems.

Table6

continued

Hypotensive

drug/nanosystem

Pharm

aceutical

form

Stud

ydesign/m

odel

Results

References

BRD-lo

aded

microspheres/carriersystem

(M/C

S)

Microspheres

poly( D,L-lactic-

co-glycolic

acid)

(PLGA)

Invitroreleasestudies

Invivo

studies(IOPmeasurementsafterBRD-lo

aded

M/C

Ssubconjunctivalim

plantation

innorm

aland

glaucomatouseyes)

Afterasingledoseof

theBRD-lo

aded

M/C

S,an

IOP

reductionof

20mmHgwas

achieved

after1day,

andwas

sustainedover

aperiod

of55

days

M/C

Sstructureremainedintactforeasyremovalafter

BRD

wasfully

released,evenaslong

as70

daysafter

implantation

[159]

Adv Ther (2020) 37:155–199 179

Page 26: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

Table7

Experim

entalevidence

obtained

withprostaglandinanalogue-lo

aded

nanoparticulatesystem

s

Hypotensive

drug/nanosystem

Pharm

aceuticalform

Stud

ydesign/m

odel

Results

References

Latanoprost-lo

aded

liposom

es(large

unilamellarvesicles)

Liposom

esIn

vivo

human

study:asingle

subconjunctivalinjection

ofliposom

al

latanoprostwas

administeredto

one

eyeof

6subjectswithocular

hypertension

orprim

aryopen-angle

glaucoma

BaselineIO

Pwas

27.55±

3.25

mmHg

[163]

MeanIO

Pdecreasedwithin1hto

14.52±

3.31

mmHg(range

10–1

8mmHg)

Aminim

um20%

IOPreductionwas

observed

through3monthsafter

injection

Noadverseeventswerereported

Latanoprostacid

(LA)-loaded

poly(lactide)/monom

ethoxy-

poly(ethyleneglycol)(PLA-PEG)

nanoparticles(N

Ps)

PLA

(40)

PEG

(5)nanoparticles

Invivo

studies:subconjunctival

injections

wereadministeredinto

the

subconjunctivalspaceof

threegroups

ofnorm

otensive

albino

rabbits:

(A)LA-lo

aded

NPs

(equivalentto

8.5mgLA);(B)Afree

LA

solution

ofthesamedrug

content;(C

)blank

NPs

IOPwas

monitored

for8consecutive

days

Invitrostudiesanalyzed

drug

entrapmentefficiency,andreleaseof

LA

Aqueous

humor

(AH)levelsof

LAwere

also

measured6days

post-

administration

IOPwas

lower

intheLA-lo

aded

PLA-

PEG

NPs

groupcomparedto

the

other3studygroups

(freedrug,blank

NP,

andcontrolgroup)

The

drug

entrapmentefficiencywas

18.3%

AH

levelsof

LAwereinitially

higher

in

thefree

LAgroupthan

the

nanoparticlegroup,butthey

decreased

over

time

Inthenanoparticlegroup,

AH

levelsof

LA

increasedwithtime,becoming

higher

onthe6thdaythan

inthefree

LA

group

[164]

180 Adv Ther (2020) 37:155–199

Page 27: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

Table7

continued

Hypotensive

drug/nanosystem

Pharm

aceuticalform

Stud

ydesign/m

odel

Results

References

Latanoprost-lo

aded

egg-

phosphatidylcholine(EggPC

)

liposom

es

Liposom

esIn

vivo

studies:afterasingle

subconjunctivalinjectionof

the

latanoprost-loaded

form

ulation,

rabbiteyes

wereclinicallymonitored

andtheIO

Precorded

Latanoprost-lo

aded

EggPC

liposom

es

wereableto

provideasustainedIO

P

loweringandgreatereffect

compared

withdaily

instillations

oftopical

latanoprostforup

to90

days

(4.8

±1.5vs

2.5?

0.9mmHg;

P=0.001),w

ithno

signsof

inflammation

[165]

Latanoprost-lo

aded

thermosensitive

chitosan-based

hydrogel(asatopical

eyedrop

form

ulation)

Hydrogel

Invitrodrug

releaseand

biocom

patibilitystudyof

the

latanoprost-loaded

hydrogel(cell

viabilityassays,hem

olysisanalysis,and

ocular

irritation

test)

Invivo

releasestudy(aqueous

humor

levelsof

thedrug)

Latanoprost-lo

aded

hydrogelwas

administeredweeklyinto

thelower

lid

ofan

experimentalglaucomamodel

(rabbit).IOPwas

assessed

Nodifference

wasfoun

din

cellviability

betweenlatanoprost-loaded

hydrogel

groupandthecontrols

Nocytotoxiceffectsweredetected

on

rabbitcornealepithelialcells

Latanoprost-lo

aded

hydrogelinstilled

once

aweekshow

edasimilarIO

P-

loweringeffect

ofcommercial

latanoprostinstilled

once

daily

[166]

Latanoprost-lo

aded

liposom

es,

thym

oquinone-lo

aded

liposom

es,and

latanoprost/thym

oquinone-lo

aded

liposom

es

Liposom

esIn

vitrodrug

release

Invivo

studies:glaucomatouswhite

albino

rabbitsweretreatedwith

latanoprosteyedropsanddiverse

liposom

eform

ulations

foraperiod

of

6weeks

Latanoprost/thymoquinone-lo

aded

liposom

esandlatanoprost-loaded

liposom

eswereableto

providea

significant

IOPloweringthat

lasted

8h

Effectof

thefree

latanoprostdidnot

persistformorethan

24h

[167]

Adv Ther (2020) 37:155–199 181

Page 28: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

Table7

continued

Hypotensive

drug/nanosystem

Pharm

aceuticalform

Stud

ydesign/m

odel

Results

References

Latanoprost-propylamino-b-

cyclodextrin

(CD)

Cyclodextrins

Invitrostability

andphasesolubility

analyses

Exvivo

cornealperm

eation

studies

Invivo

ocular

tolerabilityevaluation

Histology

study

Latanoprost-propylamino-b-CD

demonstratedasignificant

improvem

entin

itssolubilityand

stability

Clin

icalevaluationsduring

14days

show

edthat

ocular

irritation

was

15.5%

withthelatanoprostmarketed

form

ulation,

9.5%

withthe

latanoprost-propylam

ino-b-CD

form

ulation,

and7.1%

withthe

vehicleof

theform

ulations

Histologicalevaluation

ofocular

tissues

demonstratedthat

Xalatan

�induced

higher

inflammatorycellinfiltrates

than

latanoprost-propylam

ino-b-CD

form

ulationandthevehicle

[168]

Chitosanbimatoprost(BIM

)-loaded

inserts

Hydrogel

Invitrodrug

releasestudies

Biodistribution

studies(freeand

entrappedBIM

,radiolabeledwith

technetium

-99m

)

Invivo

studies(glaucom

ainducedin

Wistarrats):

Group

1:BIM

-loaded

insertswere

administeredonce

into

the

conjun

ctivalsac

Group

2:marketedBIM

drops

Group

3:placeboinserts

Biodistribution

studiesshow

edthat

a

higher

amount

of99mTc-BIM

remainedin

theeyeafterchitosan

insertim

plantation

comparedto

eye

drop

instillation

BIM

-loaded

insertswereableto

lower

IOPfor4weeks

afteroneapplication,

whereasmarketedeyedrop

couldonly

lower

IOPfor1day

[169]

182 Adv Ther (2020) 37:155–199

Page 29: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

Table7

continued

Hypotensive

drug/nanosystem

Pharm

aceuticalform

Stud

ydesign/m

odel

Results

References

Adrug-agnosticintraocularly

implantabledevice

was

used

loaded

withbimatoprost.T

hedevice

was

callednanofluidicVitrealSystem

for

TherapeuticAdm

inistration

(nViSTA)

Implantableintraoculardevice

usinga

nanochannelmem

brane

The

nViSTAim

plantabledevice

was

designed

forsustainedandcontrolled

drug

deliveryandbasedon

a

nanochannelmem

brane(which

measuresfrom

2.5to

250nm

),

without

theneed

foractuation,

pumps,o

rrepeated

clinical

intervention

Thisdevice

was

tested

withina

3-dimensionalanatom

icallysimilarin

silicohuman

eyemodelto

obtain

inform

ationon

theintraocular

pharmacokineticprofile

Resultsfrom

invitrotesting

demonstratedaburstof

approxim

ately40

lgof

bimatoprost

during

thefirst2days,followed

bya

sustainedreleaseof

bimatoprostfrom

thenV

iSTA

over

thesubsequent

18days

[170]

Bim

atoprost-lo

aded

nanosponges(N

S);

travoprost-lo

aded

nanosponges(N

S)

Nanosponge

One

travoprostnanosponge

form

ulation(50-nm

),and3

bimatoprostnanosponge

(NS)

form

ulations

weretested:(a)400-nm

NS;

(b)700-nm

NSwitham

orphous

(A-N

S)cross-lin

kers,and

(c)700-nm

NSwitham

orphous/crystalline

(AC-

NS)

cross-lin

kers

Ocularhypertensive

C57

micereceived

NSloaded

with2prostaglandin

analogue

hypotensivedrugs

(travoprostor

bimatoprost)by

a

singleintravitrealinjection

IOPwas

monitored

for7weeks

Toevaluate

thepossibility

ofretinal

deposition

andretinalganglioncell

uptake

ofNS,50-nm

NSloaded

with

Neuro-D

iOwas

injected

intravitreally

TravoprostNSform

ulationlowered

IOP19–2

9%forup

to4days

comparedto

salin

einjection

Outcomes

ofthe3bimatoprostNS

form

ulations

were:

400nm

NSlowered

IOP24–3

3%for

upto

17days

comparedto

salin

e

700nm

A-N

Slowered

IOP22–3

2%

forup

to32

days

700nm

AC-N

Slowered

IOP18–2

6%

forup

to32

days

Confocalmicroscopyandorthogonal

projectionssuggestedinternalization

ofNeuro-D

iOby

retinalganglion

cells,w

hich

means

NSmay

beeffective

attargetingthesecells

[171]

Adv Ther (2020) 37:155–199 183

Page 30: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

GLAUCOMA DRAINAGEAND NANODEVICES

Glaucoma drainage implants are oftenemployed in glaucoma care when medicaltherapy, laser procedures, and standard filtra-tion surgery fail to sufficiently control IOP[172]. The main types of glaucoma drainagedevices available commercially are the Molteno,the Baerveldt, and the Ahmed, all of themdesigned to create an alternate aqueous path-way by channeling aqueous humor from theanterior chamber to the collection plate posi-tioned under the conjunctiva [173, 174].

The long-term success of glaucoma drainageimplants, however, is reduced as a result of thefibrous reaction developing around the end-plate, which leads to encapsulation and reduc-tion of aqueous humor absorption. Epstein[175] was the first author to consider thataqueous humor in the subconjunctival space, initself, would be a stimulus for the fibrovascularproliferation in the episcleral tissue. The inten-sity of the fibrotic reaction was attributed to amultifactorial origin, although not all patho-genetic factors are well understood. Some ele-ments, such as the type of biomaterial, designand/or size of the end-plate, and the patient’simmune response, certainly influence the for-mation of fibrous tissue around the valve plate[176]. Therefore, refinement of biomaterials, aswell as the associated use of antifibrotic agentswith more advanced drug delivery nanosystemsmay meaningfully improve the long-term suc-cess of glaucoma drainage devices.

A novel double-layered porous coating forAhmed glaucoma valves based on biodegrad-able poly(lactic-co-glycolic acid) (PLGA) wasdeveloped by Ponnusamy et al. [177] to achievecontinuous drug release of antifibrotic agents[mitomycin C (MMC) and/or 5-fluorouracil (5-FU)] to the subconjunctival space. This novelporiferous coating for the Ahmed glaucomadrainage implant may have the advantage ofmodifying the degradation pattern of the poly-mer, and the drug release features. The purposeof this double-layered film is to provide a burstof MMC release followed by a slow release of5-FU, which would prevent fibroblast

proliferation over the most active period ofpostoperative wound healing (0–28 days).Results showed a very rapid release of MMCwithin 1 day, followed by the initial release of5-FU within the first 3–5 days, which lasted for 3to 4 weeks. Cytotoxicity assays have demon-strated significant cytotoxic activity from day 1which persisted until the PLGA film was almosttotally degraded.

Pan et al. [178] were able to manufacture anano-drainage device fabricated throughmicroelectromechanical systems (MEMS;miniaturized devices containing submillimeterfeatures). This nano-drainage implant is madeof a nanoporous membrane (mimicking thedrainage function of the trabecular meshwork)connected to an integrated polymeric shaftinserted through the sclera into the anteriorchamber, thus enabling a bypass route for theaqueous humor outflow. The nano-filtrationmembrane provides the designed flow resis-tance to the aqueous humor flow, but cloggingof proteins inside the nanopores from plasmaperfusion significantly increased the resistance.Further efforts will be necessary to apply modi-fications to the nanoporous membrane to reachthe ideal aqueous outflow.

Harake et al. [179] designed an anti-biofoul-ing micro-device which provides an innovativeplatform, also using MEMS, for IOP reduction inpatients with glaucoma. This novel device hasan array of parallel micro-channels built insideto yield controlled aqueous outflow resistance.Polyethylene glycol (PEG), a synthetic hydrogel,was chosen as the primary device materialowing to its favorable biocompatibility and itsantifouling properties, aiming to reduce proteinclogging of the micro-channels [180]. PEG 214and PEG 4000 were jointly used to avoid chan-nel swelling. In vitro, continuous testing underartificial flow conditions showed that the devicedesign containing 23 channels provided apressure drop of about 10 mmHg.

Ferrofluids are the colloidal mixtures ofmagnetic nanoparticles (ferri- or ferromagneticparticles) suspended in a non-magnetic, inertfluid, such as water or an organic solvent [181].As a result of their nanosize (tipically10–100 nm in diameter), the nanoparticles aresubject to Brownian motion. Brownian motion

184 Adv Ther (2020) 37:155–199

Page 31: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

or pedesis is the random movement of micro-scopic particles in suspension mixed in a fluid,and it results from the averaged impacts of fast-moving molecules of the surrounding medium[182]. Magnetite (Fe3O4) particles are composedof a single magnetic domain and show super-paramagnetic properties, i.e., a drag force alongfield gradients. If a static magnetic field exists,the interfacial force causes the ferrofluid toconform to its boundaries. Accordingly, a fer-rofluid in a capillary tube can operate as an on/off valve to flow through capillary blockade. Apermanent magnet may be utilized to keep theferrofluid in the capillary and a secondarymagnet may be utilized to regulate the forcenecessary to form the capillary barrier. When-ever the pressure exerted on the ferrofluid bythe liquid surpasses the magnetic force betweenthe ferrofluid and the secondary magnet, flowcan start after the barrier is broken. Paschaliset al. [183] designed a novel, replaceable, ferro-magnetic valve tube able to afford pressureregulation without the requirement of subcon-junctival encapsulation. Preliminary in vivoresults in rabbits showed that during the2 weeks of follow-up, there were no signs ofinflammation or infection at the surgical site.The mean IOP value in the valve implanted eye(11.8 ± 2 mmHg) was significantly lower thanthe contralateral control eye (14 ± 3 mmHg),and a continuous flow at the outlet tip of thevalve was observed until the last follow-up.

Biomaterial improvement provided bynanoparticle-coated glaucoma drainage devices,associated with design changes and innovativedrug delivery systems may potentially reducefibrous reaction, and improve their efficiency.

NANOPARTICLE-BASEDANTIMETABOLITES AND WOUNDHEALING MODULATION

Currently, trabeculectomy is still the goldstandard surgical treatment for glaucoma.Postoperative wound healing produces ahypercellular response in the subconjunctivaltissues, which may gradually close the filtrationsite, limiting its long-term success [184]. Sub-conjunctival antifibrotics, such as MMC and

5-FU, have been used to reduce the generationof scar tissue, by inhibiting fibroblast prolifera-tion [184–187].

In order to improve the efficiency of theseantifibrotics drugs, nanoformulations havebeen recently employed. Hou et al. [188] havedeveloped a new method to prepare MMC-loa-ded polylactic acid (PLA) nanoparticles,employing soybean phosphatidylcholine toimprove the liposolubility of MMC. Resultsshowed refined formulation characteristics andlonger sustained drug release. Gomes dos Santoset al. [189] designed nanosized complexes ofantisense TGFb2 phosphorothioate oligonu-cleotides (PS-ODN) with polyethylenimine(PEI), and naked PS-ODN encapsulated intopoly(lactide-co-glycolide) microspheres, basedon the hypothesis that the encapsulation ofantisense TGFb2 ODN-PEI nanosized complexesin different types of porous particles could be anefficacious system for oligonucleotide deliveryin vivo. Results showed that the MS-AsPS-ODN-PEI nanosized complex significantly improvedthe bleb survival rate following trabeculectomy(100% at 42 days) in rabbits compared to con-trols (0% of survival at day 21). The improvedefficiency of the complexes released frommicrospheres is attributed to a superior intra-cellular delivery in conjunctival cells, alongwith the inhibition of TGFb2.

Mitomycin C is considered the most effectiveantifibrotic agent used in glaucoma filteringsurgery [190]. It is used in more than 85% of thetrabeculectomies, but severe adverse effects,such as conjunctival thinning, bleb leaks,hypotony, and infection, limit its safety [190].Our group has been working on the develop-ment of a safer and effective alternativenanoparticle-based antifibrotic agent. Paclitaxelassociated with lipid nanoemulsions (LDE-PTX)was tested on rabbits undergoing trabeculec-tomy [191]. Subconjunctival injection of LDE-PTX was as effective as MMC in preventingscarring after trabeculectomy, but with sub-stantially less toxicity to the conjunctiva andciliary body. After these promising results in theanimal model, we have recently started the firstclinical trial to investigate the effectiveness ofLDE-PTX in patients with primary open-angleglaucoma (POAG) undergoing trabeculectomy.

Adv Ther (2020) 37:155–199 185

Page 32: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

Duan et al. [192] investigated the effects ofIkappaB kinase subunit beta (IKKb) inhibitionusing RNA interference (RNAi) technology onthe proliferation of human Tenon’s capsulefibroblasts (HTFs), which have a fundamentalrole in the scarring process. Ye et al. [193]developed a small interfering RNA (siRNA) tar-geting IKKb (IKKb-siRNA) associated with aternary cationic nano-copolymer called CS-g-(PEI-b-mPEG) as the vehicle delivered intoHTFs. They found that the expression of IKKbwas downregulated, the activation of nuclearfactor kappa B (NF-jB) in the HTFs was inhib-ited, and the proliferation of HTFs was sup-pressed through the blocking of the NF-jBpathway, effects that improved the surgicaloutcome in a non-human primate model oftrabeculectomy.

There is increasing interest in the use ofangiogenesis-inhibiting compounds, especiallythose that act against vascular endothelial growthfactor (VEGF), aiming to modulate the effects ofVEGF on the migration and proliferation ofhuman fibroblasts in Tenon’s capsule [194]. VEGFis upregulated in the aqueous humor of theglaucoma rabbit model as well as in patients withglaucoma, and it stimulates fibroblast prolifera-tion in vitro [195]. Bevacizumab is a full-lengthhumanized non-selective monoclonal antibodydirected against all isoforms of VEGFA, which hasbeen reported to inhibit proliferation of fibrob-lasts in vitro and to reduce angiogenesis andcollagen deposition in eyes undergoing tra-beculectomy [194–197]. Han et al. [198] investi-gated the use of bevacizumab combined withPEG-PCL-PEG (PECE) hydrogel, by intracameralinjection, after experimental glaucoma filtrationsurgery (GFS). Results demonstrated that theantifibrotic effect of bevacizumab-loaded PECEhydrogel was superior to bevacizumab alone inthe prevention of postoperative scarring after GFSin the rabbits.

NEUROPROTECTIONAND NEUROREGENERATIONAND NEUROTROPHIC FACTORS

Although IOP is considered the main risk factorfor the development and progression of

glaucoma, RGC loss may continue in spite ofIOP reduction in some patients with glaucoma[199, 200]. The physiopathology behind RGCdeath is complex and has been attributed tovarious factors, including oxidative stress,intermittent ischemia, defective axon transport,excitotoxicity, loss of electrical activity, andtrophic factor withdrawal [16], which justifiesthe development of neuroprotection/neurore-generation strategies.

Glial cell-derived neurotrophic factor(GDNF) is an important neurotrophic factorimplicated in the growth, differentiation,maintenance, and regeneration of specific neu-ronal populations in the adult brain, as well asin peripheral neurons [201].

In vitro and in vivo studies have shown thatGDNF may rescue photoreceptor and RGCfunctions during retinal degeneration [202].Previous studies have demonstrated thatexogenous GDNF is an interesting therapeuticapproach for neurodegenerative diseases affect-ing the retina, including glaucoma [203].Checa-Casalengua et al. [203] described a novelprotein microencapsulation method to ensureprotection of the biological factor during thisprocedure, allowing the controlled release ofproteins to keep their bioactivity for prolongedperiods of time. GDNF in combination withvitamin E released from the microspheres wasactive for at least 3 months after a singleintravitreal injection in an animal model ofglaucoma. There was a significant increase ofRGC survival compared with GDNF alone,vitamin E alone, or blank microspheres, whichindicates that this novel formulation may beclinically applicable as a neuroprotective tool inthe treatment of glaucomatous optic neuropa-thy. Ward et al. [204] demonstrated long-termRGC survival in a spontaneous glaucoma ani-mal model by injecting slow-release PLGAmicrospheres containing GDNF into the vitre-ous. Early treatment resulted in 3.5 timesgreater RGC density than untreated mice after15 months.

In a rat glaucoma model, animals were trea-ted with GDNF injected into their vitreouscavity. 10% GDNF-microsphere emulsionresulted in a significant reduction of optic nervehead excavation, and increased RGC survival,

186 Adv Ther (2020) 37:155–199

Page 33: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

compared with 2% GDNF-microspheres, blankmicrospheres, and saline solution [205]. Inorder to evaluate the pharmacokinetics ofGDNF, Garcıa-Caballero et al. [206] analyzed itslevels after a single intravitreal injection ofGDNF/vitamin E microspheres in rabbits. Thismethod allowed a sustained controlled releaseof GDNF for up to 6 months.

Ciliary neurotrophic factor (CNTF) has alsobeen shown to be neuroprotective on motorneurons following injury to the adult centralnervous system [207], and also for neurons inother regions in degenerative diseases[208–210]. Effective neuroprotection could bereached if sustained and local delivery withoutadverse side effects were achievable, but sus-tained CNTF delivery has proven arduous toaccomplish and control. Nkansah et al. [211]examined the effects of CNTF nanospheres onneural stem cells (NSC) and also on the factorsthat influence controlled CNTF delivery frommicrospheres and nanospheres. Protein bioac-tivity after encapsulation was studied treatingneural stem cells with CNTF released fromnanospheres and compared to those treatedwith unencapsulated CNTF. Results showedthat encapsulated CNTF provided NSC differ-entiation with no loss of potency compared tothe unencapsulated growth factor.

In addition, Pease et al. [212] demonstratedthe neuroprotective effect of virally mediatedoverexpression of CTNF and brain-derivedneurotrophic factor (BDNF) in experimentalglaucoma. Injection of adeno-associated viralvectors carrying either BDNF, CTNF, or both, inlaser-induced glaucoma eyes, was performed.Combined CNTF-BDNF and BDNF overexpres-sion alone had no noticeable improvement inRGC axon survival, but CNTF alone showed asignificant protective effect, with 15% less RGCdeath.

The induction of heat shock proteins (HSPs)has been investigated as a modality for theprotection of optic nerve damage in glaucoma[213–215]. Caprioli et al. [214] conducted thefirst demonstration of the neuroprotectiveeffects of 72-kDa HSPs in cultured RGCs andMuller cells after hyperthermia and sublethalhypoxia. Since then, many researchers havebeen attempting to apply the induction of HSPs

as a neuroprotective strategy in patients withglaucoma.

The calcium (Ca2?)-binding protein onco-modulin is a potent macrophage-derivedgrowth factor for RGCs and other neurons. Yinet al. [216] demonstrated that, in vivo, onco-modulin released from nanoparticles (micro-spheres) promotes regeneration in the maturerat optic nerve.

More recently, Jeun et al. [217] investigated anew approach to use localized HSPs—a promis-ing treatment modality for the ocular neuro-protection—without the unwanted side effectstypically occurring with the current clinicalapproaches to induce HSPs in RGCs. Theseauthors proposed the induction of HSPs by localmagnetic hyperthermia utilizing engineeredsuperparamagnetic Mn0.5Zn0.5Fe2O4 nanoparti-cle agents (EMZF-SPNPAs). The results showedthat the EMZF-SPNPAs possess all the idealcharacteristics as an in vivo localized HSPinduction agent. Moreover, the authors alsoused an innovative infusion technique, whichdelivers the silica-coated EMZF-SPNPAs throughthe vitreous body to the retina forneuroprotection.

ANTI-INFLAMMATORY DRUGS

Inflammation is a common physiological pro-tective response to injury. Inflammation isgenerally beneficial to the organism, promotingtissue survival, repair, and recovery, providedthat it occurs for a limited period of time.Otherwise, it may be detrimental when exten-sive, prolonged, or unregulated [218]. In thecentral nervous system, the activation of astro-cytes and microglia, which is indicative ofinflammation, occurs in patients with Parkin-son’s, Alzheimer’s, Huntington’s diseases, mul-tiple sclerosis, and amyotrophic lateral sclerosis[219–221]. Hence, in the nervous system, pro-longed inflammation may have a dual role,increasing neuronal loss or impeding regenera-tion. This justifies why anti-inflammatory drugsmight also have a neuroprotective activity.

In glaucoma, the neuroinflammatoryresponses during RGC degeneration have notbeen elucidated. However, microglia are

Adv Ther (2020) 37:155–199 187

Page 34: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

pleiotrophic immune cells and seem to take partin both neuroprotection and neurodegenera-tion of RGCs [222]. Thus, it is plausible thatlow-grade inflammation may occur in glaucomain response to hypoxia, ischemia, vascular dys-function, and elevated IOP. In fact, genes asso-ciated with cyclooxygenase 2 (COX-2)/PGE2[223, 224], cytokines [225], tumor necrosis fac-tor alpha (TNFa) in glial cells [226], and hypoxiainducible factor 1-alpha (HIFa) [227] are allupregulated in glaucomatous RGCs.

Curcumin [1,7-bis-(4-hydroxy-3-methox-yphenyl)-1,6-heptadiene-3,5-dione] is apolyphenol extracted from Curcuma longa [228].It has been reported that this compound canbeneficially modulate several biochemical pro-cesses involved in neurodegenerative diseases.For example, Dong et al. [229] showed thatlong-term (12-week) curcumin-supplementeddiet increased hippocampal neurogenesis andcognitive function in aged rats. Similarly, Kimet al. [230] demonstrated the beneficial effectsof low dose curcumin on mouse multi-potentneural progenitor cells, which means it canstimulate neural plasticity and repair. Further-more, Belviranlı et al. [231] concluded thatcurcumin supplementation improves cognitivefunction in aged female rats by reducing thelipid peroxidation in brain tissue, whichdemonstrates its protective effect against neuraloxidative stress.

Curcumin has been reported to protect RGCsand the microvasculature against ischemicdamage via inhibition of NF-jB, signal trans-ducer and activator of transcription 3 (STAT3),and monocyte chemotactic protein 1 (MCP-1;also known as C-C motif chemokine 2) overex-pression [232]. Wang et al. conducted a study toinvestigate the ability of curcumin to inhibitretinal ischemia/reperfusion injury. Pretreat-ment with curcumin inhibited ischemia/reper-fusion-induced cell loss in the ganglion celllayer. Also, 0.05% curcumin administered2 days after the injury showed a vasoprotectiveeffect [232]. On the basis of the hypothesis thatlocal and systemic oxidative stress participate inthe pathogenesis of glaucoma, Yue et al. [233]evaluated the antioxidant effects of curcuminboth in vitro (BV-2 microglia cell line) andin vivo and found that curcumin may improve

cell viability and decrease intracellular reactiveoxygen species and apoptosis of RGCs.

Although the therapeutic potential of cur-cumin in ophthalmology is real, its poor watersolubility [234], and low bioavailability[228, 235] are important limiting factors forclinical applicability. To surpass these limita-tions, Davis et al. [236] used a nanotechnologyapproach to provide a hydrophobic environ-ment for a poorly soluble molecule such ascurcumin, and to improve its bioavailabilitythrough the development of a nanocarriersuitable for utilization as a topical formulation.This nanoformulation was able to increase thesolubility of curcumin by a factor of 400,000,which is more than enough to overcome natu-ral ocular barriers. Topical application of cur-cumin-loaded nanocarriers twice-daily for3 weeks, in in vivo models of ocular hyperten-sion and partial optic nerve transection, signif-icantly reduced RGC loss. These results suggestthat topical curcumin nanocarriers havepotential as a neuroprotective therapy inglaucoma.

Ketorolac is a synthetic pyrrolizine car-boxylic acid derivative that belongs to thegroup of NSAIDs. Ketorolac is a non-selectiveinhibitor of the enzymes COX-1 and COX-2.The inhibition of COX-2, upregulated at sites ofinflammation, prevents conversion of arachi-donic acid to pro-inflammatory prostaglandins[237]. Cyclooxygenases are expressed by RGCsin the rodent retina [238] and are upregulatedin the retina after optic nerve injury [239] andischemia [240]. Nadal-Nicolas et al. [241] firstdescribed the neuroprotective effects of ketoro-lac on RGCs after optic nerve axotomy in rats.Two treatments were evaluated: intravitrealadministration of ketorolac tromethaminesolution and/or ketorolac-loaded PLGA micro-spheres, 1 week before the optic nerve lesionand intravitreal administration right after theoptic nerve crush. In all treated groups therewas a significant increase in the number ofRGCs compared to all control groups, whichconfirmed that an NSAID can delay neuronaldeath in an acute model of axonal injury. Pre-treatment (ketorolac solution administered pre-optic nerve lesion) resulted in 63% survival ofRGCs, while simultaneous administration of

188 Adv Ther (2020) 37:155–199

Page 35: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

ketorolac solution provided a 53% survival.However, in this study, microspheres did notshow an additional effect, contrary to otherinvestigations where intravitreal administrationof the microparticle formulation was found tobe advantageous [242].

TOXICITY OF NANOMATERIALS

Nanoparticles may generate cellular toxicitythrough oxidative stress, interaction with thecell membrane, and inflammation [243]. Incontrast to chemical toxicity, where compoundpurity and drug concentration are the essentialdeterminants, in nanotoxicity other character-istics should be taken into account, such asnanoparticle size, surface shape, and ioniccharge [244]. In addition, there are a few toxi-city forms that may be independent of the drugdelivered, namely particle toxicity, excipienttoxicity, contaminant toxicity, and inflamma-tory toxicity [243]. Each one must be indepen-dently investigated in vitro and in vivo.

Specific literature about the toxicity of ther-apeutic nanoparticles in the eye remains scarce[243]. Nanotoxicity is a subject of key impor-tance to be determined in conjunction with thetherapeutic potential of each nanoparticle[245, 246], particularly for those that requirerepeat-dose regimens [247]. All nanoparticlesproposed to be utilized for therapeutic purposesmust be thoroughly investigated in terms oflocal and systemic toxicity, before obtaining thefull regulatory approval.

CONCLUSIONS AND PERSPECTIVES

Glaucoma is a complex and multifactorial dis-ease and truly effective treatments need to bedeveloped and specifically targeted to thepathophysiological pathways that have beenidentified to date. We believe that, in the future,glaucoma treatment will include the use ofhypotensive drugs associated with neuropro-tective agents directed to the preservation ofRGCs and the optic nerve.

Nanotechnology has been shown to be arobust and valuable modality for the treatment

of ocular diseases, including glaucoma. In thisreview, we focused on the recent advances inthe development of nanosystems for glaucomatreatment. Bearing in mind the desired thera-peutic targets in glaucoma therapy, nanotech-nology has the ability to meaningfully improvethe efficacy of current treatment approaches. Byimproving the pharmacological characteristicsof drugs, or the features of surgical devices,nanotechnology has remarkable potential infuture glaucoma management. There is con-vincing experimental evidence that manynanoparticles may enhance the solubility oflipophilic drugs, which improves bioavailabil-ity, and promote sustained delivery owing totheir optimized biodegradability and physico-chemical characteristics. Nanomedicine formu-lations designed to lower IOP may not requirefrequent dosing and may reduce drug-relatedside effects, which ultimately will result inoptimizing adherence. In addition, nanofor-mulated drugs have been shown to improvewound healing modulation following filteringsurgery and at the same time reduce the toxicityto the conjunctiva and ciliary body. Nanopar-ticle-coated filtering drainage devices may alsoreduce the fibrotic reaction around the plate,increasing their long-term success.

Although promising, possible toxic effects ofnanoparticles have to be thoroughly analyzedduring preclinical evaluation. The detailedinvestigation of every possible biological reper-cussion is requisite to the future introduction ofnanomaterials. Importantly, the vast majorityof studies on the use of nanoparticles in glau-coma have been performed in animal models.Human studies are needed not only for thedetailed determination of the actual cytotoxic-ity of each nanoparticle but also to investigatetheir tolerability and efficacy.

ACKNOWLEDGEMENTS

Funding. No funding or sponsorship wasreceived for this study or publication of thisarticle.

Adv Ther (2020) 37:155–199 189

Page 36: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

Authorship. All named authors meet theInternational Committee of Medical JournalEditors (ICMJE) criteria for authorship for thisarticle, take responsibility for the integrity ofthe work as a whole and have given theirapproval for this version to be published.

Disclosures. Marcelo Luıs Occhiutto, andRaul Cavalcante Maranhao have nothing todisclose. Vital Paulino Costa has receivedresearch funding from Allergan, Alcon, Novar-tis; honoraria from Allergan, Alcon, Novartis,Aerie, Genom, Ofta, Iridex; and congressexpenses from Allergan, Alcon and Novartis.Anastasios-Georgios Konstas has receivedresearch funding from Allergan, Bayer andSanten; honoraria from Allergan, Mundipharmaand Santen; and congress expenses from Via-nex. Anastasios-Georgios Konstas is a memberof the journal’s Editorial Board.

Compliance with Ethics Guidelines. Thisarticle is based on previously conducted studiesand does not contain any studies with humanparticipants or animals performed by any of theauthors.

Data Availability. Data sharing is notapplicable to this article as no data sets weregenerated or analyzed during the current study.

Open Access. This article is distributedunder the terms of the Creative CommonsAttribution-NonCommercial 4.0 InternationalLicense (http://creativecommons.org/licenses/by-nc/4.0/), which permits any noncommer-cial use, distribution, and reproduction in anymedium, provided you give appropriate creditto the original author(s) and the source, providea link to the Creative Commons license, andindicate if changes were made.

REFERENCES

1. Thylefors B, Negrel AD. The global impact of glau-coma. Bull World Health Organ. 1994;72(3):323–6.

2. Resnikoff S, Pascolini D, Etya’ale D, et al. Globaldata on visual impairment in the year 2002. BullWorld Health Organ. 2004;82(11):844–51.

3. WHO. The global impact of glaucoma. Bull WorldHealth Organ. 1994;72(3):323–6. http://www.who.int/blindness/publications/glaucoma/en/. Accessed7 Dec 2019.

4. Stein JD, Lee PP. Screening for glaucoma. In: YanoffM, Duker JS, editors. Ophthalmology. Amsterdan,The Netherlands: Elsevier Sanders; 2009, p. 1007.

5. Heiji A, Leske MC, Bengtsson B, et al. Reduction ofintraocular pressure and glaucoma progression:results from the Early Manifest Glaucoma Trial.Arch Ophthalmol. 2002;120(10):1268–79.

6. The AGIS Investigators. The Advanced GlaucomaIntervention Study (AGIS): 7. The relationshipbetween control of intraocular pressure and visualfield deterioration Am J Ophthalmol. 2000;130(4):429–40.

7. Medeiros FA, Weinreb RN. Medical backgrounders:glaucoma. Drugs Today (Barc). 2002;38(8):563–70.

8. Kumar H, Mansoori T, Warjri GB, et al. Lasers inglaucoma. Indian J Ophthalmol. 2018;66(11):1539–53.

9. Abrams KL. Medical and surgical management ofthe glaucoma patient. Clin Tech Small Anim Pract.2001;16(1):71–6.

10. Levin LA, Crowe ME, Quigley HA. Lasker/IRRF ini-tiative on astrocytes and glaucomatous neurode-generation participants. Neuroprotection forglaucoma: requirements for clinical translation. ExpEye Res. 2017;157:34–7.

11. Cetinel S, Montemagno C. Nanotechnology appli-cations for glaucoma. Asia Pac J Ophthalmol(Phila). 2016;5(1):70–8.

12. Goyal G, Garg T, Rath G, et al. Current nanotech-nological strategies for treating glaucoma. Crit RevTher Drug Carrier Syst. 2014;31(5):365–405.

13. Cardigos J, Ferreira Q, Crisostomo S, et al. Nan-otechnology-ocular devices for glaucoma treat-ment: a literature review. Curr Eye Res. 2019;44(2):111–7.

14. Foldvari M. Noninvasive ocular drug delivery:potential transcorneal and other alternative deliv-ery routes for therapeutic molecules in glaucoma.J Glaucoma. 2014;23(8 Suppl 1):S80–2.

15. Kim NJ, Harris A, Gerber A, et al. Nanotechnologyand glaucoma: a review of the potential

190 Adv Ther (2020) 37:155–199

Page 37: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

implications of glaucoma nanomedicine. Br J Oph-thalmol. 2014;98(4):427–31.

16. Chang EE, Goldberg JL. Glaucoma 2.0: neuropro-tection, neuroregeneration, neuroenhancement.Ophthalmology. 2012;119:979–86.

17. Quigley HA. Glaucoma. Lancet. 2011;377(9774):1367–77.

18. Agarwal R, Gupta SK, Agarwal P, et al. Currentconcepts in the pathophysiology of glaucoma.Indian J Ophthalmol. 2009;57(4):257–66.

19. Weinreb RN, Khaw PT. Primary open-angle glau-coma. Lancet. 2004;363:1711–20.

20. Reardon G, Kotak S, Schwartz GF. Objective assess-ment of compliance and persistence amongpatients treated for glaucoma and ocular hyperten-sion: a systematic review. Patient Prefer Adherence.2011;5:441–63.

21. Gurwitz JH, Glynn RJ, Monane M, et al. Treatmentfor glaucoma: adherence by the elderly. Am J PublicHealth. 1993;83:711–6.

22. Sleath B, Robin AL, Covert D, et al. Patient-reportedbehavior and problems in using glaucoma medica-tions. Ophthalmology. 2006;113(3):431–6.

23. Loch C, Zakelj S, Kristl A, et al. Determination ofpermeability coefficients of ophthalmic drugsthrough different layers of porcine, rabbit andbovine eyes. Eur J Pharm Sci. 2012;47:131–8.

24. Chiang CH, Schoenwald RD. Ocular pharmacoki-netic models of clonidine-3H hydrochloride. J Phar-macokinet Biopharm. 1986;14(2):175–211.

25. Schoenwald RD. Ocular pharmacokinetics/pharma-codynamics. In: Mitra AK, editor. Ophthalmic drugdelivery systems. 2nd ed. New York: Dekker, Inc.;2003.

26. Cunha-Vaz J. The blood-ocular barriers. Surv Oph-thalmol. 1979;23(5):279–96.

27. Del Amo EM, Rimpela AK, Heikkinen E, et al.Pharmacokinetic aspects of retinal drug delivery.Prog Retin Eye Res. 2017;57:134–85.

28. Shikamura Y, Yamazaki Y, Matsunaga T, et al.Hydrogel ring for topical drug delivery to the ocularposterior segment. Curr Eye Res. 2016;41:653–61.

29. Agrahari V, Mandal A, Agrahari V, et al. A compre-hensive insight on ocular pharmacokinetics. DrugDeliv Transl Res. 2016;6:735–54.

30. Pelkonen L, Tengvall-Unadike U, Ruponen M, et al.Melanin binding study of clinical drugs with

cassette dosing and rapid equilibrium dialysisinserts. Eur J Pharm Sci. 2017;109:162–8.

31. Carreon TA, Edwards G, Wang H, et al. Segmentaloutflow of aqueous humor in mouse and human.Exp Eye Res. 2017;158:59–66.

32. Johnson M, McLaren JW, Overby DR. Unconven-tional aqueous humor outflow: a review. Exp EyeRes. 2017;158:94–111.

33. Yucel YH. Discovery of lymphatics in the humaneye and implications. Can J Ophthalmol.2010;45(2):115–7.

34. Tomczyk-Socha M, Turno-Krecicka A. A novel uve-olymphatic drainage pathway-possible new targetfor glaucoma treatment. Lymphat Res Biol.2017;15(4):360–3.

35. Yucel Y, Gupta N. Lymphatic drainage from the eye:a new target for therapy. Prog Brain Res. 2015;220:185–98.

36. Lee SJ, Kim SJ, Kim ES, et al. Trans-scleral perme-ability of Oregon Green 488. J Ocul PharmacolTher. 2008;24:579–86.

37. Del Amo EM, Urtti A. Rabbit as an animal model forintravitreal pharmacokinetics: clinical predictabil-ity and quality of the published data. Exp Eye Res.2015;137:111–24.

38. Ahmed I, Patton TF. Importance of the noncornealabsorption route in topical ophthalmic drug deliv-ery. Invest Ophthalmol Vis Sci. 1985;26:584–7.

39. Hamalainen KM, Kananen K, Auriola S, et al.Characterization of paracellular and aqueous pene-tration routes in cornea, conjunctiva, and sclera.Invest Ophthalmol Vis Sci. 1997;38:627–34.

40. Sponsel WE, Terry S, Khuu HD, Lam KW, Frenzel H.Periocular accumulation of timolol and betaxolol inpatients with glaucoma under long-term therapy.Surv Ophthalmol. 1999;43(suppl 1):S210–3.

41. Hollo G, Whitson JT, Faulkner R, et al. Concentra-tions of betaxolol in ocular tissues of patients withglaucoma and normal monkeys after 1 month oftopical ocular administration. Invest OphthalmolVis Sci. 2006;47(1):235–40.

42. Lavik E, Kuehn MH, Kwon YH. Novel drug deliverysystems for glaucoma. Eye (Lond). 2011;25:578–86.

43. Sena DF, Ramchand K, Lindsley K. Neuroprotectionfor treatment of glaucoma in adults. CochraneDatabase Syst. 2010;(2):CD006539.

44. Krupin T, Liebmann JM, Greenfield DS, et al. Arandomized trial of brimonidine versus timolol in

Adv Ther (2020) 37:155–199 191

Page 38: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

preserving visual function: results from the Low-Pressure Glaucoma Treatment Study. Am J Oph-thalmol. 2011;151:671–81.

45. Quigley HA. Clinical trials for glaucoma neuropro-tection are not impossible. Curr Opin Ophthalmol.2012;23:144–54.

46. Rosenfeld PJ, Brown DM, Heier JS, et al. Ranibizu-mab for neovascular age related macular degenera-tion. N Engl J Med. 2006;355:1419–31.

47. Dıaz-Coranguez M, Ramos C, Antonetti DA. Theinner blood-retinal barrier: cellular basis anddevelopment. Vision Res. 2017;139:123–37.

48. Liu G, Molas M, Grossmann GA, et al. Biologicalproperties of poly-L-lysine-DNA complexes gener-ated by cooperative binding of the polycation. J BiolChem. 2001;276(37):34379–87.

49. Nagarwal RC, Kant S, Singh PN, et al. Polymericnanoparticulate system: a potential approach forocular drug delivery. J Control Release. 2009;136(1):2–13.

50. Singh Y, Meher JG, Raval K, et al. Nanoemulsion:concepts, development and applications in drugdelivery. J Control Release. 2017;28(252):28–49.

51. Torchilin VP. Recent advances with liposomes aspharmaceutical carriers. Nat Rev Drug Discov.2005;4(2):145–60.

52. Akbarzadeh A, Khalilov R, Mostafavi E, et al. Role ofdendrimers in advanced drug delivery andbiomedical applications: a review. Exp Oncol.2018;40(3):178–83.

53. Cheng B, He H, Huang T, et al. Gold nanospheregated mesoporous silica nanoparticle responsive tonear-infrared light and redox potential as a thera-nostic platform for cancer therapy. J Biomed Nan-otechnol. 2016;12(3):435–49.

54. Gao W, Zhang Y, Zhang Q, et al. Nanoparticle–hy-drogel: a hybrid biomaterial system for localizeddrug delivery. Ann Biomed Eng. 2016;44(6):2049–61.

55. Lu Y, Qi J, Dong X, et al. The in vivo fate ofnanocrystals. Drug Discov Today. 2017;22(4):744–50.

56. Zhang Y, Ren K, He Z, et al. Development ofinclusion complex of brinzolamide with hydrox-ypropyl-b-cyclodextrin. Carbohydr Polym.2013;98(1):638–43.

57. Chaudhary HM, Duttagupta AS, Jadhav KR, et al.Nanodiamonds as a new horizon for

pharmaceutical and biomedical applications. CurrDrug Deliv. 2015;12(3):271–81.

58. Prajapati VD, Jani GK, Kapadia JR. Current knowl-edge on biodegradable microspheres in drug deliv-ery. Expert Opin Drug Deliv. 2015;12(8):1283–99.

59. Moghassemi S, Hadjizadeh A. Nano-niosomes asnanoscale drug delivery systems: an illustratedreview. J Control Release. 2014;10(185):22–36.

60. Haidar MK, Eroglu H. Nanofibers: new insights fordrug delivery and tissue engineering. Curr Top MedChem. 2017;17(13):1564–79.

61. Talevi A, Gantner ME, Ruiz ME. Applications ofnanosystems to anticancer drug therapy (Part I.Nanogels, nanospheres, nanocapsules). Recent PatAnticancer Drug Discov. 2014;9(1):83–98.

62. Priwitaningrum DL, Blonde JG, Sridhar A, et al.Tumor stroma-containing 3D spheroid arrays: a toolto study nanoparticle penetration. J ControlRelease. 2016;244(Pt B):257–68.

63. Hornung A, Poettler M, Friedrich RP, et al. Treat-ment efficiency of free and nanoparticle-loadedmitoxantrone for magnetic drug targeting in mul-ticellular tumor spheroids. Molecules. 2015;20(10):18016–30.

64. Yang L, Yin T, Liu Y, et al. Gold nanoparticle-cap-ped mesoporous silica-based H2O2-responsive con-trolled release system for Alzheimer’s diseasetreatment. Acta Biomater. 2016;46:177–90.

65. Muller RH, Radtke M, Wissing SA. Solid lipidnanoparticles (SLN) and nanostructured lipid carri-ers (NLC) in cosmetic and dermatological prepara-tions. Adv Drug Deliv Rev. 2002;54(Suppl. 1):S131–55.

66. Tamboli V, Mishra GP, Mitrat AK. Polymeric vectorsfor ocular gene delivery. Ther Deliv. 2011;2:523–36.

67. Wang J, Huang Y, David AE, et al. Magneticnanoparticles for MRI of brain tumors. Curr PharmBiotechnol. 2012;13(12):2403–16.

68. Chen J, Patil S, Seal S, et al. Rare earth nanoparticlesprevent retinal degeneration induced by intracel-lular peroxides. Nat Nanotechnol. 2006;1:142–50.

69. Salem HF, Ahmed SM, Omar MM. Liposomalflucytosine capped with gold nanoparticle formu-lations for improved ocular delivery. Drug DesDevel Ther. 2016;13(10):277–95.

70. Gref R, Domb A, Quellec P, et al. The controlledintravenous delivery of drugs using PEG-coatedsterically stabilized nanospheres. Adv Drug DelivRev. 1995;19:215–33.

192 Adv Ther (2020) 37:155–199

Page 39: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

71. Venishetty VK, Komuravelli R, Kuncha M, et al.Increased brain uptake of docetaxel and ketocona-zole loaded folate-grafted solid lipid nanoparticles.Nanomedicine. 2013;9(1):111–21.

72. Liu D, Lian Y, Fang Q, et al. Hyaluronic-acid-mod-ified lipid-polymer hybrid nanoparticles as an effi-cient ocular delivery platform for moxifloxacinhydrochloride. Int J Biol Macromol. 2018;17(116):1026–36.

73. Jonas JB, Aung T, Boune RR, et al. Glaucoma. Lan-cet. 2017;390:2083–93.

74. Chrai SS, Robinson JR. Corneal permeation oftopical pilocarpine nitrate in the rabbit. Am JOphthalmol. 1974;77(5):735–9.

75. Lazare R, Horlington M. Pilocarpine levels in theeyes of rabbits following topical application. ExpEye Res. 1975;21(3):281–7.

76. Plazoneet J, Grove M, Durr C, et al. Recent advancesin pilocarpine delivery. In: Saettone MF, Bucci M,Speiser P, editors. Ophthalmic drug delivery: bio-pharmaceutical, technological and clinical aspects.Padova: Springer & Liviana; 1987. p. 122.

77. Pepic I, Jalsenjak N, Jalsenjak I. Micellar solutions oftriblock copolymer surfactants with pilocarpine. IntJ Pharm. 2004;272:57–64.

78. Jarho P, Jarvinen K, Urtti A, et al. Modified beta-cyclodextrin (SBE7-beta-CyD) with viscous vehicleimproves the ocular delivery and tolerability ofpilocarpine prodrug in rabbits. J Pharm Pharmacol.1996;48(3):263–9.

79. Vandamme TF, Brobeck L. Poly(amidoamine) den-drimers as ophthalmic vehicles for ocular deliveryof pilocarpine nitrate and tropicamide. J ControlRelease. 2005;102(1):23–38.

80. Monem AS, Ali FM, Ismail MW. Prolonged effect ofliposomes encapsulating pilocarpine HCl in normaland glaucomatous rabbits. Int J Pharm. 2000;198:29–38.

81. Aktas Y, Unlu N, Orhan M, et al. Influence ofhydroxypropyl beta-cyclodextrin on the cornealpermeation of pilocarpine. Drug Dev Ind Pharm.2003;29(2):223–30.

82. Lee CH, Li YJ, Huang CC, et al. Poly(e-caprolactone)nanocapsule carriers with sustained drug release:single dose for long-term glaucoma treatment.Nanoscale. 2017;9(32):11754–64.

83. Casolaro M, Casolaro I, Lamponi S. Stimuli-re-sponsive hydrogels for controlled pilocarpine oculardelivery. Eur J Pharm Biopharm. 2012;80(3):553–61.

84. Li J, Wu L, Wu W, et al. A potential carrier based onliquid crystal nanoparticles for ophthalmic deliveryof pilocarpine nitrate. Int J Pharm. 2013;455(1–2):75–84.

85. Orasugh JT, Sarkar G, Saha NR, et al. Effect of cel-lulose nanocrystals on the performance of drugloaded in situ gelling thermo-responsive oph-thalmic formulations. Int J Biol Macromol.2019;1(124):235–45.

86. Kao HJ, Lin HR, Lo YL, et al. Characterization ofpilocarpine-loaded chitosan/Carbopol nanoparti-cles. J Pharm Pharmacol. 2006;58(2):179–86.

87. Heel RC, Brogden RN, Speight TM, Avery GF.Timolol: a review of its therapeutic efficacy in thetopical treatment of glaucoma. Drugs. 1979;17(1):38–55.

88. Fukuda M, Sasaki H. The transcorneal penetrationof commercial ophthalmic formulations containingtimolol maleate in rabbit eyes. J Ocul PharmacolTher. 2015;31(1):57–60.

89. Van Buskirk EM. Adverse reactions from timololadministration. Ophthalmology. 1980;87(5):447–50.

90. Maulvi FA, Patil RJ, Desai AR, et al. Effect of goldnanoparticles on timolol uptake and its releasekinetics from contact lenses: in vitro and in vivoevaluation. Acta Biomater. 2019;1(86):350–62.

91. Shokry M, Hathout RM, Mansour S. Exploringgelatin nanoparticles as novel nanocarriers fortimolol maleate: augmented in vivo efficacy andsafe histological profile. Int J Pharm. 2018;545(1–2):229–39.

92. Jung HJ, Chauhan A. Extended release of timololfrom nanoparticle-loaded fornix insert for glau-coma therapy. J Ocular Pharmacol Ther. 2013;29(2):229–35.

93. Shafaa MW, Sabra NM, Fouad RA. The extendedocular hypotensive effect of positive liposomalcholesterol bound timolol maleate in glaucomatousrabbits. Biopharm Drug Dispos. 2011;32(9):507–17.

94. Aggarwal D, Kaur IP. Improved pharmacodynamicsof timolol maleate from a mucoadhesive niosomalophthalmic drug delivery system. Int J Pharm.2005;290(1):155–9.

95. Gagandeep, Garg T, Malik B, Goyal AK. Develop-ment and characterization of nano-fiber patch forthe treatment of glaucoma. Eur J Pharm Sci.2014;53:10–6.

96. Fulgencio Gde O, Viana FA, Ribeiro RR, et al. Newmucoadhesive chitosan film for ophthalmic drug

Adv Ther (2020) 37:155–199 193

Page 40: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

delivery of timolol maleate: in vivo evaluation.J Ocul Pharmacol Ther. 2012;28(4):350–8.

97. Siafaka PI, Titopoulou A, Koukaras EN, et al. Chi-tosan derivatives as effective nanocarriers for ocularrelease of timolol drug. Int J Pharm. 2015;495(1):249–64.

98. Bertram JP, Saluja SS, McKain J, et al. Sustaineddelivery of timolol maleate from poly(lactic-co-gly-colic acid)/poly(lactic acid) microspheres for over 3months. J Microencapsul. 2009;26:18–26.

99. Ilka R, Mohseni M, Kianirad M, et al. Nanogel-basednatural polymers as smart carriers for the controlleddelivery of timolol maleate through the cornea forglaucoma. Int J Biol Macromol. 2018;1(109):955–62.

100. Tan G, Yu S, Pan H, et al. Bioadhesive chitosan-loaded liposomes: a more efficient and higher per-meable ocular delivery platform for timolol mal-eate. Int J Biol Macromol. 2017;94(Pt A):355–63.

101. Zhang HH, Luo QH, Yang ZJ, et al. Novel oph-thalmic timolol meleate liposomal-hydrogel and itsimproved local glaucomatous therapeutic effectin vivo. Drug Deliv. 2011;18(7):502–10.

102. Zhao R, Li J, Wang J, et al. Development of timolol-loaded galactosylated chitosan nanoparticles andevaluation of their potential for ocular drug deliv-ery. AAPS PharmSciTech. 2017;18(4):997–1008.

103. Yu S, Wang QM, Wang X, et al. Liposome incor-porated ion sensitive in situ gels for opthalmicdelivery of timolol maleate. Int J Pharm.2015;480(1–2):128–36.

104. Lindskog S. Structure and mechanism of carbonicanhydrase. Pharmacol Ther. 1997;74(1):1–20.

105. Hollo G. Carbonic anhydrase inhibitors. In: Shaar-awy TM, Sherwood MB, Hitchings RA, Crowston JG,editors. Glaucoma. 2nd ed. Philadelphia: ElsevierSaunders; 2015. p. 559.

106. Sugrue MF. Pharmacological and ocular hypoten-sive properties of topical carbonic anhydrase inhi-bitors. Prog Retin Eye Res. 2000;19(1):87–112.

107. Jonas JB, Aung T, Bourne RR, Bron AM, Ritch R,Panda-Jonas S. Glaucoma. Lancet. 2017;390(10108):2183–93.

108. Carta F, Scozzafava A, Supuran CT. Sulfonamides: apatent review (2008–2012). Expert Opin Ther Pat.2012;22(7):747–58.

109. Supuran CT, Scozzafava A. Carbonic anhydraseinhibitors and their therapeutic potential. ExpertOpin Ther Pat. 2000;10:575–600.

110. Maren TH. Carbonic anhydrase: chemistry, physi-ology, and inhibition. Physiol Rev. 1967;47(4):595–781.

111. Scozzafava A, Supuran CT. Glaucoma and theapplications of carbonic anhydrase inhibitors. Sub-cell Biochem. 2014;75:349–59.

112. Ilies M, Supuran CT, Scozzafava A, et al. Carbonicanhydrase inhibitors: sulfonamides incorporatingfuran-, thiophene- and pyrrole-carboxamido groupspossess strong topical intraocular pressure loweringproperties as aqueous suspensions. Bioorg MedChem. 2000;8(8):2145–55.

113. Winum JY, Casini A, Mincione F, et al. Carbonicanhydrase inhibitors: N-(p-sulfamoylphenyl)-alpha-D-glycopyranosylamines as topically actingantiglaucoma agents in hypertensive rabbits. BioorgMed Chem Lett. 2004;14(1):225–9.

114. DeSantis L. Preclinical overview of brinzolamide.Surv Ophthalmol. 2000;44(Suppl 2):S119–29.

115. Kaur IP, Singh M, Kanwar M. Formulation andevaluation of ophthalmic preparations of acetazo-lamide. Int J Pharm. 2000;199(2):119–27.

116. Epstein DL, Grant WM. Carbonic anhydrase inhi-bitor side effects. Serum chemical analysis. ArchOphthalmol. 1977;95(8):1378–82.

117. Kaur IP, Kapil M, Smitha R, et al. Development oftopically effective formulations of acetazolamideusing HP-beta-CD-polymer co-complexes. CurrDrug Deliv. 2004;1(1):65–72.

118. Sasaki H, Yamamura K, Mukai T, et al. Enhance-ment of ocular drug penetration. Crit Rev Ther DrugCarrier Syst. 1999;16(1):85–146.

119. Kaur IP, Smitha R, Aggarwal D, et al. Acetazolamide:future perspective in topical glaucoma therapeutics.Int J Pharm. 2002;248(1–2):1–14.

120. Friedman Z, Allen RC, Raph SM. Topical acetazo-lamide and methazolamide delivered by contactlenses. Arch Ophthalmol. 1985;103(7):963–6.

121. Loftsson T, Frithriksdottir H, Stefansson E, et al.Topically effective ocular hypotensive acetazo-lamide and ethoxyzolamide formulations in rabbits.J Pharm Pharmacol. 1994;46(6):503–4.

122. El-Gazaierly O, Hikal AH. Preparation and evalua-tion of acetazolamide liposomes as an oculardelivery system. Int J Pharm. 1997;158(2):121–7.

123. Aggarwal D, Pal D, Mitra AK, Kaur IP. Study of theextent of ocular absorption of acetazolamide from adeveloped niosomal formulation, by microdialysis

194 Adv Ther (2020) 37:155–199

Page 41: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

sampling of aqueous humor. Int J Pharm.2007;338(1–2):21–6.

124. Duarte AR, Roy C, Vega-Gonzalez A, Duarte CM,Subra-Paternault P. Preparation of acetazolamidecomposite microparticles by supercritical anti-sol-vent techniques. Int J Pharm. 2007;332(1–2):132–9.

125. Rathod LV, Kapadia R, Sawant KK. A novelnanoparticle impregnated ocular insert forenhanced bioavailability to posterior segment ofeye: in vitro, in vivo and stability studies. Mater SciEng C Mater Biol Appl. 2017;1(71):529–40.

126. Singh J, Chhabra G, Pathak K. Development ofacetazolamide-loaded, pH-triggered polymericnanoparticulate in situ gel for sustained oculardelivery: in vitro. Ex vivo evaluation and pharma-codynamic study. Drug Dev Ind Pharm. 2014;40(9):1223–32.

127. Bravo-Osuna I, Vicario-de-la-Torre M, Andres-Guerrero V, et al. Novel water-soluble mucoadhe-sive carbosilane dendrimers for ocular administra-tion. Mol Pharm. 2016;13(9):2966–76.

128. Mishra V, Jain NK. Acetazolamide encapsulateddendritic nano-architectures for effective glaucomamanagement in rabbits. Int J Pharm. 2014;461(1–2):380–90.

129. Morsi N, Ibrahim M, Refai H, et al. Nanoemulsion-based electrolyte triggered in situ gel for oculardelivery of acetazolamide. Eur J Pharm Sci.2017;15(104):302–14.

130. Verma P, Gupta RN, Jha AK, et al. Development,in vitro and in vivo characterization of Eudragit RL100 nanoparticles for improved ocular bioavail-ability of acetazolamide. Drug Deliv. 2013;20(7):269–76.

131. Pfeiffer N. Dorzolamide: development and clinicalapplication of a topical carbonic anhydrase inhi-bitor. Surv Ophthalmol. 1997;42(2):137–51.

132. Strahlman E, Tipping R, Vogel R. A double-masked,randomized 1-year study comparing dorzolamide(Trusopt), timolol, and betaxolol. InternationalDorzolamide Study Group. Arch Ophthalmol.1995;113(8):1009–16.

133. Martens-Lobenhoffer J, Banditt P. Clinical pharma-cokinetics of dorzolamide. Clin Pharmacokinet.2002;41(3):197–205.

134. Schwartz GF, Quigley HA. Adherence and persis-tence with glaucoma therapy. Surv Ophthalmol.2008;53(Suppl 1):S57–8.

135. Shinde U, Ahmed MH, Singh K. Development ofdorzolamide loaded 6-O-carboxymethyl chitosan

nanoparticles for open angle glaucoma. J DrugDeliv. 2013;2013:562727.

136. Jansook P, Stefansson E, Thorsteinsdottir M, et al.Cyclodextrin solubilization of carbonic anhydraseinhibitor drugs: formulation of dorzolamide eyedrop microparticle suspension. Eur J Pharm Bio-pharm. 2010;76(2):208–14.

137. Fu J, Sun F, Liu W, et al. Subconjunctival delivery ofdorzolamide-loaded poly(ether-anhydride)microparticles produces sustained lowering ofintraocular pressure in rabbits. Mol Pharm.2016;13(9):2987–95.

138. Katiyar S, Pandit J, Mondal RS, et al. In situ gellingdorzolamide loaded chitosan nanoparticles for thetreatment of glaucoma. Carbohydr Polym.2014;15(102):117–24.

139. Kouchak M, Malekahmadi M, Bavarsad N, et al.Dorzolamide nanoliposome as a long action oph-thalmic delivery system in open angle glaucomaand ocular hypertension patients. Drug Dev IndPharm. 2018;44(8):1239–42.

140. Gudmundsdottir BS, Petursdottir D, AsgrimsdottirGM, et al. c-Cyclodextrin nanoparticle eye dropswith dorzolamide: effect on intraocular pressure inman. J Ocul Pharmacol Ther. 2014;30(1):35–41.

141. Iester M. Brinzolamide ophthalmic suspension: areview of its pharmacology and use in the treatmentof open angle glaucoma and ocular hypertension.Clin Ophthalmol. 2008;2(3):517–23.

142. Kadam RS, Jadhav G, Ogidigben M, et al. Ocularpharmacokinetics of dorzolamide and brinzolamideafter single and multiple topical dosing: implica-tions for effects on ocular blood flow. Drug MetabDispos. 2011;39(9):1529–37.

143. Silver LH, the Brinzolamide Comfort Study Group.Ocular comfort of brinzolamide 1.0% ophthalmicsuspension compared with dorzolamide 2.0% oph-thalmic solution. Results from two multicentercomfort studies. Surv Ophthalmol. 2000;44:141–5.

144. Tsukamoto H, Noma H, Mukai S, et al. The efficacyand ocular discomfort of substituting brinzolamidefor dorzolamide in combination therapy with lata-noprost, timolol, and dorzolamide. J Ocul Pharma-col Ther. 2005;21(5):395–9.

145. Wu W, Li J, Wu L, et al. Ophthalmic delivery ofbrinzolamide by liquid crystalline nanoparticles:in vitro and in vivo evaluation. AAPS Pharm SciTech. 2013;14(3):1063–71.

146. Tuomela A, Liu P, Puranen J, et al. Brinzolamidenanocrystal formulations for ophthalmic delivery:

Adv Ther (2020) 37:155–199 195

Page 42: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

reduction of elevated intraocular pressure in vivo.Int J Pharm. 2014;467(1–2):34–41.

147. Ikuta Y, Aoyagi S, Tanaka Y, et al. Creation of nanoeye-drops and effective drug delivery to the interiorof the eye. Sci Rep. 2017;14(7):44229.

148. Mahboobian MM, Seyfoddin A, Rupenthal ID, et al.Formulation development and evaluation of thetherapeutic efficacy of brinzolamide containingnanoemulsions. Iran J Pharm Res. 2017;16(3):847–57.

149. Wang F, Bao X, Fang A, et al. Nanoliposome-en-capsulated brinzolamide-hydropropyl-b-cyclodex-trin inclusion complex: a potential therapeuticocular drug-delivery system. Front Pharmacol.2018;13(9):91.

150. Salama HA, Ghorab M, Mahmoud AA, et al. PLGAnanoparticles as subconjunctival injection formanagement of glaucoma. AAPS PharmSciTech.2017;18(7):2517–28.

151. Toris CB, Camras CB, Yablonski ME. Acute versuschronic effects of brimonidine on aqueous humordynamics in ocular hypertensive patients. Am JOphthalmol. 1999;128(1):8–14.

152. Ghate D, Edelhauser HF. Ocular drug delivery.Expert Opin Drug Deliv. 2006;3(2):275–87.

153. Konstas AG, Stewart WC, Topouzis F, et al. Bri-monidine 0.2% given two or three times daily ver-sus timolol maleate 0.5% in primary open-angleglaucoma. Am J Ophthalmol. 2001;131(6):729–33.

154. Prabhu P, Nitish KR, Koland M, et al. Preparationand evaluation of nano-vesicles of brimonidinetartrate as an ocular drug delivery system. J YoungPharm. 2010;2(4):356–61.

155. Bhagav P, Upadhyay H, Chandran S. Brimonidinetartrate-Eudragit long-acting nanoparticles: formu-lation, optimization, in vitro and in vivo evalua-tion. AAPS PharmSciTech. 2011;12(4):1087–101.

156. Chiang B, Kim YC, Doty AC, et al. Sustainedreduction of intraocular pressure by supraciliarydelivery of brimonidine-loaded poly(lactic acid)microspheres for the treatment of glaucoma. J Con-trol Release. 2016;28(228):48–57.

157. Ibrahim MM, Abd-Elgawad AH, Soliman OA, et al.Natural bioadhesive biodegradable nanoparticle-based topical ophthalmic formulations for man-agement of glaucoma. Transl Vis Sci Technol.2015;4(3):12.

158. El-Salamouni NS, Farid RM, El-Kamel AH, et al.Nanostructured lipid carriers for intraocular bri-monidine localisation: development, in vitro and

in vivo evaluation. J Microencapsul. 2018;35(1):102–13.

159. Pek YS, Wu H, Mohamed ST, et al. Long-term sub-conjunctival delivery of brimonidine tartrate forglaucoma treatment using a microspheres/carriersystem. Adv Healthc Mater. 2016;5(21):2823–31.

160. Bean GW, Camras CB. Commercially availableprostaglandin analogs for the reduction of intraoc-ular pressure: similarities and differences. SurvOphthalmol. 2008;53(Suppl 1):S69–84.

161. Tanna AP, Lin AB. Medical therapy for glaucoma:what to add after a prostaglandin analogs? CurrOpin Ophthalmol. 2015;26(2):116–20.

162. Hollo G. The side effects of the prostaglandin ana-logues. Expert Opin Drug Saf. 2007;6(1):45–52.

163. Wong TT, Novack GD, Natarajan JV, et al. Nano-medicine for glaucoma: sustained release latano-prost offers a new therapeutic option withsubstantial benefits over eyedrops. Drug DelivTransl Res. 2014;4(4):303–9.

164. Giarmoukakis A, Labiris G, Sideroudi H, et al.Biodegradable nanoparticles for controlled subcon-junctival delivery of latanoprost acid: in vitro andin vivo evaluation. Preliminary results. Exp Eye Res.2013;112:29–36.

165. Natarajan JV, Ang M, Darwitan A, et al. Nanome-dicine for glaucoma: liposomes provide sustainedrelease of latanoprost in the eye. Int J Nanomed.2012;7:123–31.

166. Cheng YH, Tsai TH, Jhan YY, et al. Thermosensitivechitosan-based hydrogel as a topical ocular drugdelivery system of latanoprost for glaucoma treat-ment. Carbohydr Polym. 2016;25(144):390–9.

167. Fahmy HM, Saad EAES, Sabra NM, et al. Treatmentmerits of latanoprost/thymoquinone—encapsu-lated liposome for glaucomatus rabbits. Int J Pharm.2018;548(1):597–608.

168. Rodriguez-Aller M, Guinchard S, Guillarme D, et al.New prostaglandin analog formulation for glau-coma treatment containing cyclodextrins forimproved stability, solubility and ocular tolerance.Eur J Pharm Biopharm. 2015;95(Pt B):203–14.

169. Franca JR, Foureaux G, Fuscaldi LL, et al. Bimato-prost-loaded ocular inserts as sustained release drugdelivery systems for glaucoma treatment: in vitroand in vivo evaluation. PLoS One. 2014;9(4):e95461.

170. Di Trani N, Jain P, Chua CYX, et al. Nanofluidicmicrosystem for sustained intraocular delivery oftherapeutics. Nanomedicine. 2019;16:1–9.

196 Adv Ther (2020) 37:155–199

Page 43: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

171. Lambert WS, Carlson BJ, van der Ende AE, et al.Nanosponge-mediated drug delivery lowersintraocular pressure. Transl Vis Sci Technol.2015;4(1):1.

172. Aref AA, Gedde SJ, Budenz DL. Glaucoma drainageimplant surgery. Dev Ophthalmol. 2017;59:43–52.

173. Ayyala RS, Duarte JL, Sahiner N. Glaucoma drainagedevices: state of the art. Expert Rev Med Devices.2006;3(4):509–21.

174. Chaudhry M, Grover S, Baisakhiya S, et al. Artificialdrainage devices for glaucoma surgery: an overview.Nepal J Ophthalmol. 2012;4(2):295–302.

175. Epstein E. Fibrosing response to aqueous. Its rela-tion to glaucoma. Br J Ophthalmol. 1959;43:641–7.

176. Hong CH, Arosemena A, Zurakowski D, et al.Glaucoma drainage devices: a systematic literaturereview and current controversies. Surv Ophthalmol.2005;50(1):48–60.

177. Ponnusamy T, Yu H, John VT, et al. A novelantiproliferative drug coating for glaucoma drai-nage devices. J Glaucoma. 2014;23(8):526–34.

178. Pan T, Brown JD, Ziaie B. An artificial nano-drainageimplant (ANDI) for glaucoma treatment. Conf ProcIEEE Eng Med Biol Soc. 2006;1:3174–7.

179. Harake RS, Ding Y, Brown JD, et al. Design, fabri-cation, and in vitro testing of an anti-biofoulingglaucoma micro-shunt. Ann Biomed Eng.2015;43(10):2394–405.

180. Popat KC, Desai TA. Poly(ethylene glycol) inter-faces: an approach for enhanced performance ofmicrofluidic systems. Biosens Bioelectron.2004;19(9):1037–44.

181. Shokrollahi H. Structure, synthetic methods, mag-netic properties and biomedical applications offerrofluids. Mater Sci Eng C Mater Biol Appl.2013;33(5):2476–87.

182. Feynman R. ‘‘The Brownian Movement’’. 1964. In:The Feynman Lectures of Physics, Volume I. pp.41–1. http://www.feynmanlectures.caltech.edu/I_41.html. Accessed 7 Dec 2019.

183. Paschalis EI, Chodosh J, Sperling RA, et al. A novelimplantable glaucoma valve using ferrofluid. PLoSOne. 2013;8(6):e67404.

184. Lama PJ, Fechtner RD. Antifibrotics and woundhealing in glaucoma surgery. Surv Ophthalmol.2003;48:314–46.

185. Costa VP, Spaeth GL, Eiferman RA, Orengo-Nania S.Wound healing modulation in glaucoma filtrationsurgery. Ophthalmic Surg. 1993;24:152–70.

186. Wilkins M, Indar A, Wormald R. Intra-operativemitomycin C for glaucoma surgery. CochraneDatabase Syst Rev. 2005;19(4):CD002897.

187. The Fluorouracil Filtering Surgery Study Group.Three-year follow-up of the Fluorouracil FilteringSurgery Study. Am J Ophthalmol. 1993;115:82–92.

188. Hou Z, Wei H, Wang Q, et al. New method to pre-pare mitomycin C loaded PLA-nanoparticles withhigh drug entrapment efficiency. Nanoscale ResLett. 2009;4(7):732–7.

189. Gomes dos Santos AL, Bochot A, Doyle A, et al.Sustained release of nanosized complexes ofpolyethylenimine and anti-TGF-beta 2 oligonu-cleotide improves the outcome of glaucoma sur-gery. J Control Release. 2006;112(3):369–81.

190. Desai MA, Gedde SJ, Feuer WJ, et al. Practice pref-erences for glaucoma surgery: a survey of theAmerican Glaucoma Society in 2008. Ophthal SurgLasers Imaging. 2011;42:202–8.

191. Occhiutto ML, Freitas FR, Lima PP, Maranhao RC,Costa VP. Paclitaxel associated with lipid nanopar-ticles as a new antiscarring agent in experimentalglaucoma surgery. Invest Ophthalmol Vis Sci.2016;57(3):971–8.

192. Duan Y, Guan X, Ge J, et al. Cationic nano-copolymers mediated IKKbeta targeting siRNAinhibit the proliferation of human Tenon’s capsulefibroblasts in vitro. Mol Vis. 2008;14:2616–28.

193. Ye H, Qian Y, Lin M, et al. Cationic nano-copoly-mers mediated IKKb targeting siRNA to modulatewound healing in a monkey model of glaucomafiltration surgery. Mol Vis. 2010;26(16):2502–10.

194. Paula JS, Ribeiro VR, Chahud F, et al. Bevacizumab-loaded polyurethane subconjunctival implants:effects on experimental glaucoma filtration surgery.J Ocul Pharmacol Ther. 2013;29(6):566–73.

195. Li Z, Van Bergen T, Van de Veire S, et al. Inhibitionof vascular endothelial growth factor reduces scarformation after glaucoma filtration surgery. InvestOphthalmol Vis Sci. 2009;50(11):5217–25.

196. Grewal DS, Jain R, Kumar H, Grewal SPS. Evaluationof subconjunctival bevacizumab as an adjunct totrabeculectomy a pilot study. Ophthalmology.2008;115(12):2141–5.

197. Vandewalle E, Abegao Pinto L, Van Bergen T, et al.Intracameral bevacizumab as an adjunct to

Adv Ther (2020) 37:155–199 197

Page 44: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

trabeculectomy: a 1-year prospective, randomisedstudy. Br J Ophthalmol. 2014;98(1):73–8.

198. Han Q, Wang Y, Li X, et al. Effects of bevacizumabloaded PEG-PCL-PEG hydrogel intracameral appli-cation on intraocular pressure after glaucoma fil-tration surgery. J Mater Sci Mater Med. 2015;26(8):225.

199. Gupta N, Yucel YH. Glaucoma as a neurodegenera-tive disease. Curr Opin Ophthalmol. 2007;18(2):110–4.

200. Brubaker RF. Delayed functional loss in glaucoma.LII Edward Jackson memorial lecture. Am J Oph-thalmol. 1996;121:473–83.

201. Allen SJ, Watson JJ, Shoemark DK, et al. GDNF, NGFand BDNF as therapeutic options for neurodegen-eration. Pharmacol Ther. 2013;138(2):155–75.

202. Frasson M, Picaud S, Leveillard T, et al. Glial cellline-derived neurotrophic factor induces histologicand functional protection of rod photoreceptors inthe rd/rd mouse. Invest Ophthalmol Vis Sci.1999;40(11):2724–34.

203. Checa-Casalengua P, Jiang C, Bravo-Osuna I, et al.Retinal ganglion cells survival in a glaucoma modelby GDNF/Vit E PLGA microspheres preparedaccording to a novel microencapsulation procedure.J Control Release. 2011;156(1):92–100.

204. Ward MS, Khoobehi A, Lavik EB, et al. Neuropro-tection of retinal ganglion cells in DBA/2 J micewith GDNF-loaded biodegradable microspheres.J Pharm Sci. 2007;96(3):558–68.

205. Jiang C, Moore MJ, Zhang X, et al. Intravitrealinjections of GDNF-loaded biodegradable micro-spheres are neuroprotective in a rat model of glau-coma. Mol Vis. 2007;24(13):1783–92.

206. Garcıa-Caballero C, Prieto-Calvo E, Checa-Casalen-gua P, et al. Six month delivery of GDNF fromPLGA/vitamin E biodegradable microspheres afterintravitreal injection in rabbits. Eur J Pharm Sci.2017;30(103):19–26.

207. Clatterbuck RE. Cliliary neurotrophic factor pre-vents retrograde neuronal death in the adult centralnervous system. Proc Natl Acad Sci USA. 1993;90:2222–6.

208. Emerich DF, Winn SR, Hantraye PM, et al. Protec-tive effect of encapsulated cells producing neu-rotrophic factor CNTF in a monkey model ofHuntington’s disease. Nature. 1997;386(6623):395–9.

209. Hagg T, Varon S. Ciliary neurotrophic factor pre-vents degeneration of adult rat substantia nigra

dopaminergic neurons in vivo. Proc Natl Acad SciUSA. 1993;90(13):6315–9.

210. Sendtner M, Schmalbruch H, Stockli KA, et al. Cil-iary neurotrophic factor prevents degeneration ofmotor neurons in mouse mutant progressive motorneuronopathy. Nature. 1992;358(6386):502–4.

211. Nkansah MK, Tzeng SY, Holdt AM, et al. Poly(lactic-co-glycolic acid) nanospheres and microspheres forshort- and long-term delivery of bioactive ciliaryneurotrophic factor. Biotechnol Bioeng.2008;100(5):1010–9.

212. Pease ME, Zack DJ, Berlinicke C, et al. Effect ofCNTF on retinal ganglion cell survival in experi-mental glaucoma. Invest Ophthalmol Vis Sci.2009;50(5):2194–200.

213. Kitagawa K, Matsumoto M, Tagaya M, et al.Hyperthermia-induced neuronal protection againstischemic injury in gerbils. J Cereb Blood FlowMetab. 1991;11(3):449e52.

214. Caprioli J, Kitano S, Morgan JE. Hyperthermia andhypoxia increase tolerance of retinal ganglion cellsto anoxia and excitotoxicity. Invest Ophthalmol VisSci. 1996;37(12):2376e81.

215. Barbe MF, Tytell M, Gower DJ, et al. Hyperthermiaprotects against light damage in the rat retina. Sci-ence. 1988;241(4874):1817e20.

216. Yin Y, Henzl MT, Lorber B, et al. Oncomodulin is amacrophage-derived signal for axon regeneration inretinal ganglion cells. Nat Neurosci. 2006;9:843–52.

217. Jeun M, Jeoung JW, Moon S, et al. Engineeredsuperparamagnetic Mn0.5Zn0.5Fe2O4 nanoparti-cles as a heat shock protein induction agent forocular neuroprotection in glaucoma. Biomaterials.2011;32(2):387–94.

218. Eming SA, Wynn TA, Martin P. Inflammation andmetabolism in tissue repair and regeneration. Sci-ence. 2017;356(6342):1026–30.

219. Pasinetti GM. Cyclooxygenase and inflammation inAlzheimer’s disease: experimental approaches andclinical interventions. J Neurosci Res. 1998;54:1–6.

220. Gonzalez-Scarano F, Baltuch G. Microglia as medi-ators of inflammatory and degenerative diseases.Annu Rev Neurosci. 1999;22:219–40.

221. Julien JP. Amyotrophic lateral sclerosis: unfoldingthe toxicity of the misfolded. Cell. 2001;104:581–91.

222. Mac Nair CE, Nickells RW. Neuroinflammation inglaucoma and optic nerve damage. Prog Mol BiolTransl Sci. 2015;134:343–63.

198 Adv Ther (2020) 37:155–199

Page 45: Nanotechnology for Medical and Surgical Glaucoma Therapy—A … · 2020-01-18 · tors, anti-inflammatory drugs, and gene thera-pies. Nanotechnology-based treatments may surpass

223. Kawano T, Anrather J, Zhou P, et al. ProstaglandinE2 EP1 receptors: downstream effectors of COX-2neurotoxicity. Nat Med. 2006;12:225–9.

224. Kolko M, DeCoster MA, de Turco EB, et al. Synergyby secretory phospholipase A2 and glutamate oninducing cell death and sustained arachidonic acidmetabolic changes in primary cortical neuronalcultures. J Biol Chem. 1996;271:32722–8.

225. Li G, Luna C, Liton PB, et al. Sustained stressresponse after oxidative stress in trabecular mesh-work cells. Mol Vis. 2007;13:2282–8.

226. Tezel G, Wax MB. Increased production of tumornecrosis factor-alpha by glial cells exposed to sim-ulated ischemia or elevated hydrostatic pressureinduces apoptosis in cocultured retinal ganglioncells. J Neurosci. 2000;20:8693–700.

227. Ergorul C, Ray A, Huang W, et al. Hypoxia induciblefactor1alpha (HIF-1alpha) and some HIF-1 targetgenes are elevated in experimental glaucoma. J MolNeurosci. 2010;42:183–91.

228. Ammon HP, Wahl MA. Pharmacology of Curcumalonga. Planta Med. 1991;57:1–7.

229. Dong S, Zeng Q, Mitchell ES, et al. Curcuminenhances neurogenesis and cognition in aged rats:implications for transcriptional interactions relatedto growth and synaptic plasticity. PLoS One.2012;7(2):e31211.

230. Kim DS, Kim JY, Han Y. Curcuminoids in neu-rodegenerative diseases. Recent Pat CNS Drug Dis-cov. 2012;7(3):184–204.

231. Belviranlı M, Okudan N, Atalık KE, et al. Curcuminimproves spatial memory and decreases oxidativedamage in aged female rats. Biogerontology.2013;14(2):187–96.

232. Wang L, Li C, Guo H, et al. Curcumin inhibitsneuronal and vascular degeneration in retina afterischemia and reperfusion injury. PLoS One.2011;6(8):e23194.

233. Yue YK, Mo B, Zhao J, et al. Neuroprotective effectof curcumin against oxidative damage in BV-2microglia and high intraocular pressure animalmodel. J Ocul Pharmacol Ther. 2014;30(8):657–64.

234. Kaminaga Y, Nagatsu A, Akiyama T, et al. Produc-tion of unnatural glucosides of curcumin withdrastically enhanced water solubility by cell sus-pension cultures of Catharanthus roseus. FEBS Lett.2003;555(2):311–6.

235. Gupta S, Patchva S, Aggarwal BB. Therapeutic rolesof curcumin: lessons learned from clinical trials.AAPS J. 2013;15(1):195–218.

236. Davis BM, Pahlitzsch M, Guo L, et al. Topical cur-cumin nanocarriers are neuroprotective in eye dis-ease. Sci Rep. 2018;8(1):11066.

237. National Cancer Institute Drug Dictionary. https://www.cancer.gov/publications/dictionaries/cancer-drug/def/ketorolac. Accessed 7 Dec 2019.

238. Ju WK, Neufeld AH. Cellular localization ofcyclooxygenase-1 and cyclooxygenase-2 in thenormal mouse, rat, and human retina. J CompNeurol. 2002;452:392–9.

239. Wang AG, Lee CM, Wang YC, et al. Up-regulation ofcytochrome oxidase in the retina following opticnerve injury. Exp Eye Res. 2002;74:651–9.

240. Ju WK, Kim KY, Neufeld AH. Increased activity ofcyclooxygenase-2 signals early neurodegenerativeevents in the rat retina following transient ische-mia. Exp Eye Res. 2003;77:137–45.

241. Nadal-Nicolas FM, Rodriguez-Villagra E, Bravo-Osuna I, et al. Ketorolac administration attenuatesretinal ganglion cell death after axonal injury.Invest Ophthalmol Vis Sci. 2016;57(3):1183–92.

242. Barcia E, Herrero-Vanrell R, Dıez A, et al. Down-regulation of endotoxin-induced uveitis by intrav-itreal injection of polylactic-glycolic acid (PLGA)microspheres loaded with dexamethasone. Exp EyeRes. 2009;89(2):238–45.

243. Prow TW. Toxicity of nanomaterials to the eye.Wiley Interdiscip Rev Nanomed Nanobiotechnol.2010;2(4):317–33.

244. Voigt N, Henrich-Noack P, Kockentiedt S, et al.Toxicity of polymeric nanoparticles in vivo andin vitro. J Nanopart Res. 2014;16(6):2379.

245. De Matteis V, Rinaldi R. Toxicity assessment in thenanoparticle era. Adv Exp Med Biol. 2018;1048:1–19.

246. El-Ansary A, Al-Daihan S, Bacha AB, et al. Toxicityof novel nanosized formulations used in medicine.Methods Mol Biol. 2013;1028:47–74.

247. De Jong WH, Van Der Ven LT, Sleijffers A, et al.Systemic and immunotoxicity of silver nanoparti-cles in an intravenous 28 days repeated dose toxic-ity study in rats. Biomaterials. 2013;34(33):8333–43.

Adv Ther (2020) 37:155–199 199