26
Full Terms & Conditions of access and use can be found at http://www.tandfonline.com/action/journalInformation?journalCode=inan20 Download by: [Texas A&M University Libraries] Date: 31 March 2016, At: 06:58 Nanotoxicology ISSN: 1743-5390 (Print) 1743-5404 (Online) Journal homepage: http://www.tandfonline.com/loi/inan20 Safety and toxicity of nanomaterials for ocular drug delivery applications Neelesh K. Mehra, Defu Cai, Lih Kuo, Travis Hein & Srinath Palakurthi To cite this article: Neelesh K. Mehra, Defu Cai, Lih Kuo, Travis Hein & Srinath Palakurthi (2016): Safety and toxicity of nanomaterials for ocular drug delivery applications, Nanotoxicology To link to this article: http://dx.doi.org/10.3109/17435390.2016.1153165 Published online: 30 Mar 2016. Submit your article to this journal View related articles View Crossmark data

48 Safety and Toxicity of Nanomaterials for ocular drug delivery applications

Embed Size (px)

Citation preview

Page 1: 48 Safety and Toxicity of Nanomaterials for ocular drug delivery applications

Full Terms & Conditions of access and use can be found athttp://www.tandfonline.com/action/journalInformation?journalCode=inan20

Download by: [Texas A&M University Libraries] Date: 31 March 2016, At: 06:58

Nanotoxicology

ISSN: 1743-5390 (Print) 1743-5404 (Online) Journal homepage: http://www.tandfonline.com/loi/inan20

Safety and toxicity of nanomaterials for oculardrug delivery applications

Neelesh K. Mehra, Defu Cai, Lih Kuo, Travis Hein & Srinath Palakurthi

To cite this article: Neelesh K. Mehra, Defu Cai, Lih Kuo, Travis Hein & Srinath Palakurthi(2016): Safety and toxicity of nanomaterials for ocular drug delivery applications,Nanotoxicology

To link to this article: http://dx.doi.org/10.3109/17435390.2016.1153165

Published online: 30 Mar 2016.

Submit your article to this journal

View related articles

View Crossmark data

Page 2: 48 Safety and Toxicity of Nanomaterials for ocular drug delivery applications

http://informahealthcare.com/nanISSN: 1743-5390 (print), 1743-5404 (electronic)

Nanotoxicology, Early Online: 1–25! 2016 Taylor & Francis. DOI: 10.3109/17435390.2016.1153165

REVIEW ARTICLE

Safety and toxicity of nanomaterials for ocular drug deliveryapplications

Neelesh K. Mehra1, Defu Cai1, Lih Kuo2,,3, Travis Hein3and Srinath Palakurthi1,

1Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M Health Science Center, Kingsville, TX, USA,2Department of Medical Physiology, College of Medicine, Texas a&M Health Science Center, Temple, TX, USA, 3Department of Surgery and Scott &

White Eye Institute, College of Medicine, Texas a&M Health Science Center, Temple, TX, USA

Abstract

Multifunctional nanomaterials are rapidly emerging for ophthalmic delivery of therapeutics tofacilitate safe and effective targeting with improved patient compliance. Because of theirextremely high area to volume ratio, nanomaterials often have physicochemical properties thatare different from those of their larger counterparts. There exists a complex relationshipbetween the physicochemical properties (composition, size, shape, charge, roughness, andporosity) of the nanomaterials and their interaction with the biological system. The eye is a verysensitive accessible organ and is subjected to intended and unintended exposure tonanomaterials. Currently, various ophthalmic formulations are available in the market, whilesome are underway in preclinical and clinical phases. However, the data on safety, efficacy, andtoxicology of these advanced nanomaterials for ocular drug delivery are sparse. Focus of thepresent review is to provide a comprehensive report on the safety, biocompatibility andtoxicities of nanomaterials in the eye.

Keywords

Carbon nanomaterials, eye, nanomaterials,nanoparticles, safety and toxicity

History

Received 23 December 2015Revised 5 February 2016Accepted 8 February 2016Published online 25 March 2016

Introduction

Nanotechnology has been a rapidly expanding field and iscontinuously being explored in the field of pharmaceutical andbiotechnology sciences to achieve the desired product or thera-peutic outcomes. Nanotoxicology is a branch of nanoscience thatdeals with the safety and toxicity of nanomaterials and evaluatingtheir interactions with biological systems. Due to the nano-size andhigh surface free area, these nanomaterials interact with the cellsand subcellular organelles rapidly and effectively, which may leadto toxic effects. The nanotechnology industry is a rapidly emergingindustry generating numerous engineered nanomaterials (ENM)that are incorporated into medical, cosmetic and consumerproducts. Thus far, more than 3000 nanomaterials have beenused in pharmaceutical companies, but toxicity data are sparse(Jain et al., 2007, 2014; Mody et al., 2014; Srivastava et al., 2015).The widespread production and use of ENM may lead tounintended human exposure, while their impact on biologicalsystems is not clearly understood. A comprehensive understandingof the nano-bio interactions would not only facilitate developingsafe and efficacious nanoproducts for therapeutic applications, butit also would aid in developing guidelines for the propermanufacturing, production, synthesis, handling, use and disposalof nanomaterials. In addition to the users of nanoproducts such ascosmetics and other consumer products, personnel involved inmanufacturing, handling, and working on nanomaterials are at riskof exposure to the nanomaterials by contact (skin and eye),

inhalation (nose and lung), and ingestion (gastrointestinal tract). Toaddress these concerns, the National Institute of EnvironmentalHealth Sciences (NIEHS) initiated a NanotechnologyEnvironmental Health and Safety (NanoEHS) program in 2004 togain fundamental understanding of the molecular and pathologicalpathways involved in mediating biological responses to ENM.

Currently, various ophthalmic formulations are available in themarket, while some are underway in preclinical and clinical phases.Herein, we attempted to present a comprehensive review on thesafety, biocompatibility and toxicity of nanomaterials in the eye.The presence of various anatomical and physiological barriers is amajor constraint on ophthalmic drug delivery, which prevents thedrug from reaching the target site. Topically applied drugs canenter into the anterior chamber by crossing the cornea or throughthe conjunctiva and sclera or via the systemic circulation, but theymust cross the blood-aqueous barrier. As a recent report suggests,topically applied ophthalmic eye drops typically deliver drugs intothe anterior chamber and negligible amounts (55%) enter into theback of the eye (Fitzpartrick et al., 2012).

In ophthalmic delivery, conventional ophthalmic formulationsare easily washed away by nasolacrimal drainage, while thenanoparticles are cleared slowly and release the drug for a longerduration upon interaction with the cornea. These nanomaterialsare expected to not only enhance the ocular bioavailability butalso offer sustained therapeutic drug concentration for a longerduration and thereby reduce frequency of administration withimproved patient compliance.

Basic structure of the eye

The eye is a complex, sensitive organ with numerous absorptionbarriers. It is composed of three main layers: the sclera and cornea

*Correspondence: Srinath Palakurthi [email protected] of Pharmaceutical Sciences, Irma Lerma Rangel College ofPharmacy Texas A&M Health Science Center, Kingsville, TX 78363,USA

Dow

nloa

ded

by [

Tex

as A

&M

Uni

vers

ity L

ibra

ries

] at

06:

58 3

1 M

arch

201

6

Page 3: 48 Safety and Toxicity of Nanomaterials for ocular drug delivery applications

(outer layer); the choroid, ciliary body, and iris (middle layer oruveal coat); and the retina (inner layer). The outer part of the eyeis mainly covered by the mobile tarsal part of eyelids. The eyeballis covered by the thin skin of lids, which permits opening andclosing of the palpebral fissure. For spontaneous blinking, eyesare under voluntary and involuntary control. Generally, thera-peutics penetrate the corneal epithelium by either paracellular ortranscellular transport mechanisms. The transcellular mechanismpredominates for lipophilic molecules, while hydrophilic mol-ecules and ions are transported by a paracellular route. The poresize of the corneal epithelium is in the range of 1–5 nm (Prausnitz& Noonan, 1998). Only about 5% of the topically administereddrug is absorbed through the corneal epithelium. The cornea is aclear, transparent, avascular tissue and it is composed ofepithelium, stroma, Descemet’s membrane, Bowman’s layer, andendothelium. The corneal epithelium consists of 5–6 layers ofcolumnar cells squeezed forward by the new cells. The cornealepithelial cell surface is susceptible to fungal, bacterial, and viralinfections and inflammation. Replacement of the epithelial cellsoccurs by mitotic division of the basal layer every 4–8 d. Thelachrymal fluid and the aqueous humor supply nutrients andoxygen to the cornea. Fungal infection of the cornea is mainlycaused by the fungal species of Aspergillus, Fusarium,Curvularia, and Candida, which may lead to cataract, loss ofvision, and blindness. The conjunctiva is a thin transparentmembrane, which lines the inner surface of eyelids. Theconjunctiva is composed of an epithelium, a highly vascularizedsubstantia propria, and a submucosa or episclera (Kim et al.,2015; Sosnik et al., 2014; Yan et al., 2012). The sclera is a fibrouslayer composed of collagen and elastic fibers. It offers resistanceto internal and external forces and provides mechanical stabilityto the eye. The scleral surface area in humans is approximately16–17 cm2 and largely influences the pharmacokinetics of drugdiffusion (Myles et al., 2005). The sclera is 10 times morepermeable than the cornea and the conjunctiva is 15–25 timesmore permeable than the sclera (Hamalainen et al., 1997). Poorcorneal penetration, short precorneal residence time, and presence

of various anatomical and physiological barriers are the mainproblems in ocular therapeutic delivery. Additionally, the blood–retinal barrier, the blood–aqueous barrier, and the extraocularepithelium represent obstacles in ocular delivery to the choroid,retina, and vitreous. Only a fraction of the topically administereddrug reaches the retina or the vitreous body following systemicadministration (Kim et al., 2015; Lezzi et al., 2012). Directintravitreal injection is performed to achieve high drug concen-trations in the vitreous and retina. However, frequent injectionsare required to sustain high drug levels because the half-life ofdrugs in the vitreous cavity is relatively short. Nanoparticlesystems that can provide sustained drug release have beenexplored to reduce the frequency of intravitreal administration.Figure 1 shows various distribution pathways for administration ofocular drugs in the eye.

Parameters to assess toxicity to the eye

There are various observable and measurable parameters fortesting the toxicity of ophthalmic formulations. A comprehensivereview on the techniques currently used for ocular toxicity testingis published by Wilson et al. (2015). While a myriad ofnanomaterials are used in FDA-regulated ocular products,immense data gaps, and conflicting reports on their toxicologycurrently prevent generalizing how nanomaterial physicochemicalproperties relate to biological activity and toxic potential (Jainet al., 2014; Srivastava et al., 2015). Moreover, little is knownabout the interaction of nanomaterials with the cells andpenetration/toxicity.

The international standard assay for acute ocular toxicity (eyeirritation test) is the rabbit in vivo Draize eye test, developed bythe FDA (Draize et al., 1994). The Draize eye irritation test is anendorsed method/assay to validate the safety and toxicity ofmaterials being used in ophthalmic preparations. Followingtopical exposure of nanomaterials (100 mL or 100 mg) to theconjunctival sac for 72 h, the rabbits are observed up to 21 d atregular intervals for signs of irritation (observable toxicity)

Figure 1. Various distribution pathways for administration of ocular drugs in the eye.

2 N.K. Mehra et al. Nanotoxicology, Early Online: 1–25

Dow

nloa

ded

by [

Tex

as A

&M

Uni

vers

ity L

ibra

ries

] at

06:

58 3

1 M

arch

201

6

Page 4: 48 Safety and Toxicity of Nanomaterials for ocular drug delivery applications

including redness, swelling, cloudiness, edema, hemorrhage,discharge, and blindness (Huhtala et al., 2008). New Zealandwhite (NZW) rabbits are most widely used model because theyhave large eyes with known anatomy and physiology, easy tohandle, available and inexpensive. Eye irritation is summarized asa ‘‘maximum average score’’ (MAS), which is an average valuescored based on the corneal injury for individual animals at thetime of scoring. Despite its gold standard status, the Draize testwas never formally validated. Moreover, since the anatomy,physiology, biochemistry of the rabbit eye differs from the humaneye (thinner cornea, lower tear production, blinking frequency andocular surface sensitivity in rabbits), differences in sensitivity toirritants can occur. Rabbits have larger conjunctival sacs and anictitating membrane, which may aid in removal of the testsubstance from the ocular surface.

The low-volume eye-irritation test (LVET) is refinement of theDraize test and was developed by Griffith et al. (1980). In theLVET, 0.01 ml/0.01 g lower volume of test substances is appliedinto the right eye of animals. The LVET test is less stressful to theanimal than the Draize test (Roggeband et al., 2000). It ischaracterized by changes in the pathological condition of cornea,conjunctiva, and iris/ciliary body. Recently, the InteragencyCoordinating Committee on the Validation of AlternativeMethods (ICCVAM) did not consider nor recommend forprospective ocular safety testing test in replacement of Draizetesting. In our opinion, we can consider the LVET toxicity testingmethod as an alternative eye toxicity test.

Ocular organotypic models or the enucleated eye test (EET)uses isolated rabbit eyes from animals used for other research orcommercial purposes (Barile, 2010). The nanomaterials may beapplied at relevant concentrations and corneal opacity andswelling are observed. Corneal opacity is an indication of proteindenaturation, swelling, vacuolation, and damage of epitheliumand stroma. Bovine and porcine cornea and chicken enucleatedeyes are widely accepted as a reliable and an accurate tissue forassessing eye irritation. EET is considered a scientifically soundtest to evaluate and identify substances that are non-irritants aswell as those materials that cause irreversible eye damage.

The test materials (including nanomaterials and formulation)that penetrate the ocular tissues or that are administeredintraocularly may show some impact on the retinal morphologyand circulation, which may experimentally be assessed usingoptical coherence tomography (OCT), ultrasound Doppler meas-urements and retinal fluorescein angiography, respectively. TheOCT is used to assess architectural changes in the nerve fiber andganglion cell layers, inner retina, photoreceptor cell layer, retinalthickness, choroidal thickness, and the vitreoretinal interface(including the development of epiretinal membranes and vitreo-macular traction) (Kjellstrom et al., 2014; Majdi et al., 2015; vanVelthoven et al., 2007). The effect of nanomaterials on ophthal-mic blood flow, which is controlled by the downstream arteriolesin the microcirculation, may be assessed using color Dopplerultrasound (Anayol et al., 2014; Dong et al., 2000; Plange et al.,2003). To detect whether exposure of test material in the eyecould cause vascular changes, such as focal constriction/dilationof the local vasculature, retinal fluorescein angiography can beperformed (Dong et al., 2000; Plange et al., 2003). To correlatethe structural and vascular changes observed in the above threetests to the neural function of the retina, toxicity to the retinalneurons can by assessed by electroretinography (ERG) (Huanget al., 2015; Kjellstrom et al., 2014). The ERG measures theelectrical and non-electrical activities generated by neural andnon-neuronal cells in the retina in response to a light stimulusfollowing exposure to the test material (Huang et al., 2015;Kjellstrom et al., 2014). Although these experimental tests are notyet validated by the regulatory agencies, they offer detailed

information on any changes in vascular and neural structure andfunctions following exposure to the test material, and guide in thedevelopment of safe ocular formulations and next generationnanomaterials.

Nanomaterials-mediated eye therapy

Several site-specific ocular therapeutic delivery systems havebeen evaluated so far, including carbon nanohorns (Miyawakiet al., 2008), fullerenes (Aoshima et al., 2009), graphenes(Angelopoulou et al., 2015; Mao et al., 2013; Yan et al., 2012),quantum dots (Kuo et al., 2011), carbon nanotubes (CNTs) (Luet al., 2013), contact lenses (Kapoor et al., 2009; Wu et al., 2015),hydrogels (Almeida et al., 2014; Kirchhof et al., 2015;Sheikholeshlami et al., 2015), corneal SkGel (SkGel implantfrom corneal laboratory) (Lopes et al., 2006), chitosan (Mannaet al., 2015), polymeric micelles (Tommaso et al., 2011),dendrimers (Holden et al., 2012; Lezzi et al., 2012; Souzaet al., 2015; Spataro et al., 2010), implants (Bozukova et al.,2010), triblock copolymers (Qiao et al., 2013), liposomes (Caiet al., 2014; Chetoni et al., 2015; Taha et al., 2014), niosomes(Marianecci et al., 2014), nanoparticles (Abrego et al., 2015;Andrei et al., 2015; Leonardi et al., 2014; Soderstjerna et al.,2014), micro- and nano-emulsions (Gan et al., 2009), nanosus-pensions (Pignatello et al., 2002), nanoaggregates (Dilbaghi et al.,2013), nanocrystals (Tuomela et al., 2014), micro-needles (Kimet al., 2015), cyclodextrins (Garcia-Fernandez et al., 2013; HalimMohamed & Mahmoud, 2011), and electric-pulse targeted(Oshima et al., 1999). The nanomaterials that have been exploredfor ocular drug delivery are illustrated in Figure 2.

Ophthalmic delivery to anterior or posterior segments of theeye has been investigated using a variety of drugs, includinghomoharringtonine (HHT) (Peng et al., 1999), mitomycin C(Kozobolis et al., 2002), autumn narcissus alkali (Fuller et al.,2002), human amniotic membrane (Rai et al., 2005), tissueplasminogen activator (Tripathi & Tfipathi, 2005), paclitaxel(Koz et al., 2007), corticosteroids (Spitzer et al., 2008), 5-fluorouracil (Wang et al., 2011), cyclosporine A (Aksungur et al.,2011; Gan et al., 2009), interferons (Wang et al., 2011), aspirin(Das et al., 2012), natamycin (Bhatta et al., 2012), timololmaleate, brimonidine (Yang et al., 2012), terbinafine hydrochlor-ide (Tayel et al., 2013), tropicamide (Dilbaghi et al., 2013),pilocarpine nitrate (Li et al., 2013), indomethacin (Nagai et al.,2014), acetazolamide (Mishra & Jain, 2014), brinzolamide(Tuomela et al., 2014), dorzolamide (Warsi et al., 2014),distamycin (Chetoni et al., 2015), and dexamethasone (Courseyet al., 2015). For the reader’s interest, Kompella et al. (2013)published a review of various nanomedicines used for delivery oftherapeutics into the back of the eye entitled ‘‘Nanomedicines forback of the eye drug delivery, gene delivery, and imaging’’.

Nanotoxicology: safety and toxicity of nanomaterials

Although a significant amount of data is available on theformulation, characterization, ocular drug delivery and targetingof nanomaterials, information on the safety and toxicity of theabove mentioned delivery systems and multifunctional nanoma-terials is sparse. Safety and toxicity are important issues that arealways of primary concern prior to approval of ophthalmicproducts for clinical trials. Although in-depth toxicology studiesof nanomaterials to the eye were summarized by Prow (2010),herein we attempt to present a detailed review on the safety andtoxicological perspectives of new nanomaterials and deliverysystems that are under various phases of preclinical and clinicalphases. Table 1 summarizes the safety and toxicity of nanomater-ials in the eye.

DOI: 10.3109/17435390.2016.1153165 Please check 3

Dow

nloa

ded

by [

Tex

as A

&M

Uni

vers

ity L

ibra

ries

] at

06:

58 3

1 M

arch

201

6

Page 5: 48 Safety and Toxicity of Nanomaterials for ocular drug delivery applications

Nanoparticles

Nanoparticles are colloidal particles having at-least one dimen-sion less than 100 nm (Mehra et al., 2015b; Rajala et al., 2014).Decrease in the particle size causes less irritation allows for ahigher surface to volume ratio, increases precorneal residencetime, and minimizes drug dose and chances of toxicity more thanmicroparticles (Das et al., 2012). Corneal toxicity and thepropensity of the drug to penetrate and permeate across thecornea increased with using smaller size nanoparticles (Conley &Naash, 2010). The tranilast nanoparticles dispersion, which wasprepared using the bead mill method, caused less damage tohuman corneal epithelium (HCE) cells. The nanoparticles alsoenhanced the corneal penetration as compared with tranilast eyedrops (RIZABEN� eye drops) (Nagai & Ito, 2014; Nagai et al.,2014).

Pilocarpine nitrate (PN) has been used in the treatment of acuteangle-closure and chronic open-angle glaucoma. However, itshows poor corneal penetration and low pre-ocular retention, andtherefore poor bioavailability. Due to the poor ocular bioavail-ability, it requires frequent administration in high concentrations,leading to side effects (myopia and miosis). Liquid crystallinenanoparticles (LCNPs) offer some unique properties like highinternal interfacial area useful for encapsulating both hydrophobicand hydrophilic as well as amphiphilic drugs in separate domains.LCNPs are a better nano-candidate for ocular delivery, whereinPNs are well entrapped. Additionally, it is non-toxic, bioadhesivein nature and depicts sustained-release behavior. Glycerylmonoolein (GMO) is a non-toxic, biocompatible and biodegrad-able amphiphilic lipid that swells in the water and thenspontaneously forms a well-ordered liquid crystalline phase (Liet al., 2013; Nair et al., 2012).

Li and et al. (2013) investigated the potential of GMO-basedLCNPs for ocular delivery of PNs (PN-LCNPs) (particle size202.28 ± 19.32 nm and percent encapsulation efficiency 61.03%)in adult New Zealand white rabbits. From the ex vivo corneal

permeation study, PN-LCNPs treatment led to a two-fold higherapparent permeability coefficient than did commercial eye dropsand decreased IOP in vivo. PN-LCNPs did not cause irritation ofocular tissues in the rabbit eye based on the conjunctival scores.

Dorzolamide (Trusopt�) is a potent and frequently prescribed

drug in the treatment of glaucoma. But dorzolamide has poorocular retention and its degree of ionization and hydrophilicnature leads to poor ocular bioavailability. It is used three to fourtimes a day. Dorzolamide-loaded PLGA nanoparticles using twoemulsifiers poly (vinyl alcohol) (PVA) and tocopheryl polyethyl-ene glycol succinate (TPGS) for glaucoma therapy weredeveloped and reported in freshly excised goat eyes and adultNew Zealand albino rabbits by Warsi et al. (2014). Ex vivo trans-corneal permeation study and pharmacoscintigraphic studiesrevealed that the PVA and TPGS nanoparticles are non-irritatingand significantly reduce the intraocular pressure by 22.81 and29.12%, respectively, via topical instillation.

Azone is one of the most widely used permeation enhancers toimprove the permeability of both hydrophilic and lipophilic drugs.Recently, the biopharmaceutical profile of pranoprofen-loadedPLGA nanoparticles (PF-F1NPs and PF-F2NPs) were developedand further dispersed into hydrogels for ocular administration.The optimized PF-NP suspensions were dispersed in freshlyprepared carbomer hydrogels (HG_PF-F1NPs and HG_PF-F2NPs) or in hydrogels containing 1% azone (HG_PF-F1NPs-Azone and HG_PF-F2NPs-Azone) to increase the ocular bio-pharmaceutical profile of pranoprofen. It is a non-steroidal anti-inflammatory drug used as a safe and effective alternative anti-inflammatory treatment following strabismus and cataract sur-gery. No signs of ocular irritancy have been observed with thehydrogels based on the in vitro and in vivo ocular tolerance andin vivo anti-inflammatory studies on New Zealand rabbits(male, weighing 2.5–3.0 kg). The ocular application of hydrogelscontaining azone was more effective than azone-free formulationsin the treatment of edema on the ocular surface (Abrego et al.,2015).

Figure 2. Nanomaterials used in ocular delivery of therapeutic agents.

4 N.K. Mehra et al. Nanotoxicology, Early Online: 1–25

Dow

nloa

ded

by [

Tex

as A

&M

Uni

vers

ity L

ibra

ries

] at

06:

58 3

1 M

arch

201

6

Page 6: 48 Safety and Toxicity of Nanomaterials for ocular drug delivery applications

Tab

le1

.S

um

mar

ize

the

safe

tyan

dto

xic

ity

of

nan

om

ater

ials

iney

e.

Nan

om

ater

ials

Ex

vivo

stu

dy

Invi

vost

ud

yT

her

apeu

tic

del

iver

edR

ou

tes

of

adm

inis

trat

ion

Tox

icit

yT

arget

site

Ou

tco

me

Ref

Hy

bri

dPA

MA

Md

end

rim

ers/

PL

GA

nan

op

arti

cles

–D

utc

h-b

elte

dm

ale

rab

bit

sB

rim

on

idin

ean

dti

mo

lol

mal

eate

To

pic

alad

min

istr

atio

nD

idn

ot

show

ocu

lar

infl

am-

mat

ion

or

dis

-co

mfo

rto

rd

idn

ot

revea

lan

ym

orp

ho

log

ical

or

stru

ctu

ral

chan

ges

Aqu

eou

sh

um

or

and

corn

eaH

DN

P,

did

no

tin

du

cean

yg

ross

tox

icit

yin

the

ocu

lar

tiss

ues

Yan

get

al.

(20

12

)

Lip

idn

ano

par

ticl

es–

Mal

eN

ewZ

eala

nd

alb

ino

rab

bit

s

–T

op

ical

No

corn

eal

op

a-ci

tyan

dn

on

-ir

rita

nt

effe

ct

Co

rnea

SL

Ns

syn

the-

size

du

sin

gC

rem

op

ho

reA

25

and

Lip

oid

S1

00

(co

nce

ntr

atio

n2

%w

)w

ere

tole

rate

dT

wee

n8

0an

dK

oll

iph

or

HS

15

(co

nce

ntr

a-ti

on

0.0

5%

)is

con

sid

erto

tall

yn

on

-ir

rita

nt

Leo

nar

di

etal

.(2

01

4)

Liq

uid

cry

stal

nan

op

arti

cles

Ex

viv

oco

rnea

lp

enet

rati

on

stu

dy

Ad

ult

New

Zea

lan

dw

hit

era

bb

its

Pil

oca

rpin

en

itra

te(P

N)

To

pic

alad

min

istr

atio

nN

on

tox

ic(g

oo

dco

rnea

lb

io-

com

pat

ibil

ity

of

PN

-L

CN

Ps)

Co

rnea

LC

NP

sw

as2

.05

-fo

ldh

igh

erin

app

aren

tp

er-

mea

bil

ity

coef

fici

ent

than

com

mer

-ci

aley

ed

rop

s

Li

etal

.(2

01

3)

Mag

net

icn

ano

par

ticl

eH

uam

nco

rnea

len

do

thel

ial

cell

s

Co

rnea

Moysi

dis

etal

.(2

01

5)

Th

iola

ted

pec

tin

nan

op

arti

cles

Fre

shly

exci

sed

go

atey

eb

all

Go

atT

imo

lol

mal

eate

(mo

del

dru

g)

–N

ot

rep

ort

edC

orn

eaH

igh

erex

vivo

corn

eal

per

-m

eati

on

wit

hp

rolo

nged

du

rati

on

Sh

arm

aet

al.

(20

12

)

Mic

ron

eed

les

bas

edE

xci

sed

eye

New

Zea

lan

dw

hit

era

bb

its

–M

icro

nee

dle

was

then

inse

rted

per

pen

dic

ula

rto

the

scle

rasu

rfac

e3

mm

No

tre

po

rted

Su

pra

cho

roid

alsp

ace

No

n-N

ewto

nia

nso

luti

on

Tar

get

edd

eliv

ery

wit

hin

Kim

etal

.(2

01

5)

(co

nti

nu

ed)

DOI: 10.3109/17435390.2016.1153165 Please check 5

Dow

nloa

ded

by [

Tex

as A

&M

Uni

vers

ity L

ibra

ries

] at

06:

58 3

1 M

arch

201

6

Page 7: 48 Safety and Toxicity of Nanomaterials for ocular drug delivery applications

Tab

le1

.C

on

tin

ued

Nan

om

ater

ials

Ex

vivo

stu

dy

Invi

vost

ud

yT

her

apeu

tic

del

iver

edR

ou

tes

of

adm

inis

trat

ion

Tox

icit

yT

arget

site

Ou

tco

me

Ref

po

ster

ior

fro

mth

eli

mb

us

sup

rach

oro

i-d

alsp

ace

Hig

hly

pu

rifi

edfu

ller

enes

(HP

Fs)

Hu

man

epid

er-

mal

ker

atin

o-

cyte

s,h

um

anfi

bro

bla

sts

Rab

bit

sN

ot

use

dT

op

ical

app

lica

tio

nC

on

jun

ctiv

are

d-

nes

s,co

rnea

lep

ith

eliu

md

efec

tsan

dm

inim

ally

irri

tati

ng

Eye

and

skin

HP

Fs

did

no

tin

du

cep

ri-

mar

y,cu

mu

la-

tive

skin

irri

tati

on

,sk

inse

nsi

tiza

tio

nan

dsk

inp

ho

tose

nsi

ti-

zati

on

and

ver

ym

ild

physi

cal

def

ects

inra

bb

itey

es

Ao

shim

aet

al.

(20

09

)

Ch

ito

san

nan

oe-

mu

lsio

nan

dn

ano

par

ticl

es

His

tolo

gy

Alb

ino

Rab

bit

Ind

om

eth

acin

To

pic

alin

stil

lati

on

No

nto

xic

,sa

feC

orn

ea,

reti

na

Eyes

of

rab

bit

str

eate

dw

ith

nan

oem

uls

ion

show

edcl

eare

rh

eal-

ing

of

corn

eal

chem

ical

ulc

erw

ith

mo

der

ate

effe

ctiv

ein

hib

itio

no

fp

oly

mo

rph

nu

clea

rle

uck

-o

cyti

cin

filt

ra-

tio

n(P

MN

Ls)

com

par

edw

ith

nan

op

ar-

ticl

esp

rep

arat

ion

Bad

awi

etal

.(2

00

8)

Ch

ito

san

hyla

uro

nic

acid

Hu

man

corn

eal

epit

hel

ial

(HC

E)

and

no

rmal

hu

man

con

jun

ctiv

al(I

OB

A-N

HC

)ce

lls

No

tp

erfo

rmed

pE

GF

Po

rp

b-g

alN

ot

per

form

edN

on

tox

icC

orn

eaH

A-C

HI

nan

o-

par

ticl

estr

ansf

ergen

esto

the

ocu

lar

surf

ace

thro

ug

hC

D4

4re

cep

tor

med

iate

dfl

uid

end

ocy

tosi

s

De

laF

uen

teet

al.

(20

08

)

Ch

ioto

san

and

mag

net

icn

ano

par

ticl

es

Ret

inal

pig

men

tep

ith

elia

lce

lls

New

Zea

lan

dw

hit

ean

dD

utc

hB

elte

dra

bb

its

Gen

eIn

trav

itre

ally

(IV

)o

rsu

bre

-ti

nal

lly

(SR

)

No

tin

flam

ma-

tory

and

did

no

tin

du

cere

tin

alp

ath

olo

gy

Ret

ina

Go

od

tran

sfec

-ti

on

wit

hle

ssto

xic

ity.

Pro

wet

al.

(20

08

)

(co

nti

nu

ed)

6 N.K. Mehra et al. Nanotoxicology, Early Online: 1–25

Dow

nloa

ded

by [

Tex

as A

&M

Uni

vers

ity L

ibra

ries

] at

06:

58 3

1 M

arch

201

6

Page 8: 48 Safety and Toxicity of Nanomaterials for ocular drug delivery applications

Nan

om

ater

ials

Ex

vivo

stu

dy

Invi

vost

ud

yT

her

apeu

tic

del

iver

edR

ou

tes

of

adm

inis

trat

ion

Tox

icit

yT

arget

site

Ou

tco

me

Ref

Go

ldN

ano

par

ticl

es(G

NP

s)

Hu

man

corn

eain

vitr

oR

abb

itco

rnea

sin

vivo

PE

I2-g

ene

To

pic

alro

ute

Low

-tox

icC

orn

eal

tiss

ue

PE

I2-G

NP

sar

elo

wto

xic

,ra

pid

up

take,

and

slow

clea

ran

cean

du

sefu

lfo

rgen

eth

erap

yin

vivo

Sh

arm

aet

al.

(20

11

)

Sil

ver

and

go

ldn

ano

par

ticl

esC

ult

ure

dp

ost

-n

atal

mo

use

reti

na.

C3

Hw

ild

-ty

pe

(wt)

mic

eN

on

eT

ox

icN

P-r

elat

edn

euro

nal

tox

icit

y

Ret

ina

low

con

cen

tra-

tio

ns

of

20

and

80

nm

size

dA

g-

and

Au

NP

sh

ave

adver

seef

fect

so

nth

ere

tin

a,u

sin

gan

org

ano

ty-

pic

reti

na

cul-

ture

mo

del

So

der

stje

rna

etal

.(2

01

4)

PL

GA

nan

op

arti

cles

–N

ewZ

eala

nd

Rab

bit

sP

ran

op

rofe

nE

xv

ivo

corn

eal

per

mea

tio

nst

ud

y

No

tox

icit

yw

asse

enC

orn

eaM

ore

effe

ctiv

ein

ocu

lar

ther

apy

con

tain

ing

azo

ne

Ab

reg

oet

al.

(20

15

)

PA

MA

Md

end

ri-

mer

sse

ries

wit

h-C

OO

Han

d–

OH

fun

ctio

nal

gro

up

s

–N

ewZ

eala

nd

alb

ino

Rab

bit

sP

ilo

carp

ine

nit

rate

and

tro

pic

amid

e–

––

Pro

lon

ged

dru

gre

sid

ence

tim

eV

and

amm

ean

dB

rob

eck

(20

05

)

5.0

GP

PI

den

dri

mer

sN

ot

per

form

edN

ewZ

eala

nd

alb

ino

Rab

bit

sA

ceta

zola

mid

e(A

CZ

)L

ow

ercu

l-d

e-sa

co

fra

bb

its

eye

Hem

oly

tic

tox

icit

y–

AC

Z-l

oad

edP

PI

den

dri

mer

sen

han

ces

ocu

lar

bio

-av

aila

bil

ity,

ocu

lar

resi

-d

ence

tim

e(c

ul-

de

sac)

of

AC

Zw

ith

less

ocu

lar

irri

tan

cy

Mis

hra

and

Jain

(20

14

)

Hy

dro

xyl-

term

i-n

ated

PA

MA

Md

end

rim

ers

–H

om

ozy

go

us

rece

ssiv

erd

yRoyal

Co

lleg

eo

fS

urg

eon

sal

bin

ora

ts(p

ron

eto

ret-

inal

Flu

oci

no

lon

eace

ton

ide

Intr

avit

real

Ou

ter

reti

na

Low

erd

ose

of

dru

gan

dle

sssi

de

effe

cts

and

clea

red

inta

ct

Lez

ziet

al.

(20

12

)

(co

nti

nu

ed)

DOI: 10.3109/17435390.2016.1153165 Please check 7

Dow

nloa

ded

by [

Tex

as A

&M

Uni

vers

ity L

ibra

ries

] at

06:

58 3

1 M

arch

201

6

Page 9: 48 Safety and Toxicity of Nanomaterials for ocular drug delivery applications

Tab

le1

.C

on

tin

ued

Nan

om

ater

ials

Ex

vivo

stu

dy

Invi

vost

ud

yT

her

apeu

tic

del

iver

edR

ou

tes

of

adm

inis

trat

ion

Tox

icit

yT

arget

site

Ou

tco

me

Ref

deg

ener

atio

n)

and

Sp

rag

ue

Daw

ley

rats

Ph

osp

ho

rou

sco

nta

inin

gd

end

rim

ers

–R

abb

its

Car

teo

lol

––

–S

pat

aro

etal

.(2

01

0)

Nan

ocr

yst

als

(hy

dro

xy

pro

pyl

met

hylc

ellu

lose

(HP

MC

),p

olo

xa-

mer

F1

27

and

F6

8,

po

lyso

rbat

e8

0)

Hu

man

corn

eal

epit

hel

ial

cell

(HC

E)

Sev

en-m

on

tho

ldm

ale

Wis

tar

rats

Bri

nzo

lam

ide

(BR

A)

To

pic

alN

o-t

ox

icE

ye

Th

ein

vivo

rat

ocu

lar

hy

per

-te

nsi

on

mo

del

show

edth

esi

gnif

ican

tly

dec

reas

edin

trao

cula

rp

ress

ure

val

ues

of

all

form

ula

tio

ns

Tu

om

ela

etal

.(2

01

4)

P1

23

/TP

GS

mix

edm

icel

les

insi

tugel

s(C

UR

-MM

-IS

Gs)

Ex

viv

oco

rnea

New

Zea

lan

dR

abb

itC

urc

um

inU

sin

gF

ran

z-d

if-

fusi

on

cel

lB

ioco

mp

atib

leC

orn

eaF

rom

exvi

voco

rnea

lp

en-

etra

tio

nst

ud

yin

dic

ated

that

the

cum

ula

-ti

ve

dru

gp

er-

mea

tio

nam

ou

nt

of

CU

R-M

M-

ISG

s(3

:1,

v/

v)

was

,re

spec

tivel

y,1

.16

-fo

ldan

d1

.32

-fo

ldh

igh

erth

anC

UR

-MM

-IS

Gs

(1:1

,v

/v

)an

dcu

rcu

-m

inso

luti

on

.

Du

anet

al.

(20

15

)

Met

hox

yp

oly

(eth

yle

ne

gly

col)

-hex

yls

ub

-st

itu

ted

po

ly(l

ac-

tid

e)m

icel

leca

rrie

rs

Imm

ort

aliz

edh

um

anco

r-n

eal

epit

hel

ial

(HC

E)

cell

s

Fem

ale

alb

ino

New

Zea

lan

dR

abb

it

Cycl

osp

ori

nA

(Cy

A)

To

pic

alN

on

tox

icE

ye

Go

od

tole

ran

ceU

sefu

lfo

rd

ryey

esy

nd

rom

e,au

toim

mu

ne

uvei

tis

and

pre

ven

tio

no

fco

rnea

lg

raft

inje

ctio

n

To

mm

aso

etal

.(2

01

1)

(co

nti

nu

ed)

8 N.K. Mehra et al. Nanotoxicology, Early Online: 1–25

Dow

nloa

ded

by [

Tex

as A

&M

Uni

vers

ity L

ibra

ries

] at

06:

58 3

1 M

arch

201

6

Page 10: 48 Safety and Toxicity of Nanomaterials for ocular drug delivery applications

Nan

om

ater

ials

Ex

vivo

stu

dy

Invi

vost

ud

yT

her

apeu

tic

del

iver

edR

ou

tes

of

adm

inis

trat

ion

Tox

icit

yT

arget

site

Ou

tco

me

Ref

Su

rfac

tan

t;la

den

po

ly-(

2-h

yd

ro-

xyet

hyl

met

h-

acry

late

)p

-H

EM

Ahy

dro

gel

s

Rel

ease

kin

etic

sN

ot

per

form

edC

yA

NA

No

nto

xic

Eye

Bo

thB

rij

97

and

mic

roem

ul-

sio

n-l

aden

hy

do

rgel

sex

hib

itsl

ow

and

exte

nd

edC

yA

rele

ase

for

20

day

san

dco

ntr

oll

ed

Kap

oo

r&

Ch

ou

nan

(20

08

)

Sh

ear-

resp

on

sive

op

hth

alm

ichy

dro

gel

Hu

man

corn

eal

epit

hel

ial

(HC

E)

and

reti

nal

pig

-m

ent

epit

he-

lial

cell

s(A

RP

E1

9ce

lls,

po

ster

-io

rse

gm

ent)

New

Zea

lan

dW

hit

e(N

ZW

)ra

bb

its

–In

trav

itre

alN

oo

pac

ific

atio

no

fle

nes

,co

rnea

or

vit

rou

s,n

or

any

fun

ctio

nal

and

mo

rph

o-

log

ical

chan

ges

toth

ep

ost

erio

rse

g-

men

to

fey

e.

Ret

ina

and

corn

eaS

hea

r-re

spo

nsi

ve

op

hth

alm

ichy

dro

gel

show

sp

rom

is-

ing

po

ten

tial

ino

ph

thal

mic

del

iver

yin

bo

than

teri

or

and

po

ster

ior

seg

men

tas

wel

lv

itre

alre

pla

cem

ent.

Sh

eik

ho

lesh

lam

iet

al.

(20

15

)

MW

CN

Ts;

MW

CN

Ts-

OH

MW

CN

Ts-

CO

OH

Ret

inal

pig

men

tep

ith

eliu

m(R

PE

)ce

lls

No

tp

erfo

rmed

NA

NA

Car

box

yl-

MW

CN

Ts

are

no

tto

xic

toey

e

Co

rnea

Th

eca

rbox

yl-

MW

CN

Ts

show

bes

to

cula

rb

io-

com

pat

ibil

ity

and

less

tox

ic.

Lu

etal

.(2

01

3)

MW

CN

Ts

of

two

dif

fere

nt

len

gth

sE

gg

Ch

ori

on

All

anto

icM

emb

ran

e(H

E-C

AM

)

Eye

irri

tati

on

test

on

rab

bit

NA

Co

nju

nct

ival

sac

of

left

eye

No

nir

rita

nt

Eye

Invi

voo

cula

rte

stre

sult

ssu

gges

tth

atM

WC

NT

of

bo

thsi

zes

wer

em

inim

-al

lyir

rita

tin

gw

her

eas,

the

invi

tro

HE

-C

AM

resu

ltis

no

n-i

rrit

ant

Kis

ho

reet

al.

(20

09

)

Sin

gle

-wal

led

carb

on

nan

oh

orn

s

Sk

inp

rim

ary

test

;C

on

jun

ctiv

alir

rita

tio

n(d

raiz

e)te

st;

Sk

inse

nsi

-ti

zati

on

test

;In

trat

rach

eal

inst

illa

tio

n

Sk

inp

rim

ary

and

eye

irri

tati

on

test

(rab

bit

s);

Sk

inse

nsi

-ti

zati

on

test

(gu

inea

pig

s);

per

ora

lad

min

istr

atio

nte

stan

d

NA

Intr

atra

chea

lin

stil

lati

on

and

per

ora

lad

min

istr

atio

nro

ute

s

Qu

ite

low

-acu

tep

ero

ral

tox

-ic

ity

was

fou

nd

(42

00

0m

g/k

gb

od

yw

eig

ht)

Co

rnea

As-

gro

wn

SW

CN

Hs

fou

nd

no

n-

irri

tan

tan

dn

on

-der

mal

sen

siti

zer

No

corn

eal

inju

ryw

aso

bse

rved

Miy

awak

iet

al.

(20

08

)

(co

nti

nu

ed)

DOI: 10.3109/17435390.2016.1153165 Please check 9

Dow

nloa

ded

by [

Tex

as A

&M

Uni

vers

ity L

ibra

ries

] at

06:

58 3

1 M

arch

201

6

Page 11: 48 Safety and Toxicity of Nanomaterials for ocular drug delivery applications

Tab

le1

.C

on

tin

ued

Nan

om

ater

ials

Ex

vivo

stu

dy

Invi

vost

ud

yT

her

apeu

tic

del

iver

edR

ou

tes

of

adm

inis

trat

ion

Tox

icit

yT

arget

site

Ou

tco

me

Ref

test

;P

ero

ral

adm

inis

trat

ion

test

intr

atra

chea

lin

stil

lati

on

(rat

s)

iney

eir

rita

-ti

on

test

Cd

Se/

Zn

Sco

re/

shel

lQ

Ds

Fre

shly

der

ived

bov

ine

corn

ea;

bov

ine

cor-

nea

lfi

bro

bla

st(B

CF

)an

db

ov

ine

skin

fib

rob

last

(BS

F)

C5

7B

L/6

Jm

ice

No

tu

sed

Intr

astr

om

alin

ject

ion

Acc

um

ula

tio

no

fQ

Ds

into

the

corn

eal

stro

-m

alce

lls

Co

rnea

Th

eQ

Ds

get

accu

mu

late

wit

hin

the

corn

eas

for

lon

ger

du

r-at

ion

(26

d)

Ku

oet

al.

(20

11

)

Hu

man

reti

nal

pig

men

tep

i-th

eliu

m(R

PE

)

Rab

bit

sN

ot

use

dIn

trav

itre

alin

ject

ion

Lit

tle

chan

ge

iney

ebal

lap

pea

ran

ce,

IOP

,an

dey

esig

ht

Vit

rou

sG

Od

idn

ot

cau

sean

ysi

gn

ifi-

can

tto

xic

ity

toth

ece

llg

row

than

dp

roli

fera

tio

n.

Intr

avit

real

inje

ctio

no

fG

Oin

tora

b-

bit

s’ey

esd

idn

ot

lead

tom

uch

chan

ge

inth

eey

ebal

lap

pea

ran

ce,

IOP

,el

ectr

o-

reti

no

gra

m,

and

his

to-

log

ical

exam

inat

ion

Yan

etal

.(2

012)

Co

llo

idal

go

ldn

ano

rod

sE

xv

ivo

on

po

r-ci

ne

eyes

Po

rcin

e–

Las

erp

uls

ead

min

istr

atio

nT

her

mal

dam

age

con

firm

edC

apsu

les

Th

erm

ald

amag

ew

ith

in5

0–

70mm

inra

dia

ld

ista

nce

Rat

toet

al.

(20

09

)

Hea

t-se

nsi

tive

or

hea

t-an

dp

H-

sen

siti

ve

lip

oso

mes

HU

VE

Can

dA

RP

ER

abb

its

Cal

cein

Intr

avit

real

inje

ctio

ns

Did

no

tsh

ow

san

yto

xic

ity

Ret

ina

Th

ep

H-

and

hea

t-se

nsi

tive

lip

oso

me

show

edti

me-

and

site

-sp

e-ci

fic

dru

gd

eliv

ery

Laj

un

enet

al.

(20

15

)

(co

nti

nu

ed)

10 N.K. Mehra et al. Nanotoxicology, Early Online: 1–25

Dow

nloa

ded

by [

Tex

as A

&M

Uni

vers

ity L

ibra

ries

] at

06:

58 3

1 M

arch

201

6

Page 12: 48 Safety and Toxicity of Nanomaterials for ocular drug delivery applications

Nan

om

ater

ials

Ex

vivo

stu

dy

Invi

vost

ud

yT

her

apeu

tic

del

iver

edR

ou

tes

of

adm

inis

trat

ion

Tox

icit

yT

arget

site

Ou

tco

me

Ref

Su

bm

icro

n-s

ized

lip

oso

mes

(ssL

ips)

Neu

ron

alp

recu

r-so

rce

llli

ne

(RG

C-5

)

Alb

ino

dd

Ym

ice

Ed

arav

on

eIn

trav

itre

alad

min

istr

atio

nR

etin

ald

amag

eR

etin

aE

dar

avo

ne-

load

edli

po

-so

mes

red

uce

dox

i-d

ativ

est

ress

-in

du

ced

ret-

inal

dam

age

mo

rest

ron

gly

rath

erth

anfr

eed

rug

su

po

nin

trav

i-tr

eal

inje

ctio

n

Hir

on

aka

etal

.(2

01

1)

Ch

ito

san

–li

po

som

esN

ewZ

eala

nd

Alb

ino

Rab

bit

s

Dic

lofe

nac

sod

ium

Low

erco

nju

nc-

tival

sac

No

irri

tati

on

and

tox

icC

orn

eaL

CH

-co

ated

lip

oso

mes

show

edsi

g-

nif

ican

tly

pro

-lo

nged

rete

n-

tio

nco

mp

ared

wit

hn

on

-co

ated

lip

o-

som

esan

dd

iclo

fen

acso

diu

mso

luti

on

Li

etal

.(2

00

9)

DOI: 10.3109/17435390.2016.1153165 Please check 11

Dow

nloa

ded

by [

Tex

as A

&M

Uni

vers

ity L

ibra

ries

] at

06:

58 3

1 M

arch

201

6

Page 13: 48 Safety and Toxicity of Nanomaterials for ocular drug delivery applications

Carbon nanomaterials

Currently, carbon-based nanomaterials (CNMs) have attractedattention in ocular therapeutics delivery and targeting. For CNMssuch as carbon nanohorns (CNHs), poly hydroxy fullerenes(PHFs), carbon nanorods (CNRs), carbon nanodots (CNDs),CNTs, and graphenes, their inherent toxicity due to the presenceof metallic and amorphous impurities restricts their application inocular delivery. On the other hand, the unique physicochemicalproperties and scalability have rendered CNTs as ideal candidatesfor targeted drug delivery, including ophthalmic administration.The CNTs are available in four categories depending on thenumber of walls, i.e., single- (SWCNTs), double- (DWCNTs),triple- (TWCNTs), and multi-walled CNTs (MWCNTs) (Lacerdaet al., 2012; Mehra & Jain, 2015a–c; Mehra & Palakurthi, 2015;Mehra et al., 2014a,b, 2015a; Sharma et al., 2015).

As we know, CNTs are present in some cosmetic products,such as the eye liner kohl (Kajal; Surma), used in Indian culture.Kohl obtained from various minerals has been used as an eyecosmetic since ancient times. It is believed by some that the kohlprotects eyes and provides strength to the infants’ eyes. Literaturesuggests that kohl (Kajal) has been used for the prevention andtreatment of eye diseases such as cataract, conjunctivitis andblepharitis (Mehra et al., 2015a–c). The increased production andbiomedical applications of new multifunctional CNTs haveincreased the exposure to the environment and public. Thus, thesafety, efficacy, and toxicity issues are crucial to ensure the safeuse of CNTs and to address the major concerns of non-biodegradability, high aspect ratio, and similarity with asbestosand vitreous fibers (Boczkowski & Lanone, 2012; Kayat et al.,2011; Jain et al., 2014,2015). Due to their needle-like tubularstructure, multifunctional CNTs may be useful in ophthalmicdelivery. Du et al (2013) explored the relationship betweenphysicochemical properties and production of CNTs, along withthe potential risks to human health with exposure to thesenanomaterials.

Two different kinds of multi-walled CNTs (MWCNT 1: 5–8mm in length with 3–8 nm inside diameter and outside diameterof 140 ± 30 nm and MWCNT 2: 1–10 mm in length with 2–6 nminside diameter and outside diameter of 10–15 nm) have beenused in an eye irritation test in the rabbit (Kishore et al., 2009).Eye irritation is summarized as a ‘‘maximum average score’’,which an average value is scored based on corneal injury forindividual animals at the time of scoring. The MWCNTs weredispersed in distilled water with vortexing and the average particlesize (nm) and polydispersity index (0 to 1 with 0¼monodisperseand 1¼ polydisperse) for MWCNT 1 was 901 nm and 1, and forMWCNT was 554 nm and 1.000 and 1.00, respectively The meantissue viability of C1 (the size of MWCNT 1 was 166 nm), C2 (thesize of MWCNT 2 was 100 nm was450%, considered to be a non-irritant in SkinEthicTM reconstructed human epidermis skincorrosion test. A total of about 18 mg (an equivalent weight to0.1 mL) of the test substance (MWCNT 1 and MWCNT 2) wasapplied into the conjunctival sac of the left eye after gently pullingthe lower lid away from the eyeball, while the right eye served asthe control. The conjunctiva, iris and cornea of both eyes wereevaluated and scored according to the Draize method up to 72 h.Both sizes of MWCNTs were minimally irritating in the in vivoocular test (Kishore et al., 2009).

Ema et al. evaluated the eye irritation of two different kinds ofMWCNTs (Nikkiso-MWCNTs and MWCNT-7) and single-walled CNTs (SWCNTs), Nikkiso-SWCNTs and super-growthSWCNTs, using an eye irritation test in rabbits. After instillationof either the MWCNTs or SWCNTs into the conjunctival sac,there were no signs of corneal opacity, abnormality of the iris orchemosis (swelling of conjunctiva) in accordance to the

Organization for Economic Co-operation and DevelopmentGuideline 405 ‘‘Acute Eye Irritation/Corrosion’’ (OECD, 2002).Hyperanemia of the conjunctival blood vessels was observed 24 hfollowing instillation of the Nikkiso-MWCNTs, but this resolvedwithin 48 h. There also was no evidence of corneal epitheliumdamage. Overall, the SWCNTs and MWCNTs in this studyappeared to exhibit minimal eye irritation (Ema et al., 2011).

The ocular biocompatibility of plasma-modified MWCNTs(pristine, hydroxyl, and carboxyl modified MWCNTs) withhuman retinal pigment epithelium (RPE) cells was reported forthe first time by Yan et al. (2013). The reactive oxygen speciesgeneration was visible but not severe; however, MWCNTs-carboxyl exposure promoted a low oxidative stress level. Theplasma- and carboxyl-modified MWCNTs showed better ocularbiocompatibility (Lu et al., 2013). Although CNTs were widelyinvestigated for drug delivery and targeting, few studies havereported their toxicological properties (Jain et al., 2007).

Carbon nanohorns

CNHs represent a new type of CNMs and comprise of singlegraphene tubes (2–5 nm in diameter and 40–50 nm in tubulelength) with a conically closed tip. The CNHs (approximately2000 horns) form spherical aggregates with a diameter of 80–120 nm, which are reminiscent of dahlia flowers. They also formbuds and seeds and can target and accumulate within tumorsbecause of an enhanced permeability and retention (EPR) effect(Guerra et al., 2012). Additionally, CNHs do not contain metallicimpurities, a common cause for any toxicity of carbonnanomaterials.

In vitro and in vivo toxicity studies of as-grown SWCNHs(tubular nanocarbons having no metallic impurity) were per-formed by Miyawaki et al. (2008). The authors reported that theas-grown SWCNHs did not show any signs of conjunctivalirritation (Draize score¼ 0) in rabbits. Figure 3 shows no cornealinjury in the rabbit eye after an eye irritation test.

The international standard assay for acute ocular toxicity is therabbit in vivo Draize eye test (Draize et al., 1994), which wasdeveloped by the Food and Drug Administration. The procedureinvolves applying 100ml (100 mg if solid) of the test substanceonto the cornea and the conjunctival sac of one eye of the

Figure 3.Photo images of rabbit eye after eye irritation test: (a) untreated;(b) 1 h after SWCNHs dose in conjunctival sac without eye washing(reproduced with copyright permission from Miyawaki et al., 2008,American Chemical Society).

12 N.K. Mehra et al. Nanotoxicology, Early Online: 1–25

Dow

nloa

ded

by [

Tex

as A

&M

Uni

vers

ity L

ibra

ries

] at

06:

58 3

1 M

arch

201

6

Page 14: 48 Safety and Toxicity of Nanomaterials for ocular drug delivery applications

conscious rabbit. Rabbits are observed up to 21 d at regularintervals for signs of irritation including redness, swelling,cloudiness, edema, hemorrhage, discharge, and blindness(Huhtala et al., 2008).

Graphenes

Graphenes are two-dimensional, monoatomic thick buildingblocks of carbon atoms packed into a honeycomb lattice. Theyshow great potential in drug delivery due to excellent biocom-patibility and aqueous dispersibility. Graphene oxide has highspecific surface area and large �-conjugated aromatic structure.Thus, various biomolecules such as doxorubicin, camptothecin,and paclitaxel can be loaded through �–� stacking interactions(Angelopoulou et al., 2015; Mao et al., 2013; Wang et al., 2011;Yang et al., 2015). However, pure graphene nanosheets areunsuitable for drug delivery and targeting due to their hydropho-bic nature and tendency to form agglomerates (Liu et al., 2015).

Up to now, reports on ocular biocompatibility of graphene andits derivatives are sparse. The toxicity and intraocular biocom-patibility of graphene oxide (GO) on human RPE cells werestudied by measuring cell viability and membrane integrity (Yanet al., 2012). The lactate dehydrogenase (LDH) assay was used tomeasure membrane integrity because LDH is a cytosolic enzymethat is released into the culture medium upon cell membranedamage (Hillegass et al., 2010). The RPE cells treated with GOsolutions displayed higher than 60% cell viability and less than 8%LDH release. An insignificant number of cells (1.5%) showedapoptosis. The authors also found that the intravitreal injection ofGO into the eyes of rabbits did not promote any changes in eyeballappearance, eyesight, intraocular pressure (IOP), electroretino-gram measurements, and histopathology (Figures 4 and 5). Theirfindings clearly revealed the potential of GO for clinicaldiagnosis, imaging and drug delivery applications in ophthalmol-ogy (Yan et al., 2012).

Figure 5. Digital camera of the rabbit’s eyeball from (a) the control eye, and (b) the 0.3 mg of GO-injected eye after injection for 49 d. Histologicalappearance of (c) the control retina and (d) the 0.3 mg of GO-injected retina after injection for 49 d (reproduced with copyright permission from Yanet al. 2012, American Chemical Society).

Figure 4. Digital photos of the experimental rabbit (top) and slit-lampfundus photo of the control eye (bottom). Slit-lamp fundus photos of (b)intravitreally injected eyes after the injection of 0.1 mg of GO, (c) 0.2 mg of GO, and (d) 0.3 mg of GO for 2 (top) and 49 d (bottom) (reproduced withcopyright permission from Yan et al., 2012, American Chemical Society).

DOI: 10.3109/17435390.2016.1153165 Please check 13

Dow

nloa

ded

by [

Tex

as A

&M

Uni

vers

ity L

ibra

ries

] at

06:

58 3

1 M

arch

201

6

Page 15: 48 Safety and Toxicity of Nanomaterials for ocular drug delivery applications

More recent reports by Chen et al. (2015) studied thenanotoxicity of GO during zebrafish embryogenesis and foundthat GO damaged the mitochondria and induced developmentalmalformation of the eyes. An alternative graphene derivative toGO that has been considered for ocular delivery is hydroxylatedgraphene (G–OH). The genotoxicity and ocular biocompatibilityof G–OH prepared by ball milling was reported by Lin et al.(2015). Genotoxicity and in vivo ocular biocompatibility studiesare essential for large-scale up and commercialization of G–OH.The possible genotoxicity (using the comet assay to measuredeoxyribonucleic acid strand breaks) was induced by G–OHat4100 mg/mL concentrations. Intravitreal injection of G–OH inrabbits gradually decreased genotoxicity after 4 weeks devoid ofdamage to cell morphology, structure, and most parts of the eyes.The authors also reported that the G–OH caused some changes ineyesight-related functions (IOP, ERG, and retinal structureschanges) (Lin et al., 2015). Thus, suitable eye protection maybe recommended for people who are dealing with graphene-basedmaterials frequently in the laboratory.

Fullerenes

Fullerenes are the third stable allotrope of carbon (in addition tographite and diamond) and were discovered in 1985. They are

spherical carbon cages with sp2 hybridized molecules and consistof 60 carbon atoms. Polyhydroxy-fullerenes (PHF) are watersoluble, negatively charged, biocompatible, functionalized fuller-enes molecules. The potent antioxidant nature of PHF hasattracted great attention in pharmaceutical applications.However, only a few studies have reported on the toxicity andsafety of highly purified fullerenes based on the screening of eyeand skin damage.

Our research group has determined the toxicity of carboxy-and hydroxy-terminal PHF in rabbit corneal epithelial (RCE) cellsand porcine retinal arterioles in vitro. The RCE cells were treatedwith different concentrations of PHF (carboxy terminal PHF; F1;hydroxy terminal PHF; F2) for 24 h at 37 �C and the cell viabilitywas assessed by the methyl thiazoletetrazolium assay andpresented in Figure 6. The IC50 values of the F1 and F2 werecalculated to be 26.8 and 27.5 mM, respectively. To study thedirect and acute impact of nanomaterials on vasomotor functionof retinal arterioles isolated from pigs, the concentration-dependent vasodilation to known endothelium-dependent nitricoxide-mediated vasodilator bradykinin (1 pM–10 nM) was exam-ined in the absence or presence of F1 and F2 for up to 2 h. Asshown in Figure 7, bradykinin at 0.1 nM produced significantdilation of the isolated retinal arteriole (i.e., the diameter wasincreased from the resting state of 45–57 mm; Figure 7A). At a

Figure 6. Percent cell viability of rabbitcorneal epithelial cells after treatment withcarboxy terminal PHF-F1; hydroxy terminalPHF-F2 nanomaterials at 0.01–10 mM. Valuesrepresented as mean ± SD.

Figure 7. (A) Retinal arteriolar images observed on the video monitor for internal diameter recordings. The diameter was recorded in the absence ofagonists (resting tone) and in the presence of bradykinin. (B) Retinal arterioles dilated to bradykinin. The vasodilations were reduced after treating thevessels with 1 mM carboxy terminal PHF-F1nanomaterials for 2 h. The dilations were not affected by 0.3 mM F1 (n¼ 4 for each group, ANOVA*p50.05).

14 N.K. Mehra et al. Nanotoxicology, Early Online: 1–25

Dow

nloa

ded

by [

Tex

as A

&M

Uni

vers

ity L

ibra

ries

] at

06:

58 3

1 M

arch

201

6

Page 16: 48 Safety and Toxicity of Nanomaterials for ocular drug delivery applications

higher concentration (10 nM) of bradykinin, the vessel furtherdilated to 83 mm. Interestingly, when vessels were treated with F1at 1mM intraluminally for 2 h, the concentration-dependentdilation to bradykinin was significantly impaired (Figure 7A).The low concentration of F1 (0.3 mM) did not alter the dilation tobradykinin (Figure 7B). The lack of toxicity was also observedwith F2 nanomaterials at 0.3 mM (data not shown). These pilotdata suggest that F1 exerts an adverse effect possibly on theendothelial cells and compromises the vasodilator function ofretinal arterioles.

Experimental studies have also examined the effect offullerene soot and highly purified fullerenes on the eye. Huczkoet al. (2001) reported no eye irritation in guinea pigs withfullerene soot in the draize eye irritation test. In addition,Aoshima et al. (2009) performed the primary eye-irritation test inrabbits and observed conjunctival redness and corneal epitheliumdefects following fullerenes application (presumed to be due toinsoluble fullerene powder with an average particle size of 100–300mm), but the symptoms disappeared within 48 h.

Metallic nanoparticles

Metallic nanoparticles including nanogold, nanosilver, quantumdots, and metallic oxides have been used in ocular delivery. Up tonow, limited studies have been performed for in vivo corneal genetherapy using gold nanoparticles (GNPs). It is known that bothacquired and genetic factors may affect the corneal transparencyand may lead to the loss of vision (Sharma et al., 2011). Readersmay refer to ‘‘silver nano-a trove for retinal therapies’’ for a moredetailed account on retinal therapies (Kalishwaralal et al., 2010).

The gene transfer potential and toxicity of 2-kDa polyethyle-neimine (PEI) conjugated gold nanoparticles (PEI2-GNPs) werestudied in the human cornea in vitro and the rabbit cornea in vivo(Sharma et al., 2011). Exposure of primary human cornealfibroblasts isolated from donor corneas to PEI2-GNPs mixed withDNA plasmids allowed for effective gene delivery withoutdamaging the cells. The topical route of application, as performedin the in vivo study with PEI2-GNPs, is the most convenient routefor delivering therapeutics into eye because of the absence ofpharmacokinetic challenges. The slit-lamp biomicroscopy clearlyrevealed the lack of edema, redness, and cellular infiltration in thePEI2-GNP-treated rabbit corneas, suggesting low toxicity withcorneal PEI2-GNP therapy (Figure 8). Transmission electronmicroscopy studies confirmed the internalization of PEI2-GNPs

in corneal cells in vivo via an endocytic mechanism after topicalapplication with gradual clearance over time (Figure 9). Thefindings suggest that GNPs did not harm corneal cells and theyslowly cleared from the rabbit cornea. Low toxicity, rapid uptake,and slow clearance of the PEI2-GNPs may open new doors in thecoming days to the ophthalmologist and researchers.

The internalization, apoptosis, oxidative stress, glial, andmicroglial activities of silver and GNPs on C3H wild-type miceretinal tissue were studied (Soderstjerna et al., 2014). The NP-related neuronal toxicity was evident by the higher number ofapoptotic cells, oxidative stress, and increased glial staining andmicroglial activation (hallmark of neural tissue insult). Lowconcentrations of Ag- (20 nm size) and AuNPs (80 nm size)showed adverse effects on the retina, using an organotypic retinaculture model.

Quantum dots

QDs are semiconductor nanocrystals of groups III–IV and II–VIin the range of 1–10 nm in diameter (Bajwa et al., 2015a,b;Volkov, 2015). Along with their small size and variable shape,they exhibit unique optical properties such as tunable emission,strong brightness, high photostability, and high resistance tophotobleaching, which has attracted attention for investigations indrug delivery, targeting, and imaging. A few studies have beenreported on the safety, routes of administration, and toxicity. In arecent study, the impact of the common CdSe/ZnS core/shell QDson the cornea was evaluated in bovine eyes in vitro and mice eyesin vivo (Kuo et al., 2011). The exposure to QDs at a concentrationof 20 nM for 48 h reduced the viability (50%) of bovine cornealfibroblasts (Figure 10). Twenty-six days after intrastromal injec-tion in the mice cornea, these QDs penetrated and were retainedwithin the corneal stroma (Figure 11). Because corneal stromacells play an important role in the maintenance of the health andtransparency of the cornea, these findings suggest potentialtoxicity with prolonged accumulation of QDs. Thus, furtherresearch is required to examine the exposure and possible risk ofQDs for ocular applications.

Dendrimers

Dendrimers (from the Greek word ‘‘Dendron’’ for tree) arehyperbranched synthetic macromolecules with monodisperse(uniform) repeating units and a globular three-dimensional

Figure 8. Representative clinical eye examination images acquired with a slit-lamp microscope in PEI2-GNP-treated and untreated (control) rabbitcorneas at 12 h and 7 d post-application. No opacity, redness, or inflammation was seen. PEI2-GNP-treated corneas show mild purple coloration at 12 h,confirming GNP uptake, and clearing of coloration detected on day 7 suggests that there was no GNP accumulation in the cornea (reproduced withcopyright permission from Sharma et al., 2011, Elsevier Pvt Ltd).

DOI: 10.3109/17435390.2016.1153165 Please check 15

Dow

nloa

ded

by [

Tex

as A

&M

Uni

vers

ity L

ibra

ries

] at

06:

58 3

1 M

arch

201

6

Page 17: 48 Safety and Toxicity of Nanomaterials for ocular drug delivery applications

morphology. They have been used as one of the promisingnanomaterials in ocular drug delivery. While the cationic natureof amine-terminated dendrimers enhances the epithelial mem-brane permeability, strong interactions with the negatively

charged membrane may result in toxicity (necrosis and apoptosis).Thus, surface engineering is needed to alleviate toxicity of thecationic dendrimers (Jain et al., 2015; Mehra et al., 2015a–c;Mishra & Jain, 2014; Yellepeddi et al., 2013).

Figure 10. Cell viability of QD-corneal fibroblast using MTT assays. *significant compared with the control (bovine cornea fibroblast incubatedwithout QDs) (reproduced with copyright permission from Kuo et al., 2011 American Chemical Society).

Figure 9. Representative TEM images of PEI2-GNP-treated rabbit corneas demonstrating the presence and intracellular trafficking of GNPs in (A)keratocytes and (B) extracellular matrix. GNPs can be seen in the endosomes near the cell surface indicating their uptake by endocytosis. Rupturedendosome represents the release of GNPs into the cytoplasm (reproduced with copyright permission from Sharma et al., 2011, Elsevier Pvt Ltd).

16 N.K. Mehra et al. Nanotoxicology, Early Online: 1–25

Dow

nloa

ded

by [

Tex

as A

&M

Uni

vers

ity L

ibra

ries

] at

06:

58 3

1 M

arch

201

6

Page 18: 48 Safety and Toxicity of Nanomaterials for ocular drug delivery applications

Experimental in vivo evidence has been provided to supportthe potential use of dendrimers as drug carriers in the eye. Acommon type for drug delivery is poly(amidoamine) (PAMAM)dendrimers. The bioadhesive properties and strong interaction ofPAMAM dendrimers with the corneal surface likely contributedto the increased bioavailability of pilocarpine (an anti-glaucomamedication) and tropicamide (anticholinergic drug used to dilatethe pupil) following ocular instillation of the PAMAM–drugsolutions in rabbits (Vandamme & Brobeck, 2005).The PAMAMdendrimers appear to be safe for topical application because theydid not cause ocular irritation. Another type of dendrimer-containing phosphorous was synthesized and tested for its abilityto improve ocular delivery of carteolol (an ocular anti-hyperten-sive drug used in treating glaucoma) in vivo in rabbits (Spataroet al., 2010). Although solubility of the carteolol–dendrimersmixture was low, this formulation did not cause irritation up to 8 hpost-instillation on the cornea and increased the amount ofcarteolol delivered to the aqueous humor. In a recent study,Mishra and Jain (2014) developed a topically effective acetazo-lamide (ACZ)-loaded poly(propylene imine) (PPI) dendrimers(ACZ/PPI) for glaucoma. The ACZ/PPI dendrimers enhancedocular residence time and showed efficacy in lowering IOP. Theocular irritation toxicity of ACZ/PPI dendrimers from themodified Draize test revealed that the dendrimer formulationswere safe for ocular delivery.

In the United States, atrophic (dry) age-related maculardegeneration (AMD) and retinitis pigmentosa (RP) are leadingcauses of blindness and no effective treatments are currentlyavailable for these chronic retinal diseases. Retinal neuroinflam-mation triggered by activated microglia appears to play a pivotalrole in the retinal pigment epithelial and photoreceptor cell loss inRP and AMD. In a recent study, hydroxyl-terminated PAMAMdendrimers were found to selectively localize within activatedmicroglia following intravitreal injection in the Royal College ofSurgeons (RCS) rat retinal degeneration model (Lezzi et al.,2012). The dendrimers were then conjugated to fluocinoloneacetonide and tested for their efficacy in drug delivery. In the RCSrats, there was a greater reduction in neuroinflammation andretinal degeneration one month after intravitreal administration ofthe dendrimers-steroid formulation than the free steroid alone.These findings along with the ability of only the dendrimers-steroid formulation to preserve neural retina function (electro-retinogram assessment) over 9 weeks suggest that these dendri-mers are safe for intravitreal delivery.

Glaucoma is a neurodegenerative disease with increased IOP,oxidative stress, and impaired ocular blood flow and it is a major

cause of irreversible blindness and visual impairment. Accordingto the World Health Organization, approximately 67 millionpeople are affected worldwide and the projected figure will reach80 million by the year 2020. A single therapeutic agent cannotoften control the IOP because of the multifactorial nature ofglaucoma and due to short duration of action. A combination oftwo or more anti-glaucoma therapeutic agents is a better way tocontrol and reduce IOP as well as to improve patient compliance.Up to now, various combinations of therapeutic agents have beendeveloped and are available in the market including latanoprost/timolol maleate, dorzolamide/timolol maleate and brimonidinetartrate/timolol maleate (Desai et al., 2010; Holden et al., 2012;Yang et al., 2012). Recently, Yang et al. (2012) co-deliveredtimolol maleate and brimonidine using a novel hybrid PAMAMdendrimers hydrogel/poly (lactic-co-glycolic acid) (PLGA) nano-particle platform (HDNP) as an eye drop formulation in normo-tensive adult Dutch-belted rabbits. After one-time topicalapplication, the HDNP formulation led to a sustained andeffective reduction in IOP for 4 d. This treatment also signifi-cantly increased the absorption of both anti-glaucoma agents inthe aqueous humor and cornea as well as absorption of timololmaleate in the conjunctiva of the eye for 1 week (Figure 12)without promoting inflammation or structural changes in the eye.Collectively, these results support the ability of a newly developedHDNP to enhance drug bioavailability and sustain IOP reductionfor several days with potential clinical application for improvinglong-term patient compliance and reducing dosing frequency aswell the enormous costs in ocular therapy.

Nanowafers

Dry eye disorder is one of the major public health issues and acommon eye disease, affecting millions of people worldwide.About 30% of patients have reported symptoms of mild or chronicdry eye disease. It is characterized by blurred vision, eyeirritation, light sensitivity, and ocular surface epithelial disease.Injuries on the surface of the eye due to dry eye disease disruptcorneal angiogenic privilege and trigger corneal neovasculariza-tion, which can lead to loss of vision (Azar, 2006; Coursey et al.,2015; Yuan et al., 2015). Dry eye is a multifactorial disease andactivates an innate immune response in the conjunctiva andcornea (Coursey et al., 2015). Currently, artificial tears and anti-inflammatory eye drops are being used for the treatment of dryeye disease and the eye drops are administered several times a dayto reach therapeutic efficacy. Because drug loaded contact lensescould not release the drug for an extended period of time,

Figure 11. (a) In vivo external photographs of mice corneas following an intrastromal injection of PBS (right eye) and QDs (left eye). (b) Themagnified image of the left eye. (c) Two-photon imaging of mice corneas 26 d following intrastromal QD injection. The arrow indicates theintracytoplasmic-localized QDs (reproduced with copyright permission from Kuo et al., 2011, American Chemical Society).

DOI: 10.3109/17435390.2016.1153165 Please check 17

Dow

nloa

ded

by [

Tex

as A

&M

Uni

vers

ity L

ibra

ries

] at

06:

58 3

1 M

arch

201

6

Page 19: 48 Safety and Toxicity of Nanomaterials for ocular drug delivery applications

nanowafers were developed. Nanowafers are readily applied bythe fingertip to the eye like a contact lens. This technology wasdeveloped by Acharya et al. (2015) in the Department ofOphthalmology at the Baylor College of Medicine. The nanowaferis a small/tiny transparent circular disc that contains arrays ofdrug-loaded nanoreservoirs. It can be applied on to the ocularsurface using a fingertip and it withstands constant blinkingdevoid of being displaced. The nanowafers delivery system couldpotentially be used for glaucoma, eye injuries, chronic dry eye,infections, and other ocular inflammatory conditions (Courseyet al., 2015; Yuan et al., 2015). Nanowafers release the drug for alonger duration of time, and subsequently the device slowlydissolves as the drug is released. Nanowafers can be easily placedonto the ocular surface, which could deliver therapeutics moreconsistently for longer duration of time to the eye as well asreduce the frequent administration of dosage forms and providebetter efficacy and feasibility. The dexamethasone (Dex) loadednanowafers (Dex-NW) were developed to deliver drugs on theocular surface for a longer duration of time (Coursey et al., 2015).Dex is a glucocorticoid with potent anti-inflammatory propertiesand is well documented for its efficacy in treating ocularinflammation and dry eye-related corneal epithelial diseases.The Dex-NW contains carboxy methyl cellulose and a 500 mmsquare drug reservoir, which were filled with Dex. The device waseffective in downregulating expression of chemokines (CXCL-10and CCL-5) and inflammatory cytokines (TNF-a and IFN-g).

Acharya et al. (2015) selected four different polymers,poly(vinyl alcohol), polyvinyl pyrrolidone, (hydroxyl pro-pyl)methyl cellulose, and carboxylmethyl cellulose (CMC), fordesigning and fabricating Nanowafers via hydrogel templatestrategy. These polymers are hydrophilic, transparent, biocom-patible, and mucoadhesive and have film-forming properties,

which allow them to readily adhere onto a wet mucosal surfaceand conform to the curvature of the eye. These fabricatedNanowafers contained sunitinib, sorafenib, and axitinib, which arealready in clinical use as anti-angiogenic therapeutics to treat late-stage renal carcinoma (Yuan et al., 2015). In a murine ocular burnmodel, once-a-day administration of axitinib-loaded nanowaferswas more effective than twice-a-day therapy with axitinib eyedrops in treating corneal neovascularization (Yuan et al., 2015).The afitinib-nanowafers were also non-toxic and did not disruptthe wound healing of the cornea. These findings suggest thatnanowafers could provide a safe and effective drug deliverysystem at a lower dosing frequency for ocular therapy.

Hydro- and nano-gel

Hydrogel is a polymer network that is expanded throughout itswhole volume by water. Nanogels are hydrogels consisting of ananoscale hydrophilic polymer network, which is loaded with adrug and can be used for delivering both hydrophilic andhydrophobic drugs. Based on how polymer networks areformed, hydrogels can be classified into physical and chemicalgels. A wide variety of synthetic, semi-synthetic and naturalpolymers such as hyaluronic acid, PVA, chitosan, acrylatecopolymers, collagen, alginate, and poly(ethylene glycol areused for hydrogel formulations (Kirchhof et al., 2015; Kompellaet al., 2013).

Toxicity evaluation of biodegradable and thermosensitivepoly(ethylene glycol)–poly(epsilon-caprolactone)–poly(ethyleneglycol) hydrogel as an in situ sustained ophthalmic drug deliverysystem showed slight corneal endothelial damage (Yin et al.,2010). The ion-sensitive hydrogels were developed using gellangum, kappa-carrageenan, and alginates (natural polysaccharides)

Figure 12. Representative fundus cameraimages of rabbits instilled with HDNP andNP formulations. FluoSpheres wereentrapped in dendrimer hydrogel or PBS pH7.4 instead of drug-encapsulating PLGAnanoparticles. Images were taken at the endof days 1, 3, 5, and 7 of instillation offormulations. Left panel of each group is theregular fundus camera image of the eyes, andright panel is a fluorescence fundus cameraimage (reproduced with copyright permissionfrom Yang et al., 2012, American ChemicalSociety).

18 N.K. Mehra et al. Nanotoxicology, Early Online: 1–25

Dow

nloa

ded

by [

Tex

as A

&M

Uni

vers

ity L

ibra

ries

] at

06:

58 3

1 M

arch

201

6

Page 20: 48 Safety and Toxicity of Nanomaterials for ocular drug delivery applications

for ophthalmic drug delivery systems. To assess the ocular safety,the ion-sensitive hydrogel was instilled into the eye of maleSprague–Dawley rates every 12 h for 3 months. No signs ofcorneal anterior surface damage, conjunctival injury, redness, orincreased tearing were observed by direct visual observation or byimmunohistochemical techniques (Figures 13 and 14)(Fernandez-Ferreiro et al., 2015).

Liposomes

Liposomes are biocompatible and biodegradable lipid assembliesand were evaluated for the first time in the 1980s with the mainobjective of enhancing corneal penetration of ocular therapeutics.Liposomes may vary in size, lipid composition, surface charge,method of preparation, and fluidity of bilayers. Liposomes havinga positive charge and nano-size range can lead to prolongedresidence time with improved therapeutic effects. The liposomescan encapsulate both hydrophilic and hydrophobic drugs and

release them through passive diffusion, vesicle fusion, and vesicledisruption (Kompella et al., 2013; Reimondez-Troitino et al.,2015). Liposomes may cause blurred vision and a possibility ofaggregation during storage or in vivo when colloidal stability ispoor. In addition, cationic surface charge of liposomes might beresponsible for inducing inflammation (Bochot & Fattal, 2012).Pattni et al. (2015) very recently reviewed ‘‘New Developments inLipsomal Drug Delivery’’ in Chemical Reviews Journal.

In 2007, positively charged multi-lamellar liposomes com-posed of egg phosphatidylcholine:cholesterol:stearylamine (7:7:1molar ratio) were prepared using the reverse-phase evaporationmethod and then loaded with the ophthalmic drug ACZ. Theliposomes showed ACZ percent encapsulation efficiency(48.27%). The authors reported that the study failed concerningthe effect of the liposomal formulation on improving ocularresidence time and ocular irritancy (Hathout et al., 2007).

Chitosan is a polysaccharide that is derived from crustaceanshells and is used in ocular drug and gene delivery. It is a natural

Figure 13. Macrophotography of the right eye of a Sprague–Dawley rat after the instillation of 7.5 mL of the ion-sensitive hydrogel containing Trypanblue (eeproduced with copyright permission from Fernandez-Ferreiro et al., 2015, Elsevier Pvt Ltd).

DOI: 10.3109/17435390.2016.1153165 Please check 19

Dow

nloa

ded

by [

Tex

as A

&M

Uni

vers

ity L

ibra

ries

] at

06:

58 3

1 M

arch

201

6

Page 21: 48 Safety and Toxicity of Nanomaterials for ocular drug delivery applications

cationic polymer with biocompatibility and mucoadhesive nature.Because of its cationic nature, chitosan nanoparticles can easilybind with the negatively charged cornea and maximize the cornealpenetration (Li et al., 2009; Prow, 2010). The low molecularweight chitosan (LCH, 8 kDa)-coated liposomes were preparedand assessed for in vitro and in vivo ocular delivery of diclofenacsodium (Li et al., 2009). From the ocular tolerance study, noirritation or toxicity was observed even after continual adminis-tration of the LCH-coated liposome in New Zealand albinorabbits for a total period of 7 d.

Poly-L-lysine (PLL) is a water-soluble cationic peptide and isused as a modifier for drugs. It enhances the cellular uptake ofdrugs by increasing the permeability of various compounds. PLLwas coated on the surface of submicronized (100 nm) liposomesand toxicity was determined in human corneal (RIKEN Cell Bank)and conjunctival (Wong–Kilbourne derivative of Chang conjunc-tiva, clone 1–5c-41, CCL-20.2) cell lines and interocular distribu-tion was assessed following eye drop administration in mice. Thefindings suggest that the PLL-modified liposomes are non-toxic incorneal or conjunctival cells and are effective for drug delivery tothe retina as eye drop formulations (Sasaki et al., 2013).

Hironaka et al. (2011) developed edaravone-loaded liposomesfor protection against oxidative stress-induced retinal damage.The retinal damage was induced by intravitreal injection of N-methyl-D-aspartate in Hank’s balanced salt solution (HBSS-Mesbuffer).The edaravone-loaded liposomes reduced oxidative stress-induced retinal damage more strongly than the free drug uponintravitreal injection. The results suggested that the edaravone-loaded liposomes remarkably improved the in vivo efficacy(Hironaka et al., 2011).

In another study, a liposomal formulation did not show anytoxic effects on human umbilical vein endothelial cells (HUVECs)and human retinal pigment epithelial (ARPE-19) cells. Theliposomes were developed employing heat-sensitive or heat- andpH-sensitive lipid composition with rod- or star -shapednanoparticles (Lajunen et al., 2015).

Micelles

Polymeric micelles are 10–100 nm self-assemblies of amphiphilicblock copolymers above critical micellar concentration (CMC)with a hydrophobic core surrounded by a hydrophilic shell.Depending on the solvent and amphiphilic molecule choice,different micellar structures are formed (standard, reverse, andunimolecular micelles). Amphiphilic block copolymers are easilytailored to prolong the micelles stability in eyefluids, inducebioadhesion, and modify drug release. The polymeric micelleshave been shown to improve drug permeation across the ocularepithelia with minimal or no irritation, leading to enhanced ocularbioavailability (Kaur & Kakkar, 2014; Kompella et al., 2013).

The itraconazole-incorporated amphiphilic block copolymer-based polymeric micelles (optimized micelles formulation) with asize of 79.99 nm, 91.32 ± 1.73% entrapment efficiency, and invitro percentage permeation (90.28 ± 0.31%) in an in situ oculargel was developed for the management of fungal keratitis (Jaiswalet al., 2015). The optimized itraconazole polymeric micellesloaded (optimized in situ gel) in an in situ ocular gel (0.5% w/vcarbopol 943 NF, 6.84 ± 0.34 pH, 47.3 ± 4.50 gelling time inseconds, and 82.22 ± 1.91% drug content) showed superior ex vivotranscorneal permeation as compared with Itral

�eye drops (Jawa

Pharmaceuticals, Gurgaon, India). No irritation was observedeven after repetitive use of the micelle formulation. Antifungalactivity was performed using the agar cup plate method withCandida albicans. The in situ gel showed a nearly 19 nm zone ofinhibition, which was higher than the zone of inhibition (about14 nm) with Itral

�eye drops.

Nanoemulsion

Nanoemulsions are composed of two immiscible phases (liquids),wherein one phase is dispersed as droplets in another phase andstabilized by surfactant (stabilizer). The nanoemulsion can beprepared using low viscosity grade surfactant, oil, and water. Thepresence of surfactant or in combination with co-surfactantsreduces the interfacial tension within nanoemulsion to facilitatedispersion, increases membrane permeability, and enhances druguptake. The non-toxic, non-irritant nature of surfactant, oil, andco-surfactants to the corneal surface and other ocular tissues/cellsis an important parameter (Daull et al., 2013; Gallarate et al.,2013; Lallemand et al., 2012; Kaur & Kakkar, 2014; Lee et al.,2015).

Difluprednate (6a, 9-difluoro-11b,17,21-trihydroxypregna-1,4-diene-3,20-dione 17-butyrate 21-acetate; DFBA) is a syn-thetic glucocorticoid used for the treatment of post-operativeinflammation and pain. DFBA is hydrolyzed into deacetylatedmetabolite 6a,9-difluoro-11b,17,21-trihydroxypregna-1,4-diene-3,20-dione 17-butyrate (DFB) in the aqueous humor afterinstillation into the eye. However, DFB shows similar activityas that of DFBA (Yamaguchi et al., 2005). DUREZOL

�(0.05%

difluprednate ophthalmic emulsion), a topical corticosteroid, isindicated for the treatment of inflammation, pain associated withocular surgery, and endogenous anterior uveitis. Safety andefficacy studies of DUREZOL

�with two or four times a day

application were conducted in 438 patients in 26 clinics in theUnited States. Based on the results, only 3% of patients showedclinically significant IOP rise in both DUREZOL

�groups

(�10 mm Hg and�21 mm Hg from baseline, respectively). Theauthors did not report any serious ocular adverse events and feweradverse events were found in difluprednate-treated groups ascompared with the placebo group (Korenfield et al., 2009).

Cyclosporine A (CsA) is a potent immunosuppressive agent. Itinhibits proliferation and action of T lymphocytes by blocking thetranscription of cytokines (Liang et al., 2012). CsA is useful forthe treatment of various ophthalmic diseases (dry eyes, uveitis inchildren and adolescents, peripheral ulcerative keratitis, andvernal keratoconjunctivitis) (Kapoor & Chauhan, 2008). Thesafety profile of three different CsA cationic emulsion formula-tions: (i) 0.05% CsA-CEm, (ii) commercial 0.05% CsA-anionicemulsion (CsA-AEm, Restasis

�), and (iii) 0.05% CsA-Oil solution

was tested in vitro with a dynamic corneal wound healing assayemploying HCE cells, and in vivo in a rabbit acute toxicity model(Liang et al., 2012). The modified Draize test scale was used forthe safety evaluation of conjunctival hyperemia, chemosis, andpurulent secretions upon instillation of the emulsions formulation.Upon repeated instillations, all three of the CsA emulsionformulations were well tolerated by the rabbit ocular surface.The acute toxicity of these emulsions was studied in a rabbit acuteirritation model. Phosphate-buffered saline (PBS) served as thenegative control, while 0.02% benzalkonium chloride (BAK) wasa positive control for toxicity. The CsA emulsion formulationsshowed a moderate irritation score (Draize test score ranging from2.6 to 4.5) at 75 min, which was higher than that obtained in thePBS-treated group (0.8) and lower than in the 0.02% BAK group.The 0.02% BAK caused the highest toxicity, chemosis withdamaged corneal epithelium, redness, and inflammatory cellinfiltration. However, CsA in anionic emulsion (CsA-A Em;Restasis

�; Allergan) and CsA in oil (CsA-Oil; medium-chain

triglycerides, Mygliol�

812; IMCD, Saint-Denis, France) inducedmoderate infiltrations of inflammatory cells near conjunctiva-associated lymphoid tissues structures (Liang et al., 2012).

Restasis is a microemulsion of cyclosporine (an immunosup-presive agent) and is used in the treatment of chronic dry eye,which may be caused by inflammation. The most common ocular

20 N.K. Mehra et al. Nanotoxicology, Early Online: 1–25

Dow

nloa

ded

by [

Tex

as A

&M

Uni

vers

ity L

ibra

ries

] at

06:

58 3

1 M

arch

201

6

Page 22: 48 Safety and Toxicity of Nanomaterials for ocular drug delivery applications

side effects of Restasis, including ocular burning (17%), conjunc-tival hypermia, epiphora, pruritus, stinging and visual disturb-ances, eye pain, and foreign body sensation, have been reported in1–5% of patients (http://www.drugs.com/restasis).

A novel pH triggered nanoemulsified in situ ophthalmic gel offluconazole (NE-ISG) was designed, developed, and tested via exvivo application to the goat eye for transcorneal permeation andcorneal toxicity. These studies revealed that NE-ISG causedgreater permeation of fluconazole in the cornea than didcommercial eye drops of the antifungal agent without showingany visual signs of ocular tissue damage (Pathak et al., 2013). Acomplete list of various ophthalmic products available in themarket is shown in Table 2.

Conclusion

Currently, several types of nanomaterials are available fortherapeutic delivery and targeting to the eye in the treatment ofvarious eye-associated diseases. Surfactants are generally used toimprove the dispersibility and to reduce the size of the emulsionin both ocular nanoemulsions and nanosuspension formulations.The concentration of surfactant used has to be carefully evaluatedas higher surfactant concentrations because it may cause irritationand corneal damage. While ionic surfactants are toxic, non-ionicsurfactants like polaxamers, polysorbabtes, polyethylene glycol,and tyloxapol may be used at pre-assessed concentrations withoutsignificant toxicity. For example, polysorbate 80 is the mostwidely used in many ocular emulsion preparations and isconsidered safe.

Restasis� is a lipid emulsion containing of 0.05% CsA. Now-a-days contact lenses (Ocusert

�and contact lenses) have been

most widely used and studied owing to their higher degree ofcomfort and biocompatibility. As we know, drawbacks with someof the existing nanomaterials, such as nanoparticles (low loadingefficiency), dendrimers (toxicity due to peripheral functionalgroups), liposomes (stability issue) and carbon nanomaterials(accumulation in lungs), need to be resolved.

The selection of a suitable animal model in assessment ofocular toxicity of the nanomaterials and the product is importantfor the development of safe and effective ocular formulations. Therabbit eye is more sensitive and susceptible to irritant nanomater-ials than the human eye and is considered to be a suitable model.Toxicity of the nanomaterials-based ophthalmic formulation canbe best evaluated by corneal opacity, eye irritation (Draize eyetest), hypersensitivity, LVET, and ocular and non-ocular organo-typic methods. The corneal opacity test is generally performed onthe cornea of rabbits and it is most widely used in the assessmentof corneal toxicity. The LVET is a refinement of the Draize eyetest wherein a tenth of the amount (10 mL or 10 mg) of the testsubstance used in the Draize test is applied to the cornea (notconjunctival sac) of the right eye of the animal with no forcedeyelid closure employed. The pathological changes are observedin the cornea, conjunctiva and iris-ciliary body. While the Draizetest is often considered for its over-prediction of the humanresponse, LVET is considered more accurate (Roggeband et al.,2000). However, LVET is not adopted by any regulatory agencyas an alternate test. The reluctance to adopt the LVET may be dueto the fact that it does not offer the element of ‘‘exaggeration’’present in the Draize test that helps to assure public safety.Recently, legislation to reduce the animal testing methods and topromote alternative methods (deceased animal tissues, 2D and 3Dcell culture models) has been introduced in many countries. Acomprehensive review on current techniques for ocular toxicitywas presented by Wilson et al. (2015).

In conclusion, as novel nanomaterials are being developed,thorough physicochemical characterization followed by a batteryT

able

2.

Cu

rren

tst

atu

so

fn

ano

mat

eria

lsb

ased

op

hth

alm

icfo

rmu

lati

on

s.

Nan

om

ater

ials

scaf

fold

sP

rod

uct

des

crip

tio

nP

rod

uct

nam

eO

cula

rth

erap

euti

csA

pp

lica

tio

nC

urr

ent

stat

us

Ph

arm

aceu

tica

lco

mp

any

Mic

roem

uls

ion

LIP

IMIX

isp

rese

rvat

ive-

free

;si

ng

le-u

sefo

rmu

lati

on

.L

ipim

ixT

M–

Dry

eye

ther

apy

Mar

ket

edin

US

Aan

dE

uro

pe

Tu

bil

ux

Ph

arm

a,It

aly

Res

tory

lm

iner

alo

ils

Ad

van

ced

lip

idre

sto

rati

ve

top

rote

ctag

ain

stte

arlo

ssS

oo

theX

P�E

mo

lien

t–

Dry

eye

ther

apy

Mar

ket

edin

US

Aan

dE

uro

pe

Bau

sch

&L

om

b

Gel

Lu

bri

cati

ng

op

hth

alm

icg

elO

ftag

el–

Mil

dd

ryey

esy

nd

rom

eM

arket

edin

Eu

rop

eS

ante

nS

AS

Cat

ion

icem

uls

ion

Cat

ion

icem

uls

ion

Cat

hio

no

rm�

–M

ild

dry

eye

syn

dro

me

Mar

ket

edin

Eu

rop

eS

ante

nS

AS

solu

tio

nO

ph

thal

mic

solu

tio

nH

yp

rosa

n�

Hy

pro

mel

lose

Dry

eye

ther

apy

Mar

ket

edin

Eu

rop

eS

ante

nS

AS

Em

uls

ion

KE

RV

IS1

mg

/mL

eye

dro

ps,

emu

lsio

nIk

erv

is�

Cy

AM

od

erat

eto

sever

ed

ryey

eP

has

eII

Icl

inic

altr

ials

San

ten

SA

S

Em

uls

ion

Cat

ion

icem

uls

ion

Vek

acia

�C

yA

Ver

nal

ker

ato

con

jun

ctiv

itis

Ap

pro

ved

Nov

agal

iP

har

mE

mu

lsio

nC

atio

nic

emu

lsio

nC

atio

pro

st�

Lat

ano

pro

stG

lau

com

aA

pp

roved

Nov

agal

iP

har

mE

mu

lsio

nO

ph

thal

mic

emu

lsio

nR

esta

sis�

CyA

Dry

eye

syndro

me

Mar

ket

edin

US

AA

ller

gan

Em

uls

ion

Op

hth

alm

icem

uls

ion

Du

rezo

l�D

iflu

pre

dn

ate

Ocu

lar

infl

amm

atio

nM

arket

edin

US

AA

lco

nL

abo

rato

ries

Tin

yp

arti

cles

Eye

spra

yO

pte

x-A

ctim

istT

M–

Dry

eye

syn

dro

me

Mar

ket

edin

Eu

rop

eO

pti

ma

Ph

arm

azeu

tisc

he

So

luti

on

So

luti

on

Vis

ud

yn

e�V

erte

po

rfin

Cla

ssic

sub

fovea

lch

oro

idal

neo

vas

cula

riza

tio

n,

resu

med

ocu

lar

his

op

lasm

osi

s

InM

arket

Bau

sch

&L

om

b

Mu

cus

pen

etra

tin

gp

arti

cles

(MP

P)

Mu

cus

pen

etra

tin

gp

arti

cles

(MP

P)

Le-

MP

PL

ote

pre

dn

ole

tab

on

ate

Ocu

lar

infl

amm

atio

nD

ryey

esy

nd

rom

eM

arket

edII

/III

clin

ical

tria

lsK

ala

Ph

arm

aceu

tica

l

Liq

uid

gel

Liq

uid

gel

Cli

nit

asG

elC

arb

om

erD

ryey

esy

nd

rom

eIn

Mar

ket

Alt

aco

rP

har

mac

euti

cal

DOI: 10.3109/17435390.2016.1153165 Please check 21

Dow

nloa

ded

by [

Tex

as A

&M

Uni

vers

ity L

ibra

ries

] at

06:

58 3

1 M

arch

201

6

Page 23: 48 Safety and Toxicity of Nanomaterials for ocular drug delivery applications

of in vitro and in vivo ocular toxicity evaluation tests are neededfor a comprehensive understanding of the interactions ofnanoparticles at the molecular, cellular, and tissue levels. Theseinteractions would determine the biocompatibility, stability andbiological performance and side effects of the nanomaterials.Such a knowledge base will in turn allow in silico hazardassessment of the nanomaterials and guide the development ofbenign, next-generation nanomaterials.

Disclosure statement

The authors report that they have no conflicts of interest. Thecontent is solely the responsibility of the authors and does notrepresent the official views of the Food and Drug Administration.

Funding information

This work was supported, in part, by a grant from the Food andDrug Administration, Center for Drug Evaluation Research(U01FD05184) to S. P.

References

Abrego G, Alvarado H, Souto EB, Guevara B, Bellowa LH, Parra A, et al2015. Biopharmaceutical profile of pranoprofen-loaded PLGA nano-particles containing hydrogels for ocular administration. Eur J PharmBiopharm J Biopharm 95:261–70.

Aksungur P, Demirbilek M, Denkbas EB, Vandervoort J, Ludwig A, UnluN. 2011. Development and characterization of cyclosporine A loadednanoparticles for ocular drug delivery: cellular toxicity, uptake andkinetic studies. J Control Release 151:286–94.

Almeida H, Amaral MH, Lobao P, Lobo JMS. 2014. In situ gellingsystems: a strategy to improve the bioavailability of ophthalmicpharmaceutical formulations. Drug Discov Today 19:400–12.

Anayol MA, Toklu Y, Kamberoglu EA, Raza S, Arifoglu HB, Simavli H,et al 2014. Short-term effects of intravitreal triamcinolone acetonideinjection on ocular blood flow evaluated with color Doppler ultrason-ography. Int J Ophthalmol 7:811–5.

Andrei F, Peptu CA, Popa M, Desbrieres J, Peptu C, Gardikiotis F, et al2015. Formulation and evaluation of cefuroxim loaded submicronparticles for ophthalmic delivery. Int J Pharm 493:16–29.

Angelopoulou A, Voulgari E, Diamanti EK, Gournis D, Avgoustakis K.2015. Graphene oxide stabilized by PLA-PEG copolymers for thecontrolled delivery of paclitaxel. Eur J Pharm Biopharm 93:18–26.

Aoshima H, Saitoh Y, Ito S, Yamana S, Miwa N. 2009. Safety evaluationof highly purified fullerenes (HPFs): based on screening of eye andskin damage. J Toxicol Sci 34:555–62.

Azar DT. 2006. Corneal angiogenic privilege: angiogenic and antiangio-genic factors in corneal avascularity, vasculogenesis, and woundhealing. Trans Am Ophthalmol Soc 104:264–302.

Badawi AA, El-Laithy HM, El Qidra EK, Mofty HE, EI Dally M. 2008.Chitosan based nanocarriers for indomethacin ocular delivery. ArchPharm Res 31:1040–9.

Bajwa N, Mehra NK, Jain K, Jain NK. 2015a. Pharmaceutical andbiomedical applications of quantum dots. Artif Cells NanomedBiotechnol. doi: 10.3109/21691401.2015.1052468).

Bajwa N, Mehra NK, Jain K, Jain NK. 2015b. Targeted anticancer drugdelivery through anthracycline antibiotic bearing functionalized quan-tum dots. Artif Cells Nanomed Biotechnol. doi: 10.3109/21691401.2015.1102740).

Barile FA. 2010. Validating and troubleshooting ocular in vitro toxicologytests. J Pharmacol Toxicol Methods 61:136–45.

Bhatta RS, Chandasana H, Chhonker YS, Rathi C, Kumar D, Mitra K,Shukla PK. 2012. Mucoadhesive nanoparticles for prolonged oculardelivery of natamycin: in vitro and pharmacokinetics studies. Int JPharm 432:105–12.

Bochot A, Fattal E. 2012. Liposomes for intravitreal drug delivery: a stateof the art. J Control Release 161:628–34.

Boczkowski J, Lanone S. 2012. Respiratory toxicities of nanomaterials –a focus on carbon nanotubes. Adv Drug Deliv Rev 64:1694–9.

Bozukova D, Pagnoulle C, Jerome R, Jerome C. 2010. Polymers inmodern ophthalmic implants-historical background and recentadvances. Mat Sci Eng R 69:63–83.

Cai D, Gao W, He B, Dai W, Zhang H, Wang X, et al 2014. Hydrophobicpenetrating peptide PFVYLi-modified stealth liposomes for doxorubi-cin delivery in breast cancer therapy. Biomaterials 35:2283–94.

Chen D, Dougherty CA, Zhu K, Hong H. 2015. Theranostic applicationsof carbon nanomaterials in cancer: focus on imaging and cargodelivery. J Control Release 210:230–45.

Chen Y, Hu X, Sun J, Zhou O. 2015. Specific nanotoxicity of grapheneoxide during zebrafish embryogenesis. Nanotoxicology. doi: 10.3109/17435390.2015.1005032.

Chetoni P, Burgalassi S, Monti D, Najarro M, Boldrini E. 2007.Liposome-encapsulated mitomycin C for the reduction of cornealhealing rate and ocular toxicity. J Drug Deliv Sci Technol 17:43–8.

Chetoni P, Monti D, Tampucci S, Matteoli B, Ceccherini-Nelli L, SubissiA, Burgalassi S. 2015. Liposomes as a potential ocular delivery systemof distamycin A. Int J Pharm 492:120–6.

Conley SM, Naash MI. 2010. Nanoparticles for retinal gene therapy. ProgRetin Eye Res 29:376–97.

Coursey TG, Henriksson JT, Marcano DC, Shin CS, Isenhart LC, AhmedF, et al 2015. Dexamethasone nanowafer as an effective therapy for dryeye disease. J Control Release 213:168–74.

Das S, Bellare JR, Banerjee R. 2012. Protein based nanoparticles asplatforms for aspirin delivery for ophthalmologic applications. ColloidsSurf B Biointerfaces 93:161–8.

Dong P, Guan Y, Yang J, He M, Wan C. 2000. Fundus microvascular flowmonitoring during retrograde cerebral perfusion: an experimentalstudy. Ann Thorac Surg 70:1478–82.

Daull P, Lallemand F, Garrigue JS. 2013. Benefits of cetalkoniumchloride cationic oil-in-water nanoemulsions for topical ophthalmicdrug delivery. J Pharm Pharmacol 66:531–41.

De la Fuente M, Seijo B, Alonso MJ. 2008. Novel hyaluronic acid-chitosan nanoparticles for ocular gene therapy. Invest Ophthalmol VisSci 49:2016–24.

Desai PN, Yuan Q, Yang H. 2010. Synthesis and characterization ofphotocurable polyamidoamine dendrimer hydrogels as a versatileplatform for tissue engineering and drug delivery. Biomacromolecules11:666–73.

Devadasu VR, BHardwaj V, Kumar MNV. 2013. Can controversialnanotechnology promise drug delivery? Chem Rev 113:1686–735.

Diebold Y, Calonge M. 2010. Applications of nanoparticles in ophthal-mology. Prog Retin Eye Res 29:596–609.

Dilbaghi N, Kaur H, Ahuja M, Kumar S. 2013. Evaluation oftropicamide-loaded tamarind seed xyloglucan nanoaggregates forophthalmic delivery. Carbohydr Polym 94:286–91.

Draize JH, Woodard G, Calvery HO. 1994. Methods for the study ofirritation and toxicity of substances applied topically to the skin andmucous membranes. J Pharmacol Exp Ther 82:377–90.

Du J, Wang S, You H, Zhao X. 2013. Understanding the toxicity ofcarbon nanotubes in the environment is crucial to the control ofnanomaterials in producing and processing and the assessment ofhealth risk for human: a review. Environ Toxicol Pharmacol 36:451–62.

Duan Y, Cai X, Du H, Zhai G. 2015. Novel in situ gel systems based onP123/TPGS mixed micelles and gellan gum for ophthalmic delivery ofcurcumin. Colloids Surf B Biointerfaces 128:322–30.

Ema M, Matsuda A, Kobayashi N, Naya M, Nakanishi J. 2011. Evaluationof dermal and eye irritation and skin sensitization due to carbonnanotubes. Regul Toxicol Pharmacol 61:276–81.

Fernandez-Ferreiro A, Barcia MG, Martinez MG, Prado AV, Lema I,Argibay B, et al 2015. In vitro and in vivo ocular safety and eye surfacepermanence determination by direct and magnetic resonance imagingof ion-sensitive hydrogels based on gellan gum and kappa-carrageenan.Eur J Pharm Biopharm 94:342–51.

Fitzpatrick SD, Mazumder MAJ, Muirhead B, Sheardown H. 2012.Development of injectable, resorbable drug-releasing copolymerscaffolds for minimally invasive sustained ophthalmic therapeutics.Acta Biomater 8:2517–28.

Fuller JR, Bevin TH, Molteno AC, Vote BJ, Herbison P. 2002. Anti-inflammatory fibrosis suppression in threatened trabeculectomy blebfailure produces good long term control of intraocular pressure withoutrisk of sight threatening complications. Br J Ophthalmol 86:1352–4.

Gallarate M, Chirio D, Bussano R, Peira E, Battaglia L, Baratta F, TrottaM. 2013. Development of O/W nanoemulsions for ophthalmicadministration of timolol. Int J Pharm 440:126–34.

Gan L, Gan Y, Zhu C, Zhang X, Zhu J. 2009. Novel micro-emulsion insitu electrolyte-triggered gelling systems for ophthalmic delivery of

22 N.K. Mehra et al. Nanotoxicology, Early Online: 1–25

Dow

nloa

ded

by [

Tex

as A

&M

Uni

vers

ity L

ibra

ries

] at

06:

58 3

1 M

arch

201

6

Page 24: 48 Safety and Toxicity of Nanomaterials for ocular drug delivery applications

lipophilic cyclosporine A: in vitro and in vivo results. Int J Pharm 365:143–9.

Griffith JF, Nixon GA, Bruce RD, Reer PJ, Bannan EA. 1980. Dose–response studies with chemical irritants in the albino rabbit eye as abasis for selecting optimum testing conditions for predicting hazard tothe human eye. Toxicol Appl Pharmacol 55:501–13.

Garcia-Fernandez MJ, Tabary N, Martel B, Cazaux F, Oliva A, Taboada ,et al 2013. Pol-(cyclo) dextrins as ethoxzolamide carriers in ophthal-mic solutions and in contact lenses. Carbohydr Polym 98:1343–52.

Guerra J, Herrero MA, Carrion B, Perez-Martinez FC, Lucio M, Rubio N,et al 2012. Carbon nanohorns functionalized with polyamidoaminedendrimers as efficient biocarrier materials for gene therapy. Carbon50:2832–44.

Habib FS, Fouad EA, Abdel-Rhaman MS, Fathalla D. 2010. Liposomesas an ocular delivery system of fluconazole: in vitro studies. ActaOphthalmol 88:901–4.

Halim Mohamed MA, Mahmoud AA. 2011. Formulation of indomethacineye drops via complexation with cyclodextrins. Curr Eye Res 36:208–16.

Hamalainen KM, Kananen K, Auriola S, Kontturi K, Urtti A. 1997.Characterization of paracellular and aqueous penetration routes incornea, conjunctiva, and sclera. Invest Ophthalmol Vis Sci 38:627–34.

Hathout RM, Mansour S, Mortada ND, Guinedi AS. 2007. Liposomes asan ocular delivery system for acetazolamide: in vitro and in vivostudies. AAPS PharmSciTech 8:E1–12.

Hillegass JM, Shukla A, Lathrop SA, MacPherson MB, Beuschel SL,Butnor KJ, et al 2010. Inflammation precedes the development ofhuman malignant mesotheliomas in a SCID mouse xenograft model.Ann N Y Acad Sci 1203:7–14.

Hironaka K, Inokuchi Y, Fujisawa T, Shimazaki H, Akane M, Tozuka Y,et al 2011. Edaravone-loaded liposomes for retinal protection againstoxidative stress-induced retinal damage. Eur J Pharm Biopharm 79:119–25.

Holden CA, Tyagi P, Thakur A, Kadam R, Jadhav G, Kompella UB, YangH. 2012. Polyamidoamine dendrimer hydrogel for enhanced delivery ofantiglaucoma drugs. Nanomedicine 8:776–83.

Hsueh PY, Edman MC, Sun G, Shi P, Xu S, Lin Y, et al 2015. Tear-mediated delivery of nanoparticles through transcytosis of the lacrimalgland. J Control Release 208:2–13.

Huang W, Collette W, Twamley M, Aguirre SA, Sacaan A. 2015.Application of electroretinography (ERG) in early drug developmentfor assessing retinal toxicity in rats. Toxicol Appl Pharmacol 289:525–33.

Available at: http://www.drugs.com/restasis.html. Accessed on 13December 2015.

Huczko A, Lange H, Calko E, Grubek-Jaworska H, Droszcz P. 2001.Physiological testing of carbon nanotubes: are they asbestos-like?Fullerene Sci Technol 9:251–4.

Huhtala A, Salminen L, Tahti H, Uusitalo H. 2008. Corneal models forthe toxicity testing of drugs and drug releasing materials. In:Ashammakhi N, ed. Topics in Multifunctional Biomaterials andDevices, 1–24.

Ibrahim HK, El-Leithy IS, Makky AA. 2010. Mucoadhesive nanoparti-cles as carrier systems for prolonged ocular delivery of gatifloxacin/prednisolone bitherapy. Mol Pharm 7:576–85.

Jain AK, Mehra NK, Lodhi N, Dubey V, Mishra DK, Jain PK, Jain NK.2007. Carbon nanotubes and their toxicity. Nanotoxicology 1:167–97.

Jain K, Mehra NK, Jain NK. 2014. Potentials and emerging trends innanopharmacology. Curr Opin Pharmacol 15:97–106.

Jain K, Mehra NK, Jain NK. 2015. Nanotechnology in drug delivery:safety and toxicity issues. Curr Pharm Des 21:4252–61.

Jaiswal M, Kumar M, Pathak K. 2015. Zero order delivery of itraconazolevia polymeric micelles incorporated in situ ocular gel for themanagement of fungal keratitis. Colloids Surf B Biointerfaces 130:23–30.

Kjellstrom U, Andreasson S, Ponjavic V. 2014. Attenuation of the retinalnerve fibre layer and reduced retinal function assessed by opticalcoherence tomography and full-field electroretinography in patientsexposed to vigabatrin medication. Acta Ophthalmol 92:149–57.

Kalishwaralal K, BarathManiKanth S, Pandian SRK, Deepak V,Gurunathan S. 2010. Silver nano – a trove for retinal therapies. JControl Release 145:76–90.

Kapoor Y, Chauhan A. 2008. Ophthalmic delivery of cyclosporine A fromBrij-97 microemulsion and surfactant-laden p-HEMA hydrogels. Int JPharm 361:222–9.

Kapoor Y, Thomas JC, Tan G, John VT, Chauhan A. 2009. Surfactant-laden soft contact lenses for extended delivery of ophthalmic drugs.Biomaterials 30:867–78.

Kayat J, Gajbhiye V, Tekade RK, Jain NK. 2011. Pulmonary toxicity ofcarbon nanotubes: a systematic report. Nanomedicine 7:40–9.

Kim YC, Oh KH, Edelhauser HF, Prausnitz MR. 2015. Formulation totarget delivery to the ciliary body and choroid via the suprachoroidalspace of the eye using microneedles. Eur J Pharm Biopharm. doi:10.1016/j.ejpb.2015.05.020.

Kirchhof S, Goepferich AM, Brandl FP. 2015. Hydrogels in ophthalmicapplications. Eur J Pharm Biopharm. doi: 10.1016/j.ejpb.2015.05.016.

Kishore AS, Surekha PS, Murthy PB. 2009. Assessment of the dermaland ocular irritation potential of multi-walled carbon nanotubes byusing in vitro and in vivo methods. Toxicol Lett 191:268–74.

Kompella UB, Amrite AC, Ravi RP, Durazo SA. 2013. Nanomedicinesfor back of the eye drug delivery, gene delivery and imaging. ProgRetin Eye Res 36:172–98.

Korenfield MS, Silverstein SM, Cooke DL, Vogel R, Crockett RS. 2009.Difluprednate ophthalmic emulsion 0.05% for postoperative inflam-mation and pain. J Cataract Refract Surg 35:26–34.

Koz OG, Ozhuy S, Tezel GG, et al 2007. The effect of paclitaxel onconjunctival wound healing: a pilot study. J Glaucoma 16:610–5.

Kozobolis VP, Christodoulakis EV, Tzanakis N, Zacharopoulos I,Pallikaris IG. 2002. Primary deep sclerectomy versus primary deepsclerectomy with the use of mitomycin C in primary open-angleglaucoma. J Glaucoma 11:287–93.

Kuo TR, Lee CF, Lin SJ, Dong CY, Chen CC, Tan HY. 2011. Studies ofintracorneal distribution and cytotoxicity of quantum dots: riskassessment of eye exposure. Chem Res Toxicol 24:253–61.

Lajunen T, Viitala L, Kontturi LS, Laaksonen T, Liang H, Vuorimaa-Laukkanen E, et al 2015. Light induced cytosolic drug delivery fromliposomes with gold nanoparticles. J Control Release 203:85–98.

Lallemand F, Duall P, Benita S, Buggage R, Garrigue JS. 2012.Successfully improving ocular drug delivery using the cationicnanoemulsion, Novasorb. J Drug Deliv 2012:604204.

Lee EH, Kim JK, Lim JS, Lim SJ. 2015. Enhancement of indocyaninegreen stability and cellular uptake by incorporating cationic lipid intoindocyanine green-loaded nanoemulsions. Colloids Surf BBiointerfaces 136:305–13.

Leonardi A, Bucolo C, Romano GL, Platania CBM, Drago F, Puglisi G,Pignaello R. 2014. Influence of different surfactants on the techno-logical properties and in vivo ocular tolerability of lipid nanoparticles.Int J Pharm 470:133–40.

Lezzi R, Guru BR, Glybina IV, Mishra MK, Kennedy A, Kannan RM.2012. Dendrimer-based targeted intravitreal therapy for sustainedattenuation of neuroinflammation in retinal degeneration. Biomaterials33:979–88.

Li J, Wu L, Wu W, Wang B, Wang Z, Xin H, Xu Q. 2013. A potentialcarrier based on liquid crystal nanoparticles for ophthalmic delivery ofpilocarpine nitrate. Int J Pharm 455:75–84.

Li N, Zhuang C, Wang M, Sun X, Nie S, Pan W. 2009. Liposome coatedwith low molecular weight chitosan and its potential use in ocular drugdelivery. Int J Pharm 379:131–8.

Liang H, Baudouin C, Duall P, Garrigue JS, Brignole-Baudouin F. 2012.Ocular safety of cationic emulsion of cyclosporine in an in vitrocorneal wound healing model and an acute in vivo rabbit model. MolVis 18:2195–204.

Lin M, Zou R, Shi H, Yu S, Li X, Guo R, et al 2015. Ocularbiocompatibility evaluation of hydroxyl-functionalized graphene.Mater Sci Eng C Mater Biol Appl 50:300–8.

Liu K, Wang Y, Li H, Duan Y. 2015. A facile one-pot synthesis of starchfunctionalized graphene as nano-carrier for pH-sensitive and starch-mediated drug delivery. Colloids Surf B Biointerfaces 128:86–93.

Lopes JF, Moster MR, Wilson RP, Altangerel U, Alvim HS, Tong MG,et al 2006. Subconjunctival sodium hyaluronate 2.3% in trabeculect-omy: a prospective randomized clinical trial. Ophthalmology 113:756–60.

Lu Y, GuoXing L, Shu Z, Fei S, XiaoJie H, Qian Z, et al 2013.Cytotoxicity and genotoxicity of multi-walled carbon nanotubes withhuman ocular cells. Mater Chem 58:2347–53.

Ludwig A. 2005. The use of mucoadhesive polymers in ocular drugdelivery. Adv Drug Deliv Rev 57:1595–639.

Manna S, Banerjee RK, Augsburger JJ, Al-Rjoub MF, Donnell A, CorreaZM. 2015. Biodegradable chitosan and polylactic acid-based intrao-cular micro-implant for sustained release of methotrexate into vitreous:

DOI: 10.3109/17435390.2016.1153165 Please check 23

Dow

nloa

ded

by [

Tex

as A

&M

Uni

vers

ity L

ibra

ries

] at

06:

58 3

1 M

arch

201

6

Page 25: 48 Safety and Toxicity of Nanomaterials for ocular drug delivery applications

analysis of pharmacokinetic and toxicity in rabbit eyes. Graefes ArchClin Exp Ophthalmol 253:1297–305.

Mao HY, Laurent S, Chen W, Akhavan O, Imani M, Ashkarran AA,Mahmoudi M. 2013. Graphene: promises, facts, opportunities andchallenges in nanomedicine. Chem Rev 113:3407–24.

Marianecci C, Marzio LD, Rinaldi F, Celia C, Paolino D, Alhaique F,et al 2014. Niosomes from 80s to present: the state of the art. AdvColloid Interface Sci 205:187–206.

Majdi JA, Qian H, Li Y, Langsner RJ, Shea KI, Agrawal A, et al 2015.The use of time-lapse optical coherence tomography to image theeffects of microapplied toxins on the retina. Invest Ophthalmol Vis Sci56:587–97.

Mehra NK, Jain K, Jain NK. 2015a. Pharmaceutical and biomedicalapplications of surface engineered carbon nanotubes. Drug DiscovToday 20:750–6.

Mehra NK, Jain K, Jain NK. 2015b. Design of multifunctionalnanocarriers for delivery of anti-cancer therapy. Curr Pharm Des 21:6157–64.

Mehra NK, Jain NK. 2015a. Multifunctional hybrid-carbon nanotubes:new horizon in drug delivery and targeting. J Drug Target. doi:10.3109/1061186X.2015.1055571.

Mehra NK, Jain NK. 2015b. Cancer targeting propensity of folateconjugated surface engineered multi-walled carbon nanotubes.Colloids Surf B Biointerfaces 132:17–26.

Mehra NK, Jain NK. 2015c. One platform comparison of estrone and folicacid anchored surface engineered MWCNTs for doxorubicin delivery.Mol Pharm 12:630–43.

Mehra NK, Mishra V, Jain NK. 2014a. A review of ligand tetheredsurface engineered carbon nanotubes. Biomaterials 35:1267–83.

Mehra NK, Palakurthi S. 2015. Interactions between carbon nanotubesand bioactives: a drug delivery perspective. Drug Discov Today. doi:10.1016/j.drudis.2015.11.011.

Mehra NK, Verma AK, Mishra PR, Jain NK. 2014b. The cancer targetingpotential of D-a-tocopheryl polyethylene glycol 1000 succinatetethered multi walled carbon nanotubes. Biomaterials 35:4573–88.

Mishra V, Jain NK. 2014. Acetazolamide encapsulated dendritic nano-architectures for effective glaucoma management in rabbits. Int JPharm 461:380–90.

Miyawaki J, Yudasaka M, Azami T, Kubo Y, Iijima S. 2008. Toxicity ofsingle-walled carbon nanohorns. ACS Nano 2:213–26.

Mody N, Tekade RK, Mehra NK, Chopdey P, Jain NK. 2014. Dendrimer,liposomes, carbon nanotubes and PLGA nanoparticles: one platformassessment of drug delivery potential. AAPS PharmSciTech 15:388–99.

Moysidis SN, Alvarez-Delfin K, Peschansky VJ, Salero E, Weisman AD,Bartakova A, et al 2015. Magnetic field-guided cell delivery withnanoparticle-loaded human corneal endothelial cells. Nanomedicine11:499–509.

Myles ME, Neumann DM, Hill JM. 2005. Recent progress in ocular drugdelivery for posterior segment disease: emphasis on transscleraliontophoresis. Adv Drug Deliv Rev 57:2063–79.

Nagai N, Ito Y. 2014. Therapeutic effects of gel ointments containingtranilast nanoparticles on paw edema in adjuvant-induced arthritis rats.Biol Pharm Bull 37:96–104.

Nagai N, Ito Y, Okamoto N, Shimomura Y. 2014. A nanoparticleformulation reduces the corneal toxicity of indomethacin eye drops andenhances its corneal permeability. Toxicology 319:53–62.

Nair KL, Vidyanand S, James J, Kumar GS. 2012. Pilocarpine-loadedpoly (dl-lactic-co-glycolic acid) nanoparticles as potential candidatesfor controlled drug delivery with enhanced ocular pharmacologicalresponse. J Appl Polym Sci 124:2030–6.

OECD (Organisation for Economic Co-operation and Development).2002. OECD guideline for the testing of chemicals: acute eye irritation/corrosion (Test No. 405).

Oshima Y, Sakamoto T, Nakamura T, Tahara Y, Goto Y, Ishibashi T,Inomata H. 1999. The comparative benefits of glaucoma filteringsurgery with an electric-pulse targeted drug delivery system demon-strated in an animal model. Ophthalmology 106:1140–6.

Pathak MK, Chhabra G, Pathak K. 2013. Design and development of anovel pH-triggered nanoemulsified in-situ ophthalmic gel of flucon-azole: ex vivo transcorneal permeation, corneal toxicity and irritationtesting. Drug Dev Ind Pharm 39:780–90.

Pattni BS, Chupin VV, Torchilin VP. 2015. New developments inliposomal drug delivery. Chem Rev 115:10938–66.

Peng D, Yu K, Zeng S, Li Y, Lin J, Wu Y, Zhou W. 1999. Anexperimental study on homoharringtonine liposome and glaucomafiltration surgery. Yan Ke Xue Bao 15:51–4.

Pignatello R, Bucolo C, Ferrara P, Maltese A, Puleo A, Puglisi G. 2002.Eudragit RS100 nanosuspensions for the ophthalmic controlleddelivery of ibuprofen

�. Eur J Pharm Sci 16:53–61.

Plange N, Remky A, Arend O. 2003. Colour Doppler imaging andfluorescein filling defects of the optic disc in normal tension glaucoma.Br J Ophthalmol 87:731–6.

Prausnitz MR, Noonan JS. 1998. Permeability of cornea, sclera, andconjunctiva: a literature analysis for drug delivery to the eye. J PharmSci 87:1479–88.

Prow TW. 2010. Toxicity of nanomaterials to the eye. Wiley InterdiscipRev Nanomed Nanobiotechnol 2:317–33.

Prow TW, Bhutto I, Kim SY, Grebe R, Merges C, McLeod S, et al 2008.Ocular nanoparticle toxicity and transfection of the retina and retinalpigment epithelium. Nanomedicine 4:340–9.

Qiao Y, Qin G, Yu L. 2013. The triblock copolymers hydrogel throughintracameral injection may be a new potential ophthalmic drugdelivery with antiscarring drugs after glaucoma filtration surgery.Med Hypotheses 80:23–5.

Rai P, Lauande-Pimentel R, Barton K. 2005. Amniotic membrane as anadjunct to donor sclera in the repair of exposed glaucoma drainagedevices. Am J Ophthalmol 140:1148–52.

Roggeband R, York M, Pericoi M, Braun W. 2000. Eye irritationresponses in rabbit and man after single application of equal volume ofundiluted model liquid detergent products. Food Chem Toxicol 38:727–34.

Rajala A, Wang Y, Zhu Y, Ranjo-Bishop M, Ma JX, Mao C, Rajala RV.2014. Nanoparticle-assisted targeted delivery of eye-specific genes toeyes significantly improves the vision of blind mice in vivo. Nano Lett14:5257–63.

Ratto F, Matteini P, Rossi F, Menabuoni L, Tiwari N, Kulkarni SK, PiniR. 2009. Photothermal effects in connective tissues mediated by laser-activated gold nanorods. Nanomedicine 5:143–51.

Sasaki H, Karasawa K, Hironaka K, Tahara K, Tozuka Y, Takeuchi H.2013. Retinal drug delivery using eyedrop preparations of poly-l-lysine-modified liposomes. Eur J Pharm Biopharm 83:364–9.

Sharma A, Tandon A, Tovey JCK, Gupta R, Robertson JD, Fortune JA,et al 2011. Polyethylenimine-conjugated gold nanoparticles: genetransfer potential and low toxicity in the cornea. Nanomedicine 7:505–13.

Sharma P, Mehra NK, Jain K, Jain NK. 2015. Biomedical applications ofcarbon nanotubes: a critical review. Curr Drug Deliv 12 (In press).

Sharma R, Ahuja M, Kaur H. 2012. Thiolated pectin nanoparticles:preparation, characterization and ex vivo corneal permeation study.Carbohydr Polym 87:1606–10.

Sheikholeshlami P, Muirhead B, Baek DSH, Wang H, Zhao X,Sivakumaran D, et al 2015. Hydrophobically-modified poly(vinylpyrrolidone) as a physically-associative, shear-responsive ophthalmichydrogel. Exp Eye Res 137:18–31.

Soderstjerna E, Bauer P, Cedervall T, Abdshill H, Johansson F, JohanssonUE. 2014. Silver and gold nanoparticles exposure to in vitro culturedretina-studies on nanoparticle internalization, apoptosis, oxidativestress, glial- and microglial activity. PLoS One 9:e105359.

Sosnik A, Neves J, Sarmento B. 2014. Mucoadhesive polymers in thedesign of nano-drug delivery systems for administration by non-parenteral routes: a review. Prog Polym Sci 39:2030–75.

Souza JG, Dias K, Silva SAM, de Rezende LCD, Rocha EM, Emery FS,Lopez RFV. 2015. Transcorneal iontophoresis of dendrimers: PAMAMcorneal penetration and dexamethasone delivery. J Control Release200:115–24.

Spataro G, Malecaze F, Turrin CO, Soler V, Duhayon C, Elena PP, et al2010. Designing dendrimers for ocular drug delivery. Eur J Med Chem45:326–34.

Spitzer MS, Yoeruek E, Kaczmarek RT, Sierra A, Aisenbrey S, GrisantiS, et al 2008. Sodium hyaluronate gels as a drug release system forcorticosteroids: release kinetics and anti-proliferative potential forglaucoma surgery. Acta Ophthalmol 86:842–8.

Srivastava V, Gusain D, Sharma YC. 2015. Critical review on the toxicityof some widely used engineered nanoparticles. Ind Eng Chem Res 54:6209–33.

Taha EI, Ei-Anazi MH, El-Bagory IM, Bayomi MA. 2014. Design ofliposomal colloidal systems for ocular delivery of ciprofloxacin. SaudiPharm J 22:231–9.

24 N.K. Mehra et al. Nanotoxicology, Early Online: 1–25

Dow

nloa

ded

by [

Tex

as A

&M

Uni

vers

ity L

ibra

ries

] at

06:

58 3

1 M

arch

201

6

Page 26: 48 Safety and Toxicity of Nanomaterials for ocular drug delivery applications

Tayel SA, El-ANabarawi MA, Tadros MI, Abd-Elsalam WH. 2013.Promising ion-sensitive in situ ocular nanoemulsion gels of terbinafinehydrochloride: design, in vitro characterization and in vivo estimationof the ocular irritation and drug pharmacokinetics in the aqueoushumor of rabbits. Int J Pharm 443:293–305.

Tommaso CD, Torriglia A, Furrer P, Behar-Cohen F, Gurny R, Moller M.2011. Ocular biocompatibility of novel cyclosporine A formulationsbased on methoxypoly(ethylene glycol)-hexylsubstituted poly(lactide)micelle carriers. Int J Pharm 416:515–24.

Tripathi RC, Tfipathi BJ. 2005. Tissue plasminogen activator therapy forthe eye. Br J Ophthalmol 89:1390–1.

Tuomela A, Liu P, Puranen J, Ronkko S, Laaksonen T, Kalesnykas G,et al 2014. Brinzolamide nanocrystal formulations for ophthalmicdelivery: reduction of elevated intraocular pressure in vivo. Int J Pharm467:34–41.

Vandamme TF, Brobeck L. 2005. Poly(amidoamine) dendrimers asophthalmic vehicles for ocular delivery of pilocarpine nitrate andtropicamide. J Control Release 102:23–38.

Varshocian R, Jeddi-Tehrani M, Mahmoudi AR, Khoshayand MR, AtyabiF, Sabzevari A, et al 2013. The protective effect of albumin onbevacizumab activity and stability in PLGA nanoparticles intended forretinal and choroidal neovascularization treatments. Eur J Pharm Sci50:341–52.

van Velthoven ME, Faber DJ, Verbraak FD, van Leeuwen TG, de SmetMD. 2007. Recent developments in optical coherence tomography forimaging the retina. Prog Retin Eye Res 26:57–77.

Volkov Y. 2015. Quantum dots in nanomedicine: recent trends, advancesand unresolved issues. Biochem Biophys Res Commun. doi: 10.1016/j.bbrc.2015.07.039.

Wong TT, Khaw PT, Aung T, Foster PJ, Htoon HM, Oen FT, et al 2009.The Singapore 5-fluorouracil trabeculectomy study: effects onintraocular pressure control and disease progression at 3 years.Ophthalmology 116:175–84.

Wang W, Zhang J, Huang Y, Huang J, Jiang J. 2011. Clinical study oninterferon treatment of early scarring in filtering bleb. Eye Sci 26:197–200.

Warsi MH, Anwar M, Garg V, Jain GK, Talegaonkar S, Ahmad FJ, KharRK. 2014. Dorzolamide-loaded PLGA/vitamin E TPGS nanoparticlesfor glaucoma therapy: pharmacoscintigraphy study and evaluation ofextended ocular hypotensive effect in rabbits. Colloids Surf BBiointerfaces 122:423–31.

Wilson SL, Ahearne M, Hopkinson A. 2015. An overview of currenttechniques for ocular toxicity testing. Toxicology 327:32–46.

Wu YT, Willcox M, Zhu H, Stapleton F. 2015. Contact lens hygienecompliance and lens case contamination: a review. Cont Lens AnteriorEye 38:307–16.

Yamaguchi M, Yasueda S, Isowaki A, Yamamoto M, Kimura M, Inada K,Ohtori A. 2005. Formulation of an ophthalmic lipid emulsioncontaining an anti-inflammatory steroidal drug, difluprednate. Int JPharm 301:121–8.

Yan L, Wang Y, Xu X, Zeng C, Hou J, Lin M, et al 2012. Can grapheneoxide cause damage to eyesight?. Chem Res Toxicol 25:1265–70.

Yang H, Tyagi P, Kadam RS, Holden CA, Kompella UB. 2012. Hybriddendrimers hydrogel/PLGA nanoparticles platform sustains drugdelivery for one week and anti-glaucoma effects for four daysfollowing one-time topical administration. ACS Nano 6:7596–606.

Yang L, Wang F, Han H, Yang L, Zhang G, Fan Z. 2015. Functionalizedgraphene oxide as a drug carrier for loading pirfenidone in treatment ofsubarachnoid hemorrhage. Colloids Surf B Biointerfaces 129:21–9.

Yellepeddi VK, Vangara KK, Palakurthi S. 2013. Poly (amido) amine(PAMAM) dendrimer-cisplatin complexes for chemotherapy of cis-platin-resistant ovarian cancer cells. J Nanopart Res 15:1897.

Yin H, Gong C, Shi S, Liu X, Wei Y, Qian Z. 2010. Toxicity evaluation ofbiodegradable and thermosensitive PEG-PCL-PEG hydrogel as apotential in situ sustained ophthalmic drug delivery system. J BiomedMater Res Part B: Appl Biomater 92:129–37.

Yuan X, Marcano DC, Shin CS, Hua X, Isenhart LC, Pflugfelder SC,Acharya G. 2015. Ocular drug delivery nanowafer with enhancedtherapeutic efficacy. ACS Nano 9:1749–58.

Zhang J, Wang S. 2009. Topical use of coenzyme Q10-loaded liposomescoated with trimethyl chitosan: tolerances, precorneal retention andanti-cataract effect. Int J Pharm 372:66–75.

DOI: 10.3109/17435390.2016.1153165 Please check 25

Dow

nloa

ded

by [

Tex

as A

&M

Uni

vers

ity L

ibra

ries

] at

06:

58 3

1 M

arch

201

6