ArticlePDF AvailableLiterature Review

How are we improving the delivery to back of the eye? Advances and challenges of novel therapeutic approaches

Authors:

Abstract and Figures

Introduction: Drug delivery to the back of the eye requires strategic approaches that guarantee the long-term therapeutic effect with patient compliance. Current treatments for posterior eye diseases suffer from significant challenges including frequent intraocular injections of anti-VEGF agents and related adverse effects in addition to the high cost of the therapy. Areas covered: Treatment challenges and promising drug delivery approaches for posterior segment eye diseases, such as age-related macular degeneration (AMD) are summarized. Advances in the development of several nanotechnology-based systems, including stimuli-responsive approaches to enhance drug bioavailability and overcome existing barriers for effective ocular delivery are discussed. Stem cell transplantation and encapsulated cell technology (ECT) approaches to treat posterior eye diseases are elaborated. Expert opinion: There are several drug delivery systems demonstrating promising results. However, a better understanding of ocular barriers, disease pathophysiology, and drug clearance mechanisms is required for better therapeutic outcomes. The stem cell transplantation strategy and ECT approach provide positive results in AMD therapy, but there are a number of challenges that must be overcome for long-term efficiency. Ultimately, there are numerous multidimensional challenges to cure vision problems and a collaborative approach among scientists is required.
Content may be subject to copyright.
Full Terms & Conditions of access and use can be found at
http://www.tandfonline.com/action/journalInformation?journalCode=iedd20
Download by: [UMKC University of Missouri Kansas City] Date: 29 September 2017, At: 07:50
Expert Opinion on Drug Delivery
ISSN: 1742-5247 (Print) 1744-7593 (Online) Journal homepage: http://www.tandfonline.com/loi/iedd20
How are we improving the delivery to back of the
eye? Advances and challenges of novel therapeutic
approaches
Vibhuti Agrahari, Vivek Agrahari, Abhirup Mandal, Dhananjay Pal & Ashim K.
Mitra
To cite this article: Vibhuti Agrahari, Vivek Agrahari, Abhirup Mandal, Dhananjay Pal & Ashim
K. Mitra (2017) How are we improving the delivery to back of the eye? Advances and challenges
of novel therapeutic approaches, Expert Opinion on Drug Delivery, 14:10, 1145-1162, DOI:
10.1080/17425247.2017.1272569
To link to this article: http://dx.doi.org/10.1080/17425247.2017.1272569
Accepted author version posted online: 14
Dec 2016.
Published online: 28 Dec 2016.
Submit your article to this journal
Article views: 197
View related articles
View Crossmark data
Citing articles: 5 View citing articles
REVIEW
How are we improving the delivery to back of the eye? Advances and challenges of
novel therapeutic approaches
Vibhuti Agrahari*, Vivek Agrahari*, Abhirup Mandal, Dhananjay Pal and Ashim K. Mitra
School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA
ABSTRACT
Introduction: Drug delivery to the back of the eye requires strategic approaches that guarantee the
long-term therapeutic effect with patient compliance. Current treatments for posterior eye diseases
suffer from significant challenges including frequent intraocular injections of anti-VEGF agents and
related adverse effects in addition to the high cost of the therapy.
Areas covered: Treatment challenges and promising drug delivery approaches for posterior segment
eye diseases, such as age-related macular degeneration (AMD) are summarized. Advances in the
development of several nanotechnology-based systems, including stimuli-responsive approaches to
enhance drug bioavailability and overcome existing barriers for effective ocular delivery are discussed.
Stem cell transplantation and encapsulated cell technology (ECT) approaches to treat posterior eye
diseases are elaborated.
Expert opinion: There are several drug delivery systems demonstrating promising results. However, a
better understanding of ocular barriers, disease pathophysiology, and drug clearance mechanisms is
required for better therapeutic outcomes. The stem cell transplantation strategy and ECT approach
provide positive results in AMD therapy, but there are a number of challenges that must be overcome
for long-term efficiency. Ultimately, there are numerous multidimensional challenges to cure vision
problems and a collaborative approach among scientists is required.
ARTICLE HISTORY
Received 1 October 2016
Accepted 12 December 2016
KEYWORDS
Ocular barriers; posterior eye
diseases; age-related
macular degeneration; drug
delivery; stimuli-responsive;
stem cell transplantation;
encapsulated cell
technology; nanocarriers;
nanotechnology
1. Introduction
Vision loss poses a major problem and it is estimated that
about 285 million people worldwide are visually impaired and
39 million are blind (http://www.who.int/mediacentre/fact
sheets/fs282/en/). The number of blind individuals increases
by approximately 7 million/year (http://www.who.int/blind
ness/Vision2020_report.pdf). In the United States, about 3.4
million individuals over the age of 40 are blind or living with
significant visual impairment [1]. The major diseases that sig-
nificantly impact vision are macular degeneration, diabetic
retinopathy, diabetic macular edema, cataract, uveitis, kerati-
tis, and glaucoma [2]. Current treatments for posterior eye
diseases suffer from significant challenges including frequent
intraocular injections, related adverse effects, and high cost of
the treatment [3]. Because of several anatomical/physiological
barriers present in the eye, drug delivery to the posterior
ocular segment is significantly impaired [4,5]. However, the
application of nanotechnology combined with various routes
of administration has been shown improvement in ocular drug
delivery. Nanotechnology offers several treatment options to
reduce the gap of limitations [6].
Considering the aforementioned facts, this review aims to
emphasize major challenges and promising drug delivery solu-
tions for the treatment of posterior eye diseases such as age-
related macular degeneration (AMD). An overview of treat-
ment strategies for AMD, highlighting the progress and
limitations in the current therapeutic approaches are
described. Various routes of drug administration to the back
of the eye and nanoformulations for the posterior eye diseases
are discussed. Further, retinal cell transplantation using stem
cells, and encapsulated cell technology (ECT) approaches are
elaborated.
1.1. Barriers and various routes of drug delivery to the
posterior eye
The interior of the human eye is subdivided into anterior and
posterior segments. Drug bioavailability in the eye is limited
due to several anatomical/static (conjunctiva, cornea, sclera,
blood aqueous and retinal barriers) and physiological/dynamic
(choroid blood flow, efflux transporters, tear washing, nasola-
crimal drainage) barriers [7]. These barriers effectively limit the
drug access to the back of the eye [8]. The usual routes for
posterior eye drug delivery are topical [9], systemic, intraocular
(suprachoroidal, intravitreal), and periocular (subconjunctival,
subtenon, retrobulbar) [10]. These routes are schematically
represented by Figure 1 and summarized below.
Topical drug administration provides high patient compli-
ance and lower side effects. Corneal and conjunctival routes
are two local pathways for drug entry from a topical adminis-
tration (Figure 2). However, topical route remains challenging
due to several constrains such as blurring of vision, precorneal
CONTACT Ashim K. Mitra mitraa@umkc.edu
*These authors contributed equally to this work
EXPERT OPINION ON DRUG DELIVERY, 2017
VOL. 14, NO. 10, 11451162
https://doi.org/10.1080/17425247.2017.1272569
© 2016 Informa UK Limited, trading as Taylor & Francis Group
Downloaded by [UMKC University of Missouri Kansas City] at 07:50 29 September 2017
drug losses, nasolacrimal drainage, and limited volume of drug
administration (~30μl). In addition, normal intraocular fluid
flow gradient is from vitreous to aqueous compartment
which further limits the passage of topically applied drugs to
the back of the eye [9]. These all limit the drug absorption and
only ~5% of the drug dose reaching the interior of the eye,
mostly entering the systemic circulation [7,11].
The drug absorption through intravenous route is con-
trolled by the presence of bloodretinal barrier (BRB), thus,
requires high drug doses to achieve adequate therapeutic
levels with the risk of several adverse effects. In addition, efflux
transporters expressed on apical and basolateral cell mem-
branes of human retinal pigment epithelium (RPE) stand
against drug permeation from choroid to retina after intrave-
nous administration [12].
Intravitreal drug delivery is one of the most efficient routes
for posterior segment due to its proximity to the retina, chor-
oid, and RPE. Though invasive, this route has the potential to
provide highest intraocular bioavailability by circumventing
several barriers of the posterior eye segment [13]. However,
intravitreal administration is associated with serious risks such
as hemorrhage, persistent discomfort, retinal detachment,
degeneration of photoreceptors (PRs), increase in intraocular
pressure (IOP), cataract formation, and bacterial endophthal-
mitis [14,15].
Periocular route involve injecting the drug outside the
globe of the eye and in the proximity of the sclera. This
route utilizes the trans-scleral pathway to deliver drugs next
to the choroid provides a significant barrier of macromole-
cules drug permeation) [16]. However, drug losses via con-
junctival, episcleral blood, and lymphatic flow are limiting
factors to posterior eye delivery of small molecules following
periocular administration [10].
Suprachoroidal injections may be the most appropriate
route to reach the choroid and vitreous humor [17]. The
suprachoroidal space lies internal to the sclera and provides
a natural route for drugs injected across the sclera along the
inner surface of the eye into the posterior segment. This route
has recently been utilized to inject bevacizumab for the treat-
ment of AMD [18]. However, postoperative inflammation and
choroid hemorrhage remain a concern.
Article highlights
Drug delivery to the eye is challenging. Conventional formulations
are unable to efficiently deliver a drug into the back of the eye due to
the presence of complex barriers, elimination mechanisms, thus,
resulting in a low ocular drug bioavailability.
Various routes of drug administration to the back of the eye, elim-
ination pathways, delivery barriers, advantages and challenges are
elaborated.
At present, frequent intravitreal injections are the preferred method
for the treatment of posterior eye diseases but have several side
effects and high treatment costs.
The current treatment options and major challenges for AMD and
future therapeutics have been described.
Several nanosystems in addition to their clinical applications for the
back of eye diseases emphasizing on AMD are discussed.
Furthermore, the current status and clinical applications of ocular
implants are reviewed. Stimuli-responsive NCs for back of the eye
diseases are also summarized.
Retinal cell transplantation using stem cells sources and ECT
approaches; their challenges with future implications for the retinal
diseases are elaborated.
This box summarizes key points contained in the article
Figure 1. Administration routes (topical, intravenous, intravitreal, periocular, and suprachoroidal) for delivering therapeutics to the posterior of the eye.
1146 V. AGRAHARI ET AL.
Downloaded by [UMKC University of Missouri Kansas City] at 07:50 29 September 2017
2. Posterior segment eye diseases and age-related
macular degeneration (AMD)
Normal aging of the posterior eye segment is characterized by
a continuous loss of PRs, Bruchs membrane (BM) thickening,
choroid thinning, scleral stiffening, vitreous degradation, and
accumulation of debris. Among the posterior segment eye
diseases, diabetic retinopathy, glaucoma, and macular degen-
eration are the leading causes of age-related vision loss, and
represents 14% of the total causes of blindness globally
[19,20]. Macular degeneration occurs when a small area in
the retina (macula) deteriorates, and it develops as person
ages, hence referred to as AMD which is a degenerative dis-
ease that damages the RPE and PRs. It is projected that the
number of people with AMD will be at about 196 million in
2020, and 288 million in 2040 [21]. About 1.75 million
Americans are affected by AMD, and this number is expected
to increase about 3 million by 2020 [21].
In earlier stages of AMD, BM slows down the transport of
metabolites resulting in druses formation. Druses are small,
yellowish extracellular subretinal deposits of lipid, cellular
debris and protein, including complement components and
categorized as small (<63 μm), medium (63124 μm), or large
(>124 μm) in diameter [22]. In early stages of AMD, druses are
small and semitransparent. However, excess druses can lead to
the RPE damage, and a chronic inflammatory response leads
to geographic atrophy (GA) and angiogenesis. The extent of
druses accumulation under the RPE and the amount of RPE
pigmentation changes (hypo or hyper) are clinical hallmark of
AMD progress [23].
As the disease progresses, AMD can be classified into the
dry and wet forms. Based on the absence or presence of
vascular growth progressing from the choroidal side toward
the retina, it is broadly subdivided into nonneovascular (NNV)
and neovascular (NV) AMD (also called as wet-AMD). In wet-
AMD, new blood vessels from the choroid may leak, resulting
in macular edema and hemorrhage. The risk of getting
advanced AMD increases from 2% at ages 5059 years, to
nearly 30% for those over the age of 75 years [24]. Multiple
factors such as oxidative stress, lipid metabolism, immune
system activation, and angiogenesis play a key role in AMD
pathogenesis. The protein aggregation and immunologic pro-
cesses are also involved, including the inflammatory molecules
in BM, recruitment of macrophages, dendritic cells, and com-
plement system components in the macula area. In addition,
smoking is a major oxidative stress factor in AMD progression
[25]. The comprehensive effects of these factors can be
explored elsewhere [26,27].
2.1. Current treatments for AMD
Over the past decade, significant progress has been made in
the treatment of AMD owing to an increased understanding of
the mechanisms of ocular angiogenesis [28]. Several factors
are involved in ocular angiogenesis, with vascular endothelial
growth factor (VEGF) playing a central role [29]. VEGF-A is a 46
kDa glycoprotein produced by ocular cells in response to
oxidative stress. VEGF-A is the most potent mediator of both
retinal and choroidal angiogenesis. It stimulates endothelial
cell growth, promotes vascular permeability and induces dis-
sociation of tight junction components. Currently, no therapy
exists for dry-AMD and only dietary modifications such as
increase in intake of antioxidants, cessation of smoking, and
control of blood pressure appears to slowdown disease pro-
gression. The only approved treatment for dry-AMD is Age-
Related Eye Disease Study (AREDS) recommended vitamin
Figure 2. Schematic representation of disposition of drug or nanocarriers (NCs) in the eye following ocular administration.
EXPERT OPINION ON DRUG DELIVERY 1147
Downloaded by [UMKC University of Missouri Kansas City] at 07:50 29 September 2017
supplements that lower the risk of developing advanced
stages of AMD [30].
In wet-AMD, a high level of VEGF has been reported to be
present below the RPE cell layer and around PRs. VEGF inhibi-
tion via intraocular anti-VEGF treatments to prevent the for-
mation of new blood vessels represents the cornerstone of
wet-AMD therapies. However, the primary atrophic compo-
nent of AMD still progresses despite anti-VEGF therapy [25].
Recombinant humanized anti-VEGF antibody fragments or
soluble receptor decoys (e.g. ranibizumab: Lucentis®;
Genentech/Roche), bevacizumab (off label drug: Avastin®;
Genentech/Roche), pegaptanib (Macugen®), and aflibercept
(Eylea®; Regeneron Pharmaceuticals) are currently Food and
Drug Administration (FDA) approved therapies for wet-AMD.
The physicochemical and pharmacokinetics parameters of
anti-VEGF therapeutics are summarized in Table 1. Another
available treatment for wet-AMD includes photodynamic ther-
apy (PDT). Studies suggested that a combination of PDT with
angiogenic inhibitors may reduce the frequency of intravitreal
injections thereby lowering the risks associated with long-term
intravitreal therapy [31]. Although PDT approach showed pro-
mising potential, more efforts are needed to develop an effi-
cient delivery system in ocular applications.
2.2. Challenges with current intravitreal anti-VEGF
treatments
Despite the facts that effective, intravitreal anti-VEGF therapies
for AMD have several drawbacks as summarized below
[3,15,38,39].
Intravitreal injections are associated with multiple
adverse effects including retinal detachment, vitreous
hemorrhage, intraocular inflammation, tachyphylaxis,
retinal vascular occlusion, cataract, and endophthalmitis.
Moreover, intravitreal injections require a high degree of
sterility to prevent infection to nonspecific ocular tissues
and cells. Furthermore, an elevation in IOP is associated
with intravitreal injections [15].
VEGF plays a protective role in the retinal tissue. The
knockout of VEGF in retina can lead to severe side effects
such as defects in the RPE choroid complex or the loss of
interaction between RPE and PRs outer segments.
A prolonged suppression of the plasma VEGF due to
anti-VEGF therapy may lead to an increased risk of car-
diovascular diseases.
The low intraocular retention of anti-VEGF drugs is a
major issue due to the high clearance rate from the
posterior section of the eye, thus requiring frequent
administration.
An effective and safe anti-VEGF therapy should not
damage normal blood vessels and preserve the physio-
logic functions of the retinal neurons and other cells.
However, this is not the case with current anti-VEGF
therapy for posterior ocular diseases.
Current anti-VEGF therapies do not address the under-
lying cause of AMD development.
Patients suffering from recurrent NV-AMD may develop
resistance to anti-VEGF therapy, which can result in a
diminished therapeutic effect [32].
The long-term anti-VEGF treatment carries a high cost
(Table 1).
To improve the safety, cost-effectiveness and impact on
patients receiving intravitreal anti-VEGF treatments, a reduc-
tion in the frequency of drug administration is required.
Hence, there remains a need for the development of new
agents, and/or novel strategies for sustained and efficient
anti-VEGF therapy. Newer anti-VEGF agents may offer longer
duration, minimize the treatment burden and overall cost of
therapy [3,38]. However, a long-term, continuous therapy with
a low financial burden remains an unmet need in AMD treat-
ment. Modifying the drug physicochemical properties, such as
the size, charge, and lipophilicity may improve the drug effi-
cacy administered through intravitreal, intraocular and supra-
choroidal routes. However, conventional formulations and
chemical modification of the drug only cannot overcome
static and dynamic barriers to ocular delivery and to reduce
the frequency of intraocular injections. In this respect, nano-
technology based approaches especially the sustained release
dosage forms can possibly overcome the current limitations of
anti-VEGF treatments [4,5].
3. Nanotechnology based drug delivery systems for
the posterior eye diseases
Nanocarriers (NCs) are colloidal systems and capable of encap-
sulating small as well as macromolecule, lipophilic and hydro-
philic drugs. Due to their small size and use of biodegradable
materials in the formulation development, NCs offer signifi-
cant advantages in ocular and other drug delivery applications
as explained below [4,5,40,41]. The therapeutic potential of
these carriers in the posterior eye diseases is summarized in
Table 2.
3.1. Liposomes
Liposomes are lipid vesicles in the size range of 0.110 µm.
These systems have been comprehensively used in ocular
therapeutics due to several advantages they provide such as
versatile surface modification chemistry, control in drug
release depending on the number of lipid bilayers and
composition, and potential of providing a stimuli-sensitive
drug release [69,70]. Also, the physiochemical properties
(size, surface charge, functional chemistry) of liposomes
can be modified by mixing different lipids during the for-
mulation development. The beneficial effects of liposomes
in prolonging the residence time of bevacizumab in the
vitreous after intravitreal administration is studied [44].
However, liposomes present certain limitations, such as
low reproducibility, instability of macromolecules during
the formulation process, manufacturing cost and scale up
issues, and variable size distribution [71].
1148 V. AGRAHARI ET AL.
Downloaded by [UMKC University of Missouri Kansas City] at 07:50 29 September 2017
Table 1. Characteristics of anti-VEGF agents in the clinical management of AMD.
Anti-VEGF agents Structure
Biological target
and mechanism K
D
,IC
50
for VEGF Mol wt. Approval Vitreous half-life Intravitreal dose Regimen
Cost
(approx.) Ref.
Pegaptanib
sodium
(Macugen®)
Pegylated RNA
aptamer (28 base)
VEGF-A
165
200 pM 50 kDa FDA (2004),
EMA (2005)
10 days (human) 0.30 mg Every 6 weeks US $995 [3235]
Bevacizumab
(Avastin®)
Recombinant
humanized mAb
lgG1
All isoforms of
VEGF-A
58 pM, 423 pM 149 kDa FDA (2004),
EMA (2005),
CFDA (2010)
6.7 days (human),
4.326.61 days (in
rabbits)
1.25 mg Every 4 weeks US $50 [32,33,35]
Ranibizumab
(Lucentis®)
Recombinant
humanized lgG1-κ
isotype mAb
fragment
All isoforms of
VEGF-A
46 pM, 343 pM 48 kDa FDA (2006),
EMA (2007),
CFDA (2012)
9 days (human), 2.88
2.89 days (in rabbits
0.50 mg Every 4 weeks US $2000 [32,33,35]
Aflibercept
(Eylea®)
Fusion protein:
domain 2 of VEGFR-
1 and domain 3 of
VEGFR-2 fused with
lgG1 Fc
All isoforms of
VEGF-A, VEGF-B,
and PIGF
0.50 pM, 8 pM 115 kDa FDA (2011),
EMA (2012)
7 days 2.0 mg Every 4 weeks for
3 months and
then once every
8 weeks
US $1850 [32,33,35]
Conbercept Fusion protein:
domain 2 of VEGFR-
1 and domains 3 &
4 of VEGFR-2 fused
with lgG1 Fc
All isoforms of
VEGF-A, VEGF-B,
VEGF-C, and
PIGF
0.50 pM, 10 pM 143 kDa CFDA (2013) 4.2 days (in rabbits) 0.50 or 2.0 mg Every 4 weeks - [35]
Brolucizumab Immunoglobulin Fv
fragments; mAb.
Humanized, single-
chain Ab fragment
inhibitor of VEGF-A
Binds all isoforms
of VEGF-A
- 26 kDa Phase III 4.9 days 1.25 mg Every 4 weeks - [3,36]
Abicipar Small molecule:
(DARPins)
Inhibits all
subtypes of
VEGF-A
- 34 kDa Phase II 6 days 1 or 2.0 mg Every 4 weeks - [3,37]
EXPERT OPINION ON DRUG DELIVERY 1149
Downloaded by [UMKC University of Missouri Kansas City] at 07:50 29 September 2017
3.2. Polymeric nanoparticles (NPs) and microparticles
(MPs)
Polymeric NPs and MPs are biodegradable/biocompatible colloi-
dal systems in the size range of 101000 nm [69]and11000 µm
[72], respectively. In addition to the advantages similar to lipo-
somes, NPs/MPs provide flexibility in routes of administration,
stimuli-responsive system, encapsulation of multiple drugs in a
single particle, and flexibility in modifying size, shape, and surface
functionality in ocular drug delivery [1,5]. However the drawbacks
are, burst drug release may lead to drug toxicity, NPs/MPs rapid
phagocytic clearance, formulations scale-up issues, particle aggre-
gation due to large surface area, and nonuniformity in size dis-
tribution. PLGA based MPs loaded with Chitosan-NPs for the
delivery of ranibizumab have been developed [45]. Results
showed that the delivery system has the potential for improved
intravitreal delivery of therapeutic proteins.
3.3. Nanomicelles
Nanomicelles are self-assembled systems in the size range
of~10100 nm made from biodegradable/biocompatible
amphiphilic block polymers. These vesicles can encapsulate
poorly and highly water soluble drugs in the core and outer
hydrophilic shell, respectively [73]. Nanomicelles offer numer-
ous advantages in ocular drug delivery applications, such as
easy and reproducible formulation, easy sterilization by filtra-
tion, prevention or minimization of drug degradation, possibi-
lities of changing polymer block arrangements as needed, and
improved drug permeation through ocular epithelia with mini-
mal or no irritation, thus, enhanced ocular bioavailability
[5,74]. A micelle formulation of lipid derivatized cidofovir is
developed [56]. The intravitreal injection provided a sustained
drug release in vitreous for chronic retinal diseases. Drawbacks
of nanomicelles include instability in the biological/
Table 2. Nanocarrier (NC) drug delivery systems in posterior eye diseases.
Delivery system and stimulus Therapeutics Polymer Route of administration Target disease Ref.
Liposomes pDNA Polyethylenimine (PEI) Topical AMD [42]
Bevacizumab Annexin A5-associated liposomes Topical Posterior eye diseases [43]
Bevacizumab Phospholipid (egg
phosphatidylcholine or 1,2-
dipalmitoyl-sn-glycero-3-
phosphocholine) and cholesterol
Intravitreal Posterior eye diseases [44]
Nanoparticles Ranibizumab PLGA MPs entrapping chitosan NPs Intravitreal AMD [45]
UV light Nintedanib Light-sensitive polymer Intravitreal Macular degeneration and DR [46]
Temperature Triamcinolone
acetonide
PEGylated PLGA NPs incorporated into
a PLGA-PEG-PLGA thermo-
reversible gel
Intravitreal AMD [47]
Bevacizumab PLGA Intravitreal AMD [48]
Ultrasound FITC-BSA Silk fibroin Transscleral Posterior eye diseases [49]
Coumairn-6 Chitosan and poloxamer 407 Topical Posterior eye diseases [50]
Microparticles/microspheres Ranibizumab PLGA Intravitreal AMD [51]
Bevacizumab PLA NPs encapsulated into PLGA MPs Intravitreal Posterior eye diseases [52]
Bevacizumab PLGA Intravitreal Posterior eye diseases [53]
Temperature Ovalbumin PLGA MPs in poly(N-isopropyl
acrylamide)-hydrogel
Intravitreal Posterior eye diseases [54]
Nanomicelles Dexamethasone Polyoxyl 40 stearate and polysorbate
80
Topical Posterior uveitis [55]
Hexadecyloxypropyl-
cidofovir
- Intravitreal Chronic retinal diseases [56]
Dendrimers Dexamethasone Poly (amidoamine) Topical and subconjunctival DR [57]
Light 5-Aminosalicylic acid G2 lysine dendrimers with a
silsesquioxane core
Intraperitoneal Retinal degeneration [58]
Nanowafers Dexamethasone Carboxymethyl cellulose polymer Topical Dry eye [59]
Axitinib PVA, polyvinylpyrrolidone,
(hydroxypropyl) methyl cellulose,
and carboxymethyl cellulose
CNV [60]
Nanocrystals Brinzolamide Different polymer stabilizers Topical Prolonged reduction of IOP [61]
Temperature Forskolin Poloxamer 407 and polycarbophil Topical Glaucoma [62]
Hydrogels Bevacizumab Oxidized alginate and glycol chitosan Intravitreal AMD [63]
UV light Bevacizumab PCL dimethacrylate and hydroxyethyl
methacrylate
Suprachoroidal CNV [64]
Temperature Bevacizumab PEG-poly-(serinolhexamethylene
urethane)
Intravitreal Posterior eye diseases [65]
Temperature Bevacizumab Poly(2-ethyl-2-oxazoline)-b-poly(ε-
caprolactone)-b-poly(2-ethyl-2-
oxazoline)
Intraocular Posterior eye diseases [66]
Bevacizumab PLGA-PEG-PLGA Intravitreal Posterior eye diseases [67]
Composite nanosystems
Temperature IgG-Fab PCL-PLA-PEG-PLA-PCL based NPs
suspended in a thermo-sensitive
gelling copolymer (mPEG-PCL-PLA-
PCL-PEGm)
Intravitreal Posterior eye diseases [14]
Temperature Octreotide, insulin,
lysozyme, IgG-Fab,
IgG, and catalase
Pentablock polymers with different
molecular weight and
arrangements.
Intravitreal Posterior eye diseases [68]
1150 V. AGRAHARI ET AL.
Downloaded by [UMKC University of Missouri Kansas City] at 07:50 29 September 2017
physiological environment, premature drug release, and diffi-
culty in formulation scale-up.
3.4. Dendrimers
Dendrimers are branched polymeric systems in the size range
of 10100 nm. Drugs can be either entrapped in the dendri-
mers network through hydrogen bonds, hydrophobic and
ionic interactions or conjugated through covalent bonds [75].
These systems have terminal end groups of amine/hydroxyl/
carboxyl functionalities which may be utilized to conjugate
targeting ligand and/or therapeutic molecules. Due to their
unique structure, dendrimers exhibit improved physicochem-
ical properties, uniform size distribution, and higher biocom-
patibility in ocular drug delivery. A dendrimer conjugate of 5-
aminosalicylic acid is studied for the treatment of retinal
degeneration [58]. However, the complexity in formulation
process may limit dendrimers applicability.
3.5. Nanowafers
Nanowafers are disk-like or rectangular membrane that con-
tains drug loaded nanoreservoirs. Nanowafers can be readily
applied in the eye and release the drug for a longer duration,
thus improving the overall therapeutic efficacy. During the
course of the drug release, nanowafer slowly dissolves in
aqueous media and fades away. Recently, topically applied
nanowafers with extended drug release and enhanced efficacy
have been developed to treat dry eye diseases [59,60]. Slow
drug release from the nanowafer increases the ocular surface
drug residence time and subsequent absorption into the sur-
rounding tissue. Although, nanowafers are in early develop-
ment, they have the potential to treat posterior eye diseases.
3.6. Nanocrystals
Nanocrystals are particles (size: 101000 nm) stabilized by the
surfactant or polymeric stabilizers. Nanocrystals possess out-
standing features enabling to overcome the solubility pro-
blems of poorly soluble drugs and provide an enhanced
bioavailability, high drug load, minimal side effects, rapid
onset of action, and an overall high efficiency and safety
[76]. Nanocrystals have been explored for the delivery of anti-
glaucoma drugs forskolin [62] and brinzolamide [61]. Thus,
nanocrystals can be designed for the treatment of posterior
eye diseases.
3.7. Hydrogels
Hydrogels have several potential applications in ophthalmol-
ogy [10]. Porous, soft nature and high water content of hydro-
gels are suitable for encapsulation of water soluble drugs
including proteins and peptides. Hydrogels are excellent for
encapsulating biomacromolecules since they are processed at
ambient temperatures and organic solvents are rarely needed
[10,77]. Hydrogels can be designed from natural or synthetic
polymers. Natural polymers provide biocompatibility, and bio-
degradability to hydrogel however, suffers from weak
mechanical strength, high batch-to-batch variability, and
immunogenicity. Synthetic polymers provide tunable mechan-
ical properties and prolonged stability. Hydrogels permit var-
ious mechanisms (diffusion/swelling/chemically controlled
and stimuli-responsive) of drug release. However, high water
content and soft nature of hydrogel may result in a rapid
release of biomolecules from the gel matrix. Moreover, the
rapidly gelling systems may affect the injectability of hydro-
gels [77].
3.8. Composite nanosystems
Composite nanosystems (NPs suspended in a gel matrix) are
made of biodegradable, FDA approved polymers. These systems
are emerging as a versatile platform in ocular drug delivery
applications [14,68]. The suspended NPs in the thermo-respon-
sive gel matrix encounter an additional diffusion barrier which
provides the long-term drug release especially for macromole-
cules. Similarly, it minimizes burst effect, and provides long-term
zero order kinetics. In addition, composite nanosystems provide
stability to macromolecules from enzymatic degradation and
helps in improving the biological half-life. The physicochemical
characteristics of composite nanosystems can be modified by
varying the chemistry, molecular weight, and block arrange-
ments of the polymers used. However, premature drug release
from the NPs and drug accumulation in the gel matrix may cause
burst effect and needs careful evaluation.
3.9. Ocular implants
Intraocular implants are designed to provide localized controlled
drug release over an extended period of time. Implants are placed
intravitreally by making an incision on the posterior globe through
minor surgery [10,78]. Though implantation is invasive, these
devices are frequently used due totheirseveraladvantagessuch
as sustained and local drug release to diseased ocular tissues at
therapeutic levels, reduced side effects and ability to circumvent
BRB [11,79]. Biodegradable ocular implants of polylactic acid (PLA),
polyglycolic acid (PGA), PLGA, and polycaprolactone (PCL) are
gaining attention due to high biocompatibility and sustained
drug release properties [79,80]. Drug release may vary depending
on the surface area, polymer degradation, swelling rates, molecu-
lar weight, and nature of the encapsulated drug [78].
Nonbiodegradable implants using polymers such as poly-
vinyl alcohol (PVA), and ethylene vinyl acetate (EVA), offer
long-lasting near zero order release kinetics [79]. However,
these devices have to be implanted and removed after drug
depletion, which renders the treatment expensive and patient
noncompliance. Moreover, adverse events such as
endophthalmitis, pseudoendophthalmitis, vitreous haze,
hemorrhage, cataract development and retinal detachment
limit their applications. In general, nonbiodegradable poly-
mers are preferred for implant fabrication to release the drug
in a more controlled manner over long-time, and the device
can be easily removed in case of adverse reactions. The ocular
implant systems for the posterior eye diseases are summarized
in Table 3.
EXPERT OPINION ON DRUG DELIVERY 1151
Downloaded by [UMKC University of Missouri Kansas City] at 07:50 29 September 2017
Table 3. Ocular implants for posterior eye diseases.
Implant Status Polymer (biodegradable/nonbiodegradable) Therapeutics Route of administration Target disease Ref.
Vitrasert FDA approval in 1996 PVA and EVA, nonbiodegradable polymers Ganciclovir Intravitreal Cytomegalo virus retinitis [78,81]
Retisert FDA approval in 2005 PVA, nonbiodegradable polymer Fluocinolone acetonide Intravitreal Chronic posterior uveitis [78,81,82]
Ozurdex (formerly
Posurdex®)
FDA approval in 2009 PLGA, biodegradable Dexamethasone Intravitreal ME [78,81,82]
Iluvien (formerly
Medidur®)
FDA approval in 2014 PVA, nonbiodegradable Fluocinolone acetonide Intravitreal DME [78,83].
I-vationPhase 2 (terminated) Poly (methyl methacrylate), EVA, nonbiodegradable Triamcinolone Intravitreal DME [78,81,83]
Brimonidine
implant
Phase 2 Biodegradable Brimonidine Intravitreal Dry-AMD and retinitis pigmentosa [78]
Verisome®(IBI-20089) Phase 2 Benzyl benzoate, biodegradable Various drugs including
proteins, triamcinolone
acetonide (with
ranibizumab)
Intravitreal NV-AMD [78,8284]
Durasert Phase I/II Bioerodible Latanoprost Subconjunctival space Ocular hypertension, glaucoma
Port delivery system Phase 1 Nonbiodegradable Ranibizumab Scleral incision CNV [78,83,84]
Cortiject Phase I completed Injectable emulsion Dexamethasone prodrug Intravitreal DME [78,84]
Abiciparpegol
(anti-VEGF
DARPin)
Phase II completed Anti-VEGF Intravitreal NV-AMD [78,84]
Mini drug pump (MEM
electronically
activated device)
Preclinical Polydimethylsiloxane, nonbiodegradable Variety of drugs via an
actuator based mechanism
Transscleral [78,85,86]
ODTx (laser activated) Preclinical Nonbiodegradable Multiple drug reservoirs Injectable Posterior eye diseases [78]
ENV705Preclinical Biodegradable Bevacizumab/trehalose [78]
Nanoporous film device Preclinical Biodegradable, impermeable Ranibizumab Intravitreal [78]
1152 V. AGRAHARI ET AL.
Downloaded by [UMKC University of Missouri Kansas City] at 07:50 29 September 2017
The route of administration of anti-VEGF based NCs sys-
tems is critical in the targeting of posterior eye diseases. The
route of injection for a given therapeutic depends upon the
nature of the disease and the drug molecule. The nanofor-
mulations administration should also not imbalance the
clear vitreous media with drug precipitates or particle aggre-
gates. Intravitreal injections are effective but, this route is
invasive and associated with serious side effects.
Subconjunctival, subtenon, retrobulbar and transscleral
(periocular) routes present challenges from both dynamic
and static anatomical barriers that pose significant hurdles
in ocular drug delivery. Although, the periocular or trans-
scleral routes avoid the physical barriers posed by the con-
junctiva, they require drugs to traverse several layers of
ocular tissues such as episclera, sclera, choroid, BM, and
RPE, before entering the posterior of the eye. The suprachor-
oidal route can overcome the static and dynamic ocular
barriers and has the advantage of providing higher drug
concentrations in the retina. This allows for the administra-
tion of lower doses and less frequent dosing of drugs. Thus,
suprachoroidal injections may be the most appropriate
option to reach the choroid and vitreous humor for the
treatment of posterior eye diseases [17]. Furthermore,
drugs injected into the suprachoroidal space are less likely
to reach the RPE, interact with the rods and cones of the PR
cells that can trigger the immunologic responses.
Considering the advantages and therapeutic potential of
the suprachoroidal route, a less invasive and efficient drug
delivery approach is required.
Microneedles have been recently introduced as a minimally
invasive means for delivering drug solutions or drug formula-
tion within the target ocular tissues through the suprachoroi-
dal route [87,88]. Microneedles can overcome the ocular
barriers with several potential advantages such as minimizing
pain, lower risk of infection, thus, high patient compliance can
be achieved. These needles help to deposit drug or carrier
system into sclera or into the suprachoroidal space which may
facilitate diffusion of drug into deeper ocular tissues, choroid
and retina. However, a number of parameters such as an
optimum microneedle design, safety, accuracy, reproducibility,
and manufacturing costs issues warrant further investigation.
4. Stimuli-responsive nanosystems for the posterior
eye diseases
Developing a stimuli-responsive NC system is an attractive
area for drug delivery to the posterior segment since vari-
able drug concentrations may be required at different time
points depending on the individuals and the disease state
[78,89]. These novel systems such as NPs, microspheres,
nanofibers, and hydrogels are able to control/trigger the
drug release in a spatial and temporal manner within a
particular site in response to a number of intrinsic (pH,
temperature, enzymes, oxidative stress) and extrinsic (mag-
netic field, light, ultrasound, heat) stimuli [67,9093]. A
schematic of stimuli-responsive delivery approaches to the
posterior eye diseases is shown by Figure 3 and are briefly
discussed below.
4.1. Light-activated systems
The transparent cornea and the lens make the eye an organ of
choice for light activated delivery system [78,94]. These sys-
tems incorporate light sensitive materials responsive to a spe-
cific wavelength. A NP drug delivery depot for on-demand
light-triggered drug release postimplantation has been formu-
lated [46]. These NPs rapidly release encapsulated drugs upon
exposure to 365 nm light. An implant system has been devel-
oped by On Demand Therapeutics (ODTx), containing several
drug reservoirs activated individually by laser light [78]. This
system provides a controlled drug delivery platform in the
treatment of posterior eye diseases. Recently, suprachoroidal
delivery of bevacizumab is performed using a light-activated
in situ gel [64].
4.2. Thermo-responsive systems
Thermosensitive polymers undergo abrupt change in solubi-
lity in response to a small change in temperature [95]. This
transition can cause conformational changes in the polymer
material that triggers drug release. An injectable thermore-
sponsive hydrogel for intravitreal sustained release of bevaci-
zumab has been designed [67]. Results show an initial burst
release followed by a sustained release of bevacizumab from
hydrogel.
4.3. In situ gelling systems
In situ gelling systems have been designed to increase the
precorneal residence time of drugs due to solgel phase
transition in the presence of various stimuli such as pH, tem-
perature, or ionic strength [96]. Several in situ gelling systems
with natural and synthetic polymers have been developed for
ocular applications [97,98]. Hydrogels are mostly preferred for
these systems because of their porous structures and swelling
properties. Although, in situ gelling system allows easy admin-
istration of sustained release materials to the desired site, it is
difficult to provide long-term release of macromolecules at a
therapeutic level. A thermosensitive in situ gelling system with
a high intraocular biocompatibility and extended bevacizu-
mab release has been developed for ocular delivery [66].
4.4. Ultrasound-responsive systems
Recent studies demonstrated the significant potential of ultra-
sound mediated drug release for ocular applications [49,99].
Ultrasound application has shown to enhance the delivery of
dexamethasone sodium phosphate, through the cornea in
vivo [100]. However, the physical effect of ultrasound on the
stability of the NCs is yet to be evaluated in addition to the risk
of sonication side effects, patients compliance and cost issues.
4.5. Conducting polymer based systems
Conducting polymers offer exciting opportunities in stimuli-
responsive implantable devices to treat posterior eye condi-
tions [78]. These polymers are biocompatible, nontoxic, pos-
sess both polymer- and metal-like properties, and offer
EXPERT OPINION ON DRUG DELIVERY 1153
Downloaded by [UMKC University of Missouri Kansas City] at 07:50 29 September 2017
excellent control over drug release through electrochemical
stimulation [101]. An electrically tunable release of dexa-
methasone from polypyrrole films has been designed for the
treatment of posterior eye diseases [102]. Results suggested
the suitability of these films for the development of electro-
responsive implants for posterior eye diseases.
4.6. Micro electro mechanical (MEM) systems
MEM systems consist of one or more drug reservoirs and
actuators which are responsible for pushing the drug out of
the reservoir by mechanical means in response to the stimulus
such as temperature, electrical stimulus, magnetic field, and
osmotic pressure [78]. A MEM based ocular implant has been
reported for phenylephrine delivery [103]. This approach has
been later modified using a mini drug pump (hydrolysis based
actuation) [85] to provide a control over the drug release
in response to the stimulus, exhibiting accurate drug delivery.
4.7. Oxidative-stress responsive systems
Oxidative stress can lead to cellular or molecular damage caused
by reactive oxygen species (ROS), which also occurs in AMD as a
result of an imbalance between the productions of ROS and the
antioxidant defense response [27,104]. Oxidative-responsive
polymers show great potential in the biomedical and drug
delivery applications to trigger the drug release [105].
Although, no known oxidative-stress responsive system has
been developed for ocular delivery, this stimulus can be targeted
in future drug delivery approaches for the posterior eye diseases.
5. Cell transplantation and delivery approaches for
AMD therapy
Cell transplantation aims to achieve the direct replacement of
endogenous cells, and/or the delivery of cells that can secrete
trophic factors to rescue degenerating cells [106]. In general,
AMD starts with the deposit of druses between RPE and BM,
leading to RPE and PRs degeneration and eventually leads to the
loss of vision [25]. RPE performs a variety of functions for proper
visual acuity including the formation of BRB by tight junctions,
transportation of nutrients from blood to PRs, light absorption,
secretion of cytokines/growth factors, and phagocytosis of outer
segments of PRs [25,107]. RPE provides nutrients, omega-3 fatty
acids for building PR outer segment-membranes and glucose for
energy metabolism needed to maintain visual function by outer-
segments of the PRs. The primary functions of BM include
regulating the diffusion of bio-molecules, minerals, antioxidants,
etc. between the choroid and RPE.
In case of dry-AMD, a gradual deterioration of RPE leads to
subsequent PR loss at the macula. The VEGF produced by the
Figure 3. (a) Nanocarriers (NCs) (liposome, nanoparticle, nanowafer, hydrogel, nanocrsytal, composite nanosystem, dendrimer, implant, and microparticle) in ocular
therapeutics. (b) Intrinsic/extrinsic stimuli and stimuli-responsive nanocarriers (NCs) approach in ocular therapeutics. Drug release from NCs can be designed to be
triggered with various stimuli (extrinsic or intrinsic) based on the NCs biomaterial (polymer) characteristics.
1154 V. AGRAHARI ET AL.
Downloaded by [UMKC University of Missouri Kansas City] at 07:50 29 September 2017
basal side of RPE cells is essential to the health of the chor-
iocapillaris. In wet-AMD, RPE produces excessive VEGF, which
contributes to the breakdown of BRB and sprouting of imma-
ture blood vessels from choroid through BM into retina. This
process is called as neovascularization. Leakage of blood from
these abnormal vessels causes an acute loss of vision. As RPE
cells age, the efficiency of recycling of waste products decline
that may lead to a build-up of toxic waste products deposited
beneath the RPE cells on BM [108,109]. Usually, BM doubles in
thickness between the age of 10 and 90 years, slowing the
import of nutrients and export of waste materials from RPE
[25,110]. Together with other BM abnormalities, this may lead
to the dysfunction and ultimately death of RPE cells [109].
The cell based AMD treatment strategies provide support
and replacement to stressed/degenerated RPE to maintain its
function. Although AMD has different origins, in general, ret-
inal diseases can be attributed to the degeneration of the RPE,
RPE cells, PR cells, and BM [25]. Thus, an enticing possibility for
AMD therapy is the replacement of PR cells, either alone or in
addition to RPE transplantation. However, replacing PRs is a
challenging task due to the highly complex organization of
the retina and PRs must integrate well in order to be func-
tional. Thus, the preservation of PRs through the replacement
of the RPE is a promising option and can potentially restore
vision [20,25,107].
5.1. Sources of RPE for cell transplantation in AMD
The replacement of RPE monolayer can be performed by stem
cells. The stem cells can be differentiated into the desired cell
type to replace damaged tissue in order to integrate and
restore function [20,25,111]. There are currently several stem
cells sources used in RPE transplantation as discussed below.
Generally, an ideal cell source should have minimal ethical
concerns, able to expand in in vitro culture easily and has
minimal costs associated [20,25,111]. Several clinical trials are
ongoing to test the safety and efficacy of stem cell transplan-
tation in the eye [106,112,113].
Embryonic stem cells (ESCs) are pluripotent cells from the
inner cell mass of blastocyst. These cells have the ability to
self-renew and differentiate into any cell type such as the PRs,
RPE, and retinal progenitor cells (RPCs) [20,25,111]. However,
the application of ESC is challenging due to ethical concern,
immunological reactions, and the risk of teratoma formation.
Efforts are also needed to explore culturing techniques and
methods for their large-scale production. Moreover, ESCs
express human leukocyte markers that mediate immune
responses, thus making ESC-RPE grafts susceptible to rejection
response by the recipient.
Induced pluripotent stem cells (iPSCs) can be derived from
the patients own tissue thus poses fewer ethical concerns
compared to ESCs. Similar to ESCs, iPSCs can be differentiated
using a combination of soluble factors into various cells of the
retina [20,25,111]. iPSC-RPE cells are shown to have a full
range of morphologic and functional properties of RPE cells
[114,115]. However, grafts of in vitro differentiated RPE from
allogenic iPSCs may evoke immune response with subsequent
rejection in addition to the challenges of the risk of malignant
tumors.
Adult-derived stem cells include mammalian ciliary body,
Müller glia derived cells and nonretinal cells (mesenchymal
stroma cells) [20,25,111]. These cells are multipotent, have no
rejection issues, and can be easily cultured with a low risk of
contamination. But, these cells are relatively difficult to harvest
and have pro-oncogenic potential.
Retinal stem cells (RSCs) such as retinal neurospheres
(RNSs) provide relatively large availability of the RPE cells
[25,107]. RPE differentiation from RNS requires limited cell
expansion in vitro and short culture time relative to ESC or
iPSC. RNS have molecular features of RPCs with a negligible
tumorigenic potential. However, further analysis is required to
fully characterize the RNS-derived cells. RNSs also show a
limited ability to be expanded in vitro.
5.2. Challenges with cell transplantation approaches
RPE cell transplantation approaches offer considerable hope
for the treatment of AMD [116]. Despite encouraging results,
there remain a number of challenges that must be overcome
for long-term efficiency. The cell delivery method must be
carefully considered since it may trigger the inflammatory
response in cell transplantation [20]. Development of low
cost, reliable and robust sources of RPE is necessary.
Optimization methods to deliver stem cell derived-RPE into
the subretinal space needs to be performed including long-
term survival and function of the implanted cells. In addition,
long-term safety such as the lack of tumor formation years
after the cell transplantation should be evaluated [117].
Further, in vivo monitoring methods are required to assess
the function of implanted cells in real time. Therapeutics
success of transplantation of stem cell-derived RPE cells may
be limited unless the transplanted cells can adhere and sur-
vive for a long period.
5.3. Cell delivery approaches in retinal cell
transplantation
There are two key approaches for retinal cell transplantation:
(i) injection of a cell suspension, and (ii) surgical implantation
of an RPE monolayer. Although, less invasive, the injection of
the cell suspension leads to poor cell survival and engraft-
ment which tend to form clusters [20]. Further, cells that do
not integrate may secrete pro-apoptotic signals and create
debris, causing further damage to the surrounding tissue.
The degeneration in AMD is accompanied by pathological
changes to BM and direct RPE transplantation onto aged BM
resultsinpoorcellsurvival,andadhesion[118,119]. This
necessitates the design of an artificial BM onto which RPE
cells can be cultured and subsequently transplanted. Various
biomaterials scaffolds have been studied for the develop-
ment of porous BM membranes designed to promote cell
survival, integration by protecting cells from the hostile host
environment [20,120122].
EXPERT OPINION ON DRUG DELIVERY 1155
Downloaded by [UMKC University of Missouri Kansas City] at 07:50 29 September 2017
5.4. Encapsulated cell technology (ECT) for AMD
ECT is a cell-based technology in which specific cells are
genetically engineered to overexpress the desired therapeutic
proteins [123,124]. These cells are then encapsulated into a
semipermeable biomaterials carrier that facilitates the diffu-
sion of nutrients and proteins to the host system (Figure 4).
ECT has the potential for continuous, long-term, and con-
trolled delivery of ocular therapeutics, bypassing the BRB with-
out any systemic exposure. In addition, a relatively low
sustained dose of drug emitted into the vitreous with an ECT
implant. Therefore, this system produces minimal adverse
events relative to the bolus injections.
NT-503 ECT implant containing genetically modified human
RPE cells can constantly secrete a controlled amount of solu-
ble VEGF-R at high levels to provide a therapeutic effect in
wet-AMD (http://www.neurotechusa.com/ect-platform.html).
Long-term cell survival due to the semipermeable hollow
fiber membrane of the device allows influx of oxygen and
nutrients and simultaneously creates an immune-isolation
chamber that prevents direct cell contact with the immune
system. In summary, ECT technology offers several advantages
such as avoidance of frequent intravitreal injections, expensive
treatment regimen as well as reduction of associated side
effects [124]. Since encapsulated cells allow continuous deliv-
ery of the product, the potential risks associated with high
concentrations of anti-VEGF therapeutics can be avoided.
6. Conclusion and future directions
Ocular drug delivery approaches to the posterior eye diseases
such as AMD are extremely challenging. Though the current
anti-VEGF treatments are effective, the financial burden on the
patient is substantial. To address the need, a sustained long-
term drug release from an ocular drug delivery platform is
required to reduce the frequency of the intravitreal injections.
The uses of nanotechnology approaches provide several
advantages in ocular applications. Thus, current research to
target posterior eye diseases is mostly focused on developing
NC drug delivery systems. However, a number of obstacles
need to be overcome prior to translation of these nanosys-
tems in clinical application such as, the reproducible and
large-scale fabrication of NCs, safety evaluation, and long-
term stability in biological fluids. The stem cell transplantation
provides positive responses but, remains questionable. ECT
offers the long-term delivery of therapeutics but, have chal-
lenges to solve.
Researchers are further investigating other delivery meth-
ods to extend the duration of action of anti-VEGF agents. A
capsule drug ring (CDR) has been designed for the treatment
of wet-AMD with a sustained delivery of anti-VEGF agent [125].
Results showed that the CDR device is biocompatible and may
release bevacizumab for more than 90 days. Other targets for
the treatment of wet-AMD such as the platelet derived growth
factor (PDGF) needs to be evaluated [126]. VEGF and PDGF
play separate but correlated roles in the formation and growth
of new blood vessels. PDGF binds to the pericytes (outside of
endothelial cells) in contrast to VEGF which binds to the inner
lining of blood vessels endothelial cells. While VEGF stimulates
abnormal blood vessel growth in the retina and macula, PDGF
stabilizes the maturing blood vessels and protects the inner
endothelial cells, despite anti-VEGF treatment. Therefore, a
combined anti-VEGF and anti-PDGF treatment may be more
effective than anti-VEGF mono therapy. Fovista, an anti-PDGF
drug from Ophthotech is under development for the potential
treatment of wet-AMD in combination with anti-VEGF
drugs [127].
This review summarized several nanosystems in addition to
the stem cell based cell transplantation approaches for the
posterior eye diseases such as AMD. Clinical pathogenesis and
Figure 4. Encapsulated cell technology (ECT) for the treatment of age-related macular degeneration (AMD). Retinal cells are encapsulated into a semi-permeable
scaffold made of biomaterials that facilitate the influx of nutrients and oxygen to provide the diffusion of therapeutic proteins directly to retina (vitreous humor)
thus, bypassing the bloodretinal barrier (BRB).
1156 V. AGRAHARI ET AL.
Downloaded by [UMKC University of Missouri Kansas City] at 07:50 29 September 2017
current treatment approaches in AMD are briefly summarized.
Several routes of ocular drug administration, elimination path-
ways, delivery barriers, their advantages and challenges are
discussed.
7. Expert opinion
The ultimate goals of a novel drug delivery system are to
improve the therapeutic efficacy and patient benefits.
Considering this, the purpose of an ocular dosage form is to
provide an optimal therapeutic effect with a minimum dose,
reduced injection frequency, and use of less invasive routes
with minimal side effects. Hence, an ideal ocular delivery
system should have several characteristic properties:
(a) Acceptability and patient compliance: the product should
be simple, nonirritating, easy to self-administer, have a conveni-
ent dosage regimen with a long-lasting action.
(b) Drug delivery system attributes: the product should allow
a high drug loading to reduce the instilled dose, appropriate size
to facilitate corneal uptake, provide no effects on vision, isotonic
and close to physiological pH to avoid irritation, provide a
controlled/sustained/stimuli-responsive drug release as required,
high specificity to the targeted tissues, and proper syringeability/
injectability to assure the administration of the prescribed dose.
(c) Therapeutic molecule characteristics: the drug molecule
should provide a long-term efficacy and higher compatibility
with other drugs, and amphipathic nature in order to pass
through different ocular tissues/cells.
(d) Safety concern, stability, and side effects: the product
should be safe with no or minimal localized and systemic toxi-
city, must be inert toward the ocular tissues, and have high
stability under diverse ocular/biological environmental and sto-
rage conditions.
(e) Feasibility of manufacturing and cost-effectiveness: the
product should be easily formulated and provide cost benefits to
the patients.
There is a rapid progress in the development of stimuli-
responsive nanoformulations such as NPs, MPs, and hydrogels
in ocular applications. However, these systems are not thor-
oughly investigated in ocular diseases. Translation is limited by
the complexity of formulation design and obstacles in scale-
up, toxicity of polymers and slow response of systems in the
presence of stimuli. For stimuli-responsive systems, the safety
and intraocular toxicity of final and degradation products
should be evaluated. Further, if stimuli-responsive approaches
can also be transformed into diagnostic application, this could
provide an emerging area of research in ocular therapeutics.
The cost-efficiency of the approach should also be carefully
evaluated along with scalability and regulatory requirements.
As the posterior ocular diseases progress, alteration in the
drug release pattern may require depending upon the treat-
ment and disease conditions. Out of several drug delivery
systems, ocular implants are promising platforms in the treat-
ment of posterior eye diseases. Currently available nonbiode-
gradable intraocular implants are designed to offer a long-
lasting near zero order release kinetics over an extended
period of time. The administration and removal procedure of
these implants increases the treatment cost significantly.
However, these nonbiodegradable implant systems could be
used for a longer duration (several years) if the drug can be
refilled using a nonsurgical approach. Moreover, controlling
the drug release using a stimuli-responsive approach based on
clinical needs may significantly enhance the treatment effi-
ciency in back of the eye diseases. Another exciting potential
strategy is the combination of delivery systems, such as NPs-
encapsulated hydrogel, for sustained and long-term drug
release. Composite nanosystems (NPs suspended in the ther-
moresponsive gel) have been formulated and in vitro results
showed positive responses [14,68]. These nanosystems pro-
vide a long-term sustained macromolecule drug release with
minimum burst effect. This can significantly reduce the dose
frequency, ultimately reduces the treatment cost burden to
the patients and could be a major breakthrough in targeting
of posterior eye diseases.
The physicochemical properties of NCs such as size, shape,
surface charge, and targeting ligand functionality are among
the key attributes that can influence their performances. These
parameters should be taken into account when designing an
effective ocular drug delivery system. The experimental design
approaches can be applied for the prescreening/optimization
of NC formulation and process variables. There are several
intraocular environments, formulation process development,
and NCs physicochemical parameters those can be considered
in the experimental design approaches to optimize NCs ther-
apeutic effects. Further studies are needed to facilitate the
understanding of the fundamentals of nanotechnology and
proper delivery routes. The efficacy of ocular delivery systems
depend not only the drug dose, but also, maintaining the
desired effects at the target site. Therefore, pharmacokinetics
models are important to assess the efficacy of ocular formula-
tions since they can significantly reduce the number of in vivo
experiments including the time and cost of the product devel-
opment. Several physicochemical properties of formulations
such as pH, viscosity, particle size, and osmolality, may affect
the ocular bioavailability and should be incorporated into
pharmacokinetics models. The selection of a suitable animal
model in the assessment of ocular toxicity is equally important
for the development of safe and effective delivery systems.
The stem cell transplantation strategy provides positive
results in AMD therapy, but, remains questionable due to the
lack of cell engraftment and survival in the host tissue after
transplantation. The success in restoring vision via cell trans-
plantation ultimately depends on the presence of residual PRs
in the retina. Hence, it is critical to accurately quantify PR cells.
Other major challenges in cell transplantation approach are to
develop an ethically acceptable treatment option without
immunological rejection issues. ECT offers the potential for
long-term and controlled delivery of therapeutics. One of the
major challenges of ECT approach is the potential for
increased macrophage activity in response to the presence
of artificial BM material and the cells must be able to survive
within the encapsulation matrix [25]. In addition, efforts
should be focused on the effects of BM scaffold thickness,
surface topography, mechanical properties, polymer degrada-
tion profile, and to find the most efficient way to sustain the
viability and functionality of the treatment [128]. Development
of diagnostic tests to assay retinal functions and transplanta-
tion methods for optimal delivery of RPE are essential. It is also
EXPERT OPINION ON DRUG DELIVERY 1157
Downloaded by [UMKC University of Missouri Kansas City] at 07:50 29 September 2017
necessary to determine the suitable storage conditions of the
ECT system to maintain the optimal function.
In summary, the most important aspects of posterior eye
disease therapeutics include the development of an effective
long-acting, and safe product that can be delivered with an
appropriate route of administration. There is a rapid develop-
ment of the technologies required to effectively deliver the
drugs to the back of the eye. However, a better understanding
of ocular barriers, and dynamic physiologic processes of the
eye such as clearance mechanisms and drug metabolism is
required. Ultimately, collaboration among scientists from aca-
demia, industry and clinics is needed for successful translation
of novel strategies to restore vision.
Funding
This study is supported by NIH R01EY09171 and NIH R01EY10659.
Declaration of interest
The authors have no other relevant affiliations or financial involvement
with any organization or entity with a financial interest in or financial
conflict with the subject matter or materials discussed in the manuscript
apart from those disclosed.
References
Papers of special note have been highlighted as either of interest ()orof
considerable interest (••) to readers.
1. Kim YC, Chiang B, Wu X, et al. Ocular delivery of macromolecules. J
Control Release. 2014 Sep;28(190):172181.
2. Gower NJ, Barry RJ, Edmunds MR, et al. Drug discovery in ophthal-
mology: past success, present challenges, and future opportunities.
BMC Ophthalmol. 2016;16:11.
3. Villegas VM, Aranguren LA, Kovach JL, et al. Current advances in
the treatment of neovascular age-related macular degeneration.
Expert Opin Drug Deliv. 2016 Aug;2:110.
4. Kompella UB, Amrite AC, Pacha Ravi R, et al. Nanomedicines for
back of the eye drug delivery, gene delivery, and imaging. Prog
Retin Eye Res. 2013 Sep;36:172198.
Key applications of nanotechnology are discussed in this
review. Additionally, nanofabricated delivery systems includ-
ing implants, films, microparticles, and nanoparticles are
described.
5. Kaur IP, Kakkar S. Nanotherapy for posterior eye diseases. J Control
Release. 2014 Nov;10(193):100112.
This review discusses various pathways available for effective
delivery to and clearance from the posterior eye. Promise held
by nanocarrier systems for effective delivery and selective
targeting is also discussed.
6. Eljarrat-Binstock E, Peer J, Domb AJ. New techniques for drug
delivery to the posterior eye segment. Pharm Res. 2010 Apr;27
(4):530543.
7. Gaudana R, Ananthula HK, Parenky A, et al. Ocular drug delivery.
AAPS J. 2010 Sep;12(3):348360.
8. Barar J, Aghanejad A, Fathi M, et al. Advanced drug delivery and
targeting technologies for the ocular diseases. Bioimpacts. 2016;6
(1):4967.
In the current study, recent advancements on ocular targeted
therapies are reviewed and discussed.
9. Boddu SH, Gupta H, Patel S. Drug delivery to the back of the eye
following topical administration: an update on research and
patenting activity. Recent Pat Drug Deliv Formul. 2014 Apr;8
(1):2736.
10. Peptu CA, Popa M, Savin C, et al. Modern drug delivery systems for
targeting the posterior segment of the eye. Curr Pharm Des.
2015;21(42):60556069.
11. Patel A, Cholkar K, Agrahari V, et al. Ocular drug delivery systems:
an overview. World J Pharmacol. 2013;2(2):4764.
•• This review summarizes the existing conventional formula-
tions for ocular delivery and their advancements followed
by current nanotechnology based formulation
developments.
12. Mandal A, Agrahari V, Khurana V, et al. Transporter effects on cell
permeability in drug delivery. Expert Opin Drug Deliv. 2016
Aug;5:117.
13. Rowe-Rendleman CL, Durazo SA, Kompella UB, et al. Drug and
gene delivery to the back of the eye: from bench to bedside.
Invest Ophthalmol Vis Sci. 2014 Apr;55(4):27142730.
14. Agrahari V, Agrahari V, Hung WT, et al. Composite nanoformulation
therapeutics for long-term ocular delivery of macromolecules. Mol
Pharm. 2016 Sep;13(9):29122922.
Discusses about the development of composite nanoformula-
tions and their application in long-term delivery of macromo-
lecule drugs.
15. Falavarjani KG, Nguyen QD. Adverse events and complications
associated with intravitreal injection of anti-VEGF agents: a review
of literature. Eye (Lond). 2013 Jul;27(7):787794.
16. Moore DJ, Clover GM. The effect of age on the macromolecular
permeability of human Bruchs membrane. Invest Ophthalmol Vis
Sci. 2001 Nov;42(12):29702975.
17. Rai Udo J, Young SA, Thrimawithana TR, et al. The suprachoroidal
pathway: a new drug delivery route to the back of the eye. Drug
Discov Today. 2015 Apr;20(4):491495.
18. Tetz M, Rizzo S, Augustin AJ. Safety of submacular suprachoroidal
drug administration via a microcatheter: retrospective analysis of
European treatment results. Ophthalmologica. 2012;227(4):183189.
19. Kang-Mieler JJ, Osswald CR, Mieler WF. Advances in ocular drug
delivery: emphasis on the posterior segment. Expert Opin Drug
Deliv. 2014 Oct;11(10):16471660.
This review summarizes currently available and recent devel-
opments for ocular drug delivery to both the anterior and
posterior segments. Modes of delivery, including topical, sys-
temic, transscleral/periocular and intravitreal, are discussed
with their corresponding examples including the advantages
and disadvantages are highlighted.
20. Delplace V, Payne S, Shoichet M. Delivery strategies for treatment
of age-related ocular diseases: from a biological understanding to
biomaterial solutions. J Control Release. 2015 Dec;10(219):652668.
•• This review provides a summary and discussion of the most
recent strategies employed for the delivery of both drugs and
cells to treat a variety of age-related eye diseases. The current
challenges and limitations to ocular delivery and how the use
of innovative materials can overcome these issues and ulti-
mately provide treatment for age-related degeneration and
regeneration of lost tissues are emphasized.
21. Wong WL, Su X, Li X, et al. Global prevalence of age-related
macular degeneration and disease burden projection for 2020
and 2040: a systematic review and meta-analysis. Lancet Glob
Health. 2014 Feb;2(2):e10616.
22. Jager RD, Mieler WF, Miller JW. Age-related macular degeneration.
N Engl J Med. 2008 Jun 12;358(24):26062617.
23. Das A, McGuire PG, Rangasamy S. Diabetic macular edema: patho-
physiology and novel therapeutic targets. Ophthalmology
2015;122:13751394.
24. Agarwal A, Rhoades WR, Hanout M, et al. Management of neovas-
cular age-related macular degeneration: current state-of-the-art
care for optimizing visual outcomes and therapies in development.
Clin Ophthalmol. 2015;9:10011015.
25. Nazari H, Zhang L, Zhu D, et al. Stem cell based therapies for age-
related macular degeneration: the promises and the challenges.
Prog Retin Eye Res. 2015 Sep;48:139.
•• This review outlines the current knowledge surrounding the
application of hESC-RPE and iPSC-RPE in AMD. Several
1158 V. AGRAHARI ET AL.
Downloaded by [UMKC University of Missouri Kansas City] at 07:50 29 September 2017
strategies to improve the stem cell-based treatments for AMD
are discussed.
26. Ambati J, Atkinson JP, Gelfand BD. Immunology of age-related
macular degeneration. Nat Rev Immunol. 2013 Jun;13(6):438451.
27. Shaw PX, Stiles T, Douglas C, et al. Oxidative stress, innate immu-
nity, and age-related macular degeneration. AIMS Mol Sci. 2016;3
(2):196221.
28. Smith AG, Kaiser PK. Emerging treatments for wet age-related
macular degeneration. Expert Opin Emerg Drugs. 2014 Mar;19
(1):157164.
29. Ozkiris A. Anti-VEGF agents for age-related macular degeneration.
Expert Opin Ther Pat. 2010 Jan;20(1):103118.
30. Age-Related Eye Disease Study Research Group. A randomized,
placebo-controlled, clinical trial of high-dose supplementation
with vitamins C and E, beta carotene, and zinc for age-related
macular degeneration and vision loss: AREDS report no. 8. Arch
Ophthalmol. 2001 Oct;119(10):14171436.
31. Rishi E, Rishi P, Sharma V, et al. Long-term outcomes of combina-
tion photodynamic therapy with ranibizumab or bevacizumab for
treatment of wet age-related macular degeneration. Oman J
Ophthalmol. 2016 May-Aug;9(2):8792.
32. Yang S, Zhao J, Sun X. Resistance to anti-VEGF therapy in neovas-
cular age-related macular degeneration: a comprehensive review.
Drug Des Devel Ther. 2016;10:18571867.
33. Eandi CM, Alovisi C, De Sanctis U, et al. Treatment for neovascular
age related macular degeneration: the state of the art. Eur J
Pharmacol. 2016 Sep;787:7883.
This review provides an overview of available data form clin-
ical trials supporting the use of anti-VEGF molecules for the
treatment of AMD.
34. Zhou B, Wang B. Pegaptanib for the treatment of age-related
macular degeneration. Exp Eye Res. 2006 Sep;83(3):615619.
35. Lu X, Sun X. Profile of conbercept in the treatment of neovascular
age-related macular degeneration. Drug Des Devel Ther.
2015;9:23112320.
36. Holz FG, Dugel PU, Weissgerber G, et al. Single-chain antibody
fragment vegf inhibitor rth258 for neovascular age-related macular
degeneration: a randomized controlled study. Ophthalmology.
2016 May;123(5):10801089.
37. Souied EH, Devin F, Mauget-Faysse M, et al. Treatment of exudative
age-related macular degeneration with a designed ankyrin repeat
protein that binds vascular endothelial growth factor: a phase I/II
study. Am J Ophthalmol. 2014 Oct;158(4):72432e2.
38. Amadio M, Govoni S, Pascale A. Targeting VEGF in eye neovascu-
larization: whats new?: A comprehensive review on current thera-
pies and oligonucleotide-based interventions under development.
Pharmacol Res. 2016 Jan;103:253269.
39. Pozarowska D, Pozarowski P. The era of anti-vascular endothelial
growth factor (VEGF) drugs in ophthalmology, VEGF and anti-VEGF
therapy. Cent Eur J Immunol. 2016;41(3):311316.
40. Mitra AK, Agrahari V, Mandal A, et al. Novel delivery approaches for
cancer therapeutics. J Control Release. 2015 Dec;10(219):248268.
41. Meng J, Agrahari V, Youm I. Advances in targeted drug delivery
approaches for the central nervous system tumors: the inspiration
of nanobiotechnology. J Neuroimmune Pharmacol. 2016 Jul 23.
42. Takashima Y, Tsuchiya T, Igarashi Y, et al. [Non-invasive ophthalmic
liposomes for nucleic acid delivery to posterior segment of eye].
Yakugaku Zasshi. 2012;132(12):13651370.
43. Davis BM, Normando EM, Guo L, et al. Topical delivery of Avastin to
the posterior segment of the eye in vivo using annexin A5-asso-
ciated liposomes. Small. 2014 Apr 24;10(8):15751584.
44. Abrishami M, Zarei-Ghanavati S, Soroush D, et al. Preparation char-
acterization, and in vivo evaluation of nanoliposomes-encapsulated
bevacizumab (avastin) for intravitreal administration. Retina. 2009
May;29(5):699703.
45. Elsaid N, Jackson TL, Elsaid Z, et al. PLGA microparticles entrapping
chitosan-based nanoparticles for the ocular delivery of ranibizu-
mab. Mol Pharm. 2016 Sep 6;13(9):29232940.
46. Huu VA, Luo J, Zhu J, et al. Light-responsive nanoparticle depot to
control release of a small molecule angiogenesis inhibitor in the
posterior segment of the eye. J Control Release. 2015 Feb;28
(200):7177.
A novel nanoparticle depot platform for on-demand drug
delivery using a far ultraviolet (UV) light-degradable polymer,
which allows noninvasively triggered drug release using brief,
low-power light exposure, is developed.
47. Hirani A, Grover A, Lee YW, et al. Triamcinolone acetonide nano-
particles incorporated in thermoreversible gels for age-related
macular degeneration. Pharm Dev Technol. 2016;21(1):6167.
48. Varshochian R, Riazi-Esfahani M, Jeddi-Tehrani M, et al.
Albuminated PLGA nanoparticles containing bevacizumab
intended for ocular neovascularization treatment. J Biomed Mater
Res A. 2015 Oct;103(10):31483156.
49. Huang D, Wang L, Dong Y, et al. A novel technology using trans-
scleral ultrasound to deliver protein loaded nanoparticles. Eur J
Pharm Biopharm. 2014 Sep;88(1):104115.
50. Ying L, Tahara K, Takeuchi H. Drug delivery to the ocular posterior
segment using lipid emulsion via eye drop administration: effect of
emulsion formulations and surface modification. Int J Pharm. 2013
Sep 10;453(2):329335.
51. Zhang L, Si T, Fischer AJ, et al. Coaxial electrospray of ranibizumab-
loaded microparticles for sustained release of anti-VEGF therapies.
Plos One. 2015;10(8):e0135608.
52. Yandrapu SK, Upadhyay AK, Petrash JM, et al. Nanoparticles in
porous microparticles prepared by supercritical infusion and pres-
sure quench technology for sustained delivery of bevacizumab.
Mol Pharm. 2013 Dec 2;10(12):46764686.
53. Ye Z, Ji YL, Ma X, et al. Pharmacokinetics and distributions of
bevacizumab by intravitreal injection of bevacizumab-PLGA micro-
spheres in rabbits. Int J Ophthalmol. 2015;8(4):653658.
54. Osswald CR, Kang-Mieler JJ. Controlled and extended release of a
model protein from a microsphere-hydrogel drug delivery system.
Ann Biomed Eng. 2015 Nov;43(11):26092617.
55. Patel S, Garapati C, Chowdhury P, et al. Development and evalua-
tion of dexamethasone nanomicelles with potential for treating
posterior uveitis after topical application. J Ocul Pharmacol Ther.
2015 May;31(4):215227.
56. Ma F, Nan K, Lee S, et al. Micelle formulation of hexadecyloxypro-
pyl-cidofovir (HDP-CDV) as an intravitreal long-lasting delivery sys-
tem. Eur J Pharm Biopharm. 2015;89:271279.
57. Yavuz B, Pehlivan SB, Vural I, et al. In vitro/in vivo evaluation of
dexamethasonePAMAM dendrimer complexes for retinal drug
delivery. J Pharm Sci. 2015 Nov;104(11):38143823.
58. Wu X, Yu G, Luo C, et al. Synthesis and evaluation of a nanoglob-
ular dendrimer 5-aminosalicylic acid conjugate with a hydrolyzable
schiff base spacer for treating retinal degeneration. ACS Nano. 2014
Jan 28;8(1):153161.
59. Coursey TG, Henriksson JT, Marcano DC, et al. Dexamethasone
nanowafer as an effective therapy for dry eye disease. J Control
Release. 2015 Sep;10(213):168174.
60. Yuan X, Marcano DC, Shin CS, et al. Ocular drug delivery nanowafer
with enhanced therapeutic efficacy. ACS Nano. 2015 Feb 24;9
(2):17491758.
61. Tuomela A, Liu P, Puranen J, et al. Brinzolamide nanocrystal
formulations for ophthalmic delivery: reduction of elevated
intraocular pressure in vivo. Int J Pharm. 2014 Jun 5;467(1
2):3441.
62. Gupta S, Samanta MK, Raichur AM. Dual-drug delivery system
based on in situ gel-forming nanosuspension of forskolin to
enhance antiglaucoma efficacy. AAPS Pharmscitech. 2010 Mar;11
(1):322335.
63. Xu X, Weng Y, Xu L, et al. Sustained release of Avastin(R) from
polysaccharides cross-linked hydrogels for ocular drug delivery. Int
J Biol Macromol. 2013 Sep;60:272276.
64. Tyagi P, Barros M, Stansbury JW, et al. Light-activated, in situ
forming gel for sustained suprachoroidal delivery of bevacizumab.
Mol Pharm. 2013 Aug 5;10(8):28582867.
65. Rauck BM, Friberg TR, Medina Mendez CA, et al. Biocompatible
reverse thermal gel sustains the release of intravitreal bevacizumab
in vivo. Invest Ophthalmol Vis Sci. 2014 Jan;55(1):469476.
EXPERT OPINION ON DRUG DELIVERY 1159
Downloaded by [UMKC University of Missouri Kansas City] at 07:50 29 September 2017
66. Wang CH, Hwang YS, Chiang PR, et al. Extended release of bevaci-
zumab by thermosensitive biodegradable and biocompatible
hydrogel. Biomacromolecules. 2012 Jan 9;13(1):4048.
67. Xie B, Jin L, Luo Z, et al. An injectable thermosensitive polymeric
hydrogel for sustained release of Avastin(R) to treat posterior seg-
ment disease. Int J Pharm. 2015 Jul 25;490(12):375383.
68. Patel SP, Vaishya R, Patel A, et al. Optimization of novel pentablock
copolymer based composite formulation for sustained delivery of
peptide/protein in the treatment of ocular diseases. J
Microencapsul. 2016;33(2):103113.
69. Agrahari V, Agrahari V, Mitra AK. Nanocarrier fabrication and
macromolecule drug delivery: challenges and opportunities. Ther
Deliv. 2016;7(4):257278.
In this excellent review, the advantages and limitations of
several routes of administration, novel delivery systems and,
fabrication approaches to make nanoformulations in different
shapes and sizes are summarized.
70. Bochot A, Fattal E. Liposomes for intravitreal drug delivery: a state
of the art. J Control Release. 2012 Jul 20;161(2):628634.
71. Akbarzadeh A, Rezaei-Sadabady R, Davaran S, et al. Liposome:
classification, preparation, and applications. Nanoscale Res Lett.
2013;8(1):102.
72. Herrero-Vanrell R, Bravo-Osuna I, Andres-Guerrero V, et al. The
potential of using biodegradable microspheres in retinal diseases
and other intraocular pathologies. Prog Retin Eye Res. 2014
Sep;42:2743.
73. Vaishya RD, Khurana V, Patel S, et al. Controlled ocular drug deliv-
ery with nanomicelles. Wiley Interdiscip Rev Nanomed
Nanobiotechnol. 2014 Sep-Oct;6(5):422437.
74. Cholkar K, Patel A, Vadlapudi AD, et al. Novel nanomicellar formu-
lation approaches for anterior and posterior segment ocular drug
delivery. Recent Pat Nanomed. 2012;2(2):8295.
75. Rodriguez Villanueva J, Navarro MG, Rodriguez Villanueva L.
Dendrimers as a promising tool in ocular therapeutics: latest
advances and perspectives. Int J Pharm. 2016 Sep 10;511(1):359
366.
76. Sharma OP, Patel V, Mehta T. Nanocrystal for ocular drug delivery:
hope or hype. Drug Deliv Transl Res. 2016 Aug;6(4):399413.
77. Buwalda SJ, Boere KW, Dijkstra PJ, et al. Hydrogels in a historical
perspective: from simple networks to smart materials. J Control
Release. 2014 Sep;28(190):254273.
In this review, an overview of the developments in hydrogel
research from simple networks to smart materials is discussed.
78. Yasin MN, Svirskis D, Seyfoddin A, et al. Implants for drug delivery
to the posterior segment of the eye: a focus on stimuli-responsive
and tunable release systems. J Control Release. 2014 Dec;28
(196):208221.
•• This article provides an overview of diseases of the posterior
segment of the eye describes currently available implants to
treat such conditions and discusses advantages and disadvan-
tages of various implant locations. Finally, stimuli-responsive
drug delivery technologies that have been investigated for, or
have the potential to be applied to, drug delivery to the back
of the eye are discussed.
79. Lee SS, Hughes P, Ross AD, et al. Biodegradable implants for
sustained drug release in the eye. Pharm Res. 2010 Oct;27
(10):20432053.
80. Bourges JL, Bloquel C, Thomas A, et al. Intraocular implants for
extended drug delivery: therapeutic applications. Adv Drug Deliv
Rev. 2006 Nov 15;58(11):11821202.
81. Thrimawithana TR, Young S, Bunt CR, et al. Drug delivery to the
posterior segment of the eye. Drug Discov Today. 2011 Mar;16(5
6):270277.
82. Haghjou N, Soheilian M, Abdekhodaie MJ. Sustained release intrao-
cular drug delivery devices for treatment of uveitis. J Ophthalmic
Vis Res. 2011 Oct;6(4):317329.
83. Wang J, Jiang A, Joshi M, et al. Drug delivery implants in the
treatment of vitreous inflammation. Mediators Inflamm.
2013;2013:780634.
84. Bansal P, Garg S, Sharma Y, et al. Posterior segment drug delivery
devices: current and novel therapies in development. J Ocul
Pharmacol Ther. 2016 Apr;32(3):135144.
85. Meng E, Hoang T. MEMS-enabled implantable drug infusion pumps
for laboratory animal research, preclinical, and clinical applications.
Adv Drug Deliv Rev. 2012 Nov;64(14):16281638.
86. Lo R, Li PY, Saati S, et al. A passive MEMS drug delivery pump for
treatment of ocular diseases. Biomed Microdevices. 2009 Oct;11
(5):959970.
87. Thakur Singh RR, Tekko I, McAvoy K, et al. Minimally invasive
microneedles for ocular drug delivery. Expert Opin Drug Deliv.
2016 Aug;25:113.
88. Park SH, Lee KJ, Lee J, et al. Microneedle-based minimally-invasive
measurement of puncture resistance and fracture toughness of
sclera. Acta Biomater. 2016;44:286294.
89. Mahlumba P, Choonara YE, Kumar P, et al. Stimuli-responsive poly-
meric systems for controlled protein and peptide delivery: future
implications for ocular delivery. Molecules. 2016 Jul;21(8):1002
1022.
90. Agrahari V, Zhang C, Zhang T, et al. Hyaluronidase-sensitive nano-
particle templates for triggered release of HIV/AIDS microbicide in
vitro. AAPS J. 2014 Mar;16(2):181193.
91. Porta IBM, Eckstein C, Xifre-Perez E, et al. Sustained, controlled and
stimuli-responsive drug release systems based on nanoporous
anodic alumina with layer-by-layer polyelectrolyte. Nanoscale Res
Lett. 2016 Dec;11(1):372.
92. Zhang T, Zhang C, Agrahari V, et al. Spray drying tenofovir loaded
mucoadhesive and pH-sensitive microspheres intended for HIV
prevention. Antiviral Res. 2013 Mar;97(3):334346.
93. Agrahari V, Meng J, Ezoulin MJ, et al. Stimuli-sensitive thiolated
hyaluronic acid based nanofibers: synthesis, preclinical safety and
in vitro anti-HIV activity. Nanomedicine (Lond). 2016 Nov;11
(22):29352958.
94. Christie JG, Kompella UB. Ophthalmic light sensitive nanocarrier
systems. Drug Discov Today. 2008 Feb;13(34):124134.
95. Matanovic MR, Kristl J, Grabnar PA. Thermoresponsive polymers:
insights into decisive hydrogel characteristics, mechanisms of gela-
tion, and promising biomedical applications. Int J Pharm. 2014 Sep
10; 472(12):262275.
This article reviews the types of thermoresponsive polymers.
Promising biomedical applications are described for injectable
formulations, which include solubilization of small hydropho-
bic drugs and delivery of labile biopharmaceutics, cell encap-
sulation, and tissue regeneration. Furthermore, combinations
of thermoresponsive hydrogels and various NCs as promising
systems for sustained drug delivery are discussed through
selected examples.
96. Agrawal AK, Das M, Jain S. In situ gel systems as smartcarriers for
sustained ocular drug delivery. Expert Opin Drug Deliv. 2012 Apr;9
(4):383402.
The present review summarizes the latest developments in
in situ gel technology, with regard to ophthalmic drug
delivery.
97. Chen X, Li X, Zhou Y, et al. Chitosan-based thermosensitive hydro-
gel as a promising ocular drug delivery system: preparation, char-
acterization, and in vivo evaluation. J Biomater Appl. 2012 Nov;27
(4):391402.
98. Yin H, Gong C, Shi S, et al. Toxicity evaluation of biodegradable and
thermosensitive PEG-PCL-PEG hydrogel as a potential in situ sus-
tained ophthalmic drug delivery system. J Biomed Mater Res B
Appl Biomater. 2010 Jan;92(1):129137.
99. Lafond M, Aptel F, Mestas JL, et al. Ultrasound-mediated ocular
delivery of therapeutic agents: a review. Expert Opin Drug Deliv.
2016 Jun;27:112.
An update on recent advances in transscleral and transcorneal
ultrasound-mediated drug delivery is provided.
100. Nabili M, Shenoy A, Chawla S, et al. Ultrasound-enhanced ocular
delivery of dexamethasone sodium phosphate: an in vivo study. J
Ther Ultrasound. 2014;2:6.
1160 V. AGRAHARI ET AL.
Downloaded by [UMKC University of Missouri Kansas City] at 07:50 29 September 2017
101. Svirskis D, Travas-Sejdic J, Rodgers A, et al. Electrochemically con-
trolled drug delivery based on intrinsically conducting polymers. J
Control Release. 2010 Aug 17;146(1):615.
102. Ramtin A, Seyfoddin A, Coutinho FP, et al. Cytotoxicity considera-
tions and electrically tunable release of dexamethasone from poly-
pyrrole for the treatment of back-of-the-eye conditions. Drug Deliv
Transl Res. 2016 Dec;6(6):793799.
103. Lo R, Li PY, Saati S, et al. A refillable microfabricated drug delivery
device for treatment of ocular diseases. Lab Chip. 2008 Jul;8
(7):10271030.
104. Jarrett SG, Boulton ME. Consequences of oxidative stress in age-related
macular degeneration. Mol Aspects Med. 2012 Aug;33(4):399417.
105. Xu Q, He C, Xiao C, et al. Reactive oxygen species (ROS) responsive
polymers for biomedical applications. Macromol Biosci. 2016
May;16(5):635646.
106. Mead B, Berry M, Logan A, et al. Stem cell treatment of degenera-
tive eye disease. Stem Cell Res. 2015 May;14(3):243257.
107. Bertolotti E, Neri A, Camparini M, et al. Stem cells as source for
retinal pigment epithelium transplantation. Prog Retin Eye Res.
2014 Sep;42:130144.
•• Different cell sources and their properties are discussed. A new
source of human RPE and the protocol for RPE differentiation
of retinal stem cells derived from adult ciliary bodies of post-
mortem donors is explained.
108. Heller JP, Martin KR. Enhancing RPE cell-based therapy outcomes
for AMD: the role of bruchs membrane. Transl Vis Sci Technol.
2014 Jun;3(3):11.
109. Kinnunen K, Petrovski G, Moe MC, et al. Molecular mechanisms of
retinal pigment epithelium damage and development of age-
related macular degeneration. Acta Ophthalmol. 2012 Jun;90
(4):299309.
A thorough explanation of RPE-derived mechanisms in AMD
pathology is provided.
110. De Jong PT. Age-related macular degeneration. N Engl J Med. 2006
Oct 5; 355(14):14741485.
•• In this excellent review, the clinical features of AMD, physiol-
ogy of the aging macula and to mechanisms implicated in the
cause is discussed.
111. Canto-Soler V, Flores-Bellver M, Vergara MN. Stem cell sources and
their potential for the treatment of retinal degenerations. Invest
Ophthalmol Vis Sci. 2016 Apr 1; 57(5):ORSFd19.
•• This paper provides a concise overview of the stem cell
sources most commonly used, weighing their therapeutic
potential on the basis of their technical strengths/limitations,
their ethical implications, and the extent of the progress
achieved to date.
112. Klassen H. Stem cells in clinical trials for treatment of retinal
degeneration. Expert Opin Biol Ther. 2016;16(1):714.
113. Zarbin M. Cell-based therapy for degenerative retinal disease.
Trends Mol Med. 2016 Feb;22(2):115134.
This review discusses about characteristics of stem cell therapy
in the eye and the challenges to clinical implementation that
are being confronted today.
114. Li Y, Tsai YT, Hsu CW, et al. Long-term safety and efficacy of human-
induced pluripotent stem cell (iPS) grafts in a preclinical model of
retinitis pigmentosa. Mol Med. 2012;18:13121319.
115. Hu Q, Friedrich AM, Johnson LV, et al. Memory in induced pluripo-
tent stem cells: reprogrammed human retinal-pigmented epithelial
cells show tendency for spontaneous redifferentiation. Stem Cells.
2010 Nov;28(11):19811991.
116. Carr AJ, Smart MJ, Ramsden CM, et al. Development of human
embryonic stem cell therapies for age-related macular degenera-
tion. Trends Neurosci. 2013 Jul;36(7):385395.
117. Kundu J, Michaelson A, Baranov P, et al. Approaches to cell deliv-
ery: substrates and scaffolds for cell therapy. Dev Ophthalmol.
2014;53:143154.
118. Gullapalli VK, Sugino IK, VanPatten Y, et al. Impaired RPE survival on
aged submacular human Bruchs membrane. Exp Eye Res. 2005
Feb;80(2):235248.
119. Sun K, Cai H, Tezel TH, et al. Bruchs membrane aging decreases
phagocytosis of outer segments by retinal pigment epithelium. Mol
Vis. 2007;13:23102319.
120. Xiang P, Wu KC, Zhu Y, et al. A novel Bruchs membrane-mimetic
electrospun substrate scaffold for human retinal pigment epithe-
lium cells. Biomaterials. 2014 Dec;35(37):97779788.
121. Shadforth AM, George KA, Kwan AS, et al. The cultivation of human
retinal pigment epithelial cells on Bombyx mori silk fibroin.
Biomaterials. 2012 Jun;33(16):41104117.
122. Treharne AJ, Thomson HA, Grossel MC, et al. Developing
methacrylate-based copolymers as an artificial Bruchsmem-
brane substitute. J Biomed Mater Res A. 2012 Sep;100
(9):23582364.
123. Emerich DF, Orive G, Thanos C, et al. Encapsulated cell therapy for
neurodegenerative diseases: from promise to product. Adv Drug
Deliv Rev. 2014 Apr;67-68:131141.
124. Tao W. Application of encapsulated cell technology for retinal
degenerative diseases. Expert Opin Biol Ther. 2006 Jul;6(7):717
726.
125. Gooch N, Burr RM, Holt DJ, et al. Design and in vitro biocompat-
ibility of a novel ocular drug delivery device. J Funct Biomater.
2013;4(1):1426.
126. Sadiq MA, Hanout M, Sarwar S, et al. Platelet-derived growth factor
inhibitors: a potential therapeutic approach for ocular neovascular-
ization. Dev Ophthalmol. 2016;55:310316.
127. Jaffe GJ, Eliott D, Wells JA, et al. 1 Study of intravitreous e10030 in
combination with ranibizumab in neovascular age-related macular
degeneration. Ophthalmology. 2016 Jan;123(1):7885.
128. Santos E, Pedraz JL, Hernandez RM, et al. Therapeutic cell encap-
sulation: ten steps towards clinical translation. J Control Release.
2013 Aug 28;170(1):114.
EXPERT OPINION ON DRUG DELIVERY 1161
Downloaded by [UMKC University of Missouri Kansas City] at 07:50 29 September 2017
1162 V. AGRAHARI ET AL.
Downloaded by [UMKC University of Missouri Kansas City] at 07:50 29 September 2017
... The cornea of the eye forms a tremendous barrier for the penetration of drug substances. To reach therapeutic drug concentrations in the retina, topical administration is usually not sufficient and intravitreal injection may be required to achieve the desired effect [28,29]. It has however been reported that lipid-based nanoparticles enhance the ocular penetration of drugs which are targeted for both anterior and posterior chambers when administered topically to the eye [30][31][32][33][34][35]. ...
Article
Recently, cGMP analogues have been investigated for the treatment of inherited retinal degenerations (IRD) using intravitreal injections. However, higher vitreous elimination rates limit the possibility to treat the retina with small molecule drugs. Here, we investigated the potential of lipid nanocapsules (LNCs) as vehicles to reduce clearance and prolong the delivery of cGMP analogue, CN03 to the retinal photoreceptors. Initially LNCs were investigated for both topical/periocular and intravitreal administration routes. While LNC-mediated drug permeation through the cornea proved to be too low for clinical applications, intravitreal application showed significant promise. Intravitreally administered LNCs containing fluorescent tracer in ex vivo porcine eyes showed complete intravitreal dispersal within 24 h. Ocular bio-distribution on histological sections showed that around 10 % of the LNCs had reached the retina, and 40 % accumulated in the ciliary body. For comparison, we used fluorescently labeled liposomes and these showed a different intraocular distribution with 48 % accumulated in the retina, and almost none were in the ciliary body. LNCs were then tested in retinal explants prepared from wild-type (WT) and rd1 mouse. In WT retina LNCs showed no significant toxic effects up to a concentration of 5 mg/mL. In rd1 retina, the LNC/CN03 formulation protected rd1 photoreceptors with similar efficacy to that of free CN03, demonstrating the usefulness of LNC/CN03 formulation in the treatment of IRD. Overall, our results indicate the suitability of LNCs for intraocular administration and drug delivery to both the retina and the ciliary body.
Chapter
Immiscible block copolymers in addition to their own assemblages have been the issue of substantial research in both basic and applied sciences. Block copolymers’ ability to construct highly ordered multimolecular frameworks, such as micelles and vesicles, with a controlled molecular weight (MW) distribution and partitioned parts for various phase separations, has prompted interest in the field. From a thermodynamic perspective, the assemblage properties of amphiphilic block copolymers can be interesting. Block copolymer segments regulate the assembly procedure, how the polymers and cargo are arranged, how stable the nanoassemblies are, how well they perform in biological environments, and more. Through the reticuloendothelial system, nanoparticles (NP) drug delivery systems can quickly remove the medication from the bloodstream while shielding it from the abrasive GI environment (RES). Moreover, drug-encapsulated nanoparticles produced from block copolymers can effectively boost drug levels at the inflamed region by actively targeting particular cells. This is accomplished by the use of ligand-specific or passive tissue targeting of changes in the tissue barrier caused by inflammation.
Article
Full-text available
Several chronic eye diseases affect the posterior segment of the eye. Among them age-related macular degeneration can cause vision loss if left untreated and is one of the leading causes of visual impairment in the world. Most treatments are based on intravitreally injected therapeutics that inhibit the action of vascular endothelial growth factor. However, due to the need for monthly injections, this method is associated with poor patient compliance. To address this problem, numerous drug delivery systems (DDSs) have been developed. This review covers a selection of particulate systems, non-stimuli responsive hydrogels, implants, and composite systems that have been developed in the last few decades. Depending on the type of DDS, polymer material, and preparation method, different mechanical properties and drug release profiles can be achieved. Furthermore, DDS development can be optimized by implementing mathematical modeling of both drug release and pharmacokinetic aspects. Several existing mathematical models for diffusion-controlled, swelling-controlled, and erosion-controlled drug delivery from polymeric systems are summarized. Compartmental and physiologically based models for ocular drug transport and pharmacokinetics that have studied drug concentration profiles after intravitreal delivery or release from a DDS are also reviewed. The coupling of drug release models with ocular pharmacokinetic models can lead to obtaining much more efficient DDSs for the treatment of age-related macular degeneration and other diseases of the posterior segment of the eye.
Article
Full-text available
Objective Therapeutic contact lenses, able to store drug and deliver it to the eye surface in a sustained fashion, gained interest as an effective and patient-friendly alternative to eye drops. Recent animal studies also demonstrated the presence of therapeutic drug levels in the back of the eye after wearing drug-loaded contact lenses, thus opening the possibility of treating the posterior segment without need of invasive intraocular injections. The drug pathways from contact lenses to the back of the eye require further investigation. Methods A mechanistic mathematical model was developed to evaluate the drug concentration over time in the tears, sclera and choroid, retina, aqueous humor and vitreous humor after the application of a therapeutic contact lens. The main drug transport mechanisms of the eye and the barrier properties of the different tissues were included in the model. Validation was performed by comparison with experimental data in literature. Results The model predictions of drug concentration over time reflected the experimental data both in the anterior and posterior segment of the eye. The model can differentiate between contributions to transport from different pathways. Conclusions The model constitutes a first step towards the possibility of predicting the ocular drug distribution and the treatment efficacy in the early stage of contact lens development, and it may help reduce both the need for in vivo tests (with ethical and economic advantages) and the gap between the lens design and clinical application. It also allows for an improved understanding of drug transport in the eye. Graphical abstract
Article
Full-text available
Ocular drug delivery is one of the most challenging endeavors among the various available drug delivery systems. Despite having suitable drugs for the treatment of ophthalmic disease, we have not yet succeeded in achieving a proper drug delivery approach with the least adverse effects. Nanotechnology offers great opportunities to overwhelm the restrictions of common ocular delivery systems, including low therapeutic effects and adverse effects because of invasive surgery or systemic exposure. The present review is dedicated to highlighting and updating the recent achievements of nano-based technologies for ocular disease diagnosis and treatment. While further effort remains, the progress illustrated here might pave the way to new and very useful ocular nanomedicines.
Article
Full-text available
This article presents current possibilities of using polyester-based materials in hard and soft tissue engineering, wound dressings, surgical implants, vascular reconstructive surgery, ophthalmology, and other medical applications. The review summarizes the recent literature on the key features of processing methods and potential suitable combinations of polyester-based materials with improved physicochemical and biological properties that meet the specific requirements for selected medical fields. The polyester materials used in multiresistant infection prevention, including during the COVID-19 pandemic, as well as aspects covering environmental concerns, current risks and limitations, and potential future directions are also addressed. Depending on the different features of polyester types, as well as their specific medical applications, it can be generally estimated that 25-50% polyesters are used in the medical field, while an increase of at least 20% has been achieved since the COVID-19 pandemic started. The remaining percentage is provided by other types of natural or synthetic polymers; i.e., 25% polyolefins in personal protection equipment (PPE).
Chapter
Drug delivery to the ocular diseases requires strategic approaches due to the existence of anatomical/static and physiological/dynamic barriers. Several ophthalmic conventional topical formulations are designed as solutions, suspensions, ointments, or emulsions to achieve an effective drug dose to the ocular tissues. In addition, various novel nanoformulation-based delivery systems have been explored through various routes of administration and showed promising results. In this book chapter, we briefly reviewed the routes of administration, ocular barriers with an emphasis on the topical route of drug administration, and the development of ocular suspension and nanosuspension formulations. The considerations in formulation development of suspension dosage forms are discussed to facilitate the development of safe, stable, and efficacious drug products. Moreover, the factors affecting the stability of suspensions/nanosuspensions and approaches to develop a stable product are summarized.
Chapter
The electrospinning method provides fabrication of nonwoven mats from nano to micrometers in size with controllable pores. Porous electrospun scaffolds have attracted increasing attention in biomedical applications especially due to their ability to mimic the extracellular matrix. This chapter presents a comprehensive overview of the advancements in the last five years through the design and preparation of porous biobased polymer mats using the electrospinning method for biomedical applications. Firstly, fundamentals of the electrospinning process are presented and followed by a discussion of the possible biomedical applications of micro- and nanostructured porous mats for drug delivery, wound dressing, tissue engineering, and other applications. Various crucial points for limitations and possible future trends are presented.
Article
Full-text available
Retinal pigment epithelial (RPE) is primarily impaired in age-related macular degeneration (AMD), leading to progressive loss of photoreceptors and sometimes choroidal neovascularization (CNV). mTOR has been proposed as a promising therapeutic target, while the usage of its specific inhibitor, rapamycin, was greatly limited. To mediate the mTOR pathway in the retina by a noninvasive approach, we developed novel biomimetic nanocomplexes where rapamycin-loaded nanoparticles were coated with cell membrane derived from macrophages (termed as MRaNPs). Taking advantage of the macrophage-inherited property, intravenous injection of MRaNPs exhibited significantly enhanced accumulation in the CNV lesions, thereby increasing the local concentration of rapamycin. Consequently, MRaNPs effectively downregulated the mTOR pathway and attenuate angiogenesis in the eye. Particularly, MRaNPs also efficiently activated autophagy in the RPE, which was acknowledged to rescue RPE in response to deleterious stimuli. Overall, we design and prepare macrophage-disguised rapamycin nanocarriers and demonstrate the therapeutic advantages of employing biomimetic cell membrane materials for treatment of AMD.
Article
Full-text available
Conditionally replicating adenoviruses have emerged as novel therapeutic agents for cancer. This study aimed to evaluate synergistic antitumor activity of replication-competent adenovirus armed with interleukin (IL)-18 (ZD55-IL-18) and dacarbazine (DTIC) against melanoma. Melanoma A375 cells or nude mouse tumor xenografts were treated with ZD55-IL-18 alone or together with DTIC. The results showed that ZD55-IL-18 competently replicated in A375 cells and expressed IL-18, and these were not affected by DTIC. ZD55-IL-18 enhanced the cytotoxicity of DTIC accompanied by increased apoptosis. Moreover, ZD55-IL-18 and DTIC synergistically inhibited the growth but promoted the apoptosis of A375 xenografts and inhibited vascular endothelial growth factor expression and lung metastasis in xenografts of nude mice. In conclusion, this is the first study to show synergistic anticancer activity of ZD55-IL-18 and DTIC for malignant melanoma. Our results provide evidence that chemo-gene-viro therapeutic approach has greater potential for malignant cancers than conventional chemotherapy or gene therapy.
Article
Full-text available
Angiogenesis is a clue process for tissue development and function, both in normal and pathological conditions. This process is regulated by multiple molecular systems. One of the most potent is vascular endothelial growth factor (VEGF) and its receptor (VEGFR) system. Members of this family are involved in new vessel formation in embryogenesis and maturation, as well as in reparative or pathological reactions in later stages. They play a substantial role in regeneration, inflammation, wound healing, as well as in cancer pathology. Nowadays it is possible to modulate VEGF-VEGFR interactions in many pathological conditions using anti-VEGF therapy. This therapy has already achieved a grounded position in the management of rheumatological disorders, tumour progression, and metastasis. Such drugs as bevacizumab, ranibizumab, aflibercept, and pegaptanib have also proven to be very effective in the treatment of several ocular diseases, such as age-related macular degeneration (AMD), macular oedema, or proliferative retinopathies and iris neovascularisation. The indications for the application of this therapy in ophthalmology are becoming wider and wider. It may also be used for corneal pathologies and in anti-glaucoma procedures.
Article
Full-text available
Controlled drug delivery systems are an encouraging solution to some drug disadvantages such as reduced solubility, deprived biodistribution, tissue damage, fast breakdown of the drug, cytotoxicity, or side effects. Self-ordered nanoporous anodic alumina is an auspicious material for drug delivery due to its biocompatibility, stability, and controllable pore geometry. Its use in drug delivery applications has been explored in several fields, including therapeutic devices for bone and dental tissue engineering, coronary stent implants, and carriers for transplanted cells. In this work, we have created and analyzed a stimuli-responsive drug delivery system based on layer-by-layer pH-responsive polyelectrolyte and nanoporous anodic alumina. The results demonstrate that it is possible to control the drug release using a polyelectrolyte multilayer coating that will act as a gate. Electronic supplementary material The online version of this article (doi:10.1186/s11671-016-1585-4) contains supplementary material, which is available to authorized users.
Article
Full-text available
Therapeutic proteins and peptides have become notable in the drug delivery arena for their compatibility with the human body as well as their high potency. However, their biocompatibility and high potency does not negate the existence of challenges resulting from physicochemical properties of proteins and peptides, including large size, short half-life, capability to provoke immune responses and susceptibility to degradation. Various delivery routes and delivery systems have been utilized to improve bioavailability, patient acceptability and reduce biodegradation. The ocular route remains of great interest, particularly for responsive delivery of macromolecules due to the anatomy and physiology of the eye that makes it a sensitive and complex environment. Research in this field is slowly gaining attention as this could be the breakthrough in ocular drug delivery of macromolecules. This work reviews stimuli-responsive polymeric delivery systems, their use in the delivery of therapeutic proteins and peptides as well as examples of proteins and peptides used in the treatment of ocular disorders. Stimuli reviewed include pH, temperature, enzymes, light, ultrasound and magnetic field. In addition, it discusses the current progress in responsive ocular drug delivery. Furthermore, it explores future prospects in the use of stimuli-responsive polymers for ocular delivery of proteins and peptides. Stimuli-responsive polymers offer great potential in improving the delivery of ocular therapeutics, therefore there is a need to consider them in order to guarantee a local, sustained and ideal delivery of ocular proteins and peptides, evading tissue invasion and systemic side-effects.
Article
Full-text available
At present, brain tumor is among the most challenging diseases to treat and the therapy is limited by the lack of effective methods to deliver anticancer agents across the blood-brain barrier (BBB). BBB is a selective barrier that separates the circulating blood from the brain extracellular fluid. In its neuroprotective function, BBB prevents the entry of toxins, as well as most of anticancer agents and is the main impediment for brain targeted drug delivery approaches. Nanotechnology-based delivery systems provide an attractive strategy to cross the BBB and reach the central nervous system (CNS). The incorporation of anticancer agents in various nanovehicles facilitates their delivery across the BBB. Moreover, a more powerful tool in brain tumor therapy has relied surface modifications of nanovehicles with specific ligands that can promote their passage through the BBB and favor the accumulation of the drug in CNS tumors. This review describes the physiological and anatomical features of the brain tumor and the BBB, and summarizes the recent advanced approaches to deliver anticancer drugs into brain tumor using nanobiotechnology-based drug carrier systems. The role of specific ligands in the design of functionalized nanovehicles for targeted delivery to brain tumor is reviewed. The current trends and future approaches in the CNS delivery of therapeutic molecules to tumors are also discussed.
Article
Aim: To develop a seminal enzyme bioresponsive, mucoadhesive nanofibers (NFs) as safe and effective nanocarriers for the prevention of HIV vaginal transmission. Methods: A novel thiolated hyaluronic acid (HA-SH) polymer was synthesized to fabricate tenofovir (TFV)-loaded electrospun NFs (HA-SH-NFs) and characterized in vitro/in vivo. Results: A triggered drug release (87% w/w) from the engineered HA-SH-NFs (mean diameter ∼75 nm) occured within 1 h under the influence of seminal hyaluronidase enzyme. HA-SH-NFs were noncytotoxic, induced no damage on the C57BL/6 mice genital-tract and other organs. No significant CD45 cell-infiltration and changes in cytokines level in cervicovaginal tissues were observed. HA-SH-NFs significantly enhanced both TFV retention and bioavailability in vaginal tissue compared with the 1% TFV-gel. The anti-HIV activity of TFV (on pseudotyped virus followed by luciferase assay) was not adversely affected by the electrospinning process. Conclusion: HA-SH-NFs developed in this study could potentially serve as a safe nanotemplate for topical intravaginal elivery of HIV/AIDS microbicides.
Article
Statement of significance: Measurement of mechanical property of soft biological tissue is challenging due to variations between tissue samples or lack of well-defined measurement techniques. Although non-invasive measurement techniques such as nano/micro indentation were employed to locally measure the elastic modulus of soft biological materials, mechanical properties such as puncture resistance or fracture toughness, which requires "invasive" measurement and is important for the application of "microneedles or hypodermic needles", has not been well studied. In this work, we report minimally-invasive measurement of puncture resistance and fracture toughness of sclera using a double MN insertion method. Parametric studies showed that use of MN proved to be advantageous because of minimally-invasive insertion into tissue as well as higher sensitivity to sub-tissue architecture during the measurement.
Article
Introduction: Anterior and posterior segment eye diseases are highly challenging to treat, due to the barrier properties and relative inaccessibility of the ocular tissues. Topical eye drops and systemically delivered treatments result in low bioavailability. Alternatively, direct injection of medication into the ocular tissues is clinically employed to overcome the barrier properties, but injections cause significant tissue damage and are associated with a number of untoward side effects and poor patient compliance. Microneedles (MNs) has been recently introduced as a minimally invasive means for localizing drug formulation within the target ocular tissues with greater precision and accuracy than the hypodermic needles. Areas covered: This review article seeks to provide an overview of a range of challenges that are often faced to achieve efficient ocular drug levels within targeted tissue(s) of the eye. It also describes the problems encountered using conventional hypodermic needle-based ocular injections for anterior and posterior segment drug delivery. It discusses research carried out in the field of MNs, to date. Expert opinion: MNs can aid in localization of drug delivery systems within the selected ocular tissue. And, hold the potential to revolutionize the way drug formulations are administered to the eye. However, the current limitations and challenges of MNs application warrant further research in this field to enable its widespread clinical application.
Article
Introduction: The role of drug transporters as one of the determinants of cellular drug permeability has become increasingly evident. Despite the lipophilicity of a drug molecule as rate-limiting factor for passive diffusion across biological membranes, carrier-mediated and active transport have gained attention over the years. A better understanding of the effects and roles of these influx transporters towards transmembrane permeability of a drug molecule need to be delineated for drug development and delivery. Areas covered: This review focuses on findings relative to role of transporters in drug absorption and bioavailability. Particularly the areas demanding further research have been emphasized. This review will also highlight various transporters expressed on vital organs and their effects on drug pharmacokinetics. Expert opinion: Significant efforts have been devoted to understand the role of transporters, their iterative interplay with metabolizing enzymes through molecular enzymology, binding and structure-activity relationship studies. A few assays such as parallel artificial membrane permeation assay (PAMPA) have been developed to analyze drug transport across phospholipid membranes. Although large web-accessible databases on tissue selective expression profiles at transcriptomic as well as proteomic are available, there is a need to collocate the scattered literature on the role of transporters in drug development and delivery.
Article
Introduction: Age-related macular degeneration (AMD) is the most common cause of permanent central visual acuity loss in persons over 65 years of age in industrialized nations. Today, intravitreal vascular endothelial growth factor (VEGF) inhibitors are the mainstay of treatment worldwide. Areas covered: The following review covers the current treatments and challenges of wet AMD management. It also covers emerging therapies including radiation, latest generation anti-VEGF agents, and combination therapies. Expert opinion: Current neovascular AMD therapy is aimed at decreasing the VEGF effect at the choroidal neovascularization (CNV) complex. The most important existing challenges in the treatment of neovascular AMD are improving visual outcomes, decreasing the treatment burden, and minimizing geographic atrophy. Clinicians are using many treatment strategies to minimize intravitreal injections without sacrificing visual outcomes. Combination of anti-VEGF therapy with other previously available treatments that target a different pathophysiological mechanism may be a reasonable clinical strategy to minimize intravitreal injections. Many exciting novel drugs that target newly discovered pathways associated with CNV development and progression hold clinical promise. The results of ongoing randomized clinical trials will answer the important concerns surrounding new drugs and delivery devices: safety and visual outcomes.