You are on page 1of 11

Advanced Drug Delivery Reviews 108 (2017) 2–12

Contents lists available at ScienceDirect

Advanced Drug Delivery Reviews

journal homepage: www.elsevier.com/locate/addr

Advances in nano-based inner ear delivery systems for the treatment of


sensorineural hearing loss☆
Lilun Li a,b, Tiffany Chao a, Jason Brant a, Bert O'Malley Jr. a, Andrew Tsourkas c, Daqing Li a,⁎
a
Department of Otorhinolaryngology—Head & Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
b
New York University School of Medicine, New York, NY 10016, USA
c
Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA

a r t i c l e i n f o a b s t r a c t

Article history: Sensorineural hearing loss (SNHL) is one of the most common diseases, accounting for about 90% of all hearing
Received 23 November 2015 loss. Leading causes of SNHL include advanced age, ototoxic medications, noise exposure, inherited and autoim-
Received in revised form 3 January 2016 mune disorders. Most of SNHL is irreversible and managed with hearing aids or cochlear implants. Although
Accepted 4 January 2016
there is increased understanding of the molecular pathophysiology of SNHL, biologic treatment options are
Available online 12 January 2016
limited due to lack of noninvasive targeted delivery systems. Obstacles of targeted inner ear delivery include
Keywords:
anatomic inaccessibility, biotherapeutic instability, and nonspecific delivery. Advances in nanotechnology may
Nanomedicine provide a solution to these barriers. Nanoparticles can stabilize and carry biomaterials across the round window
Nanohydrogel membrane into the inner ear, and ligand bioconjugation onto nanoparticle surfaces allows for specific targeting.
Targeted delivery A newer technology, nanohydrogel, may offer noninvasive and sustained biotherapeutic delivery into specific
Ototoxicity inner ear cells. Nanohydrogel may be used for inner ear dialysis, a potential treatment for ototoxicity-induced
Nanodialysis SNHL.
Inner ear © 2016 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
1.1. Systemic drug delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
1.2. Intratympanic drug delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
1.3. Hydrogel delivery system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
1.4. Nanoparticle delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
2. Inner ear anatomy and physiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
2.1. Sound transduction pathway . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
2.2. Cochlear diffusion of drugs via the round window membrane . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
3. Nanoparticle delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
4. Targeting nanoparticle delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
5. Applications of nanotechnology to sensorineural hearing loss of inner ear disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
5.1. Nanoparticle approach for noise induced hearing loss . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
5.2. Nanoparticle approach for different types of SNHL . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
5.3. Nanodialysis for cisplatin-induced ototoxicity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
6. Conclusion and future directions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10

☆ This review is part of the Advanced Drug Delivery Reviews theme issue on “Editors' Collection 2016”.
⁎ Corresponding author at: Department of Otolaryngology—Head & Neck Surgery, University of Pennsylvania Health System, CRB 145, 415 Curie Blvd, Philadelphia, PA 19104, USA.
Tel.: +1 215 746 2953; fax: +1 215 573 1934.
E-mail address: lidaqing@mail.med.upenn.edu (D. Li).

http://dx.doi.org/10.1016/j.addr.2016.01.004
0169-409X/© 2016 Elsevier B.V. All rights reserved.
L. Li et al. / Advanced Drug Delivery Reviews 108 (2017) 2–12 3

1. Introduction experienced weight gain of 10 lb or more. Although the hyperglycemia


appeared to be reversible, the weight gain persisted even after drug
The inner ear consists of the cochlea and vestibule, and it is a crucial discontinuation [7]. Aminoglycoside toxicity, too, can produce hearing
drug delivery target for the treatment of hearing and balance disorders, loss and persistent disequilibrium [9–11]. Although certain aminoglyco-
respectively. Routes for drug entry into the inner ear include the sys- sides, such as streptomycin and gentamicin, are more vestibulotoxic
temic circulation, which feeds the labyrinthine artery that supplies the than cochleotoxic, high doses can lead to cochlear damage and sensori-
cochlea and vestibule, and the round window membrane (RWM), neural hearing loss [8–10].
which connects the middle ear with the inner ear. Anatomic challenges
for drug delivery into the inner ear include the blood-inner ear barrier, 1.2. Intratympanic drug delivery
limited labyrinthine artery supply, and difficult access to the RWM.
Mechanistic challenges for drug delivery into the inner ear include var- To improve the efficacy and reduce the adverse effects of systemic
iations in RWM permeability, biotherapeutic instability, uncontrolled drug delivery, more localized drug delivery systems were developed
drug elimination, and nonspecific drug delivery. While inner ear to target inner ear disease. Schuknecht et al. was the first to introduce
diseases have historically been treated with systemic therapies, local intratympanic (IT) injection as a means of delivering streptomycin
intratympanic (IT) injection can offer more efficient and less systemical- into the inner ear for the successful treatment of Meniere's disease pa-
ly toxic drug delivery. Hydrogel, another localized drug delivery system, tients [12]. IT injection of a drug into the middle ear space allows for
has the additional advantages of sustained and controlled drug release, drug diffusion across the round window membrane (RWM) into the
which can reduce inter-patient variability in drug elimination while inner ear. In bypassing the labyrinthine artery and blood-inner ear bar-
maximizing drug diffusion. Although these forms of drug delivery rier, IT injection provides a more direct and efficient approach as com-
have been used successfully in the treatment of various inner diseases, pared to systemic drug administration. In fact, drug concentrations
such as sudden sensorineural hearing loss (SHL), autoimmune inner measured in the inner ear fluids, perilymph and endolymph, are signif-
ear disease (AIED), and Meniere's disease, they can neither stabilize icantly higher with IT injection than with oral or parenteral administra-
newer therapeutics, including biomaterials, nor target therapies to tion [6,13]. Not only can IT injection more efficiently deliver drugs into
specific cell types within the inner ear. Nanotechnology may be able the inner ear, it can also avoid many of the side effects associated with
to address these current limitations and offer a noninvasive, sustained systemic therapy.
and targeted drug delivery system. Due to these advantages and the relative ease of performing the pro-
cedure in the office, IT injection of corticosteroids and aminoglycosides
1.1. Systemic drug delivery is now commonly used for treatment of SHL and Meniere's disease.
[1,13]. For example, IT corticosteroid injections can be used as prima-
Systemic drug delivery can be accomplished via the intravenous, ry or salvage therapy for SHL and for patients with contraindications
intramuscular or oral route, and it has been successfully used to treat for systemic corticosteroid therapy [14]. A large randomized pro-
inner ear diseases such as sudden hearing loss (SHL), autoimmune spective trial by Rauch et al. found that primary IT corticosteroid
inner ear disease (AIED), and Meniere's disease. Oral corticosteroids therapy and primary oral corticosteroid therapy were equally effective
are commonly prescribed for the treatment of SHL, defined as a 30 at restoring hearing in patients with SHL, although the latter was asso-
decibels or greater hearing loss in 3 consecutive frequencies over less ciated with systemic side effects such as hyperglycemia and appetite
than 72 h [1]. A pioneering study by Wilson et al. demonstrated that changes [15]. A more recent study by Filipo et al. found that while IT
oral corticosteroids could effectively reverse SHL with a recovery rate and oral corticosteroid therapy produced similar improvement in the
of 61%, as compared with a placebo recovery rate of 32% [2]. Additional pure tone averages of patients with SHL, IT injection led to significantly
studies have since demonstrated SHL recovery rates of 57–66% with oral better hearing threshold improvement, suggesting the therapeutic su-
corticosteroid use [3,4]. More long-term courses of oral corticosteroids periority of primary IT injection [4]. IT corticosteroid injection for the
are used in the treatment of AIED, which presents as progressive treatment of AIED is less well-studied, and the few reports published in-
bilateral asymmetric sensorineural hearing loss (SNHL) occurring over volve AIED patients who have failed or could not tolerate systemic ste-
weeks to months, with possible vestibular deficit [5–7]. The current roid and/or methotrexate therapy. While high dose oral corticosteroids
treatment protocol for AIED is 4 weeks of high-dose oral prednisone continue to be the standard of care for AIED, IT corticosteroids have
and subsequent tapering to the lowest possible dose while maintaining shown promise as at least a salvage treatment option for these patients
therapeutic effect; the rate of hearing preservation using this regimen is [6].
reportedly between 53 and 70% [6]. Like SHL and AIED, Meniere's dis- Although IT delivery is efficient and has reduced the toxicities asso-
ease has also been successfully treated with systemic drug therapy. ciated with systemic drug delivery, it continues to demonstrate incon-
Aminoglycosides, which were historically delivered intra-muscularly, sistency in delivering standard doses of drugs into the inner ear.
are indicated for the treatment of vertigo in patients with who have Because IT injection relies on diffusion for the drug to reach the inner
failed more conservative medical therapies [1]. Although inherently ear from the middle ear, its success is directly related to the amount of
ototoxic, well-titrated doses of aminoglycosides can preferentially ab- drug that comes into contact with the RWM. Any drug that is not in con-
late vestibular structures that are responsible for balance, thereby re- tact with the RWM is eliminated via the Eustachian tube. Differences in
ducing vertigo symptoms in Meniere's disease patients [8,9]. However, RWM permeability can thus lead to variable rates of drug retention and
even with current titration methods, hearing loss associated with treat- elimination. This inconsistency is evident in the application of IT amino-
ment is not uncommon. glycosides for Meniere's disease treatment. As mentioned, aminoglyco-
Though drug delivery to the inner ear can be achieved via systemic sides are inherently ototoxic and can cause hearing loss and permanent
administration, limited local blood supply and poor penetration of the disequilibrium at inappropriately high doses. Early studies of IT strepto-
blood-inner ear barrier often results in subtherapeutic local concentra- mycin delivery for Meniere's disease showed that although vertigo was
tions. Administration of large doses of medication is required to produce relieved in the majority of patients, hearing loss developed in greater
the desired therapeutic effect, often leading to severe toxicities. System- than 25% of patients [12,16]. In an effort to reduce toxicity, the clinical
ic corticosteroids can lead to hypertension, hyperglycemia, osteoporosis endpoint of therapy changed over time from complete ablation to
and immunosuppression, along with more long-term adrenal suppres- alteration of the number of vertiginous attacks, and various timing
sion at high doses [1]. In a prospective study by Alexander et al. of and dosage regimens of IT gentamicin injection were tested [1,17]. A
AIED patients receiving oral prednisone, 17.6% of patients developed meta-analysis by Chia et al. evaluated the efficacy of vertigo control
of hyperglycemia in a dose-dependent manner and 39% of patients and hearing loss rate of scheduled drug delivery, including various
4 L. Li et al. / Advanced Drug Delivery Reviews 108 (2017) 2–12

timing and dosage regimens, as compared to drug titration. They con- 2.1. Sound transduction pathway
cluded that the titration method had the best rate of vertigo control
(96.3%) while multiple daily dosage had the highest rate of hearing To reach the inner ear, sound traverses the auditory canal where
loss (34.7%) [18]. The superiority of the titration method, in which mechanical energy causes vibration of the tympanic membrane at its
drug delivery is terminated with onset of vestibular symptoms, changes medial extent. This vibration is transmitted across the middle ear ossi-
in vertigo, or hearing loss, as compared with scheduled dosing empha- cles, the malleus, incus, and stapes, amplifying the energy received by
sizes the significant inter-patient variability seen with IT drug delivery. the inner ear via lever effects. The energy is further amplified (17:1)
The hearing toxicity associated with current IT aminoglycoside in- due to the difference in the surface area of the tympanic membrane
jection indicates that this approach cannot achieve targeted inner ear and oval window [6].
drug delivery. Instead of seeking a drug that is purely vestibulotoxic, it Sound energy is transmitted to the cochlea as the stapes footplate
would be prudent to develop a drug delivery system that specifically vibrates on the oval window, one of the structures that connects the
carries the drug to the cell population of interest. The ideal drug delivery middle ear with the inner ear. The cochlea is a spiral apparatus with
system for Meniere's disease would target the vestibular hair cells, approximately 2.5 turns that contains three fluid-filled longitudinal
which sense and transduce positional displacement, and the dark cells tracts: the scala vestibule, scala media and scala tympani [22]. The
of the stria vascularis and planum semilunatum, which produce the scala vestibule and scala tympani carry perilymph, while the scala
endolymph associated with vertiginous symptoms [18]. media carries endolymph. Sound vibrations along the basilar membrane
of the cochlea generate fluid waves within these cochlear tracts, and the
1.3. Hydrogel delivery system differential structural composition of the cochlea along its length causes
maximal vibration by higher frequencies at the base and lower frequen-
To address the inconsistency and inter-patient variability of IT drug cies at the apex. The mechanical energy of basilar membrane vibration is
delivery, hydrogel was developed as alternative form of localized drug converted into electrical energy via surface mechanoreceptors on highly
delivery. Paulson et al. first introduced the chitosan-glycerophosphate specialized hair cells [23,24]. Each hair cell has apical extensions called
(CGP) hydrogel delivery system, which aimed to reduce variation in stereocilia, which respond to fluid deflection by opening transduction
drug elimination by providing more sustained and controlled drug channels and causing hair cell depolarization. Hair cell depolarization
delivery into the inner ear [19]. The CGP hydrogel is applied, as a liquid, then triggers depolarization of the bipolar neurons of the spiral gangli-
directly onto the round window niche (RWN), where it then solidifies at on, which collectively form the cochlear division of CN VIII. The cochlear
body temperature to maximize RWM contact. CGP hydrogel is com- nerve travels along the internal auditory canal and eventually synapses
posed of a porous matrix, which is degraded over time by middle ear ly- with other neuronal cells in the central nervous system [23,24]. Dys-
sozymes to generate slow and continuous drug release. Studies have function or damage at any point along this neuronal pathway can lead
since shown that the CPG hydrogel system can steadily release drugs to hearing loss or dysfunction. Thus, current research is aimed at finding
such as dexamethasone and gentamicin into the perilymph of the biomaterials that will increase neuronal survival and potentiate neurite
inner ear [19,20]. In functional studies of mice who received CPG outgrowth as a means of treating specific forms of neural hearing loss.
hydrogel-delivered gentamicin, the sustained release of relatively low [25–28].
drug concentrations per day led to longer vestibular suppression and
less risk of hearing loss as compared with the IT injection group [20]. 2.2. Cochlear diffusion of drugs via the round window membrane
Not only does the CPG-hydrogel system offer a more efficient and pre-
dictable release mechanism, it can be further regulated by the enzyme Current inner ear drug delivery strategies rely on another connec-
chitosanase. Chitosanase can break down the chitosan chains in the hy- tion between the middle and inner ear, the round window, which is
drogel through a glycolytic reaction to cause faster drug elimination via covered by a three-layered, semi-permeable round window membrane
the Eustachian tube [21]. In the case of gentamicin hydrogel administra- (RWM). The RWM is composed of an outer epithelial layer, middle fi-
tion, chitosanase can act as an additional safety measure to prevent brous layer, and an inner epithelial layer [22]. As demonstrated with
hearing loss in the event of an unanticipated drug reaction. IT injection, hydrogel delivery and nanoparticle delivery (see Sections
1.2–1.4), the round window has been a successful entryway for inner
1.4. Nanoparticle delivery ear drug delivery. The exact pharmacokinetics of drug delivery across
the human RWM are difficult to study, due to the risk of permanent
While the methods discussed above can provide noninvasive, hearing loss when sampling inner ear fluids. However, animal studies
sustained and regulated drug delivery into the inner ear, they cannot and computer simulations have provided greater understanding and
deliver newer therapeutics, including biomaterials, which are rela- quantification of drug delivery into the inner ear [29–31].
tively unstable within extracellular compartments. Furthermore, Drug dispersal into the inner ear is influenced by multiple factors,
these methods cannot by themselves target delivery to specific loca- including RWM permeability, the rate of drug diffusion within the peri-
tions or cell types in the inner ear. Nanoparticle technology has lymph, and the rate of drug clearance. RWM permeability depends upon
therefore been of great interest due to its stabilizing effect on bioma- solute characteristics as well as local biochemical mediators, RWM
terials and potential for targeted therapeutic delivery. Please refer to thickness, and presence of RWM inflammation or injury [22]. Smaller,
Section 4 for further discussion about the applications of nanoparticle cationic particles preferentially pass through the RWM, and reduced
drug delivery for treatment of inner ear disease. RWM thickness increases permeability [32]. Local biochemical media-
tors, such as leukotrienes, prostaglandins, and histamine, can alter
2. Inner ear anatomy and physiology RWM permeability; in fact, histamine has been shown to facilitate up-
take of intratympanically injected dexamethasone [22,33]. Similarly,
The inner ear consists of the cochlea and the vestibule, which are re- the local inflammatory state of the RWM can affect its permeability.
sponsible for hearing and balance, respectively. These are exquisitely Goycoolea et al. found that initial otitis media infection led to increased
delicate organs encased in the hardest bone in the body with limited RWM permeability, while later stages of infection led to decreased
pathways for access. Efforts to target therapy into the inner ear are fur- permeability due to granulation tissue formation and RWM thickening
ther challenged by the need for noninvasive access, as invasion of the [32,34]. Nordang et al. found that inflammation secondary to topical ap-
inner ear leads to irreversible sensorineural hearing loss (SNHL). Fig. 1 plication of saline or hydrocortisone can, over time, lead to local edema
illustrates the anatomy of ear and the location of the round window, a and membrane leakage of the RWM, which in turn increases RWM per-
crucial site of entry for drug delivery into the inner ear. meability [35]. This finding suggests that any intratympanic injection
L. Li et al. / Advanced Drug Delivery Reviews 108 (2017) 2–12 5

Scala Vestibuli
(Perilymph)

Scala Media
(Endolymph)
Outer hair cells

Inner hair cell


Cochlear nerve
Spiral Ganglion (CN VIII)

Scala Tympani
(Perilymph)

ve
ner
lar e
stib
u erv
Ve arn
hle
oc

Semicircular
Malleus Canals

Auditory canal Oval


Tympanic Incus Window
Membrane Stapes Round Window

(Round window niche,


Round window membrane)
Eu
s
ta
ch
ia
n
Tu
b e

Fig. 1. Ear anatomy and sites for targeted inner ear delivery. Sound enters the auditory canal and delivers mechanical energy to the middle ear via vibration of the tympanic membrane and
subsequently, the malleus, incus and stapes. Vibration of the stapes against the oval window allows sound to enter the inner ear and cause fluid wave propagation within the perilymph of
the cochlea. Movement of perilymph leads to deflection of hair cells and conversion of mechanical into electrical energy, which can stimulate neuronal cells along with the auditory
pathway from the spiral ganglion to the auditory cortex. The round window is one of the two openings to access the inner ear from the middle ear and it is sealed by the round
window membrane. The round window membrane is surrounded by a bony pouch called the round window niche. Data collected recently demonstrate that different substances,
including antibiotics, steroids, local anesthetics, and biomaterials can traverse the membrane into perilymph of the inner ear.

can lead to inflammatory changes that may subsequently alter RWM the rate of drug clearance. Salt and Ma administered a marker ion,
permeability for that drug [22]. The optimal delivery system to po- trimehtylphneylammonoium (TMPA) across RWM of guinea pigs to
tentially reduce RWM permeability variations would offer slow and study inner ear drug diffusion, and found that TMPA concentration
sustained drug release to prevent drastic local environmental changes. was mainly determined by its rate of diffusion and its rate of clearance
The hydrogel system, as discussed in Section 1.3, may be able to accom- from the perilymph [29]. Furthermore, they found that TMPA unevenly
plish this goal. distributed throughout the turns of the cochlea, with the highest
Once a drug crosses the RWM into the inner ear, further distribution concentration at the base and the lowest at the apex. Interestingly, addi-
depends upon the rate of drug diffusion within the perilymph and tional studies by Salt suggested that it was not possible to achieve
6 L. Li et al. / Advanced Drug Delivery Reviews 108 (2017) 2–12

uniform drug distribution along the cochlea, and that even prolonged Click chemistry offers stereospecific and efficacious additions of li-
drug administration did not change the concentration gradient [30]. gands to nanoparticles through a Cu I-catalyzed terminal alkyne–azide
While the concentration gradient could not be eliminated, overall cycloaddition. This method has been shown to be successful in the cou-
inner ear drug concentration could be maximized by increasing the pling of peptides and fluorescent tags to SPIONs and lipases to gold NPs
time the drug remains in the middle ear, which increases diffusion rel- [64–66], but there has been little data supporting the addition of larger
ative to clearance [31]. This finding highlights the importance of a proteins or antibodies to NPs using click chemistry. There are additional
sustained drug delivery system that can maintain therapeutic doses of concerns that the click chemistry reaction may degrade or modify the
drug within the inner ear. Moreover, the persistence of the base-apex antibody of interest [65].
concentration gradient generates a need for targeted inner ear delivery To address the shortcomings of EPL and click chemistry approaches,
such that a drug may preferentially bind to apical cells without affecting expressed protein ligation and click chemistry were recently combined
basilar cells. These challenges may be addressed by the hydrogel system into a single system that permits the conjugation of targeting ligands to
and nanotechnology, respectively. nanoparticles in a site-specific and efficient manner [57]. Fig. 2 illus-
trates the chemical mechanism for the combined EPL-click chemistry
3. Nanoparticle delivery system, which produces stereospecific ligand attachment onto nanopar-
ticle surfaces. This approach was recently adapted to also allow full-
Nanoparticles (NPs) have been created from a variety of materials, length antibodies to be conjugated onto nanoparticle surfaces [67].
generally ranging from tens to several hundred nanometers in diameter,
and can be customized to encapsulate various therapeutic agents of in- 5. Applications of nanotechnology to sensorineural hearing loss of
terest [36]. Customization of NP attributes can allow for noninvasive ap- inner ear disease
plication, drug stabilization, controlled release, and surface modification
for specific targeting [37]. Many NP systems have been developed for Sensorineural hearing loss (SNHL) is one of the most common sen-
inner ear drug delivery, including poly(d,l-lactide-co-glycolide acid) sory deficits in humans, affecting millions of people worldwide. Leading
(PLGA) nanoparticles (NPs), magnetic NPs, lipid NPs, liposomes, causes of this type of hearing loss include advanced age, ototoxic
polymersomes, hydroxyapatite NPs, and silica NPs [36]. Local adminis- medications, noise exposure, inherited disorders, and immune disor-
tration of nanoparticles has been shown to successfully deliver biomate- ders. Almost all SNHL is irreversible, and current management is limited
rials into the inner ear via the RWM, although there has been limited to sound amplification devices and cochlear implants. Cochlear and
research on targeted NP delivery into the inner ear [38–41]. retrocochlear pathology are responsible for SNHL. Nanotechnology pro-
Certain classes of nanoparticles exhibit additional characteristics vides the potential for a noninvasive and targeted approach to deliver
that offer unique diagnostic and therapeutic advantages. For example, biomaterials to specific structures within the inner ear for the treatment
superparamagnetic iron oxide nanoparticles (SPIONs), gold nanoparti- of SNHL.
cles, and cationic polymer carriers may be used for bioimaging by MRI,
confocal laser microscopy, two-photon luminescence, etc. [39,42–46]. 5.1. Nanoparticle approach for noise induced hearing loss
The inherent magnetic properties of SPIONs also allows for greater con-
trol of drug delivery and dispersion within the inner ear by application Noise-induced hearing loss (NIHL) often causes permanent inner ear
of an external magnet [42,43,47–49]. Nanoparticles such as cationic damage. The initial cell type affected by NIHL is the outer hair cell, which
polymers and cationic liposomes offer non-viral vector options for modulates incoming auditory signals. Damage to these outer hair cells
gene therapy delivery, which reduces the immunogenicity, inflammatory may alter transduction of sound to the cochlear nerve, therefore causing
responses, and risk of insertional mutagenesis [50–56]. Table 1 describes SNHL.
the current nanoparticle carrier systems available and applications for One drug studied for the treatment of NIHL is the antioxidant
inner ear drug delivery. edaravone. A recent study by Gao et al. looked at nanoparticle delivery
of edaravone in vivo into the inner ear of guinea pigs with NIHL [68].
4. Targeting nanoparticle delivery Edaravone has been used clinically in Japan for treatment of acute cere-
bral infarction via inhibition of hydroxyl free radicals, and studies have
Targeted therapy involves the addition of surface ligands onto nano- demonstrated the utility of edaravone in treating NIHL by the same
particles. The efficacy of these surface ligands depends on their stability mechanism [69,70]. Gao et al. found that IT delivery of solid lipid nano-
and specificity. Bioconjugation allows for the creation of stable peptide particle (SLN)-encapsulated edaravone inhibited free radical generation
bonds, thus anchoring and immobilizing ligands to the nanoparticle in the cochlea and decreased hearing thresholds following noise-
surface. Many bioconjugation techniques have been described in the liter- induced injury, although it did not demonstrate significant hearing re-
ature including maleimide, N-hydroxysuccinimide, and carbodiimide- covery. This may be due to inefficient therapeutic effect and non-
based chemistries [57], but few have been found to consistently and effec- targeted drug delivery to the damaged cell sites.
tively add ligands with the correct orientation, surface density, and site- Using identified biomarkers for targeted delivery can significantly
specificity [58,59]. We will briefly review two more recently developed improve drug delivery efficiency. Prestin is an electromotility protein
techniques, expressed protein ligation (EPL) and click chemistry. that is solely expressed on the surface of outer hair cells (OHCs) [71].
EPL refers to a native, site-specific chemical ligation between a Discovery of prestin as an outer hair cell (OHC) biomarker paved the
recombinant protein (i.e. ligand) with a C-terminal thioester, and a way for targeted OHC drug delivery. Indeed, preliminary data has
peptide or protein with an N-terminal cysteine [60–62]. A C-terminal shown that prestin-targeted nanoparticles have increased specific bind-
thioester can be added onto a targeting ligand through the use of ing to OHCs of mouse cochlear explants. The combination of prestin-
auto-processing proteins called inteins. Inteins are placed between the targeted nanoparticles with edaravone for inner ear delivery may signif-
ligand and an affinity tag. Following bacterial expression and affinity icantly increase delivery efficiency and may lead to better therapeutic
purification, the ligand is released from the affinity tag to create a reac- effect.
tive thioester at the C-terminus. The thioester can then react with any
peptide containing an N-terminal cysteine. This reaction has been 5.2. Nanoparticle approach for different types of SNHL
shown to be highly efficient when both functional groups are at high
concentrations [63], but studies of EPL when applied to nanoparticles Disruption to retrocochlear pathways, from the spiral ganglion cells
have shown less efficacy, which may be due to limited nanoparticle con- (SGCs) to the brain, can also lead to SNHL. Although there is increased
centrations [57]. understanding of auditory neuronal survival biomolecular pathways,
L. Li et al. / Advanced Drug Delivery Reviews 108 (2017) 2–12 7

Table 1
Nanoparticle carrier systems for inner ear delivery.

Nanoparticle (NP) carrier Characteristics Advantages Select publications on NP inner ear delivery
system

Poly(d,l-lactide-co-glycolide Composed of biodegradable polymers Carry hydrophobic and hydrophilic molecules Tamura et al. (2005): local application of PLGA NPs
acid) (PLGA) nanoparticles Rate of degradation is regulated by Sustained release and stabilization of drug in vivo allowed for delivery across the RWM into
[49,83–85] the lactic:glycolic acid ratio and can during delivery the perilymph [38]
promote sustained drug release Wen et al. (2014): PLGA NPs can deliver single and
compound drugs via the RWM into the inner ear
with sustained release and increased local
bioavailability [86]
Superparamagnetic iron oxide Composed of magnetite (Fe3O4) Delivery can be directed by external magnetic Mondalek et al. (2006): SPIONs can cross an
nanoparticles (SPIONs) and/or maghemite (Fe2O3) fields in vitro tripartite model RWM under the influence
[42,48,49,83] May be used for bioimaging by MRI of a magnetic field [42]
Barnes et al. (2007): SPIONs can cross RWMs of
guinea pigs in vivo [43]
Ge et al. (2007): SPIONs embedded within PLGA
NPs can be delivered into the chinchilla cochlea
with wide distribution [39]
Lipid NPs [83,87] Composed of a hydrophobic Carry hydrophobic and hydrophilic molecules Zou et al. (2008): lipid NPs can cross the RWM via a
triglyceride core and an amphiphilic Sustained release and stabilization of drug paracellular mechanism to reach spiral ganglion
surfactant shell during delivery cells, nerve fibers and spiral ligament fibrocytes in
Good for large scale manufacturing an in vivo rat model [40]
Low production cost Chen et al. (2008, 2010): lipid NPs can deliver
dexamethasone via intratympanic injection into
the inner ear of guinea pigs [36,88]
Polymersomes [36,83,89] Amphiphilic block copolymers that Carry hydrophobic and hydrophilic molecules Buckiova (2012): Both liposome and polymersome
self-assemble to form vesicles in an Structurally similar to liposomes but with NPs are capable of carrying a disulfiram, a toxin,
aqueous environment increased drug stabilization and sustained drug into the inner ear to cause spiral ganglion cell death
release with hearing loss measurable by a functional
Controlled drug release by using polymers readout [37]
reactive to specific external stimuli such as pH, See Section 5.2 for targeted polymersome drug
temperature, or magnetic field delivery studies by Roy and Zhang [26,72,73]
Cationic polymers Examples include polyethylenimine Can bind to negatively charged siRNA and DNA Tan (2008): polyethylenimine can transfect the
[50,55,56,90] (PEI), polyamidoamine dendrimer molecules to form polyplexes and be used for perilymph of the cochlea in an in vivo guinea pig
Composed of protonable amino gene delivery model with sustained DNA delivery [50]
nitrogen atoms (“proton sponge”), Higher transfection efficiency than liposomes Praetorius (2008): cationic lipids and dendrimers
organelle-escape units, and May be used for bioimaging can transfect in the area of the organ of Corti in an
degradable fragment in vitro and in vivo mouse model and cause
Allows for release of gene therapy expression of a reporter gene [91]
within endosomes without DNA
degradation
Liposomes [12,44,55–58,92,93] Vesicular structures similar to living Carry hydrophobic and hydrophilic molecules Wareing et al. (1999): intracochlear injection or
cell membranes, formed by Sustained release and stabilization of drug infusion of a liposome-transgene complex can
phospholipid organization into a during delivery transfect cochlear cells with sustained transgene
bilayer structure Low production cost expression in an in vivo guinea pig model [51]
Cationic liposomes created by adding Cationic liposomes can bind to Jero et al. (2001): liposomes complexed with
cationic amine groups to neutral negatively-charged DNA molecules to form reporter humanized green fluorescent protein gene
helper lipids lipoplexes and be used for gene delivery can be delivered across the RWM of mice in vivo
and induce transgene expression within inner ear
structures [52]
Maeda et al. (2007): liposomes complexed with
plasmids expressing GJB2R75W, an allele that
causes deafness, were delivered across on mouse
RWMs, leading to positive transgene expression in
the cochlea. Auditory brainstem response testing
demonstrated that significant hearing loss was seen
post-delivery, illustrating the functional effects of
liposomal transgene delivery on hearing [53,94–96]
Zuris et al. (2014): cationic liposome delivery of
Cre-recombinase and Cas9-based transcription
activators into the mouse inner ear in vivo led to
90% Cre-mediated recombination and 20%
Cas9-mediated genome modification within hair
cells [54]
Gold nanoparticles [44–46,97] Composed of gold atoms Carry hydrophobic and hydrophilic molecules There are no studies in the literature on gold
Plasmon resonance, can be used for imaging nanoparticle delivery into the inner ear to date, but
with confocal laser microscopy, two-photon gold nanoparticles have been successfully used for
luminescence, and optical coherence targeted drug delivery in cancer cells
tomography
Nanohydrogel [21,74] Combination of the CGP-hydrogel Can carry a variety of nanoparticles, which can Lajud (2014): liposomal NPs delivered via
system and nanoparticle carriers encompass both hydrophobic and hydrophilic nanohydrogel in vitro persisted under physiological
molecules conditions for at least 2 weeks, and NPs could be
Significantly more sustained release mechanism detected in the scala media [74]
than individual nanoparticle carriers See Section 4.2 for additional discussion of the
Release can be controlled by the enzyme, nanohydrogel system
chitosanase
8 L. Li et al. / Advanced Drug Delivery Reviews 108 (2017) 2–12

Fluorescent dye
Ligand SCH2CH2SO3- NH 2
(D)(P)(E)(K)(D)(S)(G)(S)
+ SH
O CH
N-terminal cysteine
C-terminal thioester “Clickable” alkyne
Nanoparticle
- +
1. Expressed Protein N N N
Ligation (EPL)

Ligand NH CuSO4 -catalyzed


(D)(P)(E)(K)(D)(S)(G)(S) 2. Click Chemistry
alkyne-azide
cycloaddition
O SH CH
N N Nanoparticle
Ligand NH
(D)(P)(E)(K)(D)(S)(G)(S) N

O SH

Fig. 2. Combined expressed-protein ligation (EPL) and click chemistry bioconjugation. Expressed-protein ligation involves the site-specific reaction between a ligand bound to a C-terminal
thioester and an alkynated fluorescent peptide (AFP) containing an N-terminal cysteine. The AFP contains a “clickable” alkyne that can react with the nanoparticle surface via copper-
catalyzed alkyne–azide cycloaddition (i.e. click chemistry), allowing for stereospecific attachment of the ligand of interest to the nanoparticle.

delivery of biomaterials to targeted sites has remained challenging. delivery system. They have shown that liposomal NPs delivered via
Many current studies are aimed at delivering biomaterials using nano- nanohydrogel in vitro persisted under physiologic conditions for at
technology, and targeting them specifically to these damaged cell least 2 weeks, thus demonstrating sustained release. Furthermore,
sites, for the treatment of SNHL. they found that these NPs could be delivered in vivo across the mouse
One study by Meyer et al. demonstrated that the use of lipid NPs to RWM to reach structures in the scala media [74]. Another unique
deliver rolipram to SGCs in vitro increased neuronal survival and elonga- feature of this delivery approach is that it is noninvasive to the inner
tion of neurite outgrowth via inhibition of phosphodiesterase 4 in a ear, which is crucial for clinical application. The combination of this
dose-dependent manner. They showed that lipid NP-encapsulated nanohydrogel with targeting techniques will potentially be capable of
rolipram led to higher SGC survival rates and neurite outgrowth than delivering therapeutic biomaterials into specific molecular–cellular
rolipram delivery alone [25]. While rolipram may be studied in the fu- sites of the inner for the treatment of SNHL with different pathologies.
ture as a potential therapeutic agent, the findings of this study could Fig. 3 provides a schematic representation of a functionalized nanopar-
not be replicated in vivo, which the authors believe may be due to ticle delivered by the chitosan-glycerophosphate hydrogel system, spe-
non-sustained and nonspecific rolipram delivery. cifically targeted to an outer hair cell.
In order to target neuronal cells, Roy et al. delivered nanoparticles
conjugated with nerve growth factor-derived peptide in an in vitro 5.3. Nanodialysis for cisplatin-induced ototoxicity
inner ear model and found ligand-specific binding to Trks and p75
neurotrophin receptors of the SGCs, Schwann cells and nerve fibers Cisplatin-induced ototoxicity is not uncommon and results in per-
[26]. Trks and p75 neurotrophin receptors are part of the signaling manent inner ear damage. It is a well-known obstacle to the administra-
cascade regulating the survival of vestibular and cochlear neurons. The tion of cisplatin-based chemotherapies, which are commonly used to
potential translational application of NP-based delivery was shown in treat head and neck cancers, along with lung and genitourinary cancers.
a human ex vivo study done by the same group, which showed that Some studies suggest that the rates of cisplatin-induced ototoxicity may
the RWM of freshly frozen temporal bones is permeable to polymeric be as high as 94%, with ototoxicity occurring in one third of patients who
formulations of various sizes and chemical make-ups [72]. Nerve have received a single dose of cisplatin and worsen with further treat-
growth factor derived peptide targeted NP delivery may play a critical ment [75]. It has been shown that cisplatin reaches the inner ear within
role for the treatment of SGC lesion-based SNHL [27,28]. minutes of systemic treatment and that outer hair cells are more severe-
Another study done by Zhang et al. looked at a different targeting ap- ly affected than inner hair cells [76]. There are currently no treatments
proach to SGCs and cochlear nerve [73]. They delivered polymersome- to prevent or reverse cisplatin-induced ototoxicity. Hearing loss that
nanoparticles conjugated with the Tet1 peptide into the inner ear results from cisplatin-induced ototoxicity is irreversible; therefore, pro-
through both IT injection and cochleostomy. They found that this pep- phylactic or early treatment is a crucial component for optimized treat-
tide binds specifically to trisialoganglioside clostridial toxin receptors ment regimens.
on SGCs and the cochlear nerve. Prevention of cisplatin-induced ototoxicity has been demonstrated
Interestingly, they found that a cochleostomy approach worked for with reducing agents, such as glutathione, which decreases oxidative
targeted delivery to the SGCs and cochlear nerve while IT injection did damage to the inner ear by free radicals [77–79]. Glutathione and
not. Much like the Meyer study, these authors concluded that an IT in- other reducing agents have been studied using systemic and IT delivery
jection approach could not efficiently deliver the nanoparticles across before cisplatin administration and have been shown to decrease hear-
the RWM to the SGCs and cochlear nerve of the inner ear. Although a ing loss. However, not only are these drugs limited by inefficient inner
cochleostomy approach demonstrated nanoparticle delivery to the ear delivery, they cannot be removed from the inner ear once they are
SGCs and auditory nerve, this approach is an invasive procedure to the complexed with the toxin. Accumulation of these drug-toxin complexes
inner ear and it may worsen the existing pathology. may cause secondary toxicity to the inner ear.
In summary, although different targeted nanoparticle delivery Nanotechnology may offer a noninvasive solution to the treatment
systems demonstrate promising results, the major limitations of non- of cisplatin-induced ototoxicity. As mentioned in Table 1, biologically
sustained and/or invasive inner ear delivery hinder translational appli- coated SPIONs are able to traverse the RWM for delivery into the
cation. To address this problem, Li et al. has proposed a nanohydrogel inner ear. More importantly, their delivery can be controlled by the ap-
delivery system, which combines nanotechnology with a hydrogel plication of an external magnetic field. Subsequent removal of the
L. Li et al. / Advanced Drug Delivery Reviews 108 (2017) 2–12 9

Fig. 3. Nanohydrogel delivery into the inner ear. Schematic overview of targeted nanoparticle delivery using a chitosan-glycerophosphate (CGP) hydrogel system (nanohydrogel) to bring
therapeutic biomaterials to specific cellular structures within the inner ear. A schematic diagram of a functionalized NP is shown, with targeting ligands on the surface that can specifically
bind to prestin, an electromotility protein solely expressed on outer hair cells.

magnet leads to dispersion by Brownian motion such that these SPIONs biomaterials across the round window membrane into the inner ear in
can distribute evenly throughout the perilymph. Furthermore, in vitro models, although with more limited success in in vivo animal
pegylated coating of SPIONs can accommodate bioconjugation of models.
surface ligands such as glutathione, which has been shown to bind Furthermore, the development of the combined EPL-click chemistry
the desired target: cisplatin [80,81]. Once the SPIONs bind to cisplat- bioconjugation system allows for ligands to be effectively and stereo-
in, the external magnet can be redirected to remove the complexes specifically attached onto nanoparticle surfaces, creating a mechanism
from the inner ear. This process, as illustrated in Fig. 4, is known as for targeted delivery. Recent studies of specific inner ear biomarkers
nanodialysis. and pathways, such as prestin and the neurotrophin family receptors
Preliminary experiments studying the glutathione-SPION delivery have shown promise for targeted therapy of sensorineural hearing
system have been promising. In vivo experiments confirmed that fluo- loss. Future research may focus on even more site-specific inner ear
rescently tagged SPIONs can be passed through the RWM into the delivery systems. It has already been shown that distinct types of
inner ear and retrieved from the cochlea using an external magnet. voltage-gated potassium channels are expressed in a gradient manner
Furthermore, glutathione-bound agarose magnetic beads have been from the base to the apex in mice cochlea, and that these ion channel ex-
successful at sequestering cisplatin. Future experiments will include pressions are controlled by brain-derived neurotropic factor (BDNF)
conjugation of glutathione to SPIONs for delivery into the inner ear for and neurotrophin-3 (NT-3). A nanoparticle delivery system could be
cisplatin-binding and subsequent removal using an external magnet. used to deliver these neurotrophic factors or even ion channel proteins
and transgenes in the cochlea in vivo [82]. Being able to target specific
6. Conclusion and future directions sites and cell types of the cochlea, may translate to distinct treatments
for high frequency or low frequency SNHL, which may prevent damage
Efficient targeted inner ear delivery is the key for successful treat- to non-pathological areas and cells.
ment of sensorineural hearing loss of different pathologies. To date, Although there have been successful attempts at targeted nanopar-
there is a lack of an efficient, noninvasive and sustained delivery system ticle delivery of biomaterials to the inner ear in vitro, the findings have
that can both stabilize therapeutic biomaterials and target them to spe- not been consistent in vivo. As discussed above, the Meyer study was
cific structures within the inner ear. only able to deliver rolipram in vitro, not in vivo, and the Zhang study
Modern nanotechnology offers a promising tool for efficient and was only able to deliver the Tet1 peptide via invasive cochleostomy,
noninvasive inner ear drug and biomaterial delivery. Nanoparticle not intratympanic injection. The findings highlight the underlying
systems such as PLGA nanoparticles, lipid nanoparticles, liposomes, limitations of current targeted nanoparticle delivery systems: non-
and polymersomes have been shown to carry a variety of drugs and sustained drug release and invasive approaches for delivery. One
10 L. Li et al. / Advanced Drug Delivery Reviews 108 (2017) 2–12

Fig. 4. Application of nanodialysis system for potential treatment of cisplatin-induced ototoxicity. Glutathione-conjugated polyethylene glycol-polycaprolactone SPIO-encapsulated
nanoparticles (GPSNs) are delivered into the inner ear using chitosan-glycerophosphate (CPG) hydrogel. GPSNs disperse within the fluid compartments of the inner ear to sequester
cisplatin particles. An external magnet is applied to draw out the cisplatin-GPSN complexes and chitosanase is added to degrade the CPG hydrogel, thus removing the complexes from
the inner ear by elimination through the Eustachian tube.

possible way to resolve these issues is to use the nanohydrogel delivery Acknowledgments
system, as the hydrogel provides sustained release of biomaterials
across the RWM noninvasively. Studies on the use of the nanohydrogel This work was funded through a National Institute of Health (NIH)
system for targeted therapeutic delivery into the inner ear are currently grant: 1R01DC014464-01.
underway.
There is still much to learn about nanoparticles beyond their use as
effective carrier and targeting agents. Certain classes of nanoparticles References
display additional properties that may be used in the future for the
[1] A.A. McCall, E.E. Swan, J.T. Borenstein, W.F. Sewell, S.G. Kujawa, M.J. McKenna, Drug
treatment of cisplatin-induced ototoxicity. As discussed, SPIONs have delivery for treatment of inner ear disease: current state of knowledge, Ear Hear. 31
magnetic properties that allow their motion to be controlled by an (2010) 156–165, http://dx.doi.org/10.1097/AUD.0b013e3181c351f2.
applied external magnetic field. This property can be used for the treat- [2] W.R. Wilson, F.M. Byl, N. Laird, The efficacy of steroids in the treatment of idiopathic
sudden hearing loss. A double-blind clinical study, Arch. Otolaryngol. 106 (1980)
ment of cisplatin-induced ototoxicity, which involves removing cisplat- 772–776, http://dx.doi.org/10.1001/archotol.1980.00790360050013.
in particles that have accumulated within the inner ear. Current studies [3] W. Chen, J. Lee, C. Yuan, R. Chen, Oral steroid treatment for idiopathic sudden sen-
are aimed at attaching glutathione, a ligand known to bind to cisplatin, sorineural hearing loss, Saudi Med. J. 36 (2015) 291–296, http://dx.doi.org/10.
15537/smj.2015.3.9940.
onto SPIONs for delivery into the inner ear. The glutathione will bind to [4] R. Filipo, G. Attanasio, F.Y. Russo, G. Cartocci, A. Musacchio, A. De Carlo, et al., Oral
cisplatin within the inner ear, and the SPION-glutathione-cisplatin com- versus short-term intratympanic prednisolone therapy for idiopathic sudden hear-
plex can subsequently be retrieved using an external magnet. ing loss, Audiol. Neurootol. 19 (2014) 225–233, http://dx.doi.org/10.1159/
000360069.
Taken together, the current research on targeted nanohydrogel de-
[5] M.J. Ruckenstein, Autoimmune inner ear disease, Curr. Opin. Otolaryngol. Head Neck
livery and nanodialysis are quite promising with strong translational Surg. 12 (2004) 426–430, http://dx.doi.org/10.1097/01.moo.0000136101.95662.aa.
potential. We predict in the future that the targeted nanohydrogel deliv- [6] M.C. Buniel, K. Geelan-Hansen, P.C. Weber, V.K. Tuohy, Immunosuppressive therapy
ery system can effectively and noninvasively deliver therapeutic drugs for autoimmune inner ear disease, Immunotherapy 1 (2009) 425–434, http://dx.
doi.org/10.2217/imt.09.12.
and biomaterials to specific pathologic sites and cell types of the inner [7] T.H. Alexander, M.H. Weisman, J.M. Derebery, M.A. Espeland, B.J. Gantz, A.J. Gulya,
ear. As a consequence, it may repair defective cells and molecules in et al., Safety of high-dose corticosteroids for the treatment of autoimmune inner
specific areas of the inner ear responsible for SNHL. We hope that the ear disease, Otol. Neurotol. 30 (2009) 443–448, http://dx.doi.org/10.1097/MAO.
0b013e3181a52773.
nanodialysis system can be effectively used for detoxification of differ- [8] M.D. Graham, R. Sataloff, J. Kemink, Titration streptomycin therapy for bilateral
ent types of ototoxicity. This may greatly benefit patients who cannot Meniere's disease : A preliminary report, 921984 440–447.
compromise their critical treatment modalities despite the high poten- [9] F.R. Balyan, A. Taibah, D.G. De, A. Aslan, M. Falcioni, A. Russo, et al., Titration strepto-
mycin therapy in meniere's disease: long-term results, Otolaryngol. Head Neck Surg.
tial for ototoxicity. Future progress can be achieved by combining mod- 118 (1998) 261–266 (doi:S0194599898000291 [pii]).
ern nanotechnology with advancing knowledge of the biomolecular [10] E. Selimoglu, Aminoglycoside-induced ototoxicity, Curr. Pharm. Des. 13 (2007)
basis of sensorineural hearing loss. 119–126, http://dx.doi.org/10.2174/138161207779313731.
L. Li et al. / Advanced Drug Delivery Reviews 108 (2017) 2–12 11

[11] A.W. Langman, J.L. Kemink, M.D. Graham, Titration streptomycin therapy for bilateral [40] J. Zou, P. Saulnier, T. Perrier, Y. Zhang, T. Manninen, E. Toppila, et al., Distribution of
Meniere's disease. Follow-up report, Ann. Otol. Rhinol. Laryngol. 99 (1990) 923–926. lipid nanocapsules in different cochlear cell populations after round window mem-
[12] H.F. Schuknecht, Ablation therapy in the management of menière's disease, Acta brane permeation, J. Biomed. Mater. Res. B Appl. Biomater. 87 (2008) 10–18, http://
Otolaryngol. Suppl. 132 (1957) 1–42. dx.doi.org/10.1002/jbm.b.31058.
[13] L.E. Jackson, H. Silverstein, Chemical perfusion of the inner ear, Otolaryngol. Clin. N. [41] M. Praetorius, C. Brunner, B. Lehnert, C. Klingmann, H. Schmidt, H. Staecker,
Am. 35 (2002) 639–653, http://dx.doi.org/10.1016/S0030-6665(02)00023-3. et al., Transsynaptic delivery of nanoparticles to the central auditory nervous
[14] P.P. Lefebvre, H. Staecker, Steroid perfusion of the inner ear for sudden sensorineural system, Acta Otolaryngol. 127 (2007) 486–490, http://dx.doi.org/10.1080/
hearing loss after failure of conventional therapy: a pilot study, Acta Otolaryngol. 00016480600895102.
122 (2002) 698–702 (http://www.ncbi.nlm.nih.gov/pubmed/12484644). [42] F.G. Mondalek, Y.Y. Zhang, B. Kropp, R.D. Kopke, X. Ge, R.L. Jackson, et al., The perme-
[15] S.D. Rauch, C.F. Halpin, P.J. Antonelli, S. Babu, J.P. Carey, B.J. Gantz, et al., Oral vs ability of SPION over an artificial three-layer membrane is enhanced by external
intratympanic corticosteroid therapy for idiopathic sudden sensorineural hearing magnetic field, J. Nanobiotechnol. 4 (2006) 4, http://dx.doi.org/10.1186/1477-
loss: a randomized trial, J. Am. Med. Assoc. 305 (2011) 2071–2079, http://dx.doi. 3155-4-4.
org/10.1001/jama.2011.679. [43] A.L. Barnes, R.a. Wassel, F. Mondalek, K. Chen, K.J. Dormer, R.D. Kopke, Magnetic
[16] H. Silverstein, S.M. Hyman, J. Feldbaum, D. Silverstein, Use of streptomycin sulfate in characterization of superparamagnetic nanoparticles pulled through model mem-
the treatment of meniere's disease, Otolaryngol. Neck Surg. 92 (1984) 229–232. branes, Biomagn. Res. Technol. 5 (2007) 1, http://dx.doi.org/10.1186/1477-044X-
[17] W.E. Berryhill, M.D. Graham, Chemical and physical labyrinthectomy for meniere's 5-1.
disease, Otolaryngol. Clin. N. Am. 35 (2002) 675–682, http://dx.doi.org/10.1016/ [44] I.H. El-Sayed, X. Huang, M.A. El-Sayed, Surface plasmon resonance scattering and ab-
S0030-6665(02)00025-7. sorption of anti-EGFR antibody conjugated gold nanoparticles in cancer diagnostics:
[18] S.H. Chia, A.C. Gamst, J.P. Anderson, J.P. Harris, Intratympanic gentamicin therapy for applications in oral cancer, Nano Lett. 5 (2005) 829–834, http://dx.doi.org/10.1021/
ménière's disease: a meta-analysis, Otol. Neurotol. 25 (2004) 544–552, http://dx. nl050074e.
doi.org/10.1097/00129492-200407000-00023. [45] H. Daraee, A. Eatemadi, E. Abbasi, S. Fekri Aval, M. Kouhi, A. Akbarzadeh, Application
[19] D.P. Paulson, W. Abuzeid, H. Jiang, T. Oe, B.W. O'Malley, D. Li, A novel controlled local of gold nanoparticles in biomedical and drug delivery, Artif. Cells Nanomed.
drug delivery system for inner ear disease, Laryngoscope 118 (2008) 706–711, Biotechnol. (2014) 1–13, http://dx.doi.org/10.3109/21691401.2014.955107.
http://dx.doi.org/10.1097/MLG.0b013e31815f8e41. [46] N. Nitin, D.J. Javier, R. Richards-Kortum, Oligonucleotide-coated metallic nanoparti-
[20] L. Xu, J. Heldrich, H. Wang, T. Yamashita, S. Miyamoto, A. Li, et al., A controlled and cles as a flexible platform for molecular imaging agents, Bioconjug. Chem. 18 (2007)
sustained local gentamicin delivery system for inner ear applications, Otol. Neurotol. 2090–2096, http://dx.doi.org/10.1021/bc0701242.
31 (2010) 1115–1121, http://dx.doi.org/10.1097/MAO.0b013e3181eb32d1. [47] B. Shapiro, S. Kulkarni, a. Nacev, a. Sarwar, D. Preciado, D.a. Depireux, Shaping mag-
[21] S.a. Lajud, Z. Han, F.L. Chi, R. Gu, D.a. Nagda, O. Bezpalko, et al., A regulated delivery netic fields to direct therapy to ears and eyes, Annu. Rev. Biomed. Eng. 16 (2014)
system for inner ear drug application, J. Control. Release 166 (2013) 268–276, 455–481, http://dx.doi.org/10.1146/annurev-bioeng-071813-105206.
http://dx.doi.org/10.1016/j.jconrel.2012.12.031. [48] R.D. Kopke, R.a. Wassel, F. Mondalek, B. Grady, K. Chen, J. Liu, et al., Magnetic nano-
[22] A. Banerjee, L.S. Parnes, The biology of intratympanic drug administration and phar- particles: Inner ear targeted molecule delivery and middle ear implant, Audiol.
macodynamics of round window drug absorption, Otolaryngol. Clin. N. Am. 37 Neurootol. 11 (2005) 123–133, http://dx.doi.org/10.1159/000090685.
(2004) 1035–1051, http://dx.doi.org/10.1016/j.otc.2004.04.003. [49] X. Du, K. Chen, S. Kuriyavar, R.D. Kopke, B.P. Grady, D.H. Bourne, et al., Magnetic
[23] P. Flint, B. Haughley, V. Lund, J. Niparko, M. Richardson, K.T. Robbins, et al., Cum- targeted delivery of dexamethasone acetate across the round window membrane
mings Otolaryngology: Head and Neck Surgery, sixth ed. Saunders, 2015. in guinea pigs, Otol. Neurotol. 34 (2013) 41–47, http://dx.doi.org/10.1097/MAO.
[24] R. Pasha, Otolaryngology — Head and Neck Surgery: Clinical Reference Guide, fourth 0b013e318277a40e.
ed. Plural Publishing, 2013. [50] B.T. Tan, K.H. Foong, M.M. Lee, R. Ruan, Polyethylenimine-mediated cochlear gene
[25] H. Meyer, T. Stöver, F. Fouchet, G. Bastiat, P. Saulnier, W. Bäumer, et al., Lipidic transfer in guinea pigs, Arch. Otolaryngol. Head Neck Surg. 134 (2008) 884–891
nanocapsule drug delivery: neuronal protection for cochlear implant optimization, (http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=
Int. J. Nanomedicine 7 (2012) 2449–2464, http://dx.doi.org/10.2147/IJN.S29712. PubMed&dopt=Citation&list_uids=18711065).
[26] S. Roy, A.H. Johnston, T.a. Newman, R. Glueckert, J. Dudas, M. Bitsche, et al., [51] M. Wareing, A.N. Mhatre, R. Pettis, J.J. Han, T. Haut, M.H.F. Pfister, et al., Cationic li-
Cell-specific targeting in the mouse inner ear using nanoparticles conjugated posome mediated transgene expression in the guinea pig cochlea, Hear. Res. 128
with a neurotrophin-derived peptide ligand: Potential tool for drug delivery, (1999) 61–69, http://dx.doi.org/10.1016/S0378-5955(98)00196-8.
Int. J. Pharm. 390 (2010) 214–224, http://dx.doi.org/10.1016/j.ijpharm.2010. [52] J. Jero, a.N. Mhatre, C.J. Tseng, R.E. Stern, D.E. Coling, J.a. Goldstein, et al., Cochlear
02.003. gene delivery through an intact round window membrane in mouse, Hum. Gene
[27] D.R. Kaplan, F.D. Miller, Neurotrophin signal transduction in the nervous system, Ther. 12 (2001) 539–548, http://dx.doi.org/10.1089/104303401300042465.
Curr. Opin. Neurobiol. 10 (2000) 381–391, http://dx.doi.org/10.1016/S0959- [53] Y. Maeda, K. Fukushima, A. Kawasaki, K. Nishizaki, R.J.H. Smith, Cochlear expression
4388(00)00092-1. of a dominant-negative GJB2R75W construct delivered through the round window
[28] B. Fritzsch, K.W. Beisel, L.A. Hansen, The molecular basis of neurosensory cell forma- membrane in mice, Neurosci. Res. 58 (2007) 250–254, http://dx.doi.org/10.1016/j.
tion in ear development: a blueprint for hair cell and sensory neuron regeneration? neures.2007.03.006.
BioEssays 28 (2006) 1181–1193, http://dx.doi.org/10.1002/bies.20502. [54] J.a. Zuris, D.B. Thompson, Y. Shu, J.P. Guilinger, J.L. Bessen, J.H. Hu, et al., Cationic
[29] A.N. Salt, Y. Ma, Quantification of solute entry into cochlear perilymph through the lipid-mediated delivery of proteins enables efficient protein-based genome editing
round window membrane, Hear. Res. 154 (2001) 88–97, http://dx.doi.org/10.1016/ in vitro and in vivo, Nat. Biotechnol. 33 (2014) 73–80, http://dx.doi.org/10.1038/
S0378-5955(01)00223-4. nbt.3081.
[30] S.K.R. Plontke, A.N. Salt, Quantitative interpretation of corticosteroid pharmacoki- [55] P. Neuberg, A. Kichler, Recent Developments in Nucleic Acid Delivery with
netics in inner fluids using computer simulations, Hear. Res. 182 (2003) 34–42, Polyethylenimines, Elsevier, 2014http://dx.doi.org/10.1016/B978-0-12-800148-6.
http://dx.doi.org/10.1016/S0378-5955(03)00138-2. 00009-2.
[31] A.N. Salt, S.K.R. Plontke, Local inner-ear drug delivery and pharmacokinetics, [56] E. Wagner, Polymers for Nucleic Acid Transfer — An Overview, Elsevier, 2014http://
Drug Discov. Today 10 (2005) 1299–1306, http://dx.doi.org/10.1016/S1359- dx.doi.org/10.1016/B978-0-12-800148-6.00008-0.
6446(05)03574-9. [57] D.R. Elias, Z. Cheng, A. Tsourkas, An intein-mediated site-specific click conjugation
[32] M.V. Goycoolea, Clinical aspects of round window membrane permeability under strategy for improved tumor targeting of nanoparticle systems, Small 6 (2010)
normal and pathological conditions, Acta Otolaryngol. 121 (2001) 437–447, 2460–2468, http://dx.doi.org/10.1002/smll.201001095.
http://dx.doi.org/10.1080/000164801300366552. [58] R. Warden-Rothman, I. Caturegli, V. Popik, A. Tsourkas, Sortase-tag expressed pro-
[33] S.S.S.S.S. Chandrasekhar, R.Y. Rubinstein, J.a. Kwartler, M. Gatz, P.E. Connelly, E. tein ligation: combining protein purification and site-specific bioconjugation into
Huang, et al., Dexamethasone pharmacokinetics in the inner ear: comparison of a single step, Anal. Chem. 85 (2013) 11090–11097, http://dx.doi.org/10.1021/
route of administration and use of facilitating agents, Otolaryngol. Head Neck ac402871k.
Surg. 122 (2000) 521–528, http://dx.doi.org/10.1067/mhn.2000.102578. [59] A.D. Friedman, S.E. Claypool, R. Liu, The smart targeting of nanoparticles, Curr. Pharm.
[34] P.A. Schachern, M.M. Paparella, M.V. Goycoolea, A.J. Duvuall, Y.-B. Choo, The perme- Des. 19 (2013) 6315–6329, http://dx.doi.org/10.2174/13816128113199990375.
ability of the round window membrane during otitis media, Otolaryngol. Head Neck [60] L. Berrade, J.a. Camarero, Expressed protein ligation: a resourceful tool to study pro-
Surg. 113 (1987) 625–629 (http://www.ncbi.nlm.nih.gov/pubmed/3566945). tein structure and function, Cell. Mol. Life Sci. 66 (2009) 3909–3922, http://dx.doi.
[35] L. Nordang, B. Linder, M. Anniko, Morphologic changes in round window membrane org/10.1007/s00018–009-0122-3.
after topical hydrocortisone and dexamethasone treatment, Otol. Neurotol. 24 [61] R.R. Flavell, T.W. Muir, Expressed protein ligation (EPL) in the study of signal trans-
(2003) 339–343. duction, ion conduction, and chromatin biology, Acc. Chem. Res. 42 (2009) 107–116,
[36] G. Chen, X. Zhang, F. Yang, L. Mu, Disposition of nanoparticle-based delivery system http://dx.doi.org/10.1021/ar800129c.
via inner ear administration, Curr. Drug Metab. 11 (2010) 886–897, http://dx.doi. [62] K. Severinov, T.W. Muir, Expressed protein ligation, a novel method for studying
org/10.2174/138920010794479673. protein–protein interactions in transcription, J. Biol. Chem. 273 (1998)
[37] D. Buckiova, S. Ranjan, T.a. Newman, A.H. Johnston, R. Sood, P.K.J. Kinnunen, et al., 16205–16209, http://dx.doi.org/10.1074/jbc.273.26.16205.
Minimally invasive drug delivery to the cochlea through application of nanoparti- [63] V. Muralidharan, T.W. Muir, Protein ligation: an enabling technology for the bio-
cles to the round window membrane, 2012 1339–1354, http://dx.doi.org/10.2217/ physical analysis of proteins, Nat. Methods 3 (2006) 429–438, http://dx.doi.org/
nnm.12.5. 10.1038/nmeth886.
[38] T. Tamura, T. Kita, T. Nakagawa, T. Endo, T.-S. Kim, T. Ishihara, et al., Drug delivery to [64] H.C. Kolb, M.G. Finn, K.B. Sharpless, Click chemistry: diverse chemical function from
the cochlea using PLGA nanoparticles, Laryngoscope 115 (2005) 2000–2005, http:// a few good reactions, Angew. Chem. Int. Ed. 40 (2001) 2004–2021, http://dx.doi.
dx.doi.org/10.1097/01.mlg.0000180174.81036.5a. org/10.1002/1521-3773(20010601)40:11b2004::AID-ANIE2004N3.0.CO;2-5.
[39] X. Ge, R.L. Jackson, J. Liu, E.a. Harper, M.E. Hoffer, R.a. Wassel, et al., Distribution of [65] D.L.J. Thorek, D.R. Elias, A. Tsourkas, Comparative analysis of nanoparticle-
PLGA nanoparticles in chinchilla cochleae, Otolaryngol. Head Neck Surg. 137 antibody conjugations: carbodiimide versus click chemistry, Mol. Imaging 8
(2007) 619–623, http://dx.doi.org/10.1016/j.otohns.2007.04.013. (2009) 221–229, http://dx.doi.org/10.2310/7290.2009.00021.
12 L. Li et al. / Advanced Drug Delivery Reviews 108 (2017) 2–12

[66] J.L. Brennan, N.S. Hatzakis, T.R. Tshikhudo, N. Dirvianskyte, V. Razumas, S. Patkar, [81] T. Ishikawa, F. Ali-Osman, Glutathione-associated cis-diamminedichloroplatinum(II)
et al., Bionanoconjugation via click chemistry: the creation of functional hybrids of metabolism and ATP-dependent efflux from leukemia cells: molecular characteriza-
lipases and gold nanoparticles, Bioconjug. Chem. 17 (2006) 1373–1375, http://dx. tion of glutathione-platinum complex and its biological significance, J. Biol. Chem.
doi.org/10.1021/bc0601018. 268 (1993) 20116–20125.
[67] J.Z. Hui, A. Al Zaki, Z. Cheng, V. Popik, H. Zhang, E.T. Luning Prak, et al., Facile method [82] R.L. Davis, Gradients of neurotrophins, ion channels, and tuning in the cochlea, Neu-
for the site-specific, covalent attachment of full-length IgG onto nanoparticles, Small roscientist 9 (2003) 311–316, http://dx.doi.org/10.1177/1073858403251986.
(2014) 1–10, http://dx.doi.org/10.1002/smll.201303629. [83] C.O. Pritz, J. Dudás, H. Rask-Andersen, A. Schrott-Fischer, R. Glueckert, Nanomedicine
[68] J. Sun, G. Gao, Y. Liu, C. Zhou, P. Jiang, Solid Lipid nanoparticles loaded with strategies for drug delivery to the ear, Nanomedicine (London) 8 (2013) 1155–1172,
edaravone for inner ear protection after noise exposure, Chin. Med. J. (Engl.) 128 http://dx.doi.org/10.2217/nnm.13.104.
(2015) 203, http://dx.doi.org/10.4103/0366–6999.149202. [84] T. Ishihara, N. Izumo, M. Higaki, E. Shimada, T. Hagi, L. Mine, et al., Role of zinc in for-
[69] T. Yamamoto, S. Yuki, T. Watanabe, M. Mitsuka, K.I. Saito, K. Kogure, Delayed neuro- mulation of PLGA, J. Control. Release 105 (2005) 68–76.
nal death prevented by inhibition of increased hydroxyl radical formation in a tran- [85] J. Panyam, D. William, A. Dash, D. Leslie-Pelecky, V. Labhasetwar, Solid-state solubil-
sient cerebral ischemia, Brain Res. 762 (1997) 240–242. ity influences encapsulation and release of hydrophobic drugs from PLGA/PLA nano-
[70] T. Takemoto, K. Sugahara, T. Okuda, H. Shimogori, H. Yamashita, The clinical free particles, J. Pharm. Sci. 93 (2004) 1804–1814, http://dx.doi.org/10.1002/jps.20094.
radical scavenger, edaravone, protects cochlear hair cells from acoustic trauma, [86] X. Wen, L. Wen, N. Tirelli, H. Su, G. Chen, Enhanced Local Bioavailability of Single or
Eur. J. Pharmacol. 487 (2004) 113–116, http://dx.doi.org/10.1016/j.ejphar.2004.01. Compound Drugs Delivery to the Inner Ear Through Application of PLGA Nanoparti-
019. cles Via Round Window Administration, 2014 5591–5601.
[71] E.V. Surovtseva, A.H. Johnston, W. Zhang, Y. Zhang, A. Kim, M. Murakoshi, et al., [87] V. Scheper, M. Wolf, M. Scholl, Z. Kadlecova, T. Perrier, H.-A. Klok, et al., Potential
Prestin binding peptides as ligands for targeted polymersome mediated drug deliv- novel drug carriers for inner ear treatment: hyperbranched polylysine and lipid
ery to outer hair cells in the inner ear, Int. J. Pharm. 424 (2012) 121–127, http://dx. nanocapsules, Nanomedicine (London) 4 (2009) 623–635, http://dx.doi.org/10.
doi.org/10.1016/j.ijpharm.2011.12.042. 2217/nnm.09.41.
[72] S. Roy, R. Glueckert, A.H. Johnston, T. Perrier, T.a. Newman, P. Saulnier, et al., Strate- [88] G. Chen, S.-X. Hou, P. Hu, Q.-H. Hu, D.-D. Guo, Y. Xiao, In vitro dexamethasone re-
gies for drug delivery to the human inner ear by multifunctional nanoparticles, 7 lease from nanoparticles and its pharmacokinetics in the inner ear after administra-
(2012) 55–63, http://dx.doi.org/10.2217/nnm.11.84. tion of the drug-loaded nanoparticles via the round window, Nan Fang Yi Ke Da Xue
[73] Y. Zhang, W. Zhang, A.H. Johnston, T.a. Newman, I. Pyykkö, J. Zou, Targeted delivery Xue Bao 28 (2008) 1022–1024.
of Tet1 peptide functionalized polymersomes to the rat cochlear nerve, Int. J. [89] J.S. Lee, J. Feijen, Polymersomes for drug delivery: design, formation and character-
Nanomedicine 7 (2012) 1015–1022, http://dx.doi.org/10.2147/IJN.S28185. ization, J. Control. Release 161 (2012) 473–483, http://dx.doi.org/10.1016/j.jconrel.
[74] S.a. Lajud, D.a. Nagda, P. Qiao, N. Tanaka, A. Civantos, R. Gu, et al., A novel chitosan- 2011.10.005.
hydrogel-based nanoparticle delivery system for local inner ear application, Otol. [90] L. Jin, X. Zeng, M. Liu, Y. Deng, N. He, Current progress in gene delivery technology
Neurotol. (2014) 341–347, http://dx.doi.org/10.1097/MAO.0000000000000445. based on chemical methods and nano-carriers, Theranostics 4 (2014) 240–255,
[75] S.A. Fausti, J.A. Henry, H.I. Schaffer, D.J. Olson, R.H. Frey, G.C. Bagby, High- http://dx.doi.org/10.7150/thno.6914.
frequency monitoring for early detection of cisplatin ototoxicity, Arch. Otolaryngol. [91] M. Praetorius, S. Pfannenstiel, C. Klingmann, I. Baumann, P.K. Plinkert, H. Staecker,
Head Neck Surg. 119 (1993) 661–666, http://dx.doi.org/10.1001/archotol.1993. Expressionsmuster für nichtviral transfiziertes gfp in der cochlea der maus in vitro
01880180081015. und in vivo, HNO 56 (2008) 524–529, http://dx.doi.org/10.1007/s00106-008-1738-6.
[76] A. Tropitzsch, H. Arnold, M. Bassiouni, A. Müller, A. Eckhard, M. Müller, et al., [92] D.a. Balazs, W. Godbey, Liposomes for use in gene delivery, J. Drug Deliv. 2011
Assessing cisplatin-induced ototoxicity and otoprotection in whole organ culture (2011) 326497, http://dx.doi.org/10.1155/2011/326497.
of the mouse inner ear in simulated microgravity, Toxicol. Lett. 227 (2014) [93] A.K. Lalwani, J. Jero, A.N. Mhatre, Current issues in cochlear gene transfer, Audiol.
203–212, http://dx.doi.org/10.1016/j.toxlet.2014.03.022. Neurootol. 7 (2002) 146–151, http://dx.doi.org/10.1159/000058300.
[77] C.E. Berglin, P.V. Pierre, T. Bramer, K. Edsman, H. Ehrsson, S. Eksborg, et al., Preven- [94] D.P. Kelsell, J. Dunlop, H.P. Stevens, N.J. Lench, J.N. Liang, G. Parry, et al., Connexin 26
tion of cisplatin-induced hearing loss by administration of a thiosulfate-containing mutations in hereditary non-syndromic sensorineural deafness, Nature 387 (1997)
gel to the middle ear in a guinea pig model, Cancer Chemother. Pharmacol. 68 80–83, http://dx.doi.org/10.1038/387080a0.
(2011) 1547–1556, http://dx.doi.org/10.1007/s00280-011-1656-2. [95] F. Denoyelle, D. Weil, M.A. Maw, S.A. Wilcox, N.J. Lench, D.R. Allen-Powell, et al.,
[78] D.T. Dickey, Y.J. Wu, L.L. Muldoon, E.A. Neuwelt, Protection against cisplatin-induced Prelingual deafness: high prevalence of a 30delG mutation in the connexin 26
toxicities by N-acetylcysteine and sodium thiosulfate as assessed at the molecular, gene, Hum. Mol. Genet. 6 (1997) 2173–2177, http://dx.doi.org/10.1093/hmg/6.12.
cellular, and in vivo levels, J. Pharmacol. Exp. Ther. 314 (2005) 1052–1058, http:// 2173.
dx.doi.org/10.1124/jpet.105.087601.the. [96] T. Kudo, S. Kure, K. Ikeda, A.P. Xia, Y. Katori, M. Suzuki, et al., Transgenic expression
[79] K.C.M. Campbell, L.P. Rybak, R.P. Meech, L. Hughes, D-methionine provides excellent of a dominant-negative connexin26 causes degeneration of the organ of corti and
protection from cisplatin ototoxicity in the rat, Hear. Res. 102 (1996) 90–98, http:// non-syndromic deafness, Hum. Mol. Genet. 12 (2003) 995–1004, http://dx.doi.
dx.doi.org/10.1016/S0378-5955(96)00152-9. org/10.1093/hmg/ddg116.
[80] D. Hagrman, J. Goodisman, J.C. Dabrowiak, A.K. Souid, Kinetic study on the reaction [97] K. Sokolov, M. Follen, J. Aaron, I. Pavlova, A. Malpica, R. Lotan, et al., Real-time vital
of cisplatin with metallothionein, Drug Metab. Dispos. 31 (2003) 916–923, http:// optical imaging of precancer using anti-epidermal growth factor receptor antibodies
dx.doi.org/10.1124/dmd.31.7.916. conjugated to gold nanoparticles, Cancer Res. 63 (2003) 1999–2004.

You might also like