Current Advances in Optometry
Current Advances in Optometry
Daisy Shu
Rohan Bir Singh
Parul Ichhpujani Editors
Current
Advances in
Optometry
Current Practices in Ophthalmology
Series Editor
Parul Ichhpujani, Department of Ophthalmology
Government Medical College and Hospital
Chandigarh, India
This series of highly organized and uniform handbooks aims to cover the latest
clinically relevant developments in ophthalmology. In the wake of rapidly evolving
innovations in the field of basic research, pharmacology, surgical techniques and
imaging devices for the management of ophthalmic disorders, it is extremely
important to invest in books that help you stay updated. These handbooks are
designed to bridge the gap between journals and standard texts providing reviews on
advances that are now part of mainstream clinical practice. Meant for residents,
fellows-in-training, generalist ophthalmologists and specialists alike, each volume
under this series covers current perspectives on relevant topics and meets the CME
requirements as a go-to reference guide. Supervised and reviewed by a subject
expert, chapters in each volume provide leading-edge information most relevant and
useful for clinical ophthalmologists. This series is also useful for residents and
fellows training in various subspecialties of ophthalmology, who can read these
books while at work or during emergency duties. Additionally, these handbooks can
aid in preparing for clinical case discussions at various forums and examinations.
Daisy Shu • Rohan Bir Singh
Parul Ichhpujani
Editors
Current Advances in
Optometry
Editors
Daisy Shu Rohan Bir Singh
School of Optometry and Vision Science, Department of Ophthalmology, Harvard
Faculty of Medicine and Health Medical School
University of New South Wales (UNSW) Schepens Eye Research Institute of Mass
Sydney, NSW, Australia Eye and Ear
Boston, MA, USA
Parul Ichhpujani
Department of Ophthalmology
Government Medical College and Hospital
Chandigarh, India
© The Editor(s) (if applicable) and The Author(s), under exclusive license to Springer Nature Singapore
Pte Ltd. 2024
This work is subject to copyright. All rights are solely and exclusively licensed by the Publisher, whether
the whole or part of the material is concerned, specifically the rights of translation, reprinting, reuse of
illustrations, recitation, broadcasting, reproduction on microfilms or in any other physical way, and
transmission or information storage and retrieval, electronic adaptation, computer software, or by similar
or dissimilar methodology now known or hereafter developed.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this publication
does not imply, even in the absence of a specific statement, that such names are exempt from the relevant
protective laws and regulations and therefore free for general use.
The publisher, the authors and the editors are safe to assume that the advice and information in this book
are believed to be true and accurate at the date of publication. Neither the publisher nor the authors or the
editors give a warranty, expressed or implied, with respect to the material contained herein or for any
errors or omissions that may have been made. The publisher remains neutral with regard to jurisdictional
claims in published maps and institutional affiliations.
This Springer imprint is published by the registered company Springer Nature Singapore Pte Ltd.
The registered company address is: 152 Beach Road, #21-01/04 Gateway East, Singapore 189721,
Singapore
We welcome our readers to the fascinating subject of optometry and its dynamic
landscape, which will definitely shape the way you practice today as well as in the
future. This book delves into the intricate science and artistry behind vision care,
exploring the marvels of the intricate mechanisms that provide us sight. Whether
you are an optometry graduate, a seasoned optometrist, or an ophthalmologist seek-
ing to deepen your understanding, or simply an enthusiast curious about the novel
wonders of visual assessment and rehabilitation, this book will illuminate and
inspire you.
Optometry is a discipline at the intersection of ophthalmology, physics, and tech-
nology, continuously evolving with advancements in science and innovation.
Optometry is more than just diagnosing and correcting refractive errors. It encom-
passes a holistic approach to eye health, encompassing preventative care, disease
management, and rehabilitation. Throughout this book, we emphasize the impor-
tance of comprehensive eye examinations, early detection of eye conditions, and the
role of optometrists as primary eye care providers.
Within these pages, we endeavour to capture not only the recent advances in the
field of optometry but also the established fundamentals. Our exploration begins
with the differences in global optometry practices, advances in the management of
ocular surface disorders and cornea, as well as glaucoma. From there, we venture
into the realms of diagnostics, examining the tools and techniques used to assess
colour vision and tailoring precise prescriptions for contact lenses, vision therapy,
and low vision aids. We also delve into the relevant clinical trials for optometrists,
novel ocular drug delivery modalities, and the application of artificial intelligence in
optometry.
As we journey through these pages, it is essential to remember that optometry is
not merely a profession but a calling—a commitment to safeguarding one of our
most precious senses, enriching lives, and empowering individuals to experience
the world with clarity and confidence.
We extend our heartfelt gratitude to all the contributors, researchers, educators,
and clinicians whose dedication and expertise have enriched the field of optometry
and made this book possible. We earnestly hope that this book serves as a valuable
resource, inspiring readers to explore, learn, and contribute to the remarkable field
of optometry.
v
vi Preface
With curiosity as our compass and knowledge as our guide, let us embark on this
illuminating journey into the world of optometric practices.
I would like to extend my deepest gratitude to all those who have contributed to
the creation of this book, from the authors who shared their expertise to the editorial
team who worked tirelessly to ensure its quality and accuracy. It is my sincere hope
that this book will serve as a beacon of inspiration and a valuable resource for those
seeking to explore the advances in the realm of optometry.
Parul Ichhpujani
vii
Contents
Differences in Optometry Practices Across the Globe������������������������������������ 1
Shivani Majithia and Sahil Thakur
Advances in Dry Eye Disease Diagnosis and Management���������������������������� 11
Rohan Bir Singh and Parul Ichhpujani
Advances in Diagnosis and Management of Infectious Keratitis������������������ 19
Bharat Gurnani and Kirandeep Kaur
Advances in Glaucoma�������������������������������������������������������������������������������������� 47
Parul Ichhpujani and Shibal Bhartiya
Advances in the Management of Oculomotor Dysfunction
in Adults and Children with Concussion �������������������������������������������������������� 59
Tiong Peng Yap and Cathy Stern
Advances in Color Vision Testing �������������������������������������������������������������������� 77
Sara I. Shoushtari and Rohan Bir Singh
Advances in Low Vision Aids���������������������������������������������������������������������������� 89
Samira Mortazavi and Asha Meloottu
Advances in Contact Lenses for Ophthalmic Drug Delivery ������������������������ 99
Parvin Shokrollahi and Alex Hui
Novel Ocular Drug Delivery Methods�������������������������������������������������������������� 115
Yuan Fang, Chris Hodge, Gerard Sutton, and Jingjing You
Clinical Trials Relevant for Optometrists�������������������������������������������������������� 131
Parul Ichhpujani and Surbhi Kapoor
Artificial Intelligence and Optometry: Transforming Practice
and Patient Care������������������������������������������������������������������������������������������������ 139
Shivani Majithia and Sahil Thakur
ix
Differences in Optometry Practices
Across the Globe
Key Points
• The World Council of Optometry provides the four categories of scopes of
optometry practices.
• These include the optical technology services (OT), visual function services
(VF), ocular diagnostic services (ODx), and ocular therapeutic services (OTx).
• Level 4 services are predominantly concentrated in regions like the UK, USA,
and Oceania, while the majority of global practices are Level 2 or Level 3.
1 Introduction
In the realm of eye care, two distinct yet interdependent professions, ophthalmology,
and optometry, play pivotal roles in safeguarding and enhancing our most treasured
sense: vision. Ophthalmology stands as the medical specialization tasked with the diag-
nosis and treatment of eye diseases, with a focus on surgical interventions. Optometry,
on the other hand, focuses on the assessment and correction of refractive errors and the
prescription of corrective lenses. The collaboration between these two professions is
integral to providing comprehensive eye care, but their individual roles and practices
vary considerably across the globe. Understanding these differences is crucial, as it not
only sheds light on the evolution of healthcare practices but also illuminates the socio-
cultural and economic factors shaping the delivery of eye care services.
S. Majithia
Education and Professional Services, Essilor Luxottica, Singapore
e-mail: [email protected]
S. Thakur (*)
Singapore Eye Research Institute, Singapore, Singapore
Medical Affairs, Mediwhale, Seoul, South Korea
The World Council of Optometry (WCO) defines the scope of optometry as, “a
healthcare profession that is autonomous, educated, and regulated (licensed/regis-
tered), and optometrists are the primary healthcare practitioners of the eye and visual
system who provide comprehensive eye and vision care, which includes refraction
and dispensing, detection/diagnosis and management of disease in the eye, and the
rehabilitation of conditions of the visual system [1].” This definition plays a pivotal
role in delineating the parameters of optometry, characterizing it as an independent,
knowledge-intensive healthcare discipline that operates within a regulated frame-
work of licensure and registration. The WCO offers several guidelines, consensus
documents and educational resources that can be used by stakeholders interested in
the profession [2, 3]. The WCO has outlined its 2021–2024 strategic plan, “One
Voice of Optometry” that has outlined four themes: inspiring optometric education;
integrating optometry in broader health sector; membership support and growth; and
future planning and collaborations with international bodies like WHO and IAPB
[3]. In addition to these advocacy activities, the WCO also provides a competency
based model of Scope of Practice in Optometry to assist states and countries around
the world to reconcile the variations in the scope of optometric practice internation-
ally. The competency model includes four categories of services that provide a verti-
cal ladder for individuals seeking to expand their career. The WCO defines four
categories of practice types [3]. They are outlined in Table 1.
Differences in Optometry Practices Across the Globe 3
The WCO currently has 36 country members and is the leading global body for
advocacy, education, policy development and humanitarian outreach for optome-
trists [4]. For the subsequent chapter, we will use the four levels of practice to cat-
egorize the optometry services in different global regions.
In Europe, optometry practices vary from country to country. The European Council
of Optometry and Optics (ECOO) represents the interests of optometrists and opti-
cians across Europe. The ECOO represent more than 150,000 opticians and optom-
etrists from 22 European Union (EU) countries. The European Diploma accredited
by the ECOO is set at the Bachelor level in European Higher Education and pro-
vides a qualification defined for optometric practice at WCO Category 3 [12]. The
ECOO also provides online resources on education, advocacy and professional con-
duct for use for professionals [13]. The United Kingdom (UK), however, has a sys-
tem where optometrists are trained to diagnose and treat a range of eye conditions,
prescribe medications, and refer patients to ophthalmologists when necessary. They
are regulated by the General Optical Council (GOC) through the Opticians Act
1989, and they are distinct from medical practitioners. Registration with the GOC is
mandatory for practicing optometry in the UK [14]. The optometrists complete a
4-year undergraduate honors degree followed by a 1-year internship. Successful
completion of assessments and supervised practice qualifies them to register with
the GOC and potentially join the College of Optometrists. Members of the College
of Optometrists can use the designation MCOptom. The College of Optometrists
also provides clinical management guidelines [15] for 62 diseases and drug formu-
lary [16] of 71 drugs that the optometrists can prescribe. To sum up, the United
Kingdom also showcases a firmly established and extensively specialized domain of
optometry, providing exceptional diagnostic and therapeutic solutions for maintain-
ing ocular health. Table 2 presents the standards of practice recommended by differ-
ent organizations, the GOC provides the most detailed 19 standards in that list [18].
Table 2 (continued)
Organization Country Standards
Australian Health Australia 1. Put patients first—Safe, effective, and collaborative
Practitioner practice
Regulation Agency 2. Aboriginal and Torres Strait islander health and cultural
[20] safety
3. Respectful and culturally safe practice for all
4. Working with patients
5. Working with other practitioners
6. Working within the healthcare system
7. Minimizing risk to patients
8. Professional behavior
9. Maintaining practitioner health and wellbeing
10. Teaching, supervising, and assessing
11. Ethical research
The codes and standards are available on the official website of the concerned regulator and advi-
sory bodies
a
The general guideline from the Optometry Council of India have been adopted from the section A
of the best practice in standards of Optometry handbook available at the official website [19]
provided the model curriculum for Bachelors (4 years) and Masters of Optometry
(2 years) courses [22]. It also describes the job scope of optometrists in India. The
scope fits into WCO Category 3 with additional provision to only use diagnostic
medications, namely mydriatics and cycloplegics [22]. Recently, the National
Commission for Allied & Healthcare Professionals Act, 2021 has been passed and
optometry has been recognized as a profession in India [23]. The introduction of
PhD programs in select institutions, such as the Elite School of Optometry in
Chennai, has significantly broadened the prospects and horizons available to optom-
etrists [24]. In summary, Indian optometry is thus poised to align with global coun-
terparts in the collective pursuit of eliminating avoidable blindness and propelling
advancements within various specialized realms of optometry. This transformation
is underscored by recent legislative and regulatory reforms that have streamlined
and fortified the profession’s trajectory.
In the Oceania region, Australia and New Zealand showcase comprehensive optom-
etry practices. Optometrists in these countries are trained to diagnose, manage, and
co-manage a variety of eye conditions. The Optometry Board of Australia is respon-
sible for registering optometrists and students, establishing profession-related stan-
dards, managing complaints and investigations, evaluating overseas practitioners,
and approving accreditation standards and courses [27]. The scope of optometry
practice includes therapeutic prescribing, enabling optometrists to treat certain eye
diseases. In the “Working Together for Better Eye Care” scope of optometry docu-
ment, Optometry Australia recommends actions that can be implemented to better
utilize the skills of optometrists to enhance patient access and increase the effi-
ciency of Australia’s eye health system [28]. Australia has a total of 6175 registered
optometrists who are actively practicing. These professionals are spread across
1200 communities throughout the country. Their geographic distribution closely
mirrors the overall population, although there are certain underserved areas in rural
and remote regions. Among the optometrists, approximately 56% are women, and
an estimated 72% fall under the age of 50, indicating a relatively youthful demo-
graphic profile [28, 29]. While optometrists in New Zealand are allowed to pre-
scribe oral medications for eye conditions like infections, inflammation, and
allergies, optometrists in Australia are limited to use of topical medications [28].
Advocacy groups are also campaigning for provision of intravitreal drug injection
privileges along with greater role in national screening programs for chronic dis-
eases like cataract, glaucoma, age related macular degeneration and diabetic reti-
nopathy [28].
8 S. Majithia and S. Thakur
The Middle East has a diverse range of optometry practices. In Saudi Arabia, the
Saudi Society of Optometry (SSO) is the main advocacy organization [26]. The
SSO plays a pivotal role in establishing a comprehensive scientific and educational
platform that empowers optometrists to acquire knowledge and contribute effec-
tively. It fosters health research in eye care, benefiting both the community and
professionals in the field. The society also drives professional development, paving
the way for a brighter future for the optometry profession in Saudi Arabia. By
equipping optometrists with the skills to provide primary eye health care and com-
bat blindness, the society significantly impacts public health. It leads efforts to
enhance regulatory aspects within the optometry sector and establishes valuable
collaborations with the private sector. Additionally, the society focuses on bolster-
ing financial resources while upholding the best governance and institutional prac-
tices. Through its initiatives, the SSO promotes societal engagement, delivers health
education, and ultimately elevates the public’s perception of the optometry profes-
sion. As of 2021, there were a total of 1886 practicing optometrists in Saudi Arabia
(64% male), with 45% practicing in Riyadh, according to the Saudi Commission for
Health Specialties [34]. While in Dubai, an optometrist is defined as, ‘a primary
healthcare professional for the eye concerned especially with examining the eye for
refractive error problems and prescribing spectacle lenses, contact lenses and low
vision rehabilitation to correct these related problems’ [35]. Thus, optometry ser-
vices in Dubai mainly provide primary eye and vision care, eye examinations to
Differences in Optometry Practices Across the Globe 9
detect vision problems and prescription of corrective lenses to correct those prob-
lems under Level 3 category of the WCO.
3 Summary
References
1. Concept of Optometry: World Council of Optometry. 2020. https://worldcouncilofoptometry.
info/concept-of-optometry. Accessed 27 Dec 2023.
2. Home: World Council of Optometry. 2023. https://worldcouncilofoptometry.info. Accessed
26 Dec 2023.
3. Resources: World Council of Optometry. 2022. https://worldcouncilofoptometry.info/
resources. Accessed 27 Dec 2023.
4. Members: World Council of Optometry. 2023. https://worldcouncilofoptometry.info/our-
members/#1597164269116-cdf0cb39-5831. Accessed 26 Dec 2023.
5. Association of Schools and Colleges of Optometry’s (ASCO). Optometry programs admissions
directory. Association of Schools and Colleges of Optometry, 2023. Accessed 27 Dec 2023.
6. American Optometric Association: Find Your State Board CE Requirements and Affiliate
Contacts. 2023. https://www.aoa.org/member-center/doctors-of-optometry/find-your-state-
board-or-affiliate-contact?sso=y. Accessed 27 Dec 2023.
7. Healio. Louisiana grants optometrists surgery privileges. 2014. https://www.healio.com/
news/optometry/20140603/louisiana-grants-optometrists-surgery-privileges. Accessed on 27
Dec 2023.
8. Optometry scope of practice in the United States. Eyes On Eyecare 2022. https://eyesoneyec-
are.com/resources/optometry-scope-of-practice-united-states. Accessed 27 Dec 2023.
9. Clinical Practice Guidelines. American Optometric Association 2023. https://www.aoa.org/
practice/clinical-guidelines/clinical-practice-guidelines?sso=y. Accessed 27 Dec 2023.
10. Gyawali R, Toomey M, Stapleton F, et al. Quality of 2019 American optometric association
clinical practice guideline for diabetic eye care. Ophthalmic Physiol Opt. 2021;41(1):165–70.
11. Optometrists. US Bureau of Labour Statistics. 2023. https://www.bls.gov/oes/current/
oes291041.htm. Accessed 25 Dec 2023.
12. European Diploma in Optometry | ECOO. 2023. https://ecoo.info/ecoo-accreditation-agency/
european-diploma-in-optometry. Accessed 25 Dec 2023.
13. Resources | ECOO. European Council of Optometry and Optics, 2023.
14. Working in the United Kingdom | General Optical Council. General Optical Council, 2023.
15. Clinical management guidelines. The College of Optometrists. 2023. https://www.college-
optometrists.org/clinical-guidance/clinical-management-guidelines.
10 S. Majithia and S. Thakur
Dry eye disease (DED) is an inflammatory response of the eye caused by decreased
production of tears, excessive evaporation of tears, or both [1]. It is a very common
disorder in the USA and affects roughly 5% of adults nationally [1].
1 Epidemiology
DED is more common in people aged 50 and older and it is more common in women
than in men [1]. Previous studies have found prevalence rates to vary from 5% to
34% worldwide [2]. The difference in prevalence between men and women is due
to the effect of female hormones on the lacrimal and meibomian glands [3]. Other
studies have estimated DED to affect 3.2 million women and 1.68 million men in
the USA [4].
Global, population-based studies have found prevalence rates to vary from 5% to
50% around the world. This range is due to discrepancies in diagnostic criteria and
what actually classifies as DED [3]. Additionally, for reasons that are unknown,
prevalence is found to be higher in Asian populations as compared to White popula-
tions. Climate, geographic location, and environmental factors may explain these
discrepancies [3]. DED prevalence also increases with age; as one gets older, tear
production naturally decreases to an extent, and impaired tear production is the
underlying issue behind DED [5].
R. B. Singh (*)
Harvard Medical School, Boston, MA, USA
e-mail: [email protected]
P. Ichhpujani
Department of Ophthalmology, Government Medical College and Hospital,
Chandigarh, India
The high prevalence of DED poses a great burden in terms of healthcare costs in
the USA, especially as the population continues to age. The annual cost to the aver-
age patient with DED is estimated to be $783 [2]. This equates to a $384 million
cost to the United States health care system every year [2]. Treatments for DED are
continuing to evolve but as the population continues to age, prevalence levels will
likely remain high nationwide.
2 Pathophysiology
DED is a caused by a variety of factors affecting the ocular surface and its tears. In
a structurally functional eye, the ocular surface is kept lubricated by the lacrimal
gland producing tears superiorly, and the meibomian sebaceous glands producing
oils and lipids inferiorly [1]. Damage to any of these structures can precipitate the
effects of DED. It is thought that external stressors on the ocular surface can trigger
the body’s natural immune response and thereby cause inflammation to these cru-
cial structures of the eye [2]. These external stressors may be environmental, anti-
genic, infectious, or genetic. The inflammatory response of chemokines, cytokines,
matrix metalloproteases, and the recruited T cells will then invade the ocular surface
and its surrounding glands, rendering them nearly nonfunctional [2].
The normal tear film is composed of three main layers: the lipid layer, the aque-
ous layer, and the mucin layer [3]. The lipid layer is the most superficial layer and
is made by the meibomian glands, which sit in the eyelid. The lipid component of
the tear serves to decrease its evaporation, thereby, allowing for adequate lubrica-
tion [3]. Next, the job of the lacrimal gland, along with the accessory lacrimal
glands, is to produce the middle, aqueous layer of the tear film. This layer is the
thickest of the three layers and contributes to an overall film thickness of 2–5 μm
[3]. Lastly, the base mucin layer is composed of glycoproteins, which are produced
by goblet cells of the conjunctiva [3].
In DED, these structures, and therefore the integrity of the tears, are compro-
mised. Inflammation of the lacrimal gland will lead to decreased production of the
aqueous layer, resulting in a thinner tear film and a less hydrated eye [3]. If the
meibomian glands are damaged due to inflammation, there will be no superficial
lipid component of the tear film, making it vulnerable to increased evaporation [3].
These two mechanisms are the basis for the two subtypes of DED.
3 Subtypes
mixed ADED and EDED [2]. While the mechanism of the two subtypes differs,
both mechanisms result in an impaired tear film, which leads to inadequate lubrica-
tion of the ocular surface. This decreased lubrication is the cause of the symptoms
that patients with DED experience.
4 Symptoms
Many symptoms of DED can be vague and nonspecific; these include redness of the
eye, burning or stinging, grittiness or the feeling of a foreign body in the eye, itchi-
ness, and photophobia. These symptoms while nonspecific can greatly impair a
patient’s quality of life and their ability to carry out everyday activities comfortably;
up to 60% of patients with DED report a reduced quality of life [2]. This can cause
concurrent anxiety and/or depression for many patients. Prevalence of depression in
patients with DED was roughly 5% higher when compared to the control group, one
study found [2]. Additionally, DED can impair vision, affecting a patient’s ability to
drive or read, further impairing their quality of life and contributing to their depres-
sive symptoms. Reduced reading speed and reduced driving reaction time, tested in
a driving simulator, were both notable findings in patients with DED [2].
Additional findings in patients with DED include signs of impaired meibomian
gland function, such as obstruction of the gland by a cloudy, granular excretion [2].
This must be removed by applying significant pressure on the lower eyelid where
the gland is located. This impaired gland function may also result in blepharitis or
meibomitis, or inflammation of the eyelid and the meibomian gland, respectively
[2]. The inflammation of the ocular structures in DED may lead to damage of the
ocular surface. This presents as small epithelial erosions on the ocular surface, also
known as superficial punctate keratitis [2]. Often this is indicated by temporal con-
junctival folds that are parallel to the lid margin.
DED can present similarly to allergies or certain infections, therefore, proper diag-
nosis is crucial to insure adequate treatment is provided. The first component of
diagnosis is obtaining a detailed patient history about the symptoms, their severity,
onset, and triggers as well as additional underlying health conditions and medica-
tions [2].
Next, examination of the eyelids can provide information about the condition
and possible severity. Factors such as reduced blink rate (promoting tear evapo-
ration), lid incongruity, and insufficient lid closure can also be seen. Examination
of the lids themselves can provide insight into the functioning, or lack thereof, of
the meibomian glands; a slit lamp can be used to visualize these in greater
detail [2].
Another finding suggestive of DED is temporal lid-parallel conjunctival folds
(LIPCOFs); this is a physical exam finding that is caused by increased friction
14 R. B. Singh and P. Ichhpujani
between the eyelids and the surface of the conjunctiva [2]. This increased friction
can be due to decreased lubrication from tears, therefore, indicating DED.
The slit lamp can also be used to visualize the ocular surface for signs of damage.
One common dye, lissamine green, will stain damaged surface cells with a defective
mucin layer, while fluorescein dye will highlight erosions in the conjunctiva and
cornea [2]. Fluorescein will also stain tear film. In DED, significant staining in the
region near the palpebral fissures will be seen [2].
Lastly, the tear film itself will be examined. The height of the tear film can be
measured during a slit lamp examination and can be indicative of DED. The stabil-
ity of the tear film is also measured via the tear film break-up time (TFBUT) [2]. A
shorter TFBUT is often indicative of some underlying pathology [2]. Additional
tests are present; however, they are subject to high variability and not often required
to make the diagnosis [2].
Treatment for DED is primarily aimed at alleviating symptoms since it is a
chronic disease. Patients should also be educated about their condition and certain
aggravating factors such as dry air, smoke, or use of contact lenses [2]. Artificial
tears are the main treatment for patients with DED; no studies have been done com-
paring different types of artificial tears, however. In general, it is known that artifi-
cial tears reduce ocular surface friction, improve tear film integrity, and increase
patients’ quality of life [2]. For patients with hyperevaporative DED, artificial tears
containing a lipid component have been shown to improve tear film stability and
meibomian gland function [2].
Tear-soluble factors such as cytokines and chemokines are the best surrogate mark-
ers of DED severity and can help in disease classification and treatment planning.
Increased levels of cytokines, viz., interleukin-1β (IL-1β), IL-2, IL-4, IL-6, IL-9,
IL-12, IL-17A, interferon-gamma (IFN-γ), tumor necrosis factor-alpha (TNF-α);
chemokines (CCL2, CCL3, CCL4, C-X-C motif chemokine ligand 8; CXCL8);
MMP-9, FGF, VEGF-A; soluble receptors (sICAM-1, sTNFR1), neurotrophic fac-
tors (NGF, substance P, serotonin) and IL1RA and reduced levels of IL-7, IL-17F,
CXCL1, CXCL10, EGF, and lactoferrin have been found in DED [6].
Anti-inflammatory treatment is also recommended in patients with more severe
forms of DED [2]. This is because the underlying mechanism of DED is thought to
be an innate inflammatory response infiltrating the ocular surface structures. Chronic
inflammatory insult to the ocular surface leads to the invasion of blood and lym-
phatic vessels into the cornea, which contributes to cellular trafficking [6]. During
the inflammatory siege, granulocyte–macrophage–colony-stimulating factor
(GM-CSF) secretion adds fuel to fire by the induction of monocytes/macrophages.
Corticosteroid eye drops are also available for patients with a more severe form
of DED [2]. Again, this is likely due to reduced inflammation within the ocular
surface structures that contribute to the DED symptoms. Lastly, certain immuno-
modulators and antimicrobial agents have shown some efficacy in reducing
Advances in Dry Eye Disease Diagnosis and Management 15
6 Risk Factors
There are many potential risk factors for the development of DED, some more sup-
ported than others. Risk factors that are supported by a high level of evidence
include age and female sex, as discussed before, post-menopausal hormone replace-
ment therapy (HRT), and androgen insensitivity syndrome (AIS) [2]. Studies have
shown that the meibomian gland dysfunction (MGD) occurs because of the hor-
mones used in HRT and the lack of hormonal response in AIS [8, 9]. NOV03 (per-
fluorohexyloctane; under review with the US Food and Drug Administration [FDA])
is an emerging therapy for DED associated with MGD. This drug has shown signifi-
cant improvements in both signs and symptoms [10].
Intense pulsed light (IPL) therapy, involving treatment over the eyelids, has
emerged as a potential treatment for MGD by causing liquefaction of meibum,
thrombosis of telangiectatic blood vessels in the eyelids, changes in meibomian
gland architecture and photomodulation. A recent Cochrane review has shown that
LipiFlow performs similarly to other commonly used DED treatment modalities as
regards the DED signs and symptoms. A high level of bias was seen in the best
available evidence, resulting in low certainty evidence [11]. In addition to the rela-
tively high costs, IPL treatment may cause depigmentation or swelling of the peri-
ocular skin, eyelash loss or permanent iris transillumination defects.
Additional highly supported risk factors include anything that may affect the
integrity of the ocular surface, such as collagen vascular disease, corneal refractive
surgery, irradiation, vitamin A deficiency, and stem cell transplantation [2]. Patients
using certain antihistamine drugs may also be at an increased risk of DED due to
their mechanism of action, which decreases tear production as a side effect [12].
Lastly, for reasons that are unknown, patients with Hepatitis C are at a higher risk
for developing DED [13].
Other possible risk factors for DED that are moderately supported by research
include diabetes mellitus, isotretinoin, systemic chemotherapies, keratoplasty, sar-
coidosis, HIV infection, and certain medications such as tricyclic antidepressants,
selective serotonin reuptake inhibitors, and beta blockers [2]. Smoking, alcohol use,
Botox injections, oral contraceptives, gout, and pregnancy are other possible risk
factors for the development of DED, however, they are not well supported [2].
A point-of-care diagnostic kit, Bio-M Pathfinder (NovoMol-Dx, India, a custom-
ized version of the Ella™ Automated ELISA system, Bio-Techne®Corporation,
Minnesota, USA) has been recently introduced in the Indian market for clinical use.
This kit will help to identify the molecular status in patients with DED and addition-
ally, help define the risk of development of DED in clinically healthy eyes planned
for invasive procedures [6]. This will help in tailored management of these individu-
als (“personalized medicine”).
16 R. B. Singh and P. Ichhpujani
7 Complications
Late stages of DED can progress to more severe symptoms, leading to ocular health
complications. The consequences of the surface inflammation and the epithelial ero-
sions can progress to conjunctival scarring, permanent epithelial defects, ulceration,
or in very severe cases, corneal ulceration [2]. These complications may even lead
to vision loss [3]. However, the severe complications listed above are very rare and
often precipitated by underlying health issues and autoimmune syndromes. These
include Stevens-Johnson Syndrome, Sjögren’s Syndrome, xerophthalmia and vita-
min A deficiency, and graft-versus-host disease [2].
All in all, DED is a self-limited disease especially once symptoms are treated. Most
patients see a reduction in symptoms as time progresses with the treatment, with no
major progression of symptoms [4]. Some patients with a severe form of DED ini-
tially may report a worsening severity down the line, as well as patients with a lower
level treatment course [4]. More research is needed to accurately track the progres-
sion of DED in patients with varying degrees of severity, along with the efficacy of
different levels of treatment courses. Additionally, further research on the underly-
ing inflammatory pathways triggered in DED must be conducted in order to develop
more effective therapy for those with severe disease progression [6].
References
1. Walter K. What is dry eye disease? JAMA. 2022;328(1):84.
2. Messmer EM. The pathophysiology, diagnosis, and treatment of dry eye disease. Dtsch Arztebl
Int. 2015;112(5):71.
3. Golden MI, Meyer JJ, Patel BC. Dry eye syndrome. St. Petersburg: StatPearls; 2023. Accessed
15 Dec 2023
4. Lienert JP, Tarko L, Uchino M, Christen WG, Schaumberg DA. Long-term natural history of
dry eye disease from the patient’s perspective. Ophthalmology. 2016;123(2):425.
5. Zhang X, Wang L, Zheng Y, Deng L, Huang X. Prevalence of dry eye disease in the elderly: a
protocol of systematic review and meta-analysis. Medicine. 2020;99(37):E22234.
6. Kumar NR, Praveen M, Narasimhan R, Khamar P, D’Souza S, Sinha-Roy A, Sethu S, Shetty R,
Ghosh A. Tear biomarkers in dry eye disease: progress in the last decade. Indian J Ophthalmol.
2023;71(4):1190–202.
7. Ling J, Chan BCL, Tsang MSM, et al. Current advances in mechanisms and treatment of
dry eye disease: toward anti-inflammatory and immunomodulatory therapy and traditional
Chinese medicine. Front Med (Lausanne). 2021;8:815075.
8. Cermak JM, Krenzer KL, Sullivan RM, Dana MR, Sullivan DA. Is complete androgen insen-
sitivity syndrome associated with alterations in the meibomian gland and ocular surface?
Cornea. 2003;22(6):516–21.
9. Schaumberg DA, Buring JE, Sullivan DA, Reza Dana MR. Hormone replacement therapy and
dry eye syndrome. JAMA. 2001;286(17):2114–9.
Advances in Dry Eye Disease Diagnosis and Management 17
10. Sheppard JD, Nichols KK. Dry eye disease associated with meibomian gland dysfunc-
tion: focus on tear film characteristics and the therapeutic landscape. Ophthalmol Ther.
2023;12(3):1397–418.
11. Pucker AD, Yim TW, Rueff E, Ngo W, Tichenor AA, Conto JE. LipiFlow for the treatment of
dry eye disease. Cochrane Database Syst Rev. 2024;2(2):CD015448.
12. Katipoğlu Z, Abay RN. The relationship between dry eye disease and anticholinergic burden.
Eye (Lond). 2023;37(14):2921–5.
13. Rajalakshmy AR, Malathi J, Madhavan HN, Bhaskar S, Iyer GK. Patients with dry eye without
hepatitis C virus infection possess the viral RNA in their tears. Cornea. 2015;34(1):28–31.
Advances in Diagnosis and Management
of Infectious Keratitis
1 Introduction
Infectious keratitis (IK) is an acute or chronic infection of the cornea that poses a
significant risk of visual impairment [1, 2]. It is a serious ocular condition that can
result in corneal scarring, thinning, and perforation, potentially leading to blindness
if not correctly managed. The etiology of IK is diverse, with bacterial, viral, fungal,
and parasitic organisms all known to be causative agents [1]. The global burden of
the disease is substantial, particularly in developing countries where access to health
care is limited and risk factors, such as the use of traditional eye medications and
poor hygiene practices, are prevalent [1, 3].
Current research on the occurrence of IK is limited, with most studies being
over a decade old. The incidence rates of IK vary significantly based on geographic
location and research methodology, ranging from 2.5 to 799 cases per 100,000
people annually [3–5]. This variation is particularly notable between high-income
and low-income countries. For instance, past research indicated an incidence of
2.5–27.6 per 100,000 people per year in the USA and 2.6–40.3 per 100,000 people
per year in the United Kingdom. Recent findings from a study in Nottingham, UK,
align with these older studies, showing a steady incidence of 34.7 per 100,000
people per year from 2007 to 2019, suggesting a continuous presence of IK in
developed countries [6].
Similarly, a study in Australia reported a lower incidence of 6.6 per 100,000 people
per year between 2005 and 2015 [7]. However, these figures might be underestimates,
B. Gurnani (*)
Cataract, Cornea, External Diseases, Trauma, Ocular Surface and Refractive Surgery,
Gomabai Netralaya and Research Centre, Neemuch, India
K. Kaur
Cataract, Pediatric Ophthalmology, Strabismus and Neuro-Ophthalmology, Gomabai
Netralaya and Research Centre, Neemuch, India
as they were derived from cases requiring corneal scraping. Conversely, much higher
rates of IK have been reported in less affluent countries. For example, South India
recorded an incidence of 113 per 100,000 people per year, and Nepal reported a stag-
gering 799 per 100,000 people per year [8]. The increased incidence in these areas can
be attributed to factors like poor environmental and personal hygiene, lower educa-
tional levels, the prevalence of agricultural work, heightened risk of work-related cor-
neal injuries, and limited access to sanitation and healthcare facilities [1].
The risk factors for infectious keratitis can be varied and are often associated
with conditions that compromise the integrity or immune defense of the eye [9].
Some of the primary risk factors include contact lens use, ocular trauma, compro-
mised immune function, exposure to contaminated water, poor eyelid hygiene, pre-
vious eye infections or surgeries, use of topical corticosteroids, dry eye syndrome,
wearing decorative contact lenses, and environmental factors [10]. Addressing these
risk factors through proper eye care, hygiene, and caution in potentially hazardous
environments can significantly reduce the likelihood of developing infectious kera-
titis [2]. The diagnosis of infectious keratitis is primarily clinical but requires a high
index of suspicion, especially in individuals presenting with risk factors such as
contact lens use, recent ocular trauma, pre-existing ocular surface diseases, or sys-
temic immunodeficiency [11]. Clinically, patients with infectious keratitis may
present with a range of symptoms, including ocular pain, redness, photophobia,
decreased vision, and discharge. A thorough ophthalmic examination using a slit-
lamp microscope may reveal an epithelial defect, focal infiltrate, stromal edema,
and, in some cases, an overlying hypopyon [12].
The management of IK must be aggressive and tailored to the type of pathogen
suspected. Bacterial keratitis, often caused by Staphylococcus, Streptococcus, and
Pseudomonas species, is typically treated with broad-spectrum topical antibiotics,
which are later refined based on microbial culture and sensitivity results [9]. Viral
keratitis, predominantly due to the herpes simplex virus, requires antiviral therapy
[13]. Fungal keratitis, caused by organisms such as Fusarium or Aspergillus, is
managed with topical antifungal agents [14]. Parasitic infections like Acanthamoeba
keratitis, although less common, are particularly challenging to treat and necessitate
the use of topical anti-amoebic medications [15].
Laboratory testing plays a pivotal role in diagnosing and managing infectious
keratitis. Cultures from corneal scrapings can identify the causative organism and
guide targeted therapy. Molecular techniques like polymerase chain reaction (PCR)
offer rapid and sensitive detection of microbial DNA [16]. In vivo, confocal micros-
copy and anterior segment optical coherence tomography (ASOCT) are valuable
non-invasive imaging modalities that can provide detailed visualization of the cor-
neal layers and assist in diagnosing and monitoring treatment response [16].
The depth and severity of the infection also influence the choice of management
strategy. For superficial keratitis, topical medications may suffice. However, forti-
fied antibiotics or combination therapy may be necessary for deeper infections. In
cases where there is no response to medical therapy or in the presence of complica-
tions such as impending perforation, surgical intervention becomes essential.
Advances in Diagnosis and Management of Infectious Keratitis 21
2 Pathophysiology
3 Clinical Presentation
Infectious keratitis is a serious eye infection that affects the cornea, often caused by
bacteria, viruses, fungi, or parasites. The symptoms of infectious keratitis can vary
depending on the underlying cause but generally include severe eye pain, redness,
and irritation [2]. Patients may also experience blurred vision or sensitivity to light
(photophobia) as the infection progresses. It is common for the affected eye to pro-
duce excessive tears or a thick, sticky discharge. The cornea may appear cloudy or
have visible white spots in more advanced cases, indicating an ulcer or severe infec-
tion [22]. These symptoms can reduce vision quality and, if left untreated, might
result in permanent vision loss. Early detection and treatment are crucial to prevent
complications. If any of these symptoms are present, especially following an eye
injury or contact lens use, seeking immediate medical attention is advised [9].
Differentiating between bacterial, viral, fungal, acanthamoeba, and Pythium
keratitis is crucial for effective treatment, as the signs and symptoms vary between
these infections. Bacterial keratitis typically presents with a rapid onset of pain, red-
ness, and discharge, with the cornea showing a dense, central, white infiltrate that
may develop into an ulcer (Fig. 1). Staphylococcus and Pseudomonas are common
culprits, and they often cause a ‘hypopyon,’ a visible layer of white blood cells in
the anterior chamber of the eye [9].
Viral keratitis, often caused by the herpes simplex virus, may start with a less
aggressive presentation, including redness, tearing, and blurred vision. The key sign
is a dendritic ulcer, which can be seen on corneal staining with fluorescein and pres-
ents as branching lesions on the cornea (Fig. 2) [23].
Fungal keratitis, which can be due to various fungi like Fusarium or Aspergillus,
typically has a slower onset. It presents with a grayish, feathery infiltrate and may
include satellite lesions—small, separate opacities that surround the main infiltra-
tion. Patients may also experience a more significant sensation of a foreign body in
the eye [21]. Hypopyon is usually immobile and maybe convex (Fig. 3).
24 B. Gurnani and K. Kaur
4 Diagnostic Approach
• Clinical Examination
Initial assessment includes a patient history to identify risk factors such as con-
tact lens use, recent eye trauma, or exposure to contaminated water. Symptom
evaluation will focus on pain, redness, discharge, and visual changes [26].
• Slit-Lamp Biomicroscopy
This is a critical tool in the evaluation of keratitis. A slit lamp allows for a detailed
examination of the cornea, conjunctiva, and anterior chamber. It helps to identify
the location and depth of the corneal lesion, epithelial defect, depth and extent of
Advances in Diagnosis and Management of Infectious Keratitis 25
flap for lesions situated mid-stromal, such as those seen in infectious crystalline
keratopathy, or for deeper stromal infections like fungal keratitis. This approach
helps in obtaining a sample from the precise depth of the lesion.
5 Differential Diagnosis
Infectious keratitis typically presents with pain, photophobia, reduced vision, and
signs of infection like corneal infiltrates, edema, and ulceration. Non-infectious
keratitis may have similar symptoms but often lacks purulent discharge or marked
anterior chamber reaction. A history of trauma, contact lens wear, or prior ocular
surgery may suggest an infectious etiology, whereas autoimmune conditions or dry
eye syndrome might point toward non-infectious causes [36].
Bacterial Keratitis
It is a critical ocular condition that must be treated promptly due to its potential for
rapid progression and severe complications. Immediate initiation of empirical anti-
biotic therapy is essential. This often involves a combination regimen covering a
broad spectrum of Gram-positive and Gram-negative organisms. The standard treat-
ment protocol typically includes:
When comparing fluoroquinolones with fortified eye drops, several factors are
considered:
The Steroid for Corneal Ulcer Trial (SCUT) did not demonstrate a significant
difference in the best-corrected visual acuity at 3 months between patients treated
with topical corticosteroids versus placebo as an adjunctive therapy in the manage-
ment of bacterial corneal ulcers. Additionally, the use of corticosteroids did not
show an increased risk of corneal perforation [9].
Fungal Keratitis
Antifungal Agents
Agents like natamycin, amphotericin B, or voriconazole are used depending on the
type of fungus. Treatment duration is typically longer than for bacterial infections.
The principal categories of antifungal agents encompass:
The typical treatment regimen for fungal infections follows a stepwise approach:
Topical Antifungals: The preferred choice is 5% Natamycin, administered
every hour during the day and every 2 h at night. Based on the clinical response, the
frequency is gradually reduced after 4–7 days [14].
Surface Debridement: This procedure aims to eliminate slough and reduce the
infection load. Though its benefits are debated, it is still widely used by clinicians.
Adjustments for Worsening Conditions: If there is no improvement after
14 days, options include adding 0.15% Amphotericin B drops or 1% Voriconazole
and revisiting the culture report.
Duration of Therapy: Treatment typically spans 3–4 weeks, with complete
resolution often taking 4–8 weeks [21].
32 B. Gurnani and K. Kaur
Acanthamoeba Keratitis
Anti-amoebic Drugs
Combination therapy, including topical chlorhexidine, polyhexamethylene bigua-
nide (PHMB), and propamidine isethionate, is effective. Oral agents like itracon-
azole or miltefosine may be added for severe cases.
The treatment of Acanthamoeba keratitis is challenging and often involves a
combination of medications:
• Biguanides: The primary agents used are PHMB and chlorhexidine. These
agents are effective against both the cystic and trophozoite forms of Acanthamoeba.
• Aminoglycosides: Drugs like neomycin may be used in combination with bigu-
anides due to their efficacy against the trophozoite form [15].
• Azole Antifungals: Ketoconazole, fluconazole, or voriconazole can be effective
due to their anti-acanthamoeba properties.
• Propamidine Isethionate and Hexamidine: These are effective against the tro-
phozoite form and can be used as adjunctive therapy.
• Topical Steroids: The use of steroids is controversial and typically avoided in
the initial phase due to the potential exacerbation of the infection. Steroids may
be considered in later stages to control inflammation after infection control.
• Pain Management: Cycloplegics can be used for pain relief and to manage
photophobia. Oral analgesics may also be necessary.
Advances in Diagnosis and Management of Infectious Keratitis 33
In summary, the treatment of PIK has evolved with a greater emphasis on anti-
bacterials over antifungals, supported by recent research. Adjuvant therapies play a
significant role in managing symptoms and preventing complications. Regular
34 B. Gurnani and K. Kaur
monitoring and adjustments based on clinical response are key to successful treat-
ment outcomes [24].
Surgical Management
Indications for Surgical Intervention
• Unresponsive to medical therapy.
• Progressive corneal thinning or perforation.
• Significant visual axis opacification after the resolution of infection.
Procedures
• Debridement: Removal of infected or necrotic tissue to promote healing.
• Keratoplasty: Corneal transplantation may be necessary for tectonic support or
visual rehabilitation.
• Amniotic Membrane Transplantation: Used for promoting healing, reducing
inflammation, and as a temporary patch for small perforations [39].
Infectious keratitis, a serious eye infection affecting the cornea, can have various
complications and a varied prognosis depending on several factors, including the
cause of the infection, the patient’s overall health, and the timeliness and effective-
ness of treatment.
• Corneal Scarring: This is one of the most common complications. The infec-
tion can lead to scarring, which may cause permanent vision impairment or even
blindness if the scar is centrally located.
• Corneal Ulceration: The infection can progress to create ulcers on the cornea.
Deep ulcers can lead to corneal perforation, a severe condition that can result in
vision loss.
• Secondary Infections: If the primary infection is not adequately treated, it can
lead to secondary infections in the eye or other parts of the body.
• Chronic Eye Pain: Some patients experience ongoing eye pain, even after con-
trolling the infection.
• Hypopyon is the accumulation of white blood cells in the anterior chamber of
the eye, indicating a severe infection.
• Spread of Infection: In rare cases, the infection can spread to other parts of the
eye, leading to conditions like endophthalmitis, which is an inflammation of the
interior of the eye and can be sight-threatening [42].
• Early Diagnosis and Treatment: If diagnosed and treated early, many patients
recover completely without any permanent vision loss [11].
• Type of Infectious Agent: The causative organism affects the prognosis.
Bacterial keratitis generally has a better prognosis than fungal or parasitic kera-
titis, which are often more challenging to treat [11, 12].
• Underlying Health Conditions: Patients with pre-existing eye conditions or
systemic health issues, like diabetes or a compromised immune system, may
have a poorer prognosis.
• Adherence to Treatment: The patient’s adherence to treatment protocols,
including the use of prescribed medications and follow-up appointments, signifi-
cantly impacts the outcome.
• Severity at Presentation: More severe infections at the time of presentation
tend to have a worse prognosis, particularly if the central cornea is involved or if
there is a deep corneal ulcer.
• Recurrence: Some forms of keratitis, particularly those caused by herpes sim-
plex virus, can recur, potentially worsening the long-term prognosis [11].
Preventing infectious keratitis involves several public health strategies and personal
hygiene practices, particularly for individuals who wear contact lenses, are exposed
to environmental risks, or have pre-existing ocular conditions [17]. Here is a detailed
write-up on the prevention of infectious keratitis under the specified heads:
• Proper Hand Washing: Always wash hands with soap and water and dry them
with a lint-free towel before handling contact lenses.
• Disinfect Contact Lenses: Use only the recommended type of disinfecting solu-
tion to clean and store contact lenses. Never use tap water, saliva, or non-sterile
solutions [20].
• Replace Lenses as Directed: Follow the eye care professional’s recommenda-
tions for replacement intervals for contact lenses and cases.
• Avoid Sleeping in Contact Lenses: Unless specifically designed for overnight
wear, do not sleep in contact lenses to reduce the risk of infection [20].
• Respect Water Exposure Guidelines: Do not swim or shower while wearing
contact lenses to prevent exposure to waterborne pathogens.
Advances in Diagnosis and Management of Infectious Keratitis 37
• No Lens Sharing: Never share contact lenses with others. This can spread
microorganisms that may lead to infection.
• Regular Eye Examinations: Routine eye check-ups can help in the early detec-
tion and treatment of ocular surface diseases, preventing the progression to
keratitis.
• Manage Dry Eye: Adequate treatment of dry eye syndrome is crucial as it can
compromise the ocular surface and make it more susceptible to infections.
• Treat Lid Margin Diseases: Conditions such as blepharitis should be managed
promptly to maintain a healthy ocular surface.
• Control Inflammation: The use of anti-inflammatory medications, as prescribed,
can help control ocular surface inflammation and reduce the risk of infection [12].
9 Recent Advances
• Genetic Markers: Research into genetic markers that may predispose individu-
als to certain types of keratitis could allow for earlier identification and targeted
prevention strategies.
• Tear Fluid Analysis: The analysis of tear fluid for specific enzymes or inflam-
matory markers associated with infection can facilitate early detection and treat-
ment [48].
• In Vivo Confocal Microscopy: This imaging technique allows for the
non-invasive examination of the cornea at a cellular level, which can help in
the early diagnosis of microbial keratitis before clinical signs become
apparent [32].
• Herpes Simplex Virus (HSV) Vaccine: While still under development, there is
ongoing research into vaccines for HSV, which is a common cause of viral kera-
titis. The aim is to reduce the incidence and severity of HSV-related ocular dis-
ease [49].
• Adenovirus Vaccine: Research is also being conducted into vaccines for adeno-
virus, which causes epidemic keratoconjunctivitis and can lead to keratitis [50].
• Public Health Vaccination Programs: The implementation of widespread vac-
cination programs could significantly reduce the incidence of viral keratitis, par-
ticularly in regions where it is endemic.
10 Future Directions
The future directions in managing and understanding infectious keratitis are driven
by ongoing research and the evolving landscape of microbial resistance. Addressing
research gaps, exploring the potential of gene therapy, and the need for new antibi-
otics are at the forefront of this field. Here is a detailed write-up on these aspects:
11 Conclusion
References
1. Ting DSJ, Ho CS, Deshmukh R, Said DG, Dua HS. Infectious keratitis: an update on epide-
miology, causative microorganisms, risk factors, and antimicrobial resistance. Eye (Lond).
2021;35(4):1084–101.
Advances in Diagnosis and Management of Infectious Keratitis 43
2. Cabrera-Aguas M, Khoo P, Watson SL. Infectious keratitis: a review. Clin Exp Ophthalmol.
2022;50(5):543–62.
3. Ung L, Bispo PJM, Shanbhag SS, Gilmore MS, Chodosh J. The persistent dilemma of
microbial keratitis: global burden, diagnosis, and antimicrobial resistance. Surv Ophthalmol.
2019;64(3):255–71.
4. Erie JC, Nevitt MP, Hodge DO, Ballard DJ. Incidence of ulcerative keratitis in a defined popu-
lation from 1950 through 1988. Arch Ophthalmol. 1993;111(12):1665–71.
5. Upadhyay MP, Karmacharya PC, Koirala S, Shah DN, Shakya S, Shrestha JK, Bajracharya H,
Gurung CK, Whitcher JP. The Bhaktapur eye study: ocular trauma and antibiotic prophylaxis
for the prevention of corneal ulceration in Nepal. Br J Ophthalmol. 2001;85(4):388–92.
6. Jeng BH, Gritz DC, Kumar AB, Holsclaw DS, Porco TC, Smith SD, Whitcher JP, Margolis
TP, Wong IG. Epidemiology of ulcerative keratitis in Northern California. Arch Ophthalmol.
2010;128(8):1022–8.
7. Green M, Carnt N, Apel A, Stapleton F. Queensland Microbial Keratitis Database: 2005–2015.
Br J Ophthalmol. 2019;103(10):1481–6.
8. Gonzales CA, Srinivasan M, Whitcher JP, Smolin G. Incidence of corneal ulceration in
Madurai district, South India. Ophthalmic Epidemiol. 1996;3(3):159–66.
9. Al-Mujaini A, Al-Kharusi N, Thakral A, Wali UK. Bacterial keratitis: perspective on epi-
demiology, clinico-pathogenesis, diagnosis and treatment. Sultan Qaboos Univ Med
J. 2009;9(2):184–95.
10. Lin TY, Yeh LK, Ma DH, Chen PY, Lin HC, Sun CC, Tan HY, Chen HC, Chen SY, Hsiao
CH. Risk factors and microbiological features of patients hospitalized for microbial keratitis: a
10-year study in a referral center in Taiwan. Medicine (Baltimore). 2015;94(43):e1905.
11. Gurnani B, Kaur K. Bacterial keratitis. In: StatPearls [Internet]. Treasure Island: StatPearls
Publishing; 2023. PMID: 34662023.
12. Singh P, Gupta A, Tripathy K. Keratitis. In: StatPearls [Internet]. Treasure Island: StatPearls
Publishing; 2023. PMID: 32644440.
13. Labib BA, Chigbu DI. Clinical management of herpes simplex virus keratitis. Diagnostics.
2022;12(10):2368.
14. Atta S, Perera C, Kowalski RP, Jhanji V. Fungal keratitis: clinical features, risk factors, treat-
ment, and outcomes. J Fungi. 2022;8(9):962.
15. Varacalli G, Di Zazzo A, Mori T, Dohlman TH, Spelta S, Coassin M, Bonini S. Challenges in
acanthamoeba keratitis: a review. J Clin Med. 2021;10(5):942.
16. Gupta N, Tandon R. Investigative modalities in infectious keratitis. Indian J Ophthalmol.
2008;56(3):209–13.
17. Gokhale NS. Medical management approach to infectious keratitis. Indian J Ophthalmol.
2008;56(3):215–20.
18. Carnt N, Stapleton F. Strategies for the prevention of contact lens-related Acanthamoeba kera-
titis: a review. Ophthalmic Physiol Opt. 2016;36(2):77–92.
19. Austin A, Lietman T, Rose-Nussbaumer J. Update on the management of infectious keratitis.
Ophthalmology. 2017;124(11):1678–89.
20. Fleiszig SMJ, Kroken AR, Nieto V, Grosser MR, Wan SJ, Metruccio MME, Evans DJ. Contact
lens-related corneal infection: intrinsic resistance and its compromise. Prog Retin Eye Res.
2020;76:100804.
21. Wu J, Zhang WS, Zhao J, Zhou HY. Review of clinical and basic approaches of fungal kerati-
tis. Int J Ophthalmol. 2016;9(11):1676–83.
22. Klotz SA, Penn CC, Negvesky GJ, Butrus SI. Fungal and parasitic infections of the eye. Clin
Microbiol Rev. 2000;13(4):662–85.
23. Koganti R, Yadavalli T, Naqvi RA, Shukla D, Naqvi AR. Pathobiology and treatment of viral
keratitis. Exp Eye Res. 2021;205:108483.
24. Gurnani B, Kaur K. Understanding barriers, recommended solutions, and future prospects
for the diagnosis and management of Pythium insidiosum keratitis. Indian J Ophthalmol.
2023;71(12):3584–6.
44 B. Gurnani and K. Kaur
25. Cao B, Gonugunta VT, Radhakrishnan N, Lalitha P, Gurnani B, Kaur K, Iyer G, Agarwal S,
Srinivasan B, Keenan JD, Prajna NV. Outcomes of Pythium keratitis: a meta-analysis of indi-
vidual patient data. Curr Ophthalmol Rep. 2022;10(4):198–208.
26. Ting DSJ, Gopal BP, Deshmukh R, Seitzman GD, Said DG, Dua HS. Diagnostic armamen-
tarium of infectious keratitis: a comprehensive review. Ocul Surf. 2022;23:27–39.
27. Dahlgren MA, Lingappan A, Wilhelmus KR. The clinical diagnosis of microbial keratitis. Am
J Ophthalmol. 2007;143(6):940–4.
28. Moshirfar M, Hopping GC, Vaidyanathan U, Liu H, Somani AN, Ronquillo YC, Hoopes
PC. Biological staining and culturing in infectious keratitis: controversy in clinical utility.
Med Hypothesis Discovery Innovation Ophthalmol. 2019;8(3):145–51.
29. Toumasis P, Tsantes AG, Tsiogka A, Samonis G, Vrioni G. From clinical suspicion to diag-
nosis: a review of diagnostic approaches and challenges in fungal keratitis. J Clin Med.
2024;13(1):286.
30. Sridhar MS, Sharma S, Gopinathan U, Rao GN. Anterior chamber tap: diagnostic and thera-
peutic indications in the management of ocular infections. Cornea. 2002;21(7):718–22.
31. Gurnani B, Kaur K. Predicting prognosis based on regional prevalence, ulcer morphology
and treatment strategy in vision-threatening Pythium insidiosum keratitis. Clin Ophthalmol.
2023;17:1307–14.
32. Brasnu E, Bourcier T, Dupas B, Degorge S, Rodallec T, Laroche L, Borderie V, Baudouin C. In
vivo confocal microscopy in fungal keratitis. Br J Ophthalmol. 2007;91(5):588–91.
33. Abdelghany AA, D'Oria F, Alio Del Barrio J, Alio JL. The value of anterior segment opti-
cal coherence tomography in different types of corneal infections: an update. J Clin Med.
2021;10(13):2841.
34. Gupta N, Varshney A, Ramappa M, Basu S, Romano V, Acharya M, Gaur A, Kapur N, Singh
A, Shah G, Chaudhary I, Patel N, Tiwari A, Kate A, Sangwan V, Mathur U. Role of AS-OCT
in managing corneal disorders. Diagnostics. 2022;12(4):918.
35. Agarwal S, Khan TA, Vanathi M, Srinivasan B, Iyer G, Tandon R. Update on diagnosis and
management of refractory corneal infections. Indian J Ophthalmol. 2022;70(5):1475–90.
36. Srinivasan M, Mascarenhas J, Prashanth CN. Distinguishing infective versus noninfective
keratitis. Indian J Ophthalmol. 2008;56(3):203–7.
37. Gurnani B, Christy J, Narayana S, Rajkumar P, Kaur K, Gubert J. Retrospective mul-
tifactorial analysis of Pythium keratitis and review of literature. Indian J Ophthalmol.
2021;69(5):1095–101.
38. Gurnani B, Kaur K, Kumar T. Commentary: current concepts, recent updates, and future treat-
ment options for Pythium insidiosum keratitis. Indian J Ophthalmol. 2023;71(5):1874–6.
39. Said DG, Rallis KI, Al-Aqaba MA, Ting DSJ, Dua HS. Surgical management of infectious
keratitis. Ocul Surf. 2023;28:401–12.
40. Upadhyay MP, Srinivasan M, Whitcher JP. Diagnosing and managing microbial keratitis.
Community Eye Health. 2015;28(89):3–6.
41. Alio JL, Abbouda A, Valle DD, Del Castillo JM, Fernandez JA. Corneal cross linking and
infectious keratitis: a systematic review with a meta-analysis of reported cases. J Ophthalmic
Inflamm Infect. 2013;3(1):47.
42. Agrawal V. First aid for complications of infectious keratitis. Indian J Ophthalmol.
2008;56(3):221–2.
43. Mba IE, Nweze EI. Antimicrobial peptides therapy: an emerging alternative for treating drug-
resistant bacteria. Yale J Biol Med. 2022;95(4):445–63.
44. Rykowska I, Nowak I, Nowak R. Soft contact lenses as drug delivery systems: a review.
Molecules. 2021;26(18):5577.
45. Rathi VM, Vyas SP, Sangwan VS. Phototherapeutic keratectomy. Indian J Ophthalmol.
2012;60(1):5–14.
46. Espandar L, Carlson AN. Lamellar keratoplasty: a literature review. J Ophthalmol.
2013;2013:894319.
Advances in Diagnosis and Management of Infectious Keratitis 45
47. Dallal MMS, Nikkhahi F, Imeni SM, Molaei S, Hosseini SK, Kalafi Z, Yazdi SS, Mirzaei
HMA. Amniotic membrane transplantation for persistent epithelial defects and ulceration due
to Pseudomonas keratitis in a rabbit model. J Ophthalmic Vis Res. 2021;16(4):552–7.
48. Jung JH, Ji YW, Hwang HS, Oh JW, Kim HC, Lee HK, Kim KP. Proteomic analysis of human
lacrimal and tear fluid in dry eye disease. Sci Rep. 2017;7(1):13363.
49. Krishnan R, Stuart PM. Developments in vaccination for herpes simplex virus. Front Microbiol.
2021;12:798927.
50. Gray GC, Erdman DD. Adenovirus Vaccines. Plotkin’s Vaccines. 2018:121–133.e8.
https://doi.org/10.1016/B978-0-323-35761-6.00010-9.
51. Ong ZZ, Sadek Y, Liu X, Qureshi R, Liu SH, Li T, Sounderajah V, Ashrafian H, Ting DSW,
Said DG, Mehta JS, Burton MJ, Dua HS, Ting DSJ. Diagnostic performance of deep learn-
ing in infectious keratitis: a systematic review and meta-analysis protocol. BMJ Open.
2023;13(5):e065537.
52. Verma S, Singh A, Varshney A, Chandru RA, Acharya M, Rajput J, Sangwan VS, Tiwari AK,
Bhowmick T, Tiwari A. Infectious keratitis: an update on role of epigenetics. Front Immunol.
2021;12:765890.
53. Ting DSJ, Chodosh J, Mehta JS. Achieving diagnostic excellence for infectious keratitis: a
future roadmap. Front Microbiol. 2022;13:1020198.
54. Gurnani B, Kaur K, Venugopal A, Srinivasan B, Bagga B, Iyer G, Christy J, Prajna L, Vanathi
M, Garg P, Narayana S, Agarwal S, Sahu S. Pythium insidiosum keratitis—a review. Indian J
Ophthalmol. 2022;70(4):1107–20.
Advances in Glaucoma
1 Introduction
Glaucoma management is at the cusp of monumental change. While there are excit-
ing possibilities emerging for its diagnosis, and therapeutic interventions, clinical
scientists across the globe are looking increasingly at the quality of life of glaucoma
patients. Research is being focused at not only genetics and gene therapy for glau-
coma, but also at technologies that will enable better patient compliance with con-
ventional anti glaucoma medications (AGMs); as well safer, and more effective
surgical options [1, 2].
The clinical experience with respect to these technologies, expectedly, is lim-
ited, and so, their acceptability for both patients and glaucoma practitioners alike
is a barrier for their early adoption. It is therefore imperative that a concerted
effort be made towards education of patients and caregivers, as well as the health
care team comprising of optometrists, general ophthalmologists, and glaucoma
surgeons. It is only then that the patient will be able to benefit from the latest
advances in glaucoma management. This chapter will cover the recent advances
in glaucoma diagnostics, newer drugs as well as visual rehabilitation for glau-
coma management.
P. Ichhpujani (*)
Department of Ophthalmology, Government Medical College and Hospital,
Chandigarh, India
S. Bhartiya
Department of Ophthalmology, Marengo Asis Hospitals, Gurugram, Haryana, India
2 Glaucoma Diagnostics
-21000
Zoom A Zoom B
-21500
-22000
IOP Profile [a.u.]
-22500
-23000
-23500
-24000
-24500
10:00 11:00 12:00 13:00 14:00 15:00 16:00 17:00 18:00 19:00 20:00 21:00 22:00 23:00 00:00 01:00 02:00 03:00 04:00 05:00 06:00 07:00 08:00 09:00 10:00 11:33
Time
ing. It was CE-certified in 2017 for commercial use in the European Union
(EU). Eyemate has eight pressure and temperature sensors, designed for implan-
tation in the ciliary sulcus at the time of cataract surgery [7].
Each pressure sensor has two parallel plates, the base plate is rigid while
the plate that indents with changes in IOP is flexible. The distance between
these two plates varies with fluctuations in IOP, generating an analog signal,
which is then converted to a digital signal, that is transmitted to a small hand-
held mesograph by radiofrequency. For the reader to activate the electromag-
netic coupling sequence and the units to correspond with each other, the reader
and the intraocular transponder unit must be within 5 cm of each other. The
device can obtain up to 10 IOP measurements per second. The Eyemate (with
a GSM module) data can be shared with the physician using an internet
database.
EYEMATE-SC when implanted in patients with primary open-angle glau-
coma (POAG) undergoing simultaneous nonpenetrating glaucoma surgery
(NPGS) was found to be safe and well tolerated through 12 months [8].
Literature reassures that the Smart IOP sensors hold promise for redefining
glaucoma treatment strategies.
Digital photography:
Remote or rural areas have limited access to good ophthalmic care with the
state-of-the-art diagnostic equipment, especially for detecting preperimetric or
early glaucomatous damage [9]. Smartphone-based teleglaucoma screening can
help in early detection and treatment.
Affordable adapters that convert a smartphone camera into a miniature ante-
rior segment and fundus camera have flooded the markets globally.
Some of the adapters used in community studies include the Peek (Peek
Vision; https://www.peekvision.org) from England, the D-Eye system (Si 14;
https://www.d-eyecare.com) from Italy, and the IExaminer (Welch Allyn),
EyeGo (Stanford University and DigiSight Technologies), and Ocular CellScope
(University of California, Berkeley) from the United States.
These smart devices are being used to capture “direct” images or video of the
posterior pole (in an undilated state, without the condensing +20 D lens) not
only by the ophthalmologists and optometrists but also by the neurologists and
paediatricians. These images can be shared for expert assessment (HIPPA com-
pliant) and saved to a patient file for future reference [10, 11].
50 P. Ichhpujani and S. Bhartiya
the screen to activate the programme to show the next area with grating. Correct
and incorrect responses are recorded until the software determines the contrast
threshold in each area. The test takes 5–10 min per eye [19].
The contrast threshold is determined using a staircase strategy with reversals.
Initial correct responses advance four levels until the patient gives an incorrect
response. After the incorrect response, a contrast level two levels easier than the
previous one is presented. The algorithm advances or regresses one level at a
time until two incorrect responses are made at a specific level, which establishes
the contrast threshold. If a patient stops trying to guess the correct area and
clicks the same location repeatedly, the test terminates. Patient is instructed to
attempt to choose the location where the gratings appear. The range of contrast
tested is from 100% to 0.45% (log contrast sensitivity 0.00–2.35) and decreases
by approximately 0.15 log units between levels. The central area (CC) and four
peripheral areas (SN, ST, IN, IT) each receive separate scores. A total SPARCS
score is summated from each of the five areas (20 per area), resulting in a perfect
score of 100.
Richman et al. studied the performance of SPARCS for glaucoma patients,
and found that a total SPARCS score of <70 had a sensitivity of 79.7% and
specificity of 92.8% in the identification of glaucoma in Caucasian population
[19]. Thakur et al. have also found SPARCS to be a sensitive and reliable indica-
tor of glaucomatous damage [20]. SPARCS has also been used to test CS in
patients with refractive errors, cataract, age-related macular degeneration and
idiopathic intracranial hypertension [21–23].
D. Electrophysiology:
nGoggle™: The nGoggle™ is a “virtual reality” (VR) goggle with embed-
ded EEG sensors. It is a brain–computer interface platform (BCIP) for detecting
and monitoring neurodegenerative diseases of eye and brain. It allows detection
of steady-state visual evoked potentials (SSVEP) corresponding to pattern
reversal visual stimuli [24].
A trial (NCT03760055) is ongoing to assess the ability of the nGoggle to
detect VF loss in patients with glaucoma, as well as to differentiate different
severity stages of glaucoma, its repeatability and reliability for patient self-
testing and patient satisfaction compared to SAP, optical coherence tomography
(OCT) metrics and Diopsys NOVA (Diopsys, Inc.).
Pattern Electroretinogram (PERG):
PERG measures the activity of retinal ganglion cells (RGCs) and possibly
detects the early functional abnormalities in glaucoma. Available commercial
devices include Diopsys’ Nova and Argos Vision Testing Systems and Konan
Medical’s EvokeDx [25].
Steady-state PERG amplitude has been shown to progressively decrease over
time in a subset of glaucoma suspects, with relatively steeper slope in treated
compared to untreated glaucoma suspects [26]. EvokeDx device has been found
to be useful to help assess central vision and evaluate damage to magnocellular
(MC) pathway, however it is noncontributory in mapping specific structural
changes or peripheral defects [27].
52 P. Ichhpujani and S. Bhartiya
3 Newer Drugs
A. Rho Kinase Inhibitors: The two Rho kinase isoforms (ROCK 1 and ROCK 2)
are the effectors for the Rho A, B, and C. These G proteins get activated after
binding to guanosine triphosphate (GTP). Rho kinase inhibitors have a dual
mechanism of action, increase in aqueous outflow, along with decrease in aque-
ous outflow resistance by increasing the pores formed by the endothelial cells in
the Schlemm’s canal (SC). They also increase the outflow by relaxing smooth
muscle fibres of the trabecular meshwork (TM). In addition, Netarsudil has nor-
epinephrine transporter (NET) inhibitor activity further lowers aqueous humour
production as well as the episcleral venous pressure. Rho kinase may also be
neuroprotective, possibly via increased optic nerve head perfusion, and may
decrease postsurgical scarring. The latter action is because ROCK inhibitors
inhibit fibroblasts activation via TGF-beta [2, 28].
The two commonly used ROCK inhibitors are Ripasudil and Netarsudil.
Ripasudil hydrochloride hydrate (Glanatec®) 0.4% which requires twice a day
dosing and Netarsudil (Rhopressa®) 0.02% which is used once at bedtime [2].
They are both known to effective in lowering IOP in POAG and OHTN alike, with
the most common side effects being conjunctival hyperaemia (75%), blepharitis
(20%), and cornea verticillata (15%), with the latter being unique to Netarsudil.
The fixed drug combination (FDC) of netarsudil 0.02% and latanoprost
0.005% (ROCKLATAN®) once daily has been found to resulted in greater
mean IOP reduction as compared to monotherapy with either drug [29].
The newest ROCK inhibitor in pipeline, Fasudil, has shown good IOP lowering
even in end-stage glaucoma [30]. Use of depot formulation of Fasudil loaded
polylactide-co-glycolide (PLGA) microspheres have been proposed in the form of
intravitreal injection for glaucoma patients with coexistent retinal disease [31].
Rho kinase inhibitors hold great promise as an adjunctive therapy, since their
mechanism of action is different from the existing AGMs. Although adverse
events, especially conjunctival hyperaemia and subconjunctival haemorrhages,
may impact compliance in the long term, this class of AGM is a significant addi-
tion to the glaucoma practitioners armamentarium.
B. Latanoprostene Bunod: Latanoprostene bunod 0.024% (Vyzulta®) is a pro-
drug, which has the unique property of donating nitric oxide (NO). The drug
metabolizes into latanoprost acid, and butanediol mononitrate; and the latter is
further metabolized to 1,4-butanediol and NO.
Latanoprost acid increases uveoscleral outflow via the Prostaglandin F receptor.
Like all other prostaglandin agonists, it modulates the remodelling of the extracel-
lular matrix of the ciliary muscle via matrix metalloproteinases. Nitric oxide, the
second active metabolite of the prodrug, causes vasodilation and smooth muscle
cell relaxation in the TM and SC by decreasing cell contractility and volume, thus
increasing outflow facility [32]. LBN has shown to increase both, optic nerve head
capillary blood volume and oxygen saturation more than latanoprost [33].
The once daily dosing of LBN has been found to be more effective than both
latanoprost and timolol. The most common side effects include conjunctival
Advances in Glaucoma 53
hyperemia (17%), eyelash growth (16%), irritation and pain (10%). LBN has
been found to be as effective as netarsudil as an adjuvant therapy [34].
C. Novel Methods of Drug Delivery
Nanotechnology: Nanotechnology-based treatments may improve patient
quality of life, and consequently adherence, because of targeted drug delivery,
better bioavailability, sustained release of the AGM or anti-metabolite (in case
of glaucoma surgeries), and better therapeutic efficiency [35].
Additionally, nanomedicine formulations may have decreased side effects
due to less frequent dosing [36]. These include liposomes, nanogels, nanopar-
ticles, micelles, nanosuspensions, dendritic polymers, microneedles, etc., all of
which have a particle size ranging from 1 to 100 nm, have shown to have signifi-
cantly better therapeutic safety and efficacy than conventional ophthalmic for-
mulations in animal studies. Even though this is yet to be a part of clinical
practice, nanomedicine formulations of several AGMs that have been tried
include supraciliary injection of brimonidine-laden microspheres, subconjuncti-
val injection of dorzolamide nanoparticles and intravitreal injection of brimoni-
dine, travoprost, and bimatoprost-laden nanosponges [2, 37, 38].
Implants:
Antiglaucoma drugs dispensed as injectable systems and undertrial punctal
plug delivery systems are enlisted in Table 1.
(a) Bimatoprost implant: The bimatoprost implant (Durysta™) is a sustained
release, biodegradable implant uses the NOVADUR drug delivery system.
A 28-gauge, prefilled applicator releases this implant intracamerally. The
biodegradable polymers of the implant disintegrate within the anterior
chamber into carbon dioxide and water. The main adverse event noted was
conjunctival hyperemia in almost one in three patients, and a more than
Table 1 Antiglaucoma drugs dispensed as Injectable systems and Undertrial Punctal plug deliv-
ery systems
Drug delivery Placement of Current stage
system Drug Material location of development
ENV515/PGA or Travoprost – Anterior Phase 2 clinical
Travoprost XR chamber angle trial
Bimatoprost SR Bimatoprost – Subconjunctival Phase 3 clinical
space trial
Graybug Various – Ocular surface Preclinical
OTX-TP Travoprost Polyethylene Puncta Phase 2 clinical
glycol-based trial
hydrogel with
loaded PLA
Evolute Latanoprost – Puncta Phase 2 clinical
trial
Latanoprost Latanoprost – Puncta Phase 2 clinical
punctal plug trial
delivery system
(L-PPDS)
54 P. Ichhpujani and S. Bhartiya
20% reduction in endothelial cell density in 10% and 21% of the 10 and
15 mcg groups. About 4% and 10% of eyes in each of the two groups,
respectively, needed implant removal because of the corneal oedema, in
order to prevent further endothelial loss. Better formulations are currently
under review [39, 40].
(b) BIM ring: The bimatoprost ocular ring (Bimatoprost HELIOS™ insert,
Abbvie) is a silicone—polypropylene ring impregnated with bimatoprost
(24–29 mm in diameter) which is inserted in the fornices. It provides sus-
tained IOP control over 6 months, eluting 6–35 microns of the drug per day.
The device does not have FDA approved yet [41].
(c) Travoprost punctum plugs: Recently, Polyethylene glycol resorbable
hydrogel punctal plugs impregnated with travoprost, OTX-TP (Ocular
Therapeutix, Inc.) have been developed. The retention rates of these plugs
were 90% at 2 months, but less than 50% at 3 months. The other major
adverse event reported was foreign body sensation in almost 40% patients,
with the tolerability increasing over time [42].
Precise targeted delivery will minimize drug wastage by preventing drug loss
due to systemic absorption and first-pass metabolism. High prevalence of ocular
surface disease in patients instilling AGMs with added preservatives may be
overcome with the newer devices [43].
4 Visual Rehabilitation
(a) Virtual reality based low vision aids: For patients with advanced glaucoma
using low vision aids, augmented reality magnification can be done with devices
such as Microsoft HoloVision Hololens and a camera worn on the finger [44].
Electronic head-mounted low vision aid (e-LVA) SightPlus (GiveVision,
UK, givevision.net) improves visual function in people with low vision [45].
(b) Smartphone applications for people with low vision: Several smartphone
apps are available that can make day-to-day tasks easier for people with visual
impairment to improve their quality of life [46].
• NavCog: This iPhone based app is helpful for indoor navigation, inside
complex building spaces such as an airport or hospital. (https://www.cs.cmu.
edu/~NavCog/navcog.html)
• Seeing AI: This app gives a narration of the surrounding world around the
patient through their phone’s camera. It can help to identify and describes
people, objects (e.g., currency) and documents (reads them aloud).
Additionally, it can describe features such as colour, light, and handwriting.
• In future, patients can get customized digital devices and apps to address
their low vision.
• We are hopeful that the emerging technologies will not only help mitigate
the disease but also improve the quality of life of those with advanced or end
stage glaucoma.
Advances in Glaucoma 55
References1
1. Twa MD. Advances in glaucoma management. Optom Vis Sci. 2020;97(2):53.
2. Mohan N, Chakrabarti A, Nazm N, Mehta R, Edward DP. Newer advances in medical manage-
ment of glaucoma. Indian J Ophthalmol. 2022;70(6):1920–30.
3. Cvenkel B, Velkovska MA, Jordanova VD. Self-measurement with Icare HOME tonometer,
patients’ feasibility and acceptability. Eur J Ophthalmol. 2020;30(2):258–63.
4. Takagi D, Sawada A, Yamamoto T. Evaluation of a new rebound self-tonometer, Icare HOME. J
Glaucoma. 2017;l26(7):613–8.
5. Barbour-Hastie CC, Tatham AJ. Teaching home tonometry using a remote video link. Eye
(Lond). 2023;37(3):501–5.
6. Yang C, Huang X, Li X, Yang C, Zhang T, Wu Q, Liu D, Lin H, Chen W, Hu N, Xie X. Wearable
and implantable intraocular pressure biosensors: recent progress and future prospects. Adv Sci
(Weinheim). 2021;8(6):2002971.
7. Enders P, Cursiefen C. Device profile of the EYEMATE-IO™ system for intraocular pressure
monitoring: overview of its safety and efficacy. Expert Rev Med Devices. 2020;17(6):491–7.
8. Szurman P, Gillmann K, Seuthe AM, Dick HB, Hoffmann EM, Mermoud A, Mackert MJ,
Weinreb RN, Rao HL, Mansouri K, EYEMATE-SC Study Group. EYEMATE-SC trial:
twelve-month safety, performance, and accuracy of a suprachoroidal sensor for telemetric
measurement of intraocular pressure. Ophthalmology. 2023;130(3):304–12.
9. Morjaria P, Bergson S, Bastawrous A, Watts E, Pant S, Gudwin E, Zinn M, Chan VF. Delivering
refractive care to populations with near and distance vision impairment: 2 novel social enter-
prise models. Asia Pac J Ophthalmol. 2022;11(1):59–65.
10. Andersen T, Jeremiah M, Thamane K, Littman-Quinn R, Dikai Z, Kovarik C, Ndlovu
K. Implementing a school vision screening program in Botswana using smartphone technol-
ogy. Telemed J E Health. 2020;26(2):255–8.
11. Shukla K, Sapkota YD, Das AV, Morjaria P. Communication technology for eye care.
Community Eye Health. 2022;35(114):1–2.
12. Russo A, Mapham W, Turano R, Costagliola C, Morescalchi F, Scaroni N, Semeraro
F. Comparison of smartphone ophthalmoscopy with slit-lamp biomicroscopy for grading ver-
tical cup-to-disc ratio. J Glaucoma. 2016;25(9):e777–81.
13. Prager AJ, Kang JM, Tanna AP. Advances in perimetry for glaucoma. Curr Opin Ophthalmol.
2021;32(2):92–7.
14. Harris PA, Johnson CA, Chen Y, Fann H, Gafford G, Kim YJ, Mezgebu ED. Evaluation of the
Melbourne rapid fields test procedure. Optom Vis Sci. 2022;99(4):372–82.
15. Kumar H, Thulasidas M. Comparison of perimetric outcomes from Melbourne rapid fields
tablet perimeter software and Humphrey field analyzer in glaucoma patients. J Ophthalmol.
2020;2020:8384509.
16. Prea SM, Kong YXG, Mehta A, He M, Crowston JG, Gupta V, Martin KR, Vingrys AJ. Six-
month longitudinal comparison of a portable tablet perimeter with the Humphrey field ana-
lyzer. Am J Ophthalmol. 2018;190:9–16.
17. Kang J, De Arrigunaga S, Freeman SE, Zhao Y, Lin M, Liebman DL, Roldan AM, Kim JA,
Chang DS, Friedman DS, Elze T. Comparison of perimetric outcomes from a tablet perim-
eter, smart visual function analyzer, and Humphrey field analyzer. Ophthalmol Glaucoma.
2023;6:S2589–4196(23)00059–5.
18. Ichhpujani P, Dhillon H. Spotlight on iPad visual field tests efficacy. Clin Ophthalmol.
2022;16:2179–85.
1
Note: Some text is taken from the author’s previously published chapter in a book which is
part of the same series. Ichhpujani, P. (2020). Recent advances in glaucoma diagnostics. In:
Ichhpujani, P. (eds) Current advances in ophthalmic technology. Current Practices in
Ophthalmology. Springer, Singapore. https://doi.org/10.1007/978-981-13-9795-0_4
56 P. Ichhpujani and S. Bhartiya
19. Richman J, Zangalli C, Lu L, Wizov SS, Spaeth E, Spaeth GL. The Spaeth/Richman contrast
sensitivity test (SPARCS): design, reproducibility and ability to identify patients with glau-
coma. Br J Ophthalmol. 2015;99(1):16–20.
20. Thakur S, Ichhpujani P, Kumar S, et al. Assessment of contrast sensitivity by Spaeth Richman
contrast sensitivity test and Pelli Robson Chart Test in patients with varying severity of glau-
coma. Eye (Lond). 2018;32:1392–400.
21. Faria BM, Duman F, Zheng CX, Waisbourd M, Gupta L, Ali M, Zangalli C, Lu L, Wizov
SS, Spaeth E, Richman J, Spaeth GL. Evaluating contrast sensitivity in age-related macular
degeneration using a novel computer-based test, the Spaeth/Richman contrast sensitivity test.
Retina. 2015;35(7):1465–73.
22. Gupta L, Cvintal V, Delvadia R, Sun Y, Erdem E, Zangalli C, Lu L, Wizov SS, Richman J,
Spaeth E, Spaeth GL. SPARCS and Pelli-Robson contrast sensitivity testing in normal controls
and patients with cataract. Eye (Lond). 2017;31(5):753–61.
23. Ichhpujani P, Thakur S, Spaeth GL. Contrast sensitivity and glaucoma. J Glaucoma.
2020;29(1):71–5.
24. Nakanishi M, Wang YT, Jung TP, Zao JK, Chien YY, Diniz-Filho A, Daga FB, Lin YP, Wang
Y, Medeiros FA. Detecting glaucoma with a portable brain-computer interface for objective
assessment of visual function loss. JAMA Ophthalmol. 2017;135(6):550–7.
25. Al-Nosairy KO, Hoffmann MB, Bach M. Non-invasive electrophysiology in glaucoma, struc-
ture and function-a review. Eye (Lond). 2021;35(9):2374–85.
26. Gordon PS, Kostic M, Monsalve PF, Triolo G, Golubev L, Luna G, Ventura LM, Feuer WJ,
Porciatti V. Long-term PERG monitoring of untreated and treated glaucoma suspects. Doc
Ophthalmol. 2020;141(2):149–56.
27. https://www.aao.org/eyenet/article/does-perg-have-a-role-in-the-glaucoma-clinic. Accessed
11 Sept 2023.
28. Freiberg JC, von Spreckelsen A, Kolko M, Azuara-Blanco A, Virgili G. Rho kinase inhibi-
tor for primary open-angle glaucoma and ocular hypertension. Cochrane Database Syst Rev.
2022;6(6):CD013817.
29. Asrani S, Bacharach J, Holland E, McKee H, Sheng H, Lewis RA, Kopczynski CC, Heah
T. Fixed-dose combination of netarsudil and latanoprost in ocular hypertension and open-
angle glaucoma: pooled efficacy/safety analysis of phase 3 MERCURY-1 and -2. Adv Ther.
2020;37(4):1620–31.
30. Pakravan M, Beni AN, Ghahari E, Varshochian R, Yazdani S, Esfandiari H, Ahmadieh H. The
ocular hypotensive efficacy of topical fasudil, a rho-associated protein kinase inhibitor, in
patients with end-stage glaucoma. Am J Ther. 2017;24(6):e676–80.
31. Mietzner R, Kade C, Froemel F, Pauly D, Stamer WD, Ohlmann A, Wegener J, Fuchshofer R,
Breunig M. Fasudil loaded PLGA microspheres as potential intravitreal depot formulation for
glaucoma therapy. Pharmaceutics. 2020;12(8):706.
32. Drugs and Lactation Database (LactMed®) [Internet]. Bethesda (MD): National Institute of
Child Health and Human Development; 2006. Latanoprostene Bunod. 2022. PMID: 30000019.
33. Samaha D, Diaconu V, Bouchard JF, Desalliers C, Dupont A. Effect of latanoprostene bunod
on optic nerve head blood flow. Optom Vis Sci. 2022;99(2):172–6.
34. Lo TC, Chen YY, Hung MC, Chou P. Latanoprostene bunod 0.024% in the treatment of open-
angle glaucoma and ocular hypertension: a meta-analysis. J Clin Med. 2022;11(15):4325.
35. Occhiutto ML, Maranhão RC, Costa VP, Konstas AG. Nanotechnology for medical and surgi-
cal glaucoma therapy-a review. Adv Ther. 2020;37(1):155–99.
36. El Hoffy NM, Abdel Azim EA, Hathout RM, Fouly MA, Elkheshen SA. Glaucoma: manage-
ment and future perspectives for nanotechnology-based treatment modalities. Eur J Pharm Sci.
2021;158:105648.
37. Cardigos J, Ferreira Q, Crisóstomo S, Moura-Coelho N, Cunha JP, Pinto LA, Ferreira
JT. Nanotechnology-ocular devices for glaucoma treatment: a literature review. Curr Eye Res.
2019;44(2):111–7.
38. Wu Y, Tao Q, Xie J, Lu L, Xie X, Zhang Y, Jin Y. Advances in nanogels for topical drug deliv-
ery in ocular diseases. Gels. 2023;9(4):292.
Advances in Glaucoma 57
39. 18 month prospective efficacy and safety study of bimatoprost intracameral implant
(DURYSTA). ClinicalTrials.gov. Identifier: NCT04647214. https://clinicaltrials.gov/ct2/
show/NCT04647214. Accessed 1 May 2023
40. Craven ER, Walters T, Christie WC, Day DG, Lewis RA, Goodkin ML, et al. 24 month phase
I/ II clinical trial of bimatoprost sustained release implant (Bimatoprost SR) in glaucoma
patients. Drugs. 2020;80:167–79.
41. Brandt JD, DuBiner HB, Benza R, Sall KN, Walker GA, Semba CP, Collaborators. Long-
term safety and efficacy of a sustained-release bimatoprost ocular ring. Ophthalmology.
2017;124(10):1565–6.
42. Perera SA, Ting DS, Nongpiur ME, Chew PT, Aquino MC, Sng CC, Ho SW, Aung T. Feasibility
study of sustained-release travoprost punctum plug for intraocular pressure reduction in an
Asian population. Clin Ophthalmol. 2016;10:757–64.
43. Singh RB, Ichhpujani P, Thakur S, Jindal S. Promising therapeutic drug delivery systems for
glaucoma: a comprehensive review. Ther Adv Ophthalmol. 2020;12:2515841420905740.
44. Kinateder M, Gualtieri J, Dunn MJ, Jarosz W, Yang XD, Cooper EA. Using an augmented
reality device as a distance-based vision aid-promise and limitations. Optom Vis Sci.
2018;95(9):727–37.
45. Crossland MD, Starke SD, Imielski P, Wolffsohn JS, Webster AR. Benefit of an electronic
head-mounted low vision aid. Ophthalmic Physiol Opt. 2019;39(6):422–31.
46. https://www.aao.org/eye-health/tips-prevention/low-vision-impairment-apps-tech-assistive-
devices. Accessed 11 Sept 2023.
Advances in the Management
of Oculomotor Dysfunction in Adults
and Children with Concussion
Mild traumatic brain injury (TBI) arising from a bump, blow, shake or jolt to the
head or body is often referred to as a concussion [1]. Approximately, 70–80% of all
TBIs are classified as mild in the absence of lesions from brain imaging and/or loss
of consciousness. This may arise from road traffic accidents, falls, sports injuries,
assaults, recreational activities, or children’s playground accidents. For example,
sudden rapid changes in movement during whiplash can cause the brain to bounce,
stretch or twist within the skull. This neurological event may result in focal and/or
diffuse damage to neuronal dendrites, axons, and astrocytes, disrupting cellular
membranes and releasing neurotransmitters. This invariably affects the intracellular
and extracellular ionic equilibrium which consequently cause neuroinflammation
and cell loss [2].
Concussion patients may suffer from a wide range of issues, given the pervasive-
ness of the injury. This includes sensory, motor, perceptual, cognitive, attentional,
behavioral, somatic, academic, linguistic, and social well-being issues. Oculomotor,
vestibular, and visual processing disorders are frequently experienced. Thus, it is
necessary to investigate each of these functions carefully so that the appropriate
treatment, therapy, and rehabilitation can be rendered to the patient.
The terms ‘oculomotor’ and ‘ocular motor’ tend to be used interchangeably in
the literature despite their contextual differences in terms of nerve innervations and
eye movements respectively. Although the latter is more accurate, it is more com-
mon to use the term ‘oculomotor’ to describe eye movement function or disorders.
T. P. Yap (*)
IGARD Vision Therapy Center, Singapore, Singapore
e-mail: [email protected]
C. Stern
Pappas Rehabilitation Hospital for Children, Canton, MA, USA
Relative Rest is important for brain injury recovery. Rest during the recovery phase
allows inflammation to reduce and the brain to recover. If the condition is not wors-
ening, concussion patients are often discharged from hospitals and encouraged to
rest at home with limitations of activities involving mental concentration (e.g.,
video games and reading) and physical exertion (e.g. sports) to allow the symptoms
to subside. After the first few days, one should gradually increase physical activity
to a level that does not provoke symptoms. The duration for recovery depends on
age [4], gender [5], severity [6], and presence of comorbid conditions, such as
migraines, mood, learning, and attention-deficit disorders [1].
While full recovery is generally expected within weeks or up to 3 months, activi-
ties within the next 2–3 days should be light and kept to a level that does not pro-
voke symptoms especially when returning-to-work (or school). To avoid the risk of
a repeat concussion, return-to-sports (or play) should be conducted in a safe manner
and guided using a graduated return-to-sport strategy [7] along with baseline
Advances in Management of the Oculomotor Dysfunction… 61
Oculomotor dysfunction affects more than 60% of concussed children and adoles-
cents [12] and more than 90% of concussed adults [13]. Deficits in saccades, smooth
pursuit, vergence, and accommodation are frequently observed [14]. In the absence
of symptoms or in situations where patients are unable to recall the concussion,
oculomotor function may be used as an objective biomarker in early detection [15]
and for monitoring recovery [14].
The propensity for the visual system to be affected may be related to the large
extent of the human brain involved in visual processing. As discussed previously,
‘ocular motor function’ describes the overall quality of eye movement but ‘oculo-
motor dysfunction’ is the term more commonly used in the literature to describe eye
movement disorders. Thus, oculomotor function can be defined as the ability of the
visual system to accurately fixate and track a moving target or when the person is
moving; and to keep the target clearly in focus at all times. This entire process relies
62 T. P. Yap and C. Stern
Fig. 2 Schematic representation of the neurological elements that mediates oculomotor function
on the visual system’s capacity to align the eyes accurately in relation to the vestibu-
lar and proprioceptive systems.
Screening tools, such as the Vestibular Ocular Motor Screening (VOMS), King-
Devick test or computerized eye tracker protocols can be used for initial diagnosis
of oculomotor dysfunction but should by followed by a complete neuro optometric
evaluation. The evaluation provides more detailed information about the oculomo-
tor dysfunction, including the fixation, accommodation, vergence, saccadic eye
movements, smooth pursuit eye movements and vestibular-oculomotor func-
tion (Fig. 2). Each of these visual functions may be considered as a neurological
element that mediates oculomotor function, where each of them tend to interact
with another. For example, vergence and the accommodative systems are tightly
coupled in the process in maintaining accurate fixation, while pursuit eye move-
ments entails steady and accurate fixation in relation to the vestibular system in
order to hold the gaze. Each of these neurological elements needs to be assessed
carefully during the neuro optometric evaluation, so any deficits can be diagnosed
and properly managed in a treatment plan.
arc), and microsaccades (fast jerky involuntary eye movements with amplitudes
ranging 10–12 arc minutes) are actively occurring to fine-tune and ensure an accu-
rate fixation.
As interactions between fixational eye movements and directed visual attention
are mediated in the median cingulate gyrus, frontal eye fields (FEFs) and supplemen-
tary eye fields (SEFs) [16], fixational disorders post-concussion can affect a person’s
visual attention in a similar way to the poor fixation linked to attention deficit hyper-
activity disorder (ADHD) traits [17]. Previous experiments found that microsaccades
can result in a gaze shift from a target to another location of interest if the fixation is
poorly engaged [18], thus affecting visual attention, visual perception and cognition.
In addressing fixational disorders, two major neurological elements in the pro-
cess of fixation must be considered—vergence and accommodation. These systems
actively align and adjust the focus of each eye synchronously to maintain the fixa-
tion. Similarly, any microsaccades during each fixation are typically conjugate eye
movements with equal velocity and amplitudes, so both accommodative and ver-
gence systems are tightly coupled in the process of fixation. Therefore, both systems
need to be carefully examined in concussion patients.
the two retinae (or retinal disparity) must be readily reconciled by an adequate ver-
gence response in order to achieve sensory fusion. While viewing objects that are
nearer or further away, convergence or divergence of the two eyes works synchro-
nously with accommodation at all distances under the influence of the propriocep-
tive system which gives the internal awareness of space. Failure to maintain vergence
can result in unstable binocular vision, diplopia, asthenopia and visual discomfort.
Approximately half of those affected by a receded NPC post-concussion tend to
recover without intervention in 4.5 weeks [25], but some patients experience persis-
tent abnormalities lasting several months or longer [26]. In children with persistent
post-concussive symptoms lasting longer than 28 days, a receded NPC was observed
in 89% of them [20].
In sports-related concussion, receded NPC (>6 cm) appears to be a reliable bio-
marker for prolonged recovery (e.g. 50 days) [27]. Symptoms of convergence insuf-
ficiency tend to be similar to that reported in patients with accommodative disorder
[20]. Since vergence disorders can influence accommodation, both conditions are
often present in patients demonstrating a receded NPC. Hence, patients should ide-
ally receive a neuro optometric evaluation 4–5 weeks post-concussion when
other conditions will have abated.
Typically, the NPC breakpoints are measured as the distance from the point of
diplopia to the nasion (bridge of the nose) and the recovery point is when single
vision is re-established as the target is backed away. However, NPC measurements
can be confounded by the patient’s poor misinterpretation of blur versus diplopia.
Receded NPC tends to be more apparent using a line target on the RAF rule (e.g.
Grafton Optical, Berkhamsted, UK) compared to a pencil tip or fingertip [28], or
when the target speed is increased [29]. NPC measurements alone may not be suf-
ficient when evaluating vergence disorders but should be used as a screening test
prior to a referral for neuro optometric evaluation.
Other diagnostic factors of convergence insufficiency include a larger exophoria
at near than far and inadequate positive fusional vergence at near. Exophoria refers
to the tendency for the eyes to deviate outwards and is a common observation after
a concussion; whereas fusional vergence measurements determine if the patient has
the capacity to sustain binocular single vision as the examiner increases negative
and positive vergence demands while the patient maintains a constant level of
accommodation. This is collectively also known as binocularity, which has to be
assessed carefully by a clinician with the relevant training in optometry. While these
heterophoria measurements will determine the relative deviation of the eyes when
the two eyes are dissociated, such as during the cover test, an experienced clinician
must also consider the interplay and influence of accommodation, refractive correc-
tion, spectacle lens design, head posture, vestibular input, and the patient’s vestibu-
lar status.
Stereopsis can be measured using commercially available tests, such as the
Frisby stereotest (Stereotest Ltd., Oxfordshire, UK). Typically, positive and nega-
tive fusional vergence (PFV and NFV) ranges are measured at a fixed distance of
40 cm using a prism bar (base-out and base-in, respectively) by increasing its mag-
nitude until the blur and break (diplopia) points are found and reducing the prism
66 T. P. Yap and C. Stern
until the recovery (single vision) point. In a full binocular vision work-up, vergence
facility can be measured by alternating a 3Δ base-in/12Δ base-out prism while fix-
ating on an accommodative target at 40 cm in order to determine the number of
cycles per minute the patient can achieve while gaining single and clear vision dur-
ing each alternation of the prisms.
Saccadic disorders are estimated to occur in 20–30% of concussion patients [12, 13]
and its associated impairments tend to be more apparent under conditions of high
cognitive load, such as reading or when watching fast moving objects. For example,
patients may lose their place when reading, skip or re-read lines of print, face diffi-
culty with eye contact, and academic learning may be affected [10]. Other symp-
toms of saccadic disorders may include asthenopia, headaches, and dizziness.
Saccadic eye movements (or saccades) involves rapid changes in fixation while
shifting the gaze. In general, the process of reading tends to involve small ampli-
tudes of saccades whereas large amplitudes would typically be used during visual
scanning tasks. Following a concussion, these tasks tend to become more difficult
due to poorer accuracy (hypo- or hypermetric), slower velocities, increased laten-
cies, longer duration, and smaller peak accelerations in the saccadic eye movements.
Vergence disorders often occur together with saccadic dysfunction because these
two systems tend to interact during eye movements and may affect attention after
concussion [30].
The normal function of saccades is to scan the visual environment (e.g., visual
search or free viewing) in order to capture the image of interest as quickly as pos-
sible onto the fovea where photoreceptor density and visual resolution are the high-
est. Saccades can be voluntary or reflexive to visual, auditory, memory, and tactile
stimuli. In response to changes in fixations, the eyes shift rapidly with a velocity of
400–800°/s from one object of fixation to another. This movement is brief (short
duration of 10–100 ms), conjugate (i.e., parallel and same direction), and ballistic
(i.e., unable to change the course of the eye movement without making a separate
saccade).
Clinical examination typically evaluates the accuracy, latency, and velocity of
the saccadic eye movements by asking the patient to alternately fixate between
two targets 30–40° apart at a viewing distance of 40 cm. Latencies of 180–200 ms
are normally observed at the beginning of a saccade, then it tends to be slightly
hypometric (i.e., undershoot the target) by approximately 10% less than the
amplitude of the intended target and is fine-tuned by a microsaccade. Reading-
related eye movement can be examined using standardized rapid number naming
tests, such as the Developmental Eye Movement (DEM) test (Bernell Corp.,
Mishawaka, Indiana, USA), or the King-Devick test (King-Devick Technologies
Inc., Downers Grove, Illinois, USA). The latter is increasingly used in baseline
testing and for sideline testing when subclinical concussions are suspected among
athletes [31].
Advances in Management of the Oculomotor Dysfunction… 67
Smooth pursuit eye movement (SPEM) disorders are estimated to affect 43–60% of
patients with concussion [14]. They tend to induce visual motion sensitivity, dizzi-
ness, and nausea when viewing objects in motion. This is largely because the eyes
tend to lose track when unable to keep up with their speed, thus putting an addi-
tional load in the saccadic, vestibular, and optokinetic systems. This tends to elicit
involuntary conjugate saccades that interrupt fixation knows as saccadic intrusions
or a combination of eye and head movements in order to compensate or reduce that
target error.
The fundamental purpose of SPEM is to assist with gaze holding where the two
eyes produce conjugate movements (in the same direction) so the target remains
accurately fixated on the fovea without head movement. While the neural process of
gaze holding also involves the vestibular and optokinetic systems, SPEM maintains
gaze accurately together with the fixational and vergence systems keeping it to a
speed of 30–40°/s before shifting the gaze or moving the head. However, post-
concussion SPEM can prove to be difficult for some patients as observed by their
frequent fixations and saccades when tracking slow moving targets [32]. This affects
their attention given that the prefrontal cortex is involved in both processes. For
example, visual attention tends to be hindered when tracking multiple moving
objects due to the increased tracking load and distraction [30].
Clinical evaluation of SPEM involves observing for smoothness, amplitude,
velocity, and direction when the patient is following a 10–20°/s moving target, such
as the tip of a pencil, at a distance of approximately 1 m. The patient should be
instructed not to move his or her head, and the target should be moved in the vertical
and horizontal planes. To track a moving object, a very quick “catch-up” saccade
normally precedes the fixation and SPEM. Longer lags, smaller amplitudes,
increased eye position errors, and variabilities in gaze positions may be observed
post-concussion and SPEM may appear jerky due to saccadic intrusions and re-
fixations. Alternatively, an optokinetic drum can be used to examine the patient’s
horizontal SPEM by following the alternating white and black vertical stripe pat-
terns on the rotating drum. As the eyes moves toward the end of each excursion, a
corrective saccade would be observed in the opposite direction—this normal phe-
nomenon is known as optokinetic nystagmus and it relies on intact SPEM and sac-
cadic systems in order to produce this normal rhythmic reflex response.
• Refractive correction
• Therapeutic lenses for symptom relief, or to assist focus flexibility or
binocularity
• Prism lenses for spatial orientation, binocular compensation, balance, or pos-
tural control
• Spectral filters (color or tint) for light sensitivity or reading disability
• Selective occlusion for strabismus or diplopia
• Neuro Optometric Rehabilitation Therapy to remediate oculomotor dysfunction,
inclusive of fixation, pursuits, saccades, accommodation, vergence, and
binocularity.
While not being covered here, patients may also benefit from visual field expand-
ing devices for visual field constriction or loss, optical devices for sight loss, non-
optical aids for low vision or reading disability, and proper management or
monitoring of eye disease.
70 T. P. Yap and C. Stern
The use of lenses, prisms, selective occlusion, and spectral filters or tints is inte-
gral to treating post-concussion syndrome. The goal of the treatment is to alleviate
symptoms, achieve desired visual outcomes, meet the patient’s needs, and improve
the patient’s quality of life.
Neuro Optometric Rehabilitation Therapy may be considered at the same time as
the use of lenses, prisms, selective occlusion, and tints but may be needed alone or
after a trial of these other more passive treatments. Here, the reasons or rationale of
using each of these treatment modalities will be discussed.
Brain injury may change a patient’s refractive status. A patient may need a change
in spectacle prescription or a first prescription when they had no apparent prescrip-
tive visual need prior to their injury. A patient wearing contact lenses may no longer
be able to insert or remove the lenses and may not have a current eyeglasses pre-
scription. They may have only worn spectacles for brief periods of time for many
years and will therefore need a careful refraction that they can tolerate.
The prescription should consider both maximizing sight and patient comfort. If
there is a significant difference in the spherical or cylindrical components between the
two eyes, consider reducing the prescription in one eye to better balance the prescrip-
tion. In clinical practice, if at least one meridian of the astigmatic prescription is equal
between the two eyes, the patient can better tolerate wearing the prescription and there
is little to no compromise of binocular eyesight or binocular vision status [38].
Trial frame testing should be considered with every patient. If the refraction is
done in a phoropter, the desired prescription should be placed into a trial frame and
the patient allowed to walk around with it on for a few minutes. Giving them the
experience of near and far viewing along with body movement can be very helpful
in knowing if the patient is likely to tolerate the new prescription in everyday
activities.
Table 1 List of tinted ophthalmic lenses from the Polytrauma Brain Injury Filter Kit (Chadwick
Optical: www.chadwickoptical.com)
Green/Blue Plum
• Green/Blue 20% • Light Plum (Noir 88)
• Blue/Green 20% • Medium Plum (COI Plum)
Blue • Dark Plum (Noir 81)
• E-15 Blue (Light Blue) Rose
• E-30 Blue (Medium Blue) • Light Rose (FL-41 75%)
• E-50 Blue (Dark Blue) • Medium Rose (FL-41 50%)
• z1 Blue (Noir 26) • Dark Rose (FL-41 27%)
While many believe that neuro optometric rehabilitation is quite new, the concept of
being able to rehabilitate or enhance oculomotor, accommodative and binocular
vision skills has been in the literature since the nineteenth century. A French oph-
thalmologist, Louis Émile Javal, wrote his Manual of Strabismus in 1896 and is
considered the father of orthoptics, a precursor to modern vision therapy and neuro
optometric rehabilitation [40].
Neuro Optometric Rehabilitation Therapy is an individualized treatment pro-
gram for patients with oculomotor deficits that either do not respond to other inter-
ventions or where the other interventions mentioned still leave them with residual
deficits. There is evidence supporting vision therapy as an effective strategy for
rehabilitation. For example, Ciuffreda, et al. found that treatment with eye tracking,
accommodation and convergence exercises expedited recovery and completely alle-
viated symptoms in almost 90% of patients with mild TBI who have persistent
oculomotor defects [41].
Advances in Management of the Oculomotor Dysfunction… 73
This therapy typically involves weekly in-office sessions with supportive home
activities over an 8-month period until symptoms resolved [41]. This includes activi-
ties or techniques that are carefully planned and guided by eye doctors who are accred-
ited or board certified in vision therapy and neuro optometric rehabilitation, so as to
remediate the underlying neural deficits that limit oculomotor function. International
accreditation bodies include the Neuro Optometric Rehabilitation Association
(NORA), College of Optometrists in Vision Development (COVD)/Optometric
Vision Development and Rehabilitation Association (OVDRA), Optometric
Extension Program Foundation (OEPF), Australasian College of Behavioural
Optometrists (ACBO), British Association of Behavioural Optometrists (BABO),
and the Binocular Vision, Perception and Pediatric Optometry (BVPPO) Section of
the American Academy of Optometry (AAO).
Typically, the therapy activities begin at a level where the patient can perform the
activities without exacerbating their symptoms, and the level of the training activi-
ties is gradually increased according to the patient’s response to the treatment. This
helps in reducing the positional error of the eyes, reducing retinal slippage and
improving gaze stability. The target distances and conditions of each activity are
varied according to the treatment plan for the patient, with the goal being to normal-
ize the sensorimotor deficits of binocular vision, improve visual perceptual skills,
and integrate the oculomotor and vestibular systems. Techniques applicable to a
neuro optometric rehabilitation program are available from resources, such as the
Optometric Extension Program Foundation (OEPF) [42].
4 Summary
Oculomotor dysfunction affects more than 60% of concussed children and adoles-
cents and more than 90% of concussed adults. Deficits in saccades, smooth pursuit,
vergence, and accommodation are frequently observed. With proper refractive cor-
rection taking into account visual–vestibular interaction and the use of therapeutic
lenses, prism lenses, spectral filters, selective occlusion, and/or a program of Neuro
Optometric Rehabilitation Therapy, these patients will have their symptoms signifi-
cantly or fully resolved and be able to return to their previous daily activities. Today,
the eye and vision care community should recognize these post-concussion or mild
brain injury visual issues and treat or refer these patients for what is considered the
best medical care.
References
1. Lumba-Brown A, Yeates KO, Sarmiento K, et al. Diagnosis and management of mild traumatic
brain injury in children: a systematic review. JAMA Pediatr. 2018;172:e182847.
2. Romeu-Mejia R, Giza CC, Goldman JT. Concussion pathophysiology and injury biomechan-
ics. Curr Rev Musculoskelet Med. 2019;12:105–16.
3. McCrory P, Meeuwisse W, Johnston K, et al. Consensus statement on concussion in sport—the
3rd international conference on concussion in sport held in Zurich, November 2008. J Sci Med
Sport. 2009;12:340–51.
74 T. P. Yap and C. Stern
4. Davis GA, Anderson V, Babl FE, et al. What is the difference in concussion management in
children as compared with adults? A systematic review. Br J Sports Med. 2017;51:949–57.
5. Tanveer S, Zecavati N, Delasobera EB, Oyegbile TO. Gender differences in concussion and
postinjury cognitive findings in an older and younger pediatric population. Pediatr Neurol.
2017;70:44–9.
6. Barlow KM, Crawford S, Stevenson A, Sandhu SS, Belanger F, Dewey D. Epidemiology of post-
concussion syndrome in pediatric mild traumatic brain injury. Pediatrics. 2010;126:e374–81.
7. McCrory P, Meeuwisse W, Dvořák J, et al. Consensus statement on concussion in sport-the
5(th) international conference on concussion in sport held in Berlin, October 2016. Br J Sports
Med. 2017;51:838–47.
8. Iverson GL. Outcome from mild traumatic brain injury. Curr Opin Psychiatry. 2005;18:301–17.
9. Master CL, Master SR, Wiebe DJ, et al. Vision and vestibular system dysfunction predicts
prolonged concussion recovery in children. Clin J Sport Med. 2018;28:139–45.
10. Swanson MW, Weise KK, Dreer LE, et al. Academic difficulty and vision symptoms in chil-
dren with concussion. Optom Vis Sci. 2017;94:60–7.
11. Ellis MJ, Cordingley D, Vis S, Reimer K, Leiter J, Russell K. Vestibulo-ocular dysfunction in
pediatric sports-related concussion. J Neurosurg Pediatr. 2015;16:248–55.
12. Master CL, Scheiman M, Gallaway M, et al. Vision diagnoses are common after concussion in
adolescents. Clin Pediatr. 2016;55:260–7.
13. Ciuffreda KJ, Kapoor N, Rutner D, Suchoff IB, Han ME, Craig S. Occurrence of oculomotor
dysfunctions in acquired brain injury: a retrospective analysis. Optometry. 2007;78:155–61.
14. Hunt AW, Mah K, Reed N, Engel L, Keightley M. Oculomotor-based vision assessment in
mild traumatic brain injury: a systematic review. J Head Trauma Rehabil. 2016;31:252–61.
15. Armstrong RA. Visual problems associated with traumatic brain injury. Clin Exp Optom.
2018;101:716–26.
16. Petit L, Tzourio N, Orssaud C, Pietrzyk U, Berthoz A, Mazoyer B. Functional neuroanatomy
of the human visual fixation system. Eur J Neurosci. 1995;7:169–74.
17. Falck-Ytter T, Pettersson E, Bölte S, D'Onofrio B, Lichtenstein P, Kennedy DP. Difficulties
maintaining prolonged fixation and attention-deficit/hyperactivity symptoms share genetic
influences in childhood. Psychiatry Res. 2020;293:113384.
18. Ko HK, Poletti M, Rucci M. Microsaccades precisely relocate gaze in a high visual acuity task.
Nat Neurosci. 2010;13:1549–53.
19. Gallaway M, Scheiman M, Mitchell GL. Vision therapy for post-concussion vision disorders.
Optom Vis Sci. 2017;94:68–73.
20. Raghuram A, Cotter SA, Gowrisankaran S, et al. Postconcussion: receded near point of con-
vergence is not diagnostic of convergence insufficiency. Am J Ophthalmol. 2019;206:235–44.
21. Faucher C, De Guise D. Spasm of the near reflex triggered by disruption of normal binocular
vision. Optom Vis Sci. 2004;81:178–81.
22. Chan RV, Trobe JD. Spasm of accommodation associated with closed head trauma. J
Neuroophthalmol. 2002;22:15–7.
23. Scheiman M, Grady MF, Jenewein E, et al. Frequency of oculomotor disorders in adolescents
11 to 17 years of age with concussion, 4 to 12 weeks post injury. Vis Res. 2021;183:73–80.
24. Brahm KD, Wilgenburg HM, Kirby J, Ingalla S, Chang CY, Goodrich GL. Visual impairment
and dysfunction in combat-injured servicemembers with traumatic brain injury. Optom Vis
Sci. 2009;86:817–25.
25. Storey EP, Master SR, Lockyer JE, Podolak OE, Grady MF, Master CL. Near point of conver-
gence after concussion in children. Optom Vis Sci. 2017;94:96–100.
26. Santo AL, Race ML, Teel EF. Near point of convergence deficits and treatment following con-
cussion: a systematic review. J Sport Rehabil. 2020;29:1179–93.
27. DuPrey KM, Webner D, Lyons A, Kucuk CH, Ellis JT, Cronholm PF. Convergence insuf-
ficiency identifies athletes at risk of prolonged recovery from sport-related concussion. Am J
Sports Med. 2017;45:2388–93.
28. Adler PM, Cregg M, Viollier AJ, Margaret Woodhouse J. Influence of target type and RAF rule
on the measurement of near point of convergence. Ophthalmic Physiol Opt. 2007;27:22–30.
Advances in Management of the Oculomotor Dysfunction… 75
29. Bevilacqua ZW, Kawata K. Procedural shortcomings with near point of convergence assess-
ment may Lead to inappropriate prognosis of concussion injury in athletes: letter to the editor.
Am J Sports Med. 2018;46:Np64–np66.
30. Alnawmasi MM, Khuu SK. Deficits in multiple object-tracking and visual attention following
mild traumatic brain injury. Sci Rep. 2022;12:13727.
31. Dhawan PS, Leong D, Tapsell L, et al. King-Devick test identifies real-time concussion and
asymptomatic concussion in youth athletes. Neurol Clin Pract. 2017;7:464–73.
32. Hunfalvay M, Murray NP, Mani R, Carrick FR. Smooth pursuit eye movements as a biomarker
for mild concussion within 7-days of injury. Brain Inj. 2021;35:1682–9.
33. Bogle JM. Vestibular and balance dysfunction following sport-related concussion. Perspectives.
2019;4:1349–63.
34. Mucha A, Collins MW, Elbin RJ, et al. A brief Vestibular/Ocular Motor Screening (VOMS)
assessment to evaluate concussions: preliminary findings. Am J Sports Med. 2014;42:2479–86.
35. Gaur S, Awasthi SK, Bhadouriya SK, Saxena R, Pathak VK, Bisht M. Efficacy of Epley’s
maneuver in treating BPPV patients: a prospective observational study. Int J Otolaryngol.
2015;2015:487160.
36. Rowe FJ, Hanna K, Evans JR, et al. Interventions for eye movement disorders due to acquired
brain injury. Cochrane Database Syst Rev. 2018;3:Cd011290.
37. American Academy of Ophthalmology. Vision and concussion: symptoms, signs, evaluation,
and treatment. Pediatrics. 2022;150(2):e2021056047.
38. Yap TP, Luu CD, Suttle CM, Chia A, Boon MY. Electrophysiological and psychophysical stud-
ies of meridional anisotropies in children with and without astigmatism. Invest Ophthalmol Vis
Sci. 2019;60(6):1906–13.
39. Yap TP. Dyslexia and Vision–A review of current evidence and clinical interventions.
Singapore: Medical Grapevine, 2013; pp.10–15.
40. Roper-Hall G. Historical vignette: Louis Émile Javal (1839–1907): the father of orthoptics.
Am Orthopt J. 2006;57:131–6.
41. Ciuffreda KJ, Rutner D, Kapoor N, Suchoff IB, Craig S, Han ME. Vision therapy for ocu-
lomotor dysfunctions in acquired brain injury: a retrospective analysis. Optometry. 2008
Jan;79(1):18–22.
42. Applied concepts in vision therapy 2.0. Optometric Clinical Practice. 2022. Available through
the Optometric Extension Program Foundation (www.oepf.org/product/applied-concepts-
in-vision-therapy-2-0/). The book outlines practical guidelines on how to apply theoretical
concepts into clinical practice. Section 2 includes diagnostic and therapeutic guidelines for
traumatic brain injury.
Advances in Color Vision Testing
1 Introduction
Alterations in color vision have been recognized for almost three centuries, and
since their discovery, there have been attempts to qualify and quantify changes in
color vision perception. The theory of color vision deficiency (CVD) was first pro-
posed by John Dalton in the late eighteenth century, and reflected the recent discov-
ery of red, green, and blue as primary colors [1, 2]. The earliest tests of color vision
were developed to test vocational capacity in transportation careers [1]. Eventually,
as understanding of the perceptual changes present in CVD grew, tests were
designed to leverage the unique physiology of color vision for determination of
perceptual deficiencies.
In this chapter, we will review the basic mechanisms of color perception, the
deviations which cause CVD, and the traditional as well as the newer testing modal-
ities for screening, diagnosing, and evaluating color perception.
2 Color Vision
Vision is processed in the retina, and cone type photoreceptor cells are responsible
for color vision perception. Cones contain photopigments, comprised of an opsin
molecule and a retinol derivative [3, 4], which are capable of activating signaling
cascades upon reaction with photons. To be able to perceive complex color patterns,
the retina employs three cone types, that when combined, include most of the UV
light spectrum. Short (peak sensitivity ~419 nm), medium (peak sensitivity
~530 nm), and long (peak sensitivity ~560 nm) wavelength cones correspond to
blue, green, and red wavelengths, respectively. Activation of cone photopigments
therefore depends on the UV wavelength of the source. The relative signaling pat-
tern of cones is interpreted by the brain to form color vision perception.
There are multiple methods for assessing CVD, and thorough clinical evaluation
often relies on utilizing more than one test. The most recognized test uses the pseu-
doisochromatic plates (PIP) used in screening for CVD. More complex tests, such
as the hue arrangement testing (Farnsworth D15) and anomaloscope evaluation, are
used to diagnose specific deficiency patterns. There are also several occupationally
oriented tests used by various professional institutions to determine an individual’s
ability to complete work specific tasks. Importantly, all testing methods utilize the
unique physiology of color perception to isolate and evaluate the function of indi-
vidual cone types.
The basis of most CVD screening tests is the PIP. PIP tests are made by placing
randomly sized dots of varying color patterns to draw out numbers or shapes only
visible to individuals with color vision. The variations of the color patterns can dif-
ferentiate specific deficiencies [1, 3, 4, 13]. The first PIP test was developed by
German ophthalmologist, Dr. Jakob Stilling in the late nineteenth century [1, 4], and
in the subsequent century variations of these original tests have been made. The
straightforward nature of CVD testing using PIPs makes them ideal for screening.
Fig. 2 Ishihara plate #1 (left), the test plate, would show “12” in all individuals regardless of color
vision. Ishihara plate #9 (right) which would show “74” for normal trichromats and “21” for
dichromats
plates are considered reliable screening tests for congenital red-green deficiency,
with over 95% sensitivity and almost 100% specificity [17–19]. Ishihara plates are
unable to determine severity of deficiency though [1, 13, 20]. Additionally, the
Ishihara plates do not contain tritan sensitive plates, and therefore cannot be used to
detect tritan deficiency [1, 4].
Diagnostic Tests
Diagnostic tests have been developed for administration following screening tests to
further classify the deficiency present. Because of this, they are inclusive of all color
deficiencies, acquired and congenital, across the chromatic spectrum [1]. The most
common diagnostic test is a form of an arrangement test, in which an individual
arranges small caps filled with hues into their perceived natural order. The subjec-
tive order is then plotted to determine the specific CVD [4]. Variations of the tests
allow for more sophisticated determination of deficiency as well as recognition of
acquired variances.
82 S. I. Shoushtari and R. B. Singh
8
15
14 9
13
12 11 10
Advances in Color Vision Testing 83
3.1.4 Anomaloscope
The anomaloscope is considered the gold standard of CVD diagnostic testing. An
anomaloscope is an instrument to test color matching. An individual mixes two light
beams to adjust the proportion of each light until they feel it matches a given sample
[4, 15, 32, 33].
The results of the test are interpreted using various equations to assess the degree
and type of color deficiency. The original anomaloscope was created for evaluation
of red-green color deficiency using the Rayleigh equation, which dictates the tests
wavelength parameters [4, 32]. The Moreland test was subsequently created to
allowing for tritan deficiency testing, and similarly provides parameters and evalu-
ation criteria [32, 34]. Alternative versions, such as the Pickford-Lakowski or
Besancon test similarly can be used to assess tritan deficiency [4].
As every color on a computer display is essentially made from the base colors
red, green, and blue, an anomaloscope cannot be reproduced online. Therefore, a
simplistic reproduction, based on a different operating principle has been attempted
on portals such as, www.color-blind-test.com/anomaloscope-more.html.
The origins of CVD tests are in occupational testing, and the first standardized color
vision tests were developed to assess rail and naval employees’ abilities to safely
complete work duties [4]. Today various professional organizations continue to
include CVD testing in employee assessments to determine the ability of an indi-
vidual to perform occupational tasks [10, 31, 35].
3.2.1 Lantern
The most common occupational test in use dates to the nineteenth century, devel-
oped in response to repeated railway accidents at the time [4]. Lantern tests
involve showing various color signals and patterns to individuals and asking them
to state what they have been presented. The colors, speed, and spatial patterns of
the test should reflect relevant occupational distinction requirements and mimic
real signals the individuals would encounter [4, 27]. Thus, the lantern tests are not
reliable for screening or diagnosis of CVD and should only be used to determine
potentially hazardous visual defects [1, 4].Additionally, performance on lantern
test cannot be easily associated with performance on other CVD tests and accord-
ingly should not be used as a means of independent clinical diagnosis of CVD [13,
36, 37].
3.3.3 Electroretinography
Electroretinography (ERG) utilizes measurements of electrical signaling in the ret-
ina in response to light flashes to determine anomalies in color vision [27, 41]. ERG
offers an opportunity to further objectify CVD testing and decrease reliance on
subjective response to visual prompts for diagnosis [42, 43].
4 Limitations of Testing
The sensitivity and specificity of various CVD tests have been evaluated numerous
times and are extremely high across testing modalities and subsequent test versions
[13, 17–19, 21, 22, 31, 32, 40, 44]. However, there remains several limitations to the
currently available CVD testing methods.
Advances in Color Vision Testing 85
The most common issue with physically administered tests is the possibility for
variation. This can be a result of differences in ambient lighting during testing or
hue quality on plates and caps. Tests contain instructions on standard lighting condi-
tions to effectively administer the test [4]. There have been several studies that dem-
onstrate that changes in lighting source during testing can either improve anomalous
performance or increase failures in trichromats [45–50]. Additionally, physical
plates and hue caps have the potential to deteriorate over time, also affecting perfor-
mance of both normal and anomalous individuals. However, these limitations can
also apply to digital test, which must be similarly standardized to reduce the effect
of display screen variance on test efficacy [27].
Another limitation of current CVD testing methods is the lack of standardization
across tests [15]. Scores between modalities are not comparable and therefore grad-
ing of CVD severity is not uniform. Furthermore, there is no current procedure for
testing that allows for complete screening and diagnosis within one test. It is fre-
quently recommended that more than one test be utilized to ensure diagnose [4, 15].
A concern for many institutions that administer tests for occupational evaluation
is attempts to memorize test to ensure passing despite CVD [31]. This is especially
common with plate and lantern tests that have limited ability to randomize or adapt
to individual responses [31]. Digital tests that adapt to individual response, or more
complex tests such as hue arrangement, can provide a more reliable assessment of
color vision [15, 27, 38].
Improvement has been made to make CVD screening more accessible to indi-
viduals with intellectual disability, poor literacy, and children, however, many tests
remain inaccessible. Hue sorting test require physical mobility, and many PIP tests
require recognition of numerals [4].
5 Future Directions
Augmented reality and virtual reality technologies are being explored to create
immersive color vision testing environments. These platforms offer novel ways to
simulate real-world color discrimination tasks and evaluate color vision under vari-
ous lighting conditions, potentially improving the ecological validity of assessments.
Machine learning algorithms and AI techniques are being applied to analyze
large datasets of color vision test results [51]. These approaches can identify pat-
terns and correlations that may not be apparent to human observers, potentially
leading to new insights into color vision disorders and more accurate diagnostic
algorithms.
6 Conclusion
Color vision deficiency testing utilizes the unique physiology of human color per-
ception to isolate the function of cone types and assess their function and the prin-
ciples of color vision testing have remained the same for centuries. Developments
have been made recently to improve test administration and increase test
86 S. I. Shoushtari and R. B. Singh
References
1. French A, Rose K, Thompson K, Cornell E. The evolution of colour vision testing. Aust
Orthopt J. 2008;40:7–15.
2. Dalton J. Extraordinary facts relating to the vision of colours: with observations. England-
Manchester: Cadell and Davies; 1794.
3. Simunovic MP. Colour vision deficiency. Eye (Lond). 2010;24:747–55.
4. Melamud A, Hagstrom S, Traboulsi EI. Color vision testing. Ophthalmic Genet.
2004;25:159–87.
5. Simunovic MP. Major review acquired color vision deficiency. Surv Ophthalmol.
2016;61:132–55.
6. Weitz CJ, Went LN, Nathans J. Human tritanopia associated with a third amino acid substitu-
tion in the blue-sensitive visual pigment. Am J Hum Genet. 1992;51:444–6.
7. Baraas RC, Carroll J, Gunther KL, Chung M, Williams DR, Foster DH, Neitz M. Adaptive
optics retinal imaging reveals S-cone dystrophy in Tritan color-vision deficiency. J Opt Soc
Am. 2007;24:1438–47.
8. Carroll J, Neitz M, Hofer H, Neitz J, Williams DR, Nathans J. Functional photoreceptor
loss revealed with adaptive optics: an alternate cause of color blindness. Proc Natl Acad Sci
USA. 2004;101:8461–6.
9. Kohl S, Marx T, Giddings I, Jägle H, Jacobson SG, Apfelstedt-Sylla E, Zrenner E, Sharpe LT,
Wissinger B. Total colourblindness is caused by mutations in the gene encoding the α-subunit
of the cone photoreceptor CGMP-gated cation channel. Nat Genet. 1998;19:257–9.
10. Nathans J, Davenport CM, Maumenee IH, Lewis RA, Fielding Hejtmancik J, Litt M, Lovrien
E, Weleber R, Bachynski B, Zwas F, et al. Molecular genetics of human blue cone monochro-
macy. Science. 1989;245:831–8.
11. Schneck ME, Haegerstrom-Portnoy G, Lott LA, Brabyn JA. Comparison of Panel D-15 Tests
in a large older population. Optom Vis Sci. 2014;91:284–90.
12. Verriest G. Further studies on acquired deficiency of color discrimination. J Opt Soc Am.
1963;53:185–95.
13. Cole BL. Assessment of inherited colour vision defects in clinical practice. Clin Exp Optom.
2007;90:157–75.
14. Ishihara S. Tests for colour-blindness. Tokyo: Handaya Hongo Harukich; 1917.
15. Zarazaga AF, Gutiérrez Vásquez J, Royo VP. Revisión de Los Principales Test Clínicos Para
Evaluar La Del Color. Arch Soc Esp Oftalmol. 2019;94:25–32.
16. Miyahara E. Errors reading the Ishihara pseudoisochromatic plates made by observers with
normal colour vision. Clin Exp Optom. 2008;91:161–5.
17. Marechal M, Delbarre M, Tesson J, Lacambre C, Lefebvre H, Froussart-Maille F. Color vision
tests in pilots’ medical assessments. Aerosp Med Hum Perform. 2018;89:737–43.
18. Birch J. Efficiency of the Ishihara Test for identifying red-green colour deficiency. Ophthalmic
Physiol Opt. 1997;17:403–8.
19. Seshadri J, Christensen J, Bassi CJ. Evaluation of the new web-based “Colour Assessment and
Diagnosis” Test. Optom Vis Sci. 2005;82:882–5.
20. Hardy H, Rand G, Rittler MC. Tests for detection and analysis of color blindness: an evalua-
tion of the Ishihara Test. Arch Ophthalmol. 1945;34:295–302.
Advances in Color Vision Testing 87
21. Cole BL, Lian KY, Lakkis C. The new Richmond HRR Pseudoisochromatic Test for colour
vision is better than the Ishihara Test. Clin Exp Optom. 2006;89:73–80.
22. Birch J. Clinical use of the American Optical Company (Hardy, Rand and Rittler) pseudoiso-
chromatic plates for red-green colour deficiency. Ophthal Physiol Opt. 1997;17:248–54.
23. Alduhayan R, Almustanyir A. Comparison of two printed pseudoisochromatic tests for color
vision assessment. Optom Vis Sci. 2023;100(12):840–6.
24. Farnsworth D. The Farnsworth-Munsell 100-Hue and Dichotomous Tests for color vision*.
JOSA. 1943;33:568–78.
25. Munsell AH. A color notation: a measured color system based on the three qualities, Hue,
Values and Chroma with illustrative models, charts and a course of study arranged for teachers.
Boston: Geo H Ellis Co; 1907.
26. Hasrod N, Rubin A. Colour vision: a review of the Cambridge Colour Test and other colour
testing methods. Afr Vision Eye Health. 2015;74:23.
27. Lanthony P. The desaturated Panel D-15. Doc Ophthalmol. 1978;46:185–9.
28. Dain SJ, Adams AJ. Comparison of the Standard and Adams Desaturated D-15 Tests with
congenital colour vision deficiencies. Ophthalmic Physiol Opt. 1990;10:40–5.
29. Birch J. Clinical use of the City University Test (2nd Edition). Ophthalmic Physiol Opt.
1997;17:466–72.
30. Walsh DV, Robinson J, Jurek GM, Capó-Aponte JE, Riggs DW, Temme LA. A perfor-
mance comparison of color vision tests for military screening. Aerosp Med Hum Perform.
2016;87:382–7.
31. Zabel J, Przekoracka-Krawczyk A, Olszewski J, Michalak KP. Variability of Rayleigh and
Moreland Test results using anomaloscope in young adults without color vision disorders.
PLoS One. 2021;16:e0251903.
32. Nagel WA. Zwei Apparate Für Die Augenärztliche Funktionsprüfung Adaptometer Und Kleines
Spektralphotometer (Anomaloskop). Zeitschrift für Augenheilkunde. 1907;17:201–22.
33. Pokorny J, Smith VC, Went LN. Color matching in autosomal dominant Tritan defect. J Opt
Soc Am. 1981;71:1327.
34. Gao H, Kirkendall CD, Kinney MJ, Preston AM, Reddix MD. Color vision testing, standards,
and visual performance of the U.S. Military. Mil Med. 2023;188:49–57.
35. Cole B, Maddocks J. Can clinical colour vision test be used to predict the Farnsworth Lantern
Test? Vis Res. 1998;38:3483–5.
36. Birch J. Performance of colour-deficient people on the Holmes-Wright Lantern (Type A):
consistency of occupational colour vision standards in aviation. Ophthalmic Physiol Opt.
2008;28:253–8.
37. Melamud A, Simpson E, Traboulsi EI. Introducing a new computer-based test for the clinical
evaluation of color discrimination. Am J Ophthalmol. 2006;142:953–60.
38. Goulart PRK, Bandeira ML, Tsubota D, Oiwa NN, Costa MF, Ventura DF. A computer-
controlled color vision test for children based on the Cambridge Colour Test. Vis Neurosci.
2008;25:445–50.
39. Narayanan A, Venkadesan M, Krishnamurthy SS, Hussaindeen JR, Ramani KK. Dalton’s
pseudo-isochromatic plates and congenital colour vision deficiency. Clin Exp Optom.
2020;103:853–7.
40. Crognale MA, Teller DY, Motulsky AG, Deeb SS. Severity of color vision defects: electroreti-
nographic (ERG): molecular and behavioral studies. Vis Res. 1998;38:3377–85.
41. Aher AJ, Martins CMG, Barboni MTS, Nagy BV, Hauzman E, Bonci DMO, Ventura DF,
Kremers J. Electroretinographical determination of human color vision type. J Opt Soc Am
A. 2018;35:B92.
42. Hovis JK, Leat SJ, Heffernan S, Epp K. The validity of the University of Waterloo Colored Dot
Test for color vision testing in adults and preschool children. Optom Vis Sci. 2002;79:241–53.
43. Schmidt I. Effect of illumination in testing color vision with pseudo-isochromatic plates. J Opt
Soc Am. 1952;42:951–5.
88 S. I. Shoushtari and R. B. Singh
1 Introduction
An article published in 1964 by optometrist Dr. John Zettel discusses official ter-
minology for patients with partial vision loss, determining a need for classifying
the degree of blindness due to changes in social standards at the time. Visually
impaired patients were initially labeled as people with “subnormal vision,” how-
ever, Zettel recommended using the term “partially sighted individuals.” He also
made an outline of recommendations for these patients, including examination
techniques and low vision aids to prescribe [1].These aids included high add power
bifocals and base out prism glasses with increased power to aid in convergence and
fusion [1]. Today, “visually impaired” is the accepted terminology for this popula-
tion, and the specialty in caring for these individuals can be referred to as low
vision or vision rehabilitation. While high-power lenses are still widely prescribed,
updated technology has diversified and increased clinicians’ options in prescrip-
tion low vision aids.
The goal of low vision devices is to augment or maximize the remaining vision
that the patient has. When weighing appropriate devices for low vision patients, it is
necessary to consider their level of visual function, the type of vision loss, the
patient’s cognitive function, and the visual goal they are trying to accomplish.
Additionally, this decision-making process must consider the principle of least
effort, where options that are easier to implement and require less labor are more
favorable. Therefore, to prevent device abandonment and unnecessary patient costs,
a stepwise approach is necessary for choosing low vision devices.
Below are the general categories of devices used in low vision rehabilitation, listed
in increasing complexity.
• Spectacle correction
–– High reading adds
–– Microscopes
• Magnifiers
–– Handheld magnifiers
–– Stand magnifiers
• Telescopes
–– Handheld telescopes (Galilean, Keplerian)
–– Spectacle mounted (binocular vs. monocular)
• Electronic devices
–– Desktop CCTV (with or without OCR)
–– Portable CCTV
–– Head-mounted
–– Applications for smartphones/tablets
• Non-optical devices
–– Optical Character Recognition (OCR)
–– Tactile (e.g., Braille)
–– Audio devices (e.g., Talking watches, Siri, etc.)
These patients will fall into the visual acuity range of 20/20–20/70 (Table 1) and
will easily find improvement to their visual impairments through simple optical
devices. These include:
These patients will fall into the visual acuity range of <20/70–20/200. The patient’s
overall visual function becomes dependent on how both eyes work together. For exam-
ple, if the acuity of one eye still falls into the category of mild low vision, they may still
benefit from simple optical devices alone. However, if vision loss is complicated
by central scotomas or visual confusion the visual potential is limited and these patients
may function more similarly to someone with severe low vision. In such cases, elec-
tronic devices and smartphone applications may be more efficient at improving visual
function (See Sect. 2.3). Similarly, when patients have a hemianopsia following a
stroke or traumatic brain injury, their visual function is greatly impeded, though central
visual acuity is spared. In these scenarios, patients may benefit from a peripheral prism
(ex: Peli Prisms) to expand their awareness of the missing field.
These patients will fall into the visual acuity range of <20/200-no light perception.
At this stage, optical devices are often dependent on the acuity of both eyes indi-
vidually (as stated above). Patients will likely gain some benefit from optical devices
and will rely more on devices that augment their vision, such as electronic devices.
When the visual impairment is very severe to total blindness, the patient must rely
on other senses to function. This may mean using auditory cues from optical char-
acter recognition devices or tactile cues.
In the early 1800s, a young Louis Braille was introduced to night writing, a tactile
writing system that uses 12 raised dots in various arrangements. While night writing
was originally invented by Charles Barbier for soldiers to communicate in the dark,
Braille simplified and perfected the system by reducing the 12 dots arrangements to
6 dots and changing the symbols to stand for letters rather than sounds [3]. The
braille system is a well-known adaptation still used for visually impaired individu-
als and can be implemented presently as an accommodation. While braille is still
taught to children with vision impairment, adults are encouraged toward low vision
aids that can be seamlessly integrated into their life. Older individuals may not want
the extensive training required to learn a new way of reading or may struggle with
using tactile cues due to other comorbidities. Other available accommodations, such
as magnification, do not require learning a new language and allow patients to con-
tinue enjoying reading material they are familiar with.
Magnification-Optical magnifiers such as handheld, dome, and stand magnifiers
are commonly prescribed, due to being user-friendly, affordable, and often portable.
Traditionally, the potential magnification to read standard size print (1M) can be
determined with the patient’s recorded working distance and acuity. Based on this
92 S. Mortazavi and A. Meloottu
calculation, a low vision specialist can provide the patient with a demonstration and
a prescription for an optical magnifier. The magnification of a handheld or stand
magnifier cannot be adjusted; therefore, if vision continues to decline, a magnifier
with a lower power may be deemed useless to the patient. Video magnification, on
the other hand, such as portable digital magnifiers and CCTVs (closed circuit televi-
sions), can adjust magnification strength while providing other vision-enhancing
possibilities such as adjusting contrast and brightness. Video magnifiers can offer
magnification from 2× to 60×, providing patients the flexibility not offered by opti-
cal magnifiers. Another added benefit of digital magnifiers is comfort, primarily
through having a larger screen and a more natural reading position. Digital and
video magnifiers often include built-in stands that allow the screen to face the
patient at an angle or, in the case of desktop CCTVs, straight ahead. Unlike dome
and stand magnifiers, patients are not required to hover over the device to read on a
flat surface.
Electronic Magnification-Currently, there is a shift toward prescribing digital
magnification, such as desktop CCTVs, portable video magnifiers, and magnifying
smartphone applications. As mentioned, in addition to the possibility of adjusting
the magnification provided by the video magnifier, another substantial benefit of
such devices is the ability to change the text’s contrast. Contrast sensitivity is a
measure of visual function that describes the ability to differentiate the target lumi-
nance from the background luminance [4]. This measurement can be done with a
Pelli-Robson Chart or a MARS chart to find the logarithmic value of the contrast
sensitivity of the patient. Different values are assigned for different age groups and
are categorized as normal, moderate loss, severe loss, and profound loss. Contrast
sensitivity impairment is less commonly reported than visual acuity but may better
represent real-world goals and situations [4]. Visual acuity charts used in the exam
room are known to have 100% contrast with black letters on a white background.
However, the patient will likely encounter everyday circumstances that require high
contrast sensitivity, which become more critical on cloudy days, indoors, and in
other low lighting settings. A sign of reduced contrast can be difficulty seeing faces
or reading a newspaper, as both have a relatively low target to background
luminance ratio compared to a standard visual acuity chart. There are numerous
studies regarding the decrease in contrast sensitivity for various ocular diseases, and
even a study showing that glaucoma patients may have impaired contrast sensitivity
while having good visual acuity [5].
There are several methods to help patients with reduced contrast sensitivity.
Some digital magnifiers can increase contrast by changing the color of the reading
material or object being viewed. These color combinations can include an increased
contrast of white background with black writing, black background with white writ-
ing, yellow background with black writing, blue background with yellow writing,
and others—up to 14 different color combinations to increase the contrast of the
targeted material (e.g., See Fig. 1).
Tints-A decrease in contrast sensitivity can also be addressed with light filters or
tinted lenses. A tint assessment can be an element of a low vision examination,
depending on the patient’s vision loss and the pathology causing the loss. For
Advances in Low Vision Aids 93
a b
Fig. 1 (a and b) Textbook being magnified using a digital magnifier with “False Colors” setting
to increase contrast
interpretations of the same colors as well as other light filters scientifically proven
to be effective [7]. In addition to light filtering tints, glare reducing tints, like NoIR,
filter 99% of short wavelength light as well as infrared light to help reduce excess
glare [8].
Wearable Devices-Virtual reality (VR) headsets enable augmentation of a
patient’s visual acuity or expansion of their visual field through a wearable elec-
tronic device. Devices such as the IrisVision, Vision Buddy, eSight, and PatriotVision
all employ a camera that feeds a video image onto the internal device screen. The
head-mounted displays have a similar advantage to traditional binocular telescopes
as digital magnifiers have with optical magnifiers—flexibility. The hands-free
nature of the device affords the patient a more natural experience. The screen within
the headset allows for variable magnification using digital zoom, contrast enhance-
ments, and minification. While older VR devices are reportedly uncomfortable due
to the weight placed on the patient’s head, newer devices such as the IrisVision
Inspire and eSight Go are designed to fit more ergonomically, similar to glasses
[9–11]. While VR devices work for visual needs at all distances, they are more ben-
eficial for distance viewing. Many versions include OCR (Optical Character
Recognition) capabilities for reading, a live feed from the patient’s cable box for TV
viewing, phone connectivity for smartphone use, and voice control settings. The use
of VR devices while mobile is discouraged, as proprioception changes with magni-
fication and minification [12].
Optical Character Recognition (OCR)-Not all patients with visual impairment
can read comfortably with magnification alone. If vision reduction is more severe,
or scotomas and field loss interrupt the natural flow of reading, attempting to
increase reading ability may be frustrating and cumbersome for the patient. In these
circumstances, devices that utilize audio instead of visual support may be more
appropriate. Traditionally, patients with severe vision loss were limited to prere-
corded audiobooks and prerecorded object identification using barcode scanning.
This restricted identification to objects that had barcodes, and often would only
identify the object without providing more helpful information. For example, these
devices may be able to correctly identify a can of food but not read cooking instruc-
tions written on the back. Optical character recognition, also known as text recogni-
tion, deciphers printed text and converts it to a digital format. This technology aids
patients in reading virtually anything, including bills, newspapers, magazines, and
even text on everyday items. Devices with OCR capabilities range from desktop
CCTVs to wearable technology. Desktop devices such as Lyric or Eye-Pal Solo can
be a simple option for older patients or patients who don’t require the portability of
alternative aids. The primary benefit of these devices is the oversized tactile buttons,
which many patients find easier to utilize.
Smartphone Technology-There are many smart device applications for visually
impaired individuals, such as SeeingAI, EnVision, and Lookout-Assisted Vision.
These applications may be equipped with OCR technology, artificial intelligence to
describe a scene, options to help patients find an object in a room, and various other
features. OCR features within a smartphone application have the benefit of being an
inexpensive, more simplistic, and more immediate option when compared to
Advances in Low Vision Aids 95
desktop OCR devices Smartphones also have built-in accessibility options for visu-
ally impaired individuals that can be customized in the phone’s settings. Apple
iPhones have features such as zoom, magnifier, audio descriptions, and dictation, to
name a few [13]. Android phones are also equipped with similar accessibility
options, including customizing screen contrast, “talk back” or the screen’s ability to
be read aloud, and text magnification [14]. Both smartphone companies have tutori-
als that are available for patients and family members to make these accessibility
changes as an initial step toward independence with visual impairment. These
accessibility options allow a more seamless transition for patients to continue using
a device they are already familiar with. This also allows visually impaired individu-
als to not draw attention to their impairment should that be a concern to them.
Clinicians should familiarize themselves with these accessibility options to help
patients set up their phones for their low vision assessment in the clinic.
Similar to smartphone use described above, not all technology beneficial to visu-
ally impaired individuals was originally made for that purpose. Voice assistants
such as Apple’s Siri, Microsoft’s Cortana, Amazon’s Alexa, and Google Assistant
are available on smartphones or speaker devices. Siri and Google Assistant can send
and read texts, make phone calls, answer basic questions, set timers, and carry out
many other commands. Individuals without vision impairment use these voice
assistants regularly, but they also prove to be highly beneficial for low vision indi-
viduals. Smart home automation using Google Home and Alexa may also support
low vision individuals. Installing commands such as turning lights or appliances on/
off make previously difficult tasks for low vision individuals achievable with a few
spoken words. The thermostat may even be set to a specific temperature, rather than
manually adjusting it, negating the need for a handheld magnifier to see the small
numbers.
Mobility Aids-People with vision impairment face mobility challenges with
navigating indoor and outdoor settings [15]. For this specific functional challenge,
white canes and seeing-eye dogs continue to be commonly used among this popu-
lation. White canes provide the user beneficial feedback for obstacles in front of
them to encourage more independence while ambulating. However, learning to
use a white cane takes multiple training sessions from orientation and mobility
specialists to ensure confidence in the user. Newer technology aims to decrease
the training needed for traditional mobility aids as much as possible to encourage
seamless integration in the user’s life. This aspect is especially crucial for older
patients whose vision loss occurs later in life. Ultrasound devices on the market,
such as the Buzz clip, are designed for use with the white cane. This device uses
ultrasound to detect obstacles in the user’s path and alert them with vibrations.
The WeWALK smart handle can attach to any long cane and provides additional
mobility support, including vibration to inform the patient of low-hanging obsta-
cles that a cane alone would miss, such as a sign or tree branch. Additionally, the
patient can pair their cane to the WeWALK app on iOS or Android devices via
Bluetooth to make use of its public transportation features and other audio
alerts [16].
96 S. Mortazavi and A. Meloottu
With the rise in artificial intelligence use, wearable devices to aid in mobility,
such as the Biped, are entering the market for testing. The Biped is a harness worn
on the shoulders and is equipped with cameras, offering audio feedback to the
wearer, alerting them of any obstacles. This device advertises itself to function as a
“self-driving car” but for pedestrians, offering guidance to the wearer in real time
[17]. The Biped is currently being used in the USA and Europe and has plans to
include GPS (Global Positioning System) capabilities in upcoming updates [17]. As
mentioned earlier, these wearable devices can benefit patients because of the
reduced training required to utilize these devices.
Psychology Services-It is important to note that even with the devices and
accommodations available for visually impaired individuals, there is not a lack of
mental health difficulties associated with the loss of sight. Blindness is reported to
be one of the most feared health complications, greater than cancer or paralysis.
Loss of vision may cause anxiety, worry, embarrassment, and social withdrawal due
to negative self-perceptions [18]. Overall, vision loss can cause emotions similar to
bereavement, due to the loss of a heavily relied-on sense. Some mental health dif-
ficulties may stem from visual hallucinations that often occur with vision loss 20.
Patients may not report these hallucinations in fear of being perceived as having a
psychiatric disorder and are, therefore, less likely to receive support. In addition to
having an examination with a low vision provider, it is important to inquire about
the patient’s adaptation to their condition and offer a supportive space to share their
concerns. Providing resources available in the community, support groups, and a
referral to a psychiatric provider can offer patients with tools to better navigate the
emotional and social changes that accompany vision loss [17]. Currently there is
limited data to support the positive effects on mental health that support groups can
provide [18]. As research continues, there is a high likelihood of more data to sup-
port mental health interventions in patients with vision loss.
3 Case Scenario
A 51-year-old female presented for a low vision examination with a known history
of Stargardt Disease. The patient presented to the inherited retinal disease depart-
ment after 4 years with no reported change to her vision. She denied any issues with
her peripheral vision or night vision. Her visual acuity was 20/500 in the right eye
and 20/400 in the left eye—with a slight decrease in acuity in the right eye since last
seen. A battery of testing was performed, including a Goldman Visual Field of both
eyes, which revealed stable, dense central scotomas. Her last low vision evaluation
was several years prior and was now being referred back to the vision rehabilita-
tion department, as she had reported some difficulty with her activities of daily living.
As a previous patient of vision rehab services, she had received several devices
prior to this visit; however, she still found herself having difficulty seeing small
objects around the house. She also reported increased difficulty with reading, even
while using her current devices.
Advances in Low Vision Aids 97
The following is a review of her current devices when she presented for her low
vision evaluation:
1. Prism Magnifier Reading Glasses (+10D): Used for quick reading around the
house (e.g., reading labels in the kitchen), however found that this device
was not enough.
2. Desktop CCTV: Working well, used mostly for reading.
3. Computer enlargement software: Used for reading emails, and other computer
work - working well.
4. Ruby XL HD portable electronic magnifier: Working well, however, used infre-
quently as it was cumbersome to carry around the house or outside the house.
5. Smartphone accessibility settings: Patient is using all the accessibility settings
on her phone without difficulty. Not using any apps for magnification.
6. Eschenbach 7× handheld optical magnifier: Uses occasionally, working fine.
7. Long cane: No difficulty, no new mobility goals.
8. Beecher 5.5× binocular telescope: Working well uses this device most frequently
for distance viewing needs.
9. IrisVision head-mounted device: Using infrequently in the garden, watching TV,
reading.
Her main goal with this exam was to improve reading, and so while the above
devices did not adequately meet this need, the following devices were shown to her:
• The simpler devices were what the patient tended to use more frequently (e.g.,
Beecher binocular telescope, prism readers), following the principle of
least effort.
• Where the portable handheld CCTV (Ruby XL HD) was cumbersome to trans-
port, smartphone applications provided similar features with the addition of
other app features and ease of access (considering how consistently the patient
had her phone on her person).
4 Conclusion
Low vision device technology has advanced rapidly in recent years. To keep up with
the ever-increasing low vision population, it is necessary to have a good understand-
ing of the available devices and related resources for these patients. While optical
devices such as handheld magnifiers and telescopes remain within our baseline bat-
tery of low vision devices, newer technology such as VR headsets, OCR, and
98 S. Mortazavi and A. Meloottu
smartphone applications can give patients the most visual functional benefit and
with the most ease. Though many devices are available for patients to use, patients
are often unaware of all the tools at their disposal. Given this, it is ideal for clini-
cians to have a good knowledge of at least a few of these devices to better support
their visually impaired patients.
References
1. Zettel J Jr. The care of low vision. Am J Optom Arch Am Acad Optom. 1964;41:142–9.
2. Tan CW. Braille and the need to innovate for the blind. Ann Acad Med Singap. 2018;47(1):1–2.
3. Paulsen AJ, Pinto A, Fischer ME, Chen Y, Huang GH, Klein BEK, Klein R, Cruickshanks
KJ. Generational differences in the 10-year incidence of impaired contrast sensitivity.
Ophthalmic Epidemiol. 2021;28(2):175–82.
4. Fatehi N, Nowroozizadeh S, Henry S, Coleman AL, Caprioli J, Nouri-Mahdavi K. Association
of Structural and Functional Measures with contrast sensitivity in glaucoma. Am J Ophthalmol.
2017;178:129–39.
5. Reyes N, Huang JJ, Choudhury A, Pondelis N, Locatelli EVT, Hollinger R, Felix ER, Pattany
PM, Galor A, Moulton EA. FL-41 tint reduces activation of neural pathways of photophobia in
patients with chronic ocular pain. Am J Ophthalmol. 2024;259:172–84.
6. https://chadwickoptical.com/. Accessed 21 Jan 2024.
7. https://www.noirinsight.com/low-vision-sunglasses. Accessed 21 Jan 2024.
8. Schmidt DC, Kjølholm CDB, Torner Jordana J, Spange V, Højstrup PD, Larsen BS, Kessel
L. At-home trial of four different head-mounted displays in visual rehabilitation of people with
Stargardt disease. Clin Optom (Auckl). 2023;15:271–81.
9. https://irisvision.com/irisvision-inspire/. Accessed 21 Jan 2024.
10. https://www.esighteyewear.com. Accessed 21 Jan 2024.
11. Pur DR, Lee-Wing N, Bona MD. The use of augmented reality and virtual reality for visual
field expansion and visual acuity improvement in low vision rehabilitation: a systematic
review. Graefes Arch Clin Exp Ophthalmol. 2023;261(6):1743–55.
12. https://www.apple.com/accessibility/vision/. Accessed 21 Jan 2024.
13. https://www.android.com/accessibility/vision/. Accessed 21 Jan 2024.
14. Hoy MB. Alexa, Siri, Cortana, and more: an introduction to voice assistants. Med Ref Serv
Q. 2018;37(1):81–8.
15. Access World Home, American Foundation for the blind, March 2017 https://www.afb.org/
aw/18/3/15228. Accessed 21 Jan 2024.
16. https://www.biped.ai. Accessed on 21 Jan 2024.
17. Barasche-Berdah D, Paltiel O, Raz R. Active lifestyle and mobility of adults with vision impair-
ment: a multiphase mixedmethods study. Int J Environ Res Public Health. 2023;20(19):6839.
18. Demmin DL, Silverstein SM. Visual Impairment and Mental Health: Unmet Needs and
Treatment Options. Clin Ophthalmol. 2020;14:4229-51. https://doi.org/10.2147/OPTH.
S258783. PMID: 33299297; PMCID: PMC7721280.
Advances in Contact Lenses
for Ophthalmic Drug Delivery
In contemporary practice, more than 90% of ocular drug formulations used are eye
drops [1]. Despite their popularity, eye drops are generally inefficient drug delivery
systems due to rapid drainage of instilled drops from the ocular surface as well as
nonproductive absorption through routes leading away from the eye, resulting in
short precorneal residence time and less than 5% bioavailability, respectively [1].
This inefficiency in drug delivery necessitates multiple instillations of eye drops to
reach and maintain therapeutic concentrations, all of which can be significantly
impacted by patient compliance. Compliance is further compounded in chronic
conditions, with indefinite drug use and potential for multiple drugs being used
simultaneously. Multi use formulations also contain preservatives to protect against
microbial contamination but are known to have detrimental effects on the ocular
surface due to their cytotoxicity [2]. Nonproductive absorption systemically can
also cause detrimental side effects, with heart related complications after usage of
topical beta blockers for the treatment glaucoma a ready example [3].
It is within this context that researchers have investigated other means of deliv-
ering drugs to the eye, including use of medicated or therapeutic contact lenses
(TCLs). The field of drug delivery from contact lenses has seen important develop-
ments within the past decade, including the release of commercially available
products. This chapter will present an overview regarding the chemistry and
P. Shokrollahi
Centre for Ocular Research and Education, University of Waterloo, Waterloo, Canada
A. Hui (*)
School of Optometry and Vision Science, UNSW Sydney, Sydney, Australia
e-mail: [email protected]
material science behind contact lenses before detailing some of the methods and
means through which contact lenses have been investigated to deliver drugs to
the eye.
Table 1 Food and Drug Administration classification of the soft contact lenses
Low water content (<50%) Nonionic
High water content (>50%)
Low water content (<50%) Ionic
High water content (>50%)
— Silicone hydrogel
will be presented in this section. Some research groups have designed specific lens
formulations to fabricate TCLs [11, 12], while others have focused on developing
drug loading methods for commercially available lens formulations (with slight
modifications). The latter approach could potentially be more feasible for commer-
cialization as they use materials which are already available on the market.
2.1.3 Soaking
One of the simplest drug loading methods used in the preparation of therapeutic
contact lenses is soaking, which is a cost-effective, and widely used approach, par-
ticularly for hydrogel contact lenses. This method involves immersing a pre-formed
contact lens in a drug solution, allowing for drug diffusion into the lens via a con-
centration difference (known as osmotic pressure). Alternatively, eye drop instilla-
tion on a contact lens worn by the patient can also be used, although, sustained
release from the lens via this method is questionable [17]. When a therapeutic con-
tact lens is worn by a patient, drug release occurs via diffusion through pathways in
the contact lens to the tear film, both in the pre and post lens tear space. The overall
loading capacity of the contact lens in soaking depends on several factors, such as
drug concentration in the loading medium, lens thickness, hydrophilicity of the
drug, presence of hydrophilic/hydrophobic components in the lens, molecular
weight/size of the drug, lens incubation time, and water content [10]. However, it
should be noted that loading drugs can cause alteration of the lens critical properties
including increased swelling and decreased optical transparency, and in general,
soaking methods suffer from low drug loading and an uncontrolled, large initial
burst release of the drug of interest [10]. Chemical and environmental conditions,
such as concentration, pH, and temperature, can be adjusted to optimize loading
capacity, to some degree [10]. Innovative methods have been developed to address
some of the shortcomings of soaking, to try and increase drug loading or extend its
release. For example, Pluronic® F-68, a nonionic surfactant, has been used to dis-
solve gatifloxacin precipitates in the contact lens matrix, leading to improved drug
uptake. This approach resulted in acceptable swelling ratios (92.36%) and optical
transparency (92.84%) at carefully optimized levels and ratios of gatifloxacin to
Pluronic [18]. The presence of the surfactant also supported sustained release of
gatifloxacin for up to 72 h from these lenses, as evidenced both by in vitro and
in vivo experiments in a rabbit model [18].
to a significant increase in the drug loading capacity and a sustained release period
of bimatoprost. The GO-containing contact lenses were also transparent, with over
92% transparency, after loading with bimatoprost through soaking. This was attrib-
uted to the efficient dispersion of the drug molecules on the GO surface within the
lens matrix. Additionally, the water retention power of the GO nanoparticles
improved the swelling property of the drug-loaded composite contact lens [19].
Timolol: A similar strategy has been seen using gold nanoparticles. Silicone
hydrogel contact lenses were pre-loaded with gold nanoparticles (GNPs) during
manufacturing (GNPs-CL) and through soaking (GNPs-SS). The contact lenses
were then soaked in timolol solutions of two different concentrations (2 and 4 mg/
ml). The addition of the gold nanoparticles did not affect the swelling and optical
transmittance of the contact lenses, but the timolol loading significantly increased
using GNPs using both approaches. In vitro release profiles of timolol remained
almost unchanged for both methods, but in vivo measurements in rabbit tear fluid
showed higher timolol concentration with the GNPs-laden contact lenses at all time
points compared to those without GNPs. In vivo pharmacodynamic studies showed
a remarkable decrease in intraocular pressure (IOP) over 72 h using the GNPs-laden
contact lenses, whereas soaked contact lenses without GNPs and eye drop solutions
(0.5%w/v) only showed a decrease of 2 mmHg that lasted for only 12 h. Furthermore,
the drug deposition in the ciliary body significantly increased with the GNPs-laden
contact lenses, suggesting that incorporation of nanoparticles into these lenses may
lead to more of the timolol being bioavailable at the desired sites of action [20].
into soft hydrogel contact lenses through soaking, and then applied to freshly enu-
cleated porcine eyeballs. The hydrogel contact lenses and the hydrogel contact
lenses loaded with the cyclodextrin inclusion complexes delivered 2.8 and 5 times
more drug across the corneas, respectively, compared to econazole nitrate eye drops
at the same concentrations. Furthermore, while the eye drop led to concentrations
below the minimum inhibitory concentration (MIC) of the drug against test agents
of Fusarium semitectum and Fusarium solani (141 μg/mL) and Scolecobasidium
tshawytschae and Bipolaris hawaiiensis (283 μg/mL), the hydrogels containing the
inclusion complexes demonstrated efficient antifungal activity against these fungi,
suggesting a potential application in the treatment of keratitis [21].
causes it to swell, which allows for the mobile phase containing the drug to diffuse
more quickly. When pressure is released, the drug is deposited into the polymer via
two main mechanisms: by dispersing molecularly in the contact lens material (SFI)
or by precipitating within the hydrogel (supercritical fluid deposition or SFD).
Addition of small amounts of cosolvents can enhance the process by increasing
solvent polarity, promoting drug solubility, or by increasing polymer swelling and
pliability. Therefore, the amount of impregnated or dispersed drug can be regulated
by altering process conditions such as pressure and temperature, selecting suitable
cosolvents, and varying their compositions. While SFI results in a release profile
similar to that of soaking, SFD enables elongated and sustained release of the
drug [8].
A recent study showed that it is possible to create therapeutic contact lenses with
acetazolamide and timolol maleate using solvent mixtures of supercritical carbon
dioxide with ethanol or water (scCO2 + EtOH and scCO2 + H2O). The amount of
impregnated drug can vary depending on the type and concentration of cosolvent
used (in addition to operational conditions such as temperature and pressure), which
can be useful for controlling drug amounts for drugs with different hydrophilic/
hydrophobic character [24]. This technology has shown promise in improving drug
loading capacity and release duration for both hydrophilic and hydrophobic drugs.
However, the process can be expensive and time-consuming [25].
NP preparation
Nanoparticle method Lens material Drug(s) Disease Results and significance Ref.
mPEG-PLA Thin film pHEMA; free radical Timolol and Glaucoma In vivo (in rabbits) release was sustained for [26]
Micelle hydration polymerization of latanoprost timolol (120 h) and latanoprost (96 h); micelles
HEMA+NPs exhibited ultra-small size/narrow particle size
distribution and optical transparency; 9.8× higher
IOP reduction compared to eye drops.
PEGylated Solvent pHEMA(+DMA); free Latanoprost Glaucoma In vitro 120 h, and in vivo in rabbits 96 h; [27]
SLN evaporation radical polymerization; pegylated SLNs improved loading capacity and
soaked in NPs suspension sustained latanoprost release without
in STF, for 10 days compromising swelling, transmittance, and
oxygen permeability of the CLs. Lower protein
adherence than non-pegylated laden SLN lens.
Silica shell Microemulsion pHEMA(+MAA); free Ketotifen Ocular 240 h in vitro, and 240 h in vivo in rabbits’ eyes; [28]
nanoparticle- radical polymerization; Allergy extended drug delivery even with initial loading
laden monomers+ NPs (65 μg). In contrast to direct drug loading, NPs
laden CLs did not alter optical and swelling
properties, and ion permeability of CLs.
Drug-loaded Quasi- pHEMA(+MAA, NVP); Cyclosporine Chronic dry No drug loss was reported during monomer [29]
Eudragit emulsion CLs consisted drug laden eye syndrome extraction, 168 h drug release in vitro, 336 h
S100 NPs solvent ring and drug free center release, in vivo in rabbits’ eyes, remarkably longer
laden diffusion than in vitro, due to binding of drug to lipid (e.g.,
technique cholesterol) in tear fluid.
Poloxamer40/ Controlled Two CLs: 1. pHEMA Triamcinolone Ocular NSs with a mean particle size of 147 nm and a [30]
PVA precipitation (+MAA) radical solution acetonide inflammation drug load of 0.8 mg/ml using PVA/poloxamer
stabilized polymerization, 2. stabilizer, had no blurring effect on the CLs.
nano- Hilafilcon B (pHEMA Increased concentration of TA in the NSs improved
suspensions (+NVP) the drug-loaded dose significantly. Controlled TA
(NSs) release profiles for a period of 12 and 48 h for
Hilafilcon B and pHEMA-MAA, respectively.
P. Shokrollahi and A. Hui
PEG poly(ethylene glycol), pHEMA poly(2-hydroxyethyl methacrylate), IOP intraocular pressure, SLN solid lipid NPs, NPs nanoparticles, DMA N,N-dimethyl
acrylamide, MAA methacrylic acid, NVP N-vinylpyrrolidone, NSs nanospheres, TA Triamcinolone acetonide, PVA poly(vinyl alcohol)
Advances in Contact Lenses for Ophthalmic Drug Delivery 109
Fig. 2 (a) Host–guest inclusion complex formation between CDs ad hydrophobic drugs; (b)
Contact lens with drug-loaded implants
the adhered dead bacteria can be release and reloading of β-CD-LEV can be
achieved through immersion in a LEV solution [32].
The arguments for the development and research into drug-eluting contact lenses
center on the ubiquity of CLs having been safely worn by millions of users world-
wide over the last decades and the presumed pharmacokinetic advantages of such a
system compared to eye drops. In addition, therapeutic contact lenses can function
as a reservoir of preservative free drug that can remain in contact with the cornea
and thus facilitate ocular treatment of the drug so long as it is worn. The first drug
releasing contact lens has reached the market, specifically targeting the manage-
ment of ocular allergy with a daily disposable ketotifen releasing contact lens [23].
A vast group of commercial silicone hydrogels, conventional hydrogels, and daily
disposable contact lenses have been demonstrated to support sustained release of
ketotifen fumarate at a clinically relevant amount compared to conventional eye
drops when tested in vitro [36]. Ultimately, etafilcon A, which is a HEMA-based
soft contact lens, was utilized by Johnson & Johnson Vision Care to produce a daily-
wear, disposable, vision-correcting soft contact lens loaded with 0.019 mg ketotifen
and released to the market as Acuvue Theravision. This formulation became the first
drug-eluting contact lens to pass different phases of clinical trials successfully and
received FDA approval for the prevention of ocular itch due to allergic conjunctivi-
tis [37, 38]. Glaucoma releasing contact lenses are also in clinical trials. LLT-BMT1,
a contact lens printed with bimatoprost, has shown promising results in a Phase IIb
clinical study conducted by MediPrint™ Ophthalmics’ SIGHT-1 clinical program.
The product was well-tolerated by patients, and there was a lower incidence of
hyperemia compared to bimatoprost drops. The company has initiated [5] a Phase
IIb study for dosage optimization called SIGHT-2 and hopes to obtain dose results
by the first half of 2023, which will take the clinical study to the Phase III trial. As
further contact lens drug delivery devices become commercialized, acceptance of
these forms of alternative methods of treatment will become more of a reality for
prescribing practitioners and patients and will hopefully lead to improved patient
outcomes in treating these diseases [39, 40].
4 Conclusion
The potential for soft contact lenses, due to their water content in comparison with
rigid contact lenses, to deliver drugs was identified in the original patents for the
materials in the 1960s [41]. Research has continued in this area to demonstrate the
potential of these drug delivery devices within the laboratory with a variety of drugs,
materials, and testing conditions as well as work in vivo in animals or humans.
Matching of a material, drug, drug release kinetics, and disease to be treated will be
crucial things to identify for the future success of these devices, and the hope is that
the work published in the field will allow for this to be a successful means of ocular
drug delivery.
112 P. Shokrollahi and A. Hui
References
1. Mofidfar M, Abdi B, Ahadian S, Mostafavi E, Desai TA, Abbasi F, et al. Drug delivery to the
anterior segment of the eye: a review of current and future treatment strategies. Int J Pharm.
2021;607:120924.
2. Baudouin C. Detrimental effect of preservatives in eyedrops: implications for the treatment of
glaucoma. Acta Ophthalmol. 2008;86(7):716–26.
3. Farkouh A, Frigo P, Czejka M. Systemic side effects of eye drops: a pharmacokinetic perspec-
tive. Clin Ophthalmol. 2016;10:2433–41.
4. Musgrave CSA, Fang F. Contact lens materials: a materials science perspective. Materials
(Basel). 2019;12(2):261.
5. Akbari E, Imani R, Shokrollahi P, Heidari KS. Preparation of nanoparticle-containing ring-
implanted poly(vinyl alcohol) contact lens for sustained release of hyaluronic acid. Macromol
Biosci. 2021;21(7):e2100043.
6. Vivero-Lopez M, Pereira-da-Mota AF, Carracedo G, Huete-Toral F, Parga A, Otero A, et al.
Phosphorylcholine-based contact lenses for sustained release of resveratrol: design, anti-
oxidant and antimicrobial performances, and in vivo behavior. Acs Appl Mater Interfaces.
2022;14(50):55431–46.
7. Zhu Y, Li S, Li J, Falcone N, Cui Q, Shah S, et al. Lab-on-a-contact lens: recent advances and
future opportunities in diagnostics and therapeutics. Adv Mater. 2022;34(24):e2108389.
8. Xu J, Xue Y, Hu G, Lin T, Gou J, Yin T, et al. A comprehensive review on contact lens for
ophthalmic drug delivery. J Control Release. 2018;281:97–118.
9. Bhamra TS, Tighe BJ. Mechanical properties of contact lenses: the contribution of mea-
surement techniques and clinical feedback to 50 years of materials development. Cont Lens
Anterior Eye. 2017;40(2):70–81.
10. Rykowska I, Nowak I, Nowak R. Soft contact lenses as drug delivery systems: a review.
Molecules. 2021;26(18):5577.
11. Choi SW, Kim J. Therapeutic contact lenses with polymeric vehicles for ocular drug delivery:
a review. Materials. 2018;11(7):1125.
12. Peral A, Martinez-Aguila A, Pastrana C, Huete-Toral F, Carpena-Torres C, Carracedo
G. Contact lenses as drug delivery system for glaucoma: a review. Appl Sci. 2020;10(15):5151.
13. Jones L, Hui A, Phan CM, Read ML, Azar D, Buch J, et al. CLEAR-contact lens technologies
of the future. Cont Lens Anterior Eye. 2021;44(2):398–430.
14. Novack GD, Barnett M. Ocular drug delivery systems using contact lenses. J Ocul Pharmacol
Ther. 2020;36(8):595–601.
15. Liu Z, Overton M, Chauhan A. Transport of vitamin E from ethanol/water solution into contact
lenses and impact on drug transport. J Ocul Pharmacol Ther. 2022;38(6):396–403.
16. Maulvi FA, Singhania SS, Desai AR, Shukla MR, Tannk AS, Ranch KM, et al. Contact
lenses with dual drug delivery for the treatment of bacterial conjunctivitis. Int J Pharm.
2018;548(1):139–50.
17. Alvarez-Lorenzo C, Anguiano-Igea S, Varela-Garcia A, Vivero-Lopez M, Concheiro
A. Bioinspired hydrogels for drug-eluting contact lenses. Acta Biomater. 2019;84:49–62.
18. Maulvi FA, Parmar RJ, Desai AR, Desai DM, Shukla MR, Ranch KM, et al. Tailored gatifloxa-
cin Pluronic(R) F-68-loaded contact lens: addressing the issue of transmittance and swelling.
Int J Pharm. 2020;581:119279.
19. Maulvi FA, Soni PD, Patel PJ, Desai AR, Desai DT, Shukla MR, et al. Controlled bimatoprost
release from graphene oxide laden contact lenses: in vitro and in vivo studies. Colloids Surf B:
Biointerfaces. 2021;208:112096.
20. Maulvi FA, Patil RJ, Desai AR, Shukla MR, Vaidya RJ, Ranch KM, et al. Effect of gold
nanoparticles on timolol uptake and its release kinetics from contact lenses: in vitro and in vivo
evaluation. Acta Biomater. 2019;86:350–62.
Advances in Contact Lenses for Ophthalmic Drug Delivery 113
21. Wong A, Fallon M, Celiksoy V, Ferla S, Varricchio C, Whitaker D, et al. A composite system
based upon hydroxypropyl cyclodextrins and soft hydrogel contact lenses for the delivery of
therapeutic doses of econazole to the cornea, in vitro. Pharmaceutics. 2022;14(8):1631.
22. Pereira-da-Mota AF, Vivero-Lopez M, Topete A, Serro AP, Concheiro A, Alvarez-Lorenzo
C. Atorvastatin-eluting contact lenses: effects of molecular imprinting and sterilization on
drug loading and release. Pharmaceutics. 2021;13(5):606.
23. González-Chomón C, Silva M, Concheiro A, Alvarez-Lorenzo C. Biomimetic contact lenses
eluting olopatadine for allergic conjunctivitis. Acta Biomater. 2016;41:302–11.
24. Costa VP, Braga ME, Duarte CM, Alvarez-Lorenzo C, Concheiro A, Gil MH, et al. Anti-
glaucoma drug-loaded contact lenses prepared using supercritical solvent impregnation. J
Supercrit Fluids. 2010;53(1–3):165–73.
25. Xu J, Xue Y, Hu G. A comprehensive review on contact lens for ophthalmic drug delivery. J
Control Release. 2018;10(281):97–118.
26. Xu JW, Ge Y, Bu RX, Zhang AN, Feng SS, Wang J, et al. Co-delivery of latanoprost and
timolol from micelles-laden contact lenses for the treatment of glaucoma. J Control Release.
2019;305:18–28.
27. Dang H, Dong C, Zhang L. Sustained latanoprost release from PEGylated solid lipid nanopar-
ticle-laden soft contact lens to treat glaucoma. Pharm Dev Technol. 2022;27(2):127–33.
28. Maulvi FA, Mangukiya MA, Patel PA, Vaidya RJ, Koli AR, Ranch KM, et al. Extended release
of ketotifen from silica shell nanoparticle-laden hydrogel contact lenses: in vitro and in vivo
evaluation. J Mater Sci Mater Med. 2016;27(6):113.
29. Maulvi FA, Choksi HH, Desai AR, Patel AS, Ranch KM, Vyas BA, et al. pH triggered con-
trolled drug delivery from contact lenses: addressing the challenges of drug leaching during
sterilization and storage. Colloids Surf B Biointerfaces. 2017;157:72–82.
30. Garcia-Millan E, Quintans-Carballo M, Otero-Espinar FJ. Improved release of triamcinolone
acetonide from medicated soft contact lenses loaded with drug nanosuspensions. Int J Pharm.
2017;525(1):226–36.
31. Sun ZX, Zheng L, Wang K, Huai Z, Liu ZR. Primary vs secondary: directionalized guest coor-
dination in beta-cyclodextrin derivatives. Carbohydr Polym. 2022;297:120050.
32. Wen SM, Qin C, Shen LL, Liu D, Zhu SQ, Lin QK. Surface self-assembly construction of
therapeutic contact lens with bacterial “kill-releasing” and drug-reloading capabilities for effi-
cient bacterial keratitis treatment. ACS Biomater Sci Eng. 2022;8(3):1329–41.
33. Chatterjee S, Upadhyay P, Mishra M, Srividya M, Akshara MR, Kamali N, et al. Advances
in chemistry and composition of soft materials for drug releasing contact lenses. RSC Adv.
2020;10(60):36751–77.
34. Ross AE, Bengani LC, Tulsan R, Maidana DE, Salvador-Culla B, Kobashi H, et al.
Topical sustained drug delivery to the retina with a drug-eluting contact lens. Biomaterials.
2019;217:119285.
35. Desai AR, Maulvi FA, Desai DM, et al. Multiple drug delivery from the drug-implants-
laden silicone contact lens: Addressing the issue of burst drug release. Mater Sci Eng.
2020;112C:110885.
36. Soluri A, Hui A, Jones L. Delivery of ketotifen fumarate by commercial contact lens materials.
Optom Vis Sci. 2012;89(8):1140–9.
37. Ono J, Toshida H. Use of ketotifen fumarate-eluting daily disposable soft contact lens in man-
agement of ocular allergy: literature review and report of two cases. Cureus. 2022;14(7):e27093.
38. A drug-eluting contact lens for allergic conjunctivitis. Med Lett Drugs Ther.
2022;64(1648):60–1.
39. Balfour H. Drug-eluting contact lens shows potential in first-in-human study. Eur Pharm
Rev. 2021;
40. Ganatra S. MediPrint™ ophthalmics announces it has initiated its SIGHT-2 phase 2b clinical
study for its lead asset; 2022.
41. Sedlácek J. Possibility of the application of ophthalmic drugs with the use of gel contact
lenses. Cesk Oftalmol. 1965;21(6):509–12.
Novel Ocular Drug Delivery Methods
1 Introduction
There are many challenges involved in ocular drug delivery because of the unique
anatomical and physiological factors in the eye. Currently, 90% of all ophthalmic
drug formulations are applied as eye drops [1]. However, because of the static bar-
riers (including the cornea, conjunctiva, and sclera) and dynamic barriers (including
blinking reflex, high tear turnover rate, and rapid restoration of eye film), less than
5% of the applied dose can penetrate and reach intraocular tissue, leading to low
Y. Fang
School of Medical Sciences, University of Sydney, Camperdown, NSW, Australia
e-mail: [email protected]
C. Hodge
Vision Eye Institute Chatswood, Sydney, NSW, Australia
Save Sight Institute, The University of Sydney, Sydney, NSW, Australia
Graduate School of Health, University of Technology Sydney, Sydney, NSW, Australia
NSW Tissue Bank, Sydney, NSW, Australia
e-mail: [email protected]
G. Sutton
Vision Eye Institute Chatswood, Sydney, NSW, Australia
Save Sight Institute, The University of Sydney, Sydney, NSW, Australia
NSW Tissue Bank, Sydney, NSW, Australia
e-mail: [email protected]
J. You (*)
School of Medical Sciences, University of Sydney, Camperdown, NSW, Australia
Save Sight Institute, The University of Sydney, Sydney, NSW, Australia
e-mail: [email protected]
© The Author(s), under exclusive license to Springer Nature Singapore Pte 115
Ltd. 2024
D. Shu et al. (eds.), Current Advances in Optometry, Current Practices in
Ophthalmology, https://doi.org/10.1007/978-981-97-8140-9_9
116 Y. Fang et al.
bioavailability of approximately 1–3% [1, 2]. In the initial stages of the application
of eye drops, the majority will be washed away by tear production and blinking of
the eyes. The remaining drugs will be delivered through the corneal route and non-
corneal route as described below (Fig. 1).
The innate structure of the cornea negatively impacts drug penetration.
Anatomically, the cornea consists of five layers, from anterior to posterior; corneal
epithelium, Bowman’s layer, corneal stroma, Descemet’s membrane, and corneal
endothelium. Each layer has different priorities and potential rate-limiting structures
for drug permeability. Among the five structures of the cornea, epithelium and stroma
are considered two major drug diffusion barriers. The lipoidal epithelium cells num-
ber account for 90% of the total cornea cells, causing a significant resistance to
hydrophilic drug permeability. Furthermore, the junctional complex network of epi-
thelium also acts as a barrier to the diffusion of drugs [1]. Corneal stroma constitutes
90% thickness of the cornea that contains aligned cells guided by parallel hydrated
collagen lamellae [1]. The hydrophilic nature of stroma makes it a significant barrier
for hydrophobic drugs. Although relatively uncommon in practice, if the drugs are
targeting the posterior part of the eye, the drug will need to penetrate the cornea,
move into the anterior chamber and then through the iris, to the posterior chamber
and into posterior tissue. Among these structures, it is mainly the iris-ciliary body
and the aqueous humor that prevent further penetration of the drug from corneal
absorption. When the drug is distributed in the vascularized iris and ciliary body, it
can permeate into the blood vessels and move into the systemic circulation [3]. Drugs
in the anterior chamber may also be eliminated through aqueous humor outflow [3].
Understandably therefore, achieving therapeutic concentration levels in the pos-
terior segment is challenging through this long route. The non-cornea drug delivery
pathway involves the conjunctiva and sclera. Human conjunctiva is a highly vascu-
lar structure with approximately 17 times the surface area of the cornea, which
allows macromolecules (with a molecular weight ranging from 1000 to 20,000 Da)
to pass through with a permeability coefficient negatively related to solute size [4].
However, a large portion of the drug is absorbed by conjunctival blood capillaries
and lymphatics, and entry into system circulation before penetration into the ocular
tissue [1]. The sclera is made of connective tissue containing collagen fibers to pro-
tect and support the eye, which has comparable drug permeability with corneal
stroma and is more permeable for molecules with a smaller radius [1].
The low drug bioavailability of eye drops delivery mandates frequent administra-
tion. The highly frequent dosing schedule results in poor patient compliance and
may cause side effects in a long term. Although an extreme example, corneal micro-
bial keratitis patients need to instill eye drops every 30 min in the first 24–36 h in
mild to moderate cases [5]. Routinely, topical medication for reducing intra-
operative pressure may demand instillation between 2 and 4 times per day. The
frequent dosing schedule results in poor patient compliance and result in an extended
treatment period. Frequent instillation can lead to ocular surface disruption and irri-
tation or stinging on further application. Practically, frequent instillation has been
known also cause joint and muscle pain in the application finger, albeit in patients
with pre-existing conditions such as arthritis which may limit movement.
Many novel drug delivery systems have been explored to overcome the limita-
tions related to the topical ophthalmic formulation. The ideal drug delivery system
requires easy administration, long residence time, high biocompatibility, and does
not interfere with vision and normal eye function [2]. In this context, novel hydrogel
and nanoparticle drug delivery techniques will be discussed, as they are the most
frequently analyzed ocular drug delivery platforms that can be applied for both
anterior and posterior segment eye diseases.
2 Hydrogel
stability, but they are limited by inflammation response and cytotoxic by-products
after degradation. Hydrophilic polymers can be chemically or physically crosslinked
from liquid to gel to allow a longer residence time and show a sustained drug-releas-
ing profile representing a further possible advantage. Chemical crosslinked gels are
formed by covalently crosslinking chemical agents including N-hydroxysuccinimide
(NHS) and N,N-(3-dimethylaminopropyl)-N-ethylcarbodiimide hydrochloride
(EDC), which results in a stable structure [7]. Drug release in the covalent linkage is
usually achieved by enzyme degradation. However, the chemical crosslinker is usu-
ally cytotoxic and has potential biosafety concerns. Physically crosslinked hydrogel
is normally formed by forces including van der Waals, hydrogen bonding, and elec-
tronic bonding. Hydrogels formed by physical interactions are typically less stable
and are known to be thermally reversible [7].
High initial drug release, also known as burst release of drug molecules, has been
a challenge in hydrogel drug delivery (Fig. 2). The typical hydrogel biomaterial has
a mesh size range from 5 to 100 nm, which is larger than most small drug molecules
(molecular dimensions of 0.5–1.5 nm) [8]. Therefore, after being transferred to the
hydrogel drug delivery system in an aqueous environment, small drug molecules
can quickly diffuse from the hydrogel network in the swelling process, resulting in
an initial burst release. Burst release can result from other reasons such as the
desorption of the drug attaching to the surface of hydrogel and the poor distribution
of the drug in the network [9]. Silicone hydrogel and Poly(hydroxyethyl methacry-
late) (pHEMA) hydrogel show burst release profiles for most ophthalmic drugs with
release lasting for a few hours [10]. The burst release phenomenon usually has nega-
tive effects, including cause cytotoxicity and tissue irritation in the human body
[11]. Therefore, a constant rate of drug release, also referred to as zero-order sus-
tained drug release, is usually the preferred method of drug delivery [9]. For exam-
ple, the Retisert implant is a zero-order drug delivery system made from silicone
polymer and has been approved by Food and Drug Administration (FDA). It is used
to treat chronic non-infectious uveitis in the posterior segment of the eye, providing
a continuous release of fluocinolone acetonide for about 30 months after surgical
implantation at the back of the eye.
Fig. 2 The illustration of drug release with initial burst and zero-order kinetics
Novel Ocular Drug Delivery Methods 119
To enhance drug absorption and accomplish sustained and regulated drug release
in the hydrogel, it may be necessary to enhance molecule interactions and binding
mechanisms within the delivery system. There are various chemical, physical, and
biological factors that have been researched to improve drug delivery. Covalent con-
jugation between the hydrogel polymer and drug can allow a stable binding, which
is cleavable through enzyme degradation to allow drug release [6]. The mucoadhe-
sive effect induced by the physical interaction between polymer and mucin can also
improve drug delivery. As an example, the mucoadhesive property of chitosan poly-
mer makes it one of the most widely used drug delivery materials [12]. Applying a
chitosan-based drug delivery system to the mucosal surface results in an interaction
between chitosan and mucin, creating a strong and long-lasting adhesion that facili-
tates sustained drug release at the site. The physical factors including swelling char-
acteristics and mechanical deformation of the hydrogel can also impact the drug
delivery of hydrogel. For instance, encapsulating cyclodextrin in pHEMA hydrogel
can lead to an increase in swelling behaviors, resulting in improved drug uptake
[13]. With the ability of mechanical deformation from liquid to solid in response to
environmental changes, the in situ hydrogel has become one of the most important
hydrogel systems. A liquid formation in most cases provides an easier application
however transformation to a solid gel increases the potential time in situ further
leveraging the sustained release properties [14]. In addition to in situ hydrogel, con-
tact lenses with high bioavailability and collagen shields with high biocompatibility
have also frequently been investigated as ocular drug delivery systems and will be
discussed in the following sections.
One of the most important types of hydrogels is in situ hydrogels. In situ gelling
systems are stimuli-responsive hydrogels, which can be applied to the eye as liquid
and then mechanically transformed into gels. The in situ reversible hydrogels can
swell, or de-swell based on the change in ion concentration, temperature or pH. All
these types of stimuli-responsive hydrogels have been widely researched in
ophthalmology.
Since in situ hydrogel exists in liquid form prior to mechanical transformation, it
has been applied as a topical instillation on the corneal surface or injection into
intraocular tissues, as discussed, the transformation leads to a significant increase in
precorneal drug contact time, and consequently, an increase in bioavailability [14].
A combination of polyacrylic acid (Carbopol® 980NF) and hydroxypropyl methyl-
cellulose (Methocel® K100LV) was utilized to prepare a pH-triggered hydrogel
system for delivering ciprofloxacin [14]. To prevent any potential incompatibility
between the drug and polyacrylic acid, ciprofloxacin was complexed with an ion
exchange resin. The optimized product sustained drug delivery for 24 h in vitro,
although the release kinetics followed a zero-order pattern for only up to 10 h.
Important for both application and sustainability, it can maintain its stability for
3 months without any changes in clarity, drug content, pH, or gelling ability.
120 Y. Fang et al.
Moreover, in a rabbit model, no irritation was observed upon application of this in-
situ hydrogel.
The injectable in situ hydrogels are especially appealing to be adapted in poste-
rior eye diseases through intravitreal injection as liquid and transformed into solid
form in vivo. This process serves to not only minimize the invasive effect but will
prolong the time required for the drug to be released. Lai et al. developed an in situ
hydrogel delivery system by utilizing gelatin grafted with carboxylic end-capped
poly(N-isopropylacrylamide) to administer pilocarpine for glaucoma treatment
[15]. The hydrogel underwent a phase transition from 25 to 34 °C and exhibited a
cumulative drug release over 14 days, with an initial burst release in the first 30 min.
In a rabbit model, the hydrogel was administered intracamerally and showed excel-
lent bioadhesion, no inflammatory response, and degradation by MMP-2 over time.
The hydrogel showed three times higher drug concentration in the aqueous humor
compared to eye drop instillation after 4 h of treatment, indicating higher bioavail-
ability. This represents a potential tangible difference with practical benefits to the
patient and the treatment of the ocular condition.
The contact lens has also been proposed as a drug delivery system showing signifi-
cantly improved bioavailability than topical formulations. Contact lens delivery sys-
tems achieve an extended residence time of more than 30 min, which is significantly
higher than the normal residence time of only 1–3 min in eye drop delivery [2]. As
expected, the longer residence time increased the drug bioavailability by up to 50%,
suggesting the possibility of less frequent therapy, leading to higher patient compli-
ance and potentially a faster healing process [2]. Against this, contact lens insertion
will often require trained personnel for application. In 2022, FDA approved the first
drug-eluting contact lenses, the ACUVUE® Theravision™ made of etafilcon A
with ketotifen to prevent the itched eye induced by allergic conjunctivitis and cor-
rection of refractive ametropia [16]. Every lens contains 19 mcg of ketotifen, how-
ever, the process for how ketotifen combined with etafilcon A remains proprietary.
Containing almost 2/3 water content (59%), it is expected to have an excellent
safety profile [16]. Wetting agents such as polyvinylpyrrolidone are used in Etafilcon
A lenses to mimic natural tear films and further improve comfort through both ini-
tial and extended wear [17]. Its clinical study showed less than 2% of participants
experienced eye irritation or pain as a result of the procedure [17]. This initial prod-
uct validated the promising effect of using contact lenses as a drug delivery system.
This section will discuss various novel designs for encapsulating drugs in therapeu-
tic contact lenses.
lenses, the drug is released within a few hours. A recent study used molecular
imprinting to deliver multiple drugs simultaneously. Diclofenac sodium (DS) and
dexamethasone sodium phosphate (DMSP) or bromfenac sodium (BS) and moxi-
floxacin (MOX) were molecularly imprinted to contact lens [19]. Both DS + DMSP-
loaded lenses and BS + MOX-loaded lenses showed sustained release of loaded
drugs over 1 week. The stability of the mesh size before and after molecular imprint-
ing confirms that the controlled release is attributed to macromolecular memory
rather than a tighter structure or smaller mesh size.
With high biocompatibility and biodegradability, collagen shield has also been
explored as an ocular drug delivery system [23]. The collagen shield usually con-
tains type I and type III collagen and is reserved in dehydrated form, which needs to
be rehydrated before use [23]. The FDA has approved several collagen shields on
the market, including Biocora®, MediLens®, and ProShield®, but there is no clear
information about their composition or patient feedback. Collagen shields usually
start to dissolve after 12, 24, and 72 h [23]. After the designed dissolution period,
the collagen shield will lose 1/2 to 1/3 of its mass. By this point, its geometry will
have changed, and it will become loose on the surface of the eye, allowing it to be
easily removed.
Collagen shields can increase the drug contact time and maintain the concentra-
tion by serving as a reservoir and gradually releasing the drug as the shield dis-
solved [24]. Collagen shield has shown comparable or superior ability in the
delivery in rabbit models of many antibiotics including vancomycin, gentamicin,
tobramycin, and prednisolone acetate [25]. A 12-h collagen shield was soaked in
prednisolone acetate for 15 mins prior to application in rabbit eyes. As compared
to topical application, the drug concentration levels in the aqueous and cornea were
4–5 times higher after 30 mins and 100–200 times higher after 120 min [24].
Another study evaluated the drug delivery of dexamethasone with a 24-h collagen
shield in both the anterior and posterior segments of rabbit eyes [25]. Compared
with the single drop instillation, a 2–5-fold higher drug level was observed in the
20-min pre-soaked shield in the cornea, aqueous, iris, and vitreous over a 6-h
period. This study further compared the treatment of hourly eye drop alone to a
pre-soaked collagen shield plus hourly eye drop. The results showed collagen
shields with supplementary hourly eye drops can maintain twofold higher drug
levels in the cornea, aqueous, iris, and vitreous compared with hourly eye drops
alone over 6 h highlighting a significant potential additive impact. Despite the col-
lagen shield’s advantages over topical eye drops, it is limited by its requirement of
professional installation [23]. Similar to contact lenses, the burst release phenom-
enon still exists in the collagen shield drug delivery [26]. Furthermore, although
the collagen shield has high tolerance with regard to comfort in the eye, it has low
transparency, resulting in a reduction in visual acuity to 6/36–6/60 [23]. Although
there are some collagen shields have been approved by FDA, they are still not fre-
quently used in the clinic.
3 Nanosystems
endocytic pathways and can be used to deliver poorly permeable small molecules
and proteins [28]. In general, nanocarriers with smaller sizes remain preferred
because of lower irritation and better bioavailability [27]. Several factors affect the
mechanism of drug release from these nanocarriers, including the stability of the
carrier, drug–carrier interaction, and drug diffusion [27]. A variety of nanocarriers
have been studied for use as ocular drug delivery systems. The three most widely
studied nanocarriers are liposomes, dendrimers, and polymeric nanoparticles
including nanospheres and nanocapsules.
3.1 Liposome
3.2 Dendrimers
of terminal groups [34]. In general, those dendrimers with higher generation and
positive charges on their surface exhibited greater cytotoxicity [34].
a b
Fig. 6 Structure of (a) nanosphere and (b) nanocapsule for drug delivery application
References
1. Gaudana R, Ananthula HK, Parenky A, Mitra AK. Ocular drug delivery. AAPS
J. 2010;12:348–60.
2. Maulvi FA, Soni TG, Shah DO. A review on therapeutic contact lenses for ocular drug deliv-
ery. Drug Deliv. 2016;23:3017–26.
3. Agrahari V, et al. A comprehensive insight on ocular pharmacokinetics. Drug Deliv Transl Res.
2016;6:735–54.
4. Huang A, Tseng S, Kenyon K. Paracellular permeability of corneal and conjunctival epithelia.
Invest Ophthalmol Vis Sci. 1989;30:684–9.
5. Gokhale NS. Medical management approach to infectious keratitis. Indian J Ophthalmol.
2008;56:215–20.
6. Li J, Mooney DJ. Designing hydrogels for controlled drug delivery. Nat Rev Mater.
2016;1:16071.
7. Lynch CR, et al. Hydrogel biomaterials for application in ocular drug delivery. Front Bioeng
Biotechnol. 2020;8:228.
8. Bhattarai N, Gunn J, Zhang M. Chitosan-based hydrogels for controlled, localized drug deliv-
ery. Adv Drug Deliv Rev. 2010;62:83–99.
9. Hezaveh H, Muhamad II. Controlled drug release via minimization of burst release in pH-
response kappa-carrageenan/polyvinyl alcohol hydrogels. Chem Eng Res Des. 2013;91:508–19.
10. Karlgard CCS, Wong NS, Jones LW, Moresoli C. In vitro uptake and release studies of ocular
pharmaceutical agents by silicon-containing and p-HEMA hydrogel contact lens materials. Int
J Pharm. 2003;257:141–51.
11. Huang X, Brazel CS. Analysis of burst release of proxyphylline from poly(vinyl alcohol)
hydrogels. Chem Eng Commun. 2003;190:519–32.
12. Mohsen AM. Cationic polymeric nanoparticles for improved ocular delivery and antimycotic
activity of terconazole. J Pharm Sci. 2022;111:458–68.
13. Xu J, Li X, Sun F. Cyclodextrin-containing hydrogels for contact lenses as a platform for drug
incorporation and release. Acta Biomater. 2010;6:486–93.
14. Jain SP, Shah SP, Rajadhyaksha NS, Singh PSP, Amin PD. In situ ophthalmic gel of ciprofloxa-
cin hydrochloride for once a day sustained delivery. Drug Dev Ind Pharm. 2008;34:445–52.
15. Lai JY, Hsieh AC. A gelatin-g-poly(N-isopropylacrylamide) biodegradable in situ gelling deliv-
ery system for the intracameral administration of pilocarpine. Biomaterials. 2012;33:2372–87.
16. FDA (US Food and Drug Administration). Highlights of prescribing information ACUVUE®
Theravision™ with Ketotifen. Food and Drug Administration Web site. https://www.access-
data.fda.gov/drugsatfda_docs/label/2022/022388s000lbl.pdf. Accessed 2022.
17. Miller J, Giedd B, Subbaraman LN. Clinical comparison of a silicone hydrogel and a conven-
tional hydrogel daily disposable contact lens. Clin Ophthalmol. 2021;15:4339–45.
18. Horne RR, Rich JT, Bradley MW, Pitt WG. Latanoprost uptake and release from commercial
contact lenses. J Biomater Sci Polym Ed. 2020;31:1–19.
19. DiPasquale SA, Uricoli B, DiCerbo MC, Brown TL, Byrne ME. Controlled release of multiple
therapeutics from silicone hydrogel contact lenses for post-cataract/post-refractive surgery and
uveitis treatment. Transl Vis Sci Technol. 2021;10:5.
20. Malakooti N, Alexander C, Alvarez-Lorenzo C. Imprinted contact lenses for sustained release
of polymyxin B and related antimicrobial peptides. J Pharm Sci. 2015;104:3386–94.
21. Yokozaki Y, Sakabe J, Ng B, Shimoyama Y. Effect of temperature, pressure and depressur-
ization rate on release profile of salicylic acid from contact lenses prepared by supercritical
carbon dioxide impregnation. Chem Eng Res Des. 2015;100:89–94.
22. Ciolino JB, et al. A drug-eluting contact lens. Invest Ophthalmol Vis Sci. 2009;50:3346–52.
23. Willoughby CE, Batterbury M, Kaye SB. Collagen corneal shields. Surv Ophthalmol.
2002;47:174–82.
24. Sawusch MR, O’Brien TP, Updegraff SA. Collagen corneal shields enhance penetration of
topical prednisolone acetate. J Cataract Refract Surg. 1989;15:625–8.
130 Y. Fang et al.
25. Hwang DG, Stern WH, Hwang PH, MacGowan-Smith LA. Collagen shield enhancement of
topical dexamethasone penetration. Arch Ophthalmol. 1989;107:1375–80.
26. Zhou S, et al. Release of moxifloxacin from corneal collagen shields. Eye Contact Lens.
2018;44(Suppl 2):S143–s147.
27. Omerović N, Vranić E. Application of nanoparticles in ocular drug delivery systems. Heal
Technol. 2020;10:61–78.
28. Mosquera J, García I, Liz-Marzán LM. Cellular uptake of nanoparticles versus small mol-
ecules: a matter of size. Acc Chem Res. 2018;51:2305–13.
29. Feghhi M, Sharif Makhmalzadeh B, Farrahi F, Akmali M, Hasanvand N. Anti-microbial effect
and in vivo ocular delivery of ciprofloxacin-loaded liposome through Rabbit’s eye. Curr Eye
Res. 2020;45:1245–51.
30. Abdelbary G. Ocular ciprofloxacin hydrochloride mucoadhesive chitosan-coated liposomes.
Pharm Dev Technol. 2011;16:44–56.
31. Taha EI, El-Anazi MH, El-Bagory IM, Bayomi MA. Design of liposomal colloidal systems for
ocular delivery of ciprofloxacin. Saudi Pharm J. 2014;22:231–9.
32. Esfand R, Tomalia DA. Poly(amidoamine) (PAMAM) dendrimers: from biomimicry to drug
delivery and biomedical applications. Drug Discov Today. 2001;6:427–36.
33. Lancina MG 3rd, Wang J, Williamson GS, Yang H. DenTimol as a Dendrimeric timolol ana-
logue for glaucoma therapy: synthesis and preliminary efficacy and safety assessment. Mol
Pharm. 2018;15:2883–9.
34. Janaszewska A, Lazniewska J, Trzepiński P, Marcinkowska M, Klajnert-Maculewicz
B. Cytotoxicity of dendrimers. Biomol Ther. 2019;9(8):330.
35. Franco P, De Marco I. Contact lenses as ophthalmic drug delivery systems: a review. Polymers
(Basel). 2021;13:1102.
36. Jacinto TA, Oliveira B, Miguel SP, Ribeiro MP, Coutinho P. Ciprofloxacin-loaded zein/hyal-
uronic acid nanoparticles for ocular mucosa delivery. Pharmaceutics. 2022;14:1557.
37. Sharma S, Parveen R, Chatterji BP. Toxicology of nanoparticles in drug delivery. Curr
Pathobiol Rep. 2021;9:133–44.
38. Garhwal R, et al. Sustained ocular delivery of ciprofloxacin using nanospheres and conven-
tional contact lens materials. Invest Ophthalmol Vis Sci. 2012;53:1341–52.
39. Hosny KM. Ciprofloxacin as ocular liposomal hydrogel. AAPS PharmSciTech. 2010;11:241–6.
40. Bajgrowicz M, Phan CM, Subbaraman LN, Jones L. Release of ciprofloxacin and moxifloxa-
cin from daily disposable contact lenses from an in vitro eye model. Invest Ophthalmol Vis
Sci. 2015;56:2234–42.
Clinical Trials Relevant for Optometrists
The landmark clinical trials in Ophthalmology provide an inside look at the thera-
peutic pipeline and how the standardized treatment protocols can objectively guide
patient care, which strategies to adopt and which to discontinue. This chapter enlists
a few landmark clinical trials which may help optometry practitioners and students
to uncover better ways to prevent, diagnose, treat, and understand common ophthal-
mic disorders. For ease of the readers, the trial results and conclusions have been
tabulated.
© The Author(s), under exclusive license to Springer Nature Singapore Pte 131
Ltd. 2024
D. Shu et al. (eds.), Current Advances in Optometry, Current Practices in
Ophthalmology, https://doi.org/10.1007/978-981-97-8140-9_10
132 P. Ichhpujani and S. Kapoor
Most infants are born hyperopic, and their hyperopia reduces as the eyeball elon-
gates alongwith flattening of cornea. Both genetic and environmental factors influ-
ence the onset and progression of myopia. Increased near work, less outdoor
activities, and lifestyle ruled by smartphones/tablets are responsible for the early
development of myopia at a younger age. The prevalence rate of myopia is increas-
ing worldwide, and as per WHO, it is projected to increase to 52% by 2050 [5]. The
available treatment options for the control of myopia include spectacle correction,
contact lenses, keratorefractive surgery, and pharmaceuticals agents such as low
dose atropine. However, there are no perfect therapeutic modalities that effectively
halt and slow down the development of myopia without causing significant side
effects.
LAMP study evaluated the efficacy and safety of low-concentration atropine eye
drops at 0.05%, 0.025%, and 0.01% compared with placebo over a 1-year period.
[6] The study was a double-blinded, randomized, and placebo-controlled therefore
it provided robust evidence. It has delineated a concentration-dependent response,
both as regards efficacy and side effect profile in the low-atropine concentration
range from 0.05% to 0.01%. Higher concentration of low-concentration atropine
0.05% is most efficacious among the three concentration.
The LAMP phase 2: In a follow-up study, over 2 years, the efficacy of 0.05%
atropine observed was double that of 0.01% atropine, and it remained the optimal
concentration among the studied atropine concentrations in slowing myopia pro-
gression [7].
ATOM 2 was carried out to compare efficacy and visual side effects of three lower
doses of atropine: 0.5%, 0.1%, and 0.01%. [9]
A rebound phenomenon was seen following the cessation of atropine 1%, 0.5%,
0.1%, and 0.01%, in ATOM 1 and ATOM 2 studies.
LAMP phase 3 study plans to look more into the rebound phenomenon by ran-
domizing each of the three groups 0.05%, 0.025%, and 0.01% into wash out group
and treatment-continued group, to evaluate the efficacy of 0.05%, 0.025%, and
0.01% atropine group during 3 years; whether treatment should be stopped after
2 years of atropine.
Clinical Trials Relevant for Optometrists 135
Primary angle closure disease constitutes a spectrum that includes primary angle
closure suspects (PACS), primary angle closure (PAC, occludable angles with signs
suggestive of outflow obstruction in absence of glaucomatous optic neuropathy) and
primary angle closure glaucoma (PACG; occludable angles with glaucomatous
optic neuropathy).
Most practitioners routinely offer prophylactic laser peripheral iridotomies (LPI)
to PAC and PACG patients and only to a select few PACS ones. The ZAP trial was
conducted to assess the efficacy and safety of LPI prophylaxis for Chinese patients
with PACS [10]. The study found that although the LPI is effective in preventing
progression from PACS to PAC, given the low incidence of development of PAC
both in untreated and treated groups, LPI is not mandatorily recommended in glau-
coma screening programs. However, the low rates of progression of PACS could be
unique to Chinese population and this must be kept in mind when making clinical
decisions regarding LPI for patients with PACS.
The conclusion was that some individuals with OHTN might benefit from receiv-
ing early treatment, whereas most others were at such low risk that they did not need
antiglaucoma treatment.
Both of the original randomization groups received medication during the OHTS 2
[12]. In OHTS phase 2, participants previously randomized to treatment for a mean
of 7.5 years remained on treatment for a mean of 5.5 additional years. The data was
analyzed to determine if the cumulative incidence of POAG was greater in the
delayed treatment group and if the subsequent course after diagnosis is worse in the
delayed treatment group. OHTS 2 found that the incidence of POAG was generally
equal in the two randomization groups (HR, 1.06).
Delaying treatment in the observation group during the OHTS 1 did not initiate
a process of irreversible deterioration in most participants. Most OHT patients are
at low risk could be followed at less frequent intervals without treatment.
The OHTS Phase 3 built up on the groundwork done by the previous two studies. It
aimed to determine the incidence and severity of POAG in the cohort and develop a
20-year prediction model including the previous factors identified and potential new
risk factors [13]. In the OHTS 3, more than 70% of patients were receiving topical
Clinical Trials Relevant for Optometrists 137
ocular hypotensive treatment, and some patients had undergone glaucoma surgery.
It was noted that only one-fourth of participants in the OHTS developed visual field
loss in either eye over long-term follow-up. The cumulative incidence of POAG at
20 years, adjusted for exposure time, was 45.6%. The incidence of POAG appeared
to be generally linear over 20 years, with a possible modest increase in the rate of
conversion after 15 years. The study developed a model that identified slow versus
fast visual field progressors.
Not all OHT patients who need treatment for IOP control prefer antiglaucoma med-
ications. Selective laser trabeculoplasty (SLT) could offer an efficacious and adverse
effect free temporary solution. The answer came from the Laser in Glaucoma and
Ocular Hypertension (LiGHT) Trial, which has shown that SLT is a cost-effective
solution for both OAG and OHT at 3 years [14]. This trial reported that the health-
related quality of life (HRQoL) and clinical effectiveness of initial treatment with
SLT compared with IOP-lowering eye drops after 6 years of treatment.
Disease
entity Study group Results Conclusion
718 Group 1 = SLT * 652 (91%) returned SLT is a safe treatment for OAG and
patients group the primary outcome OHT, providing better long-term
Group questionnaire at disease control than initial drop
2 = antiglaucoma 36 month therapy, with reduced need for
drops * Average EQ-5 D incisional glaucoma and cataract
score: SLT: 0.89; eye surgery over 6 years
drops: 0.90
References
1. Holmes JM, Beck RW, Kraker RT, et al. Pediatric Eye Disease Investigator Group. Risk of
amblyopia recurrence after cessation of treatment. J AAPOS. 2004;8(5):420–8.
2. Repka MX, Beck RW, Holmes JM, et al. Pediatric Eye Disease Investigator Group. A ran-
domized trial of patching regimens for treatment of moderate amblyopia in children. Arch
Ophthalmol. 2003;121(5):603–11.
3. Louison S, Blanc J, Pallot C, Alassane S, Praudel A, Bron AM, et al. Visual outcomes and
complications of congenital cataract surgery. J Fr Ophtalmol. 2019;42(4):368–74.
4. Lenhart PD, Lambert SR. Current management of infantile cataracts. Surv Ophthalmol.
2022;67(5):1476–505.
5. Singh H, Singh H, Latief U, Tung GK, Shahtaghi NR, Sahajpal NS, Kaur I, Jain SK. Myopia,
its prevalence, current therapeutic strategy and recent developments: a review. Indian J
Ophthalmol. 2022;70(8):2788–99.
138 P. Ichhpujani and S. Kapoor
6. Yam JC, Jiang Y, Tang SM, Law AKP, et al. Low-concentration atropine for myopia progres-
sion (LAMP) study: a randomized, double-blinded, placebo-controlled trial of 0.05%, 0.025%,
and 0.01% atropine eye drops in myopia control. Ophthalmology. 2019;126(1):113–24.
7. Yam JC, Li FF, Zhang X, Tang SM, et al. Two-year clinical trial of the low-concentration atropine
for myopia progression (LAMP) study: phase 2 report. Ophthalmology. 2020;127(7):910–9.
8. Chua WH, Balakrishnan V, Chan YH, Tong L, Ling Y, Quah BL, Tan D. Atropine for the treat-
ment of childhood myopia. Ophthalmology. 2006;113(12):2285–91.
9. Chia A, Chua WH, Cheung YB, Wong WL, Lingham A, Fong A, Tan D. Atropine for the treat-
ment of childhood myopia: safety and efficacy of 0.5%, 0.1%, and 0.01% doses (atropine for
the treatment of myopia 2). Ophthalmology. 2012;119(2):347–54.
10. Han JE, Myo A, Mathew RG, Henein C. Infographic: Zhongshan angle closure prevention
(ZAP) trial. Eye (Lond). 2021;35(12):3185–6.
11. Gordon MO, Kass MA. What we have learned from the ocular hypertension treatment study.
Am J Ophthalmol. 2018;189:xxiv–xxvii.
12. Gordon MO, Gao F, Beiser JA, Miller JP, Kass MA. The 10-year incidence of glau-
coma among patients with treated and untreated ocular hypertension Arch Ophthalmol
2011;129(12):1630–1631.
13. Kass MA, Heuer DK, Higginbotham EJ, Parrish RK, Khanna CL, Brandt JD, Soltau JB,
Johnson CA, Keltner JL, Huecker JB, Wilson BS, Liu L, Miller JP, Quigley HA, Gordon
MO, Ocular Hypertension Study Group. Assessment of cumulative incidence and severity of
primary open-angle glaucoma among participants in the ocular hypertension treatment study
after 20 years of follow-up. JAMA Ophthalmol. 2021;139(5):1–9.
14. Gazzard G, Konstantakopoulou E, Garway-Heath D, Adeleke M, Vickerstaff V, Ambler
G, et al. Laser in glaucoma and ocular hypertension (LiGHT) trial. Ophthalmology.
2023;130(2):139–51.
Artificial Intelligence and Optometry:
Transforming Practice and Patient Care
1 Introduction
S. Majithia
Education and Professional Services, Essilor Luxottica, Singapore
e-mail: [email protected]
S. Thakur (*)
Singapore Eye Research Institute, Singapore, Singapore
Medical Affairs, Mediwhale, Seoul, South Korea
© The Author(s), under exclusive license to Springer Nature Singapore Pte 139
Ltd. 2024
D. Shu et al. (eds.), Current Advances in Optometry, Current Practices in
Ophthalmology, https://doi.org/10.1007/978-981-97-8140-9_11
140 S. Majithia and S. Thakur
characteristic curve (AUC-ROC), precision, recall, and F1-score. This chapter aims
to provide a focused exploration of the impact of AI on optometry, with a particular
emphasis on FDA-approved AI products, the transformative potential of early detec-
tion and screening, and the evolving scope of optometric practice in the AI era.
While acknowledging the vast landscape of AI applications in optometry, this chap-
ter prioritizes key themes and developments relevant to practitioners and stakehold-
ers. It serves as a concise overview, offering insights into the current state of AI
integration in optometry and outlining future directions for research and practice.
In recent years, the FDA has granted approval to several AI-driven technologies
designed to enhance diagnostic accuracy and streamline patient care within the field
of optometry and ophthalmology. These innovations harness the power of AI, par-
ticularly ML/DL algorithms, to analyze medical imaging data and detect ocular
abnormalities with high precision and efficiency. Notably, these FDA-approved AI
products represent a significant milestone in the integration of AI into clinical prac-
tice, offering optometrists valuable tools for early detection and management of
various ocular conditions. Notable examples include:
The EyeArt system has been designed to detect DR and vision-threatening diabetic
retinopathy (vtDR) autonomously. A prospective multicenter cross-sectional diag-
nostic study, conducted from April 17, 2017, to May 30, 2018, evaluated its safety
and accuracy in detecting mtmDR and vtDR in individuals with diabetes. Out of
942 consenting individuals aged 18 years or older, 893 patients (1786 eyes) met the
inclusion criteria and completed the study protocol across 15 primary care and eye
Artificial Intelligence and Optometry: Transforming Practice and Patient Care 141
care facilities. The system demonstrated high sensitivity and specificity in detecting
both mtmDR (sensitivity: 95.5%, specificity: 85.0%) and vtDR (sensitivity: 95.1%,
specificity: 89.0%) without dilation. After correction for enrichment, the specificity
for mtmDR increased to 87.8%, and both sensitivity (97.0%) and specificity (90.1%)
improved for vtDR [2, 3].
These FDA-approved AI products and similar country specific derivatives have
had a profound impact on the screening and diagnosis of ocular conditions, particu-
larly DR. Their deployment in screening programs has significantly enhanced the
efficiency and effectiveness of DR screening, leading to early detection and inter-
vention in at-risk populations [4]. In many countries, AI-based solutions are starting
to become integral components of national DR screening programs, offering scal-
able and cost-effective approaches to managing this sight-threatening complication
of diabetes [4–8]. Moreover, some countries, such as Singapore and Turkey, have
developed their own AI solutions tailored to their population samples, further illus-
trating the versatility and adaptability of AI across diverse scenarios [9–11].
In the realm of community vision screening, there exists a pressing need for an
efficient and accurate method to detect individuals requiring referral to specialized
eye care centers due to vision loss associated with various eye conditions. The
advent of AI and especially DL presents a promising avenue to transform this clini-
cal referral pathway. Tham et al., in their study aimed to evaluate the efficacy of a
newly developed DL algorithm in detecting disease-related visual impairment using
retinal fundus images [12]. Drawing from a dataset comprising 15,175 eyes with
complete data on visual acuity from the Singapore Epidemiology of Eye Diseases
(SEED) Study, researchers embarked on a proof-of-concept endeavor. Initially, a
single-modality DL algorithm was crafted solely from retinal photographs to dis-
cern any disease-related visual impairment, defined as eyes with major eye diseases
and best-corrected visual acuity of less than 20/40, as well as moderate or worse
142 S. Majithia and S. Thakur
3.2 Cataract
cataracts among older adults. The algorithm’s high accuracy and sensitivity suggest
its efficacy in providing more appropriate referrals to tertiary eye centers, thereby
facilitating timely interventions for individuals with cataract-related visual impair-
ment. Another study demonstrated the effectiveness of the universal AI platform that
uses anterior segment photos and multilevel collaborative pattern in diagnosing and
managing cataracts [14]. The research also explored an AI-based medical referral
pattern aimed at enhancing collaborative efficiency and resource coverage in cataract
management. The training and validation datasets were sourced from the Chinese
Medical Alliance for Artificial Intelligence, covering diverse healthcare facilities and
capture modes. The datasets underwent a three-step labeling process, which involved
recognizing capture modes, diagnosing cataracts, and detecting referable cataracts
based on etiology and severity. Additionally, the study integrated the cataract AI
agent with a real-world multilevel referral pattern that involved self-monitoring at
home, primary healthcare, and specialized hospital services. The results demon-
strated the robust diagnostic performance of the universal AI platform and multilevel
collaborative pattern across three key tasks. These tasks included capture mode rec-
ognition, cataract diagnosis (normal lens, cataract, or postoperative eye), and detec-
tion of referable cataracts. The AI platform achieved impressive AUC values ranging
from 91% to over 99% in all tests. In the real-world tertiary referral pattern, the AI
agent suggested referrals for 30.3% of individuals, significantly increasing the doc-
tor-to-population service ratio by 10.2-fold compared to the traditional pattern. The
findings suggest that this AI-based medical referral pattern could be extended to
other common disease conditions and resource-intensive situations, offering poten-
tial benefits for public health initiatives.
In recent years, deep learning systems (DLSs) have emerged as powerful tools for
screening various eye diseases, including DR, glaucoma, age-related macular
degeneration (AMD), and other fundus abnormalities. Several studies have evalu-
ated the performance of DLS in detecting these diseases, highlighting their potential
utility in optometry practice for early diagnosis and intervention. Ting et al., in a
pioneering study, evaluated the diagnostic performance of a DLS for detecting
referable diabetic retinopathy, vision-threatening diabetic retinopathy, possible
glaucoma, and AMD in multiethnic populations with diabetes [15]. Using a vast
dataset of retinal images, the DLS demonstrated high sensitivity and specificity for
identifying these conditions, paving the way for efficient screening and referral
strategies. Expanding upon this research, subsequent studies have developed DL
models capable of screening multiple abnormal findings in retinal fundus images.
These models were trained and validated using large datasets from diverse sources,
achieving high accuracy in detecting 12 major findings, including hemorrhage, hard
exudate, cotton-wool patch, drusen, and glaucomatous disc changes [16]. The DL
algorithms exhibited performance comparable to human experts, demonstrating
their potential as automated screening systems for various retinal abnormalities.
144 S. Majithia and S. Thakur
The Optometry 2040 initiative from Optometry Australia, launched in 2018, pres-
ents three distinct scenarios envisioning the future of optometry, alongside key
trends shaping the profession given in Table 1.
These scenarios highlight potential trajectories for the profession, emphasizing
the need for adaptation to technological advancements. Scenario 2, identified as
optimal, underscores the importance of embracing collaborative and technology-
enabled care delivery. Key trends identified in the report include consistently-
evolving technology, enlarged scope of practice, and consumer-centric care,
underscoring the importance of embracing AI and adapting practice models.
Table 1 Scenarios discussed under optometry 2040 initiative by Optometry Australia [19]
Scenario Description
1. Optometry fails to Technological and clinical advancements render much of optometry’s
adapt diagnostic work redundant, leading to underemployment and
decreased salaries
2. The eye care you Optometrists become trusted guides through the eye care system,
want—As well as facilitated by collaborative professional-patient relationships and
the eye care you technology-enabled care delivery. This scenario is considered optimal
need for the profession
3. Optometry beyond Optometrists lead technological innovation, becoming ‘optical
the dark room and scientists’ and integrating with other disciplines to optimize
out of the box healthcare
Artificial Intelligence and Optometry: Transforming Practice and Patient Care 145
The American Academy of Optometry (AAO) survey provides insights into optom-
etrists’ perspectives on AI integration in eye care [20]. The study was conducted
among 400 optometrists who completed a survey assessing perceived advantages
and concerns regarding AI using a 5-point Likert scale. The majority of respondents
indicated that they were familiar with AI, with approximately 66.8% reporting such
familiarity. However, approximately half of optometrists expressed concerns regard-
ing the diagnostic accuracy of AI, with around 53.0% expressing such reservations.
Despite these concerns, a significant majority, accounting for 72.0% of respondents,
believed that the integration of AI would enhance the practice of optometry.
Furthermore, optometrists’ willingness to integrate AI into their practice showed a
notable increase, rising from 53.3% before the COVID-19 pandemic to 65.5% fol-
lowing the onset of the pandemic (p < 0.001).
The convergence of insights from Optometry 2040 and the AAO survey under-
scores the transformative potential of AI in optometric practice. While Optometry
2040 provides a visionary outlook on the future of the profession and identifies key
trends shaping its trajectory, the AAO survey offers a snapshot of current percep-
tions and attitudes toward AI integration among optometrists. Together, these
resources offer valuable perspectives on the opportunities and challenges associated
with AI adoption in optometry, highlighting the importance of strategic planning,
education, and collaboration to navigate the evolving landscape of eye care delivery.
As optometry moves toward an AI-enabled future, it is essential for practitioners to
embrace innovation while upholding core principles of patient-centered care and
professional excellence.
Large language models (LLMs) have emerged as powerful tools with significant
potential to transform various aspects of optometry practice. These models, such as
ChatGPT, Google Bard (now Gemini), and Bing Chat, are trained on vast datasets
and possess human-like text generation capabilities. In the context of optometry,
LLMs can play a crucial role in enhancing diagnosis, treatment, patient care, and
expanding the scope of practice. One area where LLMs demonstrate promise is in
aiding during optometric exams and consultations. For example, eye-specific LLMs,
specifically designed for ophthalmic applications, have been evaluated for their per-
formance in assisting with the Fellowship of Royal College of Ophthalmologists
(FRCOphth) exams [21]. These models have shown varying degrees of accuracy in
answering multiple-choice questions related to ophthalmology, with differences
observed among different LLM-chatbots. This underscores the importance of con-
tinued research and refinement to optimize the performance of LLMs in supporting
eye related education and assessment. In addition to assisting with clinical decision-
making, LLMs can also facilitate patient communication and education [22].
ChatGPT, for instance, can be integrated into virtual consultations, allowing patients
146 S. Majithia and S. Thakur
to ask questions and receive personalized responses regarding their eye health con-
cerns. This not only enhances patient engagement but also promotes proactive man-
agement of ocular conditions.
Moreover, foundation models like RETFound represent a significant advance-
ment in leveraging AI for eye care. RETFound, developed by Moorfields Eye
Hospital and University College London, is an AI foundation model specifically
trained 1.6 million unlabeled retinal images by means of self-supervised learning
[23]. This model has demonstrated the ability to detect sight-threatening eye diseases
and even predict systemic health challenges such as heart attacks, stroke, and
Parkinson’s disease. One notable advantage is their ability to reduce the need for
extensive dataset collection. Unlike conventional AI models that require massive
amounts of labeled data for training, foundation models leverage self-supervised
learning techniques to learn from unannotated or minimally labeled data. This means
that RETFound can extract valuable insights from a relatively small dataset of retinal
images, thereby streamlining the data collection process and reducing the burden on
healthcare institutions to amass large datasets. Moreover, foundation models enable
faster and more customized development of AI solutions through techniques such as
few-shot and zero-shot learning. Few-shot learning allows models like RETFound to
adapt to new tasks or domains with minimal labeled examples, making it possible to
tailor the model to specific clinical applications rapidly. For example, RETFound can
be fine-tuned to detect rare or emerging eye diseases using only a handful of anno-
tated images, accelerating the development of novel diagnostic tools. Zero-shot
learning takes this concept further by enabling the model to generalize to tasks it has
never encountered before. This capability is particularly valuable for rare diseases,
where clinicians may not have enough data or samples to train models. With
RETFound, optometrists can leverage the model’s pretrained knowledge to address
new challenges without the need for additional data collection or model retraining.
Furthermore, foundation models like RETFound democratize AI by making state-of-
the-art technology accessible to a broader audience. Traditionally, AI development
has been confined to research labs and tech giants with the resources to collect and
process massive datasets [24]. However, with the advent of foundation models, even
smaller clinics or research institutions can harness the power of AI to improve patient
care. By providing open access to pretrained models and resources for model adapta-
tion, solutions like RETFound empower clinicians and researchers to innovate and
develop AI solutions tailored to their unique needs and patient populations.
However, the integration of LLMs and foundation models into current practice
necessitates upskilling and adaptation among optometrists. Continuous learning
and development programs are essential to ensure that practitioners are proficient in
leveraging and using AI tools effectively. Strategies for embracing AI, such as
incorporating LLM-driven chatbots into practice workflows and collaborating with
technologists to develop specialized models, are vital for optimizing the benefits of
AI integration. Overall, the impact of LLMs and foundation models in vision sci-
ence extends beyond conventional diagnostic and treatment paradigms. These mod-
els hold the potential to revolutionize the way we deliver care, interact with patients,
and contribute to advancements in ocular health.
Artificial Intelligence and Optometry: Transforming Practice and Patient Care 147
References
1. Abràmoff MD, Lavin PT, Birch M, Shah N, Folk JC. Pivotal trial of an autonomous AI-based
diagnostic system for detection of diabetic retinopathy in primary care offices. NPJ Digit Med.
2018;1(1):39.
2. Ipp E, Liljenquist D, Bode B, Shah VN, Silverstein S, Regillo CD, et al. Pivotal evaluation of
an artificial intelligence system for autonomous detection of referrable and vision-threatening
diabetic retinopathy. JAMA Netw Open. 2021;4(11):e2134254.
3. Lim JI, Regillo CD, Sadda SR, Ipp E, Bhaskaranand M, Ramachandra C, et al. Artificial intel-
ligence detection of diabetic retinopathy: subgroup comparison of the EyeArt system with
ophthalmologists’ dilated examinations. Ophthalmol Sci. 2023;3(1):100228.
4. Heydon P, Egan C, Bolter L, Chambers R, Anderson J, Aldington S, et al. Prospective evalua-
tion of an artificial intelligence-enabled algorithm for automated diabetic retinopathy screen-
ing of 30,000 patients. Br J Ophthalmol. 2021;105(5):723–8.
5. Cleland CR, Bascaran C, Makupa W, Shilio B, Sandi FA, Philippin H, et al. Artificial intel-
ligence-supported diabetic retinopathy screening in Tanzania: rationale and design of a ran-
domised controlled trial. BMJ Open. 2024;14(1):e075055.
6. Ruamviboonsuk P, Tiwari R, Sayres R, Nganthavee V, Hemarat K, Kongprayoon A, et al. Real-
time diabetic retinopathy screening by deep learning in a multisite national screening pro-
gramme: a prospective interventional cohort study. Lancet Digit Health. 2022;4(4):e235–e44.
7. Arenas-Cavalli JT, Abarca I, Rojas-Contreras M, Bernuy F, Donoso R. Clinical validation of
an artificial intelligence-based diabetic retinopathy screening tool for a national health system.
Eye. 2022;36(1):78–85.
8. Huang XM, Yang BF, Zheng WL, Liu Q, Xiao F, Ouyang PW, et al. Cost-effectiveness of
artificial intelligence screening for diabetic retinopathy in rural China. BMC Health Serv Res.
2022;22(1):260.
9. MOH | News Highlights 2024 [updated 2024/03/23/]. Available from: https://www.moh.gov.
sg/news-highlights/details/efficacy-of-the-selena-system.
10. Wong DCS, Kiew G, Jeon S, Ting D. Singapore eye lesions analyzer (SELENA): the deep
learning system for retinal diseases. In: Grzybowski A, editor. Artificial intelligence in oph-
thalmology. Cham: Springer International Publishing; 2021. p. 177–85.
11. Doğan ME, Bilgin AB, Sari R, Bulut M, Akar Y, Aydemir M. Head to head comparison of diag-
nostic performance of three non-mydriatic cameras for diabetic retinopathy screening with
artificial intelligence. Eye. 2024;38:1694–701.
12. Tham Y-C, Anees A, Zhang L, Goh JHL, Rim TH, Nusinovici S, et al. Referral for disease-
related visual impairment using retinal photograph-based deep learning: a proof-of-concept,
model development study. Lancet Digit Health. 2021;3(1):e29–40.
13. Tham Y-C, Goh JHL, Anees A, Lei X, Rim TH, Chee M-L, et al. Detecting visually significant
cataract using retinal photograph-based deep learning. Nat Aging. 2022;2(3):264–71.
14. Wu X, Huang Y, Liu Z, Lai W, Long E, Zhang K, et al. Universal artificial intelligence platform
for collaborative management of cataracts. Br J Ophthalmol. 2019;103(11):1553.
15. Ting DSW, Cheung CY-L, Lim G, Tan GSW, Quang ND, Gan A, et al. Development and vali-
dation of a deep learning system for diabetic retinopathy and related eye diseases using retinal
images from multiethnic populations with diabetes. JAMA. 2017;318(22):2211–23.
16. Son J, Shin JY, Kim HD, Jung KH, Park KH, Park SJ. Development and validation of
deep learning models for screening multiple abnormal findings in retinal fundus images.
Ophthalmology. 2020;127(1):85–94.
17. Cen L-P, Ji J, Lin J-W, Ju S-T, Lin H-J, Li T-P, et al. Automatic detection of 39 fundus dis-
eases and conditions in retinal photographs using deep neural networks. Nat Commun.
2021;12(1):4828.
18. Cao S, Zhang R, Jiang A, Kuerban M, Wumaier A, Wu J, et al. Application effect of an artificial
intelligence-based fundus screening system: evaluation in a clinical setting and population
screening. Biomed Eng Online. 2023;22(1):38.
148 S. Majithia and S. Thakur