Journal Pre-Proof: American Journal of Ophthalmology
Journal Pre-Proof: American Journal of Ophthalmology
PII: S0002-9394(25)00276-4
DOI: https://doi.org/10.1016/j.ajo.2025.05.040
Reference: AJOPHT 13398
Please cite this article as: Fiona Stapleton , Pablo Argüeso , Penny Asbell , Dimitri Azar ,
Charles Bosworth , Wei Chen , Joseph Ciolino , Jennifer P. Craig , Juana Gallar , Anat Galor ,
José A.P. Gomes , Isabelle Jalbert , Ying Jie , Lyndon Jones , Kenji Konomi , Yang Liu ,
Jesus Merayo-Lloves , Fabiola R. Oliveira , Victor A. Perez Quinones , Eduardo M. Rocha ,
Benjamin D. Sullivan , David A. Sullivan , Jelle Vehof , Susan Vitale , Mark Willcox ,
James Wolffsohn , Murat Dogru , TFOS DEWS III Digest Report, American Journal of Ophthal-
mology (2025), doi: https://doi.org/10.1016/j.ajo.2025.05.040
This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition
of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of
record. This version will undergo additional copyediting, typesetting and review before it is published
in its final form, but we are providing this version to give early visibility of the article. Please note that,
during the production process, errors may be discovered which could affect the content, and all legal
disclaimers that apply to the journal pertain.
1
17. Instituto Universitario Fernandez-Vega, Universidad de Oviedo, Principality
of Asturias, Spain
18. Department of Clinical Medicine, Division of Rheumatology and Clinical
Immunology, Ribeirao Preto Medical School, University of Sao Paulo,
Ribeirao Preto, SP, Brazil
19. Department of Ophthalmology, University of Miami Miller School of Medicine,
Miami, FL, USA
20. Department of Ophthalmology, Otorhinolaryngology and Head & Neck
Surgery, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao
Preto, SP, , Brazil
21. Lµbris BioPharma, LLC, Naples, FL, USA
22. Bausch & Lomb, Bridgewater, NJ, USA
23. Schepens Eye Research Institute, Department of Ophthalmology, Harvard
Medical School, and Massachusetts Eye and Ear, Boston, MA, USA
24. Departments of Ophthalmology and Epidemiology, University of Groningen,
University Medical Center Groningen, Groningen, the Netherlands
25. Department of Ophthalmology, Vestfold Hospital Trust, Tønsberg, Norway
26. Division of Epidemiology and Clinical Applications, National Eye Institute,
National Institutes of Health, Bethesda, MD, USA
27. College of Health & Life Sciences, School of Optometry, Aston University,
Birmingham, UK
28. Ichikawa General Hospital Tokyo Dental College Dept of Ophthalmology
Ichikawa, Chiba, Japan
2
Table of contents
1. Abstract ........................................................................................................... 9
2. Keywords ........................................................................................................ 9
3. Abbreviations .................................................................................................. 9
4. Introduction ................................................................................................... 10
5.7 Thyroid hormone regulation of the ocular surface and adnexa .......... 18
6. Epidemiology ................................................................................................ 22
3
6.3 Prevalence of DED ................................................................................... 26
6.3.1 Prevalence of DED based on the Women’s Health Study criteria ........... 26
6.3.2 Prevalence of symptomatic DED ............................................................ 26
6.3.3 Prevalence of DED based on signs and symptoms ................................ 27
6.3.4 Prevalence of DED based on TFOS DEWS II criteria ............................. 27
6.3.5 Prevalence based on claims data ........................................................... 27
6.3.6 Prevalence of DED based on clinical diagnosis ...................................... 27
6.3.7 Prevalence of any MGD .......................................................................... 27
6.3.8 Prevalence of clinically significant MGD ................................................. 27
7. Pathophysiology ........................................................................................... 43
7.3 Hyperosmolarity....................................................................................... 46
4
7.8 Immune cell dysfunction ......................................................................... 53
8. Tear Film........................................................................................................ 54
5
9.8 Brain imaging in DED .............................................................................. 85
6
10.5.2.2 Recommendations for management ............................................... 104
10.5.3 Other procedures ............................................................................... 105
10.5.3.1 Eye cosmetics and beauty treatments ............................................ 105
11.5 Design features to enhance clinical trial data quality and decisions 133
7
11.5.1 Biomarkers / proof of mechanism / early signal of efficacy ................ 133
11.5.2 Missing data considerations ............................................................... 134
11.5.3 Global trial designs ............................................................................ 134
11.5.4 Compliance monitoring ...................................................................... 135
8
1. Abstract
This digest summarises the interdisciplinary research in dry eye disease (DED)
published since the 2017 TFOS DEWS II reports. It comprises seven topics
including Sex, Gender, and Hormones, Epidemiology, Pathophysiology, Tear
Film, Pain and Sensation, Iatrogenic and Clinical Trial Design and explores how
each of these inform diagnostic methodology, disease subtype and management
of DED.
Sex- and gender-related differences significantly influence the ocular surface due
to hormones, sex chromosomes, sex-specific autosomal factors, epigenetics,
care-seeking behaviors, and service utilization. Epidemiological data reveal that
DED prevalence varies by age and sex, influenced by diagnostic criteria and the
multifactorial nature of the disease. New risk factors for DED include
environmental, iatrogenic, systemic diseases and lifestyle domains.
2. Keywords
3. Abbreviations
ADDE Aqueous-deficient dry eye disease
BALB/c BALB/C wildtype (mice)
CXL Corneal cross-linking
DED Dry eye disease
DEQ-5 5-Item Dry Eye Questionnaire
9
DEWS Dry eye workshops
DNA Deoxyribonucleic acid
EDE Evaporative dry eye disease
FDA Food and drug administration
(f)MRI (Functional) magnetic resonance imaging
GVHD Graft versus host disease
HLA Human leukocyte antigen
HPMC Hydroxypropyl methylcellulose
ICAM-1 Intercellular Adhesion Molecule 1
IL Interleukin
IGF Insulin-like growth factor
IVCM In vivo confocal microscopy
LASIK Laser-assisted in situ keratomileusis
LIPCOF Lid-parallel conjunctival folds
LLT Lipid layer thickness
MGD Meibomian gland dysfunction
MGYLS Meibomian glands yielding liquid secretions
MMP Matrix metalloproteinase
MUC4 Mucin 4
NGF Nerve growth factor
NIBUT Non-invasive tear film breakup time
NF-kB Nuclear factor kappa-light-chain-enhancer of activated B cells
NRS Numerical rating scale
OCT Optical coherence tomography
OR Odds ratio
OSDI Ocular Surface Disease Index
PRK Photorefractive keratectomy
QoL Quality of Life
SANDE Symptom Assessment iN Dry Eye questionnaire
SPEED Standard Patient Evaluation of Eye Dryness questionnaire
TBUT Tear film breakup time
TED Thyroid eye disease
TFOS Tear Film & Ocular Surface Society
TRPM8 Transient Receptor Potential cation channel subfamily M member 8
VAS Visual analogue scale
4. Introduction
This review updates the evidence for interdisciplinary aspects of dry eye disease
(DED). It considered novel, human and animal evidence-based research
published between 2017 and 2024. The goal of this report was to identify key
10
research published since the 2017 TFOS DEWS II Workshop reports to underpin
the evidence described in the TFOS DEWS III Diagnostic Methodology 1 and
Management and Therapy 2 reports. The topics include sex, gender, and
hormones, epidemiology including prevalence and risk factors, novel concepts
and findings associated with pathophysiology, relevant changes to the tear film
and interactions, the mechanisms related to ocular pain and sensation, iatrogenic
causes of DED and their unique management and a synthesis of clinical trial
designs to inform exploration of new treatment modalities.
5.1 Introduction
The TFOS DEWS II report on Sex, Gender, and Hormones report 3 addressed
many sex- and gender-related differences that significantly influence the ocular
surface in health and DED. Many of these differences appeared to be due to the
effects of hormones, sex chromosomes, sex-specific autosomal factors,
epigenetics, care-seeking behaviors and service utilization. 3 The purpose of this
section is to highlight some of the relevant research since the publication of that
report. The focus is primarily on studies published after July 1, 2017.
Additional studies related to sexual health and DED may be found in the recent
detailed review, entitled "TFOS Lifestyle: Impact of lifestyle challenges on the
ocular surface". 4
11
have provided critical insights into sex-related immune differences. The extent of
lacrimal gland inflammation in MRL/MpJ-Tnfrsf6lpr mice is, as in humans, far
greater in females as compared with males, whereas the magnitude of lacrimal
gland inflammation is far worse in non-obese diabetic/LtJ males. 7 Results
showed that sex significantly influences the expression of thousands of genes,
and that the immune nature of the glandular response is very dependent on the
Sjögren disease model. Lacrimal tissues of female, as compared with male,
MRL/MpJ-Tnfrsf6lpr mice featured a significant increase in the expression of
genes related to inflammatory responses, antigen processing, and chemokine
pathways. In contrast, it was the lacrimal glands of non-obese diabetic/LtJ males,
and not females, that presented with a significantly greater expression of
immune-related genes. These data suggest that factors in the lacrimal gland
microenvironment may be critically important in mediating these sex-associated
immune effects and in promoting lacrimal gland inflammation. 5
12
The prevalence of meibomian gland dysfunction (MGD) may vary by sex but
results are inconsistent. Population- and hospital-based studies report that MGD
may occur more frequently in males, or in females, or in neither sex. A recent
prospective cross-sectional study indicated that older males had more severe lid
margin abnormities and decreased gland number, height, and area compared
with females. 11 Some studies suggest that the influence of sex on MGD may
depend on the type (e.g. obstructive versus hypersecretory forms, 12 the patient's
age, 13 and/or the individual's medical condition. 14 The inconsistency in the sex-
related prevalence of MGD is notable, given that DED is more common in
females 15-24 and that MGD is a major cause of DED. 6,15,25,26 The prevalence of
any MGD and clinically significant MGD are separately reported in a meta-
analysis by age and sex (see Section 5, Figure 3 F and G). In brief, any MGD
including asymptomatic gland changes is more prevalent in men than women in
older age groups but sex-differences in clinically significant MGD are equivocal.
There remains a need to explore the impact of more detailed diagnostic criteria to
understand sex-related effects on MGD.
5.2.3 Cornea
Significant, sex-related differences exist in corneal thickness, sensitivity, re-
epithelization and DED-induced damage. As has been found previously, males
have greater corneal epithelial thickness in all but the peripheral nasal zone 27
and females have higher corneal sensitivity, 28 and corneal nerve regeneration 29
and slower corneal epithelial wound healing. 30 In addition, aqueous-deficient
DED elicits more ocular pain, anxiety and severe corneal damage in female mice.
31
13
Since that report was published, almost 6,000 articles have been cited in
PubMed addressing the phrase "sex differences and immune." These have
continued to show that sex as a biological factor significantly influences the
distribution of lymphocyte subsets, quality of T cell responses, development of
regulatory T cells, formation of the germinal centers, and the epigenetic
accessibility of B cell loci. 33,34 Sex also impacts transcriptional differences that
are often highly immune cell-specific. 35 For example, more than 50 monocyte
transcripts linked to the interferon pathway, inflammatory cytokines and
chemokines display sex-associated expression, that are prominent in some
female subjects. 35 In addition, changes in sex steroid hormone concentrations
over the course of one's life contribute to sex-related differences in immune
profiles and disease susceptibility patterns. 36
The intersection of sex and pain has been widely studied at clinical,
psychological, and social levels. However, pain research often conflates sex and
gender, and little is known about how gender identity diversity, such as gender-
affirming medical procedures (hormonal or surgical therapies), environmental
exposures, and minority status, impact pain. 38
Despite chronic pain and DED being more common in women, most pain
mechanism studies are based on male rodents. 39 With the inclusion of sex as a
biological variable in preclinical research, studies on the influence of sex on pain
and analgesia have increased. Female sex and older age are still the main
factors associated with chronic pain. Experimental pain response differences
suggest a biological mechanism rather than sociocultural gender-related issues.
40
5.5.1 Androgens
As detailed in the TFOS DEWS II report, 3 androgens are extremely important in
the regulation of the ocular surface and adnexa and appear to mediate many of
the sex-related differences in these tissues. Androgen deficiency, in turn, is
associated with, and a risk factor for, both aqueous-deficient and evaporative
DED. Recent studies are discussed below.
Anti-androgen therapy for the treatment of prostate cancer and benign prostate
hyperplasia led to a significant increase in the signs and symptoms of MGD and
DED. 46,47 Transdermal androgen therapy applied to the lower abdomen, in turn,
alleviated DED signs and symptoms in androgen-deficient individuals, 48 as well
as DED signs in rabbits with combined androgen deficiency and MGD. 49
Androgenetic alopecia was associated with decreased tear film breakup times,
increased meiboscores, ocular surface symptoms and MGD. 50 However, this
15
form of hair loss is not necessarily linked to androgen excess, but is associated
with an increased risk of polycystic ovary syndrome. 51
5.5.2 Estrogens
It has recently been reported that estrogen receptor-1 deficiency in mice induces
inflammation and lipid deposition in the meibomian gland and lacrimal gland. 52
Estrogen receptor-1 loss however, does not block estrogen receptor-activity, but
results in an abnormal endocrine environment (e.g. increased luteinizing
hormone, estradiol, testosterone, and progesterone levels) and may lead to
insulin resistance. 53-55
Ovariectomy of monkeys 56 and rats 57 engenders the signs of DED, and that
these can be reversed with estrogen administration. 57 Other studies show that
ovariectomy leads to rat anxiety and depressive-like behavior 58 conditions that
promote DED, 4, and that estrogen treatment may amplify rat ocular hyperalgesia.
59
Some of these disparate findings regarding the role of estrogens might be
explained by differences in experimental design, hormone dosage or animal
model. 60 However, complete estrogen absence does not cause lacrimal gland
inflammation, gross alterations in meibomian gland histology, or aqueous-
deficient DED in mice, and does not play a major role in the sex-related
differences of the mouse meibomian gland. 60,61
However, is it possible that this animal research does not reflect the situation in
humans? This question is prompted by the results from aromatase inhibitor
studies, which report an increased prevalence of DED. 62-65 Aromatase inhibitors
block the synthesis of estrogens, induce estrogen deficiency, and are used as
therapy in women with hormone receptor-positive breast cancer post initial
chemotherapy. 66 However, these studies had no control groups. Treatment with
aromatase inhibitors is known to promote anxiety (> 112 PubMed articles),
depression (> 243 PubMed articles), and sleep disturbance (> 25 PubMed
articles) (e.g., 67,68, all of which are associated with DED. 4 It may be that these
mental health factors and sleep disorders contribute to, and possibly account for,
these DED effects seen in those treated with aromatase inhibitors.
Investigators have also suggested that the increased estrogen levels following in
vitro fertilization are responsible for the in vitro fertilization-associated DED
symptoms 69 and/or signs. 69,70 However, there were no controls with these
studies, and the effects may also have been linked to the known in vitro
fertilization-induced anxiety, depression and sleep disturbance. 71
16
Clinicians continue to test whether topical estradiol might serve as a DED
treatment. One of the most recent clinical trials showed no significant differences
in effects between any of the estrogen dosages and the placebo. 72
Lastly, estradiol may inhibit the conjunctival goblet cell response in vitro to an
inflammatory stimulus. 73 This effect may be dose-dependent, given that the
estrogen concentration used (0.1 µM) was considerably higher than the
physiological range. Very high doses of estrogen often suppress, whereas
physiological doses often enhance, immune responses. 74
5.5.3 Progestins
Progesterone appears to suppress ocular pain and discomfort, which are
common features of DED. Within 30 minutes after application to the rat forehead,
1% progesterone gel (i.e. 10 mg/mL) produced corneal antinociception. 75
Forehead application of the same dose, twice per day, for ten weeks led to a
significant decrease in the frequency and severity of ocular symptoms in ocular
graft-versus-host patients. 76 Researchers speculated that this hormone effect
may be mediated by the V1 branch of the trigeminal nerve that innervates the
forehead skin, and that progesterone modifies the signal relay in the rostral and
caudal trigeminal nucleus to dampen nociception. 76 However, given that the
applied progesterone concentration in these studies was so much higher than
that typically found in blood (e.g. ng/ml levels), it might be possible that hormone
action also involved other receptors. Progestins are known to bind glucocorticoid,
androgen and mineralocorticoid receptors, as well membrane receptors, oxytocin
receptors and γ-aminobutyric acid (GABAA). 77,78
17
role in delivering IGF to target cells and works independently of IGF. IGF binding
protein-3 levels decrease in response to hyperosmolarity, a marker of DED,
potentially causing epithelial damage in DED. 84
Additional research has linked the DED in diabetes mellitus to insulin impairment.
For example, elevated serum opioid growth factor levels in diabetes are
associated with DED and corneal damage, which can be mitigated by controlling
glucose levels with insulin or opioid receptor antagonists. 88 Topical insulin
therapy, in turn, has shown promise for treating the signs and symptoms of DED.
2,89-91
For insulin topical therapy to be widely adopted for DED, questions about the
optimal concentration, vehicle, and suitable DED subgroups need to be
addressed. 92 Novel formulations using nanotechnology to improve insulin
permeability and exposure to the ocular surface have shown promise in
experimental models. 93,94
The association between TED and DED is debated. A study in the USA found no
link between TED and DED. 96 However, studies in India, Saudi Arabia, Spain,
Taiwan, and Russia found TED to be a risk factor for DED. 97-100 The variable
clinical manifestations of TED (Figure 1) and different definitions of DED likely
contribute to these discrepancies.
18
Figure 1. Thyroid eye disease
The mechanisms that cause DED in TED are broad and include anatomical
changes in the orbit leading to proptosis and excessive corneal exposure, as well
as inflammation of the ocular adnexa, including the main lacrimal gland and the
eyelids. 101 Animal models deprived of thyroid hormone have shown clinical
responses consistent with DED. As an example, rats made hypothyroid with
methimazole developed higher tear film osmolarity and hypoesthesis. 102
In TED, changes in the lacrimal gland have been observed via magnetic
resonance imaging, with smaller lacrimal glands correlating with more severe
clinical signs and higher inflammatory indices. 103 A study on the overlap of
Hashimoto’s thyroiditis and Sjögren disease found higher expression of four
genes involved in both diseases, which may have diagnostic value. 104 These
observations highlight the role of anatomical changes, genetic factors, and
molecular inflammation mediators in linking TED, thyroid hormone dysfunction,
and DED.
Recent studies have confirmed higher Ocular Surface Disease Index (OSDI)
scores, lower tear TBUT, and MGD in TED patients, compared to healthy
19
controls. These symptoms and signs were correlated with worse proptosis,
higher Clinical Activity Scores, incomplete blinking, as well as corneal damage.
100,105-108
Changes were also observed in euthyroid or inactive TED, indicating
mixed inflammatory and anatomical factors. 109,110 Superior limbic
keratoconjunctivitis was observed more frequently in TED patients (31%) and
was associated with worse ocular surface conditions, younger age, and smoking.
111
Worse Schirmer test results, TBUT, and goblet cell density were found in
children with Hashimoto’s thyroiditis compared to healthy children, even without
symptoms. 112 In a study of 38 individuals with moderate to severe TED in Iran,
over 70% had DED. 113
As reported in the Sex, Gender, and Hormones report of TFOS DEWS II, 3
"gender" refers to an individual's self-representation as a man or woman, and
how social institutions respond to that person based on the person's gender
presentation. 3 Gender is not the same as "sex." Sex distinguishes males and
females based upon their biological characteristics, whereas gender reflects
socially constructed characteristics such as behaviors related to being a woman,
feminine, or being a man, masculine. 3 Both sex and gender affect health and
disease, and gender also affects people's access to and interactions with the
healthcare system. 3 Many health disparities are associated with gender, and
both gender and biological sex influence DED risk and presentation, care-
seeking behaviors, and service utilization.
20
and two others were diagnosed with de novo DED due to low tear film TBUT and
punctate keratitis. 118
21
5.9 Future directions
There have been significant research advances linking sex, hormones and
gender to DED. Aging, cancer and hormone therapy increasingly broaden the
interdisciplinarity in this field over time. Despite the significant impact of gender-
affirming hormone therapy on the entire endocrine system and its effects on
physical and mental health, there is limited information on its impact on ocular
health. Variations in age, health profile, gender-affirming hormone therapy
compliance, and barriers to accessing regular healthcare limit the documentation
of side effects. Clinicians and future research should consider these variations,
as recommended in a recent systematic review on the medical aspects of the
transgender and gender diverse population. 123
6. Epidemiology
As per the TFOS DEWS II report, 129 the subcommittee examined data from a
large range of cohort studies and considered different methods of disease
ascertainment and definition, including studies involving the type, frequency and
severity of symptoms, patient self-report of a diagnosis of DED by an eyecare
practitioner, and studies that involved a clinical examination.
22
Eligible studies included those reporting prevalence of either or both dry eye
symptoms and signs. Observational studies (cross-sectional or cohort) were
included if they were population-based and presented the study outcome as DED
versus non-DED. Studies were excluded if no variance in the measure of
prevalence was available in the manuscript, if it was not possible to calculate it
from the data presented, if the sampling criteria were not explicitly stated or if no
denominator was reported. Detailed exclusion criteria are described in Figure 2.
Article author and date, setting (region; population or hospital), numbers and
characteristics of participants within each study group (age, sex, ethnicity),
prevalence, and incidence data were extracted from each article. When required,
data were extracted from manuscript figures using open-source software
(available at https://plotdigitizer.com/app). 130 Where multiple studies were
published from the same dataset, those with minimal overlap, distinctly different
diagnostic criteria and age/sex disaggregation were preferentially chosen.
23
6. Clinical diagnosis or prior diagnosis of DED
7. Any MGD reported and separately, clinically significant (grade 2 and
above, as reported) MGD
A random effects model was used to combine prevalence data. To compute the
standard error, the formula SE = √p*q/n, where p was the proportion with DED
was used. For studies where prevalence was 0, the exact Poisson confidence
limits for the proportion computed and the width of that interval divided by 3.92 to
approximate the standard error.
For descriptive analyses, box plots were produced using SAS version 9.5 (SAS
Institute, Cary, North Carolina, USA). For most diagnostic categories, the number
of studies available for each age category was small. When there was only a
single study in the category, the prevalence is reported as originally reported by
the study. If there were two or more studies in the category, a pooled rate was
computed using a suite of SAS macros models developed by Weir and Senn.
(Senn et al., 2011) These macros include summary statistics for the DerSimonian
and Laird. (DerSimonian & Laird, 1986) The Weir and Senn macro forest was
used to produce forest plots of the results.
The flowchart describes the studies identified since 2015 and reasons for
exclusion (Figure 2). A total of 2687 articles were identified through PubMed
search (updated 29 June 2024). The extracted data summary is available here
(Supplementary Table 1. Studies included in the meta-analysis). For the overall
prevalence estimation and meta-analysis, the final dataset combined both 2015
and 2024 results. Subsequently meta-analyses were conducted by age and sex
by decade where data were available.
Estimates of the prevalence of DED from 76 large international cohort studies (24
and 52 from the 2015 and 2024 datasets, respectively) are summarized in Figure
2 and below. Supplementary Table 1 describes the characteristics of the
additional included studies from the 2024 dataset.
24
Figure 2. Prisma flowchart of the literature search outcome for prevalence
studies published between 18 September 2015 and 29 June 2024.
Table 2. Prevalence range for DED for each of the diagnostic criteria
25
*Noting the lower end of the range relates to the rates in children. See Figure 3 below for
prevalence disaggregated by age
Figure 3. Prevalence of DED based on age and sex for different diagnostic
criteria
Figure 3, panel A represents prevalence by age and sex based on report of severe
symptoms and/or a diagnosis of DED by a practitioner (n=12). 132-143 The prevalence
ranges from 2.7% in those 20-29 to 30.1% in women over 80. The rate increases by
age particularly after the age of 40 in both sexes and women have a higher rate of DED
above the age of 50, with the sex difference in prevalence becoming more marked with
age. One study has shown high prevalence rates in those 10-19 years old without sex
related differences. (See supplementary Table 1 for individual study data).
Figure 3, panel B represents prevalence of symptomatic DED by age and sex. The
prevalence ranges from 7.3% in those 6-9 years old to 31.6% in females aged 20-29
years, however confidence intervals are wide (n=33). 133-135,139-142,144-168 There were
higher than expected rates of disease in the age groups under 30, except a low rate in
children aged 6-9 years. Rates were consistent in the adult groups aged 40-80 years,
with rates in men in the order of 20% and women 30%. Higher rates were observed in
the over 80s age group and in all ages above the age of 10, women had a consistently
higher prevalence.
26
6.3.3 Prevalence of DED based on signs and symptoms
Figure 3, panel C represents prevalence of DED based on the presence of signs and
symptoms by age and sex (n=10). 97,132,133,142,146,152,158,162,163,165,169 The prevalence
ranges from 4.7% in those 6-9 years old to 62.9% in females aged 20-29 years,
however confidence intervals are wide. The rates of disease are reasonably consistent
across the adult age groups with some reduction in symptomatic disease above the age
of 70 in both sexes. Sex differences were not pronounced in most age groups except for
in those aged over 70 where women have a higher rate than men.
The overall prevalence is broadly similar to diagnoses based on signs and symptoms of
DED (n=3) 132,133,170 with prevalence ranging from 5.4 (in 6-9 year olds) to 44.2%. Above
the age of 30, the prevalence is higher amongst females than males, and males show a
more obvious age-related change.
Overall prevalence based on claims, either identifying DED based on the International
Classification of Disease code or using insurance claims data based on diagnosis or
treatment code was generally low (n=1) 171, ranging from 2.8-8.5%. There was
insufficient data to disaggregate prevalence by age and/or sex.
Figure 3, panel D represents prevalence of DED based clinical diagnosis by age and
sex (n=8). 135,140,148,149,151,155,172,173 The prevalence ranges from 1.0% in men over 80, to
15.3% in women aged 50-59 years. Rates were reasonably consistent over age in
adults with lower rates in those 10-15 and over 80 years of age. Women had a higher
rate of clinically diagnosed DED at all ages.
Figure 3, panel E represents prevalence of any MGD by age and sex (n=6). 97,132,174-177
The point estimates of prevalence ranges from 0% in those under 20 years old to 66.3%
men over 80. Rates appear to markedly increase above the age of 40 and in 40 plus
age groups, MGD is significantly more prevalent in older men aged 70 and above than
in older women (p<0.05). Confidence intervals in most age groups are wide.
27
Figure 3, panel F represents prevalence of clinically significant MGD (Grade 2 or above)
by age and sex (n=2). 165,175 Rates appear to increase with age although sex differences
are equivocal. There are no studies reporting rates of clinically significant MGD in
younger individuals.
Thirty-three of 52 studies included in the systematic review from the 2024 dataset did
not contribute to the meta-analysis due to non-availability of by age decade or by sex
data, or due to duplicate data. 20,23,97,140,141,161,162,164,166,167,169,177-198 The overall
prevalence rates reported include all international cohort studies (24 and 52 from the
2015 and 2024 datasets, respectively). Meta-analysis includes only those studies
publishing disaggregated age and sex data. Supplementary Table 1 shows the
complete list of studies and data extraction for those identified in the recent dataset. The
2015 dataset is included as supplementary data previously. 129
A limited number of population studies have attempted to assess the incidence of DED
since the last TFOS DEWS II update. In a retrospective analysis of approximately 6.7
million medical claims from the United States Department of Defense Military Health
System, the annual DED incidence in those aged 2 years and above was low but to
gradually increase over time from 0.55% in 2008 to 0.87% in 2012, respectively. 171
DED annual incidence was consistently higher in women than men and increased with
age. 171
An ancillary cohort study of the Vitamin D and Omega-3 Trial (VITAL), which assessed
the incidence of DED in 12,174 men aged 50 years and older and 11 349 women aged
55 years and older during a median (range) 5.3 (3.8-6.1) years of follow-up in the United
States. All participants were initially disease free and 2% of participants experienced a
clinically incident DED. 201 The annualised incidence (average across 5 years) of
clinically diagnosed DED and of clinically diagnosed DED plus incident reports of severe
DED were 3.8 and 16 per thousand, respectively. 201 The Salnés Eye Study 2 re-
examined a cohort of 264 Spanish individuals from the Salnés Eye Study 1 now aged
51 years and older, 11 years later. 202 The annualised incidence of DED signs and
symptoms in these individuals was 2.3% (95% confidence interval 1.8-2.8). 202
28
Participants (n=1,682) from the Singapore Malay Eye Study (SiMES) were re-examined
6-years later, and symptoms of DED were evaluated using the Salisbury Eye Evaluation
Study dry eye questionnaire. 203 The 6-year incidence of DED symptoms was 5.1% (95%
confidence interval 4.1-6.4). 203
The incidence of DED is typically difficult to estimate as some with the disease will
report resolution or a reduction in symptoms over multiple sampling periods. This may
particularly occur for milder cases of DED where symptom report may vary over time.
Incidence from claims data may be influenced by changes in reimbursement or the
introduction of new therapies during the study.
Longitudinal studies of DED are rare. A cohort of 784 patients from 1000 with DED from
the Women’s Health Study and Physicians’ Health Study were surveyed about change
in their disease one year after enrolment in the study and medical records were
obtained for 261 of the participants. 204 The mean disease duration was 10.5 years, and
most participants reported no change to their disease status over time. Ocular surface
symptoms were unchanged in 32% of participants and improved in 44%. Visual
symptoms were unchanged in 52% and improved in 19%. Social impact was
unchanged in 71% and improved in 19%. Risk factors associated with progression of
ocular surface symptoms included a higher spend on treatment (more than $20 per
month), history of more severe DED symptoms and use of systemic beta-blockers.
Worsening of visual symptoms was additionally associated with a history of ocular
surgery, untreated depression and blepharitis or MGD. Disease presentation and
treatments varied by sex where women were more likely to present with corneal staining
but worsening of symptoms was not predicted by corneal staining, however treatments
beyond level 1 205, higher symptoms and having a tear breakup test performed were
associated with progression of symptoms.
In a small retrospective study of DED in the absence of treatment (n=73), 207 changes in
signs were observed with an increased bulbar redness, reduced tear meniscus height
and lipid layer thickness after 8 years. Change in symptoms over time was not reported.
29
In a registry-based study, signs of MGD and evaporative DED were apparent at an
earlier age than aqueous-deficient DED (Wang et al., 2020; Wang et al., 2019),
although there may be some confounding due to ethnic background, where up to 2/3 of
those with signs of MGD do not experience symptoms (non-obvious disease). 175 There
is indirect evidence to suggest that signs of MGD may precede other disease markers
by up to 10 years (Wang et al., 2020), which may have implications for the timing of
treatment of non-obvious MGD. In a small treatment trial of adults with symptomatic
MGD who were treated for 12 months, 1/3 of participants had improvement in MGD
signs and improvement was greater in younger participants (those under 40) and with
less ocular surface damage. 208
Conclusive and probable risk factors for DED grouped into major categories described
in Table 3 and have been divided into modifiable and non-modifiable factors.
C= consistent, P= M= possibly
probable modifiable, N =
non modifiable
1. Systemic disorders
a. Autoimmune disorders
209
Sjögren disease C N
30
209,210
Rheumatoid arthritis C N
209
Systemic sclerosis C N
209,211
Systemic lupus erythematosus C N
209
Sarcoidosis C N
210,212,213
Thyroid disease C N
d. Pain disorders
227
Irritable bowel syndrome C N
227
Fibromyalgia C N
214,228,229
Chronic pelvic pain C N
227,230
Migraine C N
230
Other headache disorders C N
212,214
Osteoarthritis C N
227
Back pain C N
228
Temperomandibular joint disorder P N
e. Psychiatric disorders
210,212,231-233
Depression C M
31
231,232
Anxiety C M
212
Stress (including post-traumatic stress disorder) C M
214
Burnout P M
214
Autism P N
f. Sleep disorders
234
Obstructive sleep apnea syndrome C M
141,235
Insomnia and poor sleep quality C M
g. Other disorders
212
Osteoporosis C M
236
Parkinson’s disease C M
214
Sinusitis P M
2. Ophthalmic disorders
237,238
Meibomian gland dysfunction C M
239-241
Anterior blepharitis C M
226
Allergic conjunctivitis C M
212
Glaucoma C M
214,242,243
Facial nerve paralysis (Bell’s palsy) C N
3. Surgery or procedures
261,262
Eyelid and periorbital surgery C N
261,262
Periocular botulinum toxin injections C N
261-263
Cosmetic periocular and ocular procedures P N
32
4. Medications
5. Environment
283
Air pollution from NO2 and CO P M
283
Low humidity C M
283
High or low temperatures P M
283
Air conditioning and wind P M
212,262,284
Contact lens wear C M
212,285-287
Screen use C M
263
Cosmetics C M
6. Demographic factors
212,288-290
Increasing age C N
212,288-290
Female sex and gender C N
212,291
Asian race / non-white race C N
33
292-294
Vitamin D deficiency P M
292
Omega-3 fatty acids deficiency C M
292,295-297
Altered gut microbiome P M
136,298,299
Genetic predisposition P N
*Consistent evidence implies the existence of multiple adequately powered and otherwise well-conducted
studies published in a peer-reviewed journal, along with the existence of a plausible biological rationale
and corroborating basic research or clinical data . Probable evidence implies the existence of at least one
adequately-powered and otherwise well-conducted study published in a peer-reviewed journal. Risk
factors with mixed or inconclusive results from multiple studies or from a systematic review or meta-
analysis are described in the Diagnostic Methodology Report. Non-modifiable = a lack of evidence that
change will impact DED.
Numerous systemic disorders (Table 3) have been associated with an increased risk of
DED, emphasising the multifactorial origin of the disease.
34
Decreased corneal sensitivity and impaired reflex tear secretion have been suggested
to be linked 308 alongside factors such as hyperglycemia, advanced glycated end
product accumulation, oxidative stress, metabolic disease and vascular disease 309.
Menopause has also been proposed as a risk factor for DED, but definitive evidence is
lacking 129,218,310.
Chronic pain disorders (the body's heightened sensitivity to non-noxious stimuli) can
present with ocular discomfort symptoms similar to those of dry eye.; fibromyalgia,
chronic pelvic pain, irritable bowel syndrome are among the most established risk
factors 227. Development of neuropathic pain within the trigeminal somatosensory
system has been proposed to underlie this link 227. A study of twins in the United
Kingdom found evidence for shared genetic factors underlying these disorders and DED,
suggesting a common chronic pain predisposition that accounts for clustering of pain
disorders 299. Other pain conditions that have been linked to DED are migraine and
other headache types, osteoarthritis, back pain, and temporomandibular joint disorder
214,227,228,314
.
There is also strong evidence for an association between DED and depression, anxiety,
burnout, and stress (including post-traumatic stress disorder) 212,214,227,231-233. A meta-
analysis of studies in patients with DED found an overall prevalence of 40% for
35
depression (OR of 1.8), and of 39% for anxiety (OR of 2.3) 231. These relationships are
likely bidirectional, and studies show an association with psychiatric disorders with DED
symptoms that is greater than with signs. Proposed mechanisms that link psychiatric
disease to DED include altered pain perception, somatization, and increased serum
inflammatory markers 227. Clinicians may find it beneficial to enquire about mental health
history in patients with DED, as appropriate, and consider referrals to mental health
professionals when indicated.
Obstructive sleep apnea has been linked to DED 212,235. It increases the risk of floppy
eyelid syndrome, which may lead to exposure keratopathy at night and to increased
conjunctival inflammation and MGD 315. Leakage of air from a continuous positive
airway pressure (CPAP) mask may also lead to irritation, conjunctivitis and DED 316,317.
Insomnia and poor sleep quality may be a consequence of DED, but could also result in
DED 141,227,235. For example, obstructive sleep apnea is associated with a persistent
low-intensity inflammatory state which may be pertinent to DED etiology 318.
Other disorders that have been convincingly linked with DED are osteoporosis (which
may reflect common underlying mechanisms such as sex hormone or vitamin D
deficiency, or a role of purinergic signalling) 319, autism (which may reflect
hypersensitivity to external stimuli) 214, Parkinson’s disease (which may be linked to
decreased blink rates and lower tear secretion) 236,320 and sinusitis (which may
sometimes involve the tear ducts, or reflect common mechanisms like allergy, or may
represent referred pain) 214.
MGD may be the most important factor associated with DED 237. Its prevalence varies
widely between studies, but has an estimated global prevalence of 35.8%, and the
majority of patients with DED have underlying MGD that is considered to be a major
cause of evaporative DED 237,238,321. Signs of MGD occur earlier in the natural history of
DED progression (typically between 24-29 years) than other clinical signs such as tear
film instability, hyperosmolarity (31-38 years) and ocular surface staining (46-52 years)
322
.
36
Other disorders of the ocular surface have also been associated with a disruption in the
tear film or inflammation at the ocular surface, potentially leading to DED (see Section
X). Conditions that have been most associated with increased risk of DED are
pterygium 160,244,324, pingueculum 245-247, conjunctivochalasis 248-250, and allergic
conjunctivitis (see section 3.1.2.1.1).
Patients with glaucoma are also at increased risk for DED, most often associated with
use of antihypertensive eye drops. Both active ingredients 269 and preservatives can
affect the ocular surface. In addition, glaucoma surgery can affect the ocular surface.
212,325,326
Facial nerve paralysis, including Bell’s palsy, can lead to exposure keratopathy through
incomplete blinking, and has also been associated with impaired lacrimation and MGD
214,242,243
. Facial nerve dysfunction may lead to lagophthalmos and reduced blink force,
compromising tear film distribution and postulated to compromise meibomian gland
expression 327.
Eye surgery has consistently been linked to an increased risk of DED. The greatest
amount of evidence is available for refractive surgery, 256-258,328, cataract surgery 255,259
and intravitreal injections, 260 but it is likely that all eye surgery comes with an increased
risk of DED. 212,214,261,262 Mechanisms that lead to DED after eye surgery include
incisional procedures leading to transectional nerve damage, surgical trauma to the
ocular surface during surgery, phototoxicity, toxicity from eyedrops (e.g. from povidone
iodine, anaesthetics, or preservatives), and damage and stress from repeated drying,
irrigation and exposure to surgical illumination. 261,329,330
Eyelid and periorbital surgery, such as blepharoplasty, ptosis surgery and brow surgery,
have also been associated with an increased risk of DED. 261 Mechanisms include
lacrimal gland injury and post-operative incomplete eyelid closure and incomplete
blinking. Similarly, periocular botulinum toxin injections can lead to lagophthalmos. The
TFOS Lifestyle Report discusses the impact of eye, eyelid and periocular procedures on
the ocular surface in further detail. 261,263
37
6.6.4 Medication use (See Section 10)
Systemic chemotherapeutic and other anticancer agents may have cytotoxic effects at
the ocular surface and affect tear film quality and reflex tear secretion. 262,270-274,331
Examples include alkylating agents such as cyclophosphamide, antimetabolites such as
5-fluorouracil and methotrexate, monoclonal antibodies such as rituximab and the
aromatase inhibitors.
Vitamin A derivatives or retinoic acids, including isotretinoin, are used for acne vulgaris
and in anti-ageing regimes, and may be administered topically or orally. They are
secreted into the tear film by the lacrimal gland 332, and are associated with tear film
instability, lower Schirmer test scores, and atrophy of meibomian glands, leading to
DED. 263,275-277,306,333
Interleukin (IL)3 and 4 receptor antagonists (such as dupilumab) are increasingly used
in patients with atopic dermatitis with good effect but may cause several adverse effects
at the ocular surface including a mild to sometimes severe conjunctivitis, punctate
keratitis, blepharitis, and loss of meibomian glands. 278-280
Hormone replacement therapy has been associated with increased prevalence of DED.
213,218,262
38
Proton pump inhibitors, antacids, and psychostimulant agents used for attention-
deficit/hyperactivity disorder have also been linked to a highly increased odds of having
DED, but biological pathways are currently unclear. 266,267
Climatic risk factors may play an important role in the etiology of DED. The TFOS
Lifestyle Environmental Conditions report has recently and comprehensively
summarized evidence for environmental risk factors for DED. 283 Both low and high
temperatures have been associated with DED, as has low humidity, in several
experimental and population-based studies. Air-conditioning and wind are well-known
risk factors that patients often report as triggering symptoms, 334 although limited
evidence is available from research studies. Air pollution such as from NO 2 and CO was
found to be probably associated with DED, and soil pollution from chromium is likely
associated with DED and Sjögren disease. Evidence for a risk of other air, soil, and
water pollutants was not conclusive. 283
Contact lens wear has been reported as a factor associated with DED in many cross-
sectional population-based studies, but prospective studies are lacking. It has been
postulated that mechanisms leading to DED may include thinning of the tear film after
insertion of the contact lenses, increased friction between the lens and the ocular
surface, leading to meibomian gland dropout, decreased tear film stability, ocular
surface staining and lid wiper epitheliopathy. 238 It is important to recognise that contact
lens discomfort symptoms overlap with DED, and symptoms do not necessarily reflect
underlying DED. 212,262 (See Section 10 Contact lenses and DED)
Screen or visual display terminal use has been consistently linked to DED. 285-287,335 As
few as 1 to 2 hours of screen use per day may even be associated with adverse ocular
surface effects. 285 Important mechanisms include decreased blink rate and incomplete
blinking that leads to increased evaporation and decreased lipid release from
meibomian glands. 336 It is also important to consider digital eye strain, independently of
DED, in the differential diagnosis of persons who are heavily exposed to a digital
environment. 286 This requires a full refractive correction to be determined for the
distances required and a binocular vision assessment to ensure suboptimal visual input
is not the cause.
Cosmetic products may be associated with adverse effects at the ocular surface and
may aggravate or initiate DED symptoms. The TFOS Lifestyle cosmetics report
identified 10 ingredients that are commonly present in cosmetics that particularly have
significant adverse effects: benzalkonium chloride, chlorphenesin, formaldehyde-
releasing compounds, parabens, phenoxyethanol, phthalates, prostaglandin analogues,
retinoids, salicylic acid, and tea tree oil. 263 Finally, recent reviews have found impacts
39
on the ocular surface from: environmental endocrine disruptors in foods, packaging and
pesticides. 337 No clear or consistent link between smoking/vaping and DED has been
identified, although effects on the tear film are evident. 227,338-341 While a direct causal
relationship between smoking and DED may not be certain, it is reasonable to suggest
that smoking cessation should be encouraged as part of a comprehensive approach to
promoting ocular surface and holistic health in patients with DED.
Female sex and gender are strong risk factors for DED (see Section 6). As discussed in
Section 5, sex-related differences in prevalence are underpinned by differences in the
ocular surface and adnexa including anatomy and immunity, a higher prevalence of
autoimmune disorders and psychiatric disease (and their related medications), higher
general pain sensitivity in women and reduced androgen levels (already lower than in
men) after menopause. 218,289,342
Increasing age is also a well-established risk factor for DED, but DED is prevalent in
both the young and the old. Systematic reviews have found that signs show a stronger
relationship with age than symptoms. MGD, also, increases in prevalence with age.
288,321,343-346
Prevalence values for DED and MGD are higher in Asian countries than in Western
countries 289 and a pooled analysis of interethnic differences suggested differences in
disease type and age at which signs manifest in Asian and Caucasian populations. 291
There is also evidence that white races are less affected than non-white races in
Western studies. 212 It is difficult to unravel precise causes for these differences, which
likely reflect socio-economic, cultural, genetic, anatomical, lifestyle and environmental
differences between groups.
Nutrition may play a role in the development of DED, but there is no strong evidence to
support recommendation of an optimal diet. Poor nutrition that drives systemic disorders
may be associated with risk factors for DED. Limited but increasing evidence links
alterations in the gut microbiome to ocular surface health, possibly by altering the
immune system. 292,296,297,347-350 The TFOS Lifestyle Nutrition report concluded that
there is evidence that deficiencies of vitamins A and C and omega 3 fatty acids are risk
factors for DED. Moderate evidence was also found for vitamin B12 and D deficiencies
as risk factors.292 There is no clear evidence that alcohol use, 227,292, a Mediterranean
diet 351 and water intake 292 represent risk or protective factors for DED.
40
Genetic factors likely contribute moderately to DED. A large twin study in the United
Kingdom established heritability accounted for 29% for DED symptoms, and 41% for
DED diagnosis. The remaining 60-70% was attributed to unique environmental factors.
136
A recent genome-wide association study in Taiwan with over 14,000 DED cases and
almost 26,000 controls found eleven independent risk loci, including MUC16, which
encodes for a mucin protein that is expressed at the ocular surface. A polygenic risk
score including 932 loci was able to detect individuals with a high-risk of DED. 298 A
limitation of this study was the use of self-reported DED only and that the findings were
not replicated in an independent cohort. This lack of replication is also a major limitation
for several, mostly small, candidate gene studies that have found a link between genetic
variations and DED. 352-358 Further studies are warranted before genetic testing
becomes clinically useful in DED.
There is debate about the impact of MGD on risk factors and prevalence of DED. It is
recognised as a risk factor for the disease and recent estimates of DED prevalence
suggest that MGD may be present in 50-70% of cases of DED. 359,360 Nonetheless,
there have been attempts to establish demographic risk factors for MGD which may or
may not be independent of those for DED more broadly. Age is frequently reported as a
risk factor in MGD, (Arita et al., 2019) particularly in elderly populations 188 and where
MGD is diagnosed using meibography to determine meibomian gland loss or drop out.
11,361
Ethnic background (Asian compared with other backgrounds) is consistently
reported as a risk factor. The impact of sex in MGD is however equivocal, in contrast to
its impact in DED more broadly where female sex predominates.
41
For the meta-analysis reported above (Section 6.3.7), large population-based studies
only have been included and two analyses report either all MGD, irrespective of
definition and clinically significant MGD signs with symptoms, broadly based on the
TFOS MGD Report. For population-based studies reporting any lid changes with or
without symptoms (Any MGD – Figure 3, F), MGD is more prevalent in older men
(above 70 years of age) than older women but there is no difference between sexes in
the rates of clinically significant MGD (Figure 3, G).
DED severely affects the lives of sufferers. It negatively impacts quality of life (QoL)
including physical, psychological and emotional well-being, social functioning, daily
living activities and independence. General and mental health, social functioning,
physical, emotional states, bodily pain and vitality are significantly poorer in those with
DED compared with those without 362-365 and that health status worsens in those with
more severe disease. 364,365 In individuals with mild and moderate disease (n =217)
blurred vision, productivity loss, and visits to eye care practitioners were increased
compared with age-matched normal individuals (n=67). 366
Adverse QoL effects appear to be consistent over time, irrespective of geography and
with variations in ethnic background 367,368 and will likely increase with population aging.
The economic cost of DED can be measured in direct resource utilisation (service
provision, medication costs), out-of-pocket costs, cost of lost productivity and reduced
QoL (quantified as a utility). There are limited studies which include an associated utility
algorithm or that meaningfully evaluate productivity impacts of DED.369-371
This update has considered the prevalence of DED in studies that have shown disease
rates by age and sex. Eight major diagnostic groups were identified and meta-analyses
reported here include DED diagnosed using the Womens Health Study criteria,
symptom report; claims data from health or insurance databases; signs and symptoms;
diagnosis according to the TFOS DEWS II criteria; clinical diagnosis; any MGD or
clinically significant MGD (grade 2 or above). Prevalence varied with diagnostic criteria
where not all disease increased with age or showed a female predominance. Broadly,
using the Womens Health Study criteria, DED increased with age and was more
common in women. A clinical diagnosis of DED showed a female preponderance but
not an age-related effect. Symptoms and signs were more common in women with
higher rates in younger and older adults. Any or severe MGD was age-related, with
males more likely to show any MGD. Some studies included in the ‘Any MGD’ analysis
reported non-obvious MGD and the age and sex effects here have not been stratified.
42
These findings are perhaps not unexpected given the multifactorial nature of DED, the
specificity of ocular symptom measurements and differences in the etiology of different
subtypes of DED 1. Prevalence data in some recent studies may be confounded by the
impact of the COVID-19 pandemic. Previous meta-analysis showed that both the
pandemic and mitigating factors (mask and screen use) were associated with greater
ocular symptoms and signs of ocular surface disease. 26,145,191,197
Studies reporting rates of DED in those under 20 are limited. Rates are lower than
adults for clinically diagnosed DED, DED with signs and symptoms (under 10 years)
and any or significant MGD. High rates of symptom-reporting are evident in those under
20 however, although it is recognised that symptom report alone is not specific for DED
and childhood anterior or posterior blepharitis, Demodex and allergy may be common
co-morbidities or contributors to ocular symptoms. Most studies reporting symptoms did
not report signs although they may be present.
Risk factors are reported as consistent or probable, and potentially modifiable or non-
modifiable. New risk factors related to environment, climate and lifestyle are included.
There is some evidence for an increase in prevalence in DED over time which may
conceivably be due to the impact of new risk factors including changes in the digital
environment. Given the differences in prevalence and age/sex associations with
different diagnostic criteria for DED, it may be important to disaggregate risk factors
particularly for DED and MGD where possible. Given the high prevalence of symptom-
reporting in childhood, appropriate triaging for other conditions and hypothesis-driven
and appropriately powered studies to explore risk factors in children would be valuable.
1. Disease severity. A limited number of studies explored the prevalence, risk factors or
natural history by disease severity, which could help to triage and manage those
more likely to experience more severe DED.
2. Geographical mapping was not considered as part of this update.
3. The generalizability of prevalence measures for DED in children and adults under 40.
These mostly originate from studies in Asia and there are limited studies in other
regions.
4. The need for appropriately powered studies to determine risk factors in those under
40.
7. Pathophysiology
7.1 Introduction
43
The consensus view of the 2017 TFOS DEWS II report envisioned a tear film centric
model of the pathophysiology of DED, 6 broadly classified by compromised quantity;
Aqueous-Deficient Dry Eye (ADDE) or quality; Evaporative Dry Eye (EDE) of the tear
film. In DED, tear hyperosmolarity is considered to set up a cascade of signaling events
within surface epithelial cells, that leads to the release of inflammatory mediators and
proteases. Such mediators, together with the tear hyperosmolarity itself, are conceived
to cause goblet cell and epithelial cell loss and damage to the epithelial glycocalyx.
Damage is reinforced by inflammatory mediators from activated T-cells, recruited to the
ocular surface. The net result is the characteristic punctate epitheliopathy of DED and a
tear film instability which leads at some point to early tear film break-up. This break-up
exacerbates and amplifies tear hyperosmolarity and completes the vicious circle events
that ultimately lead to ocular surface damage and self-perpetuation of the disease.
Epithelial injury and a defective glycocalyx, loss of tear volume and of goblet cell mucin,
lead to increased frictional damage and friction-related symptoms. The tear
hyperosmolarity and epithelial injury caused by DED, stimulates corneal nerve endings,
leading to symptoms of discomfort, increased blink rate and potentially, a compensatory,
reflex increase in lacrimal tear secretion.” The TFOS DEWS II report also highlighted
causes of ADDED that included lacrimal gland infiltration and dysfunction,
neurosecretory or reflex blocks, androgen deficiency or aging-related downregulation of
secretion, obstruction of the lacrimal ducts in cicatricial disease, and iatrogenic causes
such as prescription medication and surgical damage to trigeminal nerves. For EDED,
conditions that affected the ocular surface included MGD, anterior blepharitis,
xerophthalmia, ocular allergy, androgen deficiency as well as iatrogenic causes such as
topical preservative use, contact lens wear and certain anti-glaucoma drugs.
Subsequent research has led to a more nuanced understanding of the disease. Given
the inconsistent relationship between symptoms of DED and signs such as measurable
inflammation, hyperosmolarity, ocular surface staining, low tear volume, low TBUT, or
MGD, 360,372 the data suggest that the associated signs help identify subtypes of the
disease, with symptoms providing no predictive ability as to which etiology is active in
that patient. 360 The implications of the lack of correlation of signs and symptoms in DED
are particularly important in relation to pathophysiology, because while most historical
literature assumes that inflammation and inflammatory pathways are the common
effector of DED, evaporative subsets show a muted if any increase in inflammatory
mediators in the tear film, 372-376 and thus inflammation cannot be assumed to be active
in every patient with DED. For example, one study based on mass spectrometry found
that evaporative patients exhibited no increase in proteins associated with the
inflammatory response compared to normal controls, while 51% of differentially
upregulated proteins in the aqueous cohort were associated with inflammation. 372
Similar data showed that while aqueous- deficient subjects (Schirmer value ≤ 5 mm)
44
had elevated lipid peroxides in tears compared to healthy controls, subjects with 6–10
mm wetting were not significantly different from normal controls. 373 Another study
based on multiplex bead analysis found that evaporative patients tend to exhibit much
higher levels of epidermal growth factor, but no difference in the IL-6 and IL-8 cytokines
compared to healthy controls and essentially no detectable TNF-α in tears, 375
reinforcing the idea that not all forms of DED are driven by the same underlying
pathways. In mouse models, bilateral lacrimal gland excision showed dramatic
increases in IL-1ß, IL-6 and TNF-α protein concentration in the tear film over four weeks,
while topical benzalkonium chloride and environmental chamber-induced DED showed
no significant difference from controls in those tear film cytokine proteins, despite
concurrent increases in tear osmolarity and cytokines measured from corneal mRNA
transcripts. 377 Immune-mediated etiologies, represented by immunological diseases
that include Sjögren disease, Stevens-Johnson’s syndrome, and ocular graft versus
host disease (GVHD), are widely regarded as being more severe than evaporative
counterparts. 378 At the extreme end, severe forms of dry eye found in ocular GVHD
tend to be accompanied by fibrotic processes, 379,380 and Sjögren-related DED is
associated with lymphocytic infiltration of the lacrimal gland, neither of which is evident
in common evaporative DED 380 In a rabbit model, cauterization of the meibomian
glands results in only moderate increases in hyperosmolarity and loss of goblet cells as
compared to models in which lacrimal excretory ducts and accessory glands of rabbits
are sealed. 381 These data may help explain why anti-inflammatory medications have a
dichotomous effect, with some patients reporting improvement, 382,383 while others report
high failure rates of cyclosporine and lifitegrast in the general population. 384-387 As many
patients are still poorly served by the available therapeutic options, it may be that our
understanding of the pathophysiology of DED is incomplete.
Note that a variety of the pathophysiological aspects of the disease are discussed in
more detail elsewhere. For example, androgen deficiency is discussed in the Sex,
Gender, and Hormones section, neuropathic damage is covered in the Pain and
Sensation section, the tear proteome is outlined in the Tear Film section and iatrogenic
causes are described in detail in the TFOS Lifestyle Report (Gomes et al., 2023) and in
the Iatrogenic section of the digest report.
Except in certain situations of injury, 176 surgery 256,328 and therapy 388, establishing
causality is difficult when diagnosing DED. Metabolic disease induces mitochondrial
stress 389-394 and advanced glycation end products in the lacrimal gland 395,396, hormonal
changes that alter glandular production and fatty acid metabolism 3,397-400, and
biophysical stresses of friction 401-405, hyperosmolarity 406-414, and swelling pressure 415.
These effects are compounded by the biological activity of dysregulated or self-reactive
45
immune cells 416-419, cellular stresses from cytokines 377,420-426, proteases 427-429, reactive
aldehyde species430, extracellular deoxyribonucleic acid (DNA) and neutrophil
extracellular traps 431-433, exogenous toxins 390,411, damage or danger-associated
molecular patterns 408, gut dysbiosis 434,435, as well as neurogenic inflammation. 436-441
Typical pathways of initiation of evaporative processes include phenotypic alterations in
corneal epithelial cells that lead to a compromised glycocalyx 442, keratinization of the
meibomian gland that alters the lipid profile of the tear film, 443,444 and incomplete
blinking or reduced blink rate during screen use that exposes the ocular surface to
desiccating stress. 445 On the aqueous deficiency side, inflammatory ingress into the
lacrimal gland can be driven by androgen deficiency or autoimmunity, causing a
cascade of protease release, cytokine expression, inflammatory cell recruitment,
dendritic cell maturation, and an adaptive T-cell mediated response. 3,418,446 As disease
severity increases, the evidence suggests there is a progressive accumulation of these
mechanisms. 227,418,447,448
7.3 Hyperosmolarity
In addition to the hypothesis that inflammatory cells infiltrating either the lacrimal gland
or conjunctiva are a source of oxidative damage at the ocular surface, 458,459 In vitro
evidence suggests that hyperosmolarity directly induces reactive oxygen species
(hydroxyl and peroxyl activity) in human corneal epithelial cells, along with reductions in
the anti-oxidant superoxide dismutase-2, vitamin D, Notch ligands Dll3 and Jag 1, and a
tripling of the pro-apoptotic Bax/Bcl2 ratio. 460 Reactive oxygen species undergo lipid
peroxidation and cause reactive aldehyde species to be produced, causing a cascade
of protease release, cytokine expression, inflammatory cell recruitment, dendritic cell
maturation, and an adaptive T-cell mediated response 430,461. In vitro data have shown
that exposure to hyperosmolarity causes mitochondrial DNSA to leak into the cytoplasm
of human corneal epithelial cells, activating the cGAS-STING pathway and increasing
cytokines such as CXCL10 and IFN-β, 462 while human conjunctival impression cytology
46
samples from evaporative, short TBUT patients (2.9 s) confirmed an increase in STING
proteins compared to more normal controls. 463 Evaporative hyperosmolar patients (327
mOsm/L, 7.0 s TBUT, 10.4 mm average Schirmer value) exhibit specific upregulation of
IFN-γ, without concomitant increases in IL-2, IL-6, IL-10, TNF-a or IL-17A. 464 IFN- γ,
which is strongly associated with dose dependent CD40, MICA and MHC II expression
along with esithelial cytotoxicity in corneal epithelial cells, 465-467 increases NLRP3
oxidative stress and contributes to pyroptosis in human corneal epithelial cells under
hypertonic conditions. 468 These data mirror earlier findings that hyperosmolarity
disrupts the balance of oxygenases and antioxidant enzymes such as SOD1 and
PRDX4, stimulates lipid peroxidation (e.g., 4–hydroxynonenal & malondialdehyde),
increases COX2, and damages corneal mitochondrial DNA. 469,470 Hyperosmolarity also
potentiates the negative effects of the oxidizing blue light in corneal and conjunctival
epithelial cells, increasing H2O2 production, phototoxicity and altering the mitochondrial
membrane potential in these cells compared to those in normal media. 471 Downstream
of hyperosmolarity, a TLR-4 dependent upregulation of Dual oxidase 2, a member of the
NADPH oxidase family that regulates the production of intracellular reactive oxygen
species, increases alongside high mobility group box 1 production in human corneal
epithelial cells. 408,472 This result was consistent with data that MyD88-/- mice lacking
TLR-4 signaling exhibited significantly less corneal fluorescein staining, cytokine and
protease expression following 5 days of hyperosmolar stress and scopolamine
administration, 473 implicating TLRs and damage or danger-associated molecular
patterns (e.g., HSPs, HMGB1, S100A, tenascin-C) as essential agents in evaporative,
hyperosmolar DED pathophysiology. 473-475 Although inflammatory ingress may be an
eventual result of hyperosmolar exposure, 476 clinically, it is likely a matter of time and
severity that governs which patients progress to the inflammatory phenotype as a result
of hyperosmolarity. 381 For example, at low levels of hyperosmolarity, protective
responses such as the increase in IGF binding protein-3 are observed, but at increasing
levels of osmolarity and length of exposure, cell viability is challenged as respiration and
glycolysis are decreased alongside endoplasmic reticulum stress and caspase-3
activation, leading to unchecked mitophagy and eventual cell death. 393,477,478 Similarly,
short term exposure to low humidity environments showed an increase in damage or
danger-associated molecular patterns such as HSP-60 and a small 2.4-fold increase in
MMP-9 mRNA expression in patients with evaporative DED, but no statistically
significant increase in IL-6 or IL-8 expression. 474 These data suggest that the severity
of tear hyperosmolarity should help inform the underlying pathophysiology; wherein a
strongly elevated osmolarity will lead to oxidative stress and inflammation, while a
middling or low osmolarity is suggestive of a subclinical inflammatory state 479 and may
mitigate against prescription of anti-inflammatory therapy.
7.4 Proteases
47
Another aspect of the pathophysiology of aqueous deficient disease that separates it
from evaporative disease is the disrupted balance of protease and anti-protease activity
on the ocular surface. The normal tear film exhibits an equilibrium of protease and
protease inhibitors. 427 Excess protease is observed in Sjögren disease, where
cathepsin S catabolizes anti-proteases such as cystatin C, 428 MMP-9 is highly
expressed while thrombospondin-1 lags behind, 480 and plasmin activity is increased
ten-fold in Sjögren compared to normal tears. 481 In non-autoimmune DED, protease
activity is stratified by disease severity and is strongly associated with aqueous
deficiency. For example, mild evaporative-type subjects with low TBUT (5.0–7.2
seconds) and normal Schirmer value (13–19 mm) exhibited only slightly higher tear
MMP-9 activity (35-66 ng/mL), compared to the tears of more severe aqueous-deficient
cohorts, (Sjögren disease, Stevens-Johnson, 6 mm Schirmer value) with levels in the
101–381 ng/mL range. 378 These data are consistent with observations that 11-14% of
early stage, patients with EDE (4.9 second TBUT, 312 mOsm/L, 14.0mm Schirmer
value) presented with ELISA-validated MMP-9 positivity. 482 Increases in neutrophil
elastase, MMPs 483 and neutrophil extracellular traps are seen in severe aqueous-
deficient subjects (chronic ocular GVHD, Sjögren disease and ocular cicatricial
pemphigoid) that are not commonly observed in non-autoimmune DED and healthy
controls. 431 Similarly, the anti-protease Cystatin S was found to have a significant,
inverse relationship with aqueous severity in non-autoimmune DED subjects, falling
from about 2,000 ng/mL in controls (16 mm Schirmer value) to 400 ng/mL in moderate
aqueous-deficient DED (4 mm Schirmer value), while MMP-9 was only mildly
upregulated to the 20–40 ng/mL range in those subjects. 484 The increase in cathepsin
S, a serine protease that is known to degrade a variety of essential components of the
normal tear film and glycocalyx (lactoferrin, sIgA, proteoglycan 4), 428,485 is also stratified
by inverse tear volume; increasing monotonically as tear volume decreases. 376 Of note,
although cathepsin S is dramatically enhanced in Sjögren disease compared to non-
autoimmune subjects on average, non-autoimmune subjects with 0–5 mm wetting on
the Schirmer strip exhibited equal or higher levels of cathepsin S than patients with
Sjögren disease with > 15 mm of wetting. 376
These data invoke the question of causality, whether aqueous deficiency leads to
inflammation and protease release, or vice versa. In an animal model of severe
aqueous deficiency, excision of the lacrimal gland resulted in a significant upregulation
of serine proteases in corneal tissue including tryptase, urokinase plasminogen activator
receptor and protease activated receptor 2 expression, which upon activation, is able to
induce MAPK / ERK-1 & 2 signalling with downstream NF-κβ, ICAM-1 & cytokine
expression, 429 suggesting that aqueous deficiency can precede a measurable protease
burden at the ocular surface. In contrast, MMP-9 positivity was found in 84% of GVHD
as compared to 33% of non-autoimmune DED patients, even as the GVHD patients
48
trended towards increased Schirmer test values, lower OSDI and higher TBUT than the
DED controls, 486 suggesting that immune infiltration into the lacrimal gland precedes
the protease release in GVHD.
Ultimately, in non-autoimmune DED, the transition from low levels of protease release
to inflammatory DED with excessive protease concentrations is likely related to
progressive alterations in the lacrimal gland and ocular surface. These might result from
androgen deficiency, 3,6 accumulation of advanced glycation end-products in the
lacrimal gland, 395 diabetic keratopathy and nerve degeneration, 487 or other factors that
lead to significantly less aqueous flow, thereby tipping the homeostasis towards a
severe hyperosmolar state and associated excessive, pathogenic protease release. It is
recognised that in marked EDE or lipid deficiency, tear and ocular surface homeostasis
may be sufficiently disrupted to cause inflammation and that in advanced disease,
features of both are displayed.
The glycocalyx lubricates, 488 retains water at the ocular surface, 489 provides wetting to
the ocular surface, 490 acts as a barrier and helps remove debris from the ocular surface.
491
The hydrophilic epithelial surface provides 82.5 sec/cm of specific resistance
compared to 12.9 sec/cm for the lipid layer, 489 establishing the glycocalyx as one of the
components responsible for preventing evaporation. Electron micrographs of
intracellular, pre-expression glycocalyx showed that “the increase in numbers of
subsurface vesicles that occur in some external eye diseases may reflect an attempt to
increase the binding of the mucus to the eye surface… In late keratoconjunctivitis sicca,
however, the subsurface vesicles are absent. 492” These foundational observations
identified the difference between evaporative DED and other ocular surface conditions,
where exogenous stresses lead to a compromised epithelial glycocalyx – the proximal
cause for an increase in evaporation, hyperosmolarity and its downstream
complications in EDE.
Evidence that the glycocalyx plays a central role in disease pathogenesis has begun to
accumulate; for example, knockout of MUC4, the most abundant membrane associated
mucin in the conjunctiva with an ectodomain predicted to extend > 2 µM above the
apical cell surface, resulted in significantly reduced microplicae, tear film disruption, and
increased rose bengal dye penetrance into deeper layers of the ocular surface. 493 In
vitro evidence from corneal and conjunctival co-culture has shown that hyperosmolarity
(caused by a decreased blink rate) directly downregulated components of the epithelial
glycocalyx, increased TLR-4 expression, initiated release of cytokines and proteases
including MMP-9, and eventually, cellular apoptosis. 445 The hyperosmolarity
compromised glycocalyx led to significantly lowered TBUT, lower Schirmer value, and
49
persistently elevated fluorescein staining, whereas replenishment of the glycocalyx
using human recombinant lubricin (recombinant human proteoglycan 4) was able to
reverse these clinical indicators, as well as inhibit epithelial NF-kB translocation and
normalize IL-8, TNF-a, IL-1ß, TLR-4 and MMP-9 expression within one day of
supplementation. 445 A striking aspect of this study is that the common clinical
expressions of DED were recapitulated without immune cell involvement, establishing
the hyperosmolarity-compromised glycocalyx as a causal, initiating event in evaporative
disease. A related study found that intraperitoneal injections of streptozotocin, used to
induce metabolic hyperglycemia similar to type 1 diabetes in mice, caused dramatic
reductions in the extent of the corneal glycocalyx, with associated reductions in tear film
volume, number of goblet cells and upregulation of TLR-4, MAPK, IL-1, IL-6 and IFN-γ
genes within as little as one week following injection. 494 In a C57BL/6 mouse model,
scopolamine and desiccating stress induced an almost complete abrogation of secreted
(MUC5AC, MUC2) and transmembrane mucins (MUC1, MUC4, MUC15 & MUC16)
along with significant increases in conjunctival IFN-γ, leading to persistent epithelial
defects. 495 Importantly, an in vitro study evaluating the impact of applied hyperglycemia
in media (15 & 30 mM glucose vs. 5 mM control) showed that elevated glucose
exposure had no effect on the amount or distribution of membrane associated mucins in
either corneal or conjunctival epithelial cells, 496 suggesting that the pathophysiology of
streptozotocin-induced glycocalyx loss is more likely due to global metabolic disease
than exposure to hyperglycemia alone. In support of these data, a study of vitreo-retinal
surgery found that conjunctival MUC4 & MUC16 gene expression increased post-
surgery and tear osmolarity was reduced in normal subjects, likely as a protective
response, but older diabetic subjects exhibited a lower goblet cell density, lower
MUC5AC and increased cytokine response by comparison, once again linking systemic
metabolic disease to ocular surface glycocalyx impairment. 497 In a mouse model of
GVHD, allogeneic transplantation of a mixture of spleen and bone marrow cells resulted
in a significant reduction in the area and thickness of the corneal glycocalyx, reductions
in MUC4 and MUC5AC and a coincident reduction of tear film volume and an increase
in fluorescein staining, which were partially abrogated after application of topical
rebamipide. 498
More recent data have shown that components of the glycocalyx also regulate immune
cells499, transduce extracellular environments to intracellular signaling pathways 500, and
actively inhibit proteases such as MMP-9 501, which would further implicate the
catabolism or downregulation of the glycocalyx as an initiating event in DED
pathogenesis. As the glycocalyx is altered in ocular surface pathology, large decreases
in sialic acid 502 and increases in galectin-3 are observed in the tear film that strongly
correlate with disease severity. 503,504 When released into the tear film, galectin-3 seems
to amplify the IL-1ß mediated inflammatory response, 505 which is similar to how
50
degraded low molecular weight hyaluronic acid becomes pro-inflammatory in other
tissue systems. 506,507
By leveraging novel models of primary human meibomian gland epithelial cells, ductal
from human tissue, 510,511 3D cultures and organotypic cultures, 462,512-517 researchers
have further investigated the pathophysiology of MGD. PPARγ agonists, such as
rosiglitazone, may significantly promote cell differentiation, lipogenesis and anti-
inflammation in human meibomian gland epithelial cells. 168,518-522
51
exposure of the rat eyelid margin, 530 Soat1-null mice, 531,532 APOE KO mice, 533 and
Elovl1-deficient mice. 534
Recent studies have indicated that it may be possible to restore gland structure after
atrophy. In mice, fibroblast growth factor receptor 2 gene (FGFR2) knockout could lead
to significant gland acinar atrophy, 535 but this change is reversible if the knockout
condition is removed. Moreover, this recovery relies on the extent of ductal atrophy,
which indicates that ductal epithelia may serve as a reservoir for meibomian gland
progenitor cells for regeneration. 536 The importance of FGFR2 and other FGFRs were
also reported in humans, in which FGFR inhibiting anti-cancer drugs could induce
significant gland atrophy, and patients who use these drugs may develop MGD. 537
The relationship between systemic lipids and MGD has been a focus of several recent
studies. Patients with elevated serum total cholesterol, low-density lipoprotein and
triglyceride levels exhibited significantly higher levels of meibomian gland loss
compared to healthy controls. 538 Mice fed with a high-fat diet developed hypertrophic
meibomian glands, 539 decreased PPAR-γ expression, increased meibomian gland acini
cell apoptosis and mitochondrial damage, and activation of MAPK and NF-κB signaling
within the gland. 540 A high fat diet significantly altered the rhythmicity of meibomian
gland, which may offer new insights into the regulation of the glands by dietary lipids. 541
Patients who have long term dyslipidemia also showed significant meibomian gland
atrophy and changes in meibum quality, even while undergoing statin treatment. 542
Dietary cholesterol has a direct impact on meibum components and meibomian gland
pathophysiology. 543-545 In contrast, dyslipidemia and increased triglyceride levels were
found to be protective factors for meibomian gland atrophy in an elderly female
population. 546 A systematic review has suggested there is moderate evidence for a
beneficial effect of omega-3 supplements on MGD. 547
Dysfunctional regulatory T cells (Tregs) exhibiting reduced Foxp3, CD24 and CTLA-4
expression lose the ability to suppress the chronic Th17 mediated inflammation in an IL-
6 dependent manner. 556 Blocking the substance P / neurokinin-1 receptor interaction,
439
or the TLR4/MyD88 pathway 557, both of which are upstream of NF-κB, were shown
to normalize the Treg/Th17 balance. In mice colonized with intestinal microbiota from
patients with Sjögren disease, a reduced frequency of CD4+Foxp3+ T regulatory cells in
cervical lymph nodes was observed 558, while in aged mice, CD4+CD25+Foxp3+ T cells
were dysfunctional, lost suppressive ability, and produced significant amounts of
inflammatory cytokines IL-17 and IFN-γ. 559
53
Recent studies exploring the pathophysiology of DED have continued to define the
etiologically distinct subpopulations within the disease, recognizing limited evidence for
inflammation in evaporative DED, and expanding roles for metabolic, hormonal,
physical and molecular impacts on the ocular surface. Neural regulation of the
epithelium is taking on increasing importance, as are factors from the ocular surface
that damage nerves, such as hyperosmolarity, reactive oxygen species, and other
potentially excitotoxic stressors. Yet major questions remain about how to best classify
the subsets of disease within a single patient and to what extent observed pathways are
causal to symptomatology. The natural history of the disease is unclear, when or if
inflammation becomes involved, or when acute inflammation transitions into a chronic
state. While animal models of the pathophysiology have provided valuable mechanistic
insights, they often fail to fully replicate human disease progression. If, as the existing
data show, inflammation is most strongly associated with aqueous deficiency, whether
this due to a relative increase in hyperosmolarity compared to that seen in evaporative
disease, or whether inflammatory mediators and proteases from the diseased lacrimal
gland driving downstream changes in the epithelium underpin these effects. In part due
to these questions about pathophysiology, it is currently not possible to predict which
patients will respond to anti-inflammatory or other types of therapy and which ones will
show positive responses to lubricants. While yet to be established, there may be value
in evaluating human immune cell dynamics and cell populations using advanced
imaging methods such as functional in vivo confocal microscopy. 560 As the diversity of
drugs expands, it will be increasingly necessary to align etiology with mechanism of
action. Hundreds of millions of dollars of development rests on predicting responders
from non-responders. Thus, it is becoming ever clearer that classical indices of DED
such as corneal staining and TBUT are insufficient to support modern drug development
efforts, and that more informative methods of determining disease subtype are critical
for future research.
8. Tear Film
8.1 Introduction
The Tear Film Report of TFOS DEWS II described the biophysical and biochemical
aspects of tears and how these change in DED. 561 The report noted that DED is
characterized by loss of tear volume, more rapid breakup of the tear film and increased
evaporation of tears from the ocular surface. The tear film lipids, proteins, mucins and
electrolytes contribute to the integrity of the tear film but exactly how they interact was
an area of active research. Tear film osmolarity, proteins and mucins could potentially
be used as biomarkers for DED. Some of these themes have been explored by
research since the publication of the TFOS DEWS II report, and these are highlighted in
the current report.
54
For the current report, the authors undertook searches in Pubmed, Scopus and Web of
Science databases for keywords ‘tear film’ and ‘dry eye’ since the publication of the
TFOS DEWS II Tear Film Report in 2017. After reviewing the titles and abstracts of
documents obtained, the authors have concentrated on those areas that have produced
the greatest number of articles. This report focusses on concepts and more detail on
specific components can be found within the cited references.
Simple correlations between symptoms and tear film or ocular surface characteristics
were sought. Whilst correlations do not imply causation, they can point the way to a
greater understanding of the mechanisms involved in signs and symptoms of DED and
identify areas for further investigation, recognising that there may be confounders in
individual studies and populations may differ. There was no correlation between tear
evaporation and symptoms (OSDI score), nor between tear evaporation and tear
production. 568 However, one study found significant positive correlations between OSDI
and tear evaporation rate scores in subjects with refractive errors. Both OSDI and tear
evaporation rate were lower in those without refractive errors. 569 These findings may
have been confounded by the inclusion of participants with uncorrected refractive errors,
smokers or differences in other demographic factors, but it may be important to adjust
for refractive error in studies of tear film and symptoms.
OSDI score has been negatively correlated with NITBUT, 563,570-574 tear meniscus height
(TMH) 570,571 and meibomian gland area, 574 and positively correlated with meibomian
gland loss. 573,574 NITBUT was positively correlated with TMH, meibomian gland dropout
grade 570,571 and corneal or lid margin staining. 563 Females with refractive errors had
significantly lower tear film lipid layer (TFLL) thickness, TMH, and NITBUT scores than
those without refractive errors.570 A strong correlation was found between TMH and
55
NITBUT, but no correlations between these parameters and OSDI scores.575 These
differences may be due to difference between populations and between techniques.
TBUT has been correlated with meibomian gland irregularity determined from
meibography images taken with a keratograph.576
Tear meniscus height was significantly correlated with the Schirmer value and tear film
breakup time and negatively correlated with ocular surface staining score, but there
were no correlations with any MGD indicator. 571
A study of normal subjects and those with MGD found a positive correlation between
precorneal tear film thickness and thinning rate and negative correlations between
precorneal tear film thickness and TFLL thickness and between precorneal tear film
thinning rate and TFLL thickness. 577. There were no associations between TFLL
thinning rate and any of precorneal tear film thinning rate, precorneal tear film thickness
or TFLL thickness.577 TFLL thickness has been positively correlated with age,
meibomian gland expressibility, and negatively correlated with meibomian gland dropout
572,578
and OSDI score.572
Several studies have been conducted to determine whether tear film osmolarity
correlated with clinical signs and symptoms of DED. Across population-based studies,
tear osmolarity is generally not correlated with tear film breakup time, corneal
fluorescein staining score, lid hyperemia, tear production, blink interval, Ocular
Protection Index, Schirmer I test, meibum expressibility, meibum quality or MGD. 579-583
However, this may depend on how DED was classified, as other studies found
correlations between tear osmolarity and OSDI discomfort subscore, corneal and
conjunctival staining scores 584 OSDI score, ocular surface staining and Schirmer I.585
Data from the DREAM (Dry Eye Assessment and Management) study, whilst
demonstrating some correlations between tear osmolarity and signs and symptoms,
were interpreted as being not indicative of causation as changes in tear osmolarity were
not associated with changes in signs and symptoms of DED. 586 Other recent
longitudinal studies, however, have suggested a relationship between change in tear
osmolarity with treatment and iatrogenic effects changes, suggesting a link in certain
DED subtypes. 587-592 The Diagnostic Methodology Subcommittee report of TFOS
DEWS III 1 gives more background on correlations and the limitations of this approach,
and the hyperosmolarity section of the Pathophysiology section of the TFOS DEWS III
Digest report (Section 7) discusses the role of osmolarity in more detail.
Several studies have examined the tear film lipid layer (TFLL). These have examined
changes to the lipids in DED and/or MGD patients compared to controls, as well as
which lipids contribute to tear film stability and reduced tear evaporation.
56
Broadly, meibum, combinations of meibum lipids and a thicker TFLL were associated
with a significantly slower evaporation flux, 593-596 and significantly slower tear breakup.
597
Tear film proteins can also reduce evaporation of water, and this is enhanced when
some of the proteins (human serum albumin and lactoferrin) but not others (human
lysozyme and bovine mucin) are used in conjunction with mixture of polar and non-polar
lipids found in meibum. 598 Lysozyme can reduce the surface tension of lipids on water,
by disrupting the order of lipid molecules. 599
Patients with MGD have significantly lower peak height ratios of the CH3/CH2 bands in
NMR studies than normal subjects, and this was hypothesized to be due to changes in
branched hydrocarbon chains, which contain fewer CH2 moieties, and straight chain
hydrocarbons, which contain more CH3 moieties. 600The authors further hypothesized
that, as van der Waals interactions between CH2 moieties were responsible for lipid
ordering, the more CH2 in meibum from MGD patients could contribute to a more
ordered TFLL. A more ordered TFLL can contribute to a more patchy layer resulting in a
deterioration in spreading and decreased elasticity. 600 Another factor that is involved in
TFLL stability is saturation of lipids, with increased saturation resulting in stiffer, thicker,
and more elastic films at high surface pressures. 601 Lipid saturation is a major factor
that contributes to lipid disorder and the phase transition temperature of lipid layers. 602
Meibum of MGD patients has lower surface pressures in Langmuir trough experiments
compared to meibum in those without MGD, and formed more brittle unstable patchy
layers. 603
The cholesteryl ester to wax ester ratio decreases in patients with MGD. 604-606 An
optimal mixture of wax and cholesterol esters in the TFLL may be necessary to disrupt
the ordered packing of pure lipid species, leading to better lipid spreading, 607 a more
stable tear film, 608 and thinner lipid layer patterns. 609 In a model system, mixtures of
cholesteryl oleate (CE) and behenyl oleate (a wax ester mimic) plus phosphatidylcholine
increased the surface pressure of films on phosphate buffered saline. 610 A layer of
behenyl oleate can form a crystalline state on water and reduce its evaporation 611 Iso-
branched wax esters help to reduce evaporation and increase surface pressure. 612 On
the other hand, addition of cholesterol to cholesteryl esters increases film rigidity. 613
People with thin TFLLs (who tended to be those with EDE) and people with irregular
TFLL patterns (who tended to be those with ADDE), had lower levels of cholesteryl
esters and lysophospholipids, but higher levels of glycerolipids and phospholipids in
their tears than those with normal TFLL patterns. 614
Other meibum and tear film lipids that have been examined are the O-acyl-ω-hydroxy
fatty acids (OAHFAs). Ultra-long OAHFAs found in TLLF and meibum are capable of
integrating other species in the polar lipid layer thereby molding its properties and/or
providing a base for the creation of hierarchical structures within the TFLL. 615 In vitro,
57
OAHFAs can spread well on the surface of phosphate buffered saline to form a solid
monolayer with a crystalline structure. 616 Longer chain OAHFAs prevented evaporation
of water at a mean molecular area of approximately 18 Å2/molecule. 616
Mixtures of OAHFAs and wax esters may produce even more evaporation resistant
films. 616 Indeed, wax esters existing in the solid state under physiological conditions
were capable of forming a mixed condensed monolayer with OAHFAs, and such a
monolayer exhibited very high evaporation resistance in vitro. 617 Whilst OAHFAs can
induce stable multilamellar cholesterol ester films on water or phosphate buffered saline,
618
OAHFAs alone reduced evaporation, with cholesterol esters having no effect. 618 An
additional double bond in the hydroxy fatty acid chain of OAHFAs was accompanied by
more disordered molecular organization, which led to a loss of the evaporation
resistance.619 The ultralong chain lengths observed in naturally occurring OAHFAs may
require the presence of double bonds to achieve an appropriate balance between
spreadability and evaporation resistance.619 The observations of ordered lipids resulting
in deterioration of spreading and reduced elasticity, whereas a more disordered
molecular organisation leads to a loss of evaporation resistance require further
exploration of how the ordering of different types of or combinations of fatty acids can
optimise the evaporative resistance, spreadability and elasticity of the film.
There have also been several studies examining the contributions of sphingolipids to
the TFLL and meibum. Meibomian gland loss has been associated with loss of some
sphingosines from the TFLL. 609 Individuals with poor meibum quality had changes to
their sphingolipids in tears and meibum.622,623 The presence of both sphingomyelin and
ceramide increases surface tension due to the change their position in the TFLL under
lateral pressure.624 Sphingomyelin at the interface of the TFLL with the aqueous phase
has a role in capturing the protein lysozyme and entrapping it in the TFLL.625
Finally, several experiments have shed important light on how meibum lipids are
synthesized and the consequences of disturbance in their synthesis. Using mice with
specific genes knocked out has shown that fatty acid w-hydroxylase Cyp4f39, acyl-CoA
reductase FAR2, the elongases of very long chain fatty acids- 1i or -3, alcohol
acyltransferases Awat1 and Awat2, and sterol O-acyltransferase 1 are all involved in
various stages of the production of meibum lipids such as OAHFAs, wax esters and
58
cholesterol esters. 626-633 Some of the major findings when genes for these enzymes
were knocked out was shortening of tear film break-up time,626 plugged meibomian
gland orifices, tear film instability and increased tear evaporation, 627,631 increases in
blink frequency and evaporation from the ocular surface, 628, shorter chain, branched
and unsaturated cholesterol esters 629, and changes in the melting temperature of their
meibum.629,630
Recent findings continue to emphasize the importance of the tear proteome as a non-
invasive tool for trying to discriminate between types of DED 634,635 and also for disease
monitoring, for instance revealing changes in the levels of proteoglycan 4, in tears of
those with Sjögren disease. 636
One interesting study explored the potential of tear MUC5AC and IL-8 levels to
distinguish between Sjögren and non-Sjögren DED, indicating that lower MUC5AC and
higher IL-8 levels could serve as biomarkers for Sjögren syndrome, aiding in its
diagnosis.637 Similarly, the tear thrombospondin-1/matrix metalloproteinase-9 ratio is
significantly reduced in Sjögren disease compared to non-Sjögren DED, offering
another potential biomarker. 480 Additionally, the MMP-9/lactoferrin ratio was positively
correlated with ocular inflammation and tear film stability in stable controlled patients
with Sjögren disease. 638
Tear biomarkers related to ubiquitination (LMO7 and HUWE1) and the regulation of
intracellular vesicle dynamics (TPD52) are significantly elevated in patients with Sjögren
disease, suggesting a role for these pathways in the underlying mechanisms of the
disease. 639 Other robust protein biomarkers have been identified using LCMS/MS in
multiple cohorts, highlighting several proteases and protease inhibitors plus noting the
relevance of oxidoreductase proteins in Sjögren disease. 640 Oxidative stress
biomarkers serve as indicators for assessing the extent of ocular surface damage, and
research using both laboratory models and individuals with DED underscores redox
imbalance as one of the many pathophysiological factors driving the disease. 641
Sjögren disease may also be characterised by reduced or absent levels of complement
regulators (CD59, CD55, and CD46), alongside upregulation of C3. This imbalance may
drive excessive complement activation in ocular tissues and may suggest a possible
therapeutic pathway. 642
Ongoing research continues to shed light on the role of tear cytokines in the context of
DED. A recent meta-analysis of tear film cytokines in Sjögren disease revealed elevated
levels of inflammatory cytokines, including IFN-γ, TNF-α, IL-1α, IL-1Ra, IL-4, IL-6, IL-8,
IL-10, IL-17, IL-21, and IL-22. Interestingly, IL-23 levels were significantly lower
compared to healthy controls. 643 Additionally, reduced levels of epidermal fatty acid-
59
binding protein (E-FABP), which regulates inflammatory pathways on the ocular surface,
further suggest its potential as a biomarker for epithelial damage and altered lipid
metabolism in the disease. 644 However, despite these advances, a persistent challenge
in the field is the lack of standardization in sample collection and analytical techniques,
645
which hinders the translation of these findings into clinical practice.
Tear proteomic profiling has also revealed altered biological pathways related to corneal
sensitivity and nerve parameters in DED and neuropathic corneal pain. 646,647 (See
section 8.5)
One emerging area of interest in this field involves examining the complex interplay
between the tear proteome and the ocular surface microbiome, and its potential
contribution to the onset of dry eye. Recent data indicate that the interaction between
these two systems is beneficial to combat pathogens and maintain ocular health
through modulation of the inflammatory response. 648 Moreover, the application of
whole-metagenome sequencing has enabled researchers to pinpoint unique microbial
compositions in individuals with DED, with the relative abundances of certain bacteria
being correlated with specific tear proteins in the tear fluid. 649 Understanding the
crosstalk between these two components of the tear fluid may provide opportunities for
the development of personalized therapeutics tailored to individual patient profiles.
Interactions between the various components of the tear film are important for
maintaining its stability and protecting the ocular surface and the expression of these
components is interconnected. For instance, there is a compensatory mechanism where
the absence of lipid secretion, caused by stearoyl-CoA desaturase-1 deficiency, leads
to increased tear volume and enhanced expression of mucins, along with changes in
the expression of lipid metabolism genes. 632 The knowledge gained from understanding
the interactions between tear film components is also being used to develop dry eye
treatment strategies. Two treatment studies suggest that mucomimetic polymers can
improve the structure and functionality of the lipid layer. 650,651 Cross-linked hyaluronic
acid is particularly effective in promoting tear ferning and the spreading of meibum in
vitro, suggesting therapeutic potential of these polymers for ocular surface health. 652
8.6 Mucins
The realm of ocular mucins has witnessed several scientific discoveries, shedding
additional light on their important role in maintaining ocular surface health. Membrane-
associated mucins act as regulators of transcellular barrier function, tear film stability
and apical epithelial cell architecture. 493,653 Membrane-associated mucins are distinctly
distributed along the conjunctiva of those with and without DED, emphasizing the vital
60
role of these mucins in minimizing eyelid friction during blinking. 654 Advanced modelling
approaches have enabled researchers to establish a relationship between the loss of
membrane-associated mucins and the premature rupture of the tear film, predicting
quantitatively the shortening of NIBUT observed in DED. 655
The altered expression of miRNAs in tear fluid has positioned them as promising
candidates for non-invasive biomarkers in DED. To date, approximately 300 distinct
miRNAs have been identified in tear fluid, many of which are recently discovered and
are poorly understood in terms of their regulatory functions. 662 RNA sequencing
experiments has revealed that extracellular vesicles in the tear film in both non-Sjögren
DED and healthy controls carry distinct miRNA profiles, with 126 differentially expressed
miRNAs between the groups. 663 Among these, nine miRNAs (miR-127-5p, miR-1273h-
3p, miR-1288-5p, miR-130b-5p, miR-139-3p, miR-1910-5p, miR-203b-5p, miR-22-5p,
and miR-4632-3p) were significantly upregulated in DED and were associated with
inflammation, indicating a potential role in disease pathogenesis. Other studies, on the
other hand, have shown an inverse relationship between miRNAs in tears and
inflammation. For instance, miR-223 inhibits hyperosmolarity-induced inflammation
through downregulating NLRP3 activation in human corneal epithelial cells and those
with DED. 412 Comparison of the expression of 43 miRNAs in the tears of those with
Sjögren disease and healthy controls have revealed 14 significantly differentially
expressed miRNAs that may be involved in the pathogenesis of Sjögren disease,
though none were correlated with ocular staining scores. 664 As the precise molecular
mechanisms by which these miRNAs contribute to DED remain largely unclear, and the
lack of standardized methods hampers comparison across studies, further research is
essential to clarify the specific roles of miRNAs within the ocular environment.
61
Translational models of the tear film aim to bridge the gap between basic research and
clinical applications. Significant advances in recreating the ocular surface in vitro have
emerged using organs-on-chips and stem cell-derived organoids. Organ-on-chip
technology employs microfluidic devices to replicate the ocular surface environment,
allowing for detailed study of tear film components such as mucins and inflammatory
cytokines. By utilizing microfluidic platforms with segmented channels, researchers can
simulate dry eye conditions by exposing cultured human corneal epithelial cells to an
air-liquid interface within a chip. 665 Additionally, induced pluripotent stem cells have
facilitated the creation of organoids, offering a new platform to study the molecular
mechanisms involved in DED. These approaches have allowed the generation of
functional conjunctival epithelial lineage cells, including goblet cells. 445,666,667
Biomimetic models that rely on the enzymatic removal of mucins in cell culture have
emerged to more accurately recapitulate the pathological changes in lubrication,
adhesion, and barrier function often observed in mucin-deficient DED. 668 In addition,
hydrophilic and hydrophobic glass surfaces have been used to model the interactions of
the tear film with a healthy cornea or a hydrophobic cornea in the absence of a
glycocalyx. 669 Also noteworthy is the development of miniaturized analogs of a blinking
human eye, which utilize a dome-shaped 3D cell culture scaffold to mimic the dynamic
interface between the ocular surface and the external environment. 445 Other
investigators have used molecular modeling of the tear film and machine learning
models to better understand the contribution of proteins to tear film stability and
proteomic changes in MGD, respectively. 670,671
62
This review has identified several areas for future research that may help in the
understanding of the pathogenesis and subclassification of DED, as well as identifying
better biomarkers to help clinicians classify and monitor DED. Researchers should use
lipids that more closely align with those in meibum, rather than mimics of meibum lipids
for future in vitro experiments. Further research is needed to explore the relationship
between disordered lipids that result in spreading and increased elasticity, compared to
ordered lipids that lead to improved resistance to evaporation. Additionally,
incorporating more accurate models of the muco-aqueous layer in laboratory
experiments may help elucidate the roles of lipids, mucins, proteins, and other
components in stability and evaporation. There is also a need for a more detailed
understanding of whether tear biomarkers can be used to differentiate subtypes of DED
as described in the TFOS DEWS III Diagnostic Methodology report. 1 Analyses of
microbiome changes across individuals of different ethnicities and countries of
residence may provide further insights into its potential role in DED pathogenesis or as
a marker for the disease. Understanding the potential role of different microRNAs in
DED pathogenesis, DED subtype or as biomarkers could be a highly promising area for
future investigation.
9.1 Introduction
As described in the TFOS DEWS II Pain and Sensation Report, 40 the ocular surface,
particularly the cornea, is densely innervated with sensory fibers that have important
functions in the maintenance of ocular surface health. The dynamic nature of ocular
surface nerves is often underappreciated, as this system continuously undergoes
remodeling in adults, particularly nerve terminals in the corneal epithelium. Many
common insults can amplify this remodeling, including accidental or surgical injury or
disease processes that lead to chronic inflammation (e.g., Sjögren disease, Steven-
Johnson disease, herpes keratitis). Damage to ocular sensory nerves can result in
corneal nerve loss, a change in nerve architecture, and altered sensitivity to stimulation.
Although nerve regeneration can occur after such damage, it is typically gradual and
incomplete. As a result, it often fails to fully restore the original density, architecture, and
function of the corneal innervation, leading to persistent changes in neural excitability.
This update will consider new evidence in corneal nerve remodeling during normal
physiology and following trauma, surgery or inflammation and recent evidence for the
anatomical and functional status of the corneal nerves in diagnosing and managing
DED.
63
The development of corneal nerves begins in the fifth gestational month with the
formation of sensory axons around the cornea, followed by their radial extension into
the corneal tissue. Although the specific molecular signals controlling corneal nerve
growth are still unknown, molecules such as nerve growth factor (NGF) and various
neurotrophic factors released by corneal cells contribute to the development and
survival of corneal nerves. The growth of the corneal nerves does not stop once
development is complete but continues to occur continuously. In adults, corneal
subbasal nerves and their terminals undergo continuous morphological rearrangements
throughout life while the stromal nerves present few morphological changes. 674-676
Observations from living human eyes using in vivo confocal microscopy reveal that
subbasal nerves move centripetally at rates of 10–20 μm per day. These nerves
elongate by adding new material near the site of nerve penetration to the epithelium
from the Bowman's layer. Distal nerve segments eventually degenerate or slough into
the tear film due to the turnover of the corneal epithelium. 40,677 Additionally,
intraepithelial nerve terminals undergo spontaneous morphological changes through
long-term reconfigurations and short-term reorganization in response to outward
migrations of differentiating epithelial cells. Notably, corneal nerve remodeling is more
prominent in the central regions of the cornea than in the periphery. Research in living
transgenic and knock-in mice reveals that, over time, there are noticeable changes in
the subbasal nerve fibers and intraepithelial nerve terminals. The presence of
continuous remodeling is supported by the expression of growth-associated protein
43sub in the epithelial nerves of intact corneas. 675,677
Corneal nerves may be severed during corneal and anterior segment surgery, such as
photorefractive keratectomy (PRK), laser-assisted in situ keratomileusis (LASIK),
cataract surgery, iridectomy, trabeculectomy, and corneal transplantation. The survival
of corneal nerves relies on transporting essential substances from their parent nerve
64
cells in the trigeminal ganglion. Therefore, surgical procedures that disrupt corneal
nerve fibers can lead to rapid degeneration of the distal axons, reduced corneal
sensitivity, and impaired functional integrity of the ocular surface.
Although corneal nerves can regenerate, this process is slow and imperfect. After most
corneal surgeries, the regeneration of nerves is characterized by decreased nerve
density, changes in nerve structure, and diminished corneal sensitivity. Regeneration is
more delayed and incomplete when nerves are cut closer to their origin. 679
Consequently, surgical disruption of the subbasal and subepithelial nerve plexuses
typically results in less severe and short-term damage to corneal innervation than deep
or penetrating incisions affecting major stromal nerve bundles.
Although corneal sensitivity typically returns to preoperative levels after LASIK within 6-
12 months, 680 a significant proportion of patients may experience long-term dry eye
symptoms due to impaired nerve regeneration. 681,682 Nerve damage in small incision
lenticule extraction surgery is less marked than in LASIK, resulting in a faster nerve
regeneration three months post-surgery. However, no significant difference is observed
at six months. 257 A meta-analysis of corneal sensitivity recovery has shown a more
rapid early recovery of corneal sensitivity with small incision lenticule surgery compared
with femtosecond LASIK. 683(See also TFOS Lifestyle report 684)
In PRK, where the corneal epithelium is removed, and the corneal stroma is reshaped
with an excimer laser without creating a flap, nerve regeneration and recovery of
corneal sensitivity occur more rapidly than in LASIK. 685 Despite this, subbasal nerve
density, architecture, and corneal sensitivity may remain reduced for up to 1-2 years
post-PRK. 686
In cataract surgery, small, perilimbal incisions have minimized the risk of significant
injury to corneal innervation. However, in corneal transplantation procedures such as
penetrating keratoplasty, a full-thickness incision cuts all corneal nerves, resulting in
complete denervation of the transplanted cornea. Nerve regeneration following
penetrating keratoplasty is slow, and even years later, the innervation density of the
transplanted tissue remains lower than that of the host peripheral cornea. 687 Stromal
nerves regenerate poorly, which may be attributed to the misalignment of Schwann cell
channels in the donor cornea with the stromal nerve stumps in the host cornea. This
contrasts with the perilimbal incisions used in cataract surgery, where stromal nerves on
opposing sides of the incision remain closely aligned. Following penetrating
keratoplasty, limited nerve regeneration occurs. A few subbasal nerve fibers elongate
through the epithelium at the graft margin to enter the donor basal epithelium.
Regenerated subbasal nerves may exhibit atypical orientations and morphologies. 687
65
The corneal nerve density and corneal sensitivity remain significantly reduced
compared to healthy corneas even decades after surgery. 688
Although theoretically expected not to alter corneal innervation, other corneal transplant
techniques, such as Descemet membrane endothelial keratoplasty, show a temporary
decrease in nerve density early after transplantation. However, complete recovery of
corneal nerve density and function to preoperative values typically occurs within 6-10
months post-surgery. 689 Studies have also reported similar results with Descemet’s
stripping automated endothelial keratoplasty, where the corneal sensations were noted
to be normal within 6 months following surgery. 690 Another study comparing corneal
sensation between Descemet membrane endothelial keratoplasty and penetrating
keratoplasty showed that corneal sensation improved significantly following Descemet
membrane endothelial keratoplasty but was slightly but not significantly decreased after
penetrating keratoplasty. 691 These findings suggest better preserved corneal
sensations following endothelial transplants compared to PK.
The peripheral nervous system has impressive regenerative capabilities following injury.
However, injuries to the afferent axons of trigeminal neurons can lead to significant
morphological and functional changes, which depend on the magnitude and location of
the damage. 679,692 The regeneration of neurons after injury is influenced by a supportive
environment for axon growth and the involvement of non-neuronal cells like Schwann
cells. 693
As described in the TFOS DEWS II Pain and Sensation Report, 40 mechanical trauma or
inflammatory damage to the peripheral axons of corneal trigeminal neurons causes a
complex cellular response and changes to their spontaneous and stimulus evoked firing
rates. The expression, distribution and activation thresholds of the transduction ion
channels changes. These disturbances lead to increased responsiveness to normal
stimuli (allodynia), spontaneous firing without intended stimulation and increased
abnormal or unpleasant sensations from a stimulus that would normally elicit a
response (hyperalgesia).
The mechanisms that stimulate and direct neurite outgrowth from injured and intact
areas of corneal innervation following local nerve injury still need to be fully understood.
Corneal epithelial cells release several growth factors following an injury, which may
play essential roles. Nerve growth factor is upregulated after corneal epithelial wounding
and topical recombinant human nerve growth factor has been used to stimulate corneal
nerve regeneration and recovery of corneal sensitivity 694 in the treatment of
neurotrophic keratitis, where sensitivity is reduced. 695 The role of nerve growth factor in
66
the mechanisms resulting in allodynia or hyperalgesia, however, has not been
established, although restoration of nerve function may be advantageous.
Ocular and systemic diseases, including herpes virus keratitis, diabetes, and ADDE,
can negatively impact corneal nerves. 696 Diabetes significantly alters corneal nerve
morphology and function, reducing nerve density and sensitivity. In diabetic patients, the
appearance of subbasal nerves resemble intraepidermal small fiber neuropathy, making
in vivo confocal microscopy of corneal nerves a valuable biomarker for monitoring
diabetic neuropathy. 697
In summary, in healthy adult corneas, the subbasal nerve fibers and intraepithelial nerve
endings undergo continuous remodelling and regenerate rapidly following damage,
whereas stromal nerves maintain their structure over time. The dynamic nature of
intraepithelial nerve endings accounts for their rapid regeneration after injury, while
subbasal nerve fibers regenerate more slowly, and stromal nerve trunks may not fully
regenerate. Corneal nerve morphology is affected by trauma, ocular surgery, infections,
chronic tear deficiency, and various systemic diseases. Although damaged corneal
innervation can regenerate like other peripheral nerves, their morphology and function
are often incompletely restored, leading to reduced sensitivity, abnormal sensations,
and pain.
Many manifestations placed under the heading of “dry eye” occur because of
morphological and functional changes in ocular innervation. Changes in ocular sensory
nerves induce symptoms such as unpleasant sensations of different intensities, ranging
from dryness or ocular discomfort to lacerating and burning pain. 698 Conversely,
abnormal nerve functioning can lead to alterations in tissue trophism and in the
regulation of tear production and blinking, which in turn contributes to both symptoms
and signs.699 In other words, sensory innervation can be altered by chronic eye dryness
and can also contribute to DED pathogenesis.
Reduction of the ocular surface moistness either by reduced tearing or increased tear
evaporation is a stressful situation for the corneal epithelium exposed to a hyperosmotic
tear film and the environment (See section 7). Tear hyperosmolarity can independently
affect subbasal corneal nerves in an animal model. 700 As a response to this
hyperosmolar and desiccating stress, corneal epithelial cells and immune resident cells
produce local inflammatory mediators, primarily IL-1 and TNF- α, which stimulate the
production of matrix metalloproteases, activate dendritic cells and local inflammation
occurs, 458 leading to sensitization of nociceptive nerve terminals and development of
discomfort and pain sensations. When dryness becomes chronic, it also leads to
corneal nerve damage and, consequently, the morpho-functional changes of the
67
sensory nerves during chronic eye dryness resemble both those produced during
inflammation and those produced by nerve injury. The eyelid movement causes
mechanical friction at the ocular surface when the tear film is thin and does not lubricate
well. Together with the chronic inflammation, this movement damages the epithelium
and the intraepithelial nerve terminals, and eventually the subbasal nerve fibers,
triggering the mechanisms of nerve degeneration and regeneration. 701
Most of the alterations of ocular surface sensitivity in chronic eye dryness are due to
functional changes in corneal nerves that resemble those seen in injured nerves. There
is an increased excitability of corneal nerves consecutive to the increased activity and
expression of sodium channels,709-711 resulting in an increased spontaneous firing of
cold thermoreceptors that not only leads to dryness sensations but also to dysregulation
of protective mechanisms driven by thermal sensory input such as tearing and blinking,
contributing to increase and perpetuate the ocular surface disturbances.
68
methods of assessing nociplastic pain, but the underlying pathophysiology remains
poorly understood.
Animal models have been developed to replicate human correlates of DED. While initial
focus examined immune abnormalities, recent studies have found that various insults
that create tear and ocular surface abnormalities also impact upon corneal nerves.
Several mouse models replicate DED in Sjögren disease, including an IL-2 receptor -
chain knockout model. 713 In one study, intraepithelial corneal nerve density of knockout
was compared to wildtype mice using confocal microscopy at 4 weeks, 6 weeks, 8
weeks, and 10 to 11 weeks after birth. After combining data from each time point,
overall density was significantly decreased in the knockout compared to wildtype. These
69
results suggest that corneal nerve alterations coincide with the onset of DED, with
reduced nerve fiber density commonly noted.
Nerve alterations have also been examined in a GVHD murine model, 714 using an
allogeneic bone marrow transplant (case) group (strain B10.D2 to strain BALB/c) and a
syngeneic bone marrow transplant group (strain BALB/c to strain BALB/c, control group)
at 1-, 2-, 3-, and 4-weeks post-transplant. Corneal nerve tortuosity and branching were
increased in the allogeneic bone marrow transplant group at 4-weeks post-transplant
compared to 1-week post-transplant. Conversely, no significant differences were
observed in branching or tortuosity in the syngeneic group at any time point. These
findings support the induction of morphologic changes in corneal nerves associated with
the progression of GVHD-associated DED.
Other protocols have been developed to model DED following ocular surgery, such as
photorefractive keratectomy (PRK). 715 PRK (-9D) was performed monocularly in New
Zealand rabbits while the contralateral cornea was used as a control. Corneal nerve
fiber density was evaluated in post-PRK corneas with acetylcholinesterase
histochemistry staining at 1 day (n = 3), 1 month (n = 3), 2 months (n = 3), 3 months (n
= 3), and 6 months (n = 4) after surgery. Compared to controls (n = 3), nerve density
was significantly decreased in post-PRK corneas after 1 day, 1 month, and 2 months,
but was not different at 3- and 6-months post-surgery. Nerve morphology alterations
persisted however, (increased tortuosity and aberrant innervation) until the final time
point at 6 months. Findings that surgery may result in corneal nerve alterations that do
not fully return to baseline by 6 months post-surgery, consistent with the effects of
refractive surgery in human. 680
Other studies focused on DED prompted by iatrogenic challenges. One murine study
used confocal microscopy with staining using an anti-β3 tubulin antibody to examine
corneal nerve fiber density after repeated topical administration of benzalkonium
chloride for 14 days. 716 There was a significant reduction in density in benzalkonium
chloride-treated mice compared to controls.
Partial or complete lacrimal gland excision has also been used to model DED. 436 In one
study, the number of corneal nerve terminals in the surgical group was assessed
qualitatively by confocal microscopy and compared to a sham-surgery and control group.
The surgical group demonstrated fewer corneal nerve terminals compared to both
sham-surgery and control groups. This study also examined nerve function with
electrophysiological studies and found that post-surgical animals developed time-
dependent corneal mechanical hypersensitivity accompanied by increased spontaneous
ciliary nerve fiber electrical activity. Other investigators have found similar functional
changes after gland excision, with increased activity and expression of sodium channels
70
that lead to increased excitability of corneal nerves. This results in a greater
spontaneous firing of corneal cold thermoreceptors, causing sensations of dryness, and
an enhanced excitability of corneal nociceptors that results in low frequency
spontaneous firing and increased response to stimulation, resulting in sustained
discomfort and hyperalgesia. 710,717,718 These findings suggest that, beyond nerve
density and morphology, corneal nerve function can be impacted differentially in DED
subtypes.
In DED, alteration of corneal nerve structure and function is frequently implicated in the
onset, progression, and severity of symptoms. Disruption of the tear film in DED can
result in nerve fiber damage which stimulates regenerative processes that are often
improper or incomplete. The TFOS DEWS II Pain and Sensation Report in 2017 720
determined that while IVCM is a reliable method for detecting abnormal corneal nerve
morphology, nerve density was a somewhat unreliable marker for nerve fiber damage,
but that other morphological parameters were more useful including increased tortuosity,
reflectivity and increased beading. More recently a higher frequency of microneuromas
has been documented. 702 These structural aberrations, in turn, may further impair tear
film production and the blinking reflex resulting in additional nerve damage and
conceivably neuropathic dysfunction. 702 Typical manifestations of neuropathic
dysfunction include ocular pain, loss of sensation, and tear hyperosmolarity. Damage to
nerve fibers in DED can also result in physiologic disruption of the corneal nerve’s usual
neurotrophic functions, including regulation of nerve growth factor and substance P, that
help mediate epithelial growth, local immunoregulation, and nerve fiber regeneration.
702,721
Together, these structural and functional changes likely play a significant role in
the pathophysiology of DED.
71
Interestingly, some individuals with DED report symptoms out of proportion to ocular
surface alterations and others are found to have signs that are more severe than their
symptoms suggest. 158 One possible explanation for the symptom>sign discordance is
the presence of central abnormalities, with inappropriate processing of somatosensory
signals due to changes in the central nervous system referred to as central sensitization.
Central sensitization is associated with generalized somatosensory dysfunction and
cutaneous hypersensitivity in addition to excessive ocular pain.
Analysis:
ImageJ/NeuronJ,
OSE grading
scale
722
United States SDED (n = 22) Imaging: multi- ↑ tortuosity in SDED
image (3), central and NSDED
NSDED (n = 12) cornea compared to
controls.
Controls (n = 5)
Analysis:
ImageJ/NeuronJ,
OSE grading
scale
725
France AIDED-NCP (n = 7) Imaging: single ↑ tortuosity in
72
image, central AIDED-NCP
MGD-NCP (n = 11) cornea compared to
controls. Tortuosity
AIDED (n = 8) not different in MGD-
NCP, painless
MGD (n = 8) Analysis: AIDED, or painless
ImageJ/NeuronJ, MGD compared to
Controls (n = 10) study-designed controls.
tortuosity scale
723
China SDED (n = 22) Imaging: multi- ↑ tortuosity in SDED
image (5), central compared to
NSDED (n = 20) cornea NSDED.
Analysis:
ImageJ/NeuronJ,
OSE grading
726
France SDED (n = 71) Imaging: multi- ↑ tortuosity in SDED
image (5), central compared to MGD
MGD (n = 20) cornea and controls.
Control (n = 20)
Analysis:
ImageJ/NeuronJ,
OSE grading
Corneal nerve fiber density (CNFD) has also been studied in various DED subtypes
(Table 6) and in most studies appears to be significantly decreased in Sjögren DED,
GVHD, non-Sjögren DED and neuropathic corneal pain compared to controls, 722,728
729,730 731
although within studies density is similar between DED types. 732 One small
study has evaluated MGD and found no reduction compared with controls, 725 however
a larger study found a reduction in both ADDE and EDE compared with controls. 733
The reduction in density is greater with increased symptoms. Those with normal-to-mild
symptoms have similar CNFD to controls. 734 These studies reinforce that CNFD values
are generally lower in some DED subtypes, most notably in auto-immune associated
DED and that CNFD may be affected by degree of symptoms severity/ocular pain.
73
to controls. CNFD not
Moderate-severe different in EDE with
EDE (n = 23) normal-to-mild
Analysis: symptoms compared
Control (n=43) ACCMetrics to controls.
727
Japan GVHD (n = 12) Imaging: multi- CNFD not different in
image (3), GVHD compared to
Control (n = 10) central cornea + controls.
inferior limbal
epithelia
Analysis:
ImageJ/NeuronJ
722
United SDED (n = 22) Imaging: multi- ↓ CNFD in SDED and
States image (3), NSDED compared to
NSDED (n = 12) central cornea controls.
Control (n = 5)
Analysis:
ImageJ/NeuronJ
730
Japan NSDED (n = 25) Imaging: multi- ↓ CNFD in NSDED
image (5), compared to controls.
Control (n = 25) central cornea
Analysis:
ImageJ/NeuronJ
735
France DED (n = 32) Imaging: multi- ↓ CNFD in DED
image (5), compared to controls.
Control (n = 15) central cornea
Analysis:
ImageJ/NeuronJ
732
Italy SDED (n = 20) Imaging: multi- ↓ CNFD in SDED and
image (3), GVHD compared to
GVHD (n = 19) central cornea controls.
Control (n = 30)
74
Analysis:
ACCMetrics
736
Turkey GVHD (n = 22) Imaging: multi- ↓ CNFD decreased in
image (3), GVHD compared to
Control (n = 28) central cornea controls.
Analysis:
ImageJ/NeuronJ
729
United NCP (n = 25) Imaging: multi- ↓ CNFD in the NCP
States image (3), and DED compared to
DED (n = 30) central cornea controls.
Control (n = 16)
Analysis:
ImageJ/NeuronJ
731
United ADDE (n = 24) Imaging: multi- ↓ CNFD in ADDE and
States image (3), EDE groups to
EDE (n = 46) central cornea controls.
Control (n = 45)
Analysis:
ImageJ/NeuronJ
725
France AIDED-NCP (n = 7) Imaging: single ↓ CNFD in AIDED and
image, central MGD-NCP compared
MGD-NCP (n = 11) cornea to controls. CNFD not
different in AIDED-
AIDED (n = 8) NCP or MGD
MGD (n = 8) compared to controls.
Analysis:
ImageJ/NeuronJ
Control (n = 10)
723
China SDED (n = 22) Imaging: multi- ↓ CNFD decreased in
image (5), SDED compared to
Control (n = 20) central cornea NSDED.
Analysis:
ImageJ/NeuronJ,
737
India ADDE/EDE + Imaging: single ↓ CNFD in those with
symptoms (n = 10) image, central DE symptoms
cornea compared to controls.
Control (n = 15)
75
ADDE +/- Analysis: CNFD not different in
symptoms (n = 57) ACCMetrics ADDE (+/- symptoms)
or EDE (+/-
EDE +/- symptoms symptoms) compared
(n = 16) to controls.
Control (n = 15)
738
China DED (n = 155) Imaging: multi- ↓ CNFD in DED
image (≥10), compared to controls.
Control (n = 20) central
Analysis: CS-Net
726
France SDED (n = 71) Imaging: multi- ↓ CNFD in SDED
image (5), compared to controls.
MGD (n = 20) central cornea CNFD not different in
SDED compared to
Control (n = 20) MGD.
Analysis:
ImageJ/NeuronJ
739
China DED (n = 25) Imaging: single ↓ CNFD in DED
image, central compared to controls.
Control (n = 20) cornea
Analysis:
ImageJ/NeuronJ,
ACCMetrics
733
Turkey ADDE (n = 22) + Imaging: multi- ↓ CNFD in ADDE and
EDE (n = 21) image (3), EDE compared to
central cornea controls.
Control (n = 20)
Analysis:
ImageJ/NeuronJ
DED= dry eye disease; EDE= evaporative dry eye; GVHD= graft-versus-host disease; SDED= Sjögren
dry eye disease; NSDED= non-Sjögren dry eye disease; NCP= neuropathic corneal pain; ADDE=
aqueous-deficient dry eye; CNFD= corneal nerve fiber density; MGD = meibomian gland dysfunction.
76
been included in some studies (Table 7). Conceivably, either due to the subjective
determination of microneuromas, or the difficulty of discriminating a turn in the nerve
from an ending, there are inconsistencies in the literature. 740,741 Some studies found an
increase in microneuromas among DED subtypes compared to controls, most notably in
autoimmune DED and MGD, 725,742 with a greater increase where there was co-existing
neuropathic corneal pain. 725 While these studies suggest that microneuroma frequency
or number is increased among several DED subtypes compared to controls, other
studies have not replicated these findings in those with symptoms of DED. 743 These
findings point to the need for additional studies that standardize microneuroma
determination and further explore their utility as a biomarker for DED.
AIDED (n = 8) Analysis:
ImageJ/NeuronJ
MGD (n = 8)
Control (n = 10)
743
United DED (n = 119) Imaging: multi- MN frequency not
States image (3), different in DED or DED
DED + refractive central cornea + refractive surgery
surgery (n = 19) compared to controls.
Control (n = 18)
Analysis:
ACCMetrics
742
India ADDE/EDE + Imaging: single MN frequency ↑ in those
symptoms (n = 14) image, central with DE symptoms
cornea compared to controls.
Control (n = 27)
77
ADDE +/- Analysis: MN frequency not
symptoms (n = 24) ACCMetrics different in ADDE (+/-
symptoms) or EDE (+/-
EDE +/- symptoms symptoms) compared to
(n = 65) controls.
Control (n = 27)
726
France SDED (n = 71) Imaging: multi- MN frequency ↑ in
image (5), SDED compared to
MGD (n = 20) central cornea controls but not
compared to MGD.726
Control (n = 20)
Analysis:
ImageJ/NeuronJ
DED= dry eye disease; NCP= neuropathic corneal pain; AIDED= autoimmune dry eye disease; MGD=
meibomian gland dysfunction; ADDE= aqueous-deficient dry eye; EDE= evaporative dry eye; SDED=
Sjögren dry eye disease; MN= microneuroma.
Recent work has explored the use of AI technology in nerve analysis, particularly the
use of deep learning AI models to automatically segment corneal nerves.744. Using the
TFOS DEWS II diagnostic criteria, 131 individuals with DED had reduced CNFD
compared with controls and values were similar to that of manual annotation.738
Applying new technologies to IVCM will allow for more consistent and faster
quantification of images that can applied across centers, facilitating comparisons across
different populations.
Corneal sensitivity testing is an important diagnostic tool that can provide valuable
insights into underlying somatosensory abnormalities in DED. The TFOS DEWS II
report highlights that corneal sensitivity to mechanical stimuli tends to be reduced in
patients with ADDE.720 However, studies using air/gas esthesiometers have shown
equivocal findings, with increased 745,746, decreased 747,748, and similar 749 corneal
sensitivity in various DED subtypes. This could be due to differences in stimulus
parameters (e.g., cold, chemical stimulus included in some testing paradigms) or due to
differences in neural responses across various DED subtypes (e.g., ADDE vs MGD).750
Thus, consideration of the type of stimulus, the subtype of DED and patient factors is
critical to interpreting corneal sensitivity results. Table 7 summarizes studies evaluating
sensitivity or pain responses in DED.
78
In a study of corneal mechanical sensitivity in those with DED symptoms tested using
an air jet aesthesiometer, 13% showed hypersensitivity and 11% had hyposensitivity. 749
When grouped by DED subtype, there was an association between ADDE and reduced
sensitivity.726 There is growing awareness that some individuals with symptoms of DED
likely have neuropathic contributors to symptoms and this group, as a whole, display
corneal hypersensitivity. Individuals with hypersensitivity had more severe ocular pain
complaints while those with hyposensitivity had more severe epithelial disruption. 751
These data highlight that differences in corneal sensation and symptom reporting may
align with differences in DED profiles.751 In a study using the Cochet-Bonnet
aesthesiometer, mechanical corneal and pain sensitivity were assessed in individuals
with short tear film breakup DED, defined as TBUT<5secs and Schirmer value >5mm,
and controls (n=46). Pain sensitivity threshold but not mechanical sensitivity threshold
was higher in the DED group. 752 Corneal hyperalgesia was present in 37% of the DED
group and there was a strong relationship between pain sensitivity and subjective pain
scores. 752 Cold sensitivity measured using cooling scores was assessed in a DED and
control group and was compared with mechanical sensitivity threshold using a Cochet-
Bonnet esthesiometer. Treatment with a lubricant containing TRPM8 agonist (0.01%
menthol, a compound that activates cold thermoreceptors) or lubricant alone was
applied bilaterally in a cross-over design. While there was no difference in mechanical
sensitivity, the DED group reported a higher cooling response and score compared to
controls, suggesting mechanical and cold sensitivity may be differentially impacted in
DED. 704 The duration of disease was associated with a greater cooling response,
suggesting that nociceptor activity may change over disease duration. Taken together,
these results suggest abnormal pain and conceivably cold stimulus processing in
different DED subtypes and that different qualities of stimulus processing and other
ocular surface sites, may help to understand neurogenic changes in different subtypes
of DED.
Increased sensitivity (reduced mechanical threshold) has been reported after treatment
of DED or GVHD with autologous serum tears, 753 Sjögren DED with cyclosporine, 754
and short TBUT DED with diquafosol (Table 8).755 This underscores the dynamic nature
of corneal sensitivity, highlighting changes in the short term, with disease, and with
treatment, the implications of which need further study.
The relationship between corneal nerve structure and function is equivocal. Nerve
structure was not consistently associated with gross corneal sensitivity using a cotton
wisp test, where a significant relationship was only noted between corneal sensitivity
and corneal nerve fiber area.756 In a diabetic population, there was a stronger
relationship between anatomy and function. 757 These data highlight that relationships
between corneal nerve anatomy and function may vary with disease type and that
relying on structural findings alone may not suffice in evaluating nerve functional health.
79
Given the importance of understanding nerve function in DED, there have been efforts
to develop new esthesiometers aimed at improving precision, range, stimulus type and
portability, to facilitate routine clinical use. These are reviewed in the TFOS DEWS III
Diagnostic Methodology Report. 1 Briefly these have included non-contact airjet
esthesiometers, 758,759 liquid jet esthesiometers, 705,760,761 and a single-use filament
mechanical esthesiometer (Kerasense, Dompè farmaceutici SPA, Italy). 759,762 While the
initial reports on these esthesiometers are encouraging 705, further research is
necessary to explore their clinical utility, particularly in individuals with DED.
80
6.9±16.4 mm,
p<0.01) which
correlated with
subjective pain
scores (scale 0-4;
r=0.24, p<0.05)
749
USA DED (DEQ ≥6) Corneal detection Mean corneal
thresholds in DED detection threshold
N= 403 (Belmonte of 87 ± 46 mL/min.
esthesiometer) 13% of participants
with hypersensitivity
and 11% with
hyposensitivity
Neither lowered
detection thresholds
753
USA DED (Ocular GVHD; Corneal sensitivity Mean corneal
n=20) tested after 12 sensitivity increased
weeks of 20% AST at 12 weeks of 20%
use (Cochet- AST in 17 patients
Bonnet) with ocular GVHD
(31.1±23.8 to
51.6±12.6mm,
p=0.001)
DED= dry eye disease; GVHD = graft versus host disease; TBUT = tear film breakup time;
NCP= neuropathic corneal pain; AT = artificial tears; AST = autologous serum tears
81
sensitivity assessment into routine clinical practice could enhance the management and
therapeutic outcomes for patients with DED. Most studies have focused on corneal
sensitivity only and there is limited evidence for other ocular surface regions including
the bulbar and palpebral conjunctiva and lid margin, which may also contribute to ocular
symptoms. A commercial, quantitative and suitably sensitive, non-contact esthesiometer
is urgently needed along with evidence-based normative and reference values for
application in different subtypes of DED.
The TFOS DEWS II report emphasized the role of neurosensory abnormalities in the
etiology of DED and the need to evaluate somatosensory functions in its assessment.
720
The topical anesthetic challenge has emerged as an important tool for assessing
neurosensory abnormalities. This simple and rapid test can differentiate between pain
originating from peripheral nociceptor activation versus central (or non-ocular surface)
mediated pain arising from proximal sensory pathways or the central nervous system.
720
The test involves applying a drop of topical anesthetic (e.g., 0.5% proparacaine
hydrochloride) to the ocular surface and evaluating pain relief. Pain relief typically
indicates peripheral neuropathic or nociceptive causes of pain, as proparacaine
stabilizes nociceptor neuronal membranes, impeding initiation and conduction of nerve
impulses. 763 In contrast, the absence of relief suggests a central or non-ocular surface
etiology of pain, while partial improvement may indicate a mixed component to pain. 764
However, it is important to note that in patients with complete pain relief, the test cannot
differentiate between pain due to peripheral nerve abnormalities or nociceptive causes
like DED. 763 It is further not informative if no pain is present prior to anesthetic
placement (in individuals with pain that waxes and wanes).
Persistent pain after anesthesia can indicate central abnormalities in pain processing
pathways. 765 Specifically, individuals with persistent pain after topical anesthesia often
exhibit other features of somatosensory dysfunction. A study of veterans with DED
symptoms were categorized based on anesthetic challenge response. Those with
persistent pain had greater discordance between signs and symptoms and lower non-
ocular cutaneous cold and hot thresholds (see Section 9.7) compared with those where
ocular pain was reduced following topical anesthesia. 766 These data suggest increased
cutaneous sensitivity both at a site innervated by the trigeminal nerve (forehead) and at
a distant site (forearm), supporting the contribution of central mechanisms in individuals
with persistent ocular pain following anesthesia. Similarly, individuals with DED who
experience with central-dominant pain had higher ocular and non-ocular pain scores
compared with those who had greater reduction in pain with topical anesthesia
(peripheral-dominant).767 These findings have therapeutic implications as treatments for
peripheral vs centrally mediated pain vary. For instance, patients with peripheral
82
dominant pain may benefit more from anti-inflammatory and topical neuromodulation,
while those with central pain may benefit from systemic neuromodulation.763 Further
studies are needed, however, that examine whether results of diagnostic tests can be
used to predict therapeutic responses.
The anesthetic challenge is a simple and accessible test that can aid in the evaluation
of somatosensory (dys)function in DED. When used appropriately, it can help identify
the location of pain generation which may assist in formulating a personalized treatment
plan for individuals with ocular pain.
Pain, aftersensation
and temporal
summation of cold
pain and hot pain
stimuli at the forearm
769
United Concordant Intensity rating of Intensity rating for
States DED (n=25) pressure pain at the pressure pain and
thumbnail auditory tones were not
Discordant statistically different in
83
DED (n=23) discordant DED.
Total DED (n
=48) Intensity rating of
auditory tones in both Pressure pain and
Controls (n= ears auditory sensitivity were
26) not correlated with DED
symptoms.
770
United DED (n=326) Detection threshold Aftersensations of cold
States for vibration, cool, pain and hot pain at the
warm, cold pain, and forehead and forearm
hot pain at the positively correlated
forearm with DED discordance
scores. Intensity ratings
at threshold of cold pain
and hot pain positively
Pain, aftersensation correlated with DED
and temporal discordance scores.
summation of cold
pain and hot pain
stimuli at the forearm
771
United LASIK (n=43) MPT at the forehead MPT at the forehead
States and forearm negatively correlated
with baseline ocular
pain.
Temporal summation
and aftersensation
intensity of MPTS positively
mechanical pain at cordrelated with
the forearm baseline DE symptoms.
Presence of
aftersensations pre-
LASIK was a predictor
for chronic DE
symptoms at 6 months
post-LASIK.
772
United DED (n=235) Aftersensation Intensity of ocular pain
States intensity of cold pain due to light was a
84
and hot pain at the predictor for the
forearm presence of
aftersensations.
DED = dry eye disease; HTPS = hot pain temporal stimulation; MPT = mechanical pain
threshold; MPTS = mechanical pain temporal summation
Hot and cold thermal stimuli (hot and cold) are frequently used to evaluate
somatosensory (dys)function. Despite variation in quantitative sensory testing protocols,
increased responsiveness to hot pain has been noted in individuals with DED where
there are increased symptoms disproportionate to the signs (measured as a
discordance score). 768 DED discordance score positively correlated with hot pain
aftersensation intensity at the forehead. 770 These studies support the finding that
abnormal perception of hot pain at sites near and distant to the eye correlate with DED
symptoms in the absence of ocular surface signs and that the altered perception may
arise from central abnormalities.
85
fMRI studies have also studied the neural impact of various therapies used to treat
ocular pain. In a study examining the impact of botulinum toxin A administered to the
forehead, frontalis, procerus and corrugators, showed that in those individuals who
reported reduced unpleasantness scores when viewing the light stimulus during the
post-injection scan, exhibited activation in the spinal trigeminal nucleus in response to
light stimuli prior to injections and this was not evident in those who did not show an
improved response post-injection. 776 This suggests that photophobia may be driven by
different neural pathways, and that individuals with activity within the spinal trigeminal
nucleus during light stimulation may be the ones more likely to response to botulinum
toxin. Use of the FL-41 spectacle tint to reduce photophobia led to significant reductions
in light-evoked blood oxygen level dependent signals (used as an indirect measure of
neural activity) in the bilateral primary and secondary somatosensory, bilateral insular,
right temporal pole, precuneus, anterior cingulate cortex and paracingulate cortices as
well as bilateral cerebellar hemispheric lobule VI, although the responses in regions
associated with pain processing were not eliminated completely. 777 These findings
indicate that fMRI may play a role in predicting and monitoring responses to therapy and
suggest that more studies are needed to examine central mechanisms to pain in various
DED subtypes.
Ocular pain and sensation are intimately associated with the structural anatomy of
corneal nerves. This structure can be altered by trauma, surgery, DED, other systemic
diseases, natural aging, and incomplete or improper neural regeneration. These
alterations are pleomorphic but most commonly manifest as reduced nerve fiber density
and increased nerve tortuosity. Aberrant corneal nerve anatomy generally correlates
with altered sensation, increased ocular pain, and symptoms typical of DED. Functional
abnormalities of corneal nerves are also implicated in altered ocular sensation and
include changes in growth factor activity and increased expression of sodium channels.
Ocular pain, in some individuals can be driven by generalized dysfunction of peripheral
and central nervous systems, with studies supporting activation of the primary
somatosensory, insular, and anterior mid-cingulate cortices in individuals with chronic
ocular pain, which may be mitigated with certain therapies.
Research into this field is ongoing and future developments may include the elucidation
of specific pathways controlling corneal nerve development and regeneration,
exploration of neuroimmune crosstalk in DED, greater implementation of artificial
intelligence networks in processing large bodies of data, more sophisticated
assessments of peripheral and central nerve function, and targeted treatments to
address dysfunction in an individual patient.
86
10. Iatrogenic
10.1 Introduction
The TFOS DEWS II recognized the importance of this topic, and a specific report about
Iatrogenic DED was included.778 The impact of elective medications and procedures
was also highlighted in the "Lifestyle Epidemic: Ocular Surface Disease TFOS
Workshop".684 This review presents an update since these reports of the most common
iatrogenic causes of DED, including a summary of pathophysiology and
recommendations for management.
I. Drug-induced
A. Topical
B. Systemic
A. Refractive surgery
C. Cataract surgery
D. Lid surgery
E. Other surgeries
1. Conjunctival surgery
2. Glaucoma surgery
3. Vitreoretinal surgery
4. Strabismus surgery
5. Intrastromal corneal ring segment implantation
87
6. Others
A. Botulinum toxin
B. Crosslinking (CXL)
C. Cosmetic procedures
D. Others
V. Non-ophthalmic conditions
*Ophthalmic surgeries were extensively reviewed previously and are not included in this
update. 684,778
10.2.1 Prevalence
In a multicenter study conducted across four European countries with 9,658 patients,
over 40% of individuals treated for glaucoma reported symptoms of DED, such as pain
or discomfort during the application of eye drops, foreign body sensation, dry eye, and
burning.781. Furthermore, more than 20% of patients exhibited signs of blepharitis,
conjunctival hyperemia, or keratitis. These findings were more prevalent in patients
treated with preservative-containing eye drops. Similarly, a recent cross-sectional
comparative study with 320 patients concluded that glaucoma patients are more
affected by DED than non-glaucoma patients, showing a lower TBUT and greater
corneal staining in eyes with glaucoma using multiple eyedrops and daily doses.782
Another cross-sectional study with 101 patients undergoing anti-glaucoma treatment
detected signs and symptoms of DED in over 50% of the patients and advanced
changes in the ocular surface in 27% of the individuals examined.783 Reduced values on
the Schirmer test were observed in 61%, decreased TBUT in 78%, and staining of the
ocular surface in 22% of the subjects.
88
In a German study involving 20,506 glaucoma patients from 900 centers, the
prevalence of DED was higher in women (56.9%) than in men (45.7%), and this
difference was more pronounced in patients over 50 years old. The prevalence
increases with age and duration of glaucoma, and it occurs more frequently when three
or more medications are used.784 In another epidemiological study including 4,107
glaucomatous patients, the most frequently reported dry eye symptoms were: excessive
discomfort after applying anti-glaucoma eye drops (43%), pressure behind the eyelids
(40%); foreign body sensation (31%); dry eye sensation (23%); excessive reflex tearing
(21%); and eyelid itching (18%) of cases. These symptoms were significantly more
prevalent when using preservative-containing than preservative-free eye drops.785,786
According to TFOS DEWS II, various topical medications and excipients have been
implicated in DED. 778 (Table 11) Of particular concern is the use of chronic medications,
such as anti-glaucoma, antiallergic, and anti-inflammatory eye drops. However, there
needs to be more specific data on active compounds, as ophthalmic formulations are
often evaluated with preservatives, making it difficult to isolate the impact of the
medications, preservatives, and excipients. Also, common excipients in ophthalmic
preparations, such as solutions, ointments, suspensions, and emulsions, may cause dry
eye symptoms. 778 The chemical properties of the formulation, including
isotonicity/hypotonicity and pH, can influence the tear film and local tolerance after
application. 778
Topical drug-induced DED has mainly been studied in individuals in long-term treatment
for glaucoma and ocular hypertension. 778 Other than the presence of benzalkonium
chloride, the main risk factors for iatrogenic DED in anti-hypertensive topical drug users
are the treatment duration, higher IOP, and glaucoma severity. 787,788 The frequency of
mild or greater DED symptoms tended to increase with an increasing number of anti-
glaucoma medications. 788 Interestingly, patients on brimonidine performed the worst.
Patients on timolol reported pain induced by light, and those on latanoprost complained
of stinging. 788
89
Prostaglandin analogs (eg: Bimatoprost, Latanoprost, Travoprost)
10.2.3 Mechanism
Topical medications can affect the ocular surface through various mechanisms, causing
allergic, toxic, and/or immune-inflammatory effects or through chemical interactions with
different components of the ocular surface. 778 These effects can result from disruption
of the lipid layer of the tear film due to the detergent properties of the compounds,
reduced aqueous secretion, damage to the ocular surface epithelium, neurotoxic effects
on the corneal nerves, and injury to the eyelids, including the skin or meibomian glands.
789-792
Indirect effects may also arise if chronic inflammation induced by the topical medication
stimulates the precursors of the keratinized envelope, leading to the entrapment of
mucous cell contents and squamous metaplasia. 790 Keratinization of the eyelid margins
can further worsen MGD. 789 Additionally, destruction and/or dysfunction of goblet cells,
which are increasingly recognized as having an important role in immunomodulation of
the ocular surface, can exacerbate chronic inflammation. 793
90
establish, especially when treatment is essential for severe or vision-threatening
conditions. This is particularly true in glaucoma, where treatments are usually prolonged.
Increased tear osmolarity has also been observed in patients using preserved eye
drops compared to preservative-free topical medications. 262 Once the tear film loses its
protective properties, the compromised tear film not only leads to symptoms of DED and
corneal damage but can also spread cytotoxic inflammatory mediators across the ocular
91
surface. Consequently, increased corneal epithelial permeability has been observed in
DED with more significant deterioration when using artificial tears containing
benzalkonium chloride compared to preservative-free drops. 800 Changes in the tear film
may, therefore, initiate a series of biological alterations on the ocular surface, leading to
subsequent neurogenic inflammation and further tear film impairment, creating a vicious
cycle. New preservatives, such as Polyquad, Purite, and SofZia, have significantly
reduced cytotoxic effects compared to benzalkonium chloride. However, their impact on
DED patients needs to be further investigated. 262,684
Preservative-free eye drops are associated with better ocular surface health and higher
tolerability. 800 A systematic review of randomized clinical trials comparing beta-blockers
with and without preservatives in patients with glaucoma or ocular hypertension found
that, although the difference in intraocular pressure reduction was not clinically relevant,
preservative-free eye drops performed better in tear film breakup time and Schirmer test.
800
Additionally, in patients with DED, preservative-free eye drops demonstrated a
significant reduction in ocular inflammation symptoms and increased antioxidants in the
tear film compared to preserved eye drops. 801 Another observational study in patients
with DED who switched from preserved drops to preservative-free drops containing
hyaluronate showed significant improvement in the OSDI and reduced frequency of
superficial punctate keratitis. 802
The use of preservative-free prostaglandins has also shown significant benefits for
ocular surface health compared to preservative-containing versions. The PRAMOS
study demonstrated that patients using preservative-free prostaglandin analogs had a
lower prevalence of conjunctival hyperemia and corneal staining compared to those
using preserved eye drops. 803 A prospective study showed that switching from
preservative-containing prostaglandin-timolol fixed combinations to a preservative-free
formulation resulted in a significant improvement in ocular surface disease symptoms,
and a reduction in conjunctival hyperemia. 804 Another randomized controlled trial
revealed that transitioning from preservative-containing glaucoma therapies to
preservative-free formulations improved both ocular surface health and intraocular
pressure (IOP) control. 589 These findings are supported by studies indicating that the
absence of preservatives, such as benzalkonium chloride, prevents chronic ocular
surface toxicity, leading to improved treatment adherence and long-term efficacy. 805,806
The first step is to investigate which medication is causing DED and try to stop its use.
This subtraction can be challenging when discontinuing the treatment, which presents a
risk to the eye's health. Sometimes, multiple drugs and components are involved, or
adverse effects appear long after treatment initiation, making identification of which is
92
causing DED even more difficult. 778 In some cases, eye drops may be necessary to
treat dry eye symptoms, but adding preserved drops to eyes already suffering from
dryness caused by other drops may be ineffective and worsen the condition. Once the
responsible drug is identified, efforts should be made to discontinue using the
preservative or medication. Considering that toxicity is dose-dependent, reducing the
number of preserved eye drops can minimize adverse. 778 In glaucoma patients, more
invasive and definitive options such as laser trabeculoplasty or surgery may replace or
diminish the use of topical medication when the ocular surface and QoL are very
compromised. 778
Conceivably the adverse effects of toxic preservatives should reduce with time. There is
a significant shift in first-line primary open angle glaucoma therapy in most countries,
from topical therapies towards laser therapies. 807 Preservative-free therapies are
increasingly available and there is good evidence for their ocular surface benefits. A
significant issue related to preservative-free eye drops is their cost, as they are
generally more expensive than preserved drops. This is primarily due to higher
production costs and patent fees associated with preservative-free drops. Allocation of
healthcare and out-of-pocket costs for patients vary with region and insurers or health
system payers, and preservative-free alternatives may not be available in certain
jurisdictions.
10.3.1 Prevalence
At least 1/5 of the best-selling systemic drugs in the US have been associated with the
development of DED. In the elderly population, 62% of DED cases are related to
systemic medications, including nonsteroidal anti- inflammatory drugs (NSAIDs),
diuretics, vasodilators, analgesics/antipyretics, antiulcer agents, sulfonylureas, cardiac
glycosides, anxiolytics/benzodiazepines, anti-infectives, antidepressants/antipsychotics,
hypotensive agents, and antihistamines. 809
93
In a secondary analysis of the Dry Eye Assessment and Management Study to evaluate
whether systemic medication use is associated with DED severity, the authors found
that 160 (30%) of the 535 participants used medications for hypertension, 129 (24%)
used statins, 118 (22%) used antidepressant medications, 117 (22%) used
antihistamines, and 16 (3%) used systemic corticosteroids. 810 A multivariable analysis
demonstrated that antihistamines and corticosteroids were associated with the highest
OSDI score among these systemic medications. Users of seizure medications had a
higher composite signs severity score compared to non-users. Compared to non-users,
antihistamines, aspirin, and vitamin D3 users had significantly worse average TBUT.
Unexpectedly, MGD scores were worse in users of vitamin D3, although this could be
confounded if those users of vitamin D3 had vitamin D deficiency. Users of diuretics had
significantly better scores for MGD compared to non-users. 810
10.3.2.1 Tamsulosin
Tamsulosin, a medication that blocks alpha-1 receptors, is widely prescribed for treating
benign prostatic hyperplasia, but is also utilized in managing ureteral stones, prostatitis,
and female voiding dysfunction. Epidemiological research suggests a link between DED
and benign prostatic hyperplasia itself, as well as the medications used to treat it,
including tamsulosin (See Section 5.5.1).
A large class of systemic medications that lead to the signs and symptoms of DED are
those with anticholinergic activity. The anticholinergic class covers a wide range of
therapeutic drug categories, including antidepressants, antipsychotics or neuroleptics,
antiparkinsonians, H1 antihistamines, decongestants, and antispasmodics. 812
Anticholinergic medications, such as those used for overactive bladder, have been
linked to a reduction in tear film breakup time and an increase in DED symptoms,
including burning and foreign body sensation. Studies have shown that medications like
94
solifenacin can significantly worsen dry eye symptoms and signs compared to
placebo.813,814
Oral antihistamines and anticholinergic drugs are commonly used to treat allergies and
rhinitis. These medications, especially in combinations like pseudoephedrine and
cetirizine, can increase the sensation of dryness in the eyes and mouth. 815
Results of the comparison of DED symptoms, measured by the OSDI, between users
and non-users of medications showed that patients who used antihistamines had worse
TBUT and OSDI scores compared to non-users. Anticholinergic drugs, such as
antihistamines, contribute to DED by acting on the G-protein coupled muscarinic
receptors of the lacrimal gland acini and mucus-producing conjunctival goblet cells,
affecting the production of the aqueous and mucous components of the tear film.
Furthermore, functional cholinergic receptors have been identified in the epithelial cells
of the meibomian glands, leading to decreased tear film stability. Thus, despite being
commonly used to treat allergy-related eye symptoms, antihistamines confer a high
anticholinergic burden, contributing to the development and worsening of DED signs
and symptoms.268,810,816
10.3.2.3 Isotretinoin
Recent studies utilizing standardized tools, such as the OSDI, indicate that DED
symptoms are more commonly found in patients treated with Isotretinoin. The drug can
lead to glandular atrophy, ductal keratinization, and small fiber neuropathy, which may
reduce corneal sensitivity.818,819 Other studies have reported severe DED symptoms
and decreased tear film stability among patients who start using Isotretinoin, associated
with reduced TBUT, lower Schirmer test value, and decreased central corneal
thickness.275,820 Additionally, a few studies show that even after discontinuation of the
medication, these signs and symptoms can persist for months post-treatment.275,820 The
adverse effect of Istotretinoin on meibomian gland lipid production is likely to be through
suppression of the peroxisome proliferator-activated receptor γ pathway which inhibits
meibocyte differentiation and meibum characteristics. 821Topical isotretinoin is also
recognised as a risk factor for iatrogenic DED (see Section 6.6.4). 822
95
10.3.2.4 Chloroquine / Hydroxychloroquine
96
can significantly impact the efficacy and potential side effects of these drugs. For
instance, when administered topically, corticosteroids are effective in treating DED. 830
Additionally, many users of systemic corticosteroids have underlying autoimmune
conditions that are independently associated with DED, making it challenging to isolate
whether the worsening of DED signs and symptoms is due to medication use or the
progression of the underlying disease. 96 More prospective studies are needed to
determine the effects of systemic corticosteroid use on DED. 831
10.3.2.6 Antibiotics
Antibiotics are commonly used to treat various infections, but few studies have been
conducted regarding their impact on the ocular surface. Antimicrobials, particularly
tetracyclines and macrolides, are generally used in the treatment of MGD and have
shown promising results. 832,833
The correlation between DED and antimicrobial use is sometimes indirect, as antibiotics
can cause Stevens-Johnson syndrome. Some studies have shown that antibiotics are
notable risk factors for this condition, potentially leading to severe damage to the ocular
surface. 834,835
97
Selective serotonin reuptake inhibitors can impact the ocular surface by impairing tear
film stability. 840 Conversely, serotonin-norepinephrine reuptake inhibitors, effective in
treating chronic pain syndromes, may help alleviate DED symptoms. Patients using
these inhibitors often have lower OSDI scores, suggesting they may be safer for the
ocular surface compared to other antidepressants. Thus, while DED is common among
antidepressant users, it may be better managed with serotonin-norepinephrine reuptake
inhibitors. 841
Lithium, widely used for the maintenance and acute treatment of bipolar disorders, has
a narrow therapeutic index and various side effects, making its management
challenging. Although the adverse effects of lithium are well-documented, its ocular
impacts are less understood, which may affect patient compliance. Ocular side effects
associated with lithium use include exophthalmos, abnormal eye movements, ocular
myasthenia gravis, papilledema, photophobia, and alterations in the tear film, which can
lead to DED. 842
Given the complexity of these interactions, co-management of individuals with DED and
medicated for mental health conditions is advisable.
The relationship between contact lens wear and DED is complex. Contact lens wear is
recognised as a consistent risk factor for DED (Section 6.6). Ocular symptoms occur
more commonly in contact lens wearers than non-wearers and many large
epidemiological studies have used ocular symptom reporting as a surrogate for DED. 149
Signs such as corneal staining and tear film instability, due to the partitioning of the tear
film during wear occur. 846 Signs of meibomian gland alterations are also more common
in contact lens wearers than in age-matched non-wearers. 847 Contact lens discomfort is
characterized by episodic ocular symptoms of discomfort and dryness which resolve
98
when the contact lens is removed, as distinct from those in DED, 284 however this
distinction is not consistently considered when reporting symptoms in contact lens
wearers. A further complexity is that other complications of contact lens wear may also
lead to very similar ocular symptom-reporting. 847 Contact lens wearers may also have
existing or subsequently develop, DED. This has led to the use of series of descriptors
which are often used interchangeably, including contact lens discomfort, contact lens-
induced dry eye and contact lens-associated dry eye. With these caveats, this section
will endeavour to summarise the evidence for prevalence of, mechanisms underlying
and remedial strategies for DED associated with contact lens wear.
The prevalence of symptoms of DED in soft contact lens wearers in a series of recent
prospective and retrospective studies in select population groups ranged between 31%
to 77%. 848-850 A clinical diagnosis of DED by an eyecare professional was made in
24% of office workers in a Chinese sample 850 and in 14% of University students
wearing contact lenses in Thailand. 851. The frequency of DED in scleral lens wearers
has been more difficult to determine. These are mostly used for medical and therapeutic
indications, and an estimate of 56% of scleral lens wearers had severe DED symptoms
based on OSDI. 852,853 There is a lack of large-scale epidemiological studies on the
prevalence of DED associated with orthokeratology lenses. In a small retrospective
study of contact lens complications conducted at a tertiary hospital in China, 24% of
orthokeratology lens wearers presented with DED. 854 Understanding the pathogenesis,
and provision of mechanism-oriented treatment of DED in contact lens wearers is
important.
10.4.2 Mechanism
Contact lens use causes a variety of biophysical and biochemical changes in the tear
film, as well as alterations in the ocular surface structure and function which are
summarized in Table 12. These mechanisms collectively lead to reduced tear film
stability, increased tear evaporation, and ocular surface damage, 846,847 and likely
contribute to DED in contact lens wearers.
Rigid and soft contact lens wear increase tear film evaporation,855 although they interact
differently with the ocular surface. Mechanical interactions are different between rigid
and soft contact lenses due to material and fitting characteristics. 847,856,857 Upper lid
wiper epitheliopathy is more common in rigid than soft lens wear. 858 There is a higher
frequency of incomplete blinking, 859 leading to uneven tear distribution and
exacerbation of dry eye symptoms. Corneal staining in the 3 to 9 o'clock positions is
common among rigid lens wearers,860,861 attributed to thinning of the tear film 862
99
adjacent to the lens edge, whereas soft contact lens wearers more often exhibit inferior
arcuate cornea staining.861
Orthokeratology lenses share some mechanisms for DED with soft and rigid contact
lenses, but the unique mechanism of these lenses is related to their reverse geometry
design, 854 which not only alters corneal shape but also significantly affects the density
and distribution of corneal nerve fibers, 864,865 which may not fully recover shortly after
cessation of wear. 866,867
The evidence for changes in corneal sensitivity underpinning sensation report in contact
lens wear is limited. Contemporary rigid corneal and soft lenses do not appear to alter
corneal sensitivity, although a reduction in central corneal mechanical sensitivity is
observed in orthokeratology lens use 866,868 Reduced corneal sensitivity may reduce tear
secretion and blink frequency, thereby exacerbating symptoms of DED. 867 There is a
substantial augmentation in tear inflammatory mediators in orthokeratology lens wear,
such as IL-17A, IL-6, and prostaglandin E2. 864 The TFOS Contact Lens Discomfort
neurobiology report described the multifactorial effects of contact lens wear, include
mechanical, cooling, drying, change in osmolarity and chemical impacts on ocular
surface neurosensory processes. 869 Altered sensory processing of cooling stimuli
delivered to the cornea has recently been reported in symptomatic soft contact lens
wearers suggesting nerve sensitisation or maladaptation in the absence of sensitivity
changes, 870 which is intriguing but causality for DED has not been demonstrated.
Table 12. Effects of contact lens wear on the tear film and ocular surface
100
Tear meniscus volume Reduced
Lipid layer spreading Reduced
Tear stability Reduced
Structural ocular surface alterations
Conjunctival goblet cell density↓ Reduced
MG expressibility ↓, Reduced
MG obstruction and atrophy Increased
Basal corneal nerve density↓, nerve tortuosity↑: Reduced
Ocular surface sensitivity Corneal sensitivity unchanged in SCL use;
altered sensory processing; conjunctival
sensitivity increased
Corneal sensitivity reduced in OK use
Ocular surface friction and lid wiper epitheliopathy Increased
Biochemical tear film alterations
Tear cholesterol Increased
Malondialdehyde & 4-hydroxy 2-nonenal Increased
Beta-2 microglobulin Reduced
Proline rich protein 4 Reduced
Lacritin Reduced
Lipocalin 1D1 Reduced
Secretory IgA Effects equivocal, may depend on duration
of wear and wear modality
Albumin Increased
Deleted in Malignant Brain Tumors-1 Increased
Prolactin inducible protein Increased
MUC5AC Reduced
Inflammatory tear film/ocular surface changes
Epithelial immune cells Increased at ocular surface in SCL wear,
increased antigen capture capacity. Density
reduced at central cornea in OK
Tear cytokines IL-7,8,13,15 Increased
101
unpreserved lubricants, with or without lipid additives. 847 873 874 Discontinuing from lens
wear or reducing wear time may be successful 855 unless there is underlying DED.
In contact lens wearers with MGD and consequential evaporative DED, there is
evidence for the benefits of treatments including IPL 875, microblepharon exfoliation 876
or thermal pulsation, 877 including improved TBUT, and symptoms of DED. Low level
light therapy or photobiomodulation therapy has been used in the treatment of MGD and
evaporative DED 2, but there is no high-level evidence for benefit in contact lens
wearers, although an observational study without a control group exists in wearers with
discomfort. 878 While there are no pharmacological treatments approved for MGD in
contact lens wearers, a phase 1 879 and phase 2 880 clinical trial both demonstrated
benefit in improving comfortable wear time and meibomian gland signs in a population
of symptomatic wearers with MGD, using a topical selenium sulphide treatment.
10.5 Procedures
Paradoxically, botulinum toxin A has been used in treating DED. 881 In the literature,
BTX-A was injected into the periorbital area including the medial portion of the
orbicularis muscle and not solely into the lateral canthal region, for the treatment of
existing DED. The mechanism of action of botulinum toxin on lacrimal drainage in such
instances has been suggested to be due to a paralysis of the orbicularis oculi muscle
around the canaliculi with a decreased compression as well as weakness of apposition
of the puncta during blinking. 882,883
10.5.1.1 Mechanism
Dry eye due to botulinum toxin type-A injection for treatment of blepharospasm or after
blepharoplasty and peri-orbital surgery has been previously reported in literature. The
mechanisms by which DE is induced are multifactorial:
Botulinum toxin blocks acetylcholine release within the lacrimal gland, disrupts
neuromuscular junctions, and inhibits parasympathetic nerves, thereby suppressing
both basal and reflex tear secretion. 884 This leads to a significant decrease in TBUT
and Schirmer test results. 882,883,885
102
toxin-induced paralysis of the preseptal orbicularis oculi and Riolan's muscle weakens
eyelid closure and reduce meibum secretion, thereby decreasing lipid layer thickness
and tear film stability. 887 However, further studies are needed to examine the long-term
effects on the meibomian glands.
Botulinum toxin injections may trigger inflammatory responses at the ocular surface and
in the lacrimal glands, such as increased release of cytokines (e.g., TNF-α and IL-1β),
888
which may further impact tear stability and tear secretion.
103
10.5.2 Corneal collagen crosslinking
10.5.2.1 Mechanism
Corneal collagen cross-linking (CXL) is a treatment that uses ultraviolet A light and
riboflavin (vitamin B2) as a photosensitizer to strengthen the cornea. This technique
reinforces the chemical bonds within the cornea, thereby halting or delaying the
progression of corneal ectatic diseases such as keratoconus.890,891 Both epithelium-on
and epithelium-off CXL have been reported to have a positive effect on DED, and may
improve tear film homeostasis and reduce DED symptoms in patients with
keratoconus.892
Paradoxically, CXL can also induce DED symptoms through multiple mechanisms.
Firstly, epithelial removal and delayed healing may induce DED.893-895 This delay can
affect the uniform distribution of tears and the corneal defences,893,896,897 by
compromising tear film stability708,898 and making the ocular surface more prone to DED
symptoms.
CXL may also reduce corneal nerve density, 899 impairing corneal sensory nerve
function and decreasing corneal sensitivity, which in turn affects the tear secretion. 899
However, studies indicate that in most patients subbasal nerve regeneration occurs
within 2-3 months after CXL,899 reaching preoperative levels in 6-12 months. 900,901
Additionally, although CXL does not directly alter meibomian gland morphology, 902 the
effects on corneal sensory nerves 902, the use of eyelid speculums during surgery, post-
CXL inflammatory responses with elevation of IL-6, and medications used post-surgery
902
can indirectly affect gland function and secretion quality.
To effectively manage DED that may arise after CXL, a comprehensive approach is
essential. Firstly, bandage contact lenses, due to their material and characteristics, help
protect exposed nerve endings and the corneal epithelium, reducing frictional damage
from blinking and thereby alleviating dry eye symptoms. 903
Secondly, during the postoperative phase, the use of corneal protectants, tear
substitutes, and tear secretagogues is important. These medications should be selected
based on their ability to replace the mucin glycocalyx lost from the corneal epithelial
cells, promote the restoration of the mucin gel on the epithelial cell surface, enhance the
adhesion of the tear film to the corneal epithelium, and aid in the recovery of the lipid
layer of the precorneal tear film. 903,904
104
Lastly, advancements in modern CXL technology also help reduce the risk of
postoperative complications. These improvements include transepithelial cross-linking,
localized and personalized techniques, and accelerated procedures, which aim to
enhance surgical outcomes and minimize corneal manipulation time. 898,905-907
The use of eye cosmetic products and procedures represent a lifestyle challenge that
may exacerbate or promote the development of ocular surface and adnexal disease.
This topic has been recently explored in detail. 263
10.6.1 Radiotherapy
10.6.1.1 Mechanism
DED is associated with radiation therapy for head and neck cancer, 908,909 temporal
tumors, breast cancer, and Graves' eye disease. This condition often results from direct
damage to tissues around the eye.909-911 The incidence of radiation-related dry eye
depends on the type of radiation therapy, tumor location 912,913 and radiation
dose.909,914,915
105
10.6.1.2 Recommendations for management
The management of radiation-related DED can be divided into three key approaches:
1. Preventive measures to minimize the risk such as limiting the radiation dose to
the lacrimal glands, ideally keeping the dose below 30 Gy.914,915,921,922
2. Proper patient positioning, eye shielding, and adequate patient immobilization
during radiotherapy sessions to reduce radiation- related toxicity.912
3. Therapeutic interventions for DED. 2
Long-term management is essential for maintaining good ocular health and QoL.
Regular ophthalmologic examinations and assessments to monitor changes in DED
allow for timely adjustments in treatment plans and interventions. A multidisciplinary
team, including ophthalmologists and oncologists, can provide comprehensive care and
personalized treatment plans, thereby maximizing QoL.
10.6.3.1 Mechanism
GVHD is a common complication following human leukocyte antigen (HLA)-matched
allogeneic hematopoietic stem cell transplantation. GVHD can affect various organs,
including the skin, gastrointestinal tract, liver, lungs, and eyes, with dry eye being the
most common manifestation of ocular GVHD. 923,924 Ocular GVHD is seen in 60-90% of
transplant recipients, primarily manifesting as secondary inflammation and fibrosis of
the lacrimal and meibomian glands. 925,926 The mechanisms of DED in GVHD include
the following aspects:
1. Lacrimal Gland Fibrosis: Fibrosis of the lacrimal gland is a major cause of GVHD-
related dry eye. Studies have shown the presence of activated CD34+ fibroblasts
in the lacrimal glands of GVHD patients, with T-cell infiltration around the ducts,
thickening of blood vessels and duct basement membranes correlating with the
severity of fibrosis. 927,928
2. Renin-Angiotensin-Aldosterone: Angiotensin 1 type 1 receptor antagonists, such
as valsartan, can reduce fibrosis, decrease inflammatory cell density, and
increase tear secretion. 929 Angiotensin-mediated fibroblast activation and
upregulation of TGF-β expression via the angiotensin 1 type 1 receptor are the
primary mechanisms of fibrosis. 930
106
3. Immune-Mediated Inflammation: In GVHD patients, increased density of T cells
and dendritic cells 931 in the lacrimal glands, conjunctiva, and cornea contribute to
inflammation and fibrotic responses. 932
4. Cellular Senescence: Chemotherapy and radiotherapy-induced DNA damage
and the inflammatory environment activate senescence-related molecular
pathways. 933 Stress-induced cellular senescence promotes the secretion of
various cytokines (e.g., IL-1β, IL-6, IL-8), forming the "senescence-associated
secretory phenotype", which exacerbates inflammation. 934
GVHD patients often exhibit significant abnormalities in the meibomian glands, 935
leading to alterations in the tear film lipid layer and increased tear evaporation. 936
Corneal and conjunctival changes commonly include punctate keratopathy, filamentary
keratitis, and epithelial defects, 937 accompanied by decreased corneal nerve fiber
density, increased dendritic cell density, conjunctival epithelial squamous metaplasia,
and reduced goblet cell density. These changes collectively contribute to the onset and
progression of DED. 931,936
Iatrogenic causes underpin a significant proportion DED and may arise from topical and
systemic medications and a range of ophthalmic and non-ophthalmic surgeries.
Lifestyle choices such as contact lens wear, cosmetics and cosmetic procedures also
contribute to iatrogenic DED. Improved understanding of the range of causes and how
these contribute to different subtypes of DED and risk factors for more severe disease is
107
likely to help reduce their impact. Evaluating and managing underlying DED pre- and
post-intervention will also better support these patients. A challenge in iatrogenic
disease is understanding the impact of an intervention compared with progression of the
primary condition. This report has also identified the need to better disaggregate the
effects of topical medications alone from their preservatives and other excipients. In
DED following use of systemic medications, there is greater certainty in the adverse
effects of isotretinoin in DED and the persistence of effect once the medication ceases.
While depression and antidepressants broadly are independent risk factors for DED, it
is recognised that some classes, such as serotonin-norepinephrine reuptake inhibitors
may improve ocular symptoms and there should be further exploration of the impact of
systemic corticosteroids on the ocular surface. The natural history of DED associated
with lifestyle choices such as contact lens wear and particularly orthokeratology remains
unclear.
11.1 Introduction
The major international markets for medicines are the US, EU and Japan. These three
regions have consistent regulatory frameworks for approval of medicines following the
International Council for Harmonisation of Technical Requirements for Pharmaceuticals
for Human Use guidelines. 948 There are some variations in evidentiary standards
between these jurisdictions in clinical trials designs and features. As an example, for
new DED treatments, the EU requires a six-month time frame for primary efficacy
studies, rather than the three-months required by the FDA. In addition, EU typically
requires standard of care as one of the trial arms, in addition to vehicle as control,
where using artificial tears as standard of care. Artificial tears are considered devices in
the EU but not in the US or Japan. As far as possible, this report will provide examples
of treatments approved from of these three regions.
The development of novel treatments for DED accelerated in the US and EU following
release of both the TFOS DEWS II report in July 2017 205 and the FDA draft guidance
for industry in December 2020. 949 Five new treatments for DED were approved
between August 2018 and June of 2024 in the US, one new treatment was approved in
the EU and one new treatment was approved in Japan. Eight treatments were approved
before July of 2017, two in the US and six in Japan. 950-952 While prior TFOS DEWS
reports focused on challenges hampering the design and success of trials in DED, 953
this report will focus on how recommendations from the TFOS DEWS II report
translated into innovations in trial design, an independent analysis of trial failures
released in 2012, across ten therapeutic areas, 198 and how flexibility in application of
the FDA draft guidance have improved success rates for bringing novel treatments to
108
patients. Innovations in trial design include better matching of treatment mechanism of
action to patient sub-populations and more stringent control on concomitant treatments
and sources of trial error. 951,952,954-958
109
11.2 Approvals for DED
The recommendations for trial design from TFOS were first published in the TFOS
DEWS report 961 and were followed by the TFOS DEWS II report in 2017. 953 A
cyclosporine ophthalmic emulsion 0.05% was approved in 2003 in the US and is
indicated for an increase in tear production in patients whose tear production is
presumed to be suppressed due to ocular inflammation associated with
keratoconjunctivitis sicca. Sodium hyaluronate 0.1% and Mini 0.1% and 0.3% were
approved in 1995 in Japan for conjunctival epithelial disorders including sicca syndrome.
The TFOS DEWS report described the approval period followed by interest in the
development of additional anti-inflammatory and immunomodulatory drugs for the
treatment of DED. This time was marked by the failure of numerous follow-on
compounds and exploration of how the better control of trial design, endpoints and
operational excellence could potentially lead to a greater success rate in DED trials.
Lifitegrast ophthalmic solution (5%) was approved in the US in 2016 and is indicated for
treating the signs and symptoms of DED in adults over the age of 17 years. In Japan a
sodium hyaluronate ophthalmic solution (0.3%), Diquafosol ophthalmic solution (3%),
and Rebamipide ophthalmic suspension (2%) were approved between 2010 and 2012
for conjunctival epithelial disorders including sicca syndrome (dry eye) and
subsequently just dry eye. The TFOS DEWS II report described the approval followed
by interest in exploration of how matching the mechanism of action of a proposed
therapy to a potentially responsive population in which the treatment is likely to
demonstrate efficacy; the inclusion of biomarkers and/or surrogate markers of disease;
and recognition of strong placebo/vehicle effects that if properly controlled could
potentially lead to greater success rates in DED trials. 953 These concepts were
captured in the TFOS DEWS II report, trials for novel compounds and the FDA’s
guidance document on trial design for DED; following which a period of successive
approvals for additional DED treatments was observed (Figure 4 and Table 13).
110
Figure 4: Approved treatments for Dry Eye Disease
111
Table 13. Approved products in major markets
Cyclosporine RESTASIS® 2003 USA calcineurin inhibitor Indicated to increase tear production in patients
ophthalmic emulsion, immunosuppressant whose tear production is presumed to be suppressed
0.05% due to ocular inflammation associated with
keratoconjunctivitis sicca.
TM
Lifitegrast XIIDRA 2016 USA lymphocyte Indicated for the treatment of the signs and symptoms
ophthalmic solution, function-associated of DED
5% antigen-1 (LFA-1)
antagonist
Cyclosporine CEQUA™ 2018 USA calcineurin inhibitor Indicated to increase tear production in patients with
ophthalmic solution, immunosuppressant keratoconjunctivitis sicca (dry eye)
0.09%
Loteprednol EYSUVIS™ 2020 USA corticosteroid Indicated for the short-term (up to two weeks)
etabonate treatment of the signs and symptoms of DED
ophthalmic
suspension, 0.25%
Varenicline solution, TYRVAYA™ 2021 USA Nicotinic Indicated for the treatment of the signs and symptoms
0.03 mg nasal spray acetylcholine of DED
receptor agonist
Perfluorohexyloctane MIEBO™ 2023 USA semifluorinated Indicated for the treatment of the signs and symptoms
ophthalmic solution alkane of DED
Cyclosporine VEVYE 2023 USA calcineurin inhibitor Indicated for the treatment of moderate to severe
ophthalmic solution, immunosuppressant DED (VEVIZYE in Europe)
0.1%
Cyclosporine IKERVIS 2023 EU calcineurin inhibitor Treatment of severe keratitis in adult patients with
ophthalmic emulsion, immunosuppressant DED, which has not improved despite treatment with
0.1% tear substitutes
112
Purified sodium HYALEIN® 1995 Japan increasing tear film Indicated for the treatment of corneal and conjunctival
hyaluronate, 0.1% ophthalmic solution stability / corneal epithelial disorders (Keratoconjunctival epithelial
0.1% healing disorder resulting from the following diseases:
Intrinsic diseases such as Sjögren's syndrome,
Stevens-Johnson syndrome and sicca syndrome (dry
eye).
Extrinsic diseases caused by surgery, drugs, trauma,
contact lens wearing, etc.)
Purified sodium HYALEIN® 2010 Japan increasing tear film As above for 0.1% solution
hyaluronate, 0.3% ophthalmic solution stability / corneal
0.3% healing
Purified sodium HYALEIN® Mini 1995 Japan increasing tear film As above for 0.1% solution
hyaluronate, 0.1% ophthalmic solution stability / corneal
(preservative free) 0.1% healing
Purified sodium HYALEIN® Mini 1995 Japan increasing tear film As above for 0.1% solution
hyaluronate, 0.3% ophthalmic solution stability / corneal
(preservative free) 0.3% healing
113
Cyclosporine CEQUA® 2021 India calcineurin inhibitor Not known
ophthalmic solution, immunosuppressant
0.09%
Diquafosol sodium DIQUAS® 2017 China P2Y2 purinergic Indicated for DED diagnosed with corneal and
ophthalmic solution, receptor agonist conjunctival epithelial injury accompanied by tear
3.0% (6x Daily) abnormalities
Cyclosporine Zirun Cycloome® 2020 China calcineurin inhibitor Indicated to increase tear production in DED where
ophthalmic immunosuppressant tear production is presumed to be suppressed due to
nanoemulsion, ocular inflammation associated with
0.05% keratoconjunctivitis sicca
Diquafosol sodium RUNLIMING 2022 China P2Y2 purinergic Indicated for DED diagnosed with corneal and
ophthalmic solution, receptor agonist conjunctival epithelial injury accompanied by tear
3.0% (0.4mL:12mg) abnormalities
Varenicline solution, TYRVAYA™ 2024 China Nicotinic Indicated for the treatment of the signs and symptoms
0.03 mg nasal spray acetylcholine of DED
receptor agonist
Perfluorohexyloctane HENGQIN 2024 China Semifluorinated Indicated for the treatment of the signs and symptoms
alkane of DED
DED-dry eye disease; EU-European Union; lymphocyte LFA-1-function-associated antigen-1; TID-Three Times Daily; USA-United States of
America
Disclaimer- This is not necessarily a complete list of approved prescription medications approved in China and India
114
Globally, the availability of treatments for DED targeted at a sub-population of patients
who present predominately with signs of inflammation or its impact (e.g., hyperemia,
corneal staining, and conjunctival staining) and ADDE (e.g., decreased tear production)
speaks to a potential pathway for approval of additional products. 962-966
Three fundamental elements were identified to significantly improve trial success rates:
1) Exposure at the site of action (Pillar 1); 2) Binding to the pharmacological target
(Pillar 2); and 3) Expression of pharmacological activity from the site of action (Pillar 3).
Successful trials could achieve all 3 Pillars, achieve Pillars 1 and 3, or achieve Pillars 2
and 3. This was often combined with the inclusion of biomarkers and/or surrogate
markers of disease to achieve an early proof of mechanism and/or early signal of
efficacy. 198,967
Trials targeting patients presenting with ADDE and inflammation have consistently
linked decreased tear production with more severe disease that disrupts the ocular
115
surface, exemplified by ocular surface staining, through the concept of the lacrimal
functional unit. 951,954-957,962,963,965,966,971,972,979 The lacrimal functional unit is defined as
an integrated system comprising the lacrimal glands, ocular surface (cornea,
conjunctiva and meibomian glands), lids, and the sensory and motor nerves that
connect them. These trials have inclusion criteria and associated primary endpoints
focusing on increased ocular surface staining, decreased tear production, more severe
symptoms (e.g, VAS score ≥ 40 at screening and baseline and an OSDI score of ≥23,
and inflammation. 951,952,954-958,962,963,965,966,971,972,974,976,977,979-981 As a group, many of
these trials also excluded patients with signs of MGD, EDE or CLD. 951,955-957,962-966,971,972
The products targeting patients presenting with ADDE and inflammation can be broken
down into two main categories based upon their mechanism of action. The first group of
compounds includes immunomodulatory and anti-inflammatory agents 951,955-957,962-
966,971,972
and the second can be broadly categorized as tear stimulatory or conservation
agents for the various layers of the tear film (i.e., lipid layer, aqueous layer, and mucin
layer) (see 13X). 952,954,958,973,974,976,977,979-981 Studies involving three of these compounds
further restricted their inclusion/exclusion criteria and primary endpoints to closely
match their products’ proposed mechanism of action. 954,955,973,974,976,977,979
Studies using a semifluorinated alkane to target the lipid layer included patients
presenting with ADDE, inflammation and MGD defined as a total MGD score ≥3 [range
0-15] at screening and baseline (secretion of 5 central glands on the lower eyelid was
116
evaluated, and each was scored from 0-3: 0=normal; 1=thick/yellow, whitish, particulate;
2=paste; 3=none/occluded) and a TBUT ≤5 seconds at screening and baseline. Thus,
the inclusion criteria were tailored toward the mechanism of action of the compound
even though the primary endpoints were consistent with patients presenting with ADDE
and inflammation. 973,974,976,977
117
Table 15. Approved drugs and criteria for defining DED
Criterion for Defining DED
Signs Symptoms
Approved Drug
Corneal Staining
Meibum Quality
Ocular Dryness
Eye Redness
Studies
Conjunctival
Schiermer's
NEI-VFQ-25
Expression
Meibomian
Functional
Number of
Staining
SANDE
Glands
SPEED
Facial
Score
TBUT
Scale
OSDI
VAS
192731 -002, Sum of corneal and conj. Staining ≥ +5 in (without anaesthesia) ≤ 5 At least 9 Score ≥ 3
-003, and - the same eye with corneal staining ≥ +2 mm/5min, if 0 mm/5min responses
501 (Oxford scheme; 0-15) then nasal stimulation ≥ on the OSDI
RESTASIS®
Phase 2 and Active lid margin disease Corneal fluorescein (without anaesthesia) ≥ 1 Conjunctiveal ODS ≥ +3
Opus-1 not included staining score ≥ 2 in and ≤ 10 mm/5min redness score at 2
at least one region ≥ 1 (0-4 scale) consecutive
of either eye (0-4 in at least one time points
scale) at Screening eye (any eye intra-CAE
and Baseline; for OPUS-1)
Change in Inferior
Corneal Staining
(pre to post CAE ≥
XIIDRATM
+1)
Opus-2 and Active lid margin disease Corneal fluorescein (without anaesthesia) ≥ 1 Conjunctiveal Eye Dryness
Opus-3 not included staining score ≥ 2 in and ≤ 10 mm/5min redness score Score ≥ 40 at
at least one region ≥ 1 (0-4 scale) Screening and
of either eye (0-4 in at least one Baseline
scale) at Screening eye
and Baseline;
Inferior Corneal
Staining ≥ 0.5 at
screening and
baseline)
118
Onset-1 Blepharitis not requiring Corneal fluorescein baseline Schirmer’s Test
(OPP-002) treatment and mild staining score of ≥2 Score (STS; with topical
Meibomian gland disease in at least one anesthesia) of ≤10 mm/5
that are typically corneal region OR minutes with a cotton
associated with DED have a sum of ≥4 swab nasal stimulation
were allowed. for all corneal STS at least 7 mm
regions greater in the same eye;
<20 mm difference from
the study eye Schirmer
TYRVAYA™
CYS-002 Meibomian gland Total corneal Total lissamine Schirmer Test I score Score of ≥ 40 on
dysfunction (MGD) not fluorescein staining green between ≥ 2 mm and ≤ 8 the dryness VAS
included score of ≥ 6 (e.g. conjunctival mm at Screening and at Screening and
sum of inferior, score (sum of Baseline Baseline
superior, central, temporal and
nasal, and nasal) of ≥ 2,
temporal) according based on the
VEVYE
119
ESSENCE -1 Total corneal Total lissamine Schirmer Test I score Subjective
(CYS-003) fluorescein staining green between ≥ 1 mm and ≤ complaints of
score of ≥ 10 (e.g. conjunctival 10 mm at Screening and poor near vision
sum of inferior, score (sum of Baseline that impact
superior, central, temporal and activities of daily
nasal, and nasal) of ≥ 2, living, as defined
temporal) according based on the by at least a
to the NEI grading Oxford grading moderate impact
at Screening and at Screening (score ≥ 3) on at
Baseline and Baseline least 1 question
on the NEI VFQ-
25 Questions 5
to 7 in the main
questionnaire or
Near Vision
Subscale,
Questions A3 to
A5 in the
Appendix of
Optional
Additional
Questions, at the
screening visit
ESSENCE 2 Meibomian gland Total corneal Total lissamine Schirmer Test I score Score of ≥ 50 on
(CYS-004) dysfunction (MGD) not fluorescein staining green between ≥ 1 mm and ≤ the dryness VAS
included score of ≥ 10 (e.g. conjunctival 10 mm at Screening and at Screening and
sum of inferior, score (sum of Baseline Baseline
superior, central, temporal and
nasal, and nasal) of ≥ 2,
temporal) according based on the
to the NEI grading Oxford grading
at Screening and at Screening
Baseline and Baseline
Siccanove Severe blepharitis and/or Tear break- Corneal fluorescein Lissamine green Schirmer tear test OSDI score NOT VAS: At least
(NVG06C103) Meibomian gland disease up time staining ≥2 and ≤4 staining >4 (Van without anaesthesia of ≥23 one moderate to
(MGD) not included (TBUT) ≤8 (modified Oxford Bijsterveld ≥2 mm/5 min and <10 severe symptom of
seconds scale, scale 0-5) scale, scale 0-9) mm/5 min dry eye with a
score ≥2 (severity
graded on a 4-
point scale) i.e.,
burning/stinging,
foreign body
sensation, itching,
IKERVIS
N/A not listed not listed Corneal fluorescein not listed not listed not listed
HYALEIN®
ophthalmi
staining score
c solution
≧2+(0~3?)
0.1%
"moderate or
above"
N/A not listed not listed not listed not listed not listed not listed
"refractory or
solution 0.3%
severe"
ophthalmic
HYALEIN®
N/A not listed not listed not listed not listed not listed not listed
m
%
M
®
H
N
Y
E
L
o
p
h
u
ti
o
n
a
c
s
"moderate or
t
I
120
above"
N/A not listed not listed not listed not listed not listed not listed
hal
sol
0.3
HY
AL
op
uti
on
mi
Mi
"refractory"
ht
EI
%
ni
®
N
≤5 seconds Corneal fluorescein staining score: ≥3 (a Schirmer I test: ≤5 mm Subjective symptoms (including visual disturbance)
maximum score of 9); Rose Bengal
staining score: ≥3 (a maximum score of
DIQUAS®
A corneal fluorescein staining score of 4 Schirmer I test: ≤5 mm Score of 2 or more for 1 or more dry eye–related ocular symptom(s)
MUC
≤5 seconds Corneal fluorescein staining score ≥ 1 Schirmer I test: ≤5 mm Dryness score ≥ 1 using the DEQS questionnaire
DIQ
UA
S®
LX
121
Obstructive MGD presents as lid telangiectasia, gland orifice capping, gland dropout,
altered gland expressibility, and low TBUT). 6,984,985 This distinction from ADDE is
important as newer products in development like selenium sulfide ophthalmic ointment
975,978
and devices for treating eyelid glandular abnormalities, 986 have inclusion criteria
and associated primary endpoints focusing on glandular morphology and associated
evaporative DED. 987-993 Such trials may exclude patients with more severe symptoms,
signs of inflammation and significant ocular surface staining. 975 The inclusion/exclusion
criteria and endpoints were modelled on the clinical studies performed using a thermal
pulsation system. 994 These trial designs targeted at obstructive MGD and associated
EDE illustrate a population-targeted trial design aimed toward a group of patients who
may be underserved by some of the existing approved drug treatments.
11.3.3 US FDA clear guidance for industry and flexibility for dry eye approvals
The US FDA Draft Guidance for Industry on Dry Eye: Developing Drugs for Treatment
949
recommends the sponsor of a new DED treatment demonstrate efficacy and safety
in at least two adequate and well-controlled, multicenter independent studies. The
guidance covers trial design, comparator(s), trial population, demonstration of efficacy
and safety database requirements in detail. The following section will summarize the
key guidelines issued by the FDA. 949
The FDA guidelines are applicable to trials designed to demonstrate the efficacy and
safety of a new product in support of a new drug application and do not preclude the
use of such trial designs by companies or sponsors for the purposes of supporting
internal decisions related to a compound’s ability to progress in development based
upon measures of safety or efficacy such and proof of mechanism endpoints,
biomarkers and alternative thresholds for significance (e.g., 0.1% not 0.05%).
122
Daily exposure to environmental factors can impact DED endpoints: cigarette smoke,
smoke from wildfires, air pollution, dust, topically instilled drugs, and allergens are just a
few examples. 995-997 A model has been developed to standardize climatic, air quality
and visual conditions in a controlled environment. 996,998,999 Aspects of the environment
such as humidity, air flow and temperature can be manipulated to impact the ocular
surface, to identify individuals who may be more responsive to external environmental
factors and to create a reproducible environmental challenge. 999 This model is an
attempt to reduce the number of patients, sites, and time to demonstrate the efficacy of
an intervention. 996,998,999 Effects can be tested over one day in a controlled environment
or longer-term natural exposure trials of 2-weeks or longer. The FDA recommends that
safety trials be conducted at least 6 weeks in duration if efficacy trials are of shorter
duration. Trials in which the investigational drug is used as an add-on to a standardized
treatment regimen are also acceptable. 949
While confirmatory efficacy trials have used CAE exposure to enrich study populations
with patients who can change in severity secondary to environmental stress, the pre-
specified primary endpoints supporting approval were environmental. 964 Trials have
also used the controlled adverse environment to test an intervention’s protection against
the effects of an environmental challenge. 1000
11.3.3.2 Comparator(s)
The comparators and methods for controlling variance have evolved from the first
studies of cyclosporine in DED, 951 to the recent introduction of newer formulations and
concentrations of the active. 951,956,957,971,972 The first studies of cyclosporine in DED
evaluated cyclosporine plus artificial tears as needed to Month 4 and then less than 8
times daily after Month 4 to Month 6. 963 Numerous trials tried to replicate this
methodology and failed due to large placebo effects, lack of efficacy and regression of
signal which could have resulted from a therapeutic effect for the artificial tears. 963,1001
The subsequent approval of a novel integrin antagonist (lifitegrast) used only the vehicle
and excluded the use of artificial tears during the treatment period (See Supplementary
Figure 1 for the 0.05% ciclosporine trial design, Figure 2 for the lifitegrast controlled
environmental trial design, and Figure 3 for a lifitegrast environmental exposure trial
design).
123
The FDA acknowledged the advancement in trial methodology and encouraged trial
designers to consider that even water is known to be an effective component of topically
applied treatments for DED. Vehicle responders may be 1 in 5 of the trial population
suggesting this is a significant confounder in a study without a mechanism to screen for
vehicle response. 1002 In general, comparative clinical trials should use only the
investigational drug’s vehicle as a control agent. Trials should demonstrate statistical
and clinical superiority over a vehicle control or another treatment regimen. 949
Severe blepharitis or obvious inflammation of the lid margin can interfere with the
interpretation of trial results. In general, patients with these conditions should be studied
separately from routine DED. 949
124
2. Corneal Staining - A statistically significant difference between the percentage of
patients achieving a complete resolution of corneal staining, or
3. Tear Production - A statistically significant difference between the percentage of
patients achieving a 10-millimeter increase or more in Schirmer score
Achieving any one of these 3 endpoints can support approval of a product for the
treatment of the signs and symptoms of DED. Several marketed products have sought
approval based upon sign and symptom primary endpoint(s) and have been granted US
approval using pre-specified secondary endpoints of a known surrogate endpoint, a 10-
millimeter increase or more in Schirmer score, when the prespecified signs and
symptom endpoints failed in confirmatory efficacy studies. 951,956,957,963,971,972
The “10-millimeter increase or more in Schirmer score” was validated as part of the new
drug application for 0.05% ciclosporine and a description of the methods used to
validate the endpoint are described in the “Validation of the Clinical Relevance of the
Clinical Sign” section authored by Dr. William M Boyd from the US FDA as one of the
medical reviewers for NDA 21-023.
For reformulations of drug substances that are already approved in the same dosage
form, same route of administration, and the same or lower concentration, the FDA
recommends the sponsor ensure that a marketing application has safety information
from at least 100 patients treated for at least 6 months.
FDA recommends that the following evaluations be performed in each eye and reported
separately for each eye (regardless of which eye or eyes are treated):
Best corrected, distance visual acuity (4 meters in distance or more) at every visit.
A patient comfort examination before and after drug administration at every visit.
125
A slit lamp examination of the anterior segment that includes the cornea,
conjunctiva, anterior chamber, iris, lids, and lashes. At a minimum, examinations
should be performed at baseline, midway through the trial, the end of treatment, and
2 weeks after treatment discontinuation.
Endothelial cell count, systemic clinical and laboratory evaluations, and dilated
fundus examinations at baseline and at the end of trial or at month 3 (whichever is
later) in at least one trial.
DED also occurs in pediatric patients. 1003 Sponsors may consider a pediatric
assessment waiver request when submitting their required pediatric study plans under
the Pediatric Research Equity Act. 949
11.3.4 EMA guidance for industry and flexibility for DED approvals
EMA guidelines recommend demonstration of efficacy for 6 months, as DED is often
chronic and comparison against artificial tears, which are the standard of care is
recommended. 1004 EMA application submissions for lifitegrast and cyclosporine were
both withdrawn because of data demonstrating insufficient efficacy. 953,1005 The only
EMA-approved treatment is cyclosporine 0.1%, however, the indication is not for DED
but rather “severe keratitis in patients with DED, not improved despite treatment with
tear substitutes.” 1006
Regarding the selection of sign endpoints, the EMA considers both the target population
and the mechanism of action of the compound: Corneal staining, Schirmer score and
TBUT are established clinical endpoints. A composite measure is recommended using a
validated questionnaire for a symptom endpoint (e.g., OSDI) and use of single worst
symptom questions is not recommended. Sponsors are encouraged to consider a
relevant effect size rather than only statistical significance. For anti-inflammatory
products, since inflammation is considered a secondary manifestation, biomarkers may
be required to quantify treatment benefit.
The EMA will generally accept development programs completed in the US assuming
the studies comply with the guidelines outlined above. The EMA only requires
demonstration of safety and efficacy in one large adequately designed Phase 3 trial.
The primary endpoint for a cyclosporine 0.1% Phase 3 trial was a sign and symptom
composite responder rate. Specifically, the primary endpoint was the corneal fluorescein
staining-OSDI composite responder rate at Month 6 (i.e. end of Part 1). A corneal
fluorescein staining-OSDI responder was defined as a patient simultaneously satisfying
the following conditions: 1006
126
Improvement by 30% or more from baseline in OSDI (i.e. % change ≤-30%).
This is not an endpoint which has been acceptable to the FDA to date. The completed
confirmatory efficacy trials (SICCANOVE and SANSIKA) failed to demonstrate
consistent superiority on the primary endpoint and the product was subsequently
approved on a secondary sign endpoint by showing a statistically significant reduction in
corneal fluorescein staining versus vehicle. 951,1007,1008 No significant improvement in
symptoms was demonstrated along with the observed significant and clinically relevant
improvement in corneal staining and the indication was restricted to “severe keratitis
treatment” instead of a broader DED population.
Working with the PMDA involves a multistep process. This process is initiated with the
jizen mendan (“preliminary meeting”) which has no fee associated with it, is shorter in
length and requires less of a briefing package. The intention of this meeting is to confirm
the materials that are to be submitted and the sponsor’s questions that will be discussed
at the subsequent taimen jogen (“full consultation meeting”). At the full consultation
meeting, a more detailed briefing document is submitted in advance and written
comments are provided by the PMDA following the meeting.
127
pharmaceuticals in Japan. The PMDA typically wants to see dose ranging established in
the Japanese population specifically because of potential population differences in drug
activity and practice patterns. Confirmatory Phase 3 study is required to be completed in
Japan in part or whole. The PMDA generally requires a single, properly designed Phase
3 clinical trial to demonstrate safety and efficacy, similar to the EMA and discourages
post-hoc analysis for approval.
Phase 3 trials for the approval of diquafosol sodium ophthalmic solution 952,1012-1014 and
rebamipide ophthalmic suspension 952,1015 both required an active comparator (0.1%
hyaluronic acid ophthalmic solution was used in both) which necessitated a dosing
regimen of 6 times daily. This may have hampered interest in developing product in
Japan due the risk posed by even vehicle effects at such a high dosing frequency. The
introduction of long acting diquafasol sodium ophthalmic solution in 2022 which is only
dosed 3 times daily may have changed this risk profile. Long acting diquafasol also
employed a vehicle placebo in its Phase 3 trial. 1016
All 3 products were tested with primary endpoints at 4 weeks and all measured primary
treatment benefit using staining endpoints:
1. Diquafosol sodium ophthalmic solution: A non-inferiority margin of 0.34 for the study
(between-treatment difference in the mean change in the fluorescein & rose bengal
staining score [diquafosol - HA])
2. Rebamipide ophthalmic suspension: Change from baseline in corneal fluorescein
staining score and the lissamine green conjunctival staining score
3. Long acting diquafosol sodium ophthalmic solution: Change in corneal fluorescein
staining score from baseline to week 4
128
The primary endpoint for a hyaluronic acid Phase 3 study was a sign and symptom
composite responder rate. 980 This is not an endpoint which has been acceptable to the
FDA to date. The diquafosol ophthalmic solution Phase 3 also defined a non-inferiority
margin between-treatment difference in the mean change in the fluorescein and rose
bengal staining score (diquafosol compared to hyaluronic acid). 1012,1014 The FDA has
not accepted a non-inferiority margin to support approval of a drug for DED. Thus, the
PMDA has allowed flexibility in defining the type of comparison that will support
approval: superiority or non-inferiority to an active comparator.
The PMDA does not have the same guidance for industry on developing drugs for DED
as the FDA. In addition, the diagnostic criteria for DED were revised in 2016 1019 and no
longer include tear production or keratoconjunctival disorders in the diagnostic criteria.
Two of three approved drugs for the treatment of dry eye, diquafosol ophthalmic
solution 958,1012-1014 and rebamipide ophthalmic suspension 952,1015 were approved before
the revision and the remaining drug, long acting diquafosol sodium ophthalmic solution
1016,1017
had an additional additive that reduced the frequency of eye drops, but the
efficacy itself did not change, so the primary endpoint in trial was the fluorescein corneal
staining score. With the revision of diagnostic criteria (ie., tear production and
keratoconjunctival disorders are no longer included), it may be necessary to have a
robust discussion with regulatory authorities regarding the setting of the primary
endpoint based on the mechanism of action of a new drug.
11.4 Devices
129
Figure 5. Determining the Regulatory Pathway for a Device (from Van Norman
2016). 1021
130
approval. Class III risk devices will ultimately require a clinical trial and an
Investigational Device Exemption so that an unapproved device can be used in
clinical trials.
Trial design for Class III devices: Given that it is often difficult or unethical to have a
sham control, there is more flexibility on what type of trial is needed and discussion with
the FDA will determine what is acceptable. 1021 For the control arm of a device trial, one
may be able to use an active comparator (i.e., compare with an FDA approved device),
use metrics from pre- and post-treatment with a given device, or compare with registry
data. Typically, a double blind randomized clinical trial cannot be done with devices, as
is done with pharmaceuticals. 1022 Mixed devices, pharmaceutical and device combined,
are often reviewed by both divisions of the FDA, with one taking the lead, where
communications with the FDA determine the regulatory pathway. 953
Medical devices classified as Class III and IV are considered to have a high risk to the
human body and many of them require clinical trials. On the other hand, medical
devices used outside the body that are not implanted in the body and do not have a
high risk to the human body are often classified as Class II. With Class II, if the
probability of benefit is high when considering risk and benefit, it may be judged that
evaluation is possible without the need for clinical trials, as compared to Class III and IV.
Class I medical devices are those that are considered to have an extremely low risk to
the human body.
For medical devices that have already been approved overseas, it is possible to
extrapolate test results from overseas clinical trials, although ethnic differences must be
taken into consideration. Therefore, unlike drugs, some medical devices are approved
without domestic clinical trials by extrapolation to overseas clinical trials.
131
mucous membrane surfaces relieving ocular dryness and irritation. 953,1021 A list of
1024
approved demulcents and their concentration range has been provided by the FDA:
1. Cellulose derivatives:
a. Carboxymethyl cellulose (CMC),
b. Hydroxypropyl methylcellulose (hypromellose),
c. Hydroxyethylcellulose,
d. Methylcellulose,
2. Dextran 70
3. Gelatin
4. Polyols:
a. Glycerin,
b. Polyethylene glycol (300, 400),
c. Polysorbate 80,
5. Polymers:
a. Polyvinyl alcohol,
b. Polyvinyl pyrrolidone (povidone).
Federal law does not require premarket approval for over-the-counter eye drops,
including artificial tears, but does require eye drops to be sterile for safe use. The
existence of the FDA’s pre-approved monograph which covers artificial tears means
that if a developer lists an active ingredient that is not on the monograph, they will need
to provide clinical data supporting the safety of its use. Manufacturers can list potentially
“active ingredients” under “inactive ingredients” to avoid clinical trials.
While clinical data demonstrate that artificial tears show benefit for the management of
DED and their control is critical during drug/device trials to remove a confounding
source of variance, there are no regulatory standards for study design, clinical end
points, patient inclusion/exclusion criteria, or accepted methods to control for potential
sponsor bias when evaluating comparative efficacy claims. 1024
11.5 Design features to enhance clinical trial data quality and decisions
This section covers ways to enhance clinical trial data quality and data-supported
decisions on whether to proceed with an intervention under development for a particular
indication. (See TFOS DEWS III Management and Therapy Report, Section X, 2)
133
and Schirmer test value, are all subject to bias from the observer and /or biologic and
environmental variables-reflex tearing, time of day, humidity etc. Ophthalmology does
not have a lot of reliable, validated markers that correlate with clinically relevant findings
in DED. 953,1026 (See 953, DEWS II Clinical Trials Design Report 2017)
Recent examples of FDA cleared biomarkers include a point of care immunoassay test
for the in vitro detection of elevated levels of the MMP-9 protein in human tears, from
patients suspected of having DED 1027 Osmolarity testing is another FDA approved
biomarker.
Biomarkers were used in support of the approval of 0.1% cyclosporine and are
referenced in the EU Assessment report (level of HLA-DR expression). 1028 More
recently a genetic marker (TNFR1 marker) was used in evaluating anti-TNF alpha,
licaminlimab, and demonstrated improved outcome measures in a genetically defined
subgroup analysis. 1029 To date, no biomarker other than 10-millimeter increase or more
in Schirmer scores has supported regulatory approval for a DED treatment with the US
FDA where they could theoretically serve as a primary sign endpoint if validated as
clinically meaningful with an associated improvement in a primary symptom endpoint.
For the clinical development of drug therapies, the commonly selected sign and
symptom endpoints used in the trials have been similar across the major markets:
corneal or conjunctival staining (although using different scales), Schirmer score, and
symptom questionnaires. For symptom questionnaires there seems to be a trend
favoring overall symptom questionnaires (e.g., OSDI) and Visual Analogue Scales (i.e.,
ocular discomfort or dryness). 951,952,955-957,962-966,972,974,977,979,1012
134
production) have been approved. 951,952,955-957,962-966,972,974,977,979,1012 The major difference
between the regions is the duration of follow-up for the primary endpoints. The US
allows multiday, natural exposure trials of 2-weeks duration or longer, EMA allows
natural exposure trials of 6-months duration or longer and Japan allows natural
exposure trials of 4-weeks duration or longer. 949,1004,1009,1023
Taken together, the commonalities between the regions could allow for a single trial with
a common disease population and endpoint to support approval in more than one
region leveraging multiple statistical analysis plans specifying primarily endpoints at
different timepoints. 975,978 This allows a single trial to potentially support approval in
both the US and EU. To include Japan a sub-population of Japanese participants would
need to be included. While this could work for new compounds targeting the patient
populations and endpoints which have already been accepted in the major markets, it is
unclear how alternative endpoints (e.g., meibomian gland morphology-related endpoints)
will be accepted globally.
11.6 Conclusions
Newer market entrants have utilized clinical trial designs with enriched populations of
patients that take into consideration the mechanism of the drug under study, particularly
as it pertains to defining sign endpoints. 973,974,976,977 This has increased their likelihood
of technical success and is aligned with the US FDA Draft Guidance. This guidance
states that a statistically significant difference between the investigational treatment and
vehicle for at least one objective prespecified sign of DED (mean group score for test
versus vehicle) AND at least one subjective prespecified symptom of DED (mean group
score) can be used to support approval. A sign endpoint consistent with a drugs
mechanism of action can be selected to demonstrate that the drug under study has its
intended effect. The significance of the symptom endpoint then establishes the clinical
relevance of the observed effect the drug under study has upon how a patient feels
making it clinically relevant. 949 Thus, the recommendations from the TFOS DEWS II,
the industry report to identify three fundamental elements needed to achieve a
135
significant improvement in trial success rates (Pillars 1-3) and the US FDA Draft
Guidance for Industry on DED all coalesce on the same conclusion for achieving
regulatory and technical success for getting a new drug approved: match the sign
endpoint to the intended mechanism of the drug under study.
In conjunction with better selection of patients, trial designs have significantly evolved
over the 20 years of research. They have gone from little control over confounding
variables like artificial tear use 963 to careful control over comparator arms and run-in
periods within confirmatory efficacy studies. 954,955,966,973,974,976,977,979 Global regulatory
agencies have recognized this evolution with the FDA even encouraging trial designers
to consider that even water is known to be an effective component of topically applied
treatments for DED. Thus, comparative clinical trials should use only the vehicle from
the investigational drug as a control agent.
The agencies have also recognized that trial endpoints could change in sensitivity along
with disease severity and as a result have also evolved the requirements for replication
in confirmatory efficacy studies. The US FDA Draft Guidance recommends the sponsor
of a new DED treatment demonstrate efficacy and safety in at least two adequate and
well-controlled, multicenter independent studies. This is because the agency recognized
that different study designs, populations and timepoints may be needed to support sign
and symptom endpoints in different studies. 949 Global regulatory agencies also allow
pre-specified and alpha corrected secondary endpoints to support approval even if the
primary endpoint miss statistical significance 951,956,957,971,972,1007,1008 While major
differences still remain for trial requirements across the major markets, advances in trial
design, patient identification and statistical methods will undoubtedly continue to
advance allowing for more global confirmatory efficacy studies that can support
approvals in more than one region.
12. Summary
This report has explored key research published since the 2017 TFOS DEWS II
Workshop reports to underpin the evidence described in the TFOS DEWS III Diagnostic
Methodology 1 and Management and Therapy 2 reports and to present how new findings
from each of the topic areas considered, have informed the drivers of disease. Each
topic area has also identified key research needs for the next short and medium time
frame.
Table 16 below shows how each of the sections of the Digest report inform evidence for
the drivers of disease. Figure 6 illustrates the key findings from these interdisciplinary
areas, all of which have informed diagnosis and methodology and pathways towards
new treatment modalities. The importance of disease subtyping and the relevance of
new biomarkers has been consistently recognized.
136
Figure 6. Key findings from interdisciplinary reports
137
Table 16. Digest sections and drivers of DED
Drivers of DED
Anatomical juxtaposition
Inflammatory /oxidative
stress abnormalities
Ocular surface cell
Tear film aqueous
Blink/lid closure
Tear film lipid
abnormalities
abnormalities
abnormalities
abnormalities
abnormalities
abnormalities
abnormalities
abnormalities
Lid margin
Nerve
Sex, gender,
✔️ ✔️ ️ ️ ✔️ ️
and hormones
Epidemiology ️ ️ ️ ️ ️ ️ ️
Pathophysiology ️ ️ ️ ️ ️ ️ ️ ️ ️
Tear film ✔️ ✔️ ✔️ ✔️ ✔️ ✔️
Pain and
✔️ ✔️ ✔️
sensation
Iatrogenic ✔️ ✔️ ✔️ ✔️ ✔️ ✔️ ✔️ ✔️ ✔️
Clinical trials
✔️ ✔️ ✔️ ✔️ ✔️ ✔️
design
13. Acknowledgements
The TFOS DEWS III effort was supported by unrestricted donations from Alcon, Bausch
+ Lomb, Azura, AbbVie, CooperVision, Dompé, Espansione Group, Harrow, Laboratoire
Théa, SIFI, SINQI, Tarsus, Topcon and Trukera.
14. Figures
Figure 2. Prisma flowchart of the literature search outcome for prevalence studies
published between 18 September 2015 and 29 June 2024.
138
Figure 3. Prevalence of DED based on age and sex for different diagnostic criteria
Figure 5. Determining the regulatory pathway for a device (from Van Norman 2016)
15. References
1. Wolffsohn JS. TFOS DEWS III Diagnostic Methodology Report. Am J
Ophthalmol. 2025;
2. Jones LW. TFOS DEWS III Management and Therapy Report. Am J
Ophthalmol. 2025;
3. Sullivan DA, Rocha EM, Aragona P, et al. TFOS DEWS II Sex, Gender, and
Hormones Report. The Ocular Surface. 2017;15(3):284-333.
doi:10.1016/j.jtos.2017.04.001
4. Galor A, Britten-Jones AC, Feng Y, et al. TFOS Lifestyle: Impact of lifestyle
challenges on the ocular surface. The Ocular Surface. 2023;28:262-303.
doi:10.1016/j.jtos.2023.04.008
5. Tellefsen S, Morthen MK, Richards SM, et al. Sex Effects on Gene Expression
in Lacrimal Glands of Mouse Models of Sjögren Syndrome. Investigative
Opthalmology & Visual Science. 2018;59(13):5599. doi:10.1167/iovs.18-25772
6. Bron AJ, de Paiva CS, Chauhan SK, et al. TFOS DEWS II pathophysiology
report. The Ocular Surface. 2017;15(3):438-510. doi:10.1016/j.jtos.2017.05.011
7. Toda I, Sullivan BD, Rocha EM, Da Silveira LA, Wickham LA, Sullivan DA.
Impact of Gender on Exocrine Gland Inflammation in Mouse Models of
Sjögren's Syndrome. Experimental Eye Research. 1999;69(4):355-366.
doi:10.1006/exer.1999.0715
8. Rattner A, Heng JS, Winer BL, Goff LA, Nathans J. Normal and Sjogren’s
syndrome models of the murine lacrimal gland studied at single-cell resolution.
Proceedings of the National Academy of Sciences.
2023;120(42)doi:10.1073/pnas.2311983120
9. Hat K, Kaštelan S, Planinić A, Muller D, Ježek D. Pathohistological features of
the aging human lacrimal gland. Croatian Medical Journal. 2023;64(5):307-319.
doi:10.3325/cmj.2023.64.307
10. Chen X, Sullivan BD, Darabad RR, Liu S, Kam WR, Sullivan DA. Are BALB/c
Mice Relevant Models for Understanding Sex-Related Differences in Gene
Expression in the Human Meibomian Gland? Cornea. 2019;38(12):1554-1562.
doi:10.1097/ico.0000000000002017
11. Huang B, Fei F, Wen H, et al. Impacts of gender and age on meibomian gland
in aged people using artificial intelligence. Front Cell Dev Biol.
2023;11:1199440. doi:10.3389/fcell.2023.1199440
12. Lee PK, Chung JL, Kim DR, et al. Categorization of Meibomian Gland
Dysfunction Using Lipid Layer Thickness and Meibomian Gland Dropout in Dry
Eye Patients: A Retrospective Study. Korean Journal of Ophthalmology.
2024;38(1):64-70. doi:10.3341/kjo.2023.0126
139
13. Li Y, Lu J, Zhou Q, et al. Analysis of Clinical and Regional Distribution
Characteristics of Obstructive Meibomian Gland Dysfunction in China: A
Multicenter Study. Current Eye Research. 2020;45(11):1373-1379.
doi:10.1080/02713683.2020.1752387
14. Lin X, Wu Y, Chen Y, et al. Characterization of Meibomian Gland Atrophy and
the Potential Risk Factors for Middle Aged to Elderly Patients With Cataracts.
Translational Vision Science & Technology. 2020;9(7):48.
doi:10.1167/tvst.9.7.48
15. Stapleton F, Alves M, Bunya VY, et al. TFOS DEWS II Epidemiology Report.
The Ocular Surface. 2017;15(3):334-365. doi:10.1016/j.jtos.2017.05.003
16. García-Marqués JV, Talens-Estarelles C, García-Lázaro S, Wolffsohn JS,
Cerviño A. Systemic, environmental and lifestyle risk factors for dry eye disease
in a mediterranean caucasian population. Contact Lens and Anterior Eye.
2022;45(5):101539. doi:10.1016/j.clae.2021.101539
17. Zhao M, Yu Y, Roy NS, Ying G-s, Asbell P, Bunya VY. Sex-related differences
and hormonal effects in the Dry Eye Assessment and Management (DREAM)
study. British Journal of Ophthalmology. 2024;108(1):23-29. doi:10.1136/bjo-
2022-322238
18. Borrelli M, Frings A, Geerling G, Finis D. Gender-Specific Differences in Signs
and Symptoms of Dry Eye Disease. Current Eye Research. 2020;46(3):294-
301. doi:10.1080/02713683.2020.1801758
19. Wolffsohn JS, Wang MTM, Vidal-Rohr M, et al. Demographic and lifestyle risk
factors of dry eye disease subtypes: A cross-sectional study. The Ocular
Surface. 2021;21:58-63. doi:10.1016/j.jtos.2021.05.001
20. Li G, Garzon C, Klawe J, Akpek EK, Ahmad S. Demographic and Psychiatric
Associations With Dry Eye in a Medicare Population. Cornea.
2024;doi:10.1097/ico.0000000000003516
21. Lu P-T, Lee C-Y, Sun C-C. Sex Differences and Discordance Between
Symptoms and Signs of Dry Eye Disease. American Journal of Ophthalmology.
2024;260:14-20. doi:10.1016/j.ajo.2023.10.008
22. Wang MTM, Muntz A, Mamidi B, Wolffsohn JS, Craig JP. Modifiable lifestyle
risk factors for dry eye disease. Contact Lens and Anterior Eye.
2021;44(6):101409. doi:10.1016/j.clae.2021.01.004
23. Craig JP, Wang MTM, Ambler A, Cheyne K, Wilson GA. Characterising the
ocular surface and tear film in a population-based birth cohort of 45-year old
New Zealand men and women. The Ocular Surface. 2020;18(4):808-813.
doi:10.1016/j.jtos.2020.08.005
24. Vehof J, Sillevis Smitt-Kamminga N, Nibourg SA, Hammond CJ. Sex
differences in clinical characteristics of dry eye disease. The Ocular Surface.
2018;16(2):242-248. doi:10.1016/j.jtos.2018.01.001
25. Knop E, Knop N, Millar T, Obata H, Sullivan DA. The International Workshop on
Meibomian Gland Dysfunction: Report of the Subcommittee on Anatomy,
Physiology, and Pathophysiology of the Meibomian Gland. Investigative
Opthalmology & Visual Science. 2011;52(4):1938. doi:10.1167/iovs.10-6997c
140
26. Stapleton F, Abad JC, Barabino S, et al. TFOS Lifestyle: Impact of societal
challenges on the ocular surface. The Ocular Surface. 2023;28:165-199.
doi:10.1016/j.jtos.2023.04.006
27. Abusamak M. Corneal Epithelial Mapping Characteristics in Normal Eyes Using
Anterior Segment Spectral Domain Optical Coherence Tomography.
Translational Vision Science & Technology. 2022;11(3):6.
doi:10.1167/tvst.11.3.6
28. Abu-Romman A, Scholand KK, Govindarajan G, et al. Age-Related Differences
in the Mouse Corneal Epithelial Transcriptome and Their Impact on Corneal
Wound Healing. Investigative Ophthalmology & Visual Science. 2024;65(5):21.
doi:10.1167/iovs.65.5.21
29. Pham TL, Kakazu A, He J, Bazan HEP. Mouse strains and sexual divergence in
corneal innervation and nerve regeneration. The FASEB Journal.
2018;33(3):4598-4609. doi:10.1096/fj.201801957R
30. Abu-Romman A, Scholand KK, Pal-Ghosh S, et al. Conditional deletion of CD25
in the corneal epithelium reveals sex differences in barrier disruption. The
Ocular Surface. 2023;30:57-72. doi:10.1016/j.jtos.2023.07.008
31. Mecum NE, Demers D, Sullivan CE, Denis TE, Kalliel JR, Meng ID. Lacrimal
gland excision in male and female mice causes ocular pain and anxiety-like
behaviors. Scientific Reports. 2020;10(1)doi:10.1038/s41598-020-73945-w
32. Culoso A, Lowe C, Evinger C. Sex, blinking, and dry eye. Journal of
Neurophysiology. 2020;123(2):831-842. doi:10.1152/jn.00635.2019
33. Sakiani S, Olsen NJ, Kovacs WJ. Gonadal steroids and humoral immunity.
Nature Reviews Endocrinology. 2012;9(1):56-62. doi:10.1038/nrendo.2012.206
34. Zhao R, Chen X, Ma W, et al. A GPR174–CCL21 module imparts sexual
dimorphism to humoral immunity. Nature. 2019;577(7790):416-420.
doi:10.1038/s41586-019-1873-0
35. Schmiedel BJ, Singh D, Madrigal A, et al. Impact of Genetic Polymorphisms on
Human Immune Cell Gene Expression. Cell. 2018;175(6):1701-1715.e16.
doi:10.1016/j.cell.2018.10.022
36. McCoy SS, Sampene E, Baer AN. Association of Sjögren's Syndrome With
Reduced Lifetime Sex Hormone Exposure: A Case–Control Study. Arthritis
Care & Research. 2020;72(9):1315-1322. doi:10.1002/acr.24014
37. Lopes-Ramos CM, Chen C-Y, Kuijjer ML, et al. Sex Differences in Gene
Expression and Regulatory Networks across 29 Human Tissues. Cell Reports.
2020;31(12):107795. doi:10.1016/j.celrep.2020.107795
38. Anger JT, Case LK, Baranowski AP, et al. Pain mechanisms in the transgender
individual: a review. Frontiers in Pain Research.
2024;5doi:10.3389/fpain.2024.1241015
39. Mogil JS. Qualitative sex differences in pain processing: emerging evidence of a
biased literature. Nature Reviews Neuroscience. 2020;21(7):353-365.
doi:10.1038/s41583-020-0310-6
40. Belmonte C, Nichols JJ, Cox SM, et al. TFOS DEWS II pain and sensation
report. The Ocular Surface. 2017;15(3):404-437. doi:10.1016/j.jtos.2017.05.002
141
41. Li W, Lin MC. Sex Disparity in How Pain Sensitivity Influences Dry Eye
Symptoms. Cornea. 2019;38(10):1291-1298.
doi:10.1097/ico.0000000000002050
42. Li K, Zhang C, Yang Z, Wang Y, Si H. Evaluation of a novel dry eye model
induced by oral administration of finasteride. Molecular Medicine Reports.
2017;16(6):8763-8770. doi:10.3892/mmr.2017.7754
43. Morthen MK, Tellefsen S, Richards SM, et al. Testosterone Influence on Gene
Expression in Lacrimal Glands of Mouse Models of Sjögren Syndrome.
Investigative Opthalmology & Visual Science. 2019;60(6):2181.
doi:10.1167/iovs.19-26815
44. Sahin A, Liu Y, Kam WR, Darabad RR, Sullivan DA. Dihydrotestosterone
suppression of proinflammatory gene expression in human meibomian gland
epithelial cells. The Ocular Surface. 2020;18(2):199-205.
doi:10.1016/j.jtos.2020.02.006
45. Zibandeh N, Yıldız E, Özer B, Taş AY, Sahin A. Androgen Suppresses
Hyperosmolarity-Induced Inflammatory Mediators in Human Corneal Epithelial
Cells. Cornea. 2020;39(7):886-891. doi:10.1097/ico.0000000000002291
46. Aydin Kurna S, Oflaz Hacisalihoglu A, Altun A, et al. Effects of systemic anti-
androgen drugs on the ocular surface. Journal Français d'Ophtalmologie.
2022;45(6):619-627. doi:10.1016/j.jfo.2021.06.007
47. Shimizu S, Arita R, Kawashima M, Inoue S, Itoh K, Tsubota K. Morphological
Changes of Meibomian Glands in Men With Benign Prostate Hyperplasia.
Cornea. Apr 2021;40(4):445-448. doi:10.1097/ICO.0000000000002454
48. Supalaset S, Tananuvat N, Pongsatha S, Chaidaroon W, Ausayakhun S. A
Randomized Controlled Double-Masked Study of Transdermal Androgen in Dry
Eye Patients Associated With Androgen Deficiency. American Journal of
Ophthalmology. 2019;197:136-144. doi:10.1016/j.ajo.2018.09.021
49. Wang Lixiang, Huang L, Shuang Qiu, Deng Y. Effect of androgen replacement
therapy on mixed dry eye in a rabbit model. Zhonghua Yi Xue Za Zhi.
2021;101(32):2525-2530. doi:10.3760/cma.j.cn112137-20210322-00713
50. Kiyat P, Palamar M, Gerceker Turk B, Yagci A. Evaluation of dry eye and
Meibomian gland dysfunction in female androgenetic alopecia patients.
International Ophthalmology. 2021;41(11):3749-3757. doi:10.1007/s10792-021-
01946-3
51. Carmina E, Azziz R, Bergfeld W, et al. Female Pattern Hair Loss and Androgen
Excess: A Report From the Multidisciplinary Androgen Excess and PCOS
Committee. The Journal of Clinical Endocrinology & Metabolism.
2019;104(7):2875-2891. doi:10.1210/jc.2018-02548
52. Qi X, Yang Y, Xiong D, Wu S, Cui G, Zhang Q. ER-1 deficiency induces
inflammation and lipid deposition in meibomian gland and lacrimal gland.
Biochemical and Biophysical Research Communications. 2024;696:149526.
doi:10.1016/j.bbrc.2024.149526
53. Nalvarte I, Antonson P. Estrogen Receptor Knockout Mice and Their Effects on
Fertility. Receptors. 2023;2(1):116-126. doi:10.3390/receptors2010007
54. Hewitt SC, Couse JF, Korach KS. Estrogen receptor transcription and
transactivation Estrogen receptor knockout mice: what their phenotypes reveal
142
about mechanisms of estrogen action. Breast Cancer Research.
2000;2(5)doi:10.1186/bcr79
55. Winn NC, Jurrissen TJ, Grunewald ZI, et al. Estrogen receptor-α signaling
maintains immunometabolic function in males and is obligatory for exercise-
induced amelioration of nonalcoholic fatty liver. American Journal of Physiology-
Endocrinology and Metabolism. 2019;316(2):E156-E167.
doi:10.1152/ajpendo.00259.2018
56. Li C, Zhang X, Wang Y, Li M. Development and Long-Term Evaluation of
Ocular Surface Conditions Using Ovariectomized Cynomolgus Monkey. Eye &
Contact Lens: Science & Clinical Practice. 2019;45(4):246-253.
doi:10.1097/icl.0000000000000563
57. Kumar V, Sur VP, Guha R, Konar A, Hazra S. Estrogen Modulates Corneal
Nociception and Maintains Corneal Homeostasis in Rat Eye. Cornea.
2017;37(4):508-514. doi:10.1097/ico.0000000000001437
58. Rodríguez-Landa JF. Considerations of Timing Post-ovariectomy in Mice and
Rats in Studying Anxiety- and Depression-Like Behaviors Associated With
Surgical Menopause in Women. Frontiers in Behavioral Neuroscience.
2022;16doi:10.3389/fnbeh.2022.829274
59. Bereiter DA, Rahman M, Ahmed F, Thompson R, Luong N, Olson JK. Title:
P2x7 Receptor Activation and Estrogen Status Drive Neuroinflammatory
Mechanisms in a Rat Model for Dry Eye. Frontiers in Pharmacology.
2022;13doi:10.3389/fphar.2022.827244
60. Rahimi Darabad R, Suzuki T, Richards SM, et al. Does estrogen deficiency
cause lacrimal gland inflammation and aqueous-deficient dry eye in mice?
Experimental Eye Research. 2014;127:153-160. doi:10.1016/j.exer.2014.07.017
61. Rahimi Darabad R, Suzuki T, Richards SM, et al. Influence of Aromatase
Absence on the Gene Expression and Histology of the Mouse Meibomian
Gland. Investigative Opthalmology & Visual Science. 2013;54(2):987.
doi:10.1167/iovs.12-10992
62. Machairoudia G, Kazantzis D, Chatziralli I, et al. Prevalence and risk factors of
dry eye disease in patients treated with aromatase inhibitors for breast cancer.
Clinical and Experimental Optometry. 2023;107(4):415-419.
doi:10.1080/08164622.2023.2239247
63. Ağın A, Kocabeyoglu S, Yucel Gencoglu A, Aksoy S, Karakaya J, Irkec M. The
effects of systemic aromatase inhibitors on meibomian glands and corneal
structure. Eye. 2021;36(6):1185-1193. doi:10.1038/s41433-021-01612-z
64. Khoo P, Groeneveld T, Boyle F, O’Neill S, Forster B, Watson SL. Dry eye signs
and symptoms in patients on aromatase inhibitor therapy. Eye. 2021;36(4):766-
772. doi:10.1038/s41433-021-01538-6
65. Bicer T, Imamoglu GI, Dogan AS, et al. The effects of adjuvant hormonotherapy
on tear functions in patients with breast cancer. International Ophthalmology.
2020;40(8):2077-2083. doi:10.1007/s10792-020-01384-7
66. Kharb R, Haider K, Neha K, Yar MS. Aromatase inhibitors: Role in
postmenopausal breast cancer. Arch Pharm (Weinheim). Aug
2020;353(8):e2000081. doi:10.1002/ardp.202000081
143
67. García-Sánchez J, Mafla-España MA, Torregrosa MD, Cauli O. Adjuvant
aromatase inhibitor treatment worsens depressive symptoms and sleep quality
in postmenopausal women with localized breast cancer: A one-year follow-up
study. The Breast. 2022;66:310-316. doi:10.1016/j.breast.2022.11.007
68. Zhu Y, Huang X, Lin L, et al. Sleep Quality Is Associated With Severe
Meibomian Gland Disruption in Dry Eye. Frontiers in Medicine.
2022;9doi:10.3389/fmed.2022.812705
69. Boga A, Stapleton F, Chapman M, Golebiowski B. Effects of elevated serum
estrogen on dry eye in women undergoing in vitro fertilisation. The Ocular
Surface. 2023;29:511-520. doi:10.1016/j.jtos.2023.06.015
70. Çolak E, Ulusoy MO, Ceran MU, Taşdemir Ü, Kal A, Özçimen EE. Assessment
of tear parameters in patients undergoing ovulation induction for a short period
using optical coherence tomography of the anterior segment. Arquivos
Brasileiros de Oftalmologia. 2020;83(6)doi:10.5935/0004-2749.20200095
71. Huang L-H, Kuo C-P, Lu Y-C, Lee M-S, Lee S-H. Association of emotional
distress and quality of sleep among women receiving in-vitro fertilization
treatment. Taiwanese Journal of Obstetrics and Gynecology. 2019;58(1):168-
172. doi:10.1016/j.tjog.2018.11.031
72. Schmidl D, Szalai L, Kiss OG, Schmetterer L, Garhöfer G. A Phase II,
Multicenter, Randomized, Placebo-Controlled, Double-Masked Trial of a Topical
Estradiol Ophthalmic Formulation in Postmenopausal Women with Moderate-to-
Severe Dry Eye Disease. Advances in Therapy. 2021;38(4):1975-1986.
doi:10.1007/s12325-021-01680-3
73. Yang M, Fjærvoll HK, Fjærvoll KA, et al. Sex-based differences in conjunctival
goblet cell responses to pro-inflammatory and pro-resolving mediators.
Scientific Reports. 2022;12(1)doi:10.1038/s41598-022-20177-9
74. Straub RH. The Complex Role of Estrogens in Inflammation. Endocrine
Reviews. 2007;28(5):521-574. doi:10.1210/er.2007-0001
75. Meng ID, Barton ST, Goodney I, Russell R, Mecum NE. Progesterone
Application to the Rat Forehead Produces Corneal Antinociception.
Investigative Opthalmology & Visual Science. 2019;60(5):1706.
doi:10.1167/iovs.18-26049
76. Luo ZK, Domenech-Estarellas EA, Han A, et al. Efficacy and Safety of 1%
Progesterone Gel to the Forehead for Ocular Chronic Graft-versus-Host
Disease. Transplantation and Cellular Therapy. 2021;27(5):433.e1-433.e8.
doi:10.1016/j.jtct.2021.02.008
77. Piette PCM. The pharmacodynamics and safety of progesterone. Best Practice
& Research Clinical Obstetrics & Gynaecology. 2020;69:13-29.
doi:10.1016/j.bpobgyn.2020.06.002
78. Kontula K, Paavonen T, Luukkainen T, Andersson LC. Binding of progestins to
the glucocorticoid receptor. Biochemical Pharmacology. 1983;32(9):1511-1518.
doi:10.1016/0006-2952(83)90474-4
79. Robciuc A, Savolainen-Peltonen H, Haanpää M, Moilanen JAO, Mikkola TS.
Sex Steroid Hormone Analysis in Human Tear Fluid Using a Liquid
Chromatography—Mass Spectrometry Method. International Journal of
Molecular Sciences. 2022;23(23):14864. doi:10.3390/ijms232314864
144
80. Phan MAT, Gibson E, Golebiowski B, Stapleton F, Jenner AM, Bucknall MP.
Analysis of sex steroids in human tears using LC-MS and GC-MS:
Considerations and developments to improve method sensitivity and accuracy.
Experimental Eye Research. 2022;225:109283. doi:10.1016/j.exer.2022.109283
81. Gibson EJ, Bucknall MP, Golebiowski B, Stapleton F. Comparative limitations
and benefits of liquid chromatography – mass spectrometry techniques for
analysis of sex steroids in tears. Experimental Eye Research. 2019;179:168-
178. doi:10.1016/j.exer.2018.11.015
82. DEWS Diagnostic Methodology. Methodologies to Diagnose and Monitor Dry
Eye Disease: Report of the Diagnostic Methodology Subcommittee of the
International Dry Eye WorkShop (2007). The Ocular Surface. 2007;5(2):108-
152. doi:10.1016/s1542-0124(12)70083-6
83. Patel R, Zhu M, Robertson DM. Shifting the IGF-axis: An age-related decline in
human tear IGF-1 correlates with clinical signs of dry eye. Growth Hormone &
IGF Research. 2018;40:69-73. doi:10.1016/j.ghir.2018.02.001
84. Stuard Sambhariya W, Trautmann IJ, Robertson DM. Insulin-like growth factor
binding protein-3 mediates hyperosmolar stress–induced mitophagy through the
mechanistic target of rapamycin. Journal of Biological Chemistry.
2023;299(11):105239. doi:10.1016/j.jbc.2023.105239
85. Bisetty, Khan E. Prevalence of dry eye syndrome in a South African diabetic
paediatric population. Contact Lens and Anterior Eye. 2024:102175.
doi:10.1016/j.clae.2024.102175
86. Bayat AH, Aydemir E, Aydemir GA, Gencer H. Assessment of Tear Film
Anomalies in Childhood Obesity. Klinische Monatsblätter für Augenheilkunde.
2021;239(03):331-337. doi:10.1055/a-1668-0276
87. Kasturi N, Manchikanti V, Rajappa M, Gochhait D. Ocular surface disorder
among adult patients with type II diabetes mellitus and its correlation with tear
film markers: A pilot study. Taiwan Journal of Ophthalmology. 2021;11(2):156.
doi:10.4103/tjo.tjo_56_20
88. Zagon IS, Sassani JW, Purushothaman I, McLaughlin PJ. Dysregulation of the
OGF–OGFr pathway correlates with elevated serum OGF and ocular surface
complications in the diabetic rat. Experimental Biology and Medicine.
2020;245(15):1414-1421. doi:10.1177/1535370220940273
89. Burgos-Blasco B, Diaz-Valle D, Rego-Lorca D, et al. Topical insulin, a novel
corneal epithelial regeneration agent in dry eye disease. European Journal of
Ophthalmology. 2023;34(3):719-725. doi:10.1177/11206721231206790
90. Tahmaz V, Menghesha L, Stern ME, Holtick U, Scheid C, Steven P. Insulin eye
drops for severe refractory chronic ocular graft-versus-host disease. Bone
Marrow Transplantation. 2024;59(7):1031-1033. doi:10.1038/s41409-024-
02272-9
91. Aniah Azmi N, Bastion M-LC. Short-Term Results of Trial of Topical Insulin for
Treatment of Dry Eyes in Diabetics. Eye & Contact Lens: Science & Clinical
Practice. 2020;46(1):S25-S32. doi:10.1097/icl.0000000000000623
92. Cid-Bertomeu P, Vilaltella M, Martínez M, Mir M, Huerva V. Topical Insulin for
Ocular Surface Disease. Journal of Ocular Pharmacology and Therapeutics.
2024;40(4):204-214. doi:10.1089/jop.2024.0016
145
93. Cruz-Cazarim ELC, Cazarim MS, Ogunjimi AT, Petrilli R, Rocha EM, Lopez
RFV. Prospective insulin-based ophthalmic delivery systems for the treatment
of dry eye syndrome and corneal injuries. European Journal of Pharmaceutics
and Biopharmaceutics. 2019;140:1-10. doi:10.1016/j.ejpb.2019.04.014
94. Xia Y, Zhang Y, Du Y, Wang Z, Cheng L, Du Z. Comprehensive dry eye
therapy: overcoming ocular surface barrier and combating inflammation,
oxidation, and mitochondrial damage. Journal of Nanobiotechnology.
2024;22(1)doi:10.1186/s12951-024-02503-7
95. Dasari S, Naha K, Hande M, Vivek G. Hot and cold: coexistent Graves’ disease
and Hashimoto's thyroiditis in a patient with Schmidt's syndrome. BMJ Case
Reports. 2014:bcr2013010432. doi:10.1136/bcr-2013-010432
96. Yu K, Bunya V, Maguire M, et al. Systemic Conditions Associated with Severity
of Dry Eye Signs and Symptoms in the Dry Eye Assessment and Management
Study. Ophthalmology. Oct 2021;128(10):1384-1392.
doi:10.1016/j.ophtha.2021.03.030
97. Bikbov MM, Gilmanshin TR, Zainullin RM, et al. Prevalence and associations of
dry eye disease and meibomian gland dysfunction in the ural eye and medical
study. Scientific Reports. 2022;12(1)doi:10.1038/s41598-022-22580-8
98. Garcia-Queiruga J, Pena-Verdeal H, Sabucedo-Villamarin B, Giraldez MJ,
Garcia-Resua C, Yebra-Pimentel E. A cross-sectional study of non-modifiable
and modifiable risk factors of dry eye disease states. Contact Lens and Anterior
Eye. 2023;46(3):101800. doi:10.1016/j.clae.2022.101800
99. Dossari SK, Alkhars AZ, Albaqshi AA, et al. Prevalence of Dry Eye Disease and
Its Risk Factors Among the General Population of Saudi Arabia: A Cross-
Sectional Survey. Cureus. 2022;doi:10.7759/cureus.32552
100. Lai EW, Tai Y-H, Wu H-L, et al. The Association between Autoimmune Thyroid
Disease and Ocular Surface Damage: A Retrospective Population-Based
Cohort Study. Journal of Clinical Medicine. 2023;12(9):3203.
doi:10.3390/jcm12093203
101. Rana HS, Akella SS, Clabeaux CE, Skurski ZP, Aakalu VK. Ocular surface
disease in thyroid eye disease: A narrative review. The Ocular Surface.
2022;24:67-73. doi:10.1016/j.jtos.2022.02.001
102. Faustino-Barros JF, Saranzo Sant’Ana AM, Dias LC, et al. Distinct Inflammatory
and Oxidative Effects of Diabetes Mellitus and Hypothyroidism in the Lacrimal
Functional Unit. International Journal of Molecular Sciences. 2023;24(8):6974.
doi:10.3390/ijms24086974
103. Liao X, Aljufairi FMAA, Sebastian JU, et al. Investigation of lacrimal gland and
extraocular muscle in thyroid eye disease patients with severe subjective dry
eye disease: a multiparametric magnetic resonance imaging study. Graefe's
Archive for Clinical and Experimental Ophthalmology. 2024;262(8):2651-2659.
doi:10.1007/s00417-024-06424-x
104. Zhang K, Yu X, Zhang Y, et al. Identification of key genes in salivary gland in
Sjögren’s syndrome complicated with Hashimoto thyroiditis: Common
pathogenesis and potential diagnostic markers. Medicine.
2023;102(39):e35188. doi:10.1097/md.0000000000035188
146
105. Allam IY, Lazreg S, Shafik Shaheen M, Doheim MF, Mohammed MA. Ocular
Surface Changes in Patients with Thyroid Eye Disease: An Observational
Clinical Study. Clinical Ophthalmology. 2021;Volume 15:2481-2488.
doi:10.2147/opth.s317708
106. Park J, Baek S. Dry eye syndrome in thyroid eye disease patients: The role of
increased incomplete blinking and Meibomian gland loss. Acta
Ophthalmologica. 2018;97(5)doi:10.1111/aos.14000
107. Alanazi SA, Alomran AA, Abusharha A, et al. <p>An assessment of the ocular
tear film in patients with thyroid disorders</p>. Clinical Ophthalmology.
2019;Volume 13:1019-1026. doi:10.2147/opth.s210044
108. Liao X, Lai KKH, Aljufairi FMAA, et al. Ocular Surface Changes in Treatment-
Naive Thyroid Eye Disease. Journal of Clinical Medicine. 2023;12(9):3066.
doi:10.3390/jcm12093066
109. Lai KKH, Liao X, Aljufairi FMAA, et al. Ocular surface and meibomian gland
evaluation in euthyroid Graves’ ophthalmopathy. International Ophthalmology.
2024;44(1)doi:10.1007/s10792-024-02919-y
110. Riguetto C, Barbosa EB, Atihe CC, Reis F, Alves M, Zantut-Wittmann D. Ocular
Surface Disease Related to the Inflammatory and Non-Inflammatory Phases of
Thyroid Eye Disease. Clinical Ophthalmology. 2023;Volume 17:3465-3475.
doi:10.2147/opth.s430861
111. Takahashi Y, Vaidya A, Kakizaki H. Risk factors for development of superior
limbic keratoconjunctivitis in thyroid eye disease in Japanese. Graefe's Archive
for Clinical and Experimental Ophthalmology. 2022;261(3):833-839.
doi:10.1007/s00417-022-05827-y
112. Kiziltoprak H, Koc M, Tekin K, et al. Local Ocular Surface Alterations in Children
with Hashimoto’s Thyroiditis. Ocular Immunology and Inflammation.
2019;28(5):791-797. doi:10.1080/09273948.2019.1642495
113. Kashkouli MB, Alemzadeh SA, Aghaei H, et al. Subjective versus objective dry
eye disease in patients with moderate-severe thyroid eye disease. The Ocular
Surface. 2018;16(4):458-462. doi:10.1016/j.jtos.2018.07.003
114. Starčević A, Radojičić Z, Djurić Stefanović A, et al. Morphometric and
volumetric analysis of lacrimal glands in patients with thyroid eye disease.
Scientific Reports. 2023;13(1)doi:10.1038/s41598-023-43083-0
115. Wong NTY, Yuen KFK, Aljufairi FMAA, et al. Magnetic resonance imaging
parameters on lacrimal gland in thyroid eye disease: a systematic review and
meta-analysis. BMC Ophthalmology. 2023;23(1)doi:10.1186/s12886-023-
03008-x
116. Park J, Kim J, Lee H, Park M, Baek S. Functional and structural evaluation of
the meibomian gland using a LipiView interferometer in thyroid eye disease.
Canadian Journal of Ophthalmology. 2018;53(4):373-379.
doi:10.1016/j.jcjo.2017.11.006
117. Winter S, Diamond M, Green J, et al. Transgender people: health at the
margins of society. Lancet. Jul 23 2016;388(10042):390-400.
doi:10.1016/S0140-6736(16)00683-8
147
118. Nieves-Ríos C, Pulido JS, Thornton S, et al. Instances of ocular findings in
transgender patients undergoing hormonal therapy. American Journal of
Ophthalmology Case Reports. 2023;32:101965. doi:10.1016/j.ajoc.2023.101965
119. Berrian K, Exsted MD, Lampe NM, Pease SL, Akré ERL. Barriers to quality
healthcare among transgender and gender nonconforming adults. Health
Services Research. 2024;doi:10.1111/1475-6773.14362
120. Maoto M, Davis B. Breaking barriers: How transwomen meet their healthcare
needs. African Journal of Primary Health Care & Family Medicine.
2024;16(1)doi:10.4102/phcfm.v16i1.4598
121. Zhang X, Neuville P, Skokan AJ. Sexual health in transgender and gender
diverse people. Current Opinion in Urology.
2024;doi:10.1097/mou.0000000000001205
122. Coelho R, Gonçalves R, Mendes F, Macedo G. Gastroenterology healthcare in
LGBTQ+ individuals. European Journal of Gastroenterology & Hepatology.
2024;doi:10.1097/meg.0000000000002808
123. Ceolin C, Papa MV, Scala A, Sergi G, Garolla A. Getting old in the desired
gender: a systematic review on aging diseases in transgender people. Journal
of Endocrinological Investigation. 2024;47(8):1851-1862. doi:10.1007/s40618-
024-02353-y
124. Betsi G, Goulia P, Sandhu S, Xekouki P. Puberty suppression in adolescents
with gender dysphoria: an emerging issue with multiple implications. Frontiers in
Endocrinology. 2024;15doi:10.3389/fendo.2024.1309904
125. Heng YJ, Baker GM, Fein-Zachary VJ, et al. Effect of testosterone therapy on
breast tissue composition and mammographic breast density in trans masculine
individuals. Breast Cancer Research. 2024;26(1)doi:10.1186/s13058-024-
01867-w
126. Hashemi L, Marijic Buljubasic A, Budoff MJ, et al. Gender-Affirming Hormone
Treatment and Metabolic Syndrome Among Transgender Veterans. JAMA
Network Open. 2024;7(7):e2419696.
doi:10.1001/jamanetworkopen.2024.19696
127. de Silva NL, Dimakopoulou A, Quinton O, Jayasena CN. Metabolic and
cardiovascular risks of hormone treatment for transgender individuals. Best
Practice & Research Clinical Endocrinology & Metabolism. 2024:101907.
doi:10.1016/j.beem.2024.101907
128. Tienforti D, Savignano G, Spagnolo L, Di Giulio F, Baroni MG, Barbonetti A.
Biochemical liver damage during gender affirming therapy in trans adults
assigned female at birth: a meta-analysis. Journal of Endocrinological
Investigation. 2024;doi:10.1007/s40618-024-02418-y
129. Stapleton F, Alves M, Bunya VY, et al. TFOS DEWS II Epidemiology Report.
Ocular Surface. Jul 2017;15(3):334-365. doi:10.1016/j.jtos.2017.05.003
130. Jelicic Kadic A, Vucic K, Dosenovic S, Sapunar D, Puljak L. Extracting data
from figures with software was faster, with higher interrater reliability than
manual extraction. J Clin Epidemiol. Jun 2016;74:119-23.
doi:10.1016/j.jclinepi.2016.01.002
148
131. Wolffsohn JS, Arita R, Chalmers R, et al. TFOS DEWS II Diagnostic
Methodology report. Ocul Surf. Jul 2017;15(3):539-574.
doi:10.1016/j.jtos.2017.05.001
132. Arita R, Mizoguchi T, Kawashima M, et al. Meibomian Gland Dysfunction and
Dry Eye Are Similar but Different Based on a Population-Based Study: The
Hirado-Takushima Study in Japan. Am J Ophthalmol. Nov 2019;207:410-418.
doi:10.1016/j.ajo.2019.02.024
133. Ma J, Zhu H, Guo W, et al. Association of different digital media experiences
with paediatric dry eye in China: a population-based study. BMJ Open. Nov 30
2022;12(11):e062850. doi:10.1136/bmjopen-2022-062850
134. Stang A, Schmidt B, Schramm S, et al. Synergism between coexisting eye
diseases and sex in increasing the prevalence of the dry eye syndrome. Sci
Rep. Jan 3 2024;14(1):314. doi:10.1038/s41598-023-50871-1
135. Uchino M, Nishiwaki Y, Michikawa T, et al. Prevalence and risk factors of dry
eye disease in Japan: Koumi study. Ophthalmology. Dec 2011;118(12):2361-7.
doi:10.1016/j.ophtha.2011.05.029
136. Vehof J, Wang B, Kozareva D, Hysi PG, Snieder H, Hammond CJ. The
heritability of dry eye disease in a female twin cohort. Invest Ophthalmol Vis Sci.
Sep 23 2014;55(11):7278-83. doi:10.1167/iovs.14-15200
137. Schaumberg DA, Dana R, Buring JE, Sullivan DA. Prevalence of dry eye
disease among US men: estimates from the Physicians' Health Studies.
Archives of ophthalmology. Jun 2009;127(6):763-8.
doi:10.1001/archophthalmol.2009.103
138. Schaumberg DA, Sullivan DA, Buring JE, Dana MR. Prevalence of dry eye
syndrome among US women. Am J Ophthalmol. Aug 2003;136(2):318-26.
doi:10.1016/s0002-9394(03)00218-6
139. Zhang Y, Chen H, Wu X. Prevalence and risk factors associated with dry eye
syndrome among senior high school students in a county of Shandong
Province, China. Ophthalmic epidemiology. Aug 2012;19(4):226-30.
doi:10.3109/09286586.2012.670742
140. Ahn JH, Choi YH, Paik HJ, Kim MK, Wee WR, Kim DH. Sex differences in the
effect of aging on dry eye disease. Clin Interv Aging. 2017;12:1331-1338.
doi:10.2147/CIA.S140912
141. Magno MS, Utheim TP, Snieder H, Hammond CJ, Vehof J. The relationship
between dry eye and sleep quality. Ocul Surf. Apr 2021;20:13-19.
doi:10.1016/j.jtos.2020.12.009
142. Malet F, Le Goff M, Colin J, et al. Dry eye disease in French elderly subjects:
the Alienor Study. Acta Ophthalmol. Sep 2014;92(6):e429-36.
doi:10.1111/aos.12174
143. Paulsen AJ, Cruickshanks KJ, Fischer ME, et al. Dry eye in the beaver dam
offspring study: prevalence, risk factors, and health-related quality of life. Am J
Ophthalmol. Apr 2014;157(4):799-806. doi:10.1016/j.ajo.2013.12.023
144. Munch K, Nohre M, Westenberger A, et al. Prevalence and Correlates of Dry
Eye in a German Population Sample. Cornea. Jun 1 2024;43(6):685-692.
doi:10.1097/ICO.0000000000003374
149
145. Kai JY, Wu YB, Shi B, et al. Dry eye symptoms and health-related quality of life
among Chinese individuals: a national-based study. Br J Ophthalmol. Oct 22
2024;108(11):1500-1507. doi:10.1136/bjo-2023-324677
146. Li X, Wang Z, Mu J, et al. Prevalence and associated risk factors of dry eye
disease in Hotan, Xinjiang: a cross-sectional study. BMC Ophthalmol. May 15
2023;23(1):214. doi:10.1186/s12886-023-02955-9
147. Kim JM, Choi YJ. Impact of Dietary Nutrients on the Prevalence of Dry Eye
Syndrome among Korean Women Aged 40 and above: Evidence from the
Korea National Health and Nutrition Examination Survey. Nutrients. Jan 26
2024;16(3)doi:10.3390/nu16030372
148. Viet Vu CH, Uchino M, Kawashima M, et al. Lack of social support and social
trust as potential risk factors for dry eye disease: JPHC-NEXT study. Ocul Surf.
Apr 2019;17(2):278-284. doi:10.1016/j.jtos.2019.01.005
149. Uchino M, Schaumberg DA, Dogru M, et al. Prevalence of dry eye disease
among Japanese visual display terminal users. Ophthalmology. Nov
2008;115(11):1982-8. doi:10.1016/j.ophtha.2008.06.022
150. McCarty CA, Bansal AK, Livingston PM, Stanislavsky YL, Taylor HR. The
epidemiology of dry eye in Melbourne, Australia. Ophthalmology. Jun
1998;105(6):1114-9. doi:10.1016/S0161-6420(98)96016-X
151. Ahn JM, Lee SH, Rim TH, et al. Prevalence of and risk factors associated with
dry eye: the Korea National Health and Nutrition Examination Survey 2010-
2011. Am J Ophthalmol. Dec 2014;158(6):1205-1214 e7.
doi:10.1016/j.ajo.2014.08.021
152. Hashemi H, Khabazkhoob M, Kheirkhah A, et al. Prevalence of dry eye
syndrome in an adult population. Clin Exp Ophthalmol. Apr 2014;42(3):242-8.
doi:10.1111/ceo.12183
153. Lu P, Chen X, Liu X, et al. Dry eye syndrome in elderly Tibetans at high altitude:
a population-based study in China. Cornea. Jun 2008;27(5):545-51.
doi:10.1097/ICO.0b013e318165b1b7
154. Tong L, Saw SM, Lamoureux EL, et al. A questionnaire-based assessment of
symptoms associated with tear film dysfunction and lid margin disease in an
Asian population. Ophthalmic epidemiology. Jan-Feb 2009;16(1):31-7.
doi:10.1080/09286580802521317
155. Um SB, Kim NH, Lee HK, Song JS, Kim HC. Spatial epidemiology of dry eye
disease: findings from South Korea. Int J Health Geogr. Aug 15 2014;13:31.
doi:10.1186/1476-072X-13-31
156. Guo B, Lu P, Chen X, Zhang W, Chen R. Prevalence of dry eye disease in
Mongolians at high altitude in China: the Henan eye study. Ophthalmic
epidemiology. Aug 2010;17(4):234-41. doi:10.3109/09286586.2010.498659
157. Tan LL, Morgan P, Cai ZQ, Straughan RA. Prevalence of and risk factors for
symptomatic dry eye disease in Singapore. Clin Exp Optom. Jan 2015;98(1):45-
53. doi:10.1111/cxo.12210
158. Schein OD, Tielsch JM, Munoz B, Bandeen-Roche K, West S. Relation
between signs and symptoms of dry eye in the elderly. A population-based
perspective. Ophthalmology. Sep 1997;104(9):1395-401.
150
159. Lin PY, Tsai SY, Cheng CY, Liu JH, Chou P, Hsu WM. Prevalence of dry eye
among an elderly Chinese population in Taiwan: the Shihpai Eye Study.
Ophthalmology. Jun 2003;110(6):1096-101. doi:10.1016/S0161-
6420(03)00262-8
160. Lee AJ, Lee J, Saw SM, et al. Prevalence and risk factors associated with dry
eye symptoms: a population based study in Indonesia. Br J Ophthalmol. Dec
2002;86(12):1347-51. doi:10.1136/bjo.86.12.1347
161. Bakkar MM, Shihadeh WA, Haddad MF, Khader YS. Epidemiology of symptoms
of dry eye disease (DED) in Jordan: A cross-sectional non-clinical population-
based study. Cont Lens Anterior Eye. Jun 2016;39(3):197-202.
doi:10.1016/j.clae.2016.01.003
162. Ferrero A, Alassane S, Binquet C, et al. Dry eye disease in the elderly in a
French population-based study (the Montrachet study: Maculopathy, Optic
Nerve, nuTRition, neurovAsCular and HEarT diseases): Prevalence and
associated factors. Ocul Surf. Jan 2018;16(1):112-119.
doi:10.1016/j.jtos.2017.09.008
163. Shanti Y, Shehada R, Bakkar MM, Qaddumi J. Prevalence and associated risk
factors of dry eye disease in 16 northern West bank towns in Palestine: a cross-
sectional study. BMC Ophthalmol. Jan 13 2020;20(1):26. doi:10.1186/s12886-
019-1290-z
164. Yasir ZH, Chauhan D, Khandekar R, Souru C, Varghese S. Prevalence and
Determinants of Dry Eye Disease among 40 Years and Older Population of
Riyadh (Except Capital), Saudi Arabia. Middle East Afr J Ophthalmol. Jan-Mar
2019;26(1):27-32. doi:10.4103/meajo.MEAJO_194_18
165. Bikbov MM, Kazakbaeva GM, Rakhimova EM, et al. The prevalence of dry eye
in a very old population. Acta Ophthalmol. May 2022;100(3):262-268.
doi:10.1111/aos.14937
166. Kobia-Acquah E, Ankamah-Lomotey S, Owusu E, et al. Prevalence and
associated risk factors of symptomatic dry eye in Ghana: A cross-sectional
population-based study. Cont Lens Anterior Eye. Dec 2021;44(6):101404.
doi:10.1016/j.clae.2020.12.067
167. Choi HR, Kim NH, Lee JM, et al. Risk Factors Influencing the Occurrence and
Severity of Symptomatic Dry Eye Syndrome: A Cross-sectional Study.
Ophthalmic epidemiology. Dec 2021;28(6):488-494.
doi:10.1080/09286586.2021.1879172
168. Kim SW, Brown DJ, Jester JV. Transcriptome analysis after PPARγ activation in
human meibomian gland epithelial cells (hMGEC). Ocul Surf. Oct
2019;17(4):809-816. doi:10.1016/j.jtos.2019.02.003
169. Olaniyan SI, Fasina O, Bekibele CO, Ogundipe AO. Dry eye disease in an adult
population in South-West Nigeria. Cont Lens Anterior Eye. Oct 2016;39(5):359-
64. doi:10.1016/j.clae.2016.06.008
170. Tandon R, Vashist P, Gupta N, et al. Association of dry eye disease and sun
exposure in geographically diverse adult (>/=40 years) populations of India: The
SEED (sun exposure, environment and dry eye disease) study - Second report
of the ICMR-EYE SEE study group. Ocul Surf. Oct 2020;18(4):718-730.
doi:10.1016/j.jtos.2020.07.016
151
171. Dana R, Bradley JL, Guerin A, et al. Estimated Prevalence and Incidence of Dry
Eye Disease Based on Coding Analysis of a Large, All-age United States
Health Care System. Am J Ophthalmol. Jun 2019;202:47-54.
doi:10.1016/j.ajo.2019.01.026
172. Roh HC, Lee JK, Kim M, et al. Systemic Comorbidities of Dry Eye Syndrome:
The Korean National Health and Nutrition Examination Survey V, 2010 to 2012.
Cornea. Feb 2016;35(2):187-92. doi:10.1097/ICO.0000000000000657
173. Lim MN, Lee JY, Hyon JY, Han SB. Association of self-reported psychiatric and
systemic risk factors in dry eye disease in adult Korean population. Eye (Lond).
Apr 2024;38(5):917-922. doi:10.1038/s41433-023-02803-6
174. Siak JJK, Tong L, Wong WL, et al. Prevalence and Risk Factors of Meibomian
Gland Dysfunction: The Singapore Malay Eye Study. Cornea. Nov
2012;31(11):1223-1228. doi:10.1097/ICO.0b013e31823f0977
175. Viso E, Rodriguez-Ares MT, Abelenda D, Oubina B, Gude F. Prevalence of
asymptomatic and symptomatic meibomian gland dysfunction in the general
population of Spain. Invest Ophthalmol Vis Sci. May 4 2012;53(6):2601-6.
doi:10.1167/iovs.11-9228
176. Lin JB, Shen X, Pfeifer CW, et al. Dry eye disease in mice activates adaptive
corneal epithelial regeneration distinct from constitutive renewal in homeostasis.
Proc Natl Acad Sci U S A. Jan 10 2023;120(2):e2204134120.
doi:10.1073/pnas.2204134120
177. Hashemi H, Rastad H, Emamian MH, Fotouhi A. Meibomian gland dysfunction
and its determinants in Iranian adults: A population-based study. Cont Lens
Anterior Eye. Aug 2017;40(4):213-216. doi:10.1016/j.clae.2017.05.003
178. Aapola U, Nattinen J, Suurkuukka I, et al. Ocular surface health of the Finnish
elderly population. Acta Ophthalmol. Dec 2022;100(8):894-902.
doi:10.1111/aos.15130
179. Aljarousha M, Badarudin NE, Che Azemin MZ, Aljeesh Y, Amer A, Abdul Rahim
MAS. Prevalence and Risk Factors of Dry Eye Disease in the South of
Palestine. Malays J Med Sci. Apr 2024;31(2):72-97.
doi:10.21315/mjms2024.31.2.8
180. Alshamrani AA, Almousa AS, Almulhim AA, et al. Prevalence and Risk Factors
of Dry Eye Symptoms in a Saudi Arabian Population. Middle East Afr J
Ophthalmol. Apr-Jun 2017;24(2):67-73. doi:10.4103/meajo.MEAJO_281_16
181. Betiku AO, Oduyoye OO, Jagun OO, Olajide OS, Adebusoye SO, Aham-
Onyebuchi UO. Prevalence and risk factors associated with dry eye disease
among adults in a population-based setting in South-West Nigeria. Niger J Clin
Pract. Mar 2022;25(3):354-360. doi:10.4103/njcp.njcp_1598_21
182. Caffery B, Srinivasan S, Reaume CJ, et al. Prevalence of dry eye disease in
Ontario, Canada: A population-based survey. Ocul Surf. Jul 2019;17(3):526-
531. doi:10.1016/j.jtos.2019.02.011
183. Castro JS, Selegatto IB, Castro RS, et al. Prevalence and Risk Factors of self-
reported dry eye in Brazil using a short symptom questionnaire. Sci Rep. Feb 1
2018;8(1):2076. doi:10.1038/s41598-018-20273-9
152
184. Chatterjee S, Agrawal D, Sanowar G, Kandoi R. Prevalence of symptoms of dry
eye disease in an urban Indian population. Indian J Ophthalmol. May
2021;69(5):1061-1066. doi:10.4103/ijo.IJO_1796_20
185. Farrand KF, Fridman M, Stillman IO, Schaumberg DA. Prevalence of
Diagnosed Dry Eye Disease in the United States Among Adults Aged 18 Years
and Older. Am J Ophthalmol. Oct 2017;182:90-98.
doi:10.1016/j.ajo.2017.06.033
186. Gong YY, Zhang F, Zhou J, et al. Prevalence of Dry Eye in Uyghur and Han
Ethnic Groups in Western China. Ophthalmic epidemiology. Jun
2017;24(3):181-187. doi:10.1080/09286586.2016.1263996
187. Graue-Hernandez EO, Serna-Ojeda JC, Estrada-Reyes C, Navas A, Arrieta-
Camacho J, Jimenez-Corona A. Dry eye symptoms and associated risk factors
among adults aged 50 or more years in Central Mexico. Salud Publica Mex.
Sep-Oct 2018;60(5):520-527. doi:10.21149/9024
188. Hashemi H, Asharlous A, Aghamirsalim M, et al. Meibomian gland dysfunction
in geriatric population: tehran geriatric eye study. Int Ophthalmol. Jul
2021;41(7):2539-2546. doi:10.1007/s10792-021-01812-2
189. Kim KI, Park YS, Kim RH, Kim JH. Factors Associated with Dry Eye Symptoms
in Elderly Koreans: the Fifth Korea National Health and Nutrition Examination
Survey 2010-2012. Korean J Fam Med. Jan 2019;40(1):22-30.
doi:10.4082/kjfm.17.0058
190. Lim EWL, Chong CCY, Nusinovici S, et al. Relationship between dry eye
symptoms and quality of life: associations and mediation analysis. Br J
Ophthalmol. Nov 2023;107(11):1606-1612. doi:10.1136/bjo-2022-321246
191. Lin F, Cai Y, Fei X, Wang Y, Zhou M, Liu Y. Prevalence of dry eye disease
among Chinese high school students during the COVID-19 outbreak. BMC
Ophthalmol. Apr 26 2022;22(1):190. doi:10.1186/s12886-022-02408-9
192. Mai ELC, Lin CC, Lian I, Liao R, Chen M, Chang C. Population-based study on
the epidemiology of dry eye disease and its association with presbyopia and
other risk factors. Int Ophthalmol. Dec 2019;39(12):2731-2739.
doi:10.1007/s10792-019-01117-5
193. Marculino LGC, Hazarbassanov RM, Hazarbassanov N, et al. Prevalence and
risk factors for dry eye disease: the Sao Paulo dry eye study. Arq Bras Oftalmol.
2022;85(6):549-557. doi:10.5935/0004-2749.202200100
194. Ooi KG, Lee MH, Burlutsky G, Gopinath B, Mitchell P, Watson S. Association of
dyslipidaemia and oral statin use, and dry eye disease symptoms in the Blue
Mountains Eye Study. Clin Exp Ophthalmol. Mar 2019;47(2):187-192.
doi:10.1111/ceo.13388
195. Park HW, Park JW. The Association between Symptoms of Dry Eye Syndrome
and Metabolic Outcome in a General Population in Korea. J Korean Med Sci.
Jul 2016;31(7):1121-6. doi:10.3346/jkms.2016.31.7.1121
196. Reitmeir P, Linkohr B, Heier M, et al. Common eye diseases in older adults of
southern Germany: results from the KORA-Age study. Age Ageing. May 1
2017;46(3):481-486. doi:10.1093/ageing/afw234
197. Tonkerdmongkol D, Poyomtip T, Poolsanam C, Watcharapalakorn A,
Tawonkasiwattanakun P. Prevalence and associated factors for self-reported
153
symptoms of dry eye among Thai school children during the COVID-19
outbreak. PLoS One. 2023;18(4):e0284928. doi:10.1371/journal.pone.0284928
198. Wu SS, Fernando K, Allerton C, Jansen KU, Vincent MS, Dolsten M. Reviving
an R&D pipeline: a step change in the Phase II success rate. Drug Discov
Today. Feb 2021;26(2):308-314. doi:10.1016/j.drudis.2020.10.019
199. Donthineni PR, Kammari P, Shanbhag SS, Singh V, Das AV, Basu S.
Incidence, demographics, types and risk factors of dry eye disease in India:
Electronic medical records driven big data analytics report I. Ocul Surf. Apr
2019;17(2):250-256. doi:10.1016/j.jtos.2019.02.007
200. Lin IC, Kuo YK, Liu HY, Chien LN. Trends in Diagnosed Dry Eye Disease
Incidence, 2001 to 2015: A Nationwide Population-Based Study in Taiwan.
Cornea. Nov 1 2022;41(11):1372-1377. doi:10.1097/ICO.0000000000002987
201. Christen WG, Cook NR, Manson JE, et al. Efficacy of Marine omega-3 Fatty
Acid Supplementation vs Placebo in Reducing Incidence of Dry Eye Disease in
Healthy US Adults: A Randomized Clinical Trial. JAMA Ophthalmol. Jul 1
2022;140(7):707-714. doi:10.1001/jamaophthalmol.2022.1818
202. Millan A, Viso E, Gude F, Parafita-Fernandez A, Morana N, Rodriguez-Ares MT.
Incidence and Risk Factors of Dry Eye in a Spanish Adult Population: 11-Year
Follow-Up From the Salnes Eye Study. Cornea. Dec 2018;37(12):1527-1534.
doi:10.1097/ICO.0000000000001713
203. Man REK, Veerappan AR, Tan SP, et al. Incidence and risk factors of
symptomatic dry eye disease in Asian Malays from the Singapore Malay Eye
Study. Ocul Surf. Oct 2017;15(4):742-748. doi:10.1016/j.jtos.2017.04.004
204. Lienert JP, Tarko L, Uchino M, Christen WG, Schaumberg DA. Long-term
Natural History of Dry Eye Disease from the Patient's Perspective.
Ophthalmology. Feb 2016;123(2):425-433. doi:10.1016/j.ophtha.2015.10.011
205. Jones L, Downie LE, Korb D, et al. TFOS DEWS II Management and Therapy
Report. Ocul Surf. Jul 2017;15(3):575-628. doi:10.1016/j.jtos.2017.05.006
206. Cui D, Mathews P, Li G, VanCourt S, Akpek E. Outcomes of Sjögren’s versus
non-Sjögren’s related dry eye in a longitudinal, tertiary clinic-based sample.
PLOS ONE. 2021;16(12):e0261241. doi:10.1371/journal.pone.0261241
207. Garcia-Queiruga J, Pena-Verdeal H, Sabucedo-Villamarin B, Garcia-Resua C,
Giraldez MJ, Yebra-Pimentel E. Temporal Progression of Entry Factors into the
Vicious Circle of Dry Eye in Untreated Sufferers. Life (Basel). Jun 26
2024;14(7)doi:10.3390/life14070806
208. Wan X, Wu Y, Zhai Z, et al. Factors affecting long-term changes of meibomian
gland in MGD patients. Graefes Arch Clin Exp Ophthalmol. Feb
2024;262(2):527-535. doi:10.1007/s00417-023-06210-1
209. Turk MA, Hayworth JL, Nevskaya T, Pope JE. Ocular Manifestations in
Rheumatoid Arthritis, Connective Tissue Disease, and Vasculitis: A Systematic
Review and Metaanalysis. J Rheumatol. Jan 1 2021;48(1):25-34.
doi:10.3899/jrheum.190768
210. Yang W, Yang K, Pan Y, et al. A literature-derived dataset on risk factors for dry
eye disease. Sci Data. Jan 11 2023;10(1):21. doi:10.1038/s41597-023-01931-8
154
211. Wang L, Xie Y, Deng Y. Prevalence of dry eye in patients with systemic lupus
erythematosus: a meta-analysis. BMJ Open. Sep 29 2021;11(9):e047081.
doi:10.1136/bmjopen-2020-047081
212. Qian L, Wei W. Identified risk factors for dry eye syndrome: A systematic review
and meta-analysis. PLoS One. 2022;17(8):e0271267.
doi:10.1371/journal.pone.0271267
213. Gorimanipalli B, Khamar P, Sethu S, Shetty R. Hormones and dry eye disease.
Indian J Ophthalmol. Apr 2023;71(4):1276-1284. doi:10.4103/IJO.IJO_2887_22
214. Vehof J, Snieder H, Jansonius N, Hammond CJ. Prevalence and risk factors of
dry eye in 79,866 participants of the population-based Lifelines cohort study in
the Netherlands. Ocul Surf. Jan 2021;19:83-93. doi:10.1016/j.jtos.2020.04.005
215. Ko YT, Wu YM, Wu HL, et al. Inflammatory bowel disease and the associated
risk of dry eye and ocular surface injury: a nationwide matched cohort study.
BMC Ophthalmol. Oct 13 2023;23(1):415. doi:10.1186/s12886-023-03165-z
216. Wu PC, Ma SH, Huang YY, Chang JY, Chang YT, Dai YX. Psoriasis and Dry
Eye Disease: A Systematic Review and Meta-Analysis. Dermatology.
2022;238(5):876-885. doi:10.1159/000522167
217. Yoo TK, Oh E. Diabetes mellitus is associated with dry eye syndrome: a meta-
analysis. Int Ophthalmol. Nov 2019;39(11):2611-2620. doi:10.1007/s10792-019-
01110-y
218. Sullivan DA, Rocha EM, Aragona P, et al. TFOS DEWS II Sex, Gender, and
Hormones Report. The ocular surface. Jul 2017;15(3):284-333.
doi:10.1016/j.jtos.2017.04.001
219. Sullivan DA, Sullivan BD, Evans JE, et al. Androgen deficiency, Meibomian
gland dysfunction, and evaporative dry eye. Ann N Y Acad Sci. Jun
2002;966:211-22. doi:10.1111/j.1749-6632.2002.tb04217.x
220. Vehof J, Hysi PG, Hammond CJ. A Metabolome-Wide Study of Dry Eye
Disease Reveals Serum Androgens as Biomarkers. Ophthalmology. Apr
2017;124(4):505-511. doi:10.1016/j.ophtha.2016.12.011
221. Bonini S, Mantelli F, Moretti C, Lambiase A, Bonini S, Micera A. Itchy-dry eye
associated with polycystic ovary syndrome. Am J Ophthalmol. May
2007;143(5):763-771. doi:10.1016/j.ajo.2007.01.030
222. Sobolewska B, Schaller M, Zierhut M. Rosacea and Dry Eye Disease. Ocul
Immunol Inflamm. Apr 3 2022;30(3):570-579.
doi:10.1080/09273948.2021.2025251
223. Tavassoli S, Wong N, Chan E. Ocular manifestations of rosacea: A clinical
review. Clin Exp Ophthalmol. Mar 2021;49(2):104-117. doi:10.1111/ceo.13900
224. Koca S, Oral AY. Assessments of the ocular surface and meibomian gland
morphology in patients with treatment-naive acne vulgaris. Arq Bras Oftalmol.
Mar-Apr 2023;86(2):145-150. doi:10.5935/0004-2749.20230025
225. Huang Q, Zheng Y, Zhang C, et al. Association between asthma and dry eye
disease: a meta-analysis based on observational studies. BMJ Open. Dec 10
2021;11(12):e045275. doi:10.1136/bmjopen-2020-045275
226. Akasaki Y, Inomata T, Sung J, et al. Prevalence of Comorbidity between Dry
Eye and Allergic Conjunctivitis: A Systematic Review and Meta-Analysis. J Clin
Med. Jun 23 2022;11(13)doi:10.3390/jcm11133643
155
227. Galor A, Britten-Jones AC, Feng Y, et al. TFOS Lifestyle: Impact of lifestyle
challenges on the ocular surface. Ocul Surf. Apr 2023;28:262-303.
doi:10.1016/j.jtos.2023.04.008
228. Lee CJ, Levitt RC, Felix ER, Sarantopoulos CD, Galor A. Evidence that dry eye
is a comorbid pain condition in a U.S. veteran population. Pain Rep. Nov
2017;2(6):e629. doi:10.1097/PR9.0000000000000629
229. Vehof J, Kozareva D, Hysi PG, Hammond CJ. Prevalence and risk factors of
dry eye disease in a British female cohort. Br J Ophthalmol. Dec
2014;98(12):1712-7. doi:10.1136/bjophthalmol-2014-305201
230. Liu S, Dong H, Fang S, Zhang L. Risk of dry eye in headache patients: a
systematic review and meta-analysis. Ann Med. Dec 2022;54(1):2876-2885.
doi:10.1080/07853890.2022.2133165
231. Basilious A, Xu CY, Malvankar-Mehta MS. Dry eye disease and psychiatric
disorders: A systematic review and meta-analysis. Eur J Ophthalmol. Jul
2022;32(4):1872-1889. doi:10.1177/11206721211060963
232. Wan KH, Chen LJ, Young AL. Depression and anxiety in dry eye disease: a
systematic review and meta-analysis. Eye (Lond). Dec 2016;30(12):1558-1567.
doi:10.1038/eye.2016.186
233. Tsai CY, Jiesisibieke ZL, Tung TH. Association between dry eye disease and
depression: An umbrella review. Front Public Health. 2022;10:910608.
doi:10.3389/fpubh.2022.910608
234. Lin CW, Su YC, Liu JD, et al. Impact of Obstructive Sleep Apnea and
Continuous Positive Airway Pressure Treatment on Dry Eye Disease: A
Systematic Review and Meta-Analysis. Nat Sci Sleep. 2024;16:1921-1935.
doi:10.2147/NSS.S495883
235. Ayaki M, Tsubota K, Kawashima M, Kishimoto T, Mimura M, Negishi K. Sleep
Disorders are a Prevalent and Serious Comorbidity in Dry Eye. Invest
Ophthalmol Vis Sci. Nov 1 2018;59(14):DES143-DES150. doi:10.1167/iovs.17-
23467
236. Nagino K, Sung J, Oyama G, et al. Prevalence and characteristics of dry eye
disease in Parkinson's disease: a systematic review and meta-analysis. Sci
Rep. Nov 1 2022;12(1):18348. doi:10.1038/s41598-022-22037-y
237. Nichols KK, Foulks GN, Bron AJ, et al. The international workshop on
meibomian gland dysfunction: executive summary. Invest Ophthalmol Vis Sci.
Mar 30 2011;52(4):1922-9. doi:10.1167/iovs.10-6997a
238. Schaumberg DA, Nichols JJ, Papas EB, Tong L, Uchino M, Nichols KK. The
international workshop on meibomian gland dysfunction: report of the
subcommittee on the epidemiology of, and associated risk factors for, MGD.
Invest Ophthalmol Vis Sci. Mar 2011;52(4):1994-2005. doi:10.1167/iovs.10-
6997e
239. Lin A, Ahmad S, Amescua G, et al. Blepharitis Preferred Practice Pattern(R).
Ophthalmology. Apr 2024;131(4):P50-P86. doi:10.1016/j.ophtha.2023.12.036
240. Bron AJ, de Paiva CS, Chauhan S, et al. TFOS DEWS II Pathophysiology
Report. Ocular Surface. 2017;15(3):438-510.
156
241. Zhang AC, Muntz A, Wang MTM, Craig JP, Downie LE. Ocular Demodex: a
systematic review of the clinical literature. Ophthalmic Physiol Opt. Jul
2020;40(4):389-432. doi:10.1111/opo.12691
242. Ekin MA, Ugurlu SK, Imre SS, Kahraman HG. The role of meibomian gland
dysfunction on the development of dry eye disease in patients with facial nerve
palsy. Arq Bras Oftalmol. 2021;85(2):128-135. doi:10.5935/0004-
2749.20220021
243. Mistry RK, Hohman MH, Al-Sayed AA. Facial Nerve Trauma. StatPearls. 2024.
244. Kucuk E, Yilmaz U, Zor KR. Tear Film Functions and Dry Eye Symptoms in
Young Patients with Pterygium. Beyoglu Eye J. 2020;5(1):26-31.
doi:10.14744/bej.2019.18853
245. Napoli PE, Sanna R, Iovino C, Fossarello M. Resolution of pinguecula-related
dry eye disease after argon laser photocoagulation. Int Med Case Rep J.
2017;10:247-250. doi:10.2147/IMCRJ.S138786
246. Dong N, Li W, Lin H, et al. Abnormal epithelial differentiation and tear film
alteration in pinguecula. Invest Ophthalmol Vis Sci. Jun 2009;50(6):2710-5.
doi:10.1167/iovs.08-2905
247. Jeong J, Rand GM, Kwon T, Kwon JW. The Improvement of Dry Eye Symptoms
after Pinguecula Excision and Conjunctival Autograft with Fibrin Glue. J
Ophthalmol. 2019;2019:6438157. doi:10.1155/2019/6438157
248. Balci O. Clinical characteristics of patients with conjunctivochalasis. Clin
Ophthalmol. 2014;8:1655-60. doi:10.2147/OPTH.S61851
249. Chhadva P, Alexander A, McClellan AL, McManus KT, Seiden B, Galor A. The
impact of conjunctivochalasis on dry eye symptoms and signs. Invest
Ophthalmol Vis Sci. May 2015;56(5):2867-71. doi:10.1167/iovs.14-16337
250. Di Pascuale MA, Espana EM, Kawakita T, Tseng SC. Clinical characteristics of
conjunctivochalasis with or without aqueous tear deficiency. Br J Ophthalmol.
Mar 2004;88(3):388-92.
251. Kashkouli MB, Alemzadeh SA, Aghaei H, et al. Subjective versus objective dry
eye disease in patients with moderate-severe thyroid eye disease. Ocul Surf.
Oct 2018;16(4):458-462. doi:10.1016/j.jtos.2018.07.003
252. Park J, Baek S. Dry eye syndrome in thyroid eye disease patients: The role of
increased incomplete blinking and Meibomian gland loss. Acta Ophthalmol. Aug
2019;97(5):e800-e806. doi:10.1111/aos.14000
253. Bruscolini A, Abbouda A, Locuratolo N, Restivo L, Trimboli P, Romanelli F. Dry
Eye Syndrome in Non-Exophthalmic Graves' Disease. Semin Ophthalmol.
2015;30(5-6):372-6. doi:10.3109/08820538.2013.874491
254. Gurdal C, Sarac O, Genc I, Kirimlioglu H, Takmaz T, Can I. Ocular surface and
dry eye in Graves' disease. Curr Eye Res. Jan 2011;36(1):8-13.
doi:10.3109/02713683.2010.526285
255. Miura M, Inomata T, Nakamura M, et al. Prevalence and Characteristics of Dry
Eye Disease After Cataract Surgery: A Systematic Review and Meta-Analysis.
Ophthalmol Ther. Aug 2022;11(4):1309-1332. doi:10.1007/s40123-022-00513-y
256. Sambhi RS, Sambhi GDS, Mather R, Malvankar-Mehta MS. Dry eye after
refractive surgery: a meta-analysis. Can J Ophthalmol. Apr 2020;55(2):99-106.
doi:10.1016/j.jcjo.2019.07.005
157
257. Shen Z, Zhu Y, Song X, Yan J, Yao K. Dry Eye after Small Incision Lenticule
Extraction (SMILE) versus Femtosecond Laser-Assisted in Situ Keratomileusis
(FS-LASIK) for Myopia: A Meta-Analysis. PLoS One. 2016;11(12):e0168081.
doi:10.1371/journal.pone.0168081
258. Kobashi H, Kamiya K, Shimizu K. Dry Eye After Small Incision Lenticule
Extraction and Femtosecond Laser-Assisted LASIK: Meta-Analysis. Cornea.
Jan 2017;36(1):85-91. doi:10.1097/ICO.0000000000000999
259. Lu Q, Lu Y, Zhu X. Dry Eye and Phacoemulsification Cataract Surgery: A
Systematic Review and Meta-Analysis. Front Med (Lausanne). 2021;8:649030.
doi:10.3389/fmed.2021.649030
260. Gao M, Xia F, Wang P, Feng Z, Wang X. Influence of serial intravitreal
injections on measures of dry eye: A systemic review and meta-analysis. Cont
Lens Anterior Eye. Apr 2024;47(2):102127. doi:10.1016/j.clae.2024.102127
261. Gomes JAP, Azar DT, Baudouin C, et al. TFOS Lifestyle: Impact of elective
medications and procedures on the ocular surface. Ocul Surf. Jul 2023;29:331-
385. doi:10.1016/j.jtos.2023.04.011
262. Gomes JAP, Azar DT, Baudouin C, et al. TFOS DEWS II Iatrogenic Report. The
Ocular Surface. Jul 2017;15(3):511-538. doi:10.1016/j.jtos.2017.05.004
263. Sullivan DA, da Costa AX, Del Duca E, et al. TFOS Lifestyle: Impact of
cosmetics on the ocular surface. Ocul Surf. Jul 2023;29:77-130.
doi:10.1016/j.jtos.2023.04.005
264. Ogawa Y, Kawakami Y, Tsubota K. Cascade of Inflammatory, Fibrotic
Processes, and Stress-Induced Senescence in Chronic GVHD-Related Dry Eye
Disease. Int J Mol Sci. Jun 6 2021;22(11)doi:10.3390/ijms22116114
265. Soleimani M, Mahdavi Sharif P, Cheraqpour K, et al. Ocular graft-versus-host
disease (oGVHD): From A to Z. Surv Ophthalmol. Jul-Aug 2023;68(4):697-712.
doi:10.1016/j.survophthal.2023.02.006
266. Wolpert LE, Snieder H, Jansonius NM, Utheim TP, Hammond CJ, Vehof J.
Medication use and dry eye symptoms: A large, hypothesis-free, population-
based study in the Netherlands. Ocul Surf. Oct 2021;22:1-12.
doi:10.1016/j.jtos.2021.06.009
267. Hampel U, Schuster AK, Nickels S, et al. Schirmer test results: are they
associated with topical or systemic medication? Ocul Surf. Jan 2020;18(1):141-
147. doi:10.1016/j.jtos.2019.11.003
268. Katipoglu Z, Abay RN. The relationship between dry eye disease and
anticholinergic burden. Eye (Lond). Oct 2023;37(14):2921-2925.
doi:10.1038/s41433-023-02442-x
269. Monge-Carmona R, Caro-Magdaleno M, Sánchez-González MC. Association
between the use of prostaglandin analogues and ocular surface disease: a
systematic review. Eye (Lond). Oct 8 2024;doi:10.1038/s41433-024-03372-y
270. Stoicescu EA, Iancu RC, Popa Cherecheanu A, Iancu G. Ocular adverse effects
of anti-cancer chemotherapy. J Med Life. Jun 2023;16(6):818-821.
doi:10.25122/jml-2023-0041
271. Kam KW, Di Zazzo A, De Gregorio C, Narang P, Jhanji V, Basu S. A review on
drug-induced dry eye disease. Indian J Ophthalmol. Apr 2023;71(4):1263-1269.
doi:10.4103/IJO.IJO_2782_22
158
272. Boucher R, Haigh O, Barreau E, et al. Ocular surface toxicities associated with
modern anticancer therapies. Surv Ophthalmol. Mar-Apr 2024;69(2):198-210.
doi:10.1016/j.survophthal.2023.10.002
273. Inglis H, Boyle FM, Friedlander ML, Watson SL. Dry eyes and AIs: If you don't
ask you won't find out. Breast. Dec 2015;24(6):694-8.
doi:10.1016/j.breast.2015.08.008
274. Khoo P, Groeneveld T, Boyle F, O'Neill S, Forster B, Watson SL. Dry eye signs
and symptoms in patients on aromatase inhibitor therapy. Eye (Lond). Apr
2022;36(4):766-772. doi:10.1038/s41433-021-01538-6
275. Elubous KA, Toubasi AA, Elubous A, Alryalat SA, Abous H. Ocular
manifestations of systemic isotretinoin in patients with acne: a systemic review
and meta-analysis. Cutan Ocul Toxicol. Jun 2022;41(2):113-122.
doi:10.1080/15569527.2022.2050747
276. Caffery BE, Josephson JE. Ocular side effects of isotretinoin therapy. J Am
Optom Assoc. Mar 1988;59(3):221-4.
277. Mathers WD, Shields WJ, Sachdev MS, Petroll WM, Jester JV. Meibomian
gland morphology and tear osmolarity: changes with Accutane therapy. Cornea.
Jul 1991;10(4):286-90.
278. Serino F, Dattilo V, Cennamo M, et al. Dupilumab-Associated
Blepharoconjunctivitis: Clinical and Morphological Aspects. Biomedicines. Nov
21 2023;11(12)doi:10.3390/biomedicines11123104
279. Wu D, Daniel BS, Lai AJX, et al. Dupilumab-associated ocular manifestations: A
review of clinical presentations and management. Surv Ophthalmol. Sep-Oct
2022;67(5):1419-1442. doi:10.1016/j.survophthal.2022.02.002
280. Marolo P, Ribero S, Caselgrandi P, et al. Ocular Surface Disease in Patients
With Atopic Dermatitis Treated With Dupilumab: A Prospective Case-Control
Study. Cornea. Feb 1 2024;43(2):221-227.
doi:10.1097/ICO.0000000000003341
281. Aydin Kurna S, Oflaz Hacisalihoglu A, Altun A, et al. Effects of systemic anti-
androgen drugs on the ocular surface. J Fr Ophtalmol. Jun 2022;45(6):619-627.
doi:10.1016/j.jfo.2021.06.007
282. Chien HW, Lin CW, Lee CY, Huang JY, Yang SF, Wang K. The use of
androgen deprivation therapy for prostate cancer and its effect on the
subsequent dry eye disease: a population-based cohort study. Int J Med Sci.
2022;19(7):1103-1109. doi:10.7150/ijms.73417
283. Alves M, Asbell P, Dogru M, et al. TFOS Lifestyle Report: Impact of
environmental conditions on the ocular surface. Ocul Surf. Jul 2023;29:1-52.
doi:10.1016/j.jtos.2023.04.007
284. Nichols JJ, Willcox MD, Bron AJ, et al. The TFOS International Workshop on
Contact Lens Discomfort: executive summary. Invest Ophthalmol Vis Sci. Oct
18 2013;54(11):TFOS7-TFOS13. doi:10.1167/iovs.13-13212
285. Fjaervoll H, Fjaervoll K, Magno M, et al. The association between visual display
terminal use and dry eye: a review. Acta Ophthalmol. Jun 2022;100(4):357-375.
doi:10.1111/aos.15049
159
286. Wolffsohn JS, Lingham G, Downie LE, et al. TFOS Lifestyle: Impact of the
digital environment on the ocular surface. Ocul Surf. Apr 2023;28:213-252.
doi:10.1016/j.jtos.2023.04.004
287. Al-Mohtaseb Z, Schachter S, Shen Lee B, Garlich J, Trattler W. The
Relationship Between Dry Eye Disease and Digital Screen Use. Clin
Ophthalmol. 2021;15:3811-3820. doi:10.2147/OPTH.S321591
288. Stapleton F, Alves M, Bunya VY, et al. TFOS DEWS II Epidemiology Report.
Ocul Surf. Jul 2017;15(3):334-365. doi:10.1016/j.jtos.2017.05.003
289. Stapleton F, Abad JC, Barabino S, et al. TFOS lifestyle: Impact of societal
challenges on the ocular surface. The ocular surface. Apr 2023;28:165-199.
doi:10.1016/j.jtos.2023.04.006
290. Cai Y, Wei J, Zhou J, Zou W. Prevalence and Incidence of Dry Eye Disease in
Asia: A Systematic Review and Meta-Analysis. Ophthalmic Res.
2022;65(6):647-658. doi:10.1159/000525696
291. Wang MTM, Craig JP. Natural history of dry eye disease: Perspectives from
inter-ethnic comparison studies. Ocul Surf. Jul 2019;17(3):424-433.
doi:10.1016/j.jtos.2019.03.004
292. Markoulli M, Ahmad S, Arcot J, et al. TFOS Lifestyle: Impact of nutrition on the
ocular surface. Ocul Surf. Jul 2023;29:226-271. doi:10.1016/j.jtos.2023.04.003
293. Liu J, Dong Y, Wang Y. Vitamin D deficiency is associated with dry eye
syndrome: a systematic review and meta-analysis. Acta Ophthalmol. Dec
2020;98(8):749-754. doi:10.1111/aos.14470
294. Askari G, Rafie N, Miraghajani M, Heidari Z, Arab A. Association between
vitamin D and dry eye disease: A systematic review and meta-analysis of
observational studies. Cont Lens Anterior Eye. Oct 2020;43(5):418-425.
doi:10.1016/j.clae.2020.03.001
295. Labetoulle M, Baudouin C, Benitez Del Castillo JM, et al. How gut microbiota
may impact ocular surface homeostasis and related disorders. Prog Retin Eye
Res. May 2024;100:101250. doi:10.1016/j.preteyeres.2024.101250
296. Moon J, Yoon CH, Choi SH, Kim MK. Can Gut Microbiota Affect Dry Eye
Syndrome? Int J Mol Sci. Nov 10 2020;21(22)doi:10.3390/ijms21228443
297. Watane A, Raolji S, Cavuoto K, Galor A. Microbiome and immune-mediated dry
eye: a review. BMJ Open Ophthalmol. Jun 2022;7(1)doi:10.1136/bmjophth-
2021-000956
298. Hsu CC, Chuang HK, Hsiao YJ, et al. Predicting Risks of Dry Eye Disease
Development Using a Genome-Wide Polygenic Risk Score Model. Transl Vis
Sci Technol. May 1 2024;13(5):13. doi:10.1167/tvst.13.5.13
299. Vehof J, Zavos HMS, Lachance G, Hammond CJ, Williams FMK. Shared
genetic factors underlie chronic pain syndromes. Pain. Aug 2014;155(8):1562-
1568. doi:10.1016/j.pain.2014.05.002
300. Bjordal O, Norheim KB, Rodahl E, Jonsson R, Omdal R. Primary Sjogren's
syndrome and the eye. Surv Ophthalmol. Mar-Apr 2020;65(2):119-132.
doi:10.1016/j.survophthal.2019.10.004
301. Kiliccioglu A, Oncel D, Celebi ARC. Autoimmune Disease-Related Dry Eye
Diseases and Their Placement Under the Revised Classification Systems: An
Update. Cureus. Dec 2023;15(12):e50276. doi:10.7759/cureus.50276
160
302. Aoki T, Yokoi N, Nagata K, Deguchi H, Sekiyama Y, Sotozono C. Investigation
of the relationship between ocular sarcoidosis and dry eye. Sci Rep. Mar 2
2022;12(1):3469. doi:10.1038/s41598-022-07435-6
303. Bustamante-Arias A, Ruiz Lozano RE, Rodriguez-Garcia A. Dry eye disease, a
prominent manifestation of systemic autoimmune disorders. Eur J Ophthalmol.
Nov 2022;32(6):3142-3162. doi:10.1177/11206721221088259
304. Ruiz-Lozano RE, Hernandez-Camarena JC, Valdez-Garcia JE, et al. Ocular
involvement and complications of lichen planus, lichen planus pigmentosus, and
lichen planopilaris: A comprehensive review. Dermatol Ther. Nov
2021;34(6):e15137. doi:10.1111/dth.15137
305. Mohsenin A, Huang JJ. Ocular manifestations of systemic inflammatory
diseases. Conn Med. Oct 2012;76(9):533-44.
306. Knop E, Knop N, Millar T, Obata H, Sullivan DA. The international workshop on
meibomian gland dysfunction: report of the subcommittee on anatomy,
physiology, and pathophysiology of the meibomian gland. Investigative
ophthalmology & visual science. Mar 2011;52(4):1938-78. doi:10.1167/iovs.10-
6997c
307. Selter JH, Gire AI, Sikder S. The relationship between Graves' ophthalmopathy
and dry eye syndrome. Clin Ophthalmol. 2015;9:57-62.
doi:10.2147/OPTH.S76583
308. Achtsidis V, Eleftheriadou I, Kozanidou E, et al. Dry eye syndrome in subjects
with diabetes and association with neuropathy. Diabetes Care. Oct
2014;37(10):e210-1. doi:10.2337/dc14-0860
309. Faustino-Barros JF, Saranzo Sant'Ana AM, Dias LC, et al. Distinct Inflammatory
and Oxidative Effects of Diabetes Mellitus and Hypothyroidism in the Lacrimal
Functional Unit. Int J Mol Sci. Apr 10 2023;24(8)doi:10.3390/ijms24086974
310. Sriprasert I, Warren DW, Mircheff AK, Stanczyk FZ. Dry eye in postmenopausal
women: a hormonal disorder. Menopause. Mar 2016;23(3):343-51.
doi:10.1097/GME.0000000000000530
311. Chen X, Rao J, Zheng Z, et al. Integrated Tear Proteome and Metabolome
Reveal Panels of Inflammatory-Related Molecules via Key Regulatory
Pathways in Dry Eye Syndrome. J Proteome Res. May 3 2019;18(5):2321-
2330. doi:10.1021/acs.jproteome.9b00149
312. Leonardi A, Modugno RL, Salami E. Allergy and Dry Eye Disease. Ocul
Immunol Inflamm. Aug 18 2021;29(6):1168-1176.
doi:10.1080/09273948.2020.1841804
313. Aumond S, Bitton E. Palpebral and facial skin infestation by Demodex
folliculorum. Cont Lens Anterior Eye. Apr 2020;43(2):115-122.
doi:10.1016/j.clae.2019.09.001
314. Galor A, Covington D, Levitt AE, et al. Neuropathic Ocular Pain due to Dry Eye
Is Associated With Multiple Comorbid Chronic Pain Syndromes. The journal of
pain : official journal of the American Pain Society. Mar 2016;17(3):310-8.
doi:10.1016/j.jpain.2015.10.019
315. Salinas R, Puig M, Fry CL, Johnson DA, Kheirkhah A. Floppy eyelid syndrome:
A comprehensive review. Ocul Surf. Jan 2020;18(1):31-39.
doi:10.1016/j.jtos.2019.10.002
161
316. Acar M, Firat H, Yuceege M, Ardic S. Long-term effects of PAP on ocular
surface in obstructive sleep apnea syndrome. Can J Ophthalmol. Apr
2014;49(2):217-21. doi:10.1016/j.jcjo.2013.11.010
317. Hayirci E, Yagci A, Palamar M, Basoglu OK, Veral A. The effect of continuous
positive airway pressure treatment for obstructive sleep apnea syndrome on the
ocular surface. Cornea. Jun 2012;31(6):604-8.
doi:10.1097/ICO.0b013e31824a2040
318. Kheirandish-Gozal L, Gozal D. Obstructive Sleep Apnea and Inflammation:
Proof of Concept Based on Two Illustrative Cytokines. Int J Mol Sci. Jan 22
2019;20(3)doi:10.3390/ijms20030459
319. Jeng YT, Lin SY, Hu HY, Lee OK, Kuo LL. Osteoporosis and dry eye syndrome:
A previously unappreciated association that may alert active prevention of fall.
PLoS One. 2018;13(11):e0207008. doi:10.1371/journal.pone.0207008
320. Ungureanu L, Chaudhuri KR, Diaconu S, Falup-Pecurariu C. Dry eye in
Parkinson's disease: a narrative review. Front Neurol. 2023;14:1236366.
doi:10.3389/fneur.2023.1236366
321. Hassanzadeh S, Varmaghani M, Zarei-Ghanavati S, Heravian Shandiz J, Azimi
Khorasani A. Global Prevalence of Meibomian Gland Dysfunction: A Systematic
Review and Meta-Analysis. Ocul Immunol Inflamm. Jan 2 2021;29(1):66-75.
doi:10.1080/09273948.2020.1755441
322. Wang MTM, Muntz A, Lim J, et al. Ageing and the natural history of dry eye
disease: A prospective registry-based cross-sectional study. Ocul Surf. Oct
2020;18(4):736-741. doi:10.1016/j.jtos.2020.07.003
323. Nelson JD, Shimazaki J, Benitez-del-Castillo JM, et al. The international
workshop on meibomian gland dysfunction: report of the definition and
classification subcommittee. Invest Ophthalmol Vis Sci. Mar 2011;52(4):1930-7.
doi:10.1167/iovs.10-6997b
324. Devebacak A, Teker ME, Palamar M. The Influence of Pterygium on Meibomian
Glands and Dry Eye Parameters. Optom Vis Sci. Mar 1 2023;100(3):207-210.
doi:10.1097/OPX.0000000000001996
325. Fineide F, Lagali N, Adil MY, et al. Topical glaucoma medications - Clinical
implications for the ocular surface. Ocul Surf. Oct 2022;26:19-49.
doi:10.1016/j.jtos.2022.07.007
326. Fineide F, Magno M, Dahlo K, et al. Topical glaucoma medications - Possible
implications on the meibomian glands. Acta Ophthalmol. Jun 1
2024;doi:10.1111/aos.16728
327. Wang MTM, Tien L, Han A, et al. Impact of blinking on ocular surface and tear
film parameters. Ocul Surf. Oct 2018;16(4):424-429.
doi:10.1016/j.jtos.2018.06.001
328. Nair S, Kaur M, Sharma N, Titiyal JS. Refractive surgery and dry eye - An
update. Indian J Ophthalmol. Apr 2023;71(4):1105-1114.
doi:10.4103/IJO.IJO_3406_22
329. Naderi K, Gormley J, O'Brart D. Cataract surgery and dry eye disease: A
review. Eur J Ophthalmol. Sep 2020;30(5):840-855.
doi:10.1177/1120672120929958
162
330. Ipek T, Hanga MP, Hartwig A, Wolffsohn JS, O'Donnell C. Dry eye following
cataract surgery: The effect of light exposure using an in-vitro model. Cont Lens
Anterior Eye. Feb 2018;41(1):128-131. doi:10.1016/j.clae.2017.11.003
331. Matulonis UA, Birrer MJ, O'Malley DM, et al. Evaluation of Prophylactic
Corticosteroid Eye Drop Use in the Management of Corneal Abnormalities
Induced by the Antibody-Drug Conjugate Mirvetuximab Soravtansine. Clin
Cancer Res. Mar 15 2019;25(6):1727-1736. doi:10.1158/1078-0432.Ccr-18-
2474
332. Ge H, Di G, Song P, Han W, Chen P, Wang Y. Role of vitamin A on the ocular
surface. Exp Eye Res. Jan 2025;250:110179. doi:10.1016/j.exer.2024.110179
333. Samarawickrama C, Chew S, Watson S. Retinoic acid and the ocular surface.
Surv Ophthalmol. May-Jun 2015;60(3):183-95.
doi:10.1016/j.survophthal.2014.10.001
334. Schiffman RM, Christianson MD, Jacobsen G, Hirsch JD, Reis BL. Reliability
and validity of the Ocular Surface Disease Index. Archives of ophthalmology.
May 2000;118(5):615-21.
335. Mehra D, Galor A. Digital Screen Use and Dry Eye: A Review. Asia Pac J
Ophthalmol (Phila). Dec 2020;9(6):491-497.
doi:10.1097/APO.0000000000000328
336. Fjaervoll K, Fjaervoll H, Magno M, et al. Review on the possible
pathophysiological mechanisms underlying visual display terminal-associated
dry eye disease. Acta Ophthalmol. Dec 2022;100(8):861-877.
doi:10.1111/aos.15150
337. Pontelli RCN, Rocha BA, Garcia DM, et al. Endocrine disrupting chemicals
associated with dry eye syndrome. Ocul Surf. Jul 2020;18(3):487-493.
doi:10.1016/j.jtos.2020.01.001
338. Tariq MA, Amin H, Ahmed B, Ali U, Mohiuddin A. Association of dry eye disease
with smoking: A systematic review and meta-analysis. Indian J Ophthalmol. Jun
2022;70(6):1892-1904. doi:10.4103/ijo.IJO_2193_21
339. Bandara NA, Burgos-Blasco B, Zhou XR, et al. The Impact of Vaping on the
Ocular Surface: A Systematic Review of the Literature. J Clin Med. Apr 29
2024;13(9)doi:10.3390/jcm13092619
340. Makrynioti D, Zagoriti Z, Koutsojannis C, Morgan PB, Lagoumintzis G. Ocular
conditions and dry eye due to traditional and new forms of smoking: A review.
Cont Lens Anterior Eye. Jun 2020;43(3):277-284.
doi:10.1016/j.clae.2020.02.009
341. Md Isa NA, Koh PY, Doraj P. The Tear Function in Electronic Cigarette
Smokers. Optom Vis Sci. Sep 2019;96(9):678-685.
doi:10.1097/opx.0000000000001422
342. Vehof J, Sillevis Smitt-Kamminga N, Nibourg SA, Hammond CJ. Sex
differences in clinical characteristics of dry eye disease. The ocular surface. Apr
2018;16(2):242-248. doi:10.1016/j.jtos.2018.01.001
343. Courtin R, Pereira B, Naughton G, et al. Prevalence of dry eye disease in visual
display terminal workers: a systematic review and meta-analysis. BMJ Open.
Jan 14 2016;6(1):e009675. doi:10.1136/bmjopen-2015-009675
163
344. Song P, Xia W, Wang M, et al. Variations of dry eye disease prevalence by age,
sex and geographic characteristics in China: a systematic review and meta-
analysis. J Glob Health. Dec 2018;8(2):020503. doi:10.7189/jogh.08.020503
345. Uchino M. What We Know About the Epidemiology of Dry Eye Disease in
Japan. Invest Ophthalmol Vis Sci. Nov 1 2018;59(14):DES1-DES6.
doi:10.1167/iovs.17-23491
346. McCann P, Abraham AG, Mukhopadhyay A, et al. Prevalence and Incidence of
Dry Eye and Meibomian Gland Dysfunction in the United States: A Systematic
Review and Meta-analysis. JAMA Ophthalmol. Dec 1 2022;140(12):1181-1192.
doi:10.1001/jamaophthalmol.2022.4394
347. Goodman CF, Doan T, Mehra D, et al. Case-Control Study Examining the
Composition of the Gut Microbiome in Individuals With and Without Immune-
Mediated Dry Eye. Cornea. Nov 1 2023;42(11):1340-1348.
doi:10.1097/ICO.0000000000003195
348. Napolitano P, Filippelli M, Davinelli S, Bartollino S, dell'Omo R, Costagliola C.
Influence of gut microbiota on eye diseases: an overview. Ann Med. Dec
2021;53(1):750-761. doi:10.1080/07853890.2021.1925150
349. Labetoulle M, Baudouin C, Benitez del Castillo JM, et al. How gut microbiota
may impact ocular surface homeostasis and related disorders. Progress in
retinal and eye research. 2024/05/01/ 2024;100:101250.
doi:https://doi.org/10.1016/j.preteyeres.2024.101250
350. Pilkington M, Lloyd D, Guo B, Watson SL, Ooi KG-J. Effects of dietary
imbalances of micro- and macronutrients on the ocular microbiome and its
implications in dry eye disease. Exploration of Medicine. 2024;5(1):127-147.
doi:10.37349/emed.2024.00211
351. Wu Y, Xie Y, Yuan Y, et al. The Mediterranean Diet and Age-Related Eye
Diseases: A Systematic Review. Nutrients. Apr 23
2023;15(9)doi:10.3390/nu15092043
352. Contreras-Ruiz L, Ryan DS, Sia RK, Bower KS, Dartt DA, Masli S.
Polymorphism in THBS1 gene is associated with post-refractive surgery chronic
ocular surface inflammation. Ophthalmology. Jul 2014;121(7):1389-97.
doi:10.1016/j.ophtha.2014.01.033
353. He Y, Li X, Bao Y, Sun J, Liu J. [The correlation of polymorphism of estrogen
receptor gene to dry eye syndrome in postmenopausal women]. Yan Ke Xue
Bao. Dec 2006;22(4):233-6.
354. Imbert Y, Darling DS, Jumblatt MM, et al. MUC1 splice variants in human ocular
surface tissues: possible differences between dry eye patients and normal
controls. Exp Eye Res. Sep 2006;83(3):493-501.
doi:10.1016/j.exer.2006.01.031
355. Imbert Y, Foulks GN, Brennan MD, et al. MUC1 and estrogen receptor alpha
gene polymorphisms in dry eye patients. Exp Eye Res. Mar 2009;88(3):334-8.
doi:10.1016/j.exer.2008.05.019
356. Na KS, Mok JW, Kim JY, Joo CK. Proinflammatory gene polymorphisms are
potentially associated with Korean non-Sjogren dry eye patients. Mol Vis.
2011;17:2818-23.
164
357. Ben-Eli H, Gomel N, Aframian DJ, et al. SNP variations in IL10, TNFalpha and
TNFAIP3 genes in patients with dry eye syndrome and Sjogren's syndrome. J
Inflamm (Lond). 2019;16:6. doi:10.1186/s12950-019-0209-z
358. Polanska V, Sery O, Fojtik Z, Hlinomazova Z. [The presence of dry eye
syndrome and corneal complications in patients with rheumatoid arthritis and its
association with -174 gene polymorphism for interleukin 6]. Cesk Slov Oftalmol.
Mar 2008;64(2):77-80. Vyskyt syndromu sucheho oka a rohovkovych
komplikaci u pacientu s revmatoidni artritidou a jeho souvislost s
polymorfismem -174 genu pro interleukin 6.
359. Rabensteiner DF, Aminfar H, Boldin I, Schwantzer G, Horwath-Winter J. The
prevalence of meibomian gland dysfunction, tear film and ocular surface
parameters in an Austrian dry eye clinic population. Acta Ophthalmol. Sep
2018;96(6):e707-e711. doi:10.1111/aos.13732
360. Sullivan BD, Smith GT, Gupta A, Harman F, Ansari E. Impact of Clinician
Subjectivity on the Assessment of Dry Eye Disease Prevalence in a UK Public
Health Care Patient Population. Clin Ophthalmol. 2024;18:743-753.
doi:10.2147/OPTH.S452149
361. Arita R, Itoh K, Inoue K, Amano S. Noncontact infrared meibography to
document age-related changes of the meibomian glands in a normal population.
Ophthalmology. May 2008;115(5):911-5. doi:10.1016/j.ophtha.2007.06.031
362. Morthen MK, Magno MS, Utheim TP, Snieder H, Hammond CJ, Vehof J. The
physical and mental burden of dry eye disease: A large population-based study
investigating the relationship with health-related quality of life and its
determinants. Ocul Surf. Jul 2021;21:107-117. doi:10.1016/j.jtos.2021.05.006
363. McDonald M, Patel DA, Keith MS, Snedecor SJ. Economic and Humanistic
Burden of Dry Eye Disease in Europe, North America, and Asia: A Systematic
Literature Review. Ocul Surf. Apr 2016;14(2):144-67.
doi:10.1016/j.jtos.2015.11.002
364. Boboridis KG, Messmer EM, Benítez-Del-Castillo J, et al. Patient-reported
burden and overall impact of dry eye disease across eight European countries:
a cross-sectional web-based survey. BMJ Open. Mar 17 2023;13(3):e067007.
doi:10.1136/bmjopen-2022-067007
365. Hossain P, Siffel C, Joseph C, Meunier J, Markowitz JT, Dana R. Patient-
reported burden of dry eye disease in the UK: a cross-sectional web-based
survey. BMJ Open. Mar 4 2021;11(3):e039209. doi:10.1136/bmjopen-2020-
039209
366. McDonnell PJ, Pflugfelder SC, Stern ME, et al. Study design and baseline
findings from the progression of ocular findings (PROOF) natural history study
of dry eye. BMC Ophthalmol. Dec 28 2017;17(1):265. doi:10.1186/s12886-017-
0646-5
367. Tong L, Waduthantri S, Wong TY, et al. Impact of symptomatic dry eye on
vision-related daily activities: the Singapore Malay Eye Study. Eye (Lond). Sep
2010;24(9):1486-91. doi:10.1038/eye.2010.67
368. Li M, Gong L, Chapin WJ, Zhu M. Assessment of vision-related quality of life in
dry eye patients. Invest Ophthalmol Vis Sci. Aug 17 2012;53(9):5722-7.
doi:10.1167/iovs.11-9094
165
369. Schiffman RM, Walt JG, Jacobsen G, Doyle JJ, Lebovics G, Sumner W. Utility
assessment among patients with dry eye disease. Ophthalmology. Jul
2003;110(7):1412-9. doi:10.1016/s0161-6420(03)00462-7
370. Wlodarczyk J, Fairchild C. United States cost-effectiveness study of two dry eye
ophthalmic lubricants. Ophthalmic Epidemiol. Jan-Feb 2009;16(1):22-30.
doi:10.1080/09286580802521309
371. Brown MM, Brown GC, Brown HC, Peet J, Roth Z. Value-based medicine,
comparative effectiveness, and cost-effectiveness analysis of topical
cyclosporine for the treatment of dry eye syndrome. Arch Ophthalmol. Feb
2009;127(2):146-52. doi:10.1001/archophthalmol.2008.608
372. Perumal N, Funke S, Pfeiffer N, Grus FH. Proteomics analysis of human tears
from aqueous-deficient and evaporative dry eye patients. Sci Rep. Jul 20
2016;6:29629. doi:10.1038/srep29629
373. Augustin AJ, Spitznas M, Kaviani N, et al. Oxidative reactions in the tear fluid of
patients suffering from dry eyes. Graefes Arch Clin Exp Ophthalmol. Nov
1995;233(11):694-8. doi:10.1007/BF00164671
374. Boehm N, Funke S, Wiegand M, Wehrwein N, Pfeiffer N, Grus FH. Alterations
in the tear proteome of dry eye patients--a matter of the clinical phenotype.
Invest Ophthalmol Vis Sci. Mar 28 2013;54(3):2385-92. doi:10.1167/iovs.11-
8751
375. Enriquez-de-Salamanca A, Castellanos E, Stern ME, et al. Tear cytokine and
chemokine analysis and clinical correlations in evaporative-type dry eye
disease. Mol Vis. May 19 2010;16:862-73.
376. Hamm-Alvarez SF, Janga SR, Edman MC, et al. Tear cathepsin S as a
candidate biomarker for Sjogren's syndrome. Arthritis Rheumatol. Jul
2014;66(7):1872-81. doi:10.1002/art.38633
377. Wang HH, Chen WY, Huang YH, et al. Interleukin-20 is involved in dry eye
disease and is a potential therapeutic target. J Biomed Sci. Jun 9
2022;29(1):36. doi:10.1186/s12929-022-00821-2
378. Chotikavanich S, de Paiva CS, Li de Q, et al. Production and activity of matrix
metalloproteinase-9 on the ocular surface increase in dysfunctional tear
syndrome. Invest Ophthalmol Vis Sci. Jul 2009;50(7):3203-9.
doi:10.1167/iovs.08-2476
379. Kantor NB, Tovar A, Wang T, Galor A. How does ocular graft-versus-host
disease fit under the dry eye umbrella? A review. Clin Exp Ophthalmol. Mar
2024;52(2):167-185. doi:10.1111/ceo.14347
380. Ogawa Y. Sjogren's Syndrome, Non-Sjogren's Syndrome, and Graft-Versus-
Host Disease Related Dry Eye. Invest Ophthalmol Vis Sci. Nov 1
2018;59(14):DES71-DES79. doi:10.1167/iovs.17-23750
381. Gilbard JP, Rossi SR, Heyda KG. Tear film and ocular surface changes after
closure of the meibomian gland orifices in the rabbit. Ophthalmology. Aug
1989;96(8):1180-6. doi:10.1016/s0161-6420(89)32753-9
382. Byun YS, Rho CR, Cho K, Choi JA, Na KS, Joo CK. Cyclosporine 0.05%
ophthalmic emulsion for dry eye in Korea: a prospective, multicenter, open-
label, surveillance study. Korean J Ophthalmol. Dec 2011;25(6):369-74.
doi:10.3341/kjo.2011.25.6.369
166
383. Iaccheri B, Torroni G, Cagini C, et al. Corneal confocal scanning laser
microscopy in patients with dry eye disease treated with topical cyclosporine.
Eye (Lond). May 2017;31(5):788-794. doi:10.1038/eye.2017.3
384. Cook N, Mullins A, Gautam R, et al. Evaluating Patient Experiences in Dry Eye
Disease Through Social Media Listening Research. Ophthalmol Ther. Sep
2019;8(3):407-420. doi:10.1007/s40123-019-0188-4
385. Park Y, Song JS, Choi CY, Yoon KC, Lee HK, Kim HS. A Randomized
Multicenter Study Comparing 0.1%, 0.15%, and 0.3% Sodium Hyaluronate with
0.05% Cyclosporine in the Treatment of Dry Eye. J Ocul Pharmacol Ther. Mar
2017;33(2):66-72. doi:10.1089/jop.2016.0086
386. Schwartz LM, Woloshin S. A Clear-eyed View of Restasis and Chronic Dry Eye
Disease. JAMA Intern Med. Feb 1 2018;178(2):181-182.
doi:10.1001/jamainternmed.2017.7904
387. Williamson JF, Huynh K, Weaver MA, Davis RM. Perceptions of dry eye
disease management in current clinical practice. Eye Contact Lens. Mar
2014;40(2):111-5. doi:10.1097/ICL.0000000000000020
388. Traish AM. Health Risks Associated with Long-Term Finasteride and
Dutasteride Use: It's Time to Sound the Alarm. World J Mens Health. Jul
2020;38(3):323-337. doi:10.5534/wjmh.200012
389. Barrera MJ, Aguilera S, Castro I, et al. Dysfunctional mitochondria as critical
players in the inflammation of autoimmune diseases: Potential role in Sjogren's
syndrome. Autoimmun Rev. Aug 2021;20(8):102867.
doi:10.1016/j.autrev.2021.102867
390. Datta S, Baudouin C, Brignole-Baudouin F, Denoyer A, Cortopassi GA. The Eye
Drop Preservative Benzalkonium Chloride Potently Induces Mitochondrial
Dysfunction and Preferentially Affects LHON Mutant Cells. Invest Ophthalmol
Vis Sci. Apr 1 2017;58(4):2406-2412. doi:10.1167/iovs.16-20903
391. Li Y, Liu H, Zeng W, Wei J. Edaravone protects against hyperosmolarity-
induced oxidative stress and apoptosis in primary human corneal epithelial
cells. PLoS One. 2017;12(3):e0174437. doi:10.1371/journal.pone.0174437
392. Sacca SC, Cutolo CA, Ferrari D, Corazza P, Traverso CE. The Eye, Oxidative
Damage and Polyunsaturated Fatty Acids. Nutrients. May 24
2018;10(6)doi:10.3390/nu10060668
393. Stuard WL, Guner MK, Robertson DM. IGFBP-3 Regulates Mitochondrial
Hyperfusion and Metabolic Activity in Ocular Surface Epithelia during
Hyperosmolar Stress. Int J Mol Sci. Apr 6 2022;23(7)doi:10.3390/ijms23074066
394. Wang B, Peng L, Ouyang H, et al. Induction of DDIT4 Impairs Autophagy
Through Oxidative Stress in Dry Eye. Invest Ophthalmol Vis Sci. Jul 1
2019;60(8):2836-2847. doi:10.1167/iovs.19-27072
395. Alves M, Calegari VC, Cunha DA, Saad MJ, Velloso LA, Rocha EM. Increased
expression of advanced glycation end-products and their receptor, and
activation of nuclear factor kappa-B in lacrimal glands of diabetic rats.
Diabetologia. Dec 2005;48(12):2675-81. doi:10.1007/s00125-005-0010-9
396. Alves M, Cunha DA, Calegari VC, et al. Nuclear factor-kappaB and advanced
glycation end-products expression in lacrimal glands of aging rats. J Endocrinol.
Oct 2005;187(1):159-66. doi:10.1677/joe.1.06209
167
397. Boga A, Stapleton F, Chapman M, Golebiowski B. Effects of elevated serum
estrogen on dry eye in women undergoing in vitro fertilisation. Ocul Surf. Jul
2023;29:511-520. doi:10.1016/j.jtos.2023.06.015
398. Grasso A, Di Zazzo A, Giannaccare G, et al. Sex Hormones Related Ocular
Dryness in Breast Cancer Women. J Clin Med. Jun 14
2021;10(12)doi:10.3390/jcm10122620
399. Li K, Zhang C, Yang Z, Wang Y, Si H. Evaluation of a novel dry eye model
induced by oral administration of finasteride. Mol Med Rep. Dec
2017;16(6):8763-8770. doi:10.3892/mmr.2017.7754
400. Sullivan DA. Androgen deficiency & dry eye syndromes. Arch Soc Esp Oftalmol.
Feb 2004;79(2):49-50.
401. Kamath V, Arvind A, Bhuvana K. Lid wiper epitheliopathy in symptomatic and
asymptomatic dry eye subjects. Indian J Ophthalmol. Apr 2023;71(4):1499-
1502. doi:10.4103/IJO.IJO_2709_22
402. van Setten GB. Impact of Attrition, Intercellular Shear in Dry Eye Disease:
When Cells are Challenged and Neurons are Triggered. Int J Mol Sci. Jun 18
2020;21(12)doi:10.3390/ijms21124333
403. van Setten GB. Expression of GPR-68 in Human Corneal and Conjunctival
Epithelium. Possible indicator and mediator of attrition associated inflammation
at the ocular surface. J Fr Ophtalmol. Jan 2023;46(1):19-24.
doi:10.1016/j.jfo.2022.07.006
404. van Setten GB, Mueller-Lierheim W, Baudouin C. Dry Eye Etiology: Focus on
Friction. Klin Monbl Augenheilkd. Oct 2020;237(10):1235-1236. Atiologie des
trockenen Auges: Reibung im Fokus. doi:10.1055/a-0898-3857
405. Vu CHV, Kawashima M, Yamada M, et al. Influence of Meibomian Gland
Dysfunction and Friction-Related Disease on the Severity of Dry Eye.
Ophthalmology. Aug 2018;125(8):1181-1188. doi:10.1016/j.ophtha.2018.01.025
406. Chen K, Li Y, Zhang X, Ullah R, Tong J, Shen Y. The role of the PI3K/AKT
signalling pathway in the corneal epithelium: recent updates. Cell Death Dis.
May 31 2022;13(5):513. doi:10.1038/s41419-022-04963-x
407. Han Y, Zhang Y, Yuan K, Wu Y, Jin X, Huang X. Hyperosmolarity promotes
macrophage pyroptosis by driving the glycolytic reprogramming of corneal
epithelial cells in dry eye disease. Front Med. Aug 2023;17(4):781-795.
doi:10.1007/s11684-023-0986-x
408. Lema C, Reins RY, Redfern RL. High-Mobility Group Box 1 in Dry Eye
Inflammation. Invest Ophthalmol Vis Sci. Apr 1 2018;59(5):1741-1750.
doi:10.1167/iovs.17-23363
409. Li H, Li J, Hou C, Li J, Peng H, Wang Q. The effect of astaxanthin on
inflammation in hyperosmolarity of experimental dry eye model in vitro and in
vivo. Exp Eye Res. Aug 2020;197:108113. doi:10.1016/j.exer.2020.108113
410. Lv Y, Chu C, Liu K, et al. A combination of CMC and alpha-MSH inhibited ROS
activated NLRP3 inflammasome in hyperosmolarity stressed HCECs and
scopolamine-induced dry eye rats. Sci Rep. Jan 13 2021;11(1):1184.
doi:10.1038/s41598-020-80849-2
411. Magny R, Kessal K, Regazzetti A, et al. Lipidomic analysis of epithelial corneal
cells following hyperosmolarity and benzalkonium chloride exposure: New
168
insights in dry eye disease. Biochim Biophys Acta Mol Cell Biol Lipids. Sep
2020;1865(9):158728. doi:10.1016/j.bbalip.2020.158728
412. Ren Y, Feng J, Lin Y, et al. MiR-223 inhibits hyperosmolarity-induced
inflammation through downregulating NLRP3 activation in human corneal
epithelial cells and dry eye patients. Exp Eye Res. Jul 2022;220:109096.
doi:10.1016/j.exer.2022.109096
413. Zhang Y, Li JM, Lu R, et al. Imbalanced IL-37/TNF-alpha/CTSS signaling
disrupts corneal epithelial barrier in a dry eye model in vitro. Ocul Surf. Oct
2022;26:234-243. doi:10.1016/j.jtos.2022.10.001
414. Zhang Y, Yang M, Zhao SX, Nie L, Shen LJ, Han W. Hyperosmolarity disrupts
tight junction via TNF-alpha/MMP pathway in primary human corneal epithelial
cells. Int J Ophthalmol. 2022;15(5):683-689. doi:10.18240/ijo.2022.05.01
415. Ortiz G, Chao C, Jamali A, et al. Effect of Dry Eye Disease on the Kinetics of
Lacrimal Gland Dendritic Cells as Visualized by Intravital Multi-Photon
Microscopy. Front Immunol. 2020;11:1713. doi:10.3389/fimmu.2020.01713
416. de Souza RG, de Paiva CS, Alves MR. Age-related Autoimmune Changes in
Lacrimal Glands. Immune Netw. Feb 2019;19(1):e3. doi:10.4110/in.2019.19.e3
417. Nair AP, D'Souza S, Shetty R, et al. Altered ocular surface immune cell profile
in patients with dry eye disease. Ocul Surf. Jul 2021;21:96-106.
doi:10.1016/j.jtos.2021.04.002
418. Pflugfelder SC, de Paiva CS. The Pathophysiology of Dry Eye Disease: What
We Know and Future Directions for Research. Ophthalmology. Nov
2017;124(11S):S4-S13. doi:10.1016/j.ophtha.2017.07.010
419. Stern ME, Theofilopoulos AN, Steven P, et al. Immunologic basis for
development of keratoconjunctivitis sicca in systemic autoimmune diseases:
Role of innate immune sensors. Ocul Surf. Apr 2024;32:130-138.
doi:10.1016/j.jtos.2024.02.003
420. Benitez-Del-Castillo J, Cantu-Dibildox J, Sanz-Gonzalez SM, Zanon-Moreno V,
Pinazo-Duran MD. Cytokine expression in tears of patients with glaucoma or
dry eye disease: A prospective, observational cohort study. Eur J Ophthalmol.
Jul 2019;29(4):437-443. doi:10.1177/1120672118795399
421. Henrioux F, Navel V, Belville C, et al. Inflammation of Dry Eye Syndrome: A
Cellular Study of the Epithelial and Macrophagic Involvement of NFAT5 and
RAGE. Int J Mol Sci. Jul 4 2023;24(13)doi:10.3390/ijms241311052
422. Kelagere Y, Scholand KK, DeJong EN, et al. TNF is a critical cytokine in age-
related dry eye disease. Ocul Surf. Oct 2023;30:119-128.
doi:10.1016/j.jtos.2023.08.004
423. Kumar NR, Praveen M, Narasimhan R, et al. Tear biomarkers in dry eye
disease: Progress in the last decade. Indian J Ophthalmol. Apr
2023;71(4):1190-1202. doi:10.4103/IJO.IJO_2981_22
424. Perez VL, Stern ME, Pflugfelder SC. Inflammatory basis for dry eye disease
flares. Exp Eye Res. Dec 2020;201:108294. doi:10.1016/j.exer.2020.108294
425. Roda M, Corazza I, Bacchi Reggiani ML, et al. Dry Eye Disease and Tear
Cytokine Levels-A Meta-Analysis. Int J Mol Sci. Apr 28
2020;21(9)doi:10.3390/ijms21093111
169
426. Roy NS, Wei Y, Ying GS, et al. Association of Tear Cytokine Concentrations
with Symptoms and Signs of Dry Eye Disease: Baseline Data from the Dry Eye
Assessment and Management (DREAM) Study. Curr Eye Res. Apr
2023;48(4):339-347. doi:10.1080/02713683.2022.2162088
427. de Souza GA, Godoy LM, Mann M. Identification of 491 proteins in the tear fluid
proteome reveals a large number of proteases and protease inhibitors. Genome
Biol. 2006;7(8):R72. doi:10.1186/gb-2006-7-8-R72
428. Edman MC, Janga SR, Meng Z, et al. Increased Cathepsin S activity associated
with decreased protease inhibitory capacity contributes to altered tear proteins
in Sjogren's Syndrome patients. Sci Rep. Jul 23 2018;8(1):11044.
doi:10.1038/s41598-018-29411-9
429. Joossen C, Baan A, Moreno-Cinos C, et al. A novel serine protease inhibitor as
potential treatment for dry eye syndrome and ocular inflammation. Sci Rep. Oct
14 2020;10(1):17268. doi:10.1038/s41598-020-74159-w
430. Choi W, Lian C, Ying L, et al. Expression of Lipid Peroxidation Markers in the
Tear Film and Ocular Surface of Patients with Non-Sjogren Syndrome: Potential
Biomarkers for Dry Eye Disease. Curr Eye Res. Sep 2016;41(9):1143-9.
doi:10.3109/02713683.2015.1098707
431. An S, Raju I, Surenkhuu B, et al. Neutrophil extracellular traps (NETs)
contribute to pathological changes of ocular graft-vs.-host disease (oGVHD) dry
eye: Implications for novel biomarkers and therapeutic strategies. Ocul Surf. Jul
2019;17(3):589-614. doi:10.1016/j.jtos.2019.03.010
432. Mahajan A, Hasikova L, Hampel U, et al. Aggregated neutrophil extracellular
traps occlude Meibomian glands during ocular surface inflammation. Ocul Surf.
Apr 2021;20:1-12. doi:10.1016/j.jtos.2020.12.005
433. Sonawane S, Khanolkar V, Namavari A, et al. Ocular surface extracellular DNA
and nuclease activity imbalance: a new paradigm for inflammation in dry eye
disease. Invest Ophthalmol Vis Sci. Dec 17 2012;53(13):8253-63.
doi:10.1167/iovs.12-10430
434. Moon J, Choi SH, Yoon CH, Kim MK. Gut dysbiosis is prevailing in Sjogren's
syndrome and is related to dry eye severity. PLoS One. 2020;15(2):e0229029.
doi:10.1371/journal.pone.0229029
435. Yoon CH, Ryu JS, Moon J, Kim MK. Association between aging-dependent gut
microbiome dysbiosis and dry eye severity in C57BL/6 male mouse model: a
pilot study. BMC Microbiol. Apr 9 2021;21(1):106. doi:10.1186/s12866-021-
02173-7
436. Fakih D, Zhao Z, Nicolle P, et al. Chronic dry eye induced corneal
hypersensitivity, neuroinflammatory responses, and synaptic plasticity in the
mouse trigeminal brainstem. J Neuroinflammation. Dec 17 2019;16(1):268.
doi:10.1186/s12974-019-1656-4
437. Sonkodi B, Marsovszky L, Csorba A, et al. Neural Regeneration in Dry Eye
Secondary to Systemic Lupus Erythematosus Is Also Disrupted like in
Rheumatoid Arthritis, but in a Progressive Fashion. Int J Mol Sci. Jun 26
2023;24(13)doi:10.3390/ijms241310680
438. Sonkodi B, Resch MD, Hortobagyi T. Is the Sex Difference a Clue to the
Pathomechanism of Dry Eye Disease? Watch out for the NGF-TrkA-Piezo2
170
Signaling Axis and the Piezo2 Channelopathy. J Mol Neurosci. Aug
2022;72(8):1598-1608. doi:10.1007/s12031-022-02015-9
439. Wang S, Naderi A, Kahale F, et al. Substance P regulates memory Th17 cell
generation and maintenance in chronic dry eye disease. J Leukoc Biol. Jun 25
2024;doi:10.1093/jleuko/qiae142
440. Wu M, Downie LE, Grover LM, et al. The neuroregenerative effects of topical
decorin on the injured mouse cornea. J Neuroinflammation. May 4
2020;17(1):142. doi:10.1186/s12974-020-01812-6
441. Yu M, Lee SM, Lee H, et al. Neurokinin-1 Receptor Antagonism Ameliorates
Dry Eye Disease by Inhibiting Antigen-Presenting Cell Maturation and T Helper
17 Cell Activation. Am J Pathol. Jan 2020;190(1):125-133.
doi:10.1016/j.ajpath.2019.09.020
442. Uchino Y. The Ocular Surface Glycocalyx and its Alteration in Dry Eye Disease:
A Review. Invest Ophthalmol Vis Sci. Nov 1 2018;59(14):DES157-DES162.
doi:10.1167/iovs.17-23756
443. Goto E, Endo K, Suzuki A, Fujikura Y, Matsumoto Y, Tsubota K. Tear
evaporation dynamics in normal subjects and subjects with obstructive
meibomian gland dysfunction. Invest Ophthalmol Vis Sci. Feb 2003;44(2):533-9.
doi:10.1167/iovs.02-0170
444. McCulley JP, Shine WE. Meibomian gland function and the tear lipid layer. Ocul
Surf. Jul 2003;1(3):97-106. doi:10.1016/s1542-0124(12)70138-6
445. Seo J, Byun WY, Alisafaei F, et al. Multiscale reverse engineering of the human
ocular surface. Nat Med. Aug 2019;25(8):1310-1318. doi:10.1038/s41591-019-
0531-2
446. Sullivan DA. Tearful relationships? Sex, hormones, the lacrimal gland, and
aqueous-deficient dry eye. Ocul Surf. Apr 2004;2(2):92-123. doi:10.1016/s1542-
0124(12)70147-7
447. Baudouin C, Rolando M, Benitez Del Castillo JM, et al. Reconsidering the
central role of mucins in dry eye and ocular surface diseases. Prog Retin Eye
Res. Jul 2019;71:68-87. doi:10.1016/j.preteyeres.2018.11.007
448. Rolando M, Barabino S, Giannaccare G, Aragona P. Dealing with the Persistent
Pathogenic Issues of Dry Eye Disease: The Importance of External and Internal
Stimuli and Tissue Responses. J Clin Med. Mar 13
2023;12(6)doi:10.3390/jcm12062205
449. Gilbard JP. Tear film osmolarity and keratoconjunctivitis sicca. CLAO J. Jul-Sep
1985;11(3):243-50.
450. Luo L, Li DQ, Pflugfelder SC. Hyperosmolarity-induced apoptosis in human
corneal epithelial cells is mediated by cytochrome c and MAPK pathways.
Cornea. May 2007;26(4):452-60. doi:10.1097/ICO.0b013e318030d259
451. Chen Z, Tong L, Li Z, et al. Hyperosmolarity-induced cornification of human
corneal epithelial cells is regulated by JNK MAPK. Invest Ophthalmol Vis Sci.
Feb 2008;49(2):539-49. doi:10.1167/iovs.07-0569
452. Igarashi T, Fujimoto C, Suzuki H, et al. Short-time exposure of hyperosmolarity
triggers interleukin-6 expression in corneal epithelial cells. Cornea. Dec
2014;33(12):1342-7. doi:10.1097/ICO.0000000000000256
171
453. Luo L, Li DQ, Corrales RM, Pflugfelder SC. Hyperosmolar saline is a
proinflammatory stress on the mouse ocular surface. Eye Contact Lens. Sep
2005;31(5):186-93. doi:10.1097/01.icl.0000162759.79740.46
454. Zibandeh N, Yildiz E, Ozer B, Tas AY, Sahin A. Androgen Suppresses
Hyperosmolarity-Induced Inflammatory Mediators in Human Corneal Epithelial
Cells. Cornea. Jul 2020;39(7):886-891. doi:10.1097/ICO.0000000000002291
455. Peng F, Jiang D, Xu W, et al. AMPK/MFF Activation: Role in Mitochondrial
Fission and Mitophagy in Dry Eye. Invest Ophthalmol Vis Sci. Nov 1
2022;63(12):18. doi:10.1167/iovs.63.12.18
456. Liu H, Begley C, Chen M, et al. A link between tear instability and
hyperosmolarity in dry eye. Invest Ophthalmol Vis Sci. Aug 2009;50(8):3671-9.
doi:10.1167/iovs.08-2689
457. Peng CC, Cerretani C, Braun RJ, Radke CJ. Evaporation-driven instability of
the precorneal tear film. Adv Colloid Interface Sci. Apr 2014;206:250-64.
doi:10.1016/j.cis.2013.06.001
458. Bron AJ, de Paiva CS, Chauhan SK, et al. TFOS DEWS II pathophysiology
report. Ocul Surf. Jul 2017;15(3):438-510. doi:10.1016/j.jtos.2017.05.011
459. Wakamatsu TH, Dogru M, Tsubota K. Tearful relations: oxidative stress,
inflammation and eye diseases. Arq Bras Oftalmol. Nov-Dec 2008;71(6
Suppl):72-9. doi:10.1590/s0004-27492008000700015
460. Shetty R, Subramani M, Murugeswari P, et al. Resveratrol Rescues Human
Corneal Epithelial Cells Cultured in Hyperosmolar Conditions: Potential for Dry
Eye Disease Treatment. Cornea. Dec 2020;39(12):1520-1532.
doi:10.1097/ICO.0000000000002495
461. Ayala A, Munoz MF, Arguelles S. Lipid peroxidation: production, metabolism,
and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. Oxid
Med Cell Longev. 2014;2014:360438. doi:10.1155/2014/360438
462. Ouyang W, Wang S, Yan D, et al. The cGAS-STING pathway-dependent
sensing of mitochondrial DNA mediates ocular surface inflammation. Signal
Transduct Target Ther. Sep 21 2023;8(1):371. doi:10.1038/s41392-023-01624-
z
463. Tan X, Chen Q, Chen Z, Sun Z, Chen W, Wei R. Mitochondrial DNA-Activated
cGAS-STING Signaling in Environmental Dry Eye. Invest Ophthalmol Vis Sci.
Apr 1 2024;65(4):33. doi:10.1167/iovs.65.4.33
464. Jackson DC, Zeng W, Wong CY, et al. Tear Interferon-Gamma as a Biomarker
for Evaporative Dry Eye Disease. Invest Ophthalmol Vis Sci. Sep 1
2016;57(11):4824-4830. doi:10.1167/iovs.16-19757
465. Ko JH, Kim S, Ryu JS, Song HJ, Oh JY. Interferon-gamma elicits the ocular
surface pathology mimicking dry eye through direct modulation of resident
corneal cells. Cell Death Discov. Jun 30 2023;9(1):209. doi:10.1038/s41420-
023-01511-0
466. Wu D, Zhang J, Qian T, et al. IFN-gamma Regulates the Expression of MICA in
Human Corneal Epithelium Through miRNA4448 and NFkappaB. Front
Immunol. 2018;9:1530. doi:10.3389/fimmu.2018.01530
172
467. Zhang X, Chen W, De Paiva CS, et al. Interferon-gamma exacerbates dry eye-
induced apoptosis in conjunctiva through dual apoptotic pathways. Invest
Ophthalmol Vis Sci. Aug 9 2011;52(9):6279-85. doi:10.1167/iovs.10-7081
468. Yang X, Zuo X, Zeng H, et al. IFN-gamma Facilitates Corneal Epithelial Cell
Pyroptosis Through the JAK2/STAT1 Pathway in Dry Eye. Invest Ophthalmol
Vis Sci. Mar 1 2023;64(3):34. doi:10.1167/iovs.64.3.34
469. Deng R, Hua X, Li J, et al. Oxidative stress markers induced by hyperosmolarity
in primary human corneal epithelial cells. PLoS One. 2015;10(5):e0126561.
doi:10.1371/journal.pone.0126561
470. Li J, Ruzhi D, Hua X, et al. Blueberry Component Pterostilbene Protects
Corneal Epithelial Cells from Inflammation via Anti-oxidative Pathway. Sci Rep.
Jan 14 2016;6:19408. doi:10.1038/srep19408
471. Marek V, Melik-Parsadaniantz S, Villette T, et al. Blue light phototoxicity toward
human corneal and conjunctival epithelial cells in basal and hyperosmolar
conditions. Free Radic Biol Med. Oct 2018;126:27-40.
doi:10.1016/j.freeradbiomed.2018.07.012
472. Wang B, Zeng H, Zuo X, et al. TLR4-Dependent DUOX2 Activation Triggered
Oxidative Stress and Promoted HMGB1 Release in Dry Eye. Front Med
(Lausanne). 2021;8:781616. doi:10.3389/fmed.2021.781616
473. Reins RY, Lema C, Courson J, Kunnen CME, Redfern RL. MyD88 Deficiency
Protects Against Dry Eye-Induced Damage. Invest Ophthalmol Vis Sci. Jun 1
2018;59(7):2967-2976. doi:10.1167/iovs.17-23397
474. Alven A, Lema C, Redfern RL. Impact of Low Humidity on Damage-associated
Molecular Patterns at the Ocular Surface during Dry Eye Disease. Optom Vis
Sci. Nov 1 2021;98(11):1231-1238. doi:10.1097/OPX.0000000000001802
475. Heimann H, Coupland SE, Gochman R, Hellmich M, Foerster MH. Alterations in
expression of mucin, tenascin-c and syndecan-1 in the conjunctiva following
retinal surgery and plaque radiotherapy. Graefes Arch Clin Exp Ophthalmol. Jul
2001;239(7):488-95. doi:10.1007/s004170100301
476. Lai CT, Yao WC, Lin SY, et al. Changes of Ocular Surface and the
Inflammatory Response in a Rabbit Model of Short-Term Exposure
Keratopathy. PLoS One. 2015;10(9):e0137186.
doi:10.1371/journal.pone.0137186
477. Kam W, Sullivan DA, Sullivan BD, Venkiteshwar M. Does hyperosmolarity
induce an irreversible process leading to human corneal epithelial cell death?
Investigative Ophthalmology & Visual Science. 2016;57(12):6181-6181.
478. Martinez-Carrasco R, Fini ME. Dynasore Protects Corneal Epithelial Cells
Subjected to Hyperosmolar Stress in an In Vitro Model of Dry Eye
Epitheliopathy. Int J Mol Sci. Mar 1 2023;24(5)doi:10.3390/ijms24054754
479. Moore JE, Vasey GT, Dartt DA, et al. Effect of tear hyperosmolarity and signs of
clinical ocular surface pathology upon conjunctival goblet cell function in the
human ocular surface. Invest Ophthalmol Vis Sci. Aug 3 2011;52(9):6174-80.
doi:10.1167/iovs.10-7022
480. Masli S, Akpek EK. Reduced tear thrombospondin-1/matrix metalloproteinase-9
ratio can aid in detecting Sjogren's syndrome etiology in patients with dry eye.
Clin Transl Sci. Aug 2022;15(8):1999-2009. doi:10.1111/cts.13316
173
481. Virtanen T, Konttinen YT, Honkanen N, Harkonen M, Tervo T. Tear fluid
plasmin activity of dry eye patients with Sjogren's syndrome. Acta Ophthalmol
Scand. Apr 1997;75(2):137-41. doi:10.1111/j.1600-0420.1997.tb00109.x
482. Schargus M, Ivanova S, Kakkassery V, Dick HB, Joachim S. Correlation of Tear
Film Osmolarity and 2 Different MMP-9 Tests With Common Dry Eye Tests in a
Cohort of Non-Dry Eye Patients. Cornea. Jul 2015;34(7):739-44.
doi:10.1097/ICO.0000000000000449
483. Arafat SN, Robert MC, Abud T, et al. Elevated Neutrophil Elastase in Tears of
Ocular Graft-Versus-Host Disease Patients. Am J Ophthalmol. Apr
2017;176:46-52. doi:10.1016/j.ajo.2016.12.026
484. Benitez-Del-Castillo JM, Soria J, Acera A, Munoz AM, Rodriguez S, Suarez T.
Quantification of a panel for dry-eye protein biomarkers in tears: A comparative
pilot study using standard ELISA and customized microarrays. Mol Vis.
2021;27:243-261.
485. Regmi SC, Samsom ML, Heynen ML, et al. Degradation of proteoglycan
4/lubricin by cathepsin S: Potential mechanism for diminished ocular surface
lubrication in Sjogren's syndrome. Exp Eye Res. Aug 2017;161:1-9.
doi:10.1016/j.exer.2017.05.006
486. Berchicci L, Aragona E, Arrigo A, et al. Conjunctival Matrix Metalloproteinase-9
Clinical Assessment in Early Ocular Graft versus Host Disease. J Ophthalmol.
2021;2021:9958713. doi:10.1155/2021/9958713
487. Zhou Q, Yang L, Wang Q, Li Y, Wei C, Xie L. Mechanistic investigations of
diabetic ocular surface diseases. Front Endocrinol (Lausanne).
2022;13:1079541. doi:10.3389/fendo.2022.1079541
488. Schmidt TA, Sullivan DA, Knop E, et al. Transcription, translation, and function
of lubricin, a boundary lubricant, at the ocular surface. JAMA Ophthalmol. Jun
2013;131(6):766-76. doi:10.1001/jamaophthalmol.2013.2385
489. Iwata S, Lemp MA, Holly FJ, Dohlman CH. Evaporation rate of water from the
precorneal tear film and cornea in the rabbit. Invest Ophthalmol. Dec
1969;8(6):613-9.
490. Georgiev GA, Eftimov P, Yokoi N. Contribution of Mucins towards the Physical
Properties of the Tear Film: A Modern Update. Int J Mol Sci. Dec 5
2019;20(24)doi:10.3390/ijms20246132
491. Ablamowicz AF, Nichols JJ. Ocular Surface Membrane-Associated Mucins.
Ocul Surf. Jul 2016;14(3):331-41. doi:10.1016/j.jtos.2016.03.003
492. Dilly PN. On the nature and the role of the subsurface vesicles in the outer
epithelial cells of the conjunctiva. Br J Ophthalmol. Jul 1985;69(7):477-81.
doi:10.1136/bjo.69.7.477
493. Martinez-Carrasco R, Rachagani S, Batra SK, Argueso P, Fini ME. Roles
unveiled for membrane-associated mucins at the ocular surface using a Muc4
knockout mouse model. Sci Rep. Aug 21 2023;13(1):13558.
doi:10.1038/s41598-023-40491-0
494. Weng J, Ross C, Baker J, Alfuraih S, Shamloo K, Sharma A. Diabetes-
Associated Hyperglycemia Causes Rapid-Onset Ocular Surface Damage.
Invest Ophthalmol Vis Sci. Nov 1 2023;64(14):11. doi:10.1167/iovs.64.14.11
174
495. Lin N, Chen X, Liu H, et al. Ectoine Enhances Mucin Production Via Restoring
IL-13/IFN-gamma Balance in a Murine Dry Eye Model. Invest Ophthalmol Vis
Sci. Jun 3 2024;65(6):39. doi:10.1167/iovs.65.6.39
496. Weng J, Trinh S, Lee R, Metwale R, Sharma A. Impact of High Glucose on
Ocular Surface Glycocalyx Components: Implications for Diabetes-Associated
Ocular Surface Damage. Int J Mol Sci. Nov 18
2022;23(22)doi:10.3390/ijms232214289
497. Mani R, Shobha PS, Thilagavathi S, et al. Altered mucins and aquaporins
indicate dry eye outcome in patients undergoing Vitreo-retinal surgery. PLoS
One. 2020;15(5):e0233517. doi:10.1371/journal.pone.0233517
498. Shamloo K, Barbarino A, Alfuraih S, Sharma A. Graft Versus Host Disease-
Associated Dry Eye: Role of Ocular Surface Mucins and the Effect of
Rebamipide, a Mucin Secretagogue. Invest Ophthalmol Vis Sci. Nov 1
2019;60(14):4511-4519. doi:10.1167/iovs.19-27843
499. AbuSamra DB, Martinez-Carrasco R, Argueso P. Galectins Differentially
Regulate the Surface Glycosylation of Human Monocytes. Biomolecules. Aug
23 2022;12(9)doi:10.3390/biom12091168
500. Fini ME, Jeong S, Gong H, et al. Membrane-associated mucins of the ocular
surface: New genes, new protein functions and new biological roles in human
and mouse. Prog Retin Eye Res. Mar 2020;75:100777.
doi:10.1016/j.preteyeres.2019.100777
501. Menon NG, Goyal R, Lema C, et al. Proteoglycan 4 (PRG4) expression and
function in dry eye associated inflammation. Exp Eye Res. Jul
2021;208:108628. doi:10.1016/j.exer.2021.108628
502. Nakamura Y, Yokoi N, Tokushige H, Kinoshita S. Sialic Acid in human tear fluid
decreases in dry eye. Jpn J Ophthalmol. Nov-Dec 2004;48(6):519-23.
doi:10.1007/s10384-004-0111-x
503. Hata-Mizuno M, Uchino Y, Uchino M, et al. Analysis of the Association between
Galectin-3 Concentration in Tears and the Severity of Dry Eye Disease: A
Case-Control Study. J Clin Med. Dec 23 2021;11(1)doi:10.3390/jcm11010066
504. Uchino Y, Mauris J, Woodward AM, et al. Alteration of galectin-3 in tears of
patients with dry eye disease. Am J Ophthalmol. Jun 2015;159(6):1027-1035
e3. doi:10.1016/j.ajo.2015.02.008
505. Uchino Y, Woodward AM, Mauris J, et al. Galectin-3 is an amplifier of the
interleukin-1beta-mediated inflammatory response in corneal keratinocytes.
Immunology. Jul 2018;154(3):490-499. doi:10.1111/imm.12899
506. Hoarau A, Polette M, Coraux C. Lung Hyaluronasome: Involvement of Low
Molecular Weight Ha (Lmw-Ha) in Innate Immunity. Biomolecules. Apr 30
2022;12(5)doi:10.3390/biom12050658
507. Muller-Lierheim WGK. Why Chain Length of Hyaluronan in Eye Drops Matters.
Diagnostics (Basel). Jul 23 2020;10(8)doi:10.3390/diagnostics10080511
508. Lemp MA, Crews LA, Bron AJ, Foulks GN, Sullivan BD. Distribution of aqueous-
deficient and evaporative dry eye in a clinic-based patient cohort: a
retrospective study. Cornea. May 2012;31(5):472-8.
doi:10.1097/ICO.0b013e318225415a
175
509. Liu W, Lin T, Gong L. Analysis of Cytokine Levels in Meibum and Clinical
Correlations with Meibomian Gland Dysfunction. Dis Markers.
2022;2022:4259067. doi:10.1155/2022/4259067
510. Duong HT, Phan MAT, Madigan MC, et al. Culture of primary human
meibomian gland cells from surgically excised eyelid tissue. Exp Eye Res. Oct
2023;235:109636. doi:10.1016/j.exer.2023.109636
511. Peng X, Du YL, Liu ST, et al. Isolation and Culture of Human Meibomian Gland
Ductal Cells. Invest Ophthalmol Vis Sci. Dec 1 2023;64(15):29.
doi:10.1167/iovs.64.15.29
512. Nuwormegbe S, Park NY, Park HJ, Jin Y, Kim SW, Jester JV. Induction of
meibocyte differentiation by three-dimensional, matrigel culture of immortalized
human meibomian gland epithelial cells to form acinar organoids. Ocul Surf. Oct
2022;26:271-282. doi:10.1016/j.jtos.2022.10.004
513. Asano N, Hampel U, Garreis F, et al. Differentiation Patterns of Immortalized
Human Meibomian Gland Epithelial Cells in Three-Dimensional Culture. Invest
Ophthalmol Vis Sci. Mar 1 2018;59(3):1343-1353. doi:10.1167/iovs.17-23266
514. Xu KK, Huang YK, Liu X, Zhang MC, Xie HT. Organotypic Culture of Mouse
Meibomian Gland: A Novel Model to Study Meibomian Gland Dysfunction In
Vitro. Invest Ophthalmol Vis Sci. Apr 9 2020;61(4):30. doi:10.1167/iovs.61.4.30
515. Wang G, Xu L, Shi R, et al. Organotypic culture model of mouse meibomian
gland as a screening platform for risk factors related to meibomian gland
dysfunction. Ocul Surf. Oct 2023;30:73-84. doi:10.1016/j.jtos.2023.08.007
516. Zahn I, Garreis F, Schicht M, et al. A New Organotypic 3D Slice Culture of
Mouse Meibomian Glands Reveals Impact of Melanocortins. Int J Mol Sci. Nov
29 2022;23(23)doi:10.3390/ijms232314947
517. Liu R, Tu M, Xue J, Xiao B, Li J, Liang L. Oleic acid induces lipogenesis and
NLRP3 inflammasome activation in organotypic mouse meibomian gland and
human meibomian gland epithelial cells. Exp Eye Res. Apr 2024;241:109851.
doi:10.1016/j.exer.2024.109851
518. Kim SW, Xie Y, Nguyen PQ, et al. PPARγ regulates meibocyte differentiation
and lipid synthesis of cultured human meibomian gland epithelial cells
(hMGEC). Ocul Surf. Oct 2018;16(4):463-469. doi:10.1016/j.jtos.2018.07.004
519. Kim SW, Rho CR, Kim J, et al. Eicosapentaenoic acid (EPA) activates PPARγ
signaling leading to cell cycle exit, lipid accumulation, and autophagy in human
meibomian gland epithelial cells (hMGEC). Ocul Surf. Jul 2020;18(3):427-437.
doi:10.1016/j.jtos.2020.04.012
520. Phan MAT, Madigan MC, Willcox M, Stapleton F, Golebiowski B. Semi-
quantification of lipids in human meibomian gland epithelial cells using dual
staining microplate assays. Exp Eye Res. Sep 2021;210:108719.
doi:10.1016/j.exer.2021.108719
521. Ziemanski JF, Wilson L, Barnes S, Nichols KK. Saturation of cholesteryl esters
produced by human meibomian gland epithelial cells after treatment with
rosiglitazone. Ocul Surf. Apr 2021;20:39-47. doi:10.1016/j.jtos.2020.11.011
522. Ziemanski JF, Wilson L, Barnes S, Nichols KK. Triacylglycerol lipidome from
human meibomian gland epithelial cells: Description, response to culture
176
conditions, and perspective on function. Exp Eye Res. Jun 2021;207:108573.
doi:10.1016/j.exer.2021.108573
523. Liu Y, Chen D, Chen X, Kam WR, Hatton MP, Sullivan DA. Hypoxia: A breath of
fresh air for the meibomian gland. Ocul Surf. Apr 2019;17(2):310-317.
doi:10.1016/j.jtos.2018.12.001
524. Kim H, Cho KJ, Durkin AJ, Tromberg BJ, Park I. Quantitative measurement of
optical properties and Hb concentration in a rodent model of inflammatory
Meibomian gland dysfunction using spatial frequency domain imaging. Biomed
Opt Express. Mar 1 2022;13(3):1261-1274. doi:10.1364/boe.449150
525. Liu Y, Wang J, Chen D, Kam WR, Sullivan DA. The Role of Hypoxia-Inducible
Factor 1alpha in the Regulation of Human Meibomian Gland Epithelial Cells.
Invest Ophthalmol Vis Sci. Mar 9 2020;61(3):1. doi:10.1167/iovs.61.3.1
526. Miyake H, Oda T, Katsuta O, Seno M, Nakamura M. A Novel Model of
Meibomian Gland Dysfunction Induced with Complete Freund's Adjuvant in
Rabbits. Vision (Basel). Feb 9 2017;1(1)doi:10.3390/vision1010010
527. Lu Y, Yin Y, Gong L. Meibomian gland dysfunction model induced with
complete Freund's adjuvant in C57BL/6 mice. Int J Ophthalmol.
2020;13(11):1705-1712. doi:10.18240/ijo.2020.11.04
528. Dong ZY, Ying M, Zheng J, Hu LJ, Xie JY, Ma Y. Evaluation of a rat meibomian
gland dysfunction model induced by closure of meibomian gland orifices. Int J
Ophthalmol. 2018;11(7):1077-1083. doi:10.18240/ijo.2018.07.01
529. Jin K, Kawashima M, Ito M, Arita R, Sano K, Tsubota K. A New Modified
Experimental Meibomian Gland Injury Model: Partial Loss of Gland Due to
Orifice Cauterization and the Alleviating Potential of 22-Oxacalcitriol. J Clin
Med. Dec 22 2020;10(1)doi:10.3390/jcm10010006
530. Bu J, Wu Y, Li K, et al. Transitory alkali exposure on meibomian gland orifices
induces meibomian gland dysfunction. Ocul Surf. Jul 2023;29:406-415.
doi:10.1016/j.jtos.2023.06.007
531. Butovich IA, Wilkerson A, Yuksel S. Depletion of Cholesteryl Esters Causes
Meibomian Gland Dysfunction-Like Symptoms in a Soat1-Null Mouse Model. Int
J Mol Sci. Feb 4 2021;22(4)doi:10.3390/ijms22041583
532. Wilkerson A, Yuksel S, Acharya R, Butovich IA. Physiological Effects of Soat1
Inactivation on Homeostasis of the Mouse Ocular Surface. Invest Ophthalmol
Vis Sci. Jul 1 2024;65(8):2. doi:10.1167/iovs.65.8.2
533. Bu J, Wu Y, Cai X, et al. Hyperlipidemia induces meibomian gland dysfunction.
Ocul Surf. Oct 2019;17(4):777-786. doi:10.1016/j.jtos.2019.06.002
534. Watanabe K, Yoshida M, Okumura T, Sassa T, Kihara A, Uchiyama A.
Improvement of Evaporative Dry Eye With Meibomian Gland Dysfunction in
Model Mice by Treatment With Ophthalmic Solution Containing Mineral Oil.
Transl Vis Sci Technol. Apr 1 2021;10(4):21. doi:10.1167/tvst.10.4.21
535. Reneker LW, Wang L, Irlmeier RT, Huang AJW. Fibroblast Growth Factor
Receptor 2 (FGFR2) Is Required for Meibomian Gland Homeostasis in the Adult
Mouse. Invest Ophthalmol Vis Sci. May 1 2017;58(5):2638-2646.
doi:10.1167/iovs.16-21204
177
536. Yang X, Zhong X, Huang AJ, Reneker LW. Spontaneous acinar and ductal
regrowth after meibomian gland atrophy induced by deletion of FGFR2 in a
mouse model. Ocul Surf. Oct 2022;26:300-309. doi:10.1016/j.jtos.2021.11.005
537. Chen X, Liu Y, Zheng T, et al. Eyes on the Lid: The Impact of Fibroblast Growth
Factor Receptor Targeting Drug on Human Meibomian Gland. Eye Contact
Lens. Feb 1 2024;50(2):102-105. doi:10.1097/icl.0000000000001058
538. Serrano-Morales JM, Álvarez-Santaliestra N, Sánchez-González MC,
Ballesteros-Sánchez A, Sánchez-González JM. Impact of Dyslipidemia on Tear
Film and Meibomian Gland Dysfunction: A Cross-Sectional Study of the
Interplay between Serum Lipid Profile and Ocular Surface Health. J Ophthalmol.
2024;2024:7345270. doi:10.1155/2024/7345270
539. Osae EA, Bullock T, Chintapalati M, et al. Obese Mice with Dyslipidemia Exhibit
Meibomian Gland Hypertrophy and Alterations in Meibum Composition and
Aqueous Tear Production. Int J Mol Sci. Nov 20
2020;21(22)doi:10.3390/ijms21228772
540. Bu J, Zhang M, Wu Y, et al. High-Fat Diet Induces Inflammation of Meibomian
Gland. Invest Ophthalmol Vis Sci. Aug 2 2021;62(10):13.
doi:10.1167/iovs.62.10.13
541. Zou S, Liu J, Si H, et al. High-fat intake reshapes the circadian transcriptome
profile and metabolism in murine meibomian glands. Front Nutr.
2023;10:1146916. doi:10.3389/fnut.2023.1146916
542. Wu KI, Chen CY, Jou TS, Jimmy Juang JM, Lu JY, Wang IJ. Effect of 3-
Hydroxy-3-Methyl-Glutaryl-Coenzyme A Reductase Inhibitors on the Meibomian
Gland Morphology in Patients with Dyslipidemia. Am J Ophthalmol. Nov
2020;219:240-252. doi:10.1016/j.ajo.2020.06.029
543. Butovich IA, Wilkerson A, Yuksel S. Differential effects of dietary cholesterol
and triglycerides on the lipid homeostasis in Meibomian glands. J Steroid
Biochem Mol Biol. Jul 2021;211:105894. doi:10.1016/j.jsbmb.2021.105894
544. Guliani BP, Bhalla A, Naik MP. Association of the severity of meibomian gland
dysfunction with dyslipidemia in Indian population. Indian J Ophthalmol. Oct
2018;66(10):1411-1416. doi:10.4103/ijo.IJO_1256_17
545. Butovich IA, Suzuki T. Delineating a novel metabolic high triglycerides-low
waxes syndrome that affects lipid homeostasis in meibomian and sebaceous
glands. Exp Eye Res. Oct 2020;199:108189. doi:10.1016/j.exer.2020.108189
546. Lin X, Wu Y, Chen Y, et al. Characterization of Meibomian Gland Atrophy and
the Potential Risk Factors for Middle Aged to Elderly Patients With Cataracts.
Transl Vis Sci Technol. Jun 2020;9(7):48. doi:10.1167/tvst.9.7.48
547. Al-Namaeh M. A systematic review of the effect of omega-3 supplements on
meibomian gland dysfunction. Ther Adv Ophthalmol. Jan-Dec
2020;12:2515841420952188. doi:10.1177/2515841420952188
548. Jeyalatha MV, Qu Y, Liu Z, et al. Function of meibomian gland: Contribution of
proteins. Exp Eye Res. Oct 2017;163:29-36. doi:10.1016/j.exer.2017.06.009
549. Li S, Zhou J, Bu J, et al. Ectodysplasin A protein promotes corneal epithelial cell
proliferation. J Biol Chem. Aug 11 2017;292(32):13391-13401.
doi:10.1074/jbc.M117.803809
178
550. Alam J, Yaman E, Silva GCV, et al. Single cell analysis of short-term dry eye
induced changes in cornea immune cell populations. Front Med (Lausanne).
2024;11:1362336. doi:10.3389/fmed.2024.1362336
551. Alam J, de Paiva CS, Pflugfelder SC. Desiccation Induced Conjunctival
Monocyte Recruitment and Activation - Implications for Keratoconjunctivitis.
Front Immunol. 2021;12:701415. doi:10.3389/fimmu.2021.701415
552. Liu Z, Xie H, Li L, et al. Single-cell landscape reveals the epithelial cell-centric
pro-inflammatory immune microenvironment in dry eye development. Mucosal
Immunol. Jun 2024;17(3):491-507. doi:10.1016/j.mucimm.2023.11.008
553. Nair AP, D'Souza S, Khamar P, Nuijts R, Sethu S, Shetty R. Ocular surface
immune cell diversity in dry eye disease. Indian J Ophthalmol. Apr
2023;71(4):1237-1247. doi:10.4103/IJO.IJO_2986_22
554. Li L, Li Y, Zhu X, et al. Conjunctiva Resident gammadelta T Cells Expressed
High Level of IL-17A and Promoted the Severity of Dry Eye. Invest Ophthalmol
Vis Sci. Nov 1 2022;63(12):13. doi:10.1167/iovs.63.12.13
555. Ji YW, Lee JL, Kang HG, et al. Corneal lymphangiogenesis facilitates ocular
surface inflammation and cell trafficking in dry eye disease. Ocul Surf. Jul
2018;16(3):306-313. doi:10.1016/j.jtos.2018.03.008
556. Ortiz G, Blanco T, Singh RB, et al. IL-6 induces Treg dysfunction in desiccating
stress-induced dry eye disease. Exp Eye Res. Jul 13 2024:110006.
doi:10.1016/j.exer.2024.110006
557. Zhao D, Ji H, Zhao H, Xu Y, He A, He Y. BMSC-derived exosomes regulate the
Treg/Th17 balance through the miR-21-5p/TLR4/MyD88/NF-kappaB pathway to
alleviate dry eye symptoms in mice. In Vitro Cell Dev Biol Anim. Apr 29
2024;doi:10.1007/s11626-024-00910-6
558. Schaefer L, Trujillo-Vargas CM, Midani FS, Pflugfelder SC, Britton RA, de Paiva
CS. Gut Microbiota From Sjogren syndrome Patients Causes Decreased T
Regulatory Cells in the Lymphoid Organs and Desiccation-Induced Corneal
Barrier Disruption in Mice. Front Med (Lausanne). 2022;9:852918.
doi:10.3389/fmed.2022.852918
559. Coursey TG, Bian F, Zaheer M, Pflugfelder SC, Volpe EA, de Paiva CS. Age-
related spontaneous lacrimal keratoconjunctivitis is accompanied by
dysfunctional T regulatory cells. Mucosal Immunol. May 2017;10(3):743-756.
doi:10.1038/mi.2016.83
560. Bedggood P, Wu M, Zhang X, et al. Improved tracking of corneal immune cell
dynamics using in vivo confocal microscopy. Biomedical optics express. Nov 1
2024;15(11):6277-6298. doi:10.1364/BOE.536553
561. Willcox MDP, Argueso P, Georgiev GA, et al. TFOS DEWS II Tear Film Report.
Ocul Surf. Jul 2017;15(3):366-403. doi:10.1016/j.jtos.2017.03.006
562. Bandlitz S, Peter B, Pflugi T, et al. Agreement and repeatability of four different
devices to measure non-invasive tear breakup time (NIBUT). Cont Lens
Anterior Eye. Oct 2020;43(5):507-511. doi:10.1016/j.clae.2020.02.018
563. Lim J, Wang MTM, Craig JP. Evaluating the diagnostic ability of two automated
non-invasive tear film stability measurement techniques. Cont Lens Anterior
Eye. Aug 2021;44(4):101362. doi:10.1016/j.clae.2020.08.006
179
564. Singh S, Srivastav S, Modiwala Z, Ali MH, Basu S. Repeatability, reproducibility
and agreement between three different diagnostic imaging platforms for tear
film evaluation of normal and dry eye disease. Eye (Lond). Jul
2023;37(10):2042-2047. doi:10.1038/s41433-022-02281-2
565. Wang MT, Murphy PJ, Blades KJ, Craig JP. Comparison of non-invasive tear
film stability measurement techniques. Clin Exp Optom. Jan 2018;101(1):13-17.
doi:10.1111/cxo.12546
566. Wang MTM, Craig JP. Comparative evaluation of clinical methods of tear film
stability assessment: A randomized crossover trial. JAMA Ophthalmol. Mar 1
2018;136(3):291-294. doi:10.1001/jamaophthalmol.2017.6489
567. Szczesna-Iskander DH, Llorens-Quintana C. Agreement between invasive and
noninvasive measurement of tear film breakup time. Sci Rep. Feb 15
2024;14(1):3852. doi:10.1038/s41598-024-54219-1
568. Abusharha A, Yami AA, Alsreea K, et al. Repeatability and reproducibility of tear
film evaporation rate measurement using a new closed-chamber evaporimeter
Open J Ophthalmol. 2021;15:117-121.
569. Fagehi R, El-Hiti GA, Alsubaie MH, et al. Measurements of tear evaporation
rate in subjects with refractive errors using a portable evaporimeter. Healthcare
(Basel). Feb 21 2022;10(2)doi:10.3390/healthcare10020405
570. Alanazi MA, Alanazi SA, Baashen MA, Almutairi MS, El-Hiti GA. Assessment of
tear film parameters in females with refractive errors using a single device: An
observational study. J Pak Med Assoc. Mar 2024;74(3):599-562.
doi:10.47391/JPMA.9994
571. Ji YW, Lee J, Lee H, Seo KY, Kim EK, Kim TI. Automated measurement of tear
film dynamics and lipid layer thickness for assessment of non-Sjogren dry eye
syndrome with meibomian gland dysfunction. Cornea. Feb 2017;36(2):176-182.
doi:10.1097/ICO.0000000000001101
572. Lee Y, Hyon JY, Jeon HS. Characteristics of dry eye patients with thick tear film
lipid layers evaluated by a LipiView II interferometer. Graefes Arch Clin Exp
Ophthalmol. May 2021;259(5):1235-1241. doi:10.1007/s00417-020-05044-5
573. Qi Y, Zhang C, Zhao S, Huang Y, Yang R. A novel noninvasive ocular surface
analyzer for the assessment of dry eye with meibomian gland dysfunction. Exp
Ther Med. Jun 2017;13(6):2983-2988. doi:10.3892/etm.2017.4364
574. Shen J, Xu Z, Wang Y, et al. Influential signs of dry eye-related ocular
symptoms in participants with unstable tear film. Cornea. Jun 1 2024;43(6):751-
756. doi:10.1097/ICO.0000000000003473
575. Alanazi SA, Aldawood MA, Badawood YS, El-Hiti GA, Masmali AM. A
comparative study of the quality of non-stimulated and stimulated tears in
normal eye male subjects using the tear ferning test. Clin Optom (Auckl).
2019;11:65-71. doi:10.2147/OPTO.S214597
576. Szczesna-Iskander DH, Muzyka-Wozniak M, Llorens Quintana C. The efficacy
of ocular surface assessment approaches in evaluating dry eye treatment with
artificial tears. Sci Rep. Dec 17 2022;12(1):21835. doi:10.1038/s41598-022-
26327-3
180
577. Bai Y, Ngo W, Khanal S, Nichols KK, Nichols JJ. Human precorneal tear film
and lipid layer dynamics in meibomian gland dysfunction. Ocul Surf. Jul
2021;21:250-256. doi:10.1016/j.jtos.2021.03.006
578. Li J, Ma J, Hu M, Yu J, Zhao Y. Assessment of tear film lipid layer thickness in
patients with meibomian gland dysfunction at different ages. BMC Ophthalmol.
Oct 6 2020;20(1):394. doi:10.1186/s12886-020-01667-8
579. Yi HC, Lee YP, Shin YJ. Influence of nasal tear osmolarity on ocular symptoms
related to dry eye disease. Am J Ophthalmol. May 2018;189:71-76.
doi:10.1016/j.ajo.2018.02.008
580. Yazdani M, Fiskadal J, Chen X, et al. Tear film break-up time and dry eye
disease severity in a large Norwegian cohort. J Clin Med. Feb 22
2021;10(4)doi:10.3390/jcm10040884
581. Tashbayev B, Utheim TP, Utheim OA, et al. Utility of tear osmolarity
measurement in diagnosis of dry eye disease. Sci Rep. Mar 26
2020;10(1):5542. doi:10.1038/s41598-020-62583-x
582. Shimazaki J, Sakata M, Den S, Iwasaki M, Toda I. Tear film osmolarity
measurement in Japanese dry eye patients using a handheld osmolarity
system. Diagnostics (Basel). Oct 5
2020;10(10)doi:10.3390/diagnostics10100789
583. Nilsen C, Graae Jensen P, Gundersen M, et al. The significance of inter-eye
osmolarity difference in dry eye diagnostics. Clin Ophthalmol. 2023;17:829-835.
doi:10.2147/OPTH.S402556
584. Mathews PM, Karakus S, Agrawal D, Hindman HB, Ramulu PY, Akpek EK.
Tear osmolarity and correlation with ocular surface parameters in patients with
dry eye. Cornea. Nov 2017;36(11):1352-1357.
doi:10.1097/ICO.0000000000001364
585. Kim M, Kim HS, Na K-S. Correlation between tear osmolarity and other ocular
surface parameters in primary Sjögren’s syndrome. Korean J Ophthalmol.
2017;31:25-31.
586. Greiner JV, Ying GS, Pistilli M, et al. Association of Tear Osmolarity With Signs
and Symptoms of Dry Eye Disease in the Dry Eye Assessment and
Management (DREAM) Study. Invest Ophthalmol Vis Sci. Jan 3 2023;64(1):5.
doi:10.1167/iovs.64.1.5
587. Titiyal JS, Goswami A, Kaur M, et al. Impact of Topical Cyclosporine-A or
Topical Chloroquine on Post-LASIK Ocular Surface Stability - A Randomized
Controlled Trial. Curr Eye Res. Jun 2023;48(6):557-563.
doi:10.1080/02713683.2023.2182747
588. Rajpoot M, Singh D, Pandey K, Bhargava R. Safety and efficacy of cyclosporine
(0.05% versus 0.09%) in dry eye disease. Is it the strength of cyclosporin that
really matters? Nepalese Journal of Ophthalmology. 12/31 2022;14(2):64-77.
doi:10.3126/nepjoph.v14i2.38928
589. Konstas AG, Boboridis KG, Athanasopoulos GP, et al. Changing from
preserved, to preservative-free cyclosporine 0.1% enhanced triple glaucoma
therapy: impact on ocular surface disease-a randomized controlled trial. Eye
(Lond). Dec 2023;37(17):3666-3674. doi:10.1038/s41433-023-02578-w
181
590. Benitez-Del-Castillo JM, López-Pérez MD, Cano-Ortiz A, et al. Efficacy and
safety of intense pulsed light of upper and lower eyelids in Meibomian gland
dysfunction: A prospective multicentric study. Eur J Ophthalmol. May
2024;34(3):700-707. doi:10.1177/11206721231199121
591. Nieto-Bona A, Nombela-Palomo M, Felipe-Márquez G, Teus MA. Tear Film
Osmolarity in Response to Long-Term Orthokeratology Treatment. Eye Contact
Lens. Mar 2018;44(2):85-90. doi:10.1097/icl.0000000000000347
592. Yuksel N, Akcay E, Ayan B, Duru N. Tear-Film Osmolarity Changes Following
Dacryocystorhinostomy in Primary Acquired Nasolacrimal Duct Obstruction.
Curr Eye Res. Mar 2017;42(3):348-350. doi:10.1080/02713683.2016.1196706
593. Blanco-Campoy DG, Graue-Hernandez EO, Quiroz-Casian N, Velez-Cordero
JR, Yanez-Soto B. In-vitro evaluation of the evaporation retardation by
meibomian lipids in homogeneous and non-homogeneous evaporation. J
Colloid Interface Sci. Nov 2022;625:210-219. doi:10.1016/j.jcis.2022.06.032
594. Dursch TJ, Li W, Taraz B, Lin MC, Radke CJ. Tear-film evaporation rate from
simultaneous ocular-surface temperature and tear-breakup area. Optom Vis
Sci. Jan 2018;95(1):5-12. doi:10.1097/OPX.0000000000001156
595. Svitova TF, Lin MC. Evaporation retardation by model tear-lipid films: The roles
of film aging, compositions and interfacial rheological properties. Colloids Surf B
Biointerfaces. Jan 2021;197:111392. doi:10.1016/j.colsurfb.2020.111392
596. Xu X, Li G, Zuo YY. Effect of model tear film lipid layer on water evaporation.
Invest Ophthalmol Vis Sci. Jan 3 2023;64(1):13. doi:10.1167/iovs.64.1.13
597. Kim YH, Graham AD, Li W, et al. Tear-film evaporation flux and its relationship
to tear properties in symptomatic and asymptomatic soft-contact-lens wearers.
Cont Lens Anterior Eye. Aug 2023;46(4):101850.
doi:10.1016/j.clae.2023.101850
598. Lin MC, Svitova TF. Effects of model tear proteins and topical ophthalmic
formulations on evaporation inhibition and biophysical property of model tear
lipid nanofilm in vitro. JCIS Open. 2021;4:100028.
doi:10.1016/j.jciso.2021.100028
599. Lee D, Song S, Cho G, et al. Elucidating the molecular interactions between
lipids and lysozyme: Evaporation resistance and bacterial barriers for dry eye
disease. Nano Lett. Oct 25 2023;23(20):9451-9460.
doi:10.1021/acs.nanolett.3c02936
600. Borchman D, Ramakrishnan V, Henry C, Ramasubramanian A. Differences in
meibum and tear lipid composition and conformation. Cornea. Jan
2020;39(1):122-128. doi:10.1097/ICO.0000000000002095
601. Nencheva Y, Ramasubramanian A, Eftimov P, Yokoi N, Borchman D, Georgiev
GA. Effects of lipid saturation on the surface properties of human meibum films.
Int J Mol Sci. Jul 28 2018;19(8)doi:10.3390/ijms19082209
602. Georgiev GA, Borchman D, Eftimov P, Yokoi N. Lipid saturation and the
rheology of human tear lipids. Int J Mol Sci. Jul 12
2019;20(14)doi:10.3390/ijms20143431
603. Eftimov P, Yokoi N, Tsuji K, et al. Surface chemistry study of normal and
diseased human meibum films prior to and after supplementation with tear
182
mimetic eyedrop formulation. Appl Sci. 2024;14(8):3339.
doi:https://doi.org/10.3390/app14083339
604. Borchman D, Ramasubramanian A, Foulks GN. Human meibum cholesteryl and
wax ester variability with age, sex, and meibomian gland dysfunction. Invest
Ophthalmol Vis Sci. May 1 2019;60(6):2286-2293. doi:10.1167/iovs.19-26812
605. Hetman ZA, Borchman D. Concentration dependent cholesteryl-ester and wax-
ester structural relationships and meibomian gland dysfunction. Biochem
Biophys Rep. Mar 2020;21:100732. doi:10.1016/j.bbrep.2020.100732
606. Nagar S, Ajouz L, Nichols KK, et al. Relationship between human meibum lipid
composition and the severity of meibomian gland dysfunction: A spectroscopic
analysis. Invest Ophthalmol Vis Sci. Jul 3 2023;64(10):22.
doi:10.1167/iovs.64.10.22
607. Ewurum A, Ankem A, Georgiev G, Borchman D. A spectroscopic study of the
composition and conformation of cholesteryl and wax esters purified from
meibum. Chem Phys Lipids. Aug 2021;238:105088.
doi:10.1016/j.chemphyslip.2021.105088
608. Eftimov P, Ewurum A, Ankem A, Borchman D, Georgiev GA. Correlations
between bulk and surface properties of meibomian lipids with alteration of wax-
to-sterol esters content. Chem Phys Lipids. Jan 2022;242:105163.
doi:10.1016/j.chemphyslip.2021.105163
609. Garcia-Queiruga J, Pena-Verdeal H, Sabucedo-Villamarin B, et al. Meibum
lipidomic analysis in evaporative dry eye subjects. Int J Mol Sci. Apr 27
2024;25(9)doi:10.3390/ijms25094782
610. Rantamaki AH, Holopainen JM. The effect of phospholipids on tear film lipid
layer surface activity. Invest Ophthalmol Vis Sci. Jan 1 2017;58(1):149-154.
doi:10.1167/iovs.16-20468
611. Paananen RO, Javanainen M, Holopainen JM, Vattulainen I. Crystalline wax
esters regulate the evaporation resistance of tear film lipid layers associated
with dry eye syndrome. J Phys Chem Lett. Jul 18 2019;10(14):3893-3898.
doi:10.1021/acs.jpclett.9b01187
612. Viitaja T, Raitanen JE, Hynynen A, et al. On the importance of chain branching
in tear film lipid layer wax and cholesteryl esters. Colloids Surf B Biointerfaces.
Jun 2022;214:112429. doi:10.1016/j.colsurfb.2022.112429
613. Rubio RG, Guzmán F, Ortega F, Liggieri L. Monolayers of cholesterol and
cholesteryl stearate at the water/vapor interface: A physico-chemical study of
components of the meibum layer. Colloids Interfaces. 2021;5(2):50.
doi:https://doi.org/10.3390/colloids5020030
614. Jun I, Kim S, Kim H, et al. Evaluation of meibum lipid composition according to
tear interferometric patterns: RRH: meibum composition according to
interferometric patterns. Am J Ophthalmol. Aug 2022;240:37-50.
doi:10.1016/j.ajo.2022.02.010
615. Viitaja T, Raitanen JE, Moilanen J, Paananen RO, Ekholm FS. Biophysical
profiling of synthetic ultra-long tear film lipids. Colloids Surf B Biointerfaces. Mar
2023;223:113145. doi:10.1016/j.colsurfb.2023.113145
616. Bland HC, Moilanen JA, Ekholm FS, Paananen RO. Investigating the role of
specific tear film lipids connected to dry eye syndrome: A study on O-acyl-
183
omega-hydroxy fatty acids and diesters. Langmuir. Mar 5 2019;35(9):3545-
3552. doi:10.1021/acs.langmuir.8b04182
617. Viitaja T, Moilanen J, Svedstrom KJ, Ekholm FS, Paananen RO. Tear film lipid
layer structure: Self-assembly of O-Acyl-omega-hydroxy fatty acids and wax
esters into evaporation-resistant monolayers. Nano Lett. Sep 22
2021;21(18):7676-7683. doi:10.1021/acs.nanolett.1c02475
618. Paananen RO, Viitaja T, Olzynska A, Ekholm FS, Moilanen J, Cwiklik L.
Interactions of polar lipids with cholesteryl ester multilayers elucidate tear film
lipid layer structure. Ocul Surf. Oct 2020;18(4):545-553.
doi:10.1016/j.jtos.2020.06.001
619. Viitaja T, Raitanen JE, Moilanen J, Paananen RO, Ekholm FS. The properties
and role of O-acyl-omega-hydroxy fatty acids and Ttype I-St and Ttype II
diesters in the tear film lipid layer revealed by a combined chemistry and
biophysics approach. J Org Chem. Apr 2 2021;86(7):4965-4976.
doi:10.1021/acs.joc.0c02882
620. Khanal S, Ngo W, Nichols KK, Wilson L, Barnes S, Nichols JJ. Human meibum
and tear film derived (O-acyl)-omega-hydroxy fatty acids in meibomian gland
dysfunction. Ocul Surf. Jul 2021;21:118-128. doi:10.1016/j.jtos.2021.05.009
621. Khanal S, Bai Y, Ngo W, et al. Human meibum and tear film derived (O-acyl)-
omega-hydroxy fatty acids as biomarkers of tear film dynamics in meibomian
gland dysfunction and dry eye disease. Invest Ophthalmol Vis Sci. Jul 1
2021;62(9):13. doi:10.1167/iovs.62.9.13
622. Galor A, Sanchez V, Jensen A, et al. Meibum sphingolipid composition is
altered in individuals with meibomian gland dysfunction-a side by side
comparison of meibum and tear sphingolipids. Ocul Surf. Jan 2022;23:87-95.
doi:10.1016/j.jtos.2021.11.011
623. Paranjpe V, Tan J, Nguyen J, et al. Clinical signs of meibomian gland
dysfunction (MGD) are associated with changes in meibum sphingolipid
composition. Ocul Surf. Apr 2019;17(2):318-326. doi:10.1016/j.jtos.2018.12.006
624. Olzynska A, Cwiklik L. Behavior of sphingomyelin and ceramide in a tear film
lipid layer model. Ann Anat. Mar 2017;210:128-134.
doi:10.1016/j.aanat.2016.10.005
625. Wizert A, Iskander DR, Cwiklik L. Interaction of lysozyme with a tear film lipid
layer model: A molecular dynamics simulation study. Biochim Biophys Acta
Biomembr. Dec 2017;1859(12):2289-2296. doi:10.1016/j.bbamem.2017.08.015
626. Miyamoto M, Sassa T, Sawai M, Kihara A. Lipid polarity gradient formed by
omega-hydroxy lipids in tear film prevents dry eye disease. eLife. Apr 7
2020;9doi:10.7554/eLife.53582
627. Otsuka K, Sawai-Ogawa M, Kihara A. Formation of fatty alcohols-components
of meibum lipids-by the fatty acyl-CoA reductase FAR2 is essential for dry eye
prevention. FASEB J. Apr 2022;36(4):e22216. doi:10.1096/fj.202101733R
628. Sassa T, Tadaki M, Kiyonari H, Kihara A. Very long-chain tear film lipids
produced by fatty acid elongase ELOVL1 prevent dry eye disease in mice.
FASEB J. Jun 2018;32(6):2966-2978. doi:10.1096/fj.201700947R
184
629. Wilkerson A, Bhat N, Quoc Hai Pham H, Yuksel S, Butovich I. Physiological
effects of inactivation and the roles of Elovl3/ELOVL3 in maintaining ocular
homeostasis. FASEB J. Feb 2021;35(2):e21327. doi:10.1096/fj.202002323R
630. Sawai M, Watanabe K, Tanaka K, et al. Diverse meibum lipids produced by
Awat1 and Awat2 are important for stabilizing tear film and protecting the ocular
surface. iScience. May 21 2021;24(5):102478. doi:10.1016/j.isci.2021.102478
631. Xu X, Wilkerson A, Li G, Butovich IA, Zuo YY. Comparative biophysical study of
meibomian lipids of wild type and Soat1-null mice: Implications to meibomian
gland dysfunction and dry eye disease. Invest Ophthalmol Vis Sci. Aug 1
2023;64(11):20. doi:10.1167/iovs.64.11.20
632. Inaba T, Tanaka Y, Tamaki S, Ito T, Ntambi JM, Tsubota K. Compensatory
increases in tear volume and mucin levels associated with meibomian gland
dysfunction caused by stearoyl-CoA desaturase-1 deficiency. Sci Rep. Feb 20
2018;8(1):3358. doi:10.1038/s41598-018-21542-3
633. Ji C, Guo Y, Liu Y, et al. Inhibition of ceramide de novo synthesis ameliorates
meibomian gland dysfunction induced by SCD1 deficiency. Ocul Surf. Oct
2021;22:230-241. doi:10.1016/j.jtos.2021.08.016
634. George CT, Kurien BT, Scofield RH. The potential utility of salivary and tear
proteomics to discriminate Sjogren's disease from non-Sjogren's sicca. Int J Mol
Sci. Dec 15 2023;24(24)doi:10.3390/ijms242417497
635. Hsiao YT, Huang YT, Yu HJ, Fang PC, Kuo MT. Tear proteomics approach to
distinguishing primary from secondary Sjogren's syndrome for dry eye patients
with long-term instillation of eyedrops. Int J Mol Sci. Dec 3
2022;23(23)doi:10.3390/ijms232315239
636. Das N, Menon NG, de Almeida LGN, et al. Proteomics analysis of tears and
saliva From Sjogren's syndrome patients. Front Pharmacol. 2021;12:787193.
doi:10.3389/fphar.2021.787193
637. Akpek EK, Wu HY, Karakus S, Zhang Q, Masli S. Differential Diagnosis of
Sjogren Versus Non-Sjogren Dry Eye Through Tear Film Biomarkers. Cornea.
Aug 2020;39(8):991-997. doi:10.1097/ICO.0000000000002299
638. Kuo MT, Fang PC, Chao TL, et al. Tear Proteomics Approach to Monitoring
Sjogren Syndrome or Dry Eye Disease. Int J Mol Sci. Apr 19
2019;20(8)doi:10.3390/ijms20081932
639. Aqrawi LA, Galtung HK, Guerreiro EM, et al. Proteomic and histopathological
characterisation of sicca subjects and primary Sjogren's syndrome patients
reveals promising tear, saliva and extracellular vesicle disease biomarkers.
Arthritis Res Ther. Jul 31 2019;21(1):181. doi:10.1186/s13075-019-1961-4
640. Lepine M, Robert MC, Sleno L. Discovery and Verification of Sjogren's
Syndrome Protein Biomarkers in Tears by Targeted LC-MRM. J Proteome Res.
Jun 7 2024;23(6):2219-2229. doi:10.1021/acs.jproteome.4c00163
641. Bu J, Liu Y, Zhang R, et al. Potential New Target for Dry Eye Disease-Oxidative
Stress. Antioxidants (Basel). Mar 29 2024;13(4)doi:10.3390/antiox13040422
642. Legatowicz-Koprowska M, Nitek S, Czerwińska J. The complement system in
primary Sjögren's syndrome: the expression of certain cascade and regulatory
proteins in labial salivary glands - observational study. Reumatologia.
2020;58(6):357-366. doi:10.5114/reum.2020.102000
185
643. Aljohani S, Jazzar A. Tear Cytokine Levels in Sicca Syndrome-Related Dry Eye:
A Meta-Analysis. Diagnostics (Basel). Jun 27
2023;13(13)doi:10.3390/diagnostics13132184
644. Shinzawa M, Dogru M, Den S, et al. Epidermal Fatty Acid-Binding Protein: A
Novel Marker in the Diagnosis of Dry Eye Disease in Sjogren Syndrome. Int J
Mol Sci. Nov 4 2018;19(11)doi:10.3390/ijms19113463
645. Gijs M, van de Sande N, Bonnet C, et al. A comprehensive scoping review of
methodological approaches and clinical applications of tear fluid biomarkers.
Prog Retin Eye Res. Feb 13 2025;106:101338.
doi:10.1016/j.preteyeres.2025.101338
646. Liu C, Lin MT, Lee IXY, et al. Neuropathic corneal pain: Tear proteomic and
neuromediator profiles, imaging features, and clinical manifestations. Am J
Ophthalmol. Mar 21 2024;265:6-20. doi:10.1016/j.ajo.2024.03.015
647. Uchino Y, Uchino M, Mizuno M, Shigeno Y, Furihata K, Shimazaki J.
Morphological alterations in corneal nerves of patients with dry eye and
associated biomarkers. Exp Eye Res. May 2023;230:109438.
doi:10.1016/j.exer.2023.109438
648. Zysset-Burri DC, Schlegel I, Lincke JB, et al. Understanding the interactions
between the ocular surface microbiome and the tear proteome. Invest
Ophthalmol Vis Sci. Aug 2 2021;62(10):8. doi:10.1167/iovs.62.10.8
649. Schlegel I, De Gouyon Matignon de Pontourade CMF, Lincke JB, Keller I,
Zinkernagel MS, Zysset-Burri DC. The human ocular surface microbiome and
its associations with the tear proteome in dry eye disease. Int J Mol Sci. Sep 14
2023;24(18)doi:10.3390/ijms241814091
650. Li Y, Sang X, Yang L, et al. Low concentration of sodium hyaluronate
temporarily elevates the tear film lipid layer thickness in dry eye patients with
lipid deficiency. Int J Ophthalmol. 2018;11(3):389-394.
doi:10.18240/ijo.2018.03.07
651. Fukuoka S, Arita R. Increase in tear film lipid layer thickness after instillation of
3% diquafosol ophthalmic solution in healthy human eyes. Ocul Surf. Oct
2017;15(4):730-735. doi:10.1016/j.jtos.2017.03.005
652. Eftimov P, Yokoi N, Melo AM, Daull P, Georgiev GA. Interactions of meibum
and tears with mucomimetic polymers: A hint towards the interplay between the
layers of the tear film. Int J Mol Sci. Mar 9 2021;22(5)doi:10.3390/ijms22052747
653. Sun YC, Hung KF, Li TY, Chang YA, Yeh PT, Hu FR. Transmembrane mucin 1
blocks fluorescein ingress to corneal epithelium. Invest Ophthalmol Vis Sci. Feb
1 2022;63(2):31. doi:10.1167/iovs.63.2.31
654. Choi M, Tichenor AA. Regional conjunctival differences in glycocalyx mucin
expression in dry eye and normal subjects. Invest Ophthalmol Vis Sci. Feb 1
2024;65(2):20. doi:10.1167/iovs.65.2.20
655. Choudhury A, Dey M, Dixit HN, Feng JJ. Tear-film breakup: The role of
membrane-associated mucin polymers. Phys Rev E. Jan 2021;103(1-
1):013108. doi:10.1103/PhysRevE.103.013108
656. Matsuzawa M, Ando T, Fukase S, et al. The protective role of conjunctival
goblet cell mucin sialylation. Nat Commun. Mar 17 2023;14(1):1417.
doi:10.1038/s41467-023-37101-y
186
657. Woodward AM, Senchyna M, Argueso P. Short-term reproducibility of MUC5AC
measurement in human tear fluid. Diagnostics (Basel). Jan 2
2021;11(1)doi:10.3390/diagnostics11010057
658. Botten N, Hodges RR, Li D, et al. Resolvin D2 elevates cAMP to increase
intracellular [Ca(2+)] and stimulate secretion from conjunctival goblet cells.
FASEB J. Jul 2019;33(7):8468-8478. doi:10.1096/fj.201802467R
659. Kim CE, Kleinman HK, Sosne G, et al. RGN-259 (thymosin beta4) improves
clinically important dry eye efficacies in comparison with prescription drugs in a
dry eye model. Sci Rep. Jul 12 2018;8(1):10500. doi:10.1038/s41598-018-
28861-5
660. Choo J, Liao CH, Tseng CL, et al. Inhibition of microRNA-328 increases ocular
mucin expression and conjunctival goblet cells. Biomedicines. Jan 19
2023;11(2)doi:10.3390/biomedicines11020287
661. ClinicalTrials.gov. Assessment of the Safety and Efficacy of RGN-259
Ophthalmic Solutions for Dry Eye Syndrome : ARISE-2.
662. Altman J, Jones G, Ahmed S, Sharma S, Sharma A. Tear Film MicroRNAs as
Potential Biomarkers: A Review. Int J Mol Sci. Feb 12
2023;24(4)doi:10.3390/ijms24043694
663. Pucker AD, Ngo W, Postnikoff CK, Fortinberry H, Nichols JJ. Tear Film miRNAs
and Their Association With Human Dry Eye Disease. Curr Eye Res. Nov
2022;47(11):1479-1487. doi:10.1080/02713683.2022.2110597
664. Kim YJ, Yeon Y, Lee WJ, et al. Comparison of MicroRNA Expression in Tears
of Normal Subjects and Sjogren Syndrome Patients. Invest Ophthalmol Vis Sci.
Nov 1 2019;60(14):4889-4895. doi:10.1167/iovs.19-27062
665. Kado Abdalkader R, Chaleckis R, Fujita T, Kamei KI. Modeling dry eye with an
air-liquid interface in corneal epithelium-on-a-chip. Sci Rep. Feb 20
2024;14(1):4185. doi:10.1038/s41598-024-54736-z
666. Nomi K, Hayashi R, Ishikawa Y, et al. Generation of functional conjunctival
epithelium, including goblet cells, from human iPSCs. Cell Rep. Feb 2
2021;34(5):108715. doi:10.1016/j.celrep.2021.108715
667. Bannier-Helaouet M, Korving J, Ma Z, et al. Human conjunctiva organoids to
study ocular surface homeostasis and disease. Cell Stem Cell. Feb 1
2024;31(2):227-243 e12. doi:10.1016/j.stem.2023.12.008
668. Madl AC, Liu C, Cirera-Salinas D, Fuller GG, Myung D. A mucin-deficient ocular
surface mimetic platform for interrogating drug effects on biolubrication,
antiadhesion properties, and barrier functionality. ACS Appl Mater Interfaces.
Apr 27 2022;14(16):18016-18030. doi:10.1021/acsami.1c22280
669. Rabiah NI, Sato Y, Kannan A, Kress W, Straube F, Fuller GG. Understanding
the adsorption and potential tear film stability properties of recombinant human
lubricin and bovine submaxillary mucins in an in vitro tear film model. Colloids
Surf B Biointerfaces. Nov 2020;195:111257. doi:10.1016/j.colsurfb.2020.111257
670. Schicht M, Riedlova K, Kukulka M, et al. The potential role of SP-G as surface
tension regulator in tear film: From molecular simulations to experimental
observations. Int J Mol Sci. May 21 2022;23(10)doi:10.3390/ijms23105783
671. Storas AM, Fineide F, Magno M, et al. Using machine learning model
explanations to identify proteins related to severity of meibomian gland
187
dysfunction. Sci Rep. Dec 22 2023;13(1):22946. doi:10.1038/s41598-023-
50342-7
672. Stern ME, Pflugfelder SC. What We Have Learned From Animal Models of Dry
Eye. Int Ophthalmol Clin. Spring 2017;57(2):109-118.
doi:10.1097/IIO.0000000000000169
673. Hisey EA, Galor A, Leonard BC. A comparative review of evaporative dry eye
disease and meibomian gland dysfunction in dogs and humans. Vet
Ophthalmol. Apr 2023;26 Suppl 1(Suppl 1):16-30. doi:10.1111/vop.13066
674. Harris LW, Purves D. Rapid remodeling of sensory endings in the corneas of
living mice. J Neurosci. Jun 1989;9(6):2210-4. doi:10.1523/JNEUROSCI.09-06-
02210.1989
675. Frutos-Rincon L, Gomez-Sanchez JA, Inigo-Portugues A, Acosta MC, Gallar J.
An Experimental Model of Neuro-Immune Interactions in the Eye: Corneal
Sensory Nerves and Resident Dendritic Cells. Int J Mol Sci. Mar 10
2022;23(6)doi:10.3390/ijms23062997
676. Patel DV, Tavakoli M, Craig JP, Efron N, McGhee CN. Corneal sensitivity and
slit scanning in vivo confocal microscopy of the subbasal nerve plexus of the
normal central and peripheral human cornea. Cornea. Aug 2009;28(7):735-40.
doi:10.1097/ICO.0b013e318193e0e3
677. Stepp MA, Tadvalkar G, Hakh R, Pal-Ghosh S. Corneal epithelial cells function
as surrogate Schwann cells for their sensory nerves. Glia. Jun 2017;65(6):851-
863. doi:10.1002/glia.23102
678. Alcalde I, Inigo-Portugues A, Gonzalez-Gonzalez O, et al. Morphological and
functional changes in TRPM8-expressing corneal cold thermoreceptor neurons
during aging and their impact on tearing in mice. J Comp Neurol. Aug 1
2018;526(11):1859-1874. doi:10.1002/cne.24454
679. De Felipe C, Belmonte C. c-Jun expression after axotomy of corneal trigeminal
ganglion neurons is dependent on the site of injury. Eur J Neurosci. Mar
1999;11(3):899-906. doi:10.1046/j.1460-9568.1999.00498.x
680. Chao C, Golebiowski B, Stapleton F. The role of corneal innervation in LASIK-
induced neuropathic dry eye. Ocul Surf. Jan 2014;12(1):32-45.
doi:10.1016/j.jtos.2013.09.001
681. Belmonte C, Aracil A, Acosta MC, Luna C, Gallar J. Nerves and sensations
from the eye surface. Ocul Surf. Oct 2004;2(4):248-53. doi:10.1016/s1542-
0124(12)70112-x
682. Betz J, Behrens H, Harkness BM, et al. Ocular Pain after Refractive Surgery:
Interim Analysis of Frequency and Risk Factors. Ophthalmology. Jul
2023;130(7):692-701. doi:10.1016/j.ophtha.2023.02.016
683. He M, Huang W, Zhong X. Central corneal sensitivity after small incision
lenticule extraction versus femtosecond laser-assisted LASIK for myopia: a
meta-analysis of comparative studies. BMC Ophthalmol. Oct 24 2015;15:141.
doi:10.1186/s12886-015-0129-5
684. Gomes JAP, Azar DT, Baudouin C, et al. TFOS Lifestyle: Impact of elective
medications and procedures on the ocular surface. The Ocular Surface.
2023;29:331-385.
188
685. Bech F, Gonzalez-Gonzalez O, Artime E, et al. Functional and Morphologic
Alterations in Mechanical, Polymodal, and Cold Sensory Nerve Fibers of the
Cornea Following Photorefractive Keratectomy. Invest Ophthalmol Vis Sci. May
1 2018;59(6):2281-2292. doi:10.1167/iovs.18-24007
686. Gallar J, Acosta MC, Moilanen JA, Holopainen JM, Belmonte C, Tervo TM.
Recovery of corneal sensitivity to mechanical and chemical stimulation after
laser in situ keratomileusis. J Refract Surg. May-Jun 2004;20(3):229-35.
doi:10.3928/1081-597X-20040501-06
687. Niederer RL, Perumal D, Sherwin T, McGhee CN. Corneal innervation and
cellular changes after corneal transplantation: an in vivo confocal microscopy
study. Invest Ophthalmol Vis Sci. Feb 2007;48(2):621-6. doi:10.1167/iovs.06-
0538
688. Rao GN, John T, Ishida N, Aquavella JV. Recovery of corneal sensitivity in
grafts following penetrating keratoplasty. Ophthalmology. Oct
1985;92(10):1408-11. doi:10.1016/s0161-6420(85)33857-5
689. Shimizu T, Hayashi T, Ishida A, et al. Evaluation of corneal nerves and dendritic
cells by in vivo confocal microscopy after Descemet's membrane keratoplasty
for bullous keratopathy. Sci Rep. Apr 28 2022;12(1):6936. doi:10.1038/s41598-
022-10939-w
690. Kumar RL, Koenig SB, Covert DJ. Corneal sensation after descemet stripping
and automated endothelial keratoplasty. Cornea. Jan 2010;29(1):13-8.
doi:10.1097/ICO.0b013e3181ac052b
691. Hirayama Y, Satake Y, Hirayama M, Shimazaki-Den S, Konomi K, Shimazaki J.
Changes in corneal sensation, epithelial damage, and tear function after
descemet stripping automated endothelial keratoplasty. Cornea. Sep
2013;32(9):1255-9. doi:10.1097/ICO.0b013e318299c3b7
692. Bouheraoua N, Fouquet S, Marcos-Almaraz MT, Karagogeos D, Laroche L,
Chedotal A. Genetic Analysis of the Organization, Development, and Plasticity
of Corneal Innervation in Mice. J Neurosci. Feb 13 2019;39(7):1150-1168.
doi:10.1523/JNEUROSCI.1401-18.2018
693. Jessen KR, Mirsky R. The repair Schwann cell and its function in regenerating
nerves. J Physiol. Jul 1 2016;594(13):3521-31. doi:10.1113/JP270874
694. Matsuyama A, Takatori S, Sone Y, et al. Effect of Nerve Growth Factor on
Innervation of Perivascular Nerves in Neovasculatures of Mouse Cornea. Biol
Pharm Bull. 2017;40(4):396-401. doi:10.1248/bpb.b16-00583
695. Bonini S, Lambiase A, Rama P, Caprioglio G, Aloe L. Topical treatment with
nerve growth factor for neurotrophic keratitis. Ophthalmology. Jul
2000;107(7):1347-51; discussion 1351-2. doi:10.1016/s0161-6420(00)00163-9
696. Patel S, Hwang J, Mehra D, Galor A. Corneal Nerve Abnormalities in Ocular
and Systemic Diseases. Exp Eye Res. Jan 2021;202:108284.
doi:10.1016/j.exer.2020.108284
697. Ponirakis G, Al-Janahi I, Elgassim E, et al. Sustained corneal nerve loss
predicts the development of diabetic neuropathy in type 2 diabetes. Front
Neurosci. 2024;18:1393105. doi:10.3389/fnins.2024.1393105
189
698. Galor A, Hamrah P, Haque S, Attal N, Labetoulle M. Understanding chronic
ocular surface pain: An unmet need for targeted drug therapy. Ocul Surf. Oct
2022;26:148-156. doi:10.1016/j.jtos.2022.08.005
699. Dua HS, Said DG, Messmer EM, et al. Neurotrophic keratopathy. Prog Retin
Eye Res. Sep 2018;66:107-131. doi:10.1016/j.preteyeres.2018.04.003
700. Guzman M, Miglio M, Keitelman I, et al. Transient tear hyperosmolarity disrupts
the neuroimmune homeostasis of the ocular surface and facilitates dry eye
onset. Immunology. Oct 2020;161(2):148-161. doi:10.1111/imm.13243
701. Chen M, Seo S, Simmons X, et al. Precise longitudinal monitoring of corneal
change through in vivo confocal microscopy in a rat dry eye disease model. Mol
Vis. 2024;30:150-159.
702. Vereertbrugghen A, Galletti JG. Corneal nerves and their role in dry eye
pathophysiology. Exp Eye Res. Sep 2022;222:109191.
doi:10.1016/j.exer.2022.109191
703. Maity M, Allay MB, Ali MH, Deshmukh R, Basu S, Singh S. Association of tear
osmolarity and corneal nerves structure in dry eye disease: an in vivo study.
Graefes Arch Clin Exp Ophthalmol. Mar 2025;263(3):753-760.
doi:10.1007/s00417-024-06657-w
704. Corcoran P, Hollander DA, Ousler GW, 3rd, et al. Dynamic Sensitivity of
Corneal TRPM8 Receptors to Menthol Instillation in Dry Eye Versus Normal
Subjects. J Ocul Pharmacol Ther. Nov 2017;33(9):686-692.
doi:10.1089/jop.2017.0050
705. Talens-Estarelles C, Golebiowski B, Ehrmann K, Garcia-Lazaro S, Cervino A,
Stapleton F. Corneal hypersensitivity to cold stimuli in symptomatic computer
users. Optom Vis Sci. Sep 1 2024;101(9):571-578.
doi:10.1097/OPX.0000000000002183
706. Situ P, Simpson T, Begley C. Hypersensitivity to Cold Stimuli in Symptomatic
Contact Lens Wearers. Optom Vis Sci. Aug 2016;93(8):909-16.
doi:10.1097/OPX.0000000000000857
707. Belmonte C. Pain, Dryness, and Itch Sensations in Eye Surface Disorders Are
Defined By a Balance Between Inflammation and Sensory Nerve Injury.
Cornea. Nov 2019;38 Suppl 1:S11-S24. doi:10.1097/ICO.0000000000002116
708. Situ P, Begley CG, Simpson TL. Effects of Tear Film Instability on Sensory
Responses to Corneal Cold, Mechanical, and Chemical Stimuli. Invest
Ophthalmol Vis Sci. Jul 1 2019;60(8):2935-2941. doi:10.1167/iovs.19-27298
709. Kovacs I, Luna C, Quirce S, et al. Abnormal activity of corneal cold
thermoreceptors underlies the unpleasant sensations in dry eye disease. Pain.
Feb 2016;157(2):399-417. doi:10.1097/j.pain.0000000000000455
710. Kovacs I, Dienes L, Perenyi K, et al. Lacosamide diminishes dryness-induced
hyperexcitability of corneal cold sensitive nerve terminals. Eur J Pharmacol.
Sep 15 2016;787:2-8. doi:10.1016/j.ejphar.2016.05.044
711. Mecum NE, Russell R, Lee J, Sullivan C, Meng ID. Optogenetic Inhibition of
Nav1.8 Expressing Corneal Afferents Reduces Persistent Dry Eye Pain. Invest
Ophthalmol Vis Sci. Nov 1 2021;62(14):15. doi:10.1167/iovs.62.14.15
190
712. De Lott LB, Kaplan C, Harte S, et al. Nociplastic pain among individuals with
chronic ocular surface pain: One cause for "pain without stain"? Surv
Ophthalmol. Jan 13 2025;doi:10.1016/j.survophthal.2025.01.004
713. Stepp MA, Pal-Ghosh S, Tadvalkar G, Williams AR, Pflugfelder SC, de Paiva
CS. Reduced Corneal Innervation in the CD25 Null Model of Sjogren
Syndrome. Int J Mol Sci. Nov 30 2018;19(12)doi:10.3390/ijms19123821
714. Shimizu S, Sato S, Taniguchi H, et al. Observation of Chronic Graft-Versus-
Host Disease Mouse Model Cornea with In Vivo Confocal Microscopy.
Diagnostics (Basel). Aug 23 2021;11(8)doi:10.3390/diagnostics11081515
715. Medeiros CS, Marino GK, Lassance L, Thangavadivel S, Santhiago MR, Wilson
SE. The Impact of Photorefractive Keratectomy and Mitomycin C on Corneal
Nerves and Their Regeneration. J Refract Surg. Dec 1 2018;34(12):790-798.
doi:10.3928/1081597X-20181112-01
716. Suanno G, Fonteyne P, Ferrari G. Neurosensory abnormalities and stability of a
mouse model of dry eye disease. Exp Eye Res. Jul 2023;232:109516.
doi:10.1016/j.exer.2023.109516
717. Kurose M, Meng ID. Dry eye modifies the thermal and menthol responses in rat
corneal primary afferent cool cells. J Neurophysiol. Jul 2013;110(2):495-504.
doi:10.1152/jn.00222.2013
718. Gyenes A, Tapaszto Z, Quirce S, et al. Cyclosporine A Decreases Dryness-
Induced Hyperexcitability of Corneal Cold-Sensitive Nerve Terminals. Int J Mol
Sci. Aug 21 2023;24(16)doi:10.3390/ijms241613025
719. Tran BN, Maass M, Musial G, Stern ME, Gehlsen U, Steven P. Topical
application of cannabinoid-ligands ameliorates experimental dry-eye disease.
Ocul Surf. Jan 2022;23:131-139. doi:10.1016/j.jtos.2021.12.008
720. Belmonte C, Nichols JJ, Cox SM, et al. TFOS DEWS II pain and sensation
report. Ocul Surf. Jul 2017;15(3):404-437. doi:10.1016/j.jtos.2017.05.002
721. Eguchi H, Hiura A, Nakagawa H, Kusaka S, Shimomura Y. Corneal Nerve Fiber
Structure, Its Role in Corneal Function, and Its Changes in Corneal Diseases.
Biomed Res Int. 2017;2017:3242649. doi:10.1155/2017/3242649
722. Tepelus TC, Chiu GB, Huang J, et al. Correlation between corneal innervation
and inflammation evaluated with confocal microscopy and symptomatology in
patients with dry eye syndromes: a preliminary study. Graefes Arch Clin Exp
Ophthalmol. Sep 2017;255(9):1771-1778. doi:10.1007/s00417-017-3680-3
723. Li F, Zhang Q, Ying X, et al. Corneal nerve structure in patients with primary
Sjogren's syndrome in China. BMC Ophthalmol. May 12 2021;21(1):211.
doi:10.1186/s12886-021-01967-7
724. Oliveira-Soto L, Efron N. Morphology of corneal nerves using confocal
microscopy. Cornea. May 2001;20(4):374-84. doi:10.1097/00003226-
200105000-00008
725. Guerrero-Moreno A, Liang H, Moreau N, et al. Corneal Nerve Abnormalities in
Painful Dry Eye Disease Patients. Biomedicines. Oct 9
2021;9(10)doi:10.3390/biomedicines9101424
726. Luzu J, Labbe A, Reaux-Le Goazigo A, et al. In vivo confocal microscopic study
of corneal innervation in Sjogren's Syndrome with or without small fiber
neuropathy. Ocul Surf. Jul 2022;25:155-162. doi:10.1016/j.jtos.2022.07.003
191
727. He J, Ogawa Y, Mukai S, et al. In Vivo Confocal Microscopy Evaluation of
Ocular Surface with Graft-Versus-Host Disease-Related Dry Eye Disease. Sci
Rep. Sep 6 2017;7(1):10720. doi:10.1038/s41598-017-10237-w
728. Chen X, Graham J, Dabbah MA, Petropoulos IN, Tavakoli M, Malik RA. An
Automatic Tool for Quantification of Nerve Fibers in Corneal Confocal
Microscopy Images. IEEE Trans Biomed Eng. Apr 2017;64(4):786-794.
doi:10.1109/TBME.2016.2573642
729. Moein HR, Akhlaq A, Dieckmann G, et al. Visualization of microneuromas by
using in vivo confocal microscopy: An objective biomarker for the diagnosis of
neuropathic corneal pain? Ocul Surf. Oct 2020;18(4):651-656.
doi:10.1016/j.jtos.2020.07.004
730. Kobashi H, Kamiya K, Sambe T, Nakagawa R. Factors influencing subjective
symptoms in dry eye disease. Int J Ophthalmol. 2018;11(12):1926-1931.
doi:10.18240/ijo.2018.12.08
731. Cox SM, Kheirkhah A, Aggarwal S, et al. Alterations in corneal nerves in
different subtypes of dry eye disease: An in vivo confocal microscopy study.
Ocul Surf. Oct 2021;22:135-142. doi:10.1016/j.jtos.2021.08.004
732. Giannaccare G, Pellegrini M, Sebastiani S, Moscardelli F, Versura P, Campos
EC. In vivo confocal microscopy morphometric analysis of corneal subbasal
nerve plexus in dry eye disease using newly developed fully automated system.
Graefes Arch Clin Exp Ophthalmol. Mar 2019;257(3):583-589.
doi:10.1007/s00417-018-04225-7
733. Kasikci M, Erogul O, Polat O. Evaluation of aqueous-deficient and evaporative
dry eye cases with confocal microscopy. J Fr Ophtalmol. Dec
2023;46(10):1161-1168. doi:10.1016/j.jfo.2023.05.024
734. Shetty R, Sethu S, Deshmukh R, et al. Corneal Dendritic Cell Density Is
Associated with Subbasal Nerve Plexus Features, Ocular Surface Disease
Index, and Serum Vitamin D in Evaporative Dry Eye Disease. Biomed Res Int.
2016;2016:4369750. doi:10.1155/2016/4369750
735. Nicolle P, Liang H, Reboussin E, et al. Proinflammatory Markers, Chemokines,
and Enkephalin in Patients Suffering from Dry Eye Disease. Int J Mol Sci. Apr
17 2018;19(4)doi:10.3390/ijms19041221
736. Dikmetas O, Kocabeyoglu S, Mocan MC. Evaluation of Visual Acuity Outcomes
and Corneal Alterations of New Generation Hybrid Contact Lenses in Patients
With Advanced Keratoconus. Cornea. Nov 2020;39(11):1366-1370.
doi:10.1097/ICO.0000000000002438
737. D'Souza S, Padmanabhan Nair A, Iyappan G, et al. Clinical and Molecular
Outcomes After Combined Intense Pulsed Light Therapy With Low-Level Light
Therapy in Recalcitrant Evaporative Dry Eye Disease With Meibomian Gland
Dysfunction. Cornea. Sep 1 2022;41(9):1080-1087.
doi:10.1097/ICO.0000000000002954
738. Jing D, Liu Y, Chou Y, et al. Change patterns in the corneal sub-basal nerve
and corneal aberrations in patients with dry eye disease: An artificial intelligence
analysis. Exp Eye Res. Feb 2022;215:108851. doi:10.1016/j.exer.2021.108851
739. Zhang Y, Wu Y, Li W, Huang X. Semiautomated and Automated Quantitative
Analysis of Corneal Sub-Basal Nerves in Patients With DED With Ocular Pain
192
Using IVCM. Front Med (Lausanne). 2022;9:831307.
doi:10.3389/fmed.2022.831307
740. Chinnery HR, Rajan R, Jiao H, et al. Identification of presumed corneal
neuromas and microneuromas using laser-scanning in vivo confocal
microscopy: a systematic review. Br J Ophthalmol. Jun 2022;106(6):765-771.
doi:10.1136/bjophthalmol-2020-318156
741. Stepp MA, Pal-Ghosh S, Downie LE, et al. Corneal Epithelial "Neuromas": A
Case of Mistaken Identity? Cornea. Jul 2020;39(7):930-934.
doi:10.1097/ICO.0000000000002294
742. D'Souza S, Shetty R, Nair AP, et al. Corneal Confocal Microscopy Features and
Tear Molecular Profile in Study Participants with Discordance between Ocular
Surface Disease Clinical Signs and Discomfort. J Clin Med. Apr 25
2022;11(9)doi:10.3390/jcm11092407
743. Dermer H, Hwang J, Mittal R, Cohen AK, Galor A. Corneal sub-basal nerve
plexus microneuromas in individuals with and without dry eye. Br J Ophthalmol.
May 2022;106(5):616-622. doi:10.1136/bjophthalmol-2020-317891
744. Wei S, Shi F, Wang Y, Chou Y, Li X. A Deep Learning Model for Automated
Sub-Basal Corneal Nerve Segmentation and Evaluation Using In Vivo Confocal
Microscopy. Transl Vis Sci Technol. Jun 2020;9(2):32. doi:10.1167/tvst.9.2.32
745. Tuisku IS, Konttinen YT, Konttinen LM, Tervo TM. Alterations in corneal
sensitivity and nerve morphology in patients with primary Sjogren's syndrome.
Exp Eye Res. Jun 2008;86(6):879-85. doi:10.1016/j.exer.2008.03.002
746. Situ P, Simpson TL, Fonn D, Jones LW. Conjunctival and corneal pneumatic
sensitivity is associated with signs and symptoms of ocular dryness. Invest
Ophthalmol Vis Sci. Jul 2008;49(7):2971-6. doi:10.1167/iovs.08-1734
747. Rahman EZ, Lam PK, Chu CK, Moore Q, Pflugfelder SC. Corneal Sensitivity in
Tear Dysfunction and its Correlation With Clinical Parameters and Blink Rate.
Am J Ophthalmol. Nov 2015;160(5):858-866 e5. doi:10.1016/j.ajo.2015.08.005
748. Bourcier T, Acosta MC, Borderie V, et al. Decreased corneal sensitivity in
patients with dry eye. Invest Ophthalmol Vis Sci. Jul 2005;46(7):2341-5.
doi:10.1167/iovs.04-1426
749. Galor A, Felix ER, Feuer W, Levitt RC, Sarantopoulos CD. Corneal Nerve
Pathway Function in Individuals with Dry Eye Symptoms. Ophthalmology. Apr
2021;128(4):619-621. doi:10.1016/j.ophtha.2020.07.061
750. Golebiowski B, Papas E, Stapleton F. Assessing the sensory function of the
ocular surface: implications of use of a non-contact air jet aesthesiometer
versus the Cochet-Bonnet aesthesiometer. Exp Eye Res. May 2011;92(5):408-
13. doi:10.1016/j.exer.2011.02.016
751. Spierer O, Felix ER, McClellan AL, et al. Corneal Mechanical Thresholds
Negatively Associate With Dry Eye and Ocular Pain Symptoms. Invest
Ophthalmol Vis Sci. Feb 2016;57(2):617-25. doi:10.1167/iovs.15-18133
752. Tagawa Y, Noda K, Ohguchi T, Tagawa Y, Ishida S, Kitaichi N. Corneal
hyperalgesia in patients with short tear film break-up time dry eye. Ocul Surf.
Jan 2019;17(1):55-59. doi:10.1016/j.jtos.2018.08.004
753. Carreno-Galeano JT, Johns LK, Dana R, Yin J. Autologous serum tears
improve corneal nerve density and sensitivity in patients with ocular graft-
193
versus-host disease-associated dry eye disease. Ocul Surf. Apr 2023;28:37-39.
doi:10.1016/j.jtos.2023.01.004
754. Levy O, Labbe A, Borderie V, et al. Increased corneal sub-basal nerve density
in patients with Sjogren syndrome treated with topical cyclosporine A. Clin Exp
Ophthalmol. Jul 2017;45(5):455-463. doi:10.1111/ceo.12898
755. Kaido M, Kawashima M, Shigeno Y, Yamada Y, Tsubota K. Randomized
Controlled Study to Investigate the Effect of Topical Diquafosol Tetrasodium on
Corneal Sensitivity in Short Tear Break-Up Time Dry Eye. Adv Ther. May
2018;35(5):697-706. doi:10.1007/s12325-018-0685-1
756. Ayoubi M, Cabrera K, Locatelli EV, Felix ER, Galor A. Associations between
Corneal Nerve Structure and Function in a Veteran Population. J Clin Med. Apr
25 2024;13(9)doi:10.3390/jcm13092513
757. Qin G, Chen J, Li L, et al. Relationship between ocular surface pain and corneal
nerve loss in dry eye diabetics: a cross-sectional study in Shenyang, China.
BMJ Open. Sep 26 2023;13(9):e076932. doi:10.1136/bmjopen-2023-076932
758. Wang MTM, Meyer JJ, Xue AL, Power B, Craig JP. Predictive performance of
corneal and lid margin sensitivity for dry eye disease: An investigator-masked,
prospective, prognostic accuracy study. Ocul Surf. Jul 2024;33:11-15.
doi:10.1016/j.jtos.2024.03.010
759. Crabtree JR, Tannir S, Tran K, Boente CS, Ali A, Borschel GH. Corneal Nerve
Assessment by Aesthesiometry: History, Advancements, and Future Directions.
Vision (Basel). May 12 2024;8(2)doi:10.3390/vision8020034
760. Nosch DS, Kaser E, Bracher T, Joos RE. Clinical application of the Swiss Liquid
Jet Aesthesiometer for corneal sensitivity measurement. Clin Exp Optom. Jan
2024;107(1):14-22. doi:10.1080/08164622.2023.2191782
761. Ehrmann K, Talens-Estarelles C, Stapleton F, Truong B, Chen J, Golebiowski
B. A New Method to Measure Ocular Surface Sensitivity: Repeatability and
Reproducibility of the Liquid Jet Esthesiometer. Eye & Contact Lens.
2024:10.1097/ICL.0000000000001146. doi:10.1097/icl.0000000000001146
762. Giovannetti F, Sacchetti M, Marenco M, et al. New disposable esthesiometer
(KeraSenseⓇ) to improve diagnosis and management of neurotrophic keratitis.
Ocul Surf. Apr 2024;32:192-197. doi:10.1016/j.jtos.2024.03.009
763. Dieckmann G, Goyal S, Hamrah P. Neuropathic Corneal Pain: Approaches for
Management. Ophthalmology. Nov 2017;124(11S):S34-S47.
doi:10.1016/j.ophtha.2017.08.004
764. Goyal S, Hamrah P. Understanding Neuropathic Corneal Pain--Gaps and
Current Therapeutic Approaches. Semin Ophthalmol. 2016;31(1-2):59-70.
doi:10.3109/08820538.2015.1114853
765. Rosenthal P, Borsook D. Ocular neuropathic pain. Br J Ophthalmol. Jan
2016;100(1):128-34. doi:10.1136/bjophthalmol-2014-306280
766. Crane AM, Feuer W, Felix ER, et al. Evidence of central sensitisation in those
with dry eye symptoms and neuropathic-like ocular pain complaints: incomplete
response to topical anaesthesia and generalised heightened sensitivity to
evoked pain. Br J Ophthalmol. Sep 2017;101(9):1238-1243.
doi:10.1136/bjophthalmol-2016-309658
194
767. Kim J, Yoon HJ, You IC, Ko BY, Yoon KC. Clinical characteristics of dry eye
with ocular neuropathic pain features: comparison according to the types of
sensitization based on the Ocular Pain Assessment Survey. BMC Ophthalmol.
Nov 18 2020;20(1):455. doi:10.1186/s12886-020-01733-1
768. Galor A, Levitt RC, McManus KT, et al. Assessment of Somatosensory Function
in Patients With Idiopathic Dry Eye Symptoms. JAMA Ophthalmol. Nov 1
2016;134(11):1290-1298. doi:10.1001/jamaophthalmol.2016.3642
769. Shtein RM, Harper DE, Pallazola V, et al. Discordant Dry Eye Disease (An
American Ophthalmological Society Thesis). Trans Am Ophthalmol Soc. Aug
2016;114:T4.
770. Ong ES, Felix ER, Levitt RC, Feuer WJ, Sarantopoulos CD, Galor A.
Epidemiology of discordance between symptoms and signs of dry eye. Br J
Ophthalmol. May 2018;102(5):674-679. doi:10.1136/bjophthalmol-2017-310633
771. Levitt AE, Galor A, Small L, Feuer W, Felix ER. Pain sensitivity and autonomic
nervous system parameters as predictors of dry eye symptoms after LASIK.
Ocul Surf. Jan 2021;19:275-281. doi:10.1016/j.jtos.2020.10.004
772. Rodriguez DA, Galor A, Felix ER. Self-Report of Severity of Ocular Pain Due to
Light as a Predictor of Altered Central Nociceptive System Processing in
Individuals With Symptoms of Dry Eye Disease. J Pain. May 2022;23(5):784-
795. doi:10.1016/j.jpain.2021.11.010
773. Choudhury A, Reyes N, Galor A, Mehra D, Felix E, Moulton EA. Clinical
Neuroimaging of Photophobia in Individuals With Chronic Ocular Surface Pain.
Am J Ophthalmol. Feb 2023;246:20-30. doi:10.1016/j.ajo.2022.09.020
774. Holmes S, Reyes N, Huang JJ, et al. Disentangling the neurological basis of
chronic ocular pain using clinical, self-report, and brain imaging data: use of K-
means clustering to explore patient phenotypes. Front Neurol.
2023;14:1265082. doi:10.3389/fneur.2023.1265082
775. Glover GH. Overview of functional magnetic resonance imaging. Neurosurg Clin
N Am. Apr 2011;22(2):133-9, vii. doi:10.1016/j.nec.2010.11.001
776. Reyes N, Huang JJ, Choudhury A, et al. Botulinum toxin A decreases neural
activity in pain-related brain regions in individuals with chronic ocular pain and
photophobia. Front Neurosci. 2023;17:1202341.
doi:10.3389/fnins.2023.1202341
777. Reyes N, Huang JJ, Choudhury A, et al. FL-41 Tint Reduces Activation of
Neural Pathways of Photophobia in Patients with Chronic Ocular Pain. Am J
Ophthalmol. Mar 2024;259:172-184. doi:10.1016/j.ajo.2023.12.004
778. Gomes JAP, Azar DT, Baudouin C, et al. Tfos dews ii iatrogenic report. The
ocular surface. 2017;15(3):511-538.
779. Stapleton F, Alves M, Bunya VY, et al. Tfos dews ii epidemiology report. The
ocular surface. 2017;15(3):334-365.
780. Van Went C, Brasnu E, Hamard P, Baudouin C, Labbe A. [The influence of
ocular surface diseases in the management of glaucoma]. J Fr Ophtalmol. Apr
2011;34(4):230-7. Influence des pathologies de la surface oculaire sur le
traitement du glaucome. doi:10.1016/j.jfo.2010.11.010
781. Jaenen N, Baudouin C, Pouliquen P, Manni G, Figueiredo A, Zeyen T. Ocular
symptoms and signs with preserved and preservative-free glaucoma
195
medications. Eur J Ophthalmol. May-Jun 2007;17(3):341-9.
doi:10.1177/112067210701700311
782. Sahlu M, Giorgis AT. Dry eye disease among Glaucoma patients on topical
hypotensive medications, in a tertiary hospital, Ethiopia. BMC Ophthalmol. Mar
30 2021;21(1):155. doi:10.1186/s12886-021-01917-3
783. Leung EW, Medeiros FA, Weinreb RN. Prevalence of ocular surface disease in
glaucoma patients. J Glaucoma. Aug 2008;17(5):350-5.
doi:10.1097/IJG.0b013e31815c5f4f
784. Erb C, Gast U, Schremmer D. German register for glaucoma patients with dry
eye. I. Basic outcome with respect to dry eye. Graefes Arch Clin Exp
Ophthalmol. Nov 2008;246(11):1593-601. doi:10.1007/s00417-008-0881-9
785. Pisella PJ, Pouliquen P, Baudouin C. Prevalence of ocular symptoms and signs
with preserved and preservative free glaucoma medication. Br J Ophthalmol.
Apr 2002;86(4):418-23. doi:10.1136/bjo.86.4.418
786. Nagstrup AH. The use of benzalkonium chloride in topical glaucoma treatment:
An investigation of the efficacy and safety of benzalkonium chloride-preserved
intraocular pressure-lowering eye drops and their effect on conjunctival goblet
cells. Acta Ophthalmol. Dec 2023;101 Suppl 278:3-21. doi:10.1111/aos.15808
787. Rossi GC, Pasinetti GM, Scudeller L, Bianchi PE. Ocular surface disease and
glaucoma: how to evaluate impact on quality of life. J Ocul Pharmacol Ther.
May 2013;29(4):390-4. doi:10.1089/jop.2011.0159
788. Tirpack AR, Vanner E, Parrish JM, Galor A, Hua HU, Wellik SR. Dry Eye
Symptoms and Ocular Pain in Veterans with Glaucoma. J Clin Med. Jul 22
2019;8(7)doi:10.3390/jcm8071076
789. Fineide F, Magno M, Dahlo K, et al. Topical glaucoma medications - Possible
implications on the meibomian glands. Acta Ophthalmol. Nov 2024;102(7):735-
748. doi:10.1111/aos.16728
790. Vitoux MA, Kessal K, Melik Parsadaniantz S, et al. Benzalkonium chloride-
induced direct and indirect toxicity on corneal epithelial and trigeminal neuronal
cells: proinflammatory and apoptotic responses in vitro. Toxicol Lett. Feb 1
2020;319:74-84. doi:10.1016/j.toxlet.2019.10.014
791. Rolando M, Barabino S, Mingari C, Moretti S, Giuffrida S, Calabria G.
Distribution of conjunctival HLA-DR expression and the pathogenesis of
damage in early dry eyes. Cornea. Nov 2005;24(8):951-4.
doi:10.1097/01.ico.0000157421.93522.00
792. Zhang R, Park M, Richardson A, et al. Dose-dependent benzalkonium chloride
toxicity imparts ocular surface epithelial changes with features of dry eye
disease. Ocul Surf. Jan 2020;18(1):158-169. doi:10.1016/j.jtos.2019.11.006
793. Swamynathan SK, Wells A. Conjunctival goblet cells: Ocular surface functions,
disorders that affect them, and the potential for their regeneration. Ocul Surf.
Jan 2020;18(1):19-26. doi:10.1016/j.jtos.2019.11.005
794. Chen W, Li Z, Hu J, et al. Corneal alternations induced by topical application of
benzalkonium chloride in rabbit. PLoS One. 2011;6(10):e26103.
doi:10.1371/journal.pone.0026103
196
795. Izzotti A, La Maestra S, Micale RT, Longobardi MG, Sacca SC. Genomic and
post-genomic effects of anti-glaucoma drugs preservatives in trabecular
meshwork. Mutat Res. Feb 2015;772:1-9. doi:10.1016/j.mrfmmm.2014.11.006
796. Lou Q, Pan L, Xiang S, et al. Suppression of NLRP3/Caspase-1/GSDMD
Mediated Corneal Epithelium Pyroptosis Using Melatonin-Loaded Liposomes to
Inhibit Benzalkonium Chloride-Induced Dry Eye Disease. Int J Nanomedicine.
2023;18:2447-2463. doi:10.2147/IJN.S403337
797. Sarkar J, Chaudhary S, Namavari A, et al. Corneal neurotoxicity due to topical
benzalkonium chloride. Invest Ophthalmol Vis Sci. Apr 6 2012;53(4):1792-802.
doi:10.1167/iovs.11-8775
798. Kahook MY, Rapuano CJ, Messmer EM, Radcliffe NM, Galor A, Baudouin C.
Preservatives and ocular surface disease: A review. Ocul Surf. Oct
2024;34:213-224. doi:10.1016/j.jtos.2024.08.001
799. Tiedemann D, Mouhammad ZA, Utheim TP, et al. Conjunctival Goblet Cells, the
Overlooked Cells in Glaucoma Treatment. J Glaucoma. Apr 2019;28(4):325-
333. doi:10.1097/IJG.0000000000001168
800. Skov AG, Rives AS, Freiberg J, Virgili G, Azuara-Blanco A, Kolko M.
Comparative efficacy and safety of preserved versus preservative-free beta-
blockers in patients with glaucoma or ocular hypertension: a systematic review.
Acta Ophthalmol. May 2022;100(3):253-261. doi:10.1111/aos.14926
801. Jee D, Park SH, Kim MS, Kim EC. Antioxidant and inflammatory cytokine in
tears of patients with dry eye syndrome treated with preservative-free versus
preserved eye drops. Invest Ophthalmol Vis Sci. Jul 3 2014;55(8):5081-9.
doi:10.1167/iovs.14-14483
802. Nasser L, Rozycka M, Gomez Rendon G, Navas A. Real-life results of switching
from preserved to preservative-free artificial tears containing hyaluronate in
patients with dry eye disease. Clin Ophthalmol. 2018;12:1519-1525.
doi:10.2147/OPTH.S160053
803. Bastelica P, Renard JP, Aptel F, et al. The PRAMOS Study: PRostaglandin
Analogues Monotherapy-Awareness Survey on Ocular Surface Involvement.
Ophthalmol Ther. Jun 2024;13(6):1537-1551. doi:10.1007/s40123-024-00936-9
804. Jandrokovic S, Vidas Pauk S, Lesin Gacina D, et al. Tolerability in Glaucoma
Patients Switched from Preserved to Preservative-Free Prostaglandin-Timolol
Combination: A Prospective Real-Life Study. Clin Ophthalmol. 2022;16:3181-
3192. doi:10.2147/OPTH.S382497
805. Kim DW, Shin J, Lee CK, Kim M, Lee S, Rho S. Comparison of ocular surface
assessment and adherence between preserved and preservative-free
latanoprost in glaucoma: a parallel-grouped randomized trial. Sci Rep. Jul 22
2021;11(1):14971. doi:10.1038/s41598-021-94574-x
806. Liang H, Baudouin C, Daull P, Garrigue JS, Brignole-Baudouin F. In Vitro
Corneal and Conjunctival Wound-Healing Assays as a Tool for Antiglaucoma
Prostaglandin Formulation Characterization. Front Biosci (Landmark Ed). May 7
2022;27(5):147. doi:10.31083/j.fbl2705147
807. Gazzard G, Konstantakopoulou E, Garway-Heath D, et al. Laser in Glaucoma
and Ocular Hypertension (LiGHT) trial. A multicentre, randomised controlled
197
trial: design and methodology. Br J Ophthalmol. May 2018;102(5):593-598.
doi:10.1136/bjophthalmol-2017-310877
808. Wong J, Lan W, Ong LM, Tong L. Non-hormonal systemic medications and dry
eye. Ocul Surf. Oct 2011;9(4):212-26. doi:10.1016/s1542-0124(11)70034-9
809. Schein OD, Hochberg MC, Munoz B, et al. Dry eye and dry mouth in the elderly:
a population-based assessment. Arch Intern Med. Jun 28 1999;159(12):1359-
63. doi:10.1001/archinte.159.12.1359
810. Guo M, Diaz GM, Yu Y, et al. Association between systemic medication use
and severity of dry eye signs and symptoms in the DRy eye assessment and
management (DREAM) study. Ocul Surf. Apr 2024;32:112-119.
doi:10.1016/j.jtos.2024.01.009
811. Dana R, Bradley JL, Guerin A, Pivneva I, Evans AM, Stillman IO. Comorbidities
and Prescribed Medications in Patients With or Without Dry Eye Disease: A
Population-Based Study. Am J Ophthalmol. Feb 2019;198:181-192.
doi:10.1016/j.ajo.2018.10.001
812. Ghossein N, Kang M, Lakhkar AD. Anticholinergic medications. 2020;
813. Wellington K, Jarvis B. Cetirizine/pseudoephedrine. Drugs. 2001;61(15):2231-
40; discussion 2241-2. doi:10.2165/00003495-200161150-00009
814. Vardy MD, Mitcheson HD, Samuels TA, et al. Effects of solifenacin on
overactive bladder symptoms, symptom bother and other patient-reported
outcomes: results from VIBRANT - a double-blind, placebo-controlled trial. Int J
Clin Pract. Dec 2009;63(12):1702-14. doi:10.1111/j.1742-1241.2009.02209.x
815. Ousler GW, Wilcox KA, Gupta G, Abelson MB. An evaluation of the ocular
drying effects of 2 systemic antihistamines: loratadine and cetirizine
hydrochloride. Ann Allergy Asthma Immunol. Nov 2004;93(5):460-4.
doi:10.1016/s1081-1206(10)61413-5
816. Kam WR, Sullivan DA. Neurotransmitter influence on human meibomian gland
epithelial cells. Invest Ophthalmol Vis Sci. Nov 1 2011;52(12):8543-8.
doi:10.1167/iovs.11-8113
817. Vallerand IA, Lewinson RT, Farris MS, et al. Efficacy and adverse events of oral
isotretinoin for acne: a systematic review. Br J Dermatol. Jan 2018;178(1):76-
85. doi:10.1111/bjd.15668
818. Fouladgar N, Khabazkhoob M, Hanifnia AR, Yekta A, Mirzajani A. Evaluation of
the effects of isotretinoin for treatment of acne on corneal sensitivity. J Curr
Ophthalmol. Dec 2018;30(4):326-329. doi:10.1016/j.joco.2018.06.005
819. Caglar C, Senel E, Sabancilar E, Durmus M. Reduced ocular surface disease
index (OSDI) scores in patients with isotretinoin treatment. Int Ophthalmol. Feb
2017;37(1):197-202. doi:10.1007/s10792-016-0263-y
820. Duzgun E, Ozkur E. The effect of oral isotretinoin therapy on meibomian gland
morphology and dry eye tests. J Dermatolog Treat. Mar 2022;33(2):762-768.
doi:10.1080/09546634.2020.1774041
821. Zhang P, Tian L, Bao J, et al. Isotretinoin Impairs the Secretory Function of
Meibomian Gland Via the PPARγ Signaling Pathway. Invest Ophthalmol Vis
Sci. Mar 2 2022;63(3):29. doi:10.1167/iovs.63.3.29
198
822. Aslan Bayhan S, Bayhan HA, Çölgeçen E, Gürdal C. Effects of topical acne
treatment on the ocular surface in patients with acne vulgaris. Cont Lens
Anterior Eye. Dec 2016;39(6):431-434. doi:10.1016/j.clae.2016.06.009
823. Santaella RM, Fraunfelder FW. Ocular adverse effects associated with systemic
medications : recognition and management. Drugs. 2007;67(1):75-93.
doi:10.2165/00003495-200767010-00006
824. Wang SQ, Zhang LW, Wei P, Hua H. Is hydroxychloroquine effective in treating
primary Sjogren's syndrome: a systematic review and meta-analysis. BMC
Musculoskelet Disord. May 12 2017;18(1):186. doi:10.1186/s12891-017-1543-z
825. Chu LL, Cui K, Pope JE. Meta-Analysis of Treatment for Primary Sjogren's
Syndrome. Arthritis Care Res (Hoboken). Jul 2020;72(7):1011-1021.
doi:10.1002/acr.23917
826. Gottenberg JE, Ravaud P, Puechal X, et al. Effects of hydroxychloroquine on
symptomatic improvement in primary Sjogren syndrome: the JOQUER
randomized clinical trial. JAMA. Jul 16 2014;312(3):249-58.
doi:10.1001/jama.2014.7682
827. Fraunfelder FW, Solomon J, Mehelas TJ. Ocular adverse effects associated
with cyclooxygenase-2 inhibitors. Arch Ophthalmol. Feb 2006;124(2):277-9.
doi:10.1001/archopht.124.2.277
828. Yazici A, Sari E, Ayhan E, et al. The Effect of Low-Dose Aspirin on Dry Eye
Parameters and Ocular Surface Disease Index Questionnaire. J Ocul
Pharmacol Ther. Apr 2018;34(3):256-259. doi:10.1089/jop.2017.0064
829. Moss SE, Klein R, Klein BE. Prevalence of and risk factors for dry eye
syndrome. Arch Ophthalmol. Sep 2000;118(9):1264-8.
doi:10.1001/archopht.118.9.1264
830. Liu SH, Saldanha IJ, Abraham AG, et al. Topical corticosteroids for dry eye.
Cochrane Database Syst Rev. Oct 21 2022;10(10):CD015070.
doi:10.1002/14651858.CD015070.pub2
831. Valenzuela CV, Liu JC, Vila PM, Simon L, Doering M, Lieu JEC. Intranasal
Corticosteroids Do Not Lead to Ocular Changes: A Systematic Review and
Meta-analysis. Laryngoscope. Jan 2019;129(1):6-12. doi:10.1002/lary.27209
832. Schlatter A, Hommer N, Kallab M, et al. Effect of Treatment with Topical
Azithromycin or Oral Doxycycline on Tear Film Thickness in Patients with
Meibomian Gland Dysfunction: A Randomized Controlled Trial. J Ocul
Pharmacol Ther. Jul 2023;39(6):371-378. doi:10.1089/jop.2022.0186
833. Ben Ephraim Noyman D, Chan CC, Mimouni M, Safir M. Systemic antibiotic
treatment for meibomian gland dysfunction-A systematic review and meta-
analysis. Acta Ophthalmol. Feb 2024;102(1):e1-e10. doi:10.1111/aos.15681
834. Pejcic AV. Stevens-Johnson syndrome and toxic epidermal necrolysis
associated with the use of macrolide antibiotics: a review of published cases. Int
J Dermatol. Jan 2021;60(1):12-24. doi:10.1111/ijd.15144
835. Nappe TM, Goren-Garcia SL, Jacoby JL. Stevens-Johnson syndrome after
treatment with azithromycin: an uncommon culprit. Am J Emerg Med. Mar
2016;34(3):676 e1-3. doi:10.1016/j.ajem.2015.06.039
199
836. Rakofsky JJ, Rakofsky SI, Dunlop BW. Dry Those Crying Eyes: The Role of
Depression and Antidepressants in Dry Eye Disease. J Clin Psychopharmacol.
May-Jun 01 2021;41(3):295-303. doi:10.1097/JCP.0000000000001382
837. Isik-Ulusoy S, Ulusoy MO. Influence of Different Antidepressants on Ocular
Surface in Patients With Major Depressive Disorder. J Clin Psychopharmacol.
Jan/Feb 01 2021;41(1):49-52. doi:10.1097/JCP.0000000000001325
838. Zhang X, Yin Y, Yue L, Gong L. Selective Serotonin Reuptake Inhibitors
Aggravate Depression-Associated Dry Eye Via Activating the NF-kappaB
Pathway. Invest Ophthalmol Vis Sci. Jan 2 2019;60(1):407-419.
doi:10.1167/iovs.18-25572
839. Dixitha V. AB, Bhavya H. U., Mahesh Babu, Sudhakar N. A. Quantitative
Analysis of Tear Film in Patients on Atypical Antipsychotics. Ocul Immunol
Inflamm. Jul 2024;32(5):566-571. doi:10.1080/09273948.2023.2177175
840. Acan D, Kurtgoz P. Influence of selective serotonin reuptake inhibitors on ocular
surface. Clin Exp Optom. Jan 2017;100(1):83-86. doi:10.1111/cxo.12415
841. Ismayilov AS, Celikel G. Effects of tricyclic antidepressants, selective serotonin
reuptake inhibitors, and selective serotonin-norepinephrine reuptake inhibitors
on the ocular surface. Arq Bras Oftalmol. 2022;86(5):e20230068.
doi:10.5935/0004-2749.20230068
842. Park JSY, Sharma RA, Sharma V. Ophthalmic adverse effects of lithium. Int
Clin Psychopharmacol. Mar 2020;35(2):69-73.
doi:10.1097/YIC.0000000000000295
843. Schaumberg DA, Buring JE, Sullivan DA, Dana MR. Hormone replacement
therapy and dry eye syndrome. JAMA. Nov 7 2001;286(17):2114-9.
doi:10.1001/jama.286.17.2114
844. Nanavaty MA, Long M, Malhotra R. Transdermal androgen patches in
evaporative dry eye syndrome with androgen deficiency: a pilot study. Br J
Ophthalmol. Apr 2014;98(4):567-9. doi:10.1136/bjophthalmol-2013-304637
845. Schiffman RM, Bradford R, Bunnell B, Lai F, Bernstein P, Whitcup SW. A Multi–
Center, Double–Masked, Randomized, Vehicle–Controlled, Parallel Group
Study to Evaluate the Safety and Efficacy of Testosterone Ophthalmic Solution
in Patients With Meibomian Gland Dysfunction. Investigative Ophthalmology &
Visual Science. 2006;47(13):5608-5608.
846. Morgan PB, Murphy PJ, Gifford KL, et al. BCLA CLEAR - Effect of contact lens
materials and designs on the anatomy and physiology of the eye. Contact Lens
and Anterior Eye. 2021;44(2):192-219. doi:10.1016/j.clae.2021.02.006
847. Stapleton F, Bakkar M, Carnt N, et al. CLEAR - Contact lens complications.
Cont Lens Anterior Eye. Apr 2021;44(2):330-367.
doi:10.1016/j.clae.2021.02.010
848. Acimovic L, Stanojlovic S, Kalezic T, Dacic Krnjaja B. Evaluation of dry eye
symptoms and risk factors among medical students in Serbia. PLoS One.
2022;17(10):e0275624. doi:10.1371/journal.pone.0275624
849. Alamri A, Amer KA, Aldosari AA, et al. Assessment of Dry Eye Syndrome
Among Contact Lens Users in Asir Region, Saudi Arabia. Cureus. Jan
2022;14(1):e21526. doi:10.7759/cureus.21526
200
850. Fan Z, Du Y, Tang C, et al. Awareness, Prevalence, and Knowledge of Dry Eye
Among Internet Professionals: A Cross-sectional Study in China. Eye Contact
Lens. Mar 1 2023;49(3):92-97. doi:10.1097/icl.0000000000000968
851. Supiyaphun C, Jongkhajornpong P, Rattanasiri S, Lekhanont K. Prevalence and
risk factors of dry eye disease among University Students in Bangkok, Thailand.
PLoS One. 2021;16(10):e0258217. doi:10.1371/journal.pone.0258217
852. Schornack M, Nau C, Nau A, Harthan J, Fogt J, Shorter E. Visual and
physiological outcomes of scleral lens wear. Cont Lens Anterior Eye. Feb
2019;42(1):3-8. doi:10.1016/j.clae.2018.07.007
853. Schornack MM, Pyle J, Patel SV. Scleral lenses in the management of ocular
surface disease. Ophthalmology. Jul 2014;121(7):1398-405.
doi:10.1016/j.ophtha.2014.01.028
854. Li W, Sun X, Wang Z, Zhang Y. A survey of contact lens-related complications
in a tertiary hospital in China. Cont Lens Anterior Eye. Apr 2018;41(2):201-204.
doi:10.1016/j.clae.2017.10.007
855. Yang TT, Ma BK, Liu RJ, Qi H. [Research advances in contact lens-associated
dry eye]. Zhonghua Yan Ke Za Zhi. Feb 11 2022;58(2):149-154.
doi:10.3760/cma.j.cn112142-20210525-00256
856. Harbiyeli, II, Bozkurt B, Erdem E, et al. Associations with meibomian gland loss
in soft and rigid contact lens wearers. Cont Lens Anterior Eye. Feb
2022;45(1):101400. doi:10.1016/j.clae.2020.12.005
857. Pucker AD, Jones-Jordan LA, Marx S, et al. Clinical factors associated with
contact lens dropout. Cont Lens Anterior Eye. Jun 2019;42(3):318-324.
doi:10.1016/j.clae.2018.12.002
858. Shiraishi A, Yamaguchi M, Ohashi Y. Prevalence of upper- and lower-lid-wiper
epitheliopathy in contact lens wearers and non-wearers. Eye Contact Lens. Jul
2014;40(4):220-4. doi:10.1097/ICL.0000000000000040
859. McMonnies CW. Incomplete blinking: exposure keratopathy, lid wiper
epitheliopathy, dry eye, refractive surgery, and dry contact lenses. Cont Lens
Anterior Eye. Mar 2007;30(1):37-51. doi:10.1016/j.clae.2006.12.002
860. Muntz A, Subbaraman LN, Craig JP, Jones L. Cytomorphological assessment
of the lid margin in relation to symptoms, contact lens wear and lid wiper
epitheliopathy. Ocul Surf. Apr 2020;18(2):214-220.
doi:10.1016/j.jtos.2019.12.001
861. Pult H, Murphy PJ, Purslow C. A novel method to predict the dry eye symptoms
in new contact lens wearers. Optom Vis Sci. Sep 2009;86(9):E1042-50.
doi:10.1097/OPX.0b013e3181b598cd
862. Kojima T. Contact Lens-Associated Dry Eye Disease: Recent Advances
Worldwide and in Japan. Invest Ophthalmol Vis Sci. Nov 1
2018;59(14):Des102-des108. doi:10.1167/iovs.17-23685
863. [Chinese expert consensus on the diagnosis and treatment of contact lens-
associated dry eye (2024)]. Zhonghua Yan Ke Za Zhi. Feb 11 2024;60(2):120-
126. doi:10.3760/cma.j.cn112142-20231121-00243
864. Yang L, Zhang L, Jian Hu R, Yu PP, Jin X. The influence of overnight
orthokeratology on ocular surface and dry eye-related cytokines IL-17A, IL-6,
201
and PGE2 in children. Cont Lens Anterior Eye. Feb 2021;44(1):81-88.
doi:10.1016/j.clae.2020.04.001
865. Liu Q, Xu Z, Xu Y, et al. Changes in Corneal Dendritic Cell and Sub-basal
Nerve in Long-Term Contact Lens Wearers With Dry Eye. Eye Contact Lens.
Jul 2020;46(4):238-244. doi:10.1097/icl.0000000000000691
866. Lum E, Golebiowski B, Swarbrick HA. Reduced Corneal Sensitivity and Sub-
Basal Nerve Density in Long-Term Orthokeratology Lens Wear. Eye Contact
Lens. Jul 2017;43(4):218-224. doi:10.1097/icl.0000000000000285
867. Nombela-Palomo M, Felipe-Marquez G, Teus MA, Hernandez-Verdejo JL,
Nieto-Bona A. Long-Term Impacts of Orthokeratology Treatment on Sub-Basal
Nerve Plexus and Corneal Sensitivity Responses and Their Reversibility. Eye
Contact Lens. Mar 2018;44(2):91-96. doi:10.1097/icl.0000000000000386
868. Jin W, Ye J, Zhang J, et al. Short-Term Effects of Overnight Orthokeratology on
Corneal Sensitivity in Chinese Children and Adolescents. J Ophthalmol.
2018;2018:6185919. doi:10.1155/2018/6185919
869. Stapleton F, Marfurt C, Golebiowski B, et al. The TFOS International Workshop
on Contact Lens Discomfort: report of the subcommittee on neurobiology. Invest
Ophthalmol Vis Sci. Oct 18 2013;54(11):TFOS71-97. doi:10.1167/iovs.13-
13226
870. Situ P, Begley C, Simpson T. The roles of neural adaptation and sensitization in
contact lens discomfort. Ocul Surf. Oct 2024;34:132-139.
doi:10.1016/j.jtos.2024.07.003
871. Mobeen R, Stapleton F, Chao C, et al. Epithelial Immune Cell Response to
Initial Soft Contact Lens Wear in the Human Corneal and Conjunctival
Epithelium. Invest Ophthalmol Vis Sci. Dec 1 2023;64(15):18.
doi:10.1167/iovs.64.15.18
872. Saliman NH, Morgan PB, MacDonald AS, Maldonado-Codina C. Subclinical
Inflammation of the Ocular Surface in Soft Contact Lens Wear. Cornea. Feb
2020;39(2):146-154. doi:10.1097/ICO.0000000000002192
873. Society of Contact Lens Safety Monitoring & Vision Health of Chinese Health
Association. [Expert consensus on diagnosis and treatment of adverse
reactions of contact lens wear in China (2021)]. Zhonghua Yan Ke Za Zhi. Aug
11 2021;57(8):573-579. doi:10.3760/cma.j.cn112142-20210520-00243
874. Wolffsohn JS, Dumbleton K, Huntjens B, et al. CLEAR - Evidence-based
contact lens practice. Cont Lens Anterior Eye. Apr 2021;44(2):368-397.
doi:10.1016/j.clae.2021.02.008
875. Igarashi T, Kobayashi M, Yaguchi C, Fujimoto C, Suzuki H, Takahashi H.
Efficacy of Rebamipide Instillation for Contact Lens Discomfort With Dry Eye.
Eye Contact Lens. Nov 2018;44 Suppl 2(2):S137-s142.
doi:10.1097/icl.0000000000000438
876. Siddireddy JS, Tan J, Vijay AK, Willcox MDP. The Effect of Microblepharon
Exfoliation on Clinical Correlates of Contact Lens Discomfort. Optom Vis Sci.
Mar 2019;96(3):187-199. doi:10.1097/OPX.0000000000001354
877. Blackie CA, Coleman CA, Nichols KK, et al. A single vectored thermal pulsation
treatment for meibomian gland dysfunction increases mean comfortable contact
202
lens wearing time by approximately 4 hours per day. Clin Ophthalmol.
2018;12:169-183. doi:10.2147/OPTH.S153297
878. Pavan Michielon E, Gheller P, Piñero DP, Stanco L. Contact Lens Discomfort
(CLD) Treatment with MY MASK Light Modulation LED mask. 2024-12-31
doi:10.15626/sjovs.v17i2.4212
879. Stapleton F, Jia T, DePuy V, Bosworth C, Gleeson M, Tan J. The effect of a
biweekly novel selenium sulfide-containing topical treatment in symptomatic
contact lens wearers: An exploratory study. Ocul Surf. Apr 2025;36:190-197.
doi:10.1016/j.jtos.2025.01.015
880. Stapleton F, Hinds M, Tan J, et al. AZR-MD-001 0.5% selenium sulfide
ophthalmic ointment for the treatment of contact lens discomfort: A vehicle-
controlled, randomized, clinical trial. Ocul Surf. Dec 28
2024;doi:10.1016/j.jtos.2024.12.009
881. Choi EW, Yeom DJ, Jang SY. Botulinum Toxin A Injection for the Treatment of
Intractable Dry Eye Disease. Medicina (Kaunas). Mar 8
2021;57(3)doi:10.3390/medicina57030247
882. Ziahosseini K, Al-Abbadi Z, Malhotra R. Botulinum toxin injection for the
treatment of epiphora in lacrimal outflow obstruction. Eye (Lond). May
2015;29(5):656-61. doi:10.1038/eye.2015.18
883. Ho MC, Hsu WC, Hsieh YT. Botulinum toxin type a injection for lateral canthal
rhytids : effect on tear film stability and tear production. JAMA Ophthalmol. Mar
2014;132(3):332-7. doi:10.1001/jamaophthalmol.2013.6243
884. Žiak P, Halička J, Kapitánová K, Mojžiš P. Effect of Botulinum toxin
A application in neuro-ophtalmologic indications on Schirmers test and tears
osmolarity. Cesk Slov Oftalmol. Summer 2019;75(2):74-77. Vplyv aplikácie
botulotoxínu A v neurooftalmologických indikáciách na výsledky Schirmerovho
testu a osmolarity sĺz. doi:10.31348/2019/2/3
885. Kaynak P, Karabulut GO, Ozturker C, et al. Comparison of botulinum toxin-A
injection in lacrimal gland and conjunctivodacryocystorhinostomy for treatment
of epiphora due to proximal lacrimal system obstruction. Eye (Lond). Aug
2016;30(8):1056-62. doi:10.1038/eye.2016.88
886. Alsuhaibani AH, Eid SA. Botulinum toxin injection and tear production. Curr
Opin Ophthalmol. Sep 2018;29(5):428-433. doi:10.1097/icu.0000000000000506
887. Lipham WJ, Tawfik HA, Dutton JJ. A histologic analysis and three-dimensional
reconstruction of the muscle of Riolan. Ophthalmic Plast Reconstr Surg. Mar
2002;18(2):93-8. doi:10.1097/00002341-200203000-00002
888. Ho RW, Fang PC, Chang CH, Liu YP, Kuo MT. A Review of Periocular
Botulinum Neurotoxin on the Tear Film Homeostasis and the Ocular Surface
Change. Toxins (Basel). Jan 24 2019;11(2)doi:10.3390/toxins11020066
889. Ozgur OK, Murariu D, Parsa AA, Parsa FD. Dry eye syndrome due to botulinum
toxin type-A injection: guideline for prevention. Hawaii J Med Public Health. May
2012;71(5):120-3.
890. Wollensak G, Spoerl E, Seiler T. Riboflavin/ultraviolet-a-induced collagen
crosslinking for the treatment of keratoconus. Am J Ophthalmol. May
2003;135(5):620-7. doi:10.1016/s0002-9394(02)02220-1
203
891. Ashwin PT, McDonnell PJ. Collagen cross-linkage: a comprehensive review
and directions for future research. Br J Ophthalmol. Aug 2010;94(8):965-70.
doi:10.1136/bjo.2009.164228
892. Wang X, Bai X, Jin C, Zhao L, Wei R. Analysis of Tear Function Outcomes
following Collagen Cross-Linking Treatment in Ectatic Corneas. J Ophthalmol.
2022;2022:1910607. doi:10.1155/2022/1910607
893. Voronin GV, Bubnova IA, Averich VV, Sarkisova KG. [Precorneal tear film after
corneal collagen cross-linking in keratoconus (preliminary report)]. Vestn
Oftalmol. 2021;137(5. Vyp. 2):224-230. Sostoyanie prekorneal'noi sleznoi plenki
posle krosslinkinga rogovichnogo kollagena pri keratokonuse (predvaritel'noe
soobshchenie). doi:10.17116/oftalma2021137052224
894. Recalde JI, Acera A, Rodríguez-Agirretxe I, Sánchez-Tena MA, San-Cristóbal J,
Durán JA. Ocular Surface Disease Parameters After Collagen Cross-Linking for
Keratoconus. Cornea. Feb 2017;36(2):148-152.
doi:10.1097/ico.0000000000001085
895. Bubnova IA, Surnina ZV, Averich VV, Sarkisova KG. [The effects of corneal
collagen cross-linking on the corneal structure in keratoconus]. Vestn Oftalmol.
2020;136(5. Vyp. 2):268-276. Vliyanie krosslinkinga rogovichnogo kollagena na
strukturu rogovitsy pri keratokonuse. doi:10.17116/oftalma2020136052268
896. Akgöz H, Fındık H, Aslan MG. Evaluation of tear parameters and meibomian
gland morphology in keratoconus patients after epithelial-on corneal cross-
linking. Eur J Ophthalmol. Aug 5 2022:11206721221118740.
doi:10.1177/11206721221118740
897. Smith TM, Suzuki S, Sabat N, Rayner CL, Harkin DG, Chirila TV. Further
Investigations on the Crosslinking of Tarsal Collagen as a Treatment for Eyelid
Laxity: Optimizing the Procedure in Animal Tissue. Ophthalmic Plast Reconstr
Surg. Nov/Dec 2019;35(6):600-603. doi:10.1097/iop.0000000000001413
898. Uysal BS, Akcay E, Kilicarslan A, et al. Tear function and ocular surface
changes following corneal collagen cross-linking treatment in keratoconus
patients: 18-month results. Int Ophthalmol. Jan 2020;40(1):169-177.
doi:10.1007/s10792-019-01161-1
899. Kontadakis GA, Kymionis GD, Kankariya VP, Pallikaris AI. Effect of corneal
collagen cross-linking on corneal innervation, corneal sensitivity, and tear
function of patients with keratoconus. Ophthalmology. May 2013;120(5):917-22.
doi:10.1016/j.ophtha.2012.10.012
900. Mazzotta C, Traversi C, Baiocchi S, et al. Corneal healing after riboflavin
ultraviolet-A collagen cross-linking determined by confocal laser scanning
microscopy in vivo: early and late modifications. Am J Ophthalmol. Oct
2008;146(4):527-533. doi:10.1016/j.ajo.2008.05.042
901. Bubnova IA, Sarkisova KG. [Changes in precorneal tear film and ocular surface
in keratoconic patients after corneal collagen cross-linking]. Vestn Oftalmol.
2022;138(3):118-123. Izmenenie sostoyaniya prerogovichnoi sleznoi plenki i
glaznoi poverkhnosti posle krosslinkinga rogovichnogo kollagena pri
keratokonuse. doi:10.17116/oftalma2022138031118
204
902. El Ameen A, Majzoub S, Vandermeer G, Pisella PJ. Influence of cataract
surgery on Meibomian gland dysfunction. J Fr Ophtalmol. May
2018;41(5):e173-e180. doi:10.1016/j.jfo.2018.03.001
903. Liu L, Fukuyama T, Han T, et al. Observation and assessment of the immediate
use of a silicon hydrogel contact lens after transepithelial corneal cross linking:
a prospective study. BMC Ophthalmol. Nov 20 2023;23(1):471.
doi:10.1186/s12886-023-03217-4
904. Ondes Yilmaz F, Kepez Yildiz B, Tunc U, Kandemir Besek N, Yildirim Y,
Demirok A. Comparison of topical omega-3 fatty acids with topical sodium
hyaluronate after corneal crosslinking : Short term results. Ocul Immunol
Inflamm. May 19 2022;30(4):959-965. doi:10.1080/09273948.2020.1858117
905. Bikbov MM, Khalimov AR, Usubov EL. [Ultraviolet Corneal Crosslinking]. Vestn
Ross Akad Med Nauk. 2016;71(3):224-32. doi:10.15690/vramn562
906. Knutsson KA, Genovese PN, Paganoni G, et al. Evaluation of a Post-Operative
Therapy Protocol after Epithelium-Off Corneal Cross-Linking in Patients
Affected by Keratoconus. J Clin Med. Nov 30
2022;11(23)doi:10.3390/jcm11237093
907. Recalde JI, Duran JA, Rodriguez-Agirretxe I, et al. Changes in tear biomarker
levels in keratoconus after corneal collagen crosslinking. Mol Vis. 2019;25:12-
21.
908. Jeganathan VS, Wirth A, MacManus MP. Ocular risks from orbital and
periorbital radiation therapy: a critical review. Int J Radiat Oncol Biol Phys. Mar
1 2011;79(3):650-9. doi:10.1016/j.ijrobp.2010.09.056
909. Bhandare N, Moiseenko V, Song WY, Morris CG, Bhatti MT, Mendenhall WM.
Severe dry eye syndrome after radiotherapy for head-and-neck tumors. Int J
Radiat Oncol Biol Phys. Mar 15 2012;82(4):1501-8.
doi:10.1016/j.ijrobp.2011.05.026
910. Durkin SR, Roos D, Higgs B, Casson RJ, Selva D. Ophthalmic and adnexal
complications of radiotherapy. Acta Ophthalmol Scand. May 2007;85(3):240-50.
doi:10.1111/j.1600-0420.2006.00822.x
911. Gordon KB, Char DH, Sagerman RH. Late effects of radiation on the eye and
ocular adnexa. Int J Radiat Oncol Biol Phys. Mar 30 1995;31(5):1123-39.
doi:10.1016/0360-3016(95)00062-4
912. Soni M, Walia S, Jain P. Dry eye disease in head and neck cancer patients
undergoing radiotherapy. Indian J Ophthalmol. Apr 2023;71(4):1556-1560.
doi:10.4103/ijo.Ijo_2673_22
913. Thariat J, Maschi C, Lanteri S, et al. Dry Eye Syndrome After Proton Therapy of
Ocular Melanomas. Int J Radiat Oncol Biol Phys. May 1 2017;98(1):142-151.
doi:10.1016/j.ijrobp.2017.01.199
914. Parsons JT, Bova FJ, Fitzgerald CR, Mendenhall WM, Million RR. Severe dry-
eye syndrome following external beam irradiation. Int J Radiat Oncol Biol Phys.
Nov 15 1994;30(4):775-80. doi:10.1016/0360-3016(94)90348-4
915. Bessell EM, Henk JM, Whitelocke RA, Wright JE. Ocular morbidity after
radiotherapy of orbital and conjunctival lymphoma. Eye (Lond). 1987;1 ( Pt
1):90-6. doi:10.1038/eye.1987.14
205
916. Groenewald C, Konstantinidis L, Damato B. Effects of radiotherapy on uveal
melanomas and adjacent tissues. Eye (Lond). Feb 2013;27(2):163-71.
doi:10.1038/eye.2012.249
917. Konstantinidis L, Roberts D, Errington RD, Kacperek A, Heimann H, Damato B.
Transpalpebral proton beam radiotherapy of choroidal melanoma. Br J
Ophthalmol. Feb 2015;99(2):232-5. doi:10.1136/bjophthalmol-2014-305313
918. Tiwari S, Bhatt A, Nagamodi J, et al. Aqueous Deficient Dry Eye Syndrome Post
Orbital Radiotherapy: A 10-Year Retrospective Study. Transl Vis Sci Technol.
Jun 2017;6(3):19. doi:10.1167/tvst.6.3.19
919. Sia S, Harper C, McAllister I, Perry A. Iodine-I25 episcleral plaque therapy in
uveal melanoma. Clin Exp Ophthalmol. Dec 2000;28(6):409-13.
doi:10.1046/j.1442-9071.2000.00354.x
920. Karp LA, Streeten BW, Cogan DG. Radiation-induced atrophy of the Meibomian
gland. Arch Ophthalmol. Feb 1979;97(2):303-5.
doi:10.1001/archopht.1979.01020010155013
921. Parsons JT, Bova FJ, Mendenhall WM, Million RR, Fitzgerald CR. Response of
the normal eye to high dose radiotherapy. Oncology (Williston Park). Jun
1996;10(6):837-47; discussion 847-8, 851-2.
922. Chen X, Badian RA, Hynne H, et al. Alterations in meibomian glands in patients
treated with intensity-modulated radiotherapy for head and neck cancer. Sci
Rep. Nov 17 2021;11(1):22419. doi:10.1038/s41598-021-01844-9
923. Ferrara JL, Levine JE, Reddy P, Holler E. Graft-versus-host disease. Lancet.
May 2 2009;373(9674):1550-61. doi:10.1016/s0140-6736(09)60237-3
924. Franklin RM, Kenyon KR, Tutschka PJ, Saral R, Green WR, Santos GW. Ocular
manifestations of graft-vs-host disease. Ophthalmology. Jan 1983;90(1):4-13.
doi:10.1016/s0161-6420(83)34604-2
925. Hirst LW, Jabs DA, Tutschka PJ, Green WR, Santos GW. The eye in bone
marrow transplantation. I. Clinical study. Arch Ophthalmol. Apr
1983;101(4):580-4. doi:10.1001/archopht.1983.01040010580010
926. Westeneng AC, Hettinga Y, Lokhorst H, Verdonck L, van Dorp S, Rothova A.
Ocular graft-versus-host disease after allogeneic stem cell transplantation.
Cornea. Jul 2010;29(7):758-63. doi:10.1097/ICO.0b013e3181ca321c
927. Ogawa Y, Yamazaki K, Kuwana M, et al. A significant role of stromal fibroblasts
in rapidly progressive dry eye in patients with chronic GVHD. Invest Ophthalmol
Vis Sci. Jan 2001;42(1):111-9.
928. Ogawa Y, Kuwana M, Yamazaki K, et al. Periductal area as the primary site for
T-cell activation in lacrimal gland chronic graft-versus-host disease. Invest
Ophthalmol Vis Sci. May 2003;44(5):1888-96. doi:10.1167/iovs.02-0699
929. Yaguchi S, Ogawa Y, Shimmura S, et al. Presence and physiologic function of
the renin-angiotensin system in mouse lacrimal gland. Invest Ophthalmol Vis
Sci. Aug 13 2012;53(9):5416-25. doi:10.1167/iovs.12-9891
930. Yaguchi S, Ogawa Y, Kawakita T, Shimmura S, Tsubota K. Tissue Renin-
Angiotensin System in Lacrimal Gland Fibrosis in a Murine Model of Chronic
Graft-Versus-Host Disease. Cornea. Nov 2015;34 Suppl 11:S142-52.
doi:10.1097/ico.0000000000000586
206
931. Kheirkhah A, Rahimi Darabad R, Cruzat A, et al. Corneal Epithelial Immune
Dendritic Cell Alterations in Subtypes of Dry Eye Disease: A Pilot In Vivo
Confocal Microscopic Study. Invest Ophthalmol Vis Sci. Nov 2015;56(12):7179-
85. doi:10.1167/iovs.15-17433
932. Chatterjee D. A brief account on ocular graft versus host disease. Indian J
Ophthalmol. Apr 2023;71(4):1115-1122. doi:10.4103/ijo.Ijo_2839_22
933. Yamane M, Sato S, Shimizu E, et al. Senescence-associated secretory
phenotype promotes chronic ocular graft-vs-host disease in mice and humans.
Faseb j. Aug 2020;34(8):10778-10800. doi:10.1096/fj.201900218R
934. Rodier F, Coppé JP, Patil CK, et al. Persistent DNA damage signalling triggers
senescence-associated inflammatory cytokine secretion. Nat Cell Biol. Aug
2009;11(8):973-9. doi:10.1038/ncb1909
935. Yang F, Hayashi I, Sato S, et al. Eyelid blood vessel and meibomian gland
changes in a sclerodermatous chronic GVHD mouse model. Ocul Surf. Oct
2022;26:328-341. doi:10.1016/j.jtos.2021.10.006
936. Ban Y, Ogawa Y, Ibrahim OM, et al. Morphologic evaluation of meibomian
glands in chronic graft-versus-host disease using in vivo laser confocal
microscopy. Mol Vis. 2011;17:2533-43.
937. Sinha S, Singh RB, Dohlman TH, Taketani Y, Yin J, Dana R. Prevalence and
Risk Factors Associated With Corneal Perforation in Chronic Ocular Graft-
Versus-Host-Disease. Cornea. Jul 1 2021;40(7):877-882.
doi:10.1097/ico.0000000000002526
938. Lee GA, Chen SX. Autologous serum in the management of recalcitrant dry eye
syndrome. Clin Exp Ophthalmol. Mar 2008;36(2):119-22. doi:10.1111/j.1442-
9071.2008.01680.x
939. Wolff D, Gerbitz A, Ayuk F, et al. Consensus conference on clinical practice in
chronic graft-versus-host disease (GVHD): first-line and topical treatment of
chronic GVHD. Biol Blood Marrow Transplant. Dec 2010;16(12):1611-28.
doi:10.1016/j.bbmt.2010.06.015
940. Dausch D, Lee S, Dausch S, Kim JC, Schwert G, Michelson W. [Comparative
study of treatment of the dry eye syndrome due to disturbances of the tear film
lipid layer with lipid-containing tear substitutes]. Klin Monbl Augenheilkd. Dec
2006;223(12):974-83. Vergleichende Studie zur Therapie des Trockenen Auges
bedingt durch Lipidphasenstörungen mit lipidhaltigen Tränenpräparaten.
doi:10.1055/s-2006-927266
941. Pflugfelder SC. Antiinflammatory therapy for dry eye. Am J Ophthalmol. Feb
2004;137(2):337-42. doi:10.1016/j.ajo.2003.10.036
942. Sabbe LJ, Van De Merwe D, Schouls L, Bergmans A, Vaneechoutte M,
Vandamme P. Clinical spectrum of infections due to the newly described
Actinomyces species A. turicensis, A. radingae, and A. europaeus. J Clin
Microbiol. Jan 1999;37(1):8-13. doi:10.1128/jcm.37.1.8-13.1999
943. Ogawa Y, Okamoto S, Kuwana M, et al. Successful treatment of dry eye in two
patients with chronic graft-versus-host disease with systemic administration of
FK506 and corticosteroids. Cornea. May 2001;20(4):430-4.
doi:10.1097/00003226-200105000-00020
207
944. Noble BA, Loh RS, MacLennan S, et al. Comparison of autologous serum eye
drops with conventional therapy in a randomised controlled crossover trial for
ocular surface disease. Br J Ophthalmol. May 2004;88(5):647-52.
doi:10.1136/bjo.2003.026211
945. Ogawa Y, Okamoto S, Mori T, et al. Autologous serum eye drops for the
treatment of severe dry eye in patients with chronic graft-versus-host disease.
Bone Marrow Transplant. Apr 2003;31(7):579-83. doi:10.1038/sj.bmt.1703862
946. Pezzotta S, Del Fante C, Scudeller L, et al. Long-term safety and efficacy of
autologous platelet lysate drops for treatment of ocular GvHD. Bone Marrow
Transplant. Jan 2017;52(1):101-106. doi:10.1038/bmt.2016.221
947. Tarnawska D, Wylegała E. Corneal grafting and aggressive medication for
corneal defects in graft-versus-host disease following bone marrow
transplantation. Eye (Lond). Dec 2007;21(12):1493-500.
doi:10.1038/sj.eye.6702589
948. ICH. ICH Guidelines. https://www.ich.org/page/ich-guidelines
949. Dry Eye: Developing Drugs for Treatment Guidance for Industry (Center for
Drug Evaluation and Research,) (2020).
950. Cui D, Saldanha IJ, Li G, Mathews PM, Lin MX, Akpek EK. United States
Regulatory Approval of Topical Treatments for Dry Eye. Am J Ophthalmol. Feb
2024;258:14-21. doi:10.1016/j.ajo.2023.09.024
951. Akpek EK, Wirta DL, Downing JE, et al. Efficacy and Safety of a Water-Free
Topical Cyclosporine, 0.1%, Solution for the Treatment of Moderate to Severe
Dry Eye Disease: The ESSENCE-2 Randomized Clinical Trial. JAMA
Ophthalmol. May 1 2023;141(5):459-466.
doi:10.1001/jamaophthalmol.2023.0709
952. Kinoshita S, Oshiden K, Awamura S, et al. A randomized, multicenter phase 3
study comparing 2% rebamipide (OPC-12759) with 0.1% sodium hyaluronate in
the treatment of dry eye. Ophthalmology. Jun 2013;120(6):1158-65.
doi:10.1016/j.ophtha.2012.12.022
953. Novack GD, Asbell P, Barabino S, et al. TFOS DEWS II Clinical Trial Design
Report. Ocul Surf. Jul 2017;15(3):629-649. doi:10.1016/j.jtos.2017.05.009
954. Wirta D, Torkildsen GL, Boehmer B, et al. ONSET-1 Phase 2b Randomized
Trial to Evaluate the Safety and Efficacy of OC-01 (Varenicline Solution) Nasal
Spray on Signs and Symptoms of Dry Eye Disease. Cornea. Oct 1
2022;41(10):1207-1216. doi:10.1097/ICO.0000000000002941
955. Korenfeld M, Nichols KK, Goldberg D, et al. Safety of KPI-121 Ophthalmic
Suspension 0.25% in Patients With Dry Eye Disease: A Pooled Analysis of 4
Multicenter, Randomized, Vehicle-Controlled Studies. Cornea. May 1
2021;40(5):564-570. doi:10.1097/ICO.0000000000002452
956. Tauber J, Schechter BA, Bacharach J, et al. A Phase II/III, randomized, double-
masked, vehicle-controlled, dose-ranging study of the safety and efficacy of
OTX-101 in the treatment of dry eye disease. Clin Ophthalmol. 2018;12:1921-
1929. doi:10.2147/OPTH.S175065
957. Goldberg DF, Malhotra RP, Schechter BA, Justice A, Weiss SL, Sheppard JD.
A Phase 3, Randomized, Double-Masked Study of OTX-101 Ophthalmic
208
Solution 0.09% in the Treatment of Dry Eye Disease. Ophthalmology. Sep
2019;126(9):1230-1237. doi:10.1016/j.ophtha.2019.03.050
958. Takamura E, Tsubota K, Watanabe H, Ohashi Y, Diquafosol Ophthalmic
Solution Phase 3 Study G. A randomised, double-masked comparison study of
diquafosol versus sodium hyaluronate ophthalmic solutions in dry eye patients.
Br J Ophthalmol. Oct 2012;96(10):1310-5. doi:10.1136/bjophthalmol-2011-
301448
959. White DE, Zhao Y, Ogundele A, et al. Real-World Treatment Patterns Of
Cyclosporine Ophthalmic Emulsion And Lifitegrast Ophthalmic Solution Among
Patients With Dry Eye. Clin Ophthalmol. 2019;13:2285-2292.
doi:10.2147/OPTH.S226168
960. Mah F, Milner M, Yiu S, Donnenfeld E, Conway TM, Hollander DA. PERSIST:
Physician's Evaluation of Restasis((R)) Satisfaction in Second Trial of topical
cyclosporine ophthalmic emulsion 0.05% for dry eye: a retrospective review.
Clin Ophthalmol. 2012;6:1971-6. doi:10.2147/OPTH.S30261
961. DEWS Clinical Trials. Design and conduct of clinical trials: report of the Clinical
Trials Subcommittee of the International Dry Eye WorkShop (2007). Ocul Surf.
Apr 2007;5(2):153-62. doi:10.1016/s1542-0124(12)70084-8
962. Holland EJ, Luchs J, Karpecki PM, et al. Lifitegrast for the Treatment of Dry Eye
Disease: Results of a Phase III, Randomized, Double-Masked, Placebo-
Controlled Trial (OPUS-3). Ophthalmology. Jan 2017;124(1):53-60.
doi:10.1016/j.ophtha.2016.09.025
963. Sall K, Stevenson OD, Mundorf TK, Reis BL. Two multicenter, randomized
studies of the efficacy and safety of cyclosporine ophthalmic emulsion in
moderate to severe dry eye disease. CsA Phase 3 Study Group.
Ophthalmology. Apr 2000;107(4):631-9. doi:10.1016/s0161-6420(99)00176-1
964. Semba CP, Torkildsen GL, Lonsdale JD, et al. A phase 2 randomized, double-
masked, placebo-controlled study of a novel integrin antagonist (SAR 1118) for
the treatment of dry eye. Am J Ophthalmol. Jun 2012;153(6):1050-60 e1.
doi:10.1016/j.ajo.2011.11.003
965. Sheppard JD, Torkildsen GL, Lonsdale JD, et al. Lifitegrast ophthalmic solution
5.0% for treatment of dry eye disease: results of the OPUS-1 phase 3 study.
Ophthalmology. Feb 2014;121(2):475-83. doi:10.1016/j.ophtha.2013.09.015
966. Tauber J, Karpecki P, Latkany R, et al. Lifitegrast Ophthalmic Solution 5.0%
versus Placebo for Treatment of Dry Eye Disease: Results of the Randomized
Phase III OPUS-2 Study. Ophthalmology. Dec 2015;122(12):2423-31.
doi:10.1016/j.ophtha.2015.08.001
967. Morgan P, Van Der Graaf PH, Arrowsmith J, et al. Can the flow of medicines be
improved? Fundamental pharmacokinetic and pharmacological principles
toward improving Phase II survival. Drug Discov Today. May 2012;17(9-
10):419-24. doi:10.1016/j.drudis.2011.12.020
968. Craig JP, Nichols KK, Akpek EK, et al. TFOS DEWS II Definition and
Classification Report. Ocul Surf. Jul 2017;15(3):276-283.
doi:10.1016/j.jtos.2017.05.008
209
969. Asbell PA, Stapleton FJ, Wickstrom K, et al. The international workshop on
meibomian gland dysfunction: report of the clinical trials subcommittee. Invest
Ophthalmol Vis Sci. Mar 2011;52(4):2065-85. doi:10.1167/iovs.10-6997h
970. Foulks G, Chalmers R, Keir N, et al. The TFOS International Workshop on
Contact Lens Discomfort: report of the subcommittee on clinical trial design and
outcomes. Invest Ophthalmol Vis Sci. Oct 18 2013;54(11):TFOS157-83.
doi:10.1167/iovs.13-13189
971. Wirta DL, Torkildsen GL, Moreira HR, et al. A Clinical Phase II Study to Assess
Efficacy, Safety, and Tolerability of Waterfree Cyclosporine Formulation for
Treatment of Dry Eye Disease. Ophthalmology. Jun 2019;126(6):792-800.
doi:10.1016/j.ophtha.2019.01.024
972. Sheppard JD, Wirta DL, McLaurin E, et al. A Water-free 0.1% Cyclosporine A
Solution for Treatment of Dry Eye Disease: Results of the Randomized Phase
2B/3 ESSENCE Study. Cornea. Oct 1 2021;40(10):1290-1297.
doi:10.1097/ICO.0000000000002633
973. Sheppard JD, Nichols KK. Dry Eye Disease Associated with Meibomian Gland
Dysfunction: Focus on Tear Film Characteristics and the Therapeutic
Landscape. Ophthalmol Ther. Jun 2023;12(3):1397-1418. doi:10.1007/s40123-
023-00669-1
974. Tauber J, Berdy GJ, Wirta DL, Krosser S, Vittitow JL, Group GS. NOV03 for Dry
Eye Disease Associated with Meibomian Gland Dysfunction: Results of the
Randomized Phase 3 GOBI Study. Ophthalmology. May 2023;130(5):516-524.
doi:10.1016/j.ophtha.2022.12.021
975. Watson SL, Jones LW, Stapleton F, et al. Efficacy and safety of AZR-MD-001
selenium sulfide ophthalmic ointment in adults with meibomian gland
dysfunction: A vehicle-controlled, randomized clinical trial. Ocul Surf. Jul
2023;29:537-546. doi:10.1016/j.jtos.2023.07.002
976. Fahmy AM, Harthan JS, Evans DG, et al. Perfluorohexyloctane ophthalmic
solution for dry eye disease: pooled analysis of two phase 3 clinical trials. Front
Ophthalmol (Lausanne). 2024;4:1452422. doi:10.3389/fopht.2024.1452422
977. Protzko EE, Segal BA, Korenfeld MS, Krosser S, Vittitow JL. Long-Term Safety
and Efficacy of Perfluorohexyloctane Ophthalmic Solution for the Treatment of
Patients With Dry Eye Disease: The KALAHARI Study. Cornea. Sep 1
2024;43(9):1100-1107. doi:10.1097/ICO.0000000000003418
978. Downie LE, Craig JP, Stapleton F, et al. Efficacy and safety of AZR-MD-001
selenium sulfide ophthalmic ointment in adults with meibomian gland
dysfunction over six months of treatment: A Phase 2, vehicle-controlled,
randomized extension trial. Ocul Surf. Jan 2025;35:15-24.
doi:10.1016/j.jtos.2024.11.008
979. Wirta D, Vollmer P, Paauw J, et al. Efficacy and Safety of OC-01 (Varenicline
Solution) Nasal Spray on Signs and Symptoms of Dry Eye Disease: The
ONSET-2 Phase 3 Randomized Trial. Ophthalmology. Apr 2022;129(4):379-
387. doi:10.1016/j.ophtha.2021.11.004
980. Hynnekleiv L, Magno M, Vernhardsdottir RR, et al. Hyaluronic acid in the
treatment of dry eye disease. Acta Ophthalmol. Dec 2022;100(8):844-860.
doi:10.1111/aos.15159
210
981. Watanabe H. Medical Treatment for Dry Eye in Japan. Invest Ophthalmol Vis
Sci. Nov 1 2018;59(14):DES116-DES120. doi:10.1167/iovs.18-24130
982. Greaves MW. Anti-inflammatory action of corticosteroids. Postgrad Med J. Oct
1976;52(612):631-3. doi:10.1136/pgmj.52.612.631
983. Coutinho AE, Chapman KE. The anti-inflammatory and immunosuppressive
effects of glucocorticoids, recent developments and mechanistic insights. Mol
Cell Endocrinol. Mar 15 2011;335(1):2-13. doi:10.1016/j.mce.2010.04.005
984. Tomlinson A, Bron AJ, Korb DR, et al. The international workshop on
meibomian gland dysfunction: report of the diagnosis subcommittee. Invest
Ophthalmol Vis Sci. Mar 2011;52(4):2006-49. doi:10.1167/iovs.10-6997f
985. Geerling G, Tauber J, Baudouin C, et al. The international workshop on
meibomian gland dysfunction: report of the subcommittee on management and
treatment of meibomian gland dysfunction. Invest Ophthalmol Vis Sci. Mar 30
2011;52(4):2050-64. doi:10.1167/iovs.10-6997g
986. Kent C. Devices for Treating The Meibomian Glands. Review of
Ophthalmology2018.
987. Valencia-Nieto L, Novo-Diez A, Blanco-Vazquez M, Lopez-Miguel A.
Therapeutic Instruments Targeting Meibomian Gland Dysfunction. Ophthalmol
Ther. Dec 2020;9(4):797-807. doi:10.1007/s40123-020-00304-3
988. Tauber J, Owen J, Bloomenstein M, Hovanesian J, Bullimore MA. Comparison
of the iLUX and the LipiFlow for the Treatment of Meibomian Gland Dysfunction
and Symptoms: A Randomized Clinical Trial. Clin Ophthalmol. 2020;14:405-
418. doi:10.2147/OPTH.S234008
989. Li S, Yang K, Wang J, et al. Effect of a Novel Thermostatic Device on
Meibomian Gland Dysfunction: A Randomized Controlled Trial in Chinese
Patients. Ophthalmol Ther. Feb 2022;11(1):261-270. doi:10.1007/s40123-021-
00431-5
990. Laufenbock C. [Thermal pulsation system (LipiFlow(R)) for treatment of
meibomian gland dysfunction (MGD) from the perspective of an ophthalmologist
in private practice]. Ophthalmologie. Jun 2022;119(6):605-610. Thermo-
Pulsations-System (LipiFLow(R)) zur Behandlung der Meibom-Drusen-
Dysfunktion (MDD) aus der Sicht eines niedergelassenen Augenfacharztes.
doi:10.1007/s00347-021-01541-y
991. Finis D, Konig C, Hayajneh J, Borrelli M, Schrader S, Geerling G. Six-month
effects of a thermodynamic treatment for MGD and implications of meibomian
gland atrophy. Cornea. Dec 2014;33(12):1265-70.
doi:10.1097/ICO.0000000000000273
992. Ballesteros-Sanchez A, Gargallo-Martinez B, Gutierrez-Ortega R, Sanchez-
Gonzalez JM. Eyelid Exfoliation Treatment Efficacy and Safety in Dry Eye
Disease, Blepharitis, and Contact Lens Discomfort Patients: A Systematic
Review. Asia Pac J Ophthalmol (Phila). May-Jun 01 2023;12(3):315-325.
doi:10.1097/APO.0000000000000607
993. Badawi D. A novel system, TearCare((R)), for the treatment of the signs and
symptoms of dry eye disease. Clin Ophthalmol. 2018;12:683-694.
doi:10.2147/OPTH.S160403
211
994. Lane SS, DuBiner HB, Epstein RJ, et al. A new system, the LipiFlow, for the
treatment of meibomian gland dysfunction. Cornea. Apr 2012;31(4):396-404.
doi:10.1097/ICO.0b013e318239aaea
995. Galperin G, Berra M, Marquez MI, Mandaradoni M, Tau J, Berra A. Impact of
environmental pollution on the ocular surface of Sjogren's syndrome patients.
Arq Bras Oftalmol. Nov./Dec. 2018;81(6):481-489. doi:10.5935/0004-
2749.20180091
996. Gonzalez-Garcia MJ, Gonzalez-Saiz A, de la Fuente B, et al. Exposure to a
controlled adverse environment impairs the ocular surface of subjects with
minimally symptomatic dry eye. Invest Ophthalmol Vis Sci. Sep
2007;48(9):4026-32. doi:10.1167/iovs.06-0817
997. Youssouf H, Liousse C, Roblou L, et al. Non-accidental health impacts of
wildfire smoke. Int J Environ Res Public Health. Nov 14 2014;11(11):11772-804.
doi:10.3390/ijerph111111772
998. Calonge M, Labetoulle M, Messmer EM, et al. Controlled Adverse Environment
Chambers in Dry Eye Research. Curr Eye Res. Apr 2018;43(4):445-450.
doi:10.1080/02713683.2017.1420197
999. Ousler GW, 3rd, Rimmer D, Smith LM, Abelson MB. Use of the Controlled
Adverse Environment (CAE) in Clinical Research: A Review. Ophthalmol Ther.
Dec 2017;6(2):263-276. doi:10.1007/s40123-017-0110-x
1000. Meerovitch K, Torkildsen G, Lonsdale J, et al. Safety and efficacy of MIM-D3
ophthalmic solutions in a randomized, placebo-controlled Phase 2 clinical trial in
patients with dry eye. Clin Ophthalmol. 2013;7:1275-85.
doi:10.2147/OPTH.S44688
1001. Liew SH, Nichols KK, Klamerus KJ, Li JZ, Zhang M, Foulks GN. Tofacitinib (CP-
690,550), a Janus kinase inhibitor for dry eye disease: results from a phase 1/2
trial. Ophthalmology. Jul 2012;119(7):1328-35.
doi:10.1016/j.ophtha.2012.01.028
1002. Montecchi-Palmer M, Wu M, Rolando M, Lau C, Perez Quinones VL, Dana R.
Possible Strategies to Mitigate Placebo or Vehicle Response in Dry Eye
Disease Trials: A Narrative Review. Ophthalmol Ther. Aug 2023;12(4):1827-
1849. doi:10.1007/s40123-023-00720-1
1003. Stapleton F, Velez FG, Lau C, Wolffsohn JS. Dry eye disease in the young: A
narrative review. Ocul Surf. Jan 2024;31:11-20. doi:10.1016/j.jtos.2023.12.001
1004. Wickström K. Dry Eyes – Regulatory perspectives. 2011.
1005. Withdrawal of application for the marketing authorisation of Xiidra (lifitegrast)
(2020).
1006. Ikervis (2023).
1007. Baudouin C, de la Maza MS, Amrane M, et al. One-Year Efficacy and Safety of
0.1% Cyclosporine a Cationic Emulsion in the Treatment of Severe Dry Eye
Disease. Eur J Ophthalmol. Nov 8 2017;27(6):678-685.
doi:10.5301/ejo.5001002
1008. Baudouin C, Figueiredo FC, Messmer EM, et al. A randomized study of the
efficacy and safety of 0.1% cyclosporine A cationic emulsion in treatment of
moderate to severe dry eye. Eur J Ophthalmol. Aug 30 2017;27(5):520-530.
doi:10.5301/EJO.5000952
212
1009. Chapin M, Matsuda H, Kawaba T. Considerations for Development in Japan.
Review of Ophthalmology2017.
1010. [Basic principles for conducting phase 1 studies in Japanese prior to initiating
multi-regional clinical trials including Japan for drugs in which early clinical
development is preceding outside Japan] (Ministry of Health, Labour and
Welfare ) (2023).
1011. [Q&A for basic principles for conducting phase 1 studies in Japanese prior to
initiating multi-regional clinical trials including Japan for drugs in which early
clinical development is preceding outside Japan] (Ministry of Health, Labour and
Welfare ) (2023).
1012. Keating GM. Diquafosol ophthalmic solution 3 %: a review of its use in dry eye.
Drugs. May 2015;75(8):911-22. doi:10.1007/s40265-015-0409-7
1013. Matsumoto Y, Ohashi Y, Watanabe H, Tsubota K, Diquafosol Ophthalmic
Solution Phase 2 Study G. Efficacy and safety of diquafosol ophthalmic solution
in patients with dry eye syndrome: a Japanese phase 2 clinical trial.
Ophthalmology. Oct 2012;119(10):1954-60. doi:10.1016/j.ophtha.2012.04.010
1014. Yokoi N, Kato H, Kinoshita S. Facilitation of tear fluid secretion by 3%
diquafosol ophthalmic solution in normal human eyes. Am J Ophthalmol. Jan
2014;157(1):85-92 e1. doi:10.1016/j.ajo.2013.09.009
1015. Kinoshita S, Awamura S, Oshiden K, et al. Rebamipide (OPC-12759) in the
treatment of dry eye: a randomized, double-masked, multicenter, placebo-
controlled phase II study. Ophthalmology. Dec 2012;119(12):2471-8.
doi:10.1016/j.ophtha.2012.06.052
1016. Hori Y, Oka K, Inai M. Efficacy and Safety of the Long-Acting Diquafosol
Ophthalmic Solution DE-089C in Patients with Dry Eye: A Randomized, Double-
Masked, Placebo-Controlled Phase 3 Study. Adv Ther. Aug 2022;39(8):3654-
3667. doi:10.1007/s12325-022-02194-2
1017. Arita R, Fukuoka S, Kaido M. Tolerability of Diquas LX on tear film and
meibomian glands findings in a real clinical scenario. PLoS One.
2024;19(9):e0305020. doi:10.1371/journal.pone.0305020
1018. Fukuoka S, Arita R. Tear film lipid layer increase after diquafosol instillation in
dry eye patients with meibomian gland dysfunction: a randomized clinical study.
Sci Rep. Jun 24 2019;9(1):9091. doi:10.1038/s41598-019-45475-7
1019. Shimazaki J. Definition and Diagnostic Criteria of Dry Eye Disease: Historical
Overview and Future Directions. Invest Ophthalmol Vis Sci. Nov 1
2018;59(14):DES7-DES12. doi:10.1167/iovs.17-23475
1020. Novack GD. The development of drugs vs devices. Ocul Surf. Jan 2011;9(1):56-
7. doi:10.1016/s1542-0124(11)70010-6
1021. Van Norman GA. Drugs, Devices, and the FDA: Part 2: An Overview of
Approval Processes: FDA Approval of Medical Devices. JACC Basic Transl Sci.
Jun 2016;1(4):277-287. doi:10.1016/j.jacbts.2016.03.009
1022. Van Norman GA. Drugs, Devices, and the FDA: Part 1: An Overview of
Approval Processes for Drugs. JACC Basic Transl Sci. Apr 2016;1(3):170-179.
doi:10.1016/j.jacbts.2016.03.002
213
1023. Konishi A, Isobe S, Sato D. New Regulatory Framework for Medical Devices in
Japan: Current Regulatory Considerations Regarding Clinical Studies. J Vasc
Interv Radiol. May 2018;29(5):657-660. doi:10.1016/j.jvir.2017.12.022
1024. Kathuria A, Shamloo K, Jhanji V, Sharma A. Categorization of Marketed
Artificial Tear Formulations Based on Their Ingredients: A Rational Approach for
Their Use. J Clin Med. Mar 21 2021;10(6)doi:10.3390/jcm10061289
1025. Ren Y, Chen J, Zheng Q, Chen W. Short-term effect of a developed warming
moist chamber goggle for video display terminal-associated dry eye. BMC
Ophthalmol. Feb 7 2018;18(1):33. doi:10.1186/s12886-018-0700-y
1026. Roy NS, Wei Y, Kuklinski E, Asbell PA. The Growing Need for Validated
Biomarkers and Endpoints for Dry Eye Clinical Research. Invest Ophthalmol Vis
Sci. May 1 2017;58(6):BIO1-BIO19. doi:10.1167/iovs.17-21709
1027. Lanza NL, Valenzuela F, Perez VL, Galor A. The Matrix Metalloproteinase 9
Point-of-Care Test in Dry Eye. Ocul Surf. Apr 2016;14(2):189-95.
doi:10.1016/j.jtos.2015.10.004
1028. Leonardi A, Messmer EM, Labetoulle M, et al. Efficacy and safety of 0.1%
ciclosporin A cationic emulsion in dry eye disease: a pooled analysis of two
double-masked, randomised, vehicle-controlled phase III clinical studies. Br J
Ophthalmol. Jan 2019;103(1):125-131. doi:10.1136/bjophthalmol-2017-311801
1029. Donnenfeld E, Baudouin C, Galor A, Weissgerber G, He Y, Perez VL.
Pharmacogenomic Analysis of Response to Topical Tumor Necrosis Factor
alpha Antagonist Licaminlimab (OCS-02) in Dry Eye Disease [RETRACTED].
Cornea. Oct 1 2024;43(10):e36-e42. doi:10.1097/ICO.0000000000003510
214