You are on page 1of 16

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/324565100

Otic Drug Delivery Systems: Formulation Principles and Recent Developments

Article  in  Drug Development and Industrial Pharmacy · April 2018


DOI: 10.1080/03639045.2018.1464022

CITATIONS READS

15 20,318

4 authors, including:

Xu Liu Mingshuang Li
University of Texas at Austin California State University, Northridge
11 PUBLICATIONS   425 CITATIONS    19 PUBLICATIONS   56 CITATIONS   

SEE PROFILE SEE PROFILE

Hugh David Charles Smyth


University of Texas at Austin
248 PUBLICATIONS   4,751 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Psychophysical mechanisms for the Mandarin congenital amusics View project

Control of ciprofloxacin lung residency time after aerosolization by formation of complex with copper View project

All content following this page was uploaded by Mingshuang Li on 27 April 2018.

The user has requested enhancement of the downloaded file.


Drug Development and Industrial Pharmacy

ISSN: 0363-9045 (Print) 1520-5762 (Online) Journal homepage: http://www.tandfonline.com/loi/iddi20

Otic drug delivery systems: formulation principles


and recent developments

Xu Liu, Mingshuang Li, Hugh Smyth & Feng Zhang

To cite this article: Xu Liu, Mingshuang Li, Hugh Smyth & Feng Zhang (2018): Otic drug delivery
systems: formulation principles and recent developments, Drug Development and Industrial
Pharmacy, DOI: 10.1080/03639045.2018.1464022

To link to this article: https://doi.org/10.1080/03639045.2018.1464022

Accepted author version posted online: 16


Apr 2018.
Published online: 25 Apr 2018.

Submit your article to this journal

Article views: 16

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=iddi20
DRUG DEVELOPMENT AND INDUSTRIAL PHARMACY, 2018
https://doi.org/10.1080/03639045.2018.1464022

REVIEW ARTICLE

Otic drug delivery systems: formulation principles and recent developments


Xu Liua, Mingshuang Lib, Hugh Smytha and Feng Zhanga
a
College of Pharmacy, The University of Texas at Austin, Austin, TX, USA; bDepartment of Communication Sciences and Disorders, The University
of Texas at Austin, Austin, TX, USA

ABSTRACT ARTICLE HISTORY


Disorders of the ear severely impact the quality of life of millions of people, but the treatment of these dis- Received 11 February 2018
orders is an ongoing, but often overlooked challenge particularly in terms of formulation design and prod- Revised 3 April 2018
uct development. The prevalence of ear disorders has spurred significant efforts to develop new Accepted 4 April 2018
therapeutic agents, but perhaps less innovation has been applied to new drug delivery systems to improve
KEYWORDS
the efficacy of ear disease treatments. This review provides a brief overview of physiology, major diseases, Otic drug delivery;
and current therapies used via the otic route of administration. The primary focuses are on the various intratympanic drug delivery;
administration routes and their formulation principles. The article also presents recent advances in otic intracochlear drug delivery;
drug deliveries as well as potential limitations. Otic drug delivery technology will likely evolve in the next ear disease
decade and more efficient or specific treatments for ear disease will arise from the development of less
invasive drug delivery methods, safe and highly controlled drug delivery systems, and biotechnology tar-
geting therapies.

Introduction Local drug delivery systems may have significant advantages


over systemic drug delivery systems depending on the disease
Millions of people worldwide suffer from various ear disorders and
and therapeutic. Local drug delivery, in most cases, maximizes
associated conditions such as otitis media, hearing loss, tinnitus,
drug concentration in the inner ear while minimizing systemic
and Meniere’s disease. For example, the chronic otitis media or
exposure. Because of this ‘pharmacokinetic advantage,’ local drug
otitis externa affects 3–5% of the US population, with an esti-
delivery systems are becoming the most common clinical therapy
mated annual cost of more than $2.98 billion [1–3]. Potentially,
method for the treatment of inner ear diseases [12]. Currently, the
life-threatening infections can spread to surrounding tissues if
three main pathways for local drug delivery to the inner ear are
these diseases are not optimally treated, especially for immuno-
the transtympanic, intratympanic, and intracochlear routes.
compromised patients. According to a WHO survey, 250 million Developments in gene and stem cell therapy have also shown
people worldwide have moderate to severe hearing loss. The that these therapies may be able to promote the regeneration of
prevalence of hearing loss in the USA increases as the population the neural and hearing sensory cells in the inner ear, which pro-
ages and causes serious social and economic burden [4,5]. vide another strategy for the treatment of hearing loss and other
Tinnitus, Meniere’s disease, and autoimmune inner ear disease are ear diseases [13,14].
also common disorders that significantly reduce an individual’s Otic drug delivery has gained increasing interest and has
quality of life [6–8]. advanced rapidly over the past few decades. Several review
The prevalence of ear disorders has spurred significant effort to articles on otic drug delivery have been published in the past
develop new therapeutic agents and some effort has also been 10 years, and they have covered the various aspects of otic drug
directed toward development of otic drug delivery systems to delivery (Table 1). However, most of these reviews focus on only
improve the efficacy of treatments for diseases of the ear. The one particular drug delivery strategy or a specific disease type.
demand for therapeutic treatments for ear disease is significant, Only a very limited number of review papers have directly
with a market of approximately $10 billion [4]. Fortunately, if addressed drug delivery to the ear. Moreover, there is a lack of
many of these diseases are properly diagnosed and treated in critical reviews that have summarized formulation design princi-
their early stages, half of all hearing impairment including deaf- ples of the various local drug administration methods that guide
ness may be successfully treated [9,10]. the development of otic drug products. Thus, this review high-
The ear is a very delicate and anatomically protected organ, lights the basic formulation principles of the various local ear drug
and this poses many challenges for otic drug delivery, especially administrations. In this article, we first provide a brief overview of
for the treatment of inner ear diseases. Traditional drug adminis- the anatomy and physiology of the ear as it relates to drug deliv-
tration routes (e.g. oral routes, injection, and topical routes) have ery. The major ear diseases and drug products are then discussed.
been mostly ineffective for treating inner ear diseases, due to The advantages and disadvantages of otic administration routes
three major physical barriers for drug delivery to the ear: the tym- are reviewed and compared. This review also discusses promising
panic membrane, the round and oval windows (OW), and the drug delivery systems for future clinical applications as well as the
blood–perilymph barrier (BPB) [11,12]. current challenges that remain to be solved.

CONTACT Feng Zhang feng.zhang@austin.utexas.edu College of Pharmacy, The University of Texas at Austin, 2409 University Avenue, A1920, Austin, TX
78712, USA
ß 2018 Informa UK Limited, trading as Taylor & Francis Group
2 X. LIU ET AL.

Table 1. Key review papers of the past decade on otic drug delivery.
Review topics References
Topical ear treatment Harmes et al. [15], Kaushik et al. [16]
Nanoparticles drug delivery to inner ear Li et al. [17], Valente et al. [12], Pyykko et al. [18,19], Pritz et al. [20], Gang
et al. [21]
Patent review about the treatment of inner ear diseases Nguyen et al. [22]
Summary about inner ear drug delivery strategies, clinical applications and the El Kechai et al. [23], Staecker et al. [24], Liu et al. [11] Teresa et al. [25], Hoskison
current hurdles et al. [26], Shibata et al. [27], McCall et al. [28], Salt et al. [29], Swan et al. [30]
Alles et al. [31]
Intracochlear drug delivery Peppi et al. [32]. Ayoob et al. [33], Borenstein et al. [34], Borkholder et al. [35]
Intratympanic drug delivery Pai et al. [36], Seggas et al. [37], Meyer and Thomas [7], Herraiz et al. [38]
Gene and stem cell therapy for the inner ear disease Hideto et al. [13], Kohrman et al. [39]
Biodegradable materials for local drug delivery to the inner ear Nakagawa et al. [40]

Figure 1. Anatomy of the human ear. Image was adapted from reference [22]. RWM: round window membrane; OW: oval window; IHCs: inner hair cells; OHCs: outer
hair cells.

Anatomy and physiology of the ear membrane is the transmission of sound waves to the ossicular
chain. These vibrations cause the movement of the ossicular chain,
The human ear consists of three major parts: the outer ear, the
and the piston-like motion of the stapes transfers the pressure
middle ear, and the inner ear (Figure 1). The outer ear extends
wave to the inner ear fluids via the OW. The vibrations of the
from the auricle, or pinna, along the ear canal and ends at the inner ear hair cells convert the acoustic information to a bioelec-
tympanic membrane (eardrum). A healthy ear canal is slightly tric signal. This signal is then transmitted to the brainstem via the
acidic, with a pH value between 5.0 and 5.7, which can inhibit cochlear nerve and ultimately to the cerebral cortex, where the
bacterial growth [41,42]. At the end of the ear canal lies the tym- information is translated into auditory sensations.
panic membrane, which separates the middle ear from the outer The first major barrier to drug delivery to the ear is the tym-
ear. The middle ear includes the ossicular chain, which consists of panic membrane, which is elliptical and slightly conical in shape
the malleus, incus, stapes, and a mucosal lining that keeps the and separates the middle ear from the outer ear. The tympanic
middle ear environment moist. The Eustachian tube provides a membrane is around 0.1 mm thick and consists of three layers: an
conduit between the middle ear and the upper airway to allow for outer epidermal layer, an inner mucosal layer, and a middle
equalization of middle ear pressure [43]. fibrous layer with collagen [44]. The tympanic membrane is
The inner ear includes the cochlea, the organs of balance, the impenetrable to all but relatively small and moderately lipophilic
vestibule (utricle and saccule), and the semicircular canals. The molecules due to its keratin and lipid-rich stratum corneum [45].
cochlea is separated into three fluid-filled compartments. The mid- The tympanic membrane is the first barrier to drug delivery to the
dle compartment, called the scala media, is filled with endolymph; middle and inner ear, so it must be breached if drugs are deliv-
while the lower and upper compartments, the scala tympani and ered through intratympanic injection or intracochlear injection.
scala vestibule, are both filled with perilymph. The basilar mem- Because of its structural similarity to skin, transtympanic delivery
brane supports the organ of Corti, which consists of highly organ- can transport a drug across an intact tympanic membrane from
ized mechanosensory cells: the inner and outer hair cells (OHC) the ear canal into the middle or inner ear [46].
and their supporting cells (SC). The second major barrier to drug delivery to the ear consists of
In the auditory process, sound waves are generally collected by the round window membrane (RWM) and the oval window (OW)
the pinna into the auditory canal, which causes vibration of the [47]. These are semipermeable membranes, located at the base of
tympanic membrane. The major function of the tympanic the cochlea, that separate the cochlea from the middle ear. They
DRUG DEVELOPMENT AND INDUSTRIAL PHARMACY 3

are considered either as barriers or as pathways in terms of drug Table 2. The major diseases related to the parts of the human ear.
delivery to the cochlea. Outer ear Middle ear Inner ear
The round window consists of three layers: (1) the outer epithe- Diseases Acute external otitis Otitis media Hearing loss (sensorineural
lium, which faces the middle ear; (2) the inner epithelium, which (swimmer’s ear) hearing loss, noise inducing
bounds the inner ear; (3) and a middle connective tissue layer. hearing loss, sudden sensori-
neural hearing loss), auto-
The outer and inner epithelial layers are separated by the connect- immune inner ear disease,
ive tissue, which consists of fibroblasts, blood vessels, collagen, Meniere’s disease, Tinnitus
and elastic fibers [48,49]. The round window has an average thick-
ness of 70 mm in humans, with a surface area around 2.2 mm2 [24].
Various animal models are used to evaluate otic drug delivery.
or fungal infection (10% of cases). In certain cases, it may also be
However, there are many differences between humans and other
associated with a variety of noninfectious dermatologic processes
species in the anatomy and physiology of the RWM [50]. The main
[61,62]. Symptoms include ear discomfort, itchiness, discharge, and
interspecies difference of the RWM is the overall thickness [51].
impaired hearing.
Substantial differences in RWM thickness exist between humans
Based on the latest clinical guidelines on the diagnosis and
70 mm, and rodents, with thicknesses of 10–14 lm for the chin-
management of acute otitis externa published by The American
chilla, and 12 lm for rats and 10–30 lm for guinea pigs. As the
Academy of Otolaryngology-Head and Neck Surgery Foundation,
physical barrier, the RWM isolates the cochlear perilymph from the
the primary treatment of otitis externa includes management of
middle ear cavity. The diffusion of a drug across this membrane is
pain, administration of topical medications to control edema and
not only influenced by the thickness of the RWM, but also by its
infection, removal of debris from the external auditory canal, and
physical conditions such as inflammation or morphological integ-
avoidance of contributing factors [63]. Most acute otitis externa
rity [52]. In another aspect, the elastic and semipermeable proper-
can be successfully treated with over-the-counter analgesics and
ties allow the round window to transport drugs into the cochlea
topical eardrops. Commonly used eardrops include acetic acid
and to be used as the major pathway for drug delivery to the
drops, which lower the pH of the ear canal; antibacterial or anti-
inner ear from the middle ear [53]. Some studies have shown that
fungal preparations, which control bacterial or fungal growth. In
the factors that influence the permeability of the round window
severe cases, treatment may require oral or intravenous antibiotic
include molecular size, concentration, solubility, and the electrical
therapy and narcotic analgesics [63].
charge of the molecule being transported and the thickness and
Otitis media is the most common disorder of the middle ear
the condition of the RWM [51,52,54,55].
[64]. It is characterized by inflammation of the middle ear without
The OW is located near the scala vestibuli and contains the
reference to etiology or pathogenesis. It is very common in chil-
footplate of the stapes and the annular ligament (Figure 1). This
dren. Otitis media can be subdivided into acute otitis media, otitis
structure has been suggested as a secondary route for drugs to
media with effusion, chronic suppurative otitis media, and adhesive
enter the perilymph of the vestibule [56]. However, it is difficult to
otitis media [46]. Guidance for the medical and surgical manage-
measure the amount of drug entering the inner ear through this ment of the different types of otitis media is well established
route [29]. Kang et al. used fluorescence technology to study and [2,65,66]. According to this guidance, the management of otitis
quantify the intracochlear drug delivery through the OW. They media should include an assessment of pain. Analgesics should be
find that the fluorescently labeled bisphosphonate compound can used to treat pain regardless of whether antibiotic therapy is pre-
be delivered to the apical cochlear turn via the OW in the human scribed. Antibiotic therapy and appropriate choices of antibiotic
cochlea, and interstellar communication likely plays an important agents should be carefully considered before the assessment of
role in determining patterns of drug delivery in the inner ear [57]. the patient’s condition. Compared to systemic administration, local
The third major barrier to drug delivery to the ear is the BPB. delivery of antibiotics directly to the middle ear as a treatment for
The BPB is a physical and biochemical barrier between systemic otitis media could be an efficient strategy, and a safer one, in view
circulation and the cochlea. It is formed by a continuous capillary of the risk of systemic toxicity and emerging antibiotic resist-
endothelium that links blood vessels in the cochlea. The endothe- ance [67,68].
lial cells are connected by tight junctions with no fenestrations Inner ear diseases mainly include hearing loss, Meniere’s disease,
[48]. Like the blood–brain barrier, the BPB is permeable only to tinnitus, and autoimmune inner ear disease. These are common
small, lipid-soluble molecules. Molecules that are charged, water- disorders that significantly influence an individual’s quality of life.
soluble, or have a high molecular weight may have low passive Hearing loss is one of the most common ear disorders in humans,
diffusion across the barrier [58]. The BPB protects the ear from the and it becomes increasingly prevalent with age [69,70]. Hearing
damage caused by exogenous and endogenous toxins in systemic loss is the third most common chronic condition in the older
circulation, but it also presents challenges for efficient drug deliv- population (after arthritis and high blood pressure) [71]. The types
ery from systemic circulation to the cochlea for treatment [59]. of hearing loss include sensorineural hearing loss, conductive hear-
ing loss, retrocochlear hearing loss, and mixed hearing loss. The dif-
ferent types of hearing loss result from different etiologies [25,72].
Major ear diseases and therapies
Meniere’s disease is a disorder of the inner ear with unknown
Many diseases can affect any of the three main compartments of etiology. It is characterized by endolymphatic hydrops that occur
the human ear. The major diseases are summarized in Table 2. due to the dysfunction of types I and II spiral ligament fibrocytes.
Otitis externa is the most common disorder of the outer ear [60]. The symptoms of Meniere’s disease include episodes of spontan-
It affects the ear canal, with or without infection. This inflamma- eous, recurrent vertigo accompanied by fluctuating hearing loss
tion is usually generalized throughout the ear canal and can affect and intermittent tinnitus. The treatment of Meniere’s disease
the outer ear. It can be subdivided into acute (less than 6 weeks), includes the systemic and local administration of corticosteroids
recurrent acute, and chronic (more than 3 months) [16]. Acute otitis and aminoglycoside (i.e. gentamicin, gadolinium, and kanamycin)
externa is a common clinical problem encountered in general [31,73,74]. Tinnitus is defined as the experience of auditory percep-
practice. It results from either a bacterial infection (90% of cases) tion in the absence of an external stimulus. It occurs in
4 X. LIU ET AL.

Table 3. Therapeutic agents and routes of administration for major ear diseases.
Type Molecule Diseases Route of administration References
Antibiotics Amoxicillin Acute otitis externa otitis media Systemic/local [26,79]
Ciprofloxacin
Neomycin
Antifungal Clotrimazole Otitis externa Systemic/local [60,80]
Tolnaftate
Antiviral Acyclovir Hearing loss due to viral infection Local [81,82]
Cidofovir Meniere’s disease
Ganciclovir
Anesthetic agents Benzocaine Pain from otitis Local [79,83]
Lidocaine Tinnitus
Steroids Dexamethasone Otitis Local [37,78,84]
Methylprednisolone Hearing loss
Prednisone Meniere’s disease
Tinnitus
Autoimmune inner ear disease
Aminoglycoside Gentamicin Meniere’s disease Local [6,85,86]
Streptomycin
Kanamycin
Antioxidants D-methionine Hearing loss Local [87–88]
Thiourea
n-Acetyl cysteine
Apoptosis inhibitors AM 111 Hearing loss Local [89,90]
(JNK inhibitor)
NMDA inhibitors Gacyclidine Tinnitus Local [91,92]
Ifenprodil
Esketamine
Gene Atonal homolog 1 (Atoh1) Hearing loss Local [13,39]
Stem cells

approximately 10–15% of the population and severely impairs the Systemic drug delivery
quality of life in 1–2% of cases [33]. The pathology of tinnitus is
Systemic drug delivery has multiple limitations, such as the BPB
still unclear, but there are indications of potential involvement of
and undesired side effects, but systemic drug delivery through
both peripheral and central neural components [75]. Results from
oral and intramuscular routes is still considered the most conveni-
some studies indicate a beneficial effect on tinnitus after the use
ent method of drug administration to the various parts of ear. It is
of intratympanic dexamethasone injections in the treatment of
currently accepted as the first-line approach in the treatment of
sudden hearing loss [76]. Autoimmune ear diseases result in hear-
ing loss when the immune regulation system is compromised inner ear disorders [11]. Systemic drug delivery can be divided
[8,77]. High-dose corticosteroids and the cytotoxic agent cyclo- into oral drug delivery and intravenous drug delivery.
phosphamide are both safe and effective treatments of auto- Oral drug delivery is commonly used to treat middle ear and
immune ear diseases [78]. inner ear disorders due to its convenience and easier compliance
Various treatments have been proposed for treating diseases of by patients [15,16]. The bioavailability of oral delivery is limited
the ear, whether they are infectious, inflammatory, metabolic, or compared to local drug delivery, but it has wide appeal for many
autoimmune conditions. Drugs can be delivered systemically, or patients. Oral antibiotics are commonly prescribed for treating
they can be directed locally to specific ear positions. Diseases of acute external otitis and otitis media [96]. However, the disadvan-
the outer ear and some middle ear diseases are commonly treated tages of oral antibiotics include the possibility of multidrug resist-
with topical antibiotics, antifungal agents, steroids, or anesthetic ance and imbalance of intestinal flora. Oral steroids, antioxidants,
agents. Inner ear diseases can be treated with steroids, aminogly- and neuroprotective agents are commonly used to treat sudden
coside, antioxidants, apoptosis inhibitors, or N-methyl-D-aspartate sensorineural hearing loss to improve hearing [26,97].
(NMDA) inhibitors. Table 3 summarizes the main medicines used The other common systemic method is intravenous drug deliv-
for the treatment of ear diseases. In addition, an increasing num- ery (i.e. intravenous injection), which has been used for severe
ber of new drugs now in development, either in the preclinical or middle ear infections such as acute mastoiditis and necrotizing oti-
clinical research stage, are showing promising results for the treat- tis externa. Intravenous steroids have also been used to treat hear-
ment of inner ear diseases [22,24,33,93]. ing loss and Meniere’s disease [98,99]. The side effects of
intravenous steroids are similar to those of oral steroids. Some
research has shown that steroid nanoparticles can reduce poten-
Administration routes for otic drug delivery tial systemic side effects by improving oral bioavailabil-
Various administration routes have been developed for ear drug ity [100,101].
delivery. The selection of the administration route depends on the
disease and the physicochemical properties of the drug substan-
Local drug delivery
ces. Generally, the administration routes for ear drug delivery can
be divided into systemic delivery routes and local drug delivery During the past decade, interest in local drug delivery for the ther-
routes. Table 4 summarizes the routes for ear drug administration. apy of ear diseases, especially for the inner ear, has increased sig-
Ear implants are beyond the scope of this article and readers are nificantly. Compared to systemic administration, local application
directed to other recent review articles for detailed discussion of has many advantages, especially for drugs that have a narrow
ear implants [33,58,94,95]. therapeutic window, significant first-pass metabolism, or serious
DRUG DEVELOPMENT AND INDUSTRIAL PHARMACY 5

Table 4. Summary of the drug delivery routes to the ear.


Route Dosage form Disease Advantages Disadvantages
Systemic Oral Tablet Otitis media Self-administered Blood–cochlear barrier
Capsule Hearing loss Short-course Multidrug resistance
Meniere disease No interventions required Low local vascularization
Intravenous Solution Hearing loss High-dose Side effects
Nanomedicine Meniere disease
Local Topical Solution Acute Otitis externa Self-administered Limited applications
Suspension Otitis media Noninvasive Drops are unsuitable
Cream Short-course
Gel No interventions required
Nanomedicine
Transtympanic Hydrogels Otitis media Noninvasive Lower bioavailability
Nanomedicine Hearing loss
Meniere disease
Tinnitus
Intratympanic Suspension Hearing loss Easy access Invasive
Hydrogels Meniere disease Minimal risk of hearing loss Lower bioavailability
Nanomedicine Tinnitus Less surgical trauma Variability in dosage control
Autoimmune inner ear disease Local anesthesia Unknown pharmacokinetic profiles of drug
Intracochlear Solutions Hearing loss Dose control Highly invasive
Meniere disease Increased bioavailability Fluid leakage
Tinnitus Minor residual loss Difficult to access
Autoimmune inner ear disease General anesthesia

side effects. The advantages of local drug delivery are (1) the abil- membrane of rats that have otitis media. This offers a new drug
ity to bypass the BPB, (2) higher local drug concentration, and (3) delivery platform for the noninvasive delivery of gene therapy vec-
reduced side effects. Four types of local delivery systems are cur- tor to the middle ear [109,110]. What we need to pay an attention
rently in use: topical, transtympanic, intratympanic, and coch- is that currently most of these methods are still in the preclinical
lear delivery. stage and have not advanced to clinical evaluation. One potential
disadvantage of transtympanic delivery is the limited amount of
drug that can be transported across the tympanic membrane.
Topical drug delivery
Topical drug delivery has long played an important role in treating
ear disorders. This route involves direct administration of a drug Intratympanic drug administration
into the ear canal. Commonly used topical medications include The middle ear has been used as a reservoir for drugs that can
topical antibiotic, and antifungal drugs in the form of drops [102], then diffuse through the round window into the inner ear.
gels [103], or foams [104]. Otic topical drug administration pro- Intratympanic drug delivery involves injecting the drug in the mid-
vides a variety of advantages [105]. The most significant advan- dle ear cavity, and the drug substances then diffuse from the mid-
tage is the ability to achieve a local drug concentration much dle ear cavity into the cochlea (Figure 3). Drug diffusion across the
higher than what could be achieved via systemic administration. RWM is driven by the concentration gradient between the middle
This is often required for therapeutic purposes; for example, the ear and the perilymph-filled scala tympani on the opposite side of
elimination of biofilm bacteria requires 10–1000 times higher con- the RWM. The drug diffusion rate depends on various factors, such
centrations of antibiotics than planktonic bacteria. Other advan- as the molecular weight, configuration, concentration gradient, lip-
tages of topical drug delivery include rapid delivery, good patient ophilicity, and electrical charge of the drug, as well as the thick-
compliance, and the ability to combine different drugs into a sin- ness of the RWM [7]. The volume of intratympanic injection in
gle formulation [106]. The major limitation of topical drug delivery published studies ranges from 0.3 to 0.5 ml [7]. The RWM as a por-
is the potential ototoxicity for some drugs, especially if the drug tal to inner ear therapy has been studied extensively in animals;
concentration is very high [106]. however, very few study focused on the influence of inter-species
differences on intratympanic delivery. The thickness of the human
RWM, the presence of false membranes, the presence of tissue
Transtympanic drug administration plugs, and bony obliteration might influence the passage of sub-
The tympanic membrane is the first barrier to the delivery of stances through human RWM [25]. The influence of inter-species
drugs to the middle ear and inner ear. Successful transtympanic differences on intratympanic drug delivery should be carefully
delivery relies on drug diffusion through the intact tympanic considered when designing human clinical trials. Table 4 summa-
membrane from the ear canal into the middle and inner ear rizes the advantages and disadvantages of this administra-
(Figure 2) [46]. This process can be facilitated by permeation tion route.
enhancers [103,107]. Localized and sustained antibiotic delivery Small pharmaceutical devices such as the osmotic pump,
directly to the middle ear by transtympanic administration to treat microcatheter, and microWick have also been used for intratym-
otitis media has been achieved using chemical permeation panic drug delivery [25,33]. A number of clinical studies have been
enhancers contained within a hydrogel. Using magnetic fields to published on the intratympanic injection of steroids for the treat-
delivery magnetic drug nanoparticles has been demonstrated to ment of hearing loss and Meniere’s disease [37,84,111]. The limita-
be a viable method to facilitate drug permeation through the tym- tions of intratympanic drug delivery include the anatomic barriers
panic membrane [108]. Recent studies have shown that peptides to drug absorption from the middle ear to the inner ear, the loss
can be actively transported through the intact tympanic of drug in the middle ear through the Eustachian tube, and the
6 X. LIU ET AL.

Figure 2. Different strategies for promoting transtympanic drug delivery. Image was adapted from reference [103].

Figure 3. Different ear components with intratympanic and intracochlear routes for local drug delivery to the inner ear. Image was adapted from reference [22].

unknown pharmacokinetic profiles of medications administered by delivery for acute drug application to the base of the cochlea. In
this route [29,112]. Controlled delivery systems, such as biodegrad- addition, the use of microneedle for drug delivery across RWM
able hydrogel and nanoparticles, were developed to overcome could create temporary microperforation in the RWM to greatly
these limitations of intratympanic drug delivery [23,113,114]. enhance diffusion and duration of therapeutic agents.
Furthermore, the measurement of sealing the injection site could
reduce the fluid leaks that result from cochlear perforation,
Intracochlear drug administration improved the drug retention in perilymph [116]. The application
Compared with intratympanic drug delivery, intracochlear drug of microneedle minimizes damage to the RWM and prevents the
delivery bypasses the middle ear and allows drugs to reach the unintended disruption of endocochlear pressure fluctuation [117].
intended sites directly. Intracochlear delivery can achieve better Furthermore, microneedles are being designed to detect precisely
drug bioavailability than other delivery methods [34]. Intracochlear the moment of penetration without tearing the RWM [118]. The
administration methods include direct injections, cochlear amount of the injected substance retained in cochlear perilymph
implants, osmotic mini-pumps, and reciprocating perfusion [11]. following an intracochlear injection could be measured using
The simplest method for drug delivery to the inner ear is the dir- fluorescein technology [57,116,119].
ect injection of drugs into the cochlea using a syringe. Small vol- Cochlear implants and osmotic pumps offer some of the best
ume (approximately 5 mL) of drug solutions can be injected methods for controlled, automatic, complex dosing of numerous
directly into the cochlea through the RWM using 30–36 G fine- compounds into the cochlea [28]. Although intracochlear adminis-
gauge microneedle [115,116]. This method provides accurate tration is more effective than intratympanic administration,
DRUG DEVELOPMENT AND INDUSTRIAL PHARMACY 7

intracochlear drug delivery still has some limitations: It is invasive, Table 5. The common excipients used for topical otic drug delivery systems.
and it has potential toxicity due to the complex pharmacokinetics Function category Excipients Dosage forms
of the inner ear with locally delivered drugs [32,120]. pH modifier Acetic acid Solution, suspension,
Calcium carbonate liquid, drops
Citric acid
Strategies for developing otic formulations Hydrochloric acid
Benzethonium chloride
Topical drug delivery Benzyl alcohol
Hydrochloric acid
Typically, the dosage forms of topical otic drug administration Lactic acid
include ear drops, foams, gels, creams, and ointments. Drugs can Monopotassium phosphate
Sodium acetate
be dissolved or dispersed in water, glycerol, diluted alcohol, or
Sodium borate
propylene glycol. The selection of solvent vehicles depends on the Sodium citrate
solubility of the drug, because the concentration must be high Sodium phosphate, dibasic,
enough to account for the low-dose volume. monobasic
Sodium hydroxide
The viscosity of a topical formulation is also important, because Sulfuric acid
of the effect that viscosity has on the ability to effectively deliver Tromethamine
a drug to the site of infection. Excipients to increase the formula- Antimicrobial Aluminum acetate
tion viscosity can be added to retain formulations within the ear. preservative Benzalkonium chloride
Otic foam is commonly used to improve drug retention time in Benzethonium chloride
Benzyl alcohol
the ear canal. Studies have shown that the ciprofloxacin foam for- Boric acid
mulation is more efficacious than the solution for acute otitis Chlorobutanol
externa [104,121]. Isopropyl alcohol
Another important factor in formulating a topical agent is pH. Phenethyl alcohol
Methylparaben
Traditionally, topical otic preparations are acidic solutions or sus- Potassium metabisulfite
pensions with a pH of 3–4. Bacterial growth is inhibited at this pH Propylparaben
level [102]. Based on FDA inactive ingredient database and Thiomersal
approved topical otic products on market, the common used exci- Suspending agent Aluminum sulfate
pients are summarized in Table 5. Cetyl alcohol
Hydroxyethyl cellulose
Methylparaben
Polyvinyl alcohol
Intratympanic drug delivery
Stabilizing agent/thickening Creatinine
The cochlea is extremely sensitive to the fluid volume and ionic agent Hydrogenated soybean lecithin
Povidone K30
composition change, so intratympanic drug administration must Povidone K90
maintain the homeostasis of the inner fluids. The solution used for Poloxamer 407
intratympanic drug delivery must be within physiological pH range Emollient Cupric sulfate
(7.1–7.4). In addition, the formulation must be sterile, isotonic, and Glycerol
preservative free, as preservatives may affect round window per- Polyoxyl 40 Stearate
meability and lead to toxicity in the cochlear cells [55]. Solubilizing agent/wetting Polysorbate 20
agent Polysorbate 80
Formulations for intratympanic drug delivery can be solutions, sus-
Tyloxapol
pensions, hydrogels, or nanoparticles [12,38].
Tonicity agent Sodium chloride
The major advantage of hydrogels and nanoparticles is their Sodium sulfite
ability to deliver drugs for extended periods of time, thereby Ointment base Mineral oil Oil, ointment
greatly increasing the period of performance over solution formu- Peanut oil
lations. In addition, the hydrogel-based delivery system can enable Petrolatum
precise targeting of tissues and organs through the use of trigger-
ing mechanisms. These triggers such as pH, temperature, pressure,
or electrical potential will cause the hydrogel to swell and release OtiprioV is the first and only commercial intratympanic injection
R

the drug in a controlled fashion. Chitosan [30,113], hyaluronic acid for treating otitis media. It is a single-dose otic ciprofloxacin sus-
[23], poloamer 407 [111,122], and PLGA [68,123] are commonly pension with thermosensitive liquid-to-gel technology developed
used to prepare hydrogel formulations. When developing hydro- by Otonomy, Inc. Poloxamer 407 is used in order to achieve a
gels for intratympanic drug delivery, several important parameters hydrogel at body temperature. Another product OtividexV from
R

must be evaluated: (1) injectability through long and fine needles, Otonomy, using the same technology for Meniere’s disease treat-
(2) residence time of the hydrogel in the middle ear, (3) drug ment, is in a phase-3 clinical trial.
release and diffusion profiles, and (4) the ototoxicity of the hydro- Nanoparticle drug delivery systems have garnered much atten-
gel. Computer mathematical simulation shows that a major factor tion in the past decades for drug delivery to the inner ear. These
controlling both the amount of drug entering the inner ear and systems are of particular interest due to several advantages, such
the distribution of drug loading along the length of the ear is the as improving the solubility and bioavailability of poorly water-sol-
duration the drug remains in the middle ear space [124]. Several uble drugs delivered to the ear as well as providing for more effi-
studies revealed that drug formulated in hydrogel shared signifi- cient and more accurate treatment [21]. Multiple nanoparticle
cantly more prolonged exposure than those formulated in aque- drug delivery systems have been studied, including liposome
ous solutions in the inner ear in both in vitro and in vivo [126], lipid nanocapsules [127], PLGA nanoparticles [128], polymer-
models [111,125]. somes [114,129], and superparamagnetic nanoparticles. The critical
8 X. LIU ET AL.

Table 6. List of drugs and excipients that may cause ototoxicity.


Drugs Solvents Other excipients
Topical and systemic antibiotics Polyethylene glycol Antiseptics
All aminoglycosides Propylene glycol Acetic acid
Platinum compounds Benzalkonium chloride Alcohol
Chloramphenicol Chlorhexidine
Polymyxin B Cresylate
Amphotericin B Gentian violet
Bacitracin Povidone iodine
Chloramphenicol
Macrolides
Nystatin
Nonsteroidal anti-inflammatory drugs
Salicylates
Indomethacin
Ibuprofen
Phenylbutazone
Paracetamol
Topical combinations
Polymyxin/neomycin/hydrocortisone
Ticarcilline/clavulanate
Figure 4. Cell types present in the cochlea and the endogenous or transgenic
expression pattern of several genes required for hearing function. SV is the stria
vascularis cell, IHC is the inner ear hair cell, OHC is the outer hair cell, SP is the
spiral prominence cell, SC is the supporting cell, SG is the spiral ganglion cell, Biological formulations
and MC is the mesothelial cell. Image was adapted from reference [39].
Gene delivery and stem cell therapy are also promising
approaches to treat inner ear diseases [13,132]. Various biological
formulations are either in the preclinical or clinical stage, and no
properties of nanoparticles for intratympanic administration are
product has yet been approved by the FDA.
their permeability through the RWM, their distribution in the coch-
Studies have shown that specific genes play a pivotal role in
lea, their drug release profile, and their potential ototoxicity governing hair cell differentiation and regeneration. Genetic
[18,19]. Currently, no commercial nanoparticle product for intra- manipulation of these genes in humans would lead to protection,
tympanic administration exists; however, the application of nano- replacement, and regeneration of the function cells relating to
particles to guide drugs to the inner ear after they are hearing, including inner hair cells (IHC), OHC, and the spiral gan-
administered to the middle ear shows great potential in ani- glion cells (SG) of the auditory nerve [39]. Successful gene therapy
mal models. must target a range of cell types using viral, nanoparticle, or lipo-
somes vectors. Multiple types of epithelial SC, stria vascularis cells
(SV), and spiral prominence cells play important roles in hearing
Intracochlear drug delivery function (Figure 4). When determining the optimal vector, one
should consider potential side effects, cell specificity, duration of
In intracochlear administration, the drug is introduced directly into
gene expression, and access for delivery [13]. The major limitations
the cochlea via injection to overcome the barriers of the middle
of gene therapy include immune response, lack of access to the
ear (Figure 2) [35]. Similar to intratympanic drug delivery, the solu- scala media, and the targeting efficiency of the gene [11].
tion used for intracochlear drug delivery should be sterile, isotonic, Stem cell therapy provides a good option for treating hearing
preservative free, and have a pH ranging from 7.1–7.4; since even loss [121]. Stem cell therapy uses embryonic stem cells, fetal dorsal
small changes in the inner ear fluids (e.g. composition, pH, osmo- root ganglia, and otocyst cells in the inner ear to restore damaged
lality, and volume) could lead to deafness and ototoxicity. hair cells.
A safe and robust technique for intracochlear delivery of drugs
is still under development. To date, most of the published studies
were conducted in animal models [116,130]. Very few clinical Potential ototoxicity
study has evaluated intracochlear formulations to treat inner ear When delivered directly to the ear in large quantities, the drug
disorders [94]. The first clinical trials focus on intracochlear drug and its delivery carrier have the potential for ototoxicity regardless
delivery is underway [131]. Based on pharmacokinetic data from in of the administration route [20,133]. Toxicity in the middle and
vivo investigations in guinea pigs, dexamethasone concentration inner ear is a major cause of sensorineural hearing loss, tinnitus,
in the perilymph of the scala tympani was estimated for different and dizziness or vertigo [93,134]. It is essential to evaluate the oto-
application strategies in humans by means of a validated com- toxic effect of otic formulations, not only on the inner ear but also
puter model for calculation of substance concentrations in inner on the middle ear and tympanic membrane [135,136]. Table 6 pro-
ear fluids [131]. The simulation results indicated the continuous vides the wide range of substances that can cause middle and
intracochlear application of OzurdexV in addition to a cochlear
R
inner ear toxicity, including therapeutic drugs, antiseptics, and the
implant electrode carrier resulted in a stable drug concentration. inert components of vehicles [137–139]. Aminoglycoside antibiot-
Although intracochlear drug delivery system is highly invasive ics and platinum-based chemotherapeutic agents are the two
with lack of control over drug release profiles, intracochlear drug major classes of indispensable drugs that can cause serious oto-
delivery will likely become a promising approach to treat a host of toxicity [140,141]. The mechanisms underlying these side effects
currently intractable diseases of the inner ear, given the develop- are believed to involve the production of reactive oxygen species
ment of precision injection technology and our improved under- in the cochlea, which can trigger hair cell-death pathways
standing of the mechanisms of inner ear diseases. [142,143]. The strategies for minimizing ototoxicity include local
DRUG DEVELOPMENT AND INDUSTRIAL PHARMACY 9

Table 7. Summary of the latest local otic drug delivery product pipeline developments.
Company Product/description Research stage Administration route Indication
OtiprioV (ciprofloxacin otic suspenstion)
R
Otonomy Approved by FDA (2015) Intratympanic injection Otitis media
OtividexV (dexamethasone)
R
Phase III Meniere disease
OTO-311 (gacyclidine) Phase I Tinnitus
Auris/Xigen AM-111 (brimapitide) Phase III Intratympanic injection Hearing loss
KeyzilenV (esketamine)
R
Auris Phase III Intratympanic injection Tinnitus
Edison pharmaceuticals EPI-743 (antioxidant) Phase II Oral Hearing loss
Sound pharmaceuticals SPI-1005 (ebselen) Phase II Oral Hearing loss
Meniere disease
SPI-3005 (ebselen and allopurinol) Phase I Oral Chemotherapy or aminoglycoside-induced
ototoxicity
Autifony AUT00063 Phase I Systemic injection Hearing loss
GenVec/Novartis CGF166 (Atoh1 gene) Phase I intralabyrinthine injection Hearing loss
Sensorion SENS-401 (R-azasetron besylate/ Phase I Oral Hearing loss
A 5-HT3 receptor antagonist)
Nordmark Ancord (fibrinogenase) Phase II Systemic injection Hearing loss
PedmarkV (sodium thiosulfate)
R
Fennec Phase III Intratympanic injection Cisplatin-induced hearing loss
GSK Vestipitant (NK1 receptor antagonist) Phase II Oral Tinnitus
Synphora Latanoprost (PGF2 alpha agonist) Phase II Intratympanic injection Meniere disease
Strekin STR-001 (peroxisome proliferator-activated Phase II Intratympanic injection Hearing loss
receptor-gamma) agonist

administration of antioxidants or steroids to protect the inner ear and sustained manner, and their in vivo study demonstrated that
[144–146]. Next generation aminoglycoside antibiotics and plat- intact liposomes were delivered into the perilymphatic space and
inum compounds with less ototoxicity are being devel- reached cellular structures in the scala media.
oped [147,148]. An increasing number of new chemical entities are in the pre-
clinical or clinical stage. Table 7 summarizes the product pipeline
for local otic drug delivery. Many of these compounds have dem-
Recent advances in otic drug delivery onstrated promising results for treating inner ear disorders in an
Significant advances in otic drug delivery have been achieved in effective and targeted manner [27,36]. Significant advances in
the past decade. In the past few years, several patents have been characterizing the drug release study in the inner ear will promote
filed on new compounds and drug delivery systems for the treat- the development of inner ear drug delivery [151,152].
ment of inner ear diseases [22]. An improved understanding of
the mechanisms of inner ear diseases has led to new drugs that
target the voltage-gated potassium ion channels, glutamate recep- Current challenges in otic drug delivery
tors, and inhibitors of the notch developmental signaling pathway Although significant advances in otic drug delivery were achieved
in order to treat hearing loss, tinnitus, and peripheral vestibular in the last decade, many challenges have yet to be overcome. The
dysfunction [22,149]. difficulties are mainly related to the biology of the ear and the
The development of sustained drug delivery to the RWM is also molecular mechanisms of the inner ear diseases [149]. The path-
crucial for local delivery to the inner ear. El Kechai et al. [23] eval- ology of some of inner ear diseases is poorly understood, includ-
uated a hyaluronic acid liposomal gel for the sustained delivery of ing sensorineural hearing loss, Meniere’s disease, and tinnitus
a corticoid to the inner ear after local injection into the middle ear
[132,149,153]. Our limited knowledge of these inner ear diseases
in a guinea pig model. Prolonged residence time at the site of
makes it challenging to find an effective drug and delivery strat-
injection, as well as in the round window, was achieved without
egy. With a better understanding of the anatomy of the inner ear
negative effect on the hearing thresholds of the animal. The pres-
and the pathology of inner ear diseases, more effective otic drug
ence of liposomes in the formulation resulted in sustained drug
products will be available on the market [154,155].
release in the perilymph for 30 d.
In addition, the pharmacokinetics of drugs in the inner ear are
Yu et al. developed an injectable PEG-Silk hydrogel to achieve
not well defined, and data are limited regarding drug distribution,
sustained release of glucocorticoid. The glucocorticoid concentra-
round window permeability, and local bioavailability in the inner
tion in the perilymph remained above 100 ng/mL for at least 10 d
for the PEG-Silk formulation, but less than 12 h for the control for- ear [120,156]. There are no standard testing methods for evaluat-
mulation of free glucocorticoid [150]. ing inner ear drug delivery. For example, Franz diffusion cells are
The advancement of new polymer materials and nanoparticle commonly used to evaluate transdermal and topical drug delivery
therapies also provide alternative strategies to improve drug per- formulations. However, this method, as well as the use of other
meation through the inner ear barriers and target particular cells existing in vitro diffusion cell systems, may not be suitable to
in the inner ear [17,40]. Yoon et al. developed Arg8-conjugated evaluate drug diffusion through the middle ear and the RWM. A
nanoparticles as a controlled drug release system for inner ear convenient and efficient in vitro diffusion cell method for evaluat-
therapy. The study results show that the nanoparticle is a promis- ing intratympanic formulations for inner ear drug delivery is not
ing candidate for drug or gene delivery [114]. It has been reported yet available [20].
that the incorporation of nanoparticle systems into a hydrogel Furthermore, because the fluid volume in the inner ear is
increases the residence time of the nanocarrier in the middle ear, extremely small, it is difficult to provide adequate samples for ana-
thus enhancing drug concentration in the inner ear. lysis by conventional analytical methods. Highly sensitive techni-
Lajud et al. [113] have developed a nanohydrogel in which the ques, such as LC-MS, are more adapted for assays in perilymph
liposome was incorporated into a chitosan hydrogel. They discov- samples, but the technique is complex and expensive.
ered that the nanohydrogel releases the liposome in a controlled Measurement of drug concentration at different locations in the
10 X. LIU ET AL.

cochlea presents a technical challenge for studying the perilymph systems, including hydrogels, nanoparticles, and other minimally
pharmacokinetics [156]. invasive drug delivery methods. These delivery systems offer bet-
Although local drug delivery to the inner ear has been widely ter treatment options with reduced side effects and a better qual-
accepted in clinical practice, the choice of drugs and application ity of life for individuals who suffer from diseases of the ear.
protocols in human is largely empirically derived [157]. Study of
the pharmacokinetics of drugs in inner ear will become increas-
ingly important for optimization of clinical practice and the devel- Disclosure statement
opment of otic drugs [35]. There are significant obstacles to study
the pharmacokinetics of drugs in inner ear. The pharmacokinetic No potential conflict of interest was reported by the authors.
properties derived from animal experiments might not represent
those in humans. Pharmacokinetic is also affected by the inner ear
ORCID
diseases. The computer simulation software developed by Salt has
demonstrated significant promise in facilitating the understanding Hugh Smyth http://orcid.org/0000-0002-6582-5869
of drug pharmacokinetic in the inner ear [120,158–161]. To date,
the cochlear fluids simulator software is one of the most compre-
hensive simulation programs of the ear. It can be used to calculate References
the effective drug concentration achieved when drug is delivered
[1] Osguthorpe JD, Nielsen DR. Otitis externa: review and clin-
directly to the inner ear of guinea pig, mouse, and human. The
ical update. Am Fam Physician. 2006;74:1510–1516.
simulation mechanism is based on the mathematical calculation of
[2] Shekelle PG, Takata G, Newberry SJ, et al. Management of
physical process, which involves solute dispersal through diffusion,
acute otitis media: update. Evid Rep Technol Assess (Full
longitudinal fluid flow, and clearances to other compartments.
Rep). 2010;198:1–426.
This approach has been validated in numerous experiments in
[3] Coco AS. Cost-effectiveness analysis of treatment options
which ion-selective electrodes were used to quantify the spread of
marker substances in the cochlear fluids [162]. for acute otitis media. Ann Fam Med. 2007;5:29–38.
Researchers have employed both in vitro and in vivo methods [4] Vio MM, Holme RH. Hearing loss and tinnitus: 250 million
to investigate otic drug delivery systems, but the in vitro–in vivo people and a US$10 billion potential market. Drug Discov
correlation study is overlooked, and few reports compare the in Today. 2005;10:1263–1265.
vitro and in vivo methods [11]. Generally, in vitro–in vivo correl- [5] Lin FR, Niparko JK, Ferrucci L. Hearing loss prevalence in
ation for otic drugs is poor due to (1) the difference in the anat- the United States. Arch Intern Med. 2011;171:1851–1853.
omy and physiology of the inner ear between the human and [6] Syed I, Aldren C. Meniere’s disease: an evidence based
other species, (2) the difference in RWM permeability in vitro and approach to assessment and management. Int J Clin Pract.
in vivo, and (3) the lack of cochlear clearance in vitro. Future 2012;66:166–170.
research and development of an effective in vitro evaluation [7] Meyer T. Intratympanic treatment for tinnitus: a review.
method for inner ear drug delivery is needed. Noise Health. 2013;15:83–90.
Last but not least, the toxicity and safety of new inner ear drug [8] Ruckenstein MJ. Autoimmune inner ear disease. Curr Opin
delivery systems are generally not well established. The inner ear Otolaryngol Head Neck Surg. 2004;12:426–430.
is a complex and subtle organ. Adverse effects and complications [9] WHO.int [Internet]. New York: World Health Organization;
of inner ear drug delivery can lead to severe side effects such as 2018. Prevention of blindness and deafness. Available
hearing loss. The potential ototoxicity of novel drug delivery sys- from: http://www.who.int/pbd/deafness/facts/en/.
tems should be carefully examined. [10] Esterberg R, Coffin AB, Ou H, et al. Fish in a dish: drug dis-
covery for hearing habilitation. Drug Discov Today.
2013;10:e23–e29.
Conclusion [11] Liu H, Hao J, Li KS. Current strategies for drug delivery to
Otic drug delivery, especially for the inner ear, is undergoing rapid the inner ear. Acta Pharmaceutica Sinica B. 2013;3:86–96.
development. A wide range of drug delivery systems have been [12] Valente F, Astolfi L, Simoni E, et al. Nanoparticle drug
developed in the last decade. Otic drug delivery can be divided delivery systems for inner ear therapy: an overview. J Drug
into systemic and local drug delivery. Deliv Sci Technol. 2017;39:28–35.
Despite potential side effects, systemic drug delivery is a min- [13] Fukui H, Raphael Y. Gene therapy for the inner ear. Hear
imally invasive approach and is very convenient for self-adminis- Res. 2013;297:99–105.
tration. Therefore, systematic delivery continues to play an [14] Kojima K, Murata M, Nishio T, et al. Survival of fetal rat
important role in otic drug delivery. otocyst cells grafted into the damaged inner ear. Acta
Local drug delivery can be further categorized as topical, intra- Oto-Laryngologica. 2004;124:53–55.
tympanic, and intracochlear. Local drug delivery has the unique [15] Harmes KM, Blackwood RA, Burrows HL, et al. Otitis media:
advantages of achieving high therapeutic concentration in the ear diagnosis and treatment. Am Fam Physician. 2013;88:
and minimizing systemic side effects. 435–440.
The most suitable drug delivery route should be optimized [16] Kaushik V, Malik T, Saeed SR. Interventions for acute otitis
based on the disease, the potential side effects, and the require- externa. Cochrane Database Syst Rev. 2010;1:Cd004740.
ments of the individual patient. Development of otic formulations [17] Li L, Chao T, Brant J, et al. Advances in nano-based inner
should follow these basic formulation principles to ensure efficient ear delivery systems for the treatment of sensorineural
and specific treatment for each type of ear disease. hearing loss. Adv Drug Deliv Rev. 2017;108:2–12.
Although many challenges in otic drug development still [18] Pyykko I, Zou J, Schrott-Fischer A, et al. An overview of
remain, continued progress toward understanding the biology of nanoparticle based delivery for treatment of inner ear dis-
the ear has resulted in the development of novel drug delivery orders. Methods Mol Biol. 2016;1427:363–415.
DRUG DEVELOPMENT AND INDUSTRIAL PHARMACY 11

[19] Pyykko € I, Zou J, Zhang Y, et al. Nanoparticle-based delivery [42] Kim JK, Cho JH. Change of external auditory canal pH in
for the treatment of inner ear disorders. Curr Opin acute otitis externa. Ann Otol Rhinol Laryngol. 2009;118:
Otolaryngol Head Neck Surg. 2011;19:388–396. 769–772.
[20] Pritz CO, Roy S, Dudas J, et al. Nanomedicine strategies for [43] Ballachanda B. The human ear canal. San Diego (CA):
drug delivery to the ear. Nanomedicine (Lond). Plural Publishing, Incorporated; 2013.
2013;8:1155–1172. [44] Salvinelli F, Maurizi M, Calamita S, et al. The external ear
[21] Chen G, Zhang X, Yang F, et al. Disposition of nanopar- and the tympanic membrane: a three-dimensional study.
ticle-based delivery system via inner ear administration. Scand Audiol. 1991;20:253–256.
Curr Drug Metab. 2010;11:886–897. [45] Yang R, Wei T, Goldberg H, et al. Getting drugs across bio-
[22] Nguyen K, Kempfle JS, Jung DH, et al. Recent advances in logical barriers. Adv Mater. 2017;29:1606596–160659.
therapeutics and drug delivery for the treatment of inner [46] Qureishi A, Lee Y, Belfield K, et al. Update on otitis media –
ear diseases: a patent review (2011–2015). Expert Opin prevention and treatment. Infect Drug Resist. 2014;7:
Ther Pat. 2016;27:191–202. 15–24.
[23] El Kechai N, Mamelle E, Nguyen Y, et al. Hyaluronic acid [47] Salt AN, Hirose K. Communication pathways to and from
liposomal gel sustains delivery of a corticoid to the inner the inner ear and their contributions to drug delivery.
ear. J Control Release. 2016;226:248–257. Hear Res. 2017;362:25–37.
[24] Staecker H, Rodgers B. Developments in delivery of medi- [48] Juhn SK. Barrier systems in the inner ear. Acta Oto-
cations for inner ear disease. Expert Opin Drug Deliv. Laryngologica. 1988;105:79–83.
2013;10:639–650. [49] Carpenter AM, Muchow D, Goycoolea MV. Ultrastructural
[25] Rivera T, Sanz L, Camarero G, et al. Drug delivery to the studies of the human round window membrane. Arch
inner ear: strategies and their therapeutic implications for Otolaryngol Head Neck Surg. 1989;115:585–590.
sensorineural hearing loss. Curr Drug Deliv. [50] Becvarovski Z, Bojrab DI, Michaelides EM, et al. Round win-
2012;9:231–242. dow gentamicin absorption: an in vivo human model.
[26] Hoskison E, Daniel M, Al-Zahid S, et al. Drug delivery to Laryngoscope. 2002;112:1610–1613.
the ear. Ther Deliv. 2012;4:115–124. [51] Goycoolea MV, Lundman L. Round window membrane:
[27] Shibata SB, Raphael Y. Future approaches for inner ear structure function and permeability: a review. Microsc Res
protection and repair. J Commun Dis. 2010;43:295–310. Tech. 1997;36:201–211.
[28] McCall AA, Swan EE, Borenstein JT, et al. Drug delivery for [52] Goycoolea MV. The round window membrane under nor-
treatment of inner ear disease: current state of knowledge. mal and pathological conditions. Acta Otolaryngol Suppl.
Ear Hear. 2010;31:156. 1992;493:43–55.
[29] Salt AN, Plontke SK. Principles of local drug delivery to the [53] Duan M-l, Zhi-qiang C. Permeability of round window
inner ear. Audiol Neurootol. 2009;14:350–360. membrane and its role for drug delivery: our own findings
[30] Paulson DP, Abuzeid W, Jiang H, et al. A novel controlled and literature review. J Otol. 2009;4:34–43.
local drug delivery system for inner ear disease. [54] Okuno H, Sando I. Anatomy of the round window: a histo-
Laryngoscope. 2008;118:706–711. pathological study with a graphic reconstruction method.
[31] Alles MJRC, der Gaag MA, Stokroos RJ. Intratympanic ster- Acta Oto-Laryngologica. 1988;106:55–63.
oid therapy for inner ear diseases, a review of the litera- [55] Mikulec AA, Hartsock JJ, Salt AN. Permeability of the round
ture. Eur Arch Otorhinolaryngol. 2006;263:791–797. window membrane is influenced by the composition of
[32] Peppi M, Marie A, Belline C, et al. Intracochlear drug deliv- applied drug solutions and by common surgical proce-
ery systems: a novel approach whose time has come. dures. Otol Neurotol. 2008;29:1020–1026.
Expert Opin Drug Deliv. 2018;15:319–324. [56] Saijo S, Kimura RS. Distribution of HRP in the inner ear
[33] Ayoob AM, Borenstein JT. The role of intracochlear drug after injection into the middle ear cavity. Acta Otolaryngol.
delivery devices in the management of inner ear disease. 1984;97:593–610.
Expert Opinion Drug Deliv. 2015;12:465–479. [57] Kang WS, Nguyen K, McKenna CE, et al. Intracochlear
[34] Borenstein JT. Intracochlear drug delivery systems. Expert drug delivery through the oval window in fresh cadaveric
Opin Drug Deliv. 2011;8:1161–1174. human temporal bones. Otol Neurotol. 2016;37:218–222.
[35] Borkholder DA. State-of-the-art mechanisms of intraco- [58] Swan EE, Mescher MJ, Sewell WF, et al. Inner ear drug
chlear drug delivery. Curr Opin Otolaryngol Head Neck delivery for auditory applications. Adv Drug Deliv Rev.
Surg. 2008;16:472–477. 2008;60:1583–1599.
[36] Pai Y-C, Lee C-F. Intratympanic steroid injection for inner [59] Shi X. Pathophysiology of the cochlear intrastrial fluid-
ear disease. Tzu Chi Med J. 2013;25:146–149. blood barrier (review). Hear Res. 2016;338:52–63.
[37] Seggas I, Koltsidopoulos P, Bibas A, et al. Intratympanic [60] Fedorova OV, Shadrin GB. The current views of the treat-
steroid therapy for sudden hearing loss: a review of the lit- ment of diffuse external otitis. Vestn Otorinolaringol.
erature. Otol Neurotol. 2011;32:29–35. 2016;81:51–53.
[38] Herraiz C, Miguel Aparicio J, Plaza G. Intratympanic drug [61] Sander R. Otitis externa: a practical guide to treatment
delivery for the treatment of inner ear diseases. Acta and prevention. Am Fam Physician. 2001;63:927–936.
Otorrinolaringologica. 2010;61:225–232. [62] Schaefer P, Baugh RF. Acute otitis externa: an update. Am
[39] Kohrman DC, Raphael Y. Gene therapy for deafness. Gene Fam Physician. 2012;86:1055–1061.
Ther. 2013;20:1119–1123. [63] Rosenfeld RM, Schwartz SR, Cannon CR, et al. Clinical prac-
[40] Nakagawa T, Ito J. Local drug delivery to the inner ear tice guideline: acute otitis externa. Otolaryngol Head Neck
using biodegradable materials. Ther Deliv. 2011;2:807–814. Surg. 2014;150:S1–S24.
[41] Martinez Devesa P, Willis CM, Capper JWR. External audi- [64] Granath A. Recurrent acute otitis media: what are the
tory canal pH in chronic otitis externa. Clin Otolaryngol. options for treatment and prevention? Curr
2003;28:320–324. Otorhinolaryngol Rep. 2017;5:93–100.
12 X. LIU ET AL.

[65] Lieberthal AS, Carroll AE, Chonmaitree T, et al. The diagno- [87] Heman-Ackah SE, Juhn SK, Huang TC, et al. A combination
sis and management of acute otitis media. Pediatrics. antioxidant therapy prevents age-related hearing loss in
2013;131:e964–e999. C57BL/6 mice. Otolaryngol Head Neck Surg.
[66] Rosenfeld RM, Shin JJ, Schwartz SR, et al. Clinical practice 2010;143:429–434.
guideline: otitis media with effusion (update). Otolaryngol [88] Clifford RE, Coleman JK, Balough BJ, et al. Low-dose D-
Head Neck Surg. 2016;154:S1–S41. methionine and N-acetyl-L-cysteine for protection from
[67] Daniel M, Chessman R, Al-Zahid S, et al. Biofilm eradication permanent noise-induced hearing loss in chinchillas.
with biodegradable modified-release antibiotic pellets: a Otolaryngol Head Neck Surg. 2011;145:999–1006.
potential treatment for glue ear. Arch Otolaryngol Head [89] Suckfuell M, Lisowska G, Domka W, et al. Efficacy and
Neck Surg. 2012;138:942–949. safety of AM-111 in the treatment of acute sensorineural
[68] Feng L, Ward JA, Li SK, et al. Assessment of PLGA-PEG- hearing loss: a double-blind, randomized, placebo-con-
PLGA copolymer hydrogel for sustained drug delivery in trolled phase II study. Otology & Neurotology.
the ear. Curr Drug Deliv. 2014;11:279–286. 2014;35:1317–1326.
[69] Cunningham LL, Tucci DL. Hearing loss in adults. N Engl J [90] Omotehara Y, Hakuba N, Hato N, et al. Protection against
Med. 2017;377:2465–2473. ischemic cochlear damage by intratympanic administration
[70] Atar O, Avraham KB. Therapeutics of hearing loss: expecta- of AM-111. Otol Neurotol. 2011;32:1422–1427.
tions vs reality. Drug Discov Today. 2005;10:1323–1330. [91] Salvi R, Lobarinas E, Sun W. Pharmacological treatments
[71] Shargorodsky J, Curhan SG, Curhan GC, et al. Change in for tinnitus: new and old. Drugs Future. 2009;34:381–400.
prevalence of hearing loss in us adolescents. JAMA. [92] Wenzel GI, Warnecke A, Sto €ver T, et al. Effects of extraco-
2010;304:772–778. chlear gacyclidine perfusion on tinnitus in humans: a case
[72] Crowson MG, Hertzano R, Tucci DL. Emerging therapies for series. Eur Arch Otorhinolaryngol. 2010;267:691–699.
sensorineural hearing loss. Otol Neurotol. 2017;38:792–803. [93] Mukherjea D, Rybak LP, Sheehan KE, et al. The design and
[73] Hill SL 3rd, Digges EN, Silverstein H. Long-term follow-up screening of drugs to prevent acquired sensorineural hear-
after gentamicin application via the Silverstein MicroWick ing loss. Expert Opin Drug Discov. 2011;6:491–505.
in the treatment of Meniere’s disease. Ear Nose Throat J. [94] El Kechai N, Agnely F, Mamelle E, et al. Recent advances in
2006;85:494–496. local drug delivery to the inner ear. Int J Pharm.
[74] Nakashima T, Naganawa S, Sugiura M, et al. Visualization 2015;494:83–101.
of endolymphatic hydrops in patients with Meniere’s dis- [95] Pararas EEL, Borkholder DA, Borenstein JT. Microsystems
ease. Laryngoscope. 2007;117:415–420. technologies for drug delivery to the inner ear. Adv Drug
[75] Kaltenbach JA. Tinnitus: models and mechanisms. Hear Deliv Rev. 2012;64:1650–1660.
Res. 2011;276:52–60. [96] Acton QA. Otitis – advances in research and treatment:
[76] Garduno-Anaya MA, Couthino De Toledo H, Hinojosa GR, 2013 Edition: ScholarlyBrief. 2013: Scholarly Editions.
et al. Dexamethasone inner ear perfusion by intratympanic [97] Rauch SD, Halpin CF, Antonelli PJ, et al. Oral vs intratym-
injection in unilateral Meniere’s disease: a two-year pro- panic corticosteroid therapy for idiopathic sudden sensori-
spective, placebo-controlled, double-blind, randomized neural hearing loss: a randomized trial. JAMA.
trial. Otolaryngol Head Neck Surg. 2005;133:285–294. 2011;305:2071–2079.
[77] Bovo R, Aimoni C, Martini A. Immune-mediated inner ear [98] Kakehata S, Sasaki A, Oji K, et al. Comparison of intratym-
disease. Acta Otolaryngol. 2006;126:1012–1021. panic and intravenous dexamethasone treatment on sud-
[78] Alexander TH, Weisman MH, Derebery JM, et al. Safety of den sensorineural hearing loss with diabetes. Otol
high-dose corticosteroids for the treatment of auto- Neurotol. 2006;27:604–608.
immune inner ear disease. Otol Neurotol. 2009;30:443–448. [99] Shea PF, Richey PA, Wan JY, et al. Hearing results and
[79] Subcommittee on management of acute otitis media. quality of life after streptomycin/dexamethasone perfusion
Diagnosis and management of acute otitis media. for Meniere’s disease. Laryngoscope. 2012;122:204–211.
Pediatrics. 2004;113:1451–1465. [100] Horie RT, Sakamoto T, Nakagawa T, et al. Stealth-nanopar-
[80] Okovityi SV, Ivkin D, Malygin SV. Drug therapy of otitis ticle strategy for enhancing the efficacy of steroids in mice
externa and otitis media. Vestn Otorinolaringol. 2012;1: with noise-induced hearing loss. Nanomedicine.
52–56. 2010;5:1331–1340.
[81] Awad Z, Huins C, Pothier DD. Antivirals for idiopathic sud- [101] Sun C, Wang X, Chen D, et al. Dexamethasone loaded
den sensorineural hearing loss. Cochrane Database Syst nanoparticles exert protective effects against Cisplatin-
Rev. 2012;8:Cd006987. induced hearing loss by systemic administration. Neurosci
[82] Gacek RR. Recovery of hearing in Meniere’s disease after Lett. 2016;619:142–148.
antiviral treatment. Am J Otolaryngol. 2015;36:315–323. [102] Eng CY, El-Hawrani AS. The pH of commonly used topical
[83] Horie RT, Sakamoto T, Nakagawa T, et al. Sustained deliv- ear drops in the treatment of otitis externa. Ear Nose
ery of lidocaine into the cochlea using poly lactic/glycolic Throat J. 2011;90:160–162.
acid microparticles. Laryngoscope. 2010;120:377–383. [103] Khoo X, Simons EJ, Chiang HH, et al. Formulations for
[84] Dispenza F, De Stefano A, Costantino C, et al. Sudden sen- trans-tympanic antibiotic delivery. Biomaterials.
sorineural hearing loss: results of intratympanic steroids as 2013;34:1281–1288.
salvage treatment. Am J Otolaryngol. 2013;34:296–300. [104] Marom T, Yelin R, Goldfarb A, et al. Comparison of safety
[85] Blakley BW. Update on intratympanic gentamicin for and efficacy of foam-based versus solution-based cipro-
Meniere’s disease. Laryngoscope. 2000;110:236–240. floxacin for acute otitis externa. Otolaryngol Head Neck
[86] Monsell EM, Cass SP, Rybak LP. Therapeutic use of amino- Surg. 2010;143:492–499.
glycosides in Meniere’s disease. Otolaryngol Clin North [105] Rosenfeld RM, Singer M, Wasserman JM, et al.
Am. 1993;26:737–746. Systematic review of topical antimicrobial therapy for
DRUG DEVELOPMENT AND INDUSTRIAL PHARMACY 13

acute otitis externa. Otolaryngol Head Neck Surg. clinical drug delivery protocols. Laryngoscope.
2006;134:S24–S48. 2008;118:1793–1800.
[106] Haynes DS. Topical antibiotics: strategies for avoiding oto- [125] Salt AN, Hartsock JJ, Plontke SK, et al. Distribution of dexa-
toxicity. Ear Nose Throat J. 2004;83:12–14. methasone and preservation of inner ear function follow-
[107] Yang R, Sabharwal V, Okonkwo OS, et al. Treatment of oti- ing intratympanic delivery of a gel-based formulation.
tis media by transtympanic delivery of antibiotics. Sci Audiol Neurotol. 2011;16:323–335.
Transl Med. 2016;8:356ra120–356ra120. [126] Bozzuto G, Molinari A. Liposomes as nanomedical devices.
[108] Shapiro B, Kulkarni S, Nacev A, et al. Shaping magnetic Int J Nanomed. 2015;10:975–999.
fields to direct therapy to ears and eyes. Annu Rev [127] Roy S, Glueckert R, Johnston AH, et al. Strategies for drug
Biomed Eng. 2014;16:455–481. delivery to the human inner ear by multifunctional nano-
[109] Kurabi A, Beasley KA, Chang L, et al. Discovery of a bio- particles. Nanomedicine. 2011;7:55–63.
logical mechanism of active transport through the tym- [128] Cai H, Wen X, Wen L, et al. Enhanced local bioavailability of
panic membrane to the middle ear. Sci Rep. 2016;6:22663. single or compound drugs delivery to the inner ear through
[110] Kurabi A, Beasley KA, Chang L, et al. Peptides actively application of PLGA nanoparticles via round window
transported across the tympanic membrane: functional administration. Int J Nanomed. 2014;9:5591.
and structural properties. PLos One. 2017;12:e0172158. [129] Kim D-K, Park SN, Park KH, et al. Development of a drug
[111] Wang X, Dellamary L, Fernandez R, et al. Principles of delivery system for the inner ear using poly(amino acid)-
inner ear sustained release following intratympanic admin- based nanoparticles. Drug Deliv. 2015;22:367–374.
istration. Laryngoscope. 2011;121:385–391. [130] Hahn H, Salt AN, Biegner T, et al. Dexamethasone levels
[112] Bowe SN, Jacob A. Round window perfusion dynamics: and base to apex concentration gradients in scala tympani
implications for intracochlear therapy. Curr Opin perilymph following intracochlear delivery in the guinea
Otolaryngo Head Neck Surg. 2010;18:377–385. pig. Otol Neurotol. 2012;33:660–665.
[113] Lajud SA, Han Z, Chi FL, et al. A regulated delivery system [131] Plontke SK, Gotze G, Rahne T, et al. Intracochlear drug
for inner ear drug application. J Control Release. delivery in combination with cochlear implants: current
2013;166:268–276. aspects. HNO. 2017;65:19–28.
[114] Yoon JY, Yang KJ, Kim DE, et al. Intratympanic delivery of [132] Mittal R, Nguyen D, Patel AP, et al. Recent advancements
oligoarginine-conjugated nanoparticles as a gene (or drug) in the regeneration of auditory hair cells and hearing res-
toration. Front Mol Neurosci. 2017;10:236.
carrier to the inner ear. Biomaterials. 2015;73:243–253.
[133] Tom LWC. Ototoxicity of common topical antimycotic
[115] Sellick P, Layton MG, Rodger J, et al. A method for intro-
preparations. Laryngoscope. 2000;110:509–516.
ducing non-silencing siRNA into the guinea pig cochlea in
[134] Wang KW. Bailey’s head and neck surgery: otolaryngology.
vivo. J Neurosci Method. 2008;167:237–245.
Philadephia (PA): Lippincott Williams & Wilkins; 2013.
[116] Plontke SK, Hartsock JJ, Gill RM, et al. Intracochlear drug
[135] Vural A, Şahin M_I, Aydın M, et al. The effect of nystatin
injections through the round window membrane: meas-
solution on otoacoustic emissions in rats. J Int Adv Otol.
ures to improve drug retention. Audiol Neurotol.
2017;13:105–109.
2016;21:72–79.
[136] Roland PS, Rutka JA. Ototoxicity. Raleigh (NC): PMPH-USA;
[117] Watanabe H, Kysar JW, Lalwani AK. Round window mem-
2004.
brane as a portal for inner ear therapy. Recent Adv
[137] Roland PS. Characteristics of systemic and topical agents
Otolaryngol Head Neck Surg. 2017;6:39.
implicated in toxicity of the middle and inner ear. Ear
[118] Wazen JM, Stevens JP, Watanabe H, et al. Silver/silver
Nose Throat J. 2003;82:3.
chloride microneedles can detect penetration through the [138] Taneja MK, Varshney H, Taneja V, et al. Ototoxicity, drugs,
round window membrane. J Biomed Mater Res. chemicals, mobile phones and deafness. Indian J Otol.
2017;105:307–311. 2015;21:161–164.
[119] Lee JJ, Jang JH, Choo OS, et al. Steroid intracochlear distri- [139] Haynes DS, Rutka J, Hawke M, et al. Ototoxicity of ototopi-
bution differs by administration method: systemic versus cal drops-an update. Otolaryngol Clin North Am.
intratympanic injection. Laryngoscope. 2018;128:189–194. 2007;40:669–683.
[120] Salt AN, Plontke SK. Pharmacokinetic principles in the [140] Waissbluth S, Daniel SJ. Cisplatin-induced ototoxicity:
inner ear: influence of drug properties on intratympanic transporters playing a role in cisplatin toxicity. Hearing
applications. Hear Res. 2018;1:1–13. Res. 2013;299:37–45.
[121] Havenith S, Versnel H, Agterberg MJ, et al. Spiral ganglion [141] Xie J, Talaska AE, Schacht J. New developments in amino-
cell survival after round window membrane application of glycoside therapy and ototoxicity. Hear Res.
brain-derived neurotrophic factor using gelfoam as carrier. 2011;281:28–37.
Hear Res. 2011;272:168–177. [142] Selimoglu E. Aminoglycoside-induced ototoxicity. Curr
[122] Engleder E, Honeder C, Klobasa J, et al. Preclinical evalu- Pharm Des. 2007;13:119–126.
ation of thermoreversible triamcinolone acetonide hydro- [143] Paken J, Govender CD, Paken J, et al. Cisplatin-associated
gels for drug delivery to the inner ear. Int J Pharm. ototoxicity: a review for the health professional. J Toxicol.
2014;471:297–302. 2016;2016:13.
[123] Sidell D, Ward JA, Pordal A, et al. Combination therapies [144] Rybak LP, Ramkumar V. Ototoxicity. Kidney Int.
using an intratympanic polymer gel delivery system in the 2007;72:931–935.
guinea pig animal model: a safety study. Int J Pediatr [145] Rybak LP, Whitworth CA. Ototoxicity: therapeutic opportu-
Otorhinolaryngol. 2016;84:132–136. nities. Drug Discov Today. 2005;10:1313–1321.
[124] Salt AN, Gill RM, Plontke SK. Dependence of hearing [146] Gianoli GJ, Li JC. Transtympanic steroids for treatment of
changes on the dose of intratympanically-applied genta- sudden hearing loss. Otolaryngol Head Neck Surg.
micin: a meta-analysis using mathematical simulations of 2001;125:142–146.
14 X. LIU ET AL.

[147] O’Sullivan ME, Perez A, Lin R, et al. Towards the preven- [155] De Paolis A, Watanabe H, Nelson JT, et al. Human cochlear
tion of aminoglycoside-related hearing loss. Front Cell hydrodynamics: a high-resolution lCT-based finite element
Neurosci. 2017;11:325. study. J Biomech. 2017;50:209–216.
[148] Karasawa T, Steyger PS. An integrated view of cisplatin- [156] Salt AN, Plontke SKR. Local inner-ear drug delivery and
induced nephrotoxicity and ototoxicity. Toxicol Lett. pharmacokinetics. Drug Discov Today. 2005;10:1299–1306.
2015;237:219–227. [157] Sakamoto T, Nakagawa T, Horie RT, et al. Inner ear drug
[149] Goodall AF, Siddiq MA. Current understanding of the delivery system from the clinical point of view. Acta Oto-
pathogenesis of autoimmune inner ear disease: a review. Laryngologica. 2010;130:101–104.
Clin Otolaryngol. 2015;40:412–419. [158] Liebau A, Pogorzelski O, Salt AN, et al. Hearing changes
[150] Yu D, Sun C, Zheng Z, et al. Inner ear delivery of dexa- after intratympanically applied steroids for primary therapy
methasone using injectable silk-polyethylene glycol (PEG) of sudden hearing loss: a meta-analysis using mathemat-
hydrogel. Int J Pharm. 2016;503:229–237. ical simulations of drug delivery protocols. Otol Neurotol.
[151] Liu H, Feng L, Tolia G, et al. Evaluation of intratympanic 2017;38:19–30.
formulations for inner ear delivery: methodology and sus- [159] Plontke S, Siedow N, Hahn H, et al. 1D- and 3D-computer
tained release formulation testing. Drug Dev Ind Pharm. simulation for experimental planning and interpretation of
2014;40:896–903. pharmacokinetic studies in the inner ear after local drug
[152] Kanzaki S, Fujioka M, Yasuda A, et al. Novel in vivo imag- delivery. Altex. 2004;21:77–85.
ing analysis of an inner ear drug delivery system in mice: [160] Salt AN, Plontke SK. Endolymphatic hydrops: pathophysi-
comparison of inner ear drug concentrations over time ology and experimental models. Otolaryngol Clin North
after transtympanic and systemic injections. PLos One. Am. 2010;43:971–983.
2012;7:e48480. [161] Plontke SKR, Salt AN. Quantitative interpretation of cortico-
[153] Ryan AF, Harris JP, Keithley EM. Immune-mediated hearing steroid pharmacokinetics in inner fluids using computer
loss: basic mechanisms and options for therapy. Acta simulations. Hear Res. 2003;182:34–42.
Otolaryngol Suppl. 2002;122:38–43. [162] Salt AN. Simulation of methods for drug delivery to the
[154] Holley MC. Keynote review: the auditory system, hearing cochlear fluids. In: Felix D, Oestreicher E, editors. Rational
loss and potential targets for drug development. Drug pharmacotherapy of the inner ear. Basel (Switzerland):
Discov Today. 2005;10:1269–1282. Karger Publishers; 2002. p. 140–148.

View publication stats

You might also like