You are on page 1of 29

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

net/publication/350398362

CLEAR - Effect of contact lens materials and designs on the anatomy and
physiology of the eye

Article in Contact Lens and Anterior Eye · April 2021


DOI: 10.1016/j.clae.2021.02.006

CITATIONS READS

29 2,344

14 authors, including:

Philip Morgan Paul J Murphy


The University of Manchester University of Waterloo
358 PUBLICATIONS 8,218 CITATIONS 139 PUBLICATIONS 3,678 CITATIONS

SEE PROFILE SEE PROFILE

Paul Gifford Blanka Golebiowski


UNSW Sydney UNSW Sydney
37 PUBLICATIONS 664 CITATIONS 69 PUBLICATIONS 2,213 CITATIONS

SEE PROFILE SEE PROFILE

All content following this page was uploaded by Helmer Schweizer on 20 April 2021.

The user has requested enhancement of the downloaded file.


Contact Lens and Anterior Eye 44 (2021) 192–219

Contents lists available at ScienceDirect

Contact Lens and Anterior Eye


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

CLEAR - Effect of contact lens materials and designs on the anatomy and
physiology of the eye
Philip B. Morgan a, *, Paul J. Murphy b, Kate L. Gifford c, Paul Gifford d, Blanka Golebiowski d,
Leah Johnson e, Dimitra Makrynioti f, Amir M. Moezzi g, Kurt Moody h,
Maria Navascues-Cornago a, Helmer Schweizer i, Kasandra Swiderska a, Graeme Young j,
Mark Willcox d
a
Eurolens Research, Division of Pharmacy and Optometry, University of Manchester, UK
b
University of Waterloo, School of Optometry and Vision Science, Waterloo, Canada
c
School of Optometry and Vision Science, Queensland University of Technology, Brisbane, Australia
d
School of Optometry and Vision Science, UNSW Sydney, Australia
e
CooperVision Specialty EyeCare, Gilbert, AZ, United States
f
School of Health Rehabilitation Sciences, University of Patras (Aigio), Greece
g
Centre for Ocular Research and Education, University of Waterloo, Canada
h
Johnson & Johnson Vision Care, Jacksonville, FL, United States
i
Alcon Vision Care, IMG, Vernier-Geneva, Switzerland
j
Visioncare Research Ltd, Farnham, UK

A R T I C L E I N F O A B S T R A C T

Keywords: This paper outlines changes to the ocular surface caused by contact lenses and their degree of clinical signifi­
Contact lens evidence-based academic reports cance. Substantial research and development to improve oxygen permeability of rigid and soft contact lenses has
(CLEAR) meant that in many countries the issues caused by hypoxia to the ocular surface have largely been negated. The
Eyelid
ability of contact lenses to change the axial growth characteristics of the globe is being utilised to help reduce the
Cornea
myopia pandemic and several studies and meta-analyses have shown that wearing orthokeratology lenses or soft
Conjunctiva
Myopia control multifocal contact lenses can reduce axial length growth (and hence myopia).
Inflammation However, effects on blinking, ptosis, the function of Meibomian glands, fluorescein and lissamine green
staining of the conjunctiva and cornea, production of lid-parallel conjunctival folds and lid wiper epitheliopathy
have received less research attention. Contact lens wear produces a subclinical inflammatory response man­
ifested by increases in the number of dendritiform cells in the conjunctiva, cornea and limbus. Papillary
conjunctivitis is also a complication of all types of contact lenses. Changes to wear schedule (daily disposable
from overnight wear) or lens materials (hydrogel from SiHy) can reduce papillary conjunctivitis, but the effect of
such changes on dendritic cell migration needs further study. These changes may be associated with decreased
comfort but confirmatory studies are needed. Contact lenses can affect the sensitivity of the ocular surface to
mechanical stimulation, but whether these changes affect comfort requires further investigation.
In conclusion, there have been changes to lens materials, design and wear schedules over the past 20+ years
that have improved their safety and seen the development of lenses that can reduce the myopia development.
However, several changes to the ocular surface still occur and warrant further research effort in order to optimise
the lens wearing experience.

circumstances, a key aim is for a contact lens to achieve its desired


performance whilst either (a) leaving the anatomy and physiology of the
1. Introduction
eye unaffected or (b) altering ocular characteristics only as intended (e.
g. the programmed, structural change of the eye during in myopia
Contact lenses are medical devices worn to offer refractive correction
control with contact lenses). Given the complexity of the anatomical
or a medical solution to a clinical problem at the ocular surface. In all

* Corresponding author.
E-mail address: philip.morgan@manchester.ac.uk (P.B. Morgan).

https://doi.org/10.1016/j.clae.2021.02.006

1367-0484/© 2021 British Contact Lens Association. Published by Elsevier Ltd. All rights reserved.
P.B. Morgan et al. Contact Lens and Anterior Eye 44 (2021) 192–219

appears to be little difference between the sexes [22,23] and although


Abbreviations blink-rate increases with age, this may be due to age-related dry eye
disease issues [9].
CEDC corneal endothelial dendritic cells Increased blink-rate and unaltered blink completeness has been re­
CI confidence interval ported in the early stages of hard and rigid corneal lens wear [11,24,25],
ECP eye care practitioner whereas no difference in overall blink-rate was found between long-term
LIPCOF lid parallel conjunctival folds rigid corneal lens wearers and non-wearers [26]. However, long-term
LWE lid wiper epitheliopathy rigid corneal lens wearers showed fewer complete blinks and more
Ortho-k orthokeratology blink attempts than non-wearers. In addition, rigid corneal lens wearers
PMMA poly methyl methacrylate with 3- and 9-o’clock staining showed more incomplete blinks and more
SICS solution-induced corneal staining blink attempts than wearers with minimal staining and non-wearers
SiHy silicone hydrogel [26]. A trend toward an increased blink-rate was also shown in neo­
phytes fitted with soft lenses [27,28], as well as in adapted soft contact
lens wearers [7,29,30]. There was no clear effect of soft contact lens
wear on blink completeness [27–29], which appeared to be more
structures and physiological processes with which a contact lens in­ influenced by the task performed during the assessment [7].
teracts, this has proven to be a high threshold and one not yet fully met There is limited evidence of the effect of different soft lens materials
by modern lenses despite significant improvements in designs and ma­ and designs on blink characteristics. A shorter inter-blink interval (i.e.
terials, especially over the past 50 years. increased blink-rate) was reported after 10 min of soft contact lens wear,
This paper outlines the various changes caused by contact lens wear, particularly for toric lenses (a periballast design and a double slab-off
how these alter with different contact lens types and their degree of design), although none of the changes were statistically significant
clinical significance. The information is provided on a structure-by- [25]. An increased blink-rate was found in subjects wearing a hydrogel
structure basis and broadly follows the order in which an eye care contact lens (etafilcon A) after exposure to controlled adverse environ­
practitioner might examine the integrity of the eye during a contact lens mental conditions, whilst no change was observed in subjects wearing a
examination. This paper aims to review information which is different to SiHy lens (narafilcon A) [31]. The authors suggested that the higher
the CLEAR Complications Report [1] which features as a sister paper in blink frequency was ‘a compensation mechanism to alleviate the rela­
the CLEAR initiative. In general terms, the CLEAR Complications Report tively higher dryness over the lens surface.’ A higher blink-rate for SiHy
[1] describes changes to the eye which require clinical intervention. The lens wear (comfilcon A), compared with hydrogel lens wear (omafilcon
current paper covers physiological and anatomical alterations which do A), was seen during exposure to controlled standard and adverse envi­
not require such intervention either because this is not considered to be ronmental conditions [32]. According to the authors, the higher dehy­
helpful for patient care, or because the described ocular change has only dration observed for the SiHy lens in the study could be the reason for
recently been described and/or the appropriate clinical management is the rise in blink-rate ‘in an attempt to refresh the tear film more
not yet established. Inevitably there is modest overlap in these papers frequently’, although other work has found dehydration to be greater
but due to the deliberately different approaches taken (this paper takes with conventional hydrogels [33,34]. Contrary to these studies, other
an anatomy-based approach whereas the CLEAR Complications Report investigators found no significant difference in the increment of blink
[1] adopts an aetiology-based structure), where this occurs it is relevant, rate after two months of lens wear between hydrogel (hilafilcon B) and
helpful and additive. silicone hydrogel (lotrafilcon B) materials [28]. The effects of contact
lens wear on other aspects of blink dynamics, such as velocity and
2. The eyelids and adnexa duration, have not yet been studied.
The notion of incomplete blinking may be relevant to contact lens
2.1. Blinking wear as incomplete blinking accounts for a two-fold increase in the risk
of DED, meibomian gland atrophy and poor tear film stability [3].
Blinking is an important ocular surface physiological mechanism, Incomplete blinking might be more problematic for patients with low
maintaining physiology and providing good optics [2]. It involves both blink-rates, as this combination of effects will increase the exposure of
the upper and lower eyelids: the upper lid moves in the vertical and the inferior ocular surface. This means that potential contact lens pa­
inward, whereas the lower lid moves in a temporal-to-nasal direction tients who are more predisposed to incomplete blinking, those who are
[3]. Blinking is either voluntary (a conscious and deliberate blink), re­ using computers or ‘digital devices’ [9] or some ethnic groups (e.g.
flex (elicited by external tactile, light, sound or electrical stimulation), Asian patients [35]), may require closer clinical attention prior to fitting
or spontaneous (an unconscious blink in the absence of deliberate and during the aftercare process.
stimuli), with the latter the most common and most relevant to contact The measurement of blink characteristics has been challenging and
lens wear. The spontaneous blink-rate varies between 8 and 21 blinks complex using traditional methods [17,36]. However, the increased
per minute (in primary gaze) [4], has a duration of about 300 ms and an availability and accessibility of technologies such as high-speed digital
upper blink excursion of 7− 10 mm [5,6]. cameras [10,37,38] and mobile phones [39] have facilitated the inves­
These key blink variables can be influenced by various factors. For tigation of human blinking. Additionally, commercially available in­
example, both dry eye disease (DED) and contact lens wear cause an struments designed for tear film analysis, such as the LipiView II
increase in blink-rate [7–9]. Substantial variability in spontaneous blink interferometer or the IDRA ocular surface analyser, have the capability
rate has been reported in the literature, which may be attributed to a to measure some aspects of blink dynamics, allowing eye care practi­
number of factors including methodology employed [10], task per­ tioners (ECPs) to assess blink characteristics in the clinical setting.
formed during blink assessment [7,11–14], gaze direction [12,15,16],
cognitive and emotional factors [17] and inter-participant variability 2.2. Ptosis
[18,19]. The exact nature of the stimulus responsible for the increase in
blink-rate during contact lens wear is not clear, but tear film instability, Eyelid ptosis is the prolapse of the upper eyelid below its normal
visual disturbance and symptoms of ocular irritation may provide position [40]. Blepharoptosis is the more specific term for this
stimulation for blinking [7,20]. One report has noted an association ophthalmic condition and it can be either congenital or acquired [41].
between greater subjective dryness and increased blink-rate [21]. There Ptosis related to contact lens use is described.
Typically, the distance between the upper lid margin and the eyelid

193
P.B. Morgan et al. Contact Lens and Anterior Eye 44 (2021) 192–219

fold is minimal, but in contact lens induced ptosis this is enlarged, which signs of Meibomian gland dropout or had dropout of less than 25 %.
may be of cosmetic concern [3]. Since the vast majority of patients wear However, the pre-lens lipid layer thickness was strongly associated with
contact lenses bilaterally, this condition may not be noticeable and so its dry eye status [57].
prevalence may be higher than that reported in clinical practice. A A 2009 cross-sectional study found greater Meibomian gland
systematic review has suggested that there is an increased risk of ptosis dropout in 121 contact lens wearers than in 137 non-contact lens
in rigid corneal (OR 17.4x) and soft contact (OR 8.1x) lens wearers wearers, with the upper eyelid more affected than the lower. This work
compared to non-wearers [42]. Previous studies have highlighted the also noted that Meibomian gland dropout started not from the orifice
association of prolonged rigid corneal lens wear with acquired ptosis side but from the distal side in contact lens wearers [63]. Another
[43–50]. Although the exact mechanism remains unknown, most au­ outcome of this study was the significant correlation between the
thors agree that excessive physical manipulation of the eyelids during duration of contact lens wear and Meibomian gland dropout [63]. The
insertion and removal of rigid corneal lenses may be responsible for study results were compared to the earlier findings of age-related
inducing damage to the levator aponeurosis [43,46,48,51]. Other pro­ changes of the Meibomian glands in a normal population where the
posed mechanisms include eyelid oedema or inflammation [44] and authors found that aging increases the severity of Meibomian gland
contact lens-induced irritation [46,49]. There are fewer reports of con­ dropout. On average the Meibomian gland changes in contact lens
tact lens induced ptosis in soft contact lens wearers [49,52]. Contact lens wearers (mean age = 31.8 ± 8.0 years) from this study could be
application and removal and contact lens induced-irritation may play a observed in a 60- to 69-year-old age group of non-contact lens wearers
role in the pathogenesis of ptosis in soft contact lens wearers [47,49,52]. from the previous study [64]. There was no significant difference in
The vertical palpebral aperture size of rigid corneal lens wearers is average Meibomian gland dropout between rigid corneal lens wearers
significantly smaller than non-wearers, but this phenomenon does not and hydrogel lens wearers [63]. Another, more recent study which also
occur with soft lens use [51]. This observation has been confirmed in evaluated the effect of contact lens wear on Meibomian glands in an
long-term adapted rigid corneal and soft lens wearers, when compared Asian population supports these findings in that contact lens wear
to non-lens wearers. The palpebral aperture size of the rigid corneal lens, negatively affects Meibomian glands and, furthermore, the structural
soft lens and non-lens wearer groups were: 9.76 ± 0.99 mm, 10.25 ± changes worsen with years of wear [65]. Other researchers have also
0.94 mm and 10.10 ± 1.11 mm, respectively [53]. Ptosis is a feature of reported apparent changes to Meibomian glands related to contact lens
the upper eyelid and rigid corneal lenses cause a reduction in palpebral wear [66,67]. However, the methodology used (Meibomian gland acini
aperture size of about 0.5 mm [51,53]. reflectivity and acinar unit diameter measured by in vivo laser scanning
Subjects fitted on an overnight wear basis with a rigid corneal lens in confocal microscopy) is now considered to not image the Meibomian
one eye and a soft lens in the other eye for 13 weeks had a maximal glands but rete ridges present at the dermal-epidermal junction [68].
reduction in palpebral aperture size of 12 % with the rigid corneal lens at Furthermore, there is no association between rete ridges parameters
the 4–6 week time point versus 3 % for the soft lens eye [41]. At 13 measured by laser scanning confocal microscopy and actual Meibomian
weeks, the rigid corneal lens wearing eye had demonstrated a 3 % glands seen in meibography images [69].
reduction in palpebral aperture size compared to a 7 % increase for the The relationship between contact lens-related allergic conjunctivitis
soft lens eye. These results suggest there may be an adaptation for many and morphological changes in the Meibomian glands has been investi­
patients wearing contact lenses, with an initial reaction that may gated. It has been shown that allergic reaction, rather than contact lens
diminish over time, although the rigid corneal lens/soft lens contralat­ wear, causes Meibomian gland distortion in patients with contact lens-
eral nature of the study design may have influenced the findings. Most related allergic conjunctivitis. However, contact lens wearers both
cases of ptosis can be managed by refitting into an alternative lens type with and without contact lens-related allergic conjunctivitis showed
or discontinuing lens wear, although surgery is an option in extreme higher Meibomian gland dropout in contrast to non-wearers even
cases where ptosis does not resolve after discontinuation of contact lens though it was (marginally) not significant (p = 0.051). There was no
wear [3]. significant difference between the mean Meibomian gland distortion
between rigid corneal and hydrogel lens wearers [58].
2.3. Meibomian gland changes Other workers have found no association between changes in the
Meibomian gland morphology (both in Meibomian gland distortion and
Meibomian glands are large sebaceous glands located in the upper dropout level) and contact lens use. Here, contact lens wearers had
and lower eyelids just posterior to the tarsal plate. They contribute most significantly worse meibum quality and orifice plugging and further­
of the tear film lipid layer which protects the aqueous phase from more, abnormal meibum quality was strongly correlated to the duration
evaporating too quickly and also stabilises the tear film by lowering of contact lens wear [59]. This group challenged previous findings [58]
surface tension. Any abnormalities in Meibomian gland function and/or suggesting that contact lens replacement schedule and wearing time
anatomy lead to reduced meibum secretion and/or altered lipid should be considered when assessing the effect of contact lens wear on
composition which in turn disrupt the ocular surface integrity and in­ Meibomian gland morphology [59]. Other studies have also failed to
fluence contact lens success [54–56]. show that contact lens use affects Meibomian gland structure and
There is no consensus on the impact of contact lenses on Meibomian function [60,70].
glands (see Table 1). However, most recent data suggest that contact The aforementioned findings suggesting that the duration of contact
lens wear is not associated with Meibomian gland atrophy although it lens wear correlates with characteristics of the Meibomian glands stand
may affect Meibomian gland function [57–61]. Early findings on the in contrast to another study that did not find that correlation [71]. Ac­
relationship between contact lens wear and Meibomian glands showed cording to these authors, functional and structural Meibomian gland
that meibum in contact lens wearers has a 3 ◦ C higher melting point than changes in soft contact lens wearers occur within the first two years of
in non-wearers, with no difference between the three types of contact wear but do not worsen thereafter; however, the changes seem to be
lenses (polymethyl methacrylate [PMMA] corneal, soft or rigid gas permanent as contact lens dropouts did not show signs of improvement
permeable corneal) [62]. Another study examined the relationship be­ [71]. These results are consistent with other findings that found Mei­
tween various ocular factors and self-reported contact lens-associated bomian gland characteristics in SiHy contact lens wearers worsen sig­
dry eye. The data did not show correlation between Meibomian gland nificantly after three years of contact lens wear but remain stable after
dropout (i.e. apparent atrophy or loss of Meibomian glands when seven years of wear [72]. The earliest change that can be observed in
imaged) and dry eye in contact lens wearers suggesting that structural Meibomian gland appearance caused by contact lens wear is thickening
changes do not lead to altered or reduced meibum secretion. Further­ of the upper eyelid glands [72].
more, most patients (both contact lens wearers and non-wearers) had no A detailed analysis of various characteristics of Meibomian glands,

194
P.B. Morgan et al. Contact Lens and Anterior Eye 44 (2021) 192–219

Table 1
Effect of contact lenses on Meibomian glands. The blank spaces indicate that either evaluation was not performed, or it was not possible to make a clear judgement
whether results were relevant and appropriate. *Orthokeratology study. See also [86]. NIBUT = non-invasive breakup time; FBUT = fluorescein tear film breakup time;
LLT = lipid layer thickness; MG = meibomian gland; CL = contact lens.
Study Subjects Symptoms Plugging, Meibum quality also NIBUT, LLT Evaporation MG
obstruction expressibility FBUT rate appearance

Ong and Larke (1990) [62] CL wearers 70


yes
Non-wearers 70
Nichols and Sinnott (2006) [57] CL wearers 360 yes yes no
Arita et al. (2009) [63] CL wearers 121
yes yes
Non-wearers 137
Villani et al. (2011) [66] CL wearers 20
yes yes yes
Non-wearers 20
Arita et al. (2012) [58] CL wearers 64 + 77
no yes no
Non-wearers 55 + 47
Michalinska et al. (2015) [59] CL wearers 41 yes (quality) / no
no yes no no
Non-wearers 31 (expressibility)
Pucker et al. (2015) [60] CL wearers 70
no no no no
Non-wearers 70
Alghmandi et al. (2016) [71] CL wearers 60
Non-wearers 20 no yes yes yes no no yes
CL dropouts 20
Na et al. (2016) [61]* CL wearers 58 yes no no
87 (173
Ucakhan et al. (2018) [72] CL wearers
eyes)
yes yes yes yes
55 (103
Non-wearers
eyes)
Wang et al. (2019) [83]* CL wearers 59 no no no no
Pucker et al. (2019) [74] CL wearers 56
no no
CL dropouts 56
Gu et al. (2020) [65] CL wearers 85
yes yes yes
Non-wearers 63
Yang et al. (2020) [84]* CL wearers 60
no yes yes
Non-wearers 60
Llorens-Quintana et al. (2020) [73] CL wearers 33
yes
CL dropouts 8

such as area of dropout, number of glands, Meibomian gland length, Meibomian glands is also ambiguous. One study found that disturbed
Meibomian gland width and Meibomian gland irregularity has been Meibomian gland function characteristics (foam at Meibomian gland
provided [73]. This showed that experienced contact lens wearers had orifices, expressibility, meibum quality, lipid layer thickness, fluorescein
larger areas of dropout and shorter glands when compared to tear film breakup time and evaporation rate) were associated with
non-contact lens wearers, but those changes were not correlated to years symptoms of discomfort among the symptomatic contact lens wearers
of wear. However, neophytes fitted with daily disposable soft contact [67] whereas another did not find a difference in lipid layer patterns
lenses did not show any structural changes in Meibomian glands within between asymptomatic and symptomatic contact lens wearers [81]. In
the first 12 months of wear suggesting that changes happen later in time. addition, there was no difference in pre-lens tear break-up time between
Differences between contact lens materials were also examined. Here, symptomatic or asymptomatic groups [82]. Many other studies have
hydrogel contact lens wearers showed some significant variations in the shown that subjective symptoms are related to contact lens wear [57,61,
total number of glands and the area of gland atrophy in contrast to SiHy 65,66,72] but on the other hand, there are also some that did not
wearers. Non-invasive tear film breakup time also appeared to be observe this relation [59,60,71,83,84].
dependent on the lens material. Furthermore, the changes in the per­ The influence of overnight orthokeratology (ortho-k) on Meibomian
centage area of gland atrophy correlated with the fluorescein tear film glands has also received some attention in the literature. No significant
breakup time in SiHy contact lens wearers. Moreover, the preferred differences in Meibomian gland appearance and fluorescein tear film
habitual lens modality (monthly/fortnightly) seems to have an impact breakup time after 3 years of ortho-k wear in children and adolescents
on the area of gland atrophy and gland width, although this relationship have been reported [61]. These findings are supported by another study
was not clearly explained by the authors [73]. Meibomian gland width is that did not find significant changes in non-invasive tear film breakup
a characteristic that does not seem to be affected by contact lens wear. time, orifice plugging, meibum quality, difficulty of meibum excretion
Two studies found no correlation between this particular Meibomian and Meibomian gland dropout level when comparing time points prior
gland characteristic and contact lens wear when successful contact lens to and 2 years after the ortho-k wear in teenagers [83]. These outcomes
wearers were compared to both contact lens dropouts and non-wearers are contrary to that one study that found that Meibomian gland
[73,74]. appearance in the upper eyelid got gradually worse and non-invasive
Larger areas of Meibomian gland dropout are associated with shorter tear film breakup time significantly decreased within 12 months of
fluorescein tear film breakup time [75]. A moderate negative correlation ortho-k wear [84].
has been reported between daily lens wear duration and FBUT [76]. No Overall, then, the mechanism for Meibomian gland loss in contact
significant change in non-invasive tear film breakup time has been found lens wear is not fully understood. Possible explanations involve me­
after six months of SiHy contact lens wear, a similar finding to other chanical trauma, chronic irritation and aggregation of desquamated
studies in hydrogel contact lens wearers, but in contrast to one other epithelial cells at the orifices of the glands [62,63,85].
study that reported reduced non-invasive tear film breakup time in
neophytes after being fitted with hydrogel contact lenses [77–80].
The relation between subjective symptoms in contact lens wear and

195
P.B. Morgan et al. Contact Lens and Anterior Eye 44 (2021) 192–219

3. Conjunctiva ● Pre-lens surface deposits are a feature of all contact lens wear mo­
dalities, including daily disposable, although with this modality the
3.1. Bulbar and limbal conjunctiva clinical consequences of deposits are negligible. Deposit formation
occurs as a result of chemical interactions between the lens material
3.1.1. Hyperaemia and the tear film [119]. The deposits produce an allergic-type in­
Hyperaemia is a visible response to the wearing of a contact lens (or flammatory reaction [120,121]. Treatment is by initiating or
to some other irritating or inflammatory factor) that is expressed as increasing the frequency of the surface cleaning regime, or by
dilation of the conjunctival blood vessels [87]. This dilation changes the changing lens wear modality.
appearance of the exposed sclera and overlying bulbar and limbal con­ ● Post-lens hypoxia is produced by insufficient gas-exchange through
junctiva within the palpebral aperture from a quiescent ‘white’ to a the lens, principally due to low lens Dk [122]. Hypoxia was a
provoked ‘red’. The shift in hyperaemia is a sign that some underlying particular feature of early low Dk soft lens materials [93]. Silicone
factor has altered the homeostatic conjunctival blood flow balance. No hydrogel (SiHy) lens materials have effectively removed hypoxia
eye is ever perfectly ‘white’ as the conjunctiva contains visible blood (and thus hypercapnia) as a source for limbal and bulbar hyperaemia
vessels. There is a normal range in the hyperaemia appearance for the [123,124]. Post-lens hypoxia is treated by choosing a lens material
general population, reflecting physiological variation between in­ with a higher Dk [125,126]. Hypoxia is still an issue for scler­
dividuals and non-irritative influences on the homeostatic balance al/overnight medical wear due to the effect of the post-lens fluid
[88–90]. It is therefore important, when assessing change in hyperaemia reservoir [127,128].
with contact lens wear, to establish the non-lens wear baseline for each ● A less stable tear film can be produced in contact lens wear, which
patient and to compare future change to that baseline. induces increased tear evaporation [129–131], leading to partial
The hyperaemia is produced by increased dilation of the arterioles in dehydration of the lens material [132,133]. This may produce me­
the limbal corneal arcades and/or the bulbar conjunctival arteries [91]. chanical effects from a tighter lens fit or increased friction from the
The arteriolar walls are encircled by smooth muscle cells that control the lens surface [131,134]. Treatment is by changing the lens material,
diameter of the arteriole and thus blood flow through the arteriole. lens wear modality, environmental conditions (if possible) and other
When stimulated, the smooth muscle relaxes leading to an increase in lens wearer adaptations.
the arteriole diameter [91]. This changes the ratio between the hyper­ ● Lens care solutions can produce limbal and bulbar hyperaemia
aemia of the blood vessels to the whiteness of the scleral background and [135–138]. This may be a direct effect from a biologically incom­
the eye appears redder. The smooth muscle cells are innervated by patible reaction between solution component and ocular surface, or
sympathetic nerves [92], which provide central autonomic control over indirectly through a failure of the product to work effectively, e.g. an
the arteriole diameter. The muscle cells are also affected by local factors. ineffective protein cleaner. Treatment is by changing lens care so­
These locally-derived factors are moderated by chemical agents, such as lution modality or by lens wear modality.
prostaglandins or cytokines, that form part of the inflammatory response ● Lid-related infections, such as blepharitis or meibomian gland
[90,93,94]. dysfunction, can be related to poor lid hygiene [139]. For these
Increased dilation of the arterioles can be caused by mechanical cases, improved lid hygiene can produce a significant improvement
irritation, hypoxia, hypercapnia, acidic shift (increase in lactic and in ocular hyperaemia.
carbonic acids), increased osmolarity, increased potassium, toxic re­
actions to a noxious agent, (e.g. preservatives, hydrogen peroxide), or as 3.1.2. Sodium fluorescein, lissamine green and rose bengal staining
part of the inflammatory response to allergens or infection [95–97]. Two types of dye, sodium fluorescein and lissamine green, are
Many of these factors can be present in contact lens wear and can be currently used to examine the conjunctiva as part of contact lens pre-
acute or chronic in their expression. fitting and aftercare examinations [140]. Experimentally, fluorescein
Hyperaemia is such a common response to contact lens wear [93, is actively taken up by healthy cells in cell culture [141,142]. Clinically,
98–101] that it is easy to forget that hyperaemia can be a sign the eye is it is thought to permeate the cytoplasm of living but damaged cells,
experiencing stress [90,102,103]. It is therefore important to include whereas lissamine green stains the cell membrane of dead or damaged
questions about any reported or observed ocular hyperaemia as part of cells. The presence of lissamine green staining is thus highly specific for
the patient’s lens wear history during a clinical examination [104]. The dry eye disease [143]. Both stains are enhanced by the use of filters: a
clinician should identify the potential causes for the hyperaemia and yellow filter with blue light in the case of fluorescein and a red filter with
make suitable changes to the lens specifications, wear schedule, or lens white light for lissamine green. Lissamine green has largely replaced
care solution to prevent the condition becoming chronic. The clinician rose bengal, which is toxic, even in relatively low concentrations and
can use visual scales to grade hyperaemia severity and to monitor uncomfortable, if not painful, for the patient [144].
treatment effect [105]. Significant efforts have been made to develop Two main types of conjunctival staining are noted in soft contact lens
standardised grading scales that are easy to use by the clinician or that wearers: i) dryness-related staining, primarily located on the nasal and
rely on computer analysis [106–112]. temporal bulbar conjunctiva and ii) circumlimbal mechanical staining
The main areas identified as possible causes for bulbar and limbal from contact lens edges.
hyperaemia in contact lens wear are: lens surface/ocular surface me­ Conjunctival fluorescein staining is commonly seen in non-contact
chanical interactions, pre-lens surface deposits, post-lens hypoxia, lens wearers, but typically at lower levels than in contact lens
altered tear film, lens care solutions and ocular hygiene. All of these wearers. Some conjunctival staining was seen in 98 % of a mixed group
produce some form of inflammatory response from the ocular surface. of contact lens wearers and non-wearers; however the proportion of
subjects showing greater than Grade 1 staining (0–4 scale) was much
● Mechanical interactions can be produced by both tight-fitting and higher in the contact lens group: 62 % versus 12 % [145]. Another study
loose-fitting lenses, with a particular effect on the limbal arcades found conjunctival fluorescein staining in approximately half (53 %) of
producing limbal hyperaemia [113,114]. Lens edge design can have non-wearers versus 63 % of lens wearers [146]. With both groups, the
a particular effect on lens movement and on limbus/lens interaction incidence of staining was significantly higher for those classified as
[115]. Movement of the lens over the limbal area is the primary symptomatic.
source of the mechanical interaction [116] and can be visibly Lissamine green conjunctival staining is less common than fluores­
observed as limbal hyperaemia and increased staining in the peril­ cein staining in both contact lens wearers and non-wearers. However,
imbal area [117]. Treatment is by modification of lens specifications lissamine green staining (outside of the limbal area normally covered by
to improve the fit, with close attention to lens edge design [118]. the lens edge) is more discriminating in identifying symptomatic

196
P.B. Morgan et al. Contact Lens and Anterior Eye 44 (2021) 192–219

patients, particularly contact lens wearers [146]. The authors hypoth­ contact lens discomfort [82,161,170–172].
esised that reduced blinking in soft lens wearers results in greater The aetiology of LIPCOF in contact lens wear is thought to be similar
evaporation and poorer conjunctival lubrication which, in turn, leads to to that in dry eye disease - increased friction between the moving eyelid
increased friction during the blink and tissue damage. and the ocular surface. The presence of the contact lens in the eye alters
There has been little research on the effect of soft lens wear on the normal spreading and stability of the tear film over the ocular sur­
conjunctival staining. However, two studies have thrown useful light on face and the normal apposition of the eyelid against the bulbar
the significance of edge design [117,147]. Soft lens edge profiles broadly conjunctival surface. These changes lead to an increase in friction at the
fit into three categories: rounded, knife and chisel edge designs. Edge ocular surface between the eyelid and conjunctiva during blinking,
design was the primary factor in controlling circumlimbal fluorescein particularly in the 4 and 8 o’clock area where the shear forces are
staining for SiHy lenses [117]. A rounded edge produced the least cir­ thought to be greatest [170,173]. Within this model, improving the
cumlimbal staining, while a thin knife edge design produced the most, wettability of the lens surface and thus the distribution of the tear film
with an inverse association between staining and comfort. Comfort was over the lens and exposed conjunctiva, should also reduce friction and
poorest with the rounded design and highest with the knife edge (72 vs. the incidence of LIPCOF. However, it should be noted that a clear
87 out of 100) [117]. The study also noted lens rigidity as a secondary relationship between the coefficient of friction and LIPCOF is yet to be
factor, finding that a lens of higher modulus generated more circum­ proven [118].
limbal staining than a similar design of low modulus. Another study There is a limited literature on the effects of different contact lens
found broadly similar results with a wider range of lens types [147]. types, materials and designs on LIPCOF. Most studies report a positive
With both chisel and knife edge designs, the higher modulus SiHy de­ correlation between the presence of LIPCOF and discomfort symptoms
signs showed significantly more conjunctival staining than their or with the extent of lens wear experience. In a series of studies on
hydrogel counterparts. subjects experiencing discomfort when using low to medium water
Conjunctival staining induced by the lens edge is rarely symptomatic content (24–62 %) monthly disposable hydrogel contact lenses, LIPCOF
or accompanied by hyperaemia and, therefore, does not necessarily was strongly positively correlated to discomfort symptoms, older age, lid
require a change of lens. An exception might be instances of significant wiper epitheliopathy (LWE) and to a lower mucin production [82,171,
conjunctival indentation which has been imaged by optical coherence 174].
tomography in soft lens wearers [148–151]. Clinically, this is revealed The effect is also seen with neophyte lens wearers. One study re­
by pooling of fluorescein in circumlimbal indentations corresponding to ported that the main discriminators for contact lens-induced dry eye in
the positioning of the lens edge. In one study of nine different soft lens neophyte contact lens wearers wearing vifilcon A hydrogel contact lens
types, conjunctival indentation was associated with poorer comfort and senofilcon A SiHy contact lenses was LIPCOF [160]. Similarly,
[152]. This can be alleviated by switching to a lens of thinner edge neophytes who wore SiHy lenses full time for six months showed an
design and/or lower modulus. increase in LIPCOF [80]. The extent of lens wear experience is also a
factor in the development of LIPCOF. Using the term ‘con­
3.1.3. Lid-parallel conjunctival folds junctivochalasis’, but using the Hoh LIPCOF scale to describe conjunc­
Lid-parallel conjunctival folds (LIPCOF) are observed as small folds tival changes (suggesting LIPCOF might have actually been reported), an
on the bulbar conjunctival surface, close to the lower lid margin and increase in LIPCOF with lens wear experience and with lens wearer age
near to the limbus. They occur in both the lower temporal and nasal has been reported [175]. Also, a positive correlation between LIPCOF
bulbar conjunctival areas (at around 4 and 8-o’clock of the corneal and duration of contact lens wear (with LIPCOF findings higher after at
location), while the patient is looking in the primary gaze [118,153]. least a year of SiHy usage) has been found [176].
The term lid-parallel conjunctival fold was first introduced in 1995 There is one contrasting report that found no relationship between
[154] and the feature has been the subject of research by others sub­ LIPCOF and contact lens materials (hydrogel contact lens and SiHy
sequently [82,153,155,156]. contact lenses) or lens wear modality (yearly disposable contact lens and
LIPCOF are thought to be caused by increased shearing forces during monthly disposable contact lens) [155], although this could be due to
blinking, as a result of increased friction between the ocular surface and the small number of study subjects in this work.
the lids, which, in turn, has been caused by reduced lubrication due to a It has been proposed that LIPCOF should be incorporated into the
deficient tear film [157]. These causal factors are particularly found in clinical assessment of lens wearers to identify those wearers at greater
dry eye disease and studies have shown that LIPCOF is highly correlated risk of developing contact lens discomfort symptoms. This is based on
with dry eye disease and associated symptoms [153,156–159]. The the finding that ocular dryness symptoms are strongly linked to a
movement of the eyelids on the conjunctiva causes it to wrinkle into the combination of LIPCOF and non-invasive tear film breakup time [156].
folds. The model proposes that the greater the friction, the greater the However, even though the specificity and sensitivity of LIPCOF ‘sum’
size or extent of the LIPCOF. The folds are not permanent, but are scores (a combination of nasal and temporal score) has proved to be
maintained by the position of the eyelid. If the lower lid is retracted, the excellent [160], the repeatability of LIPCOF is limited, although prob­
LIPCOF will disappear, but will reappear after normal blinking [157]. ably better than other measures that may be associated with contact lens
LIPCOF can also occur in subjects showing no other signs or symptoms of discomfort such as tear break-up time, Schirmer test, tear meniscus
dry eye disease [160]. height or phenol red thread test [177].
LIPCOF are classified in two ways: by assessing the height of the folds It is important to note that, clinically, LIPCOF should not be mistaken
[154] or by counting the number of folds [161–163]. The number of for conjunctivochalasis or conjunctival flaps. LIPCOF refers to small
folds approach has been adopted for both slit-lamp biomicroscopy and conjunctival folds of about 0.08 mm height [162,178], visible under
optical coherence tomography assessment [153,155,164]. white light (slit-lamp magnification >18x) [179,180] and caused by
LIPCOF interfere with tear meniscus assessment, either of the tear friction forces during blinking [181]. Conjunctivochalasis may partly
meniscus radius [165], height [165,166] volume [167,168], curvature, share the same aetiology as LIPCOF, but it is also age-related and can be
depth or cross-sectional area [162]. LIPCOF may affect tear film mixing, located anywhere on the bulbar surface [157,181–183]. ‘Conjunctival
spreading and thus ocular surface lubrication, although the precise flaps’ are epithelial sheets, containing goblet cells, which are detached
mechanism for this is unclear [169]. from the conjunctiva, are sized of 0.01 mm height, can be located
‘Contact lens discomfort’ is an adverse clinical response to contact anywhere on the bulbar surface in irregular directions and are mainly
lens wear, characterised by the wearer reporting symptoms of discom­ reported after SiHy contact lens wear [157,184–187].
fort and possible reduced lens wear time. Symptoms are not always The management of LIPCOF in contact lens wearers has been aimed
associated with clinical signs. LIPCOF has been proposed as a feature of at reducing the amount of friction between the lids and the contact lens,

197
P.B. Morgan et al. Contact Lens and Anterior Eye 44 (2021) 192–219

either by recommending the use of artificial tears or by changing to a play a role in initiating such a response. Lens material or lens design
contact lens with improved lens surface wettability (see CLEAR Main­ cannot be excluded and the role of these factors in upregulation of
tenance and Biochemistry Report [188] for discussion on contact lens conjunctival inflammation requires further investigation; as does the
wettability). A recent study demonstrated that moving experienced impact of rigid corneal lenses. Nevertheless, the return towards pre-lens
monthly replacement daily contact lens wearers to senofilcon A levels [194] suggests the immune response is transient and adaptation
two-weekly replacement lenses, or to spectacle wear, significantly occurs.
improved LIPCOF after three months [173]. However, a clear frame­ In the normal cornea, the highest density of dendritiform cells at the
work for such intervention is yet to be formulated [180]. ocular surface is found at the limbus [191]. In this region, dendritiform
cells are activated to mature and migrate via the lymphatic vessels be­
3.1.4. Sub-clinical inflammatory response tween the cornea and lymph node [196]. They are also nearby the limbal
The inflammatory response to contact lens wear, which occurs in blood vessels, enabling travel in the circulation to further facilitate the
response to a myriad of factors including mechanical interactions be­ immune responses. They also have a role in intravascular surveillance
tween the lens and ocular surface, hypoxia, tear film alterations, de­ within the limbal vessels [197]. The impact of contact lenses and spe­
posits on the lens surface, lens care solutions and hygiene, is observed cifically materials and edge design on limbal dendritic cells has not yet
clinically as hyperaemia at the conjunctiva as discussed above. Sub­ been studied, but this area is worthy of research given the essential role
clinical biomarkers, such as dendritiform cells which may initiate and these cells play in ocular surface defence.
modulate the immune response by stimulation of T and B cells (see also
review [189]), can be imaged using in vivo confocal microscopy [190] 3.1.5. Sensitivity changes
(Fig. 1). Although the effects of contact lens wear and ocular surface Before considering conjunctival sensitivity, it is helpful to differen­
conditions on dendritiform cells in the corneal epithelium have been tiate the neural architecture in the conjunctiva, which consists of
studied (see Section 4.1.5) [190,191], less is known about any impact on epithelial free nerve endings and specialised receptor bodies [92], from
dendritic cells within the conjunctiva and limbus. the cornea, which consists of epithelial free nerve endings [198–201].
The density of dendritiform cells in the temporal bulbar conjunctiva The density of sensory nerve fibres in the cornea is the highest in the
was increased following one week daily wear of SiHy and hydrogel body. These nerve fibres are arranged in large overlapping receptive
reusable lenses (disinfected with hydrogen peroxide), but not of the fields, providing an extremely high level of sensitivity [202,203]. The
same hydrogel lenses replaced daily [192]. This increase was of similar conjunctival neural density is much less than the cornea and provides a
magnitude to that observed at the lid margin (Section 3.2.3) and central reduced sensitivity as a result [204]. It is also helpful to define the limits
cornea (Section 4.1.5) in the same study. Increased numbers of immune of bulbar and limbal conjunctiva. For this review, bulbar is considered to
cells in the bulbar conjunctiva were confirmed using impression begin 2 mm from the visible limbal/cornea junction and the limbal
cytology and flow cytometry [192]. In contrast, another report conjunctiva to be contained within a 2 mm annular zone around the
demonstrated that one week wear of daily disposable hydrogel lenses visible junction [205]. A soft contact lens can be expected to cover and
resulted in a rise in the number of dendritiform cells in the nasal con­ move over the limbal area during wear [114].
junctiva [193]. This difference may be attributed to lens type or to
methodological differences as dendritic cell numbers are shown to differ 3.1.5.1. Bulbar sensitivity changes. There is a limited literature on
across bulbar conjunctival regions; here, cell numbers were reduced bulbar and limbal conjunctiva sensitivity changes with contact lens wear
after four weeks and returned to similar numbers to pre-lens wear levels and the results are mixed. Early studies showed a reduction in bulbar
at six months of lens wear [194]. There was no difference in the number sensitivity with PMMA corneal, rigid gas permeable corneal and low Dk
of conjunctival dendritiform cells between experienced wearers of soft hydrogel lens wear [206,207]. In contrast, both an increase [208]
reusable hydrogel lenses (with a mean of 10 years wear experience) and and no change [209] in sensitivity with contact lenses have also been
non-contact lens wearers [194]. reported. An increase in sensitivity in lens wearers who discontinued
Upregulation of the immune response may be more likely to occur wear due to discomfort has been reported [206]. No studies have re­
when contact lenses are re-worn and proteins and lipids on the lens ported on bulbar sensitivity changes with scleral lens wear.
surface [195] or microbial contamination of the lens storage case may A convincing mechanism to explain the process that produces an

Fig. 1. In vivo confocal microscopic images depicting: Left: dendritiform cells with long dendrites found in the bulbar conjunctival epithelium (400 μm × 400 μm
image captured at depth of ~20 μm from the epithelial surface in the temporal quadrant), Right: dendritiform cells in the sub-basal epithelium of mid-peripheral
cornea (1 mm × 1 mm montage) [190].

198
P.B. Morgan et al. Contact Lens and Anterior Eye 44 (2021) 192–219

altered bulbar sensitivity is lacking for both rigid and soft lens wear. This variation [223] and a normal baseline should be obtained for each pa­
may be due to the large overlapping receptive fields of the ocular surface tient before lens wear to allow comparison with subsequent lens wear.
receptors and the imprecision of current sensitivity measurement tech­ Assessment should only be made across the central area of the everted
niques. The conjunctival blood supply facilitates gas exchange and en­ lid as other areas of the conjunctiva can be affected by non-contact
sures a normal metabolism for the bulbar conjunctiva and since the lens lens-related effects or by the lid eversion process, which can distort
is not physically present, a mechanical interaction does not occur. A the lid along the lid margin [224].
possible effect from conjunctival drying, associated with 3 and 9 o’clock Changes to the papillary conjunctiva are a significant complication
staining or with insufficient blinking during lens wear [210] affecting of all types of contact lens wear and are caused by the interaction be­
conjunctival epithelial nerve fibre endings is possible, but this has not tween the front surface of the contact lens and the back surface of the
been tested. The lack of an obvious mechanism for sensitivity loss in the eyelid [101,120]. The interactions can be mechanical or allergic in na­
bulbar conjunctiva, in combination with the current results, suggests ture, producing an inflammatory response that alters the homeostatic
that sensitivity is not significantly changed in bulbar conjunctiva for any balance in the conjunctival blood vessel diameter (hyperaemia) [91]
modern lens wear type. and an inflammatory cell response that produces localised swelling in
the conjunctival epithelium and stroma (roughness) [94,224]. The me­
3.1.5.2. Limbal conjunctiva sensitivity changes. The general evidence for chanical interaction is produced by increased friction between the
the limbal conjunctiva is that short-term and long-term wear of soft palpebral conjunctiva and lens surface, due to lens dehydration or lens
hydrogel and SiHy lenses produces an increased limbal conjunctival surface deposition [170,225]. A papillary reaction can also be caused by
sensitivity [116,211]. An increase in sensitivity was also found in pre­ surface deposits or by chronic leakage of lens care solution ingredients
vious lens wearers who discontinued wear due to discomfort [211]. In (e.g. PHMB) from the lens during wear [136]. These clinical signs and
contrast, other studies have found no change in inferior limbal their cause are characteristic of contact lens-induced papillary
conjunctival sensitivity for both low Dk soft hydrogel and SiHy lens conjunctivitis [226]. Differential diagnosis should also be made for
wearers [212,213]. allergic, atopic and vernal conjunctivitis, which can show similar clin­
The presence and movement of the soft lens edge over the limbal ical appearance and symptomatology [227].
conjunctival zone during lens wear suggests a mechanical interaction The appearance of contact lens-induced papillary conjunctivitis can
between the lens and the limbal conjunctival sensory nerves [214]. vary with time from onset, lens wear type or modality and inflammation
Evidence for this mechanical interaction on the limbal zone has been severity [228]. Onset can be within a few weeks (from initial lens wear)
observed as circumlimbal staining following SiHy lens wear [117]. for soft contact lenses, or up to 14 months in rigid corneal lens wear
However, in contrast to the reduced sensitivity produced by adaptation [224]. Studies indicate that 6–12 % of soft hydrogel lens wearers will
to a mechanical stimulus as reported for the corneal nerves (Section present with contact lens-induced papillary conjunctivitis at some stage
4.1.6), the mechanical interaction with the limbal zone increases in their lens wear lifetime [229–233], that overnight lens wear increases
sensitivity. This suggests that the mechanical interaction has a different the incidence rate (up to 18 %) [234,235] and that daily disposable lens
form which promotes an increased neural response or that the neuro­ wear reduces the rate (as low as 2 %) [236]. Silicone hydrogel lens wear
receptors respond differently [201,215]. Moreover, the neural archi­ produces a slightly higher incidence rate than soft lens wear [100,235].
tecture is different in this area, possessing specialised pressure sensors A contact lens-induced papillary conjunctivitis incidence rate of 2 % has
[201,216,217]. It is unclear whether this difference has a role in the been reported in overnight rigid corneal lens wear [229].
altered neural response. Contact lens-induced papillary conjunctivitis presents as both
Increased limbal sensitivity may lead to symptoms of discomfort by increased hyperaemia, which appears first, and increased lid roughness.
the lens wearer and so form part of the contact lens discomfort model In soft hydrogel lens wear, papillae are more numerous than in rigid
[218]. The form that this symptomatic limbal zone/lens interaction corneal lens wear, are located towards the upper tarsal plate and the
takes is unclear, but lens edge design, lens modulus and lens thickness apex of the papillae has a rounded, flatter form [120,224]. In rigid
might all influence the strength of the mechanical interaction [117]. corneal lens wear, the papillae are crater-like and are located towards
Significantly, these factors have also been linked to lens wear discomfort the eyelid margin [120,224]. In severe cases, papillae can develop up to
[118,137,219]. 1 mm in diameter and extend over a wide area of the conjunctiva. Here,
The alternative mechanism for how the lens could affect limbal patients can complain of discomfort/itching (which may be sufficient to
conjunctival sensitivity (hypoxia) can be rejected since this would lead cause the patient to stop lens wear) and blurred vision from increased
to a depression in neural activity [220] rather than the observed in­ mucin production. Treatment may involve stopping lens wear for a
crease. An alteration in corneal or conjunctival metabolism is, in any period of time, along with the use of anti-inflammatory therapy, a
case, unlikely with the presence of the extensive blood supply through change in lens wear type/modality and/or lens care solution [224,228].
the limbal arcades [205].
3.2.2. Lid wiper epitheliopathy
The lid wiper is the portion of the eyelid marginal conjunctiva that is
3.2. Palpebral and marginal conjunctiva in contact with the globe and wipes the ocular or contact lens surface
during blinking. Lid wiper epitheliopathy (LWE) is a clinical condition
3.2.1. Papillary hyperaemia and roughness observed through the staining of the lid wiper region with fluorescein,
The palpebral conjunctiva forms the back surface of both eyelids lissamine green or rose bengal [237], which extends beyond the physi­
[221]. It is a key part of the ocular lubricating system, in conjunction ological staining of Marx’s line [238]. Lid wiper epitheliopathy is
with the tear film, that assists with eyeball and eyelid movement [222]. observed in both upper and lower eyelid and has been shown to occur in
It is formed into an epithelial layer, containing mucin secreting cells contact lens wearers as well as in non-wearers [209,237,239–247]. The
(goblet cells) and a stromal layer that contains the blood supply, derived severity of lid wiper staining is most commonly graded subjectively
from the episcleral arteries and some inflammatory cells. The normal based on the horizontal and vertical extent of lid margin staining [248],
appearance of the palpebral conjunctiva is satin or smooth in 14 % of although objective/automated techniques have also been used [249,
people, containing small uniformly sized ‘micropapillae’ (<0.3 mm 250].
diameter) in 85 % of people, or containing non-uniform papillae (up to Although the exact aetiology of LWE remains unclear, the primary
0.5 mm diameter) in <1 % of people [120]. Palpebral hyperaemia and hypothesis is that LWE results from increased friction between the lid
roughness can be graded using visual grading scales [105]. Similarly to wiper and the ocular or contact lens surface during blinking in the
bulbar and limbal hyperaemia, there is considerable inter-participant absence of adequate lubrication [237]. LWE is likely to be a

199
P.B. Morgan et al. Contact Lens and Anterior Eye 44 (2021) 192–219

multifactorial condition with different underlying causes [134], in some instances, histological widths of the lid wiper conjunctiva were
including deficient tear film [251], altered mucin production [171], significantly correlated with lissamine green staining grades [262], no
increased tear osmolarity [252–254], inflammation [134], incomplete correlation was found between lissamine green staining and morpho­
blinking [255] and eyelid pressure [256,257]. A recent study further logical changes in the lid wiper epithelium [246].
supports the primary hypothesis that inadequate lubrication causes Only two studies have investigated the vascular response of the lid
friction factors and results in LWE [258]. Upper LWE may have a wiper to contact lens wear. Redness of the upper and lower lid wiper (as
different aetiology to lower LWE [239,253]. During contact lens wear, an analogue of vascular response) in soft contact lens wearers and non-
the frictional properties of contact lenses may contribute to the aetiology wearers did not significantly change through the day or following ≥6 h
of LWE [170]. of lens wear [244]. In contrast, increased microvascular network density
Lid wiper staining was observed in 25 % of patients presenting to an has been reported in the upper lid wiper after 6 h of SiHy lens wear in
eye clinic, including contact lens wearers and non-wearers [259]. Some neophytes, whilst no change was observed through a day of no lens wear
reports suggest a greater prevalence and grade of LWE staining in con­ [266].
tact lens wearers (52 %–84 %) compared with non-wearers (13 %–40 %) While LWE has been established as a diagnostic marker for dry eye
[239,241,244], whilst others have found no difference [242,243,245, disease [162], the relationship between LWE and contact lens discom­
246]. Studies investigating longitudinal changes in LWE have shown fort is not clear. Several studies have reported greater LWE prevalence
increased upper lid wiper staining with lens wear [80,160,254,260]. and grade in symptomatic contact lens wearers (67 % and 90 %,
Upper LWE staining significantly increased in neophytes after four respectively) compared with asymptomatic lens wearers (13%–32%),
weeks of hydrogel contact lens wear [160]. Similarly, increased upper primarily for the upper lid wiper [67,82,171,237,240–243]. In contrast,
LWE grades were found in neophytes following six months of SiHy an approximately similar number of studies have been unable to show a
contact lens wear [80]. Increased upper lid wiper staining was reported relationship between LWE staining and contact lens associated dry­
after 10 days of SiHy lens wear, compared to spectacle wear at baseline, ness/discomfort [80,134,209,244,247,254,262]. This could be partly
whilst no change was found for the lower lid [254]. Regarding diurnal due to differences in methodology, population characteristics and clas­
variation, upper lid wiper staining significantly increased throughout sification criteria for contact lens discomfort. Histological widths of the
the day in contact lens wearers, whilst no change was observed in lid wiper conjunctiva have not been associated with contact lens comfort
non-wearers [261]. An increased area of lower lid wiper staining has [262]. No association was also observed between lid wiper hyperaemia
been reported after 12 h of soft contact lens wear, whilst no change was and subjective comfort [244]. On the other hand, increased microvas­
found in non-wearers [209]. cular density in the lid wiper was significantly correlated with decreased
There is limited evidence of the effect of different contact lens ma­ contact lens comfort [266]. Recently, a study investigating the use of
terials and designs on LWE. Greater LWE prevalence and grade in rigid fluorescein-labelled wheat germ agglutinin as a marker for ocular sur­
corneal lens wearers compared with soft contact lens wearers was re­ face mucins found that symptomatic lens wearers showed reduced
ported for the upper lid, whilst lower LWE was similar between groups wheat germ agglutinin fluorescence in the lid wiper, which may suggest
[239]. Similarly, rigid corneal lens wear was associated with greater altered density and/or structure of mucins [267].
LWE grades compared with soft contact lens wear, particularly at the Contradictory results in the literature may be partly due to discrep­
upper lid margin [262]. These findings would suggest a mechanical ef­ ancies in staining protocol and grading technique. Lid wiper staining has
fect possibly related to lens-related factors such as modulus, edge design, been shown to be influenced by dye concentration [250,268], instilled
movement. Conversely, a study on daily wearers of hydrogel, SiHy or volume [250], frequency of lid eversion [268,269], sequential instilla­
rigid corneal contact lenses failed to demonstrate an effect of lens type tion and timing of assessment [270], which highlights the need for a
on upper lid wiper staining [263]. In addition, a number of studies standardised methodology. The limited repeatability of subjective
showed no significant differences in LWE grades or patterns between grading of LWE staining [177,271] and variations between brands [272]
SiHy lens types [254,260,264]. In contrast, in a study of a large sample may have also contributed to inconsistencies in the literature.
of soft contact lens wearers, habitual senofilcon A wearers were found to Whether LWE really represents a pathological process and its clinical
have the lowest LWE grades. However, according to the authors, the significance are still under debate. Nonetheless, subjects showing severe
clinical significance of the differences observed was unclear [265]. To lid wiper staining may benefit from treatments aimed at increasing
date, no studies have demonstrated a link between contact lens coeffi­ lubrication during contact lens wear [241]. Management strategies
cient of friction and LWE. could include altering lens type and wearing modalities, improving
A limited number of studies have investigated the impact of contact blinking behaviour [255] and the use of lubricant eye drops [263,273,
lens wear on the lid wiper at a cellular level using impression cytology 274].
techniques. No differences were found in the appearance of epithelial
cells and density of goblet cells of the upper marginal epithelium be­ 3.2.3. Sub-clinical inflammatory response
tween contact lens wearers and non-wearers [245]. Expression of Given the nature of the interaction between the tissues of the
keratinization-related proteins (filaggrin, transglutaminase-1 and cyto­ palpebral conjunctiva and lid margin with the contact lens edge and
keratin 1/10) was demonstrated in the lid margin epithelium, which surface, it would be reasonable to speculate that recruitment of immune
may indicate that a pathological process occurs at the lid wiper. In cells would be seen in these tissues.
contrast, soft contact lens wearers with short and moderate experience The density of dendritic cells at the lid margin was increased
had altered cytoplasmic and nuclear characteristics, as well as reduced following one week of daily SiHy and hydrogel reusable lens wear, but
goblet cell density in the upper lid wiper compared with non-wearers not in daily disposable wear of the same hydrogel lenses [192]. This
[246]. Interestingly, the lid wiper epithelium of previous contact lens increase was of similar magnitude to that observed on the bulbar con­
wearers was similar to that of non-wearers suggesting that changes junctiva and central cornea in the same study. The rise in number of
during contact lens wear might be reversible to some extent. Rigid dendritic cells was supported by a rise in leukocytes shown with
corneal contact lens wear has been associated with a wider, more ker­ impression cytology and flow cytometry and suggests an upregulation in
atinised, lid wiper conjunctiva compared with soft contact lens wearers inflammation occurs at the lid margin with the wear of reusable lenses,
and non-wearers, both at the upper and the lower lid margin [262]. but not when these lenses are replaced daily. It is hypothesised that
These findings lend support to the proposed mechanical or frictional degraded proteins and lipids on the lens surface or microbial contami­
nature of LWE. nation of the lens storage case and then transfer of microbial product to
The relationship between vital staining of the lid wiper and the the lens may play a role in initiating such an inflammatory response
specific underlying morphological changes remains unclear. Although, [195].

200
P.B. Morgan et al. Contact Lens and Anterior Eye 44 (2021) 192–219

Interestingly, another study of daily disposable hydrogel lens wear Despite its long history of use in ophthalmology, the mechanisms of
reported elevated density of dendritic cells at the lid margin after 6 corneal staining are not fully understood [287,288], although recent
months of wear in patients with symptoms of contact lens discomfort, research has expanded this. Sodium fluorescein is water soluble and is
but not in asymptomatic wearers [275]. Pre-lens wear dendritic cell more accurately described as a dye rather than a stain. Early theories
density was not measured, so it is impossible to surmise whether the suggested that punctate staining represented an accumulation of fluo­
recruitment of immune cells into the lid margin was a response to the rescein in intercellular spaces or pooling in sites of missing cells. How­
presence of the lens itself or a subclinical inflammation due to contact ever, these have been largely discredited. Several observations point
lens dry eye. No relationship was found between lid margin dendritic towards the fact that fluorescein enters the epithelial cells themselves: i)
cells and lens comfort [192]. Nevertheless, the demonstrated relation­ rinsing with saline fails to eliminate punctate staining [289], ii) punctate
ship between contact lens-induced discomfort and higher numbers of staining matches the size and shape of epithelial cells [290], iii) cells
dendritic cells at the lid margin is worthy of further study. shed by apoptosis show fluorescein staining and iv) even healthy
Immune cells on the palpebral conjunctiva have not been observed in epithelial cells fluoresce, albeit at a much lower level than damaged cells
the context of contact lens wear. Examination of the palpebral con­ [291]. In the case of deeper corneal damage, fluorescein can diffuse in
junctiva conducted in normal patients and in those with severe allergy the stroma and show a background glow [292].
(vernal keratoconjunctivitis) has suggested that dendritic cells can be An extensive study of contact lens wearers (91.5 % soft, 8.5 % rigid
observed on the palpebral conjunctiva using in vivo confocal microscopy corneal) noted corneal staining in 54 % of patients [293]. Factors related
[276,277]. It is not clear whether the reported cells were observed in the to increased corneal staining included: increased daily wearing times,
thin 2–3 layer epithelium of the palpebral conjunctiva or in deeper tissue lissamine green conjunctival staining, contact lens deposition, increased
[277]. A study which examined the palpebral conjunctiva in normal tear meniscus height, decreased hydrogel nominal water content and
eyes and in trachoma showed no association between the number of lower financial income. The wearing of SiHy (as opposed to hydrogels)
dendritiform cells viewed in vivo in the palpebral conjunctiva and the gave lower levels of corneal staining.
number of cells labelled as dendritic cells in an immunohistochemical
examination of biopsy specimens from the same patients [278] sug­ 4.1.1.1. Desiccation. With both rigid corneal and soft contact lenses,
gesting that the dendritiform cells may not all be true dendritic cells. desiccation induced staining is the most commonly encountered type of
corneal staining. Desiccation staining occurs in locations where the tear
3.2.4. Sensitivity changes film is thinnest and/or least stable. Soft lens smile-shaped staining oc­
There is a very limited literature on changes in the palpebral and lid curs parallel and adjacent to the lower lid. Not surprisingly, desiccation
margin conjunctiva sensitivity in contact lens wear. In part, this may be staining is more common with partial blinking and often marks the
due to difficulties in measurement of this area - for example, lid lowest extent of the upper lid during the blink. Desiccation staining can
manipulation and exposure of the tissue under measurement may also coincide with the edge of the lower tear meniscus due to a thin band
impact on the values obtained. In general, studies indicate that soft in the tear film from surface tension [29].
contact lens wear produces a reduction in upper lid palpebral conjunc­ Low levels of desiccation staining can be tolerated since this can
tival sensitivity, but a possible increase in sensitivity in symptomatic often be present in the normal non-lens wearing eye [294]. Low to
lens wearers. A reduction in palpebral conjunctiva sensitivity in rigid moderate levels of corneal desiccation staining can be mitigated by
corneal lens wearers and in low Dk soft lens wearers has been reported improved blinking, the use of wetting drops, reduced wearing time, or
[279]. A decrease in palpebral sensitivity with soft contact lens wear, the use of dehydration-resistant lens materials [295].
but an increase in sensitivity in symptomatic soft lens wearers has also Rigid lens 3 & 9 o’clock staining is also a type of desiccation staining
been reported [280]. There was an increase in palpebral sensitivity for but occurring through instability of the pre-corneal rather than pre-lens
soft lens wearers compared to non-lens wearers [281]. One study re­ tear film. The characteristic triangular shape of 3 & 9 o’clock staining
ported reduction in upper lid palpebral sensitivity in soft lens wearers, can be explained in terms of poor corneal wetting during the blink due to
but an increase in lower lid palpebral sensitivity in the morning [209]. vaulting of the lid between the lens edge and ocular surface. The largest
The mechanism for these effects appears to be the mechanical interac­ gap occurs when the lid crosses the widest part of the lens. Less wide­
tion between the anterior lens surface and the overlying palpebral spread 3 & 9 o’clock staining, closer to the lens edge can be explained by
conjunctiva during eyelid blinking producing an adaptation in the localised tear film thinning adjacent to the tear meniscus surrounding
neural response to the repeated stimulation. The increase in sensitivity the lens. Both types of staining are best mitigated by modifying the lens
in symptomatic lens wearers suggests a possible effect on lens wear design to improve corneal wetting, i.e. decreasing edge clearance,
discomfort from this route [280]. decreasing edge thickness, decreasing diameter, or increasing diameter
Lid margin sensitivity is the highest of all the conjunctival areas [296].
[282–284]. It is generally considered to be reduced in both rigid corneal
and soft lens wear [209,279,281,282,285,286]. The mechanism for this 4.1.1.2. Trauma. The simplest example of corneal trauma is foreign
effect is thought to be linked to a neural adaptation from the mechanical body abrasion caused by dust or an eyelash being trapped beneath the
interaction between the eyelid margin and the edge of the contact lens lens. Epithelial abrasions can heal at more than one square millimetre
during blinking. per hour, depending on the initial wound size; larger wounds healing
faster [297]. Based on the results of animal studies [292], a temporary
4. Cornea discontinuation of lens wear has been proposed based on the depth and
extent of corneal staining: 24 h if slight stromal diffusion, two to three
4.1. Epithelium days if moderate stromal diffusion.
Superior arcuate corneal staining (also termed Superior Arcuate
4.1.1. Fluorescein staining Epithelial Lesion) was relatively common with early hydrogel designs
Fluorescein is versatile enough to reveal most disruptions to the and perplexed ECPs for some time, partly due to its similarity to superior
ocular surface and is helpful in detecting a wide range of contact lens- limbic keratitis. However, it became clear that Superior Arcuate
related stress factors, including: desiccation, trauma, infection, allergic Epithelial Lesions are due to trauma from relatively stiff soft lenses
and toxic effects. As well as differentiating various corneal disorders [298]. Superior Arcuate Epithelial Lesions are typically located on the
through the pattern of staining (Fig. 2), it indicates the severity through cornea under the upper lid approximately a millimetre from the limbus.
the depth and extent of staining and, therefore, is an invaluable pointer Lenses of high modulus or thick lenses of any modulus that fail to align
towards management.

201
P.B. Morgan et al. Contact Lens and Anterior Eye 44 (2021) 192–219

severe cases may be associated with reduced comfort [300–302] and an


increased incidence of infiltrative events [303]. The phenomenon of
SICS shows a number of interesting features. First, the severity varies
greatly according to the lens-solution combination [304]. Group 2
hydrogel lenses in conjunction with polyhexamethylene biguanide
(PHMB) solutions induce the greatest level of staining. This can be
explained in terms of the preservative uptake-release characteristics;
being highly temperature sensitive, Group 2 materials discharge a
relatively high proportion of solution into the post-lens tear film, thus
increasing the dosage. Second, the staining usually reaches a maximum
two hours after insertion but then disappears after a further two hours
[305]. This is at least partly explained by an associated increase in
shedding of corneal cells with SICS, suggesting a temporary increase in
epithelial apoptosis [306]. SICS may be related to an increase of
dynamin-mediated uptake of fluorescein into cells which in turn is due
to the presence of Tetronic 1107, a block co-polymer included in a
number of multi-purpose solutions [142].

4.1.2. Hypoxia-related responses: microcysts and epithelial oedema


Contact lens-induced hypoxia has inspired a large body of research
and led to many insights into corneal physiology [122,307,308]. In
relation to the corneal epithelium, hypoxia leads to: decreased epithelial
metabolic rate [309,310], reduced basal cell mitosis [311,312],
epithelial cell enlargement and increased cell life [313], epithelial
thinning [309,314,315], lactate accumulation, acidic shift and increased
bacterial binding [316,317]. The main clinical manifestations of this
include: epithelial microcysts, compromise in junctional integrity, neo­
vascularisation and decreased corneal sensation (Section 4.1.6).
Soft contact lens wear has been reported to suppress corneal basal
epithelial cell mitosis in rabbits [312]. Subsequent work showed a cor­
relation between reduced oxygen supply and reduced exfoliation of
epithelial cells with both rigid corneal and soft lens wear [318–320]. A
correlation with the ability of Pseudomonas aeruginosa to bind to exfo­
liated epithelial cells was reported that was hypothesised, but not
proven, to be a factor in increased risk of corneal infection [321].
Fig. 2. Schematic representation of various patterns of corneal fluorescein
Binding of P. aeruginosa to corneal cells was greater with hydrogel lenses
staining. A. ‘Smile’ desiccation staining; B. Desiccation staining; C. 3 & 9 than SiHy lenses [319] but, by contrast, hyper-oxygen permeable rigid
o’clock staining; D. Foreign body tracks; E. Superior arcuate staining (SEAL); F. corneal lenses did not increase P. aeruginosa binding. However, a sub­
Inferior staining from soft lens edge; G. Limbal staining and follicles from sequent study has shown a reversal of this effect over a longer period
thimerosal hypersensitivity; H. Solution-induced corneal staining (SICS); I. Se­ (6–12 months) of wear [322].
vere SICS; J. Microbial keratitis (MK); K. Adenovirus/Epidemic Keratocon­ There is no evidence for corneal epithelial swelling due to anoxia
junctivitis; L. Epithelial microcysts breaking the corneal surface. [323,324] or hypoxia [325,326]. Corneal epithelial oedema is a rare
manifestation of contact lens wear, usually arising from significant
trauma or being exposed to a hypotonic environment. Fluid accumulates
with the corneal shape can result in pressure points. Additional pressure in the intercellular spaces rather than within cells. Increased lactate
from the upper lid and lens motion during the blink produce an abrasion production during hypoxia may accumulate between basal cells and
which is invariably severe enough to warrant a change of lens design. draws water out of the cells by osmosis [327]. Light scattering from
The edges of both rigid corneal and soft lenses, when in contact with these vacuoles of lower refractive index can result in cloudy vision and
the corneal surface, can cause corneal staining of varying degrees. Rigid coloured haloes. Visual phenomena with epithelial oedema are similar
lenses with inadequate edge clearance can produce arcuate staining to those experienced with stromal oedema but occur much earlier, i.e.
close to the limbus. This can be confused with 3 & 9 o’clock desiccation with relatively low levels of oedema. With high levels of epithelial
staining but is differentiated by examining the peripheral lens fit. When oedema, vacuoles may be visible on slit lamp examination and are
the lens is moved close to the limbus, the fluorescein fit should show distinguished from epithelial microcysts by marginal retro illumination;
some peripheral clearance, even if only minimal. Soft lenses whose vacuoles show unreversed illumination [328].
edges encroach onto the peripheral cornea can also result in mild Epithelial microcysts are an indicator of disordered epithelial
staining. In aligning with the ocular surface, soft lenses show the metabolism and in contact lens wearers, occurs as a delayed response to
greatest deformation at the edge, which typically stretches about 4 % high levels of chronic hypoxia [329]. Since they are associated with
causing a band of pressure [299]. In both cases, the remedy is to modify overnight wear or contact lenses of low oxygen transmissibility, they are
the lens fit to avoid corneal contact with the lens edge. rarely seen with daily wear of current SiHy lenses.
Epithelial microcysts are translucent, typically 15–50 μm in diameter
4.1.1.3. Toxicity. Solution-induced corneal staining (SICS) is a common and show reversed illumination, indicating a higher refractive index
side effect of soft lens multipurpose disinfection systems. Unlike other than surrounding tissue [328]. They form in the basal layers, gradually
types of staining, SICS is usually evident in three or more peripheral moving to the corneal surface, at which point they may stain with
corneal sectors. Often the punctate staining follows a circular pattern fluorescein. Non-contact lens wearers can show small numbers (<10) of
but, in more severe cases, can involve the whole of the cornea. Gener­ microcyst but, in lens wearers, larger numbers, even hundreds, may be
ally, the level of corneal staining in SICS is low grade, although more present. They are usually asymptomatic and, except in severe cases,

202
P.B. Morgan et al. Contact Lens and Anterior Eye 44 (2021) 192–219

have no effect on vision. There is an inverse relation between lens ox­ lens, as has been shown in animal models of lens wear [348,349]. A
ygen transmissibility and the number of microcysts [329]. Hydrogel summary of studies which have to date examined the effects of contact
lenses are associated with a greater number of microcysts compared to lens wear on corneal dendritic cell density presented in Table 2 illus­
rigid corneal lenses with similar Dk/t [329]. trates much variability in cell numbers reported by different in­
Their aetiology and morphology is not fully understood but micro­ vestigators who may have used differing methodologies and criteria.
cysts are believed to represent degenerated (apoptotic) cells [330] due However, from the available evidence, it can be surmised that CEDC
to altered mitosis of the basal epithelial cells [329]. A curious feature of numbers increase during lens wear, that this increase is temporary and
microcysts is their time-course; their onset is slow, reaching a peak after that there is an effect of lens type.
several months and, when lens wear is ceased, first increase in number The corneal CEDC response occurs as quickly as after 2 h of hydrogel
before disappearing over a period of 2–3 months. The return of basal cell lens wear in non-wearers [350]. These rapid changes are perhaps not
mitosis to normal levels may accelerate the process of bringing cellular surprising as measurements of DC kinetics in mice show track speeds of
debris to the surface [309]. the order of 3 μm/min [351]. CEDC in the central cornea returned to
baseline levels after 6 months of wear [193], which agrees with findings
4.1.3. Corneal wrinkling of other investigators who found central CEDC density not to be elevated
Corneal Wrinkling is a rare but notable complication of thin hydrogel in experienced lens wearers compared to non-wearers [190,346].
lenses [331,332]. The prevalence has been estimated to be 1.7 % in In contrast, another report found that CEDC density was not signif­
wearers of disposable hydrogel overnight wear lenses [233], whereas icantly higher than baseline until 4 weeks wear of hydrogel lenses and
others have argued that it is limited to those using mis-designed custom had not returned to baseline by 6 months [347]. Only two studies have
lenses and may be unfamiliar to the contemporary contact lens practi­ quantified the distribution of CEDC away from the corneal centre. One
tioner [333]. report showed an increased density of CEDC in the peripheral cornea
Wrinkles in the thin central zone of these lenses are temporarily after 1 week of reusable hydrogel lenses, which had not returned to
transferred to the corneal surface through lid pressure and so wrinkling baseline at 6 months [347]. Another report showed CEDC density in the
is generally considered to have a mechanical aetiology, although an mid-peripheral cornea to be increased in lens wearers [190].
osmotic cause has also been suggested [334]. Clinically, wrinkling can These differences between studies are likely to be due to the lens
result in a reduction in visual acuity and is evident from pooling of type, replacement schedule and use of disinfecting solution, as well as
fluorescein and deformation of keratoscope images. contact-lens dry eye status. When considering only asymptomatic lens
wearers, only a small (not significant) rise in CEDC was found [193].
4.1.4. Corneal warpage Daily replacement of lenses also reduces the CEDC response – no
Throughout the history of contact lenses, there have been many re­ elevation of CEDC occurred at 1 week of wear when hydrogel lenses
ports of corneal warpage, however, these have tended to relate to non- were replaced daily, but CEDC density increased when the same lens was
standard lenses, such as rigid corneal PMMA, opaque tinted, toric and reused and disinfected [192]. The effect of lens material and oxygen
overnight wear contact lenses. Corneal warpage can manifest as: i) an transmissibility remain unclear, with higher, lower and no differences
increase in corneal astigmatism, ii) central irregular astigmatism, iii) reported between SiHy and hydrogel lenses [192,345,346].
loss of radial symmetry and iv) change from normal corneal asphericity.
A review of early case reports (1965–1988) noted that, out of 473 cases, 4.1.6. Sensitivity and nerve changes
343 (72.5 %) were associated with rigid corneal lenses and 130 (27.5 %) Since the earliest introduction of contact lenses, changes in corneal
with hydrogel lenses [335]. Another report found corneal warpage was sensation have been a feature of contact lens wear. Although an optical
associated with the fit of rigid corneal lenses [336]. Several reports benefit is produced by lens wear, the mechanical interaction of the
discussed corneal warpage with early examples of opaque tinted lenses contact lens with the cornea and eyelids can produce a strong foreign
[337,338]. In a more recent study of prospective refractive surgery pa­ body sensation, particularly in rigid corneal lens wear. Thus adaptation
tients [339], contact lens wearers were required to cease lens wear for 5 to the presence of the lens is a desired side-effect for successful rigid
days to 3 weeks depending on lens type; 6.7 % (11/165) patients showed corneal lens wear [21,282,286,352–354]. Adaptation to soft lens wear is
significant corneal warpage. The recovery rates varied with lens type, not needed to the same extent, but discomfort from soft lens wear raises
soft overnight wearing eyes taking the longest (11.6 ± 8.5 weeks) and a different question about how the corneal nerves respond to lens wear
rigid corneal lenses the shortest (8.8 ± 6.8 weeks). and whether a lack of adaptation may be contributing to discomfort
Corneal warpage is a rare occurrence with contemporary high Dk [21].
contact lenses, but remains a possibility with low Dk lenses. Notably, Wearing PMMA or rigid corneal lenses will produce a marked
there is one report of keratoconus being wrongly diagnosed in the case of reduction in corneal sensitivity in the wearer, with the magnitude of the
corneal warpage due to soft lens wear [340] and the correct discrimi­ effect depending on the length of lens wear, both in the short-term (over
nation of these two conditions is still a subject of study [341]. a period of a day) and in the long-term (over a prolonged period of daily
lens wear), up to a maximum effect for that wearer [355–362]. There is
4.1.5. Sub-clinical inflammatory response also reduced sensitivity in wearers of reverse geometry ortho-k lenses
Clinically observed corneal infiltrates and inflammatory events occur [363]. A total loss of sensitivity does not occur, although sensitivity may
in up to 10 % of all soft contact lens wearers per year [342]. However, be sufficiently reduced to increase the risk of an undetected foreign body
sub-clinical inflammation is much more common and possibly ubiqui­ [357,359]. Peripheral corneal sensitivity, not covered by the lens, is
tous when contact lens wear is first commenced (see Section 3.1.4). unaltered [364]. Recovery of sensation after lens wear follows a similar
Changes in the density, morphology and distribution (e.g. migration into effect - a shorter period of lens wear allows for an earlier recovery, with
the corneal centre) of corneal epithelial dendritic cells (CEDC) are full recovery appearing to occur [355,365]. No significant morpholog­
considered pathognomonic of activation of the corneal immune ical changes in corneal nerve structure have been noted with high Dk
response [343] and have been observed in the presence of corneal rigid corneal lens wear [366].
infection, ocular surface disease including dry eye and allergy and Ortho-k lenses, by virtue of being rigid, and also of being worn
during contact lens wear. overnight, produce a reduction in sensitivity, with similar changes
in vivo confocal microscopy investigations have shown elevated related to duration of wear as those seen in standard rigid corneal lenses.
CEDC density in the central cornea in soft contact lens wearers [192,193, Central sensitivity is reduced by approximately 50 % and recovers to
344–347], suggesting these cells migrate into the centre in response to pre-lens wear levels after cessation of lens wear [367–369]. Corneal
an inflammatory stimulus, perhaps due to the presence of the contact nerve morphology is affected by ortho-k, with a reduction in nerve fibre

203
P.B. Morgan et al. Contact Lens and Anterior Eye 44 (2021) 192–219

density and re-organisation of the nerve fibre orientation [368–372]. suggests that no change in sensitivity is likely. This is supported by ev­
The nerve fibre changes also resolve with cessation of ortho-k lens wear, idence that corneal neural structures are unchanged with scleral lens
in sync with the recovery in sensitivity [369,372]. These features wear [366,373]. However, scleral lens materials with an insufficient Dk
strongly indicate a direct link between sensitivity loss and the me­ [374], or with an excessive post-lens tear reservoir [127,128] will pro­
chanical effect of the ortho-k lens on the corneal epithelium during duce oedema, suggesting that either scenario may produce a reduced
overnight lens wear [367]. A metabolic effect from hypoxia under the sensitivity.
lens is not considered to be a factor, since very high Dk lens materials are For low Dk soft hydrogel lenses, both a mild reduction in sensitivity
used and no corneal oedema is observed. [116,362,375,376] and no change in sensitivity has been reported [211,
No studies have reported on corneal sensitivity change in scleral lens 377,378]. For studies that reported a reduction, a full recovery to
wear, but the vaulting of the lens over the cornea, combined with good pre-lens wear levels occurs when lens wear is stopped [375,376]. The
Dk lens materials and an appropriate post-lens fluid reservoir thickness, duration of wear does not appear to produce an increase in effect beyond

Table 2
Summary of studies reporting corneal epithelial dendritic cell density in contact lens wearers. HCL = hard (PMMA) contact lenses; SCL = soft contact lenses; Hy = soft
hydrogel contact lenses; SiHy = silicone hydrogel contact lenses; DD = daily disposable contact lenses; w = week.
Study Subjects Corneal epithelial dendritic cell density (cells/mm2)

Centre Periphery

Zhivov et al 2007 [344] Contact lens HCL: 36 ± 22 HCL: 189 ± 34


wearers SCL: 114 ± 41 SCL: 228 ± 35
(n=54)

Non-wearers 34 ± 3 98 ± 8
(n=70)

Sindt et al 2012 [345] Contact lens All: 64 ± 71


wearers significantly higher than non-wearers
(n=53)

Hy: 47 ± 44
SiHy DD: 69 ± 77
no significant difference between lens types

Non-wearers 29 ± 23

(n=10)

Lopéz-De La Rosa et al 2018 [346] Contact lens Hy: 132 ± 83


wearers significantly higher than SiHy & no wear
(n=40) –
SiHy: 68 ± 77
not significantly different to no wear

Non-wearers 58 ± 20

(n=20)

Alzahrani et al 2017 [193] Contact lens 1w Hy DD: 47 ± 25


wearers significantly higher than no wear
(n=60) –
24w Hy DD: 32 ± 29
not significantly different to no wear

Non-wearers 27 ± 19

(n=23)

Golebiowski et al 2020 [190] Contact lens 20.1 ± 26.3 15.8 ± 13.6


wearers not significantly different to no wear
(n=20)

Non-wearers 18.6 ± 23.9 12.3 ± 10.8


(n=20)

Saliman et al 2020 [192] Contact lens 1w Hy DD: 15.2 ± 7.1


wearers not significantly different to BL
(n=20)

1w Hy: 24.1 ± 11.1
1w SiHy: 22.7 ± 7.8
significantly higher than BL

Baseline 14 ± 7.5
(n=20) –
(experienced wearers)

Liu et al 2020 [347] Contact lens 1w Hy: 32.29 ± 5.47 1w Hy: 53.54 ± 6.84
wearers 4w Hy: 60.52 ± 12.29 4w Hy: 90.42 ± 8.80
(n=20) 12w Hy: 62.71 ± 13.25 12w Hy: 57.81 ± 6.40
[data reported as mean ± SE] 24w Hy: 55.94 ± 10.63 24w Hy: 47.40 ± 4.35
4-24w significantly higher than no wear all significantly higher than no wear

Non-wearers 16.25 ± 2.58


(n=20) 19.06 ± 2.53

204
P.B. Morgan et al. Contact Lens and Anterior Eye 44 (2021) 192–219

the maximum effect for the wearer [362]. Depending on the water mechanisms [401]:
content of the lens and the duration of wearing, some loss in sensitivity is
likely. ● Hypoxic route: the hypoxia itself may trigger the release of vascular
For high Dk soft hydrogel, daily disposable and SiHy lenses, no sig­ endothelial growth factor in an attempt to rescue the oxygen-thriving
nificant loss in sensitivity has been noted using any type of instrument epithelial and endothelial cells.
[211–213,379], although differences in sensory processing have been ● Hypoxia mediated inflammatory route: hypoxia-induced corneal
noted [379,380]. The consequences for this latter finding are unclear, inflammation may signal the release of inflammatory vaso­
but may have a role in contact lens discomfort. No significant changes in stimulating factors in the stroma.
corneal neural architecture have been noted with soft contact lens wear
in the central or mid-peripheral cornea, although the changes may be Corneal limbal vasculature has an insignificant role in corneal
more subtle than those visible with confocal microscopy [213,366, oxygenation. The vascular arcade does not penetrate the normal clear
381–383]. cornea due to barrier function of limbal stem cells among other things.
The different patterns of effect with each lens type illustrate the two However, as a result of the imbalance from hypoxia the limbal vessels
main mechanisms proposed to explain sensitivity loss: sub-lens corneal may penetrate and continue to encroach the stroma towards the corneal
hypoxia and mechanical pressure or rubbing [201,214,384,385]. The apex. The depth of corneal vascularisation may depend on the location
importance of gas exchange through the lens material is demonstrated of the angiogenic site in the cornea hence deeper stromal vascularisation
by the reduction in sensitivity loss with increasing lens Dk and/or tear could be expected with higher degrees of corneal hypoxia. The posterior
exchange [214]. This matches with experimental studies that demon­ oxygen supply to the peripheral endothelial cells from the aqueous
strate a reduced corneal sensitivity with a reduced pre-corneal oxygen could, at least in part, explain the uncommonness of contact lens-
concentration (compared to normal) [386,387]. The exact mechanism induced deep stromal vascularisation. Deeper stromal vascularisation
for this reduction has not been established, but has been linked to an could pose a higher risk of corneal opacification from leaking lipid into
altered corneal metabolism, i.e. the change in gas exchange rate leads to the stroma [399], plus the additional risk of intracorneal haemorrhage
an altered metabolic level within the cornea that produces a new [402].
sensitivity baseline. This change may be moderated by hypoxia and/or Clinically, the state of corneal oxygenation can be gauged from its
hypercapnia [220], as well as altered stromal pH [220], neurotrans­ inverse relation with contact lens-induced corneal swelling. Hypoxia-
mitter [213,388,389] and acetylcholine transferase activity [386,390, induced corneal vascularisation may rarely occur with rigid corneal
391]. Evidence for a hypoxia-related mechanism is shown by the re­ overnight wear lenses, or with PMMA daily wear. This is attributed to
covery in corneal sensitivity in PMMA lens wearers refitted into rigid the smaller diameter of hard lenses hence greater exposure of peripheral
corneal lenses [392] and in low-Dk soft lens wearers refitted into SiHy cornea to oxygen. The risk of corneal vascularisation is increased with
lenses [208,212]. overnight wear, compared to daily wear, of conventional hydrogel
The mechanical effect is revealed by the different findings between lenses [99,403]. The same is true with SiHy lenses. Although, on
the rigid and soft lens material, each of which delivers significantly average, the risk of corneal vascularisation has been minimised with
different stimulation to the corneal nerves. The mechanical interaction greater oxygen availability in SiHy lenses [125,404,405], a cautious
of the lens with the corneal surface produces a sustained response from approach to identify and monitor high-swellers is still suggested
the corneal nerves (and thus symptoms of discomfort in the lens wearer), [406–408]. This is to prevent potentially sight-threatening outcomes
which gradually reduces with neural adaptation [393]. This adaptation from silent vessel ingrowth, as well as to lower the risk of possible in­
may be at the receptor level or at a higher level [393]. flammatory events associated with vascularisation in these patients
[409]. This is particularly important in lenses with higher minus power,
4.2. Stroma thicker lens edge profile and especially in SiHy overnight wear [407,
408,410].
4.2.1. Neovascularisation
The limbal vasculature does not provide the required nutrition and 4.2.2. Hypoxia-related responses: swelling, striae, folds, haze
oxygen for corneal metabolism [394]. The cornea receives the required Hypoxia-induced corneal swelling mainly occurs in the stroma which
oxygen for its respiration from the anterior surface through diffusion of accounts for around 90 % of corneal thickness [411]. The stroma plays
atmospheric oxygen that is dissolved in the tear film. The required an important role in maintaining corneal transparency because of its
nutrition (e.g. glucose and amino acids) for the avascular cornea diffuse uniquely organised lamella structure of collagen fibrils [412] with
from the aqueous humour across the leaky corneal endothelium [395, anterior stromal keratocytes contain a crystalline protein to minimise
396]. backscattering of the light [413]. Collagen fibrils in the anterior stroma
Peripheral corneal oxygen deprivation by contact lens wear may lead are more tightly packed than the posterior stroma to provide adequate
to limbal hyperaemia that is inversely related to peripheral lens oxygen rigidity and strength for maintaining the anterior corneal curvature and
transmissibility (Dk/t) [123,397]. The increased limbal hyperaemia by shape against the stress from stromal swelling [414].
contact lens wear could be a precursor to corneal neovascularisation. In open-eye conditions at sea level the bare cornea is exposed to the
However, a direct link between limbal hyperaemia and the neovascular atmospheric partial oxygen pressure (PO2) of 155 mmHg [415]. The PO2
ingrowth has not been established [307]. The absence of vessels in the is reduced to one third or ~55 mmHg that is largely supplied by the
cornea is a major factor in maintaining its transparency. The loss of palpebral conjunctival blood vessels in closed eye conditions [415]. A
corneal transparency by progressive growth of invading new vessels in contact lens barrier will reduce the available PO2 leading to increased
the cornea could be sight threatening [398]. A suggested mechanism for anaerobic metabolism of glucose [416,417] in corneal epithelial cells.
maintaining corneal avascularity is through an active balance between Lactic acid, the product of the anaerobic metabolism, diffuses posteri­
corneal angiogenic and antiangiogenic factors [399]. Peripheral corneal orly into the stroma. The increased osmotic pressure caused by elevated
swelling can facilitate limbal vessel penetration into the cornea but the levels of lactic acid production from anaerobic corneal metabolism leads
presence of stromal swelling by itself is not a sufficient trigger to the to stromal swelling [418–420]. This is because the hydrophilic compo­
growth of new vessels. Contact lens-induced corneal neovascularisation nents of the stromal ground substance will expand by diffusion of
may be triggered by corneal hypoxia through down-regulating anti-­ additional water from the anterior chamber, across the leaky corneal
angiogenic factors and up-regulating angiogenic factors [399,400]. endothelium by osmosis [421]. This phenomenon can physiologically
However, corneal hypoxia may affect the balance between corneal occur when eyelids are closed during sleep (overnight corneal swelling)
angiogenic and antiangiogenic factors through two different and is in the range of 3–4 % of corneal thickening [422–424]. Typically,

205
P.B. Morgan et al. Contact Lens and Anterior Eye 44 (2021) 192–219

the cornea is at its maximum thickness upon waking in the morning and [457]. Striae and folds gradually disappear as a result of stromal
it gradually recovers to its daytime thickness within a few hours after deswelling after removal of hypoxic stress. Striae are clinically seen as
eye opening [422,423,425–429]. A contact lens barrier can also induce vertical lines in the posterior stroma because of increased fluid separa­
corneal swelling. There is a direct linear relationship between corneal tion of the posterior stromal fibrils due to posterior stromal swelling.
hydration and thickness [430,431]. Therefore, corneal swelling is often Folds are seen in the posterior corneal surface as a result of buckling/­
used to determine the level of oxygen supply through a contact lens flattening of the posterior limiting lamina from further increase in the
[407,432]. posterior stromal swelling as the stroma cannot expand laterally. Striae
Assuming an average of 4 % overnight central corneal swelling and folds of the swollen posterior stroma are reported to be visible in
without lens wear in their original study, a minimum Dk/t criterion of different angular directions, including horizontally, using confocal mi­
87 Dk/t units was established to avoid lens-induced overnight corneal croscopy [458]. Results from a clinical study suggested significantly less
swelling in overnight wear [433]. Although a higher lens trans­ flattening of the posterior corneal surface with a SiHy lens (lotrafilcon A)
missibility requirement of at least 125 Dk/t units was reported after compared to a hydrogel lens (etafilcon A) after one week of overnight
updating the assumption of no-lens overnight swelling to 3 % [434]. wear [459]. There was less posterior corneal flattening with the
However, based on clinical studies with SiHy lenses, even at higher lens hydrogel lens in their study compared to an initial report from another
Dk/t levels of 175 [428] and 211 [410] Dk/t units, closed-eye lens-in­ group who examined 3 h closed-eye wear of a thick hydrogel and a
duced corneal swelling could not be eliminated. PMMA contact lens, respectively [460]. This is in line with lower levels
It was also suggested that 24 Dk/t units was needed to prevent of corneal swelling in their study because they used lenses of higher Dk/t
average central corneal swelling in daily wear [433]. This criterion was in overnight wear and examined corneal swelling and posterior curva­
re-evaluated in a more recent study and determined to be 19.8 and 32.6 ture later in the day (instead of immediately after eye opening in the
Dk/t units, respectively, to avoid average central and peripheral corneal initial 3 h closed-eye study). Although a more recent open-eye study
swelling in soft lens daily wear [435]. The results of current short-term [461] measured posterior corneal steepening with a hydrogel toric lens
studies suggest expecting an average minimal ocular physiological in daily wear this was attributed to the regional pattern of corneal
impact in open-eye wear with a minimum central Dk/t ~25 units and swelling in this study showing higher swelling in the corneal periphery
peripheral Dk/t of ~11 units [436,437]. It is noteworthy that the sug­ than the centre because of lower oxygen transmissibility in the thicker
gested minimum Dk/t values for closed or open-eye lens wear can only peripheral stabilization zones of the lens. Stromal striae and folds are
predict the population average corneal swelling values and are inher­ less frequently expected with daily wear of hydrogel or with overnight
ently incapable of reflecting individual Dk/t requirements in practice wear of SiHy lenses. However, they can still occur in those with high
[408]. levels of swelling [408]. Stromal striae were 4 times less frequent with
There is a greater degree of swelling in the posterior cornea [438] overnight wear of a SiHy lens (balafilcon A) compared to overnight wear
compared to its anterior region [439]. Differential corneal swelling is of a hydrogel lens (etafilcon A) in a 12-month study [462].
supported by the relevant physiological [440,441], physical [439,442,
443] and ultrastructural [414,444–446] differences between stromal 4.2.3. Thinning
anterior and posterior properties. This is in line with the reported [438] Early studies revealed an asymptomatic progressive corneal thinning
higher water content of the mammalian posterior than anterior stroma phenomenon with overnight wear of hydrogel lenses [463,464]. A
[447] attributed to higher ratio [448] of keratan sulphate, a more hy­ landmark 5-year study [309] of unilateral soft lens overnight wear
drophilic glycosaminoglycan [449], in the posterior stroma compared to showed that, 7 days after the discontinuation of lens wear (recovery
higher ratio of dermatan sulphate, a less hydrophilic glycosaminoglycan period of the chronic swelling), the average stroma was ~11 microns or
in the anterior stroma. In this process, stromal collagen fibrils do not 2.3 % thinner than the baseline (~2 microns thinning/year). This
have the capability to absorb water. They are only being more separated finding was attributed to the possible effects of the chronic stromal
because of the expansion of their surrounding ground substance in direct swelling on morphology and/or the function of stromal keratocytes
relation to the hypoxia. This can lead to increased light scattering that thereby reducing their ability for synthesis of the stromal components
may affect corneal transparency. and/or to the possible effects of hypoxia-induced acidosis on breaking
Corneal transparency depends on the precise ultrastructure of the down the components of the stromal grand substance [309]. However,
corneal stroma, which remains steady under aerobic conditions [446, similar progressive thinning effects were also reported in daily wear of
450]. In the presence of sufficient oxygen, the corneal stroma can stay at hydrogel and rigid corneal lenses, as well as in overnight wear of SiHy
a constant level of 78 % hydration (deturgescence), ensuring trans­ lenses [465,466].
parency [451,452]. Stromal light scatter occurs because of increased A novel study [467] using confocal microscopy investigated the ef­
distances and decreased uniformity of the collagen fibrils in the fect of 6-month overnight wear on the stromal keratocyte density in
oedematous stroma [412]. Intense light scattering was reported with neophytes where subjects wore a hydrogel lens (etafilcon A) in one eye
slight swelling of the anterior stroma, but relatively slight light scat­ and a SiHy lens (balafilcon A) in the other eye. They found that the
tering has been observed with a higher degree of posterior stromal posterior stromal keratocyte density was reduced in both eyes equally,
swelling [442]. The stromal haze may occur as a result of an increased independent of lens oxygen transmissibility [467]. Therefore, they
amount of back scattered light from the anterior stroma [453]. There­ concluded that the reduction of the keratocyte density in overnight wear
fore, the haze is more likely to occur with higher levels of corneal was not dependent on lens-induced hypoxia and/or swelling [467].
swelling (>15 %), for example after closed-eye wear of a low Dk/t lens Based on these results they suggested a mechanical aetiology for the loss
and/or in high-swellers. However, haze is more commonly seen in of keratocytes from the physical presence of the lens on the cornea
corneal pathological conditions (e.g. Fuchs’ dystrophy). Corneal haze [467].
can clinically be measured with customised slit-lamp biomicroscopy, In a further investigation in this regard, a study [468] found that, in a
confocal microscopy or using a Scheimpflug device [453]. group of neophyte subjects, keratocyte densities were lower in the SiHy
Stromal striae and folds are two important short-term clinical indices and rigid corneal lens-wearing eyes compared to their contralateral
of corneal swelling that are more commonly seen with low Dk/t controls, despite similar levels of anoxia-induced corneal swelling for 2 h
hydrogel lenses and especially after closed-eye lens wear [454,455]. in the lens-wearing and control eyes. They also found higher levels of
Striae and folds are reported at swelling levels as low as ~4 and ~7 %, tear inflammatory markers after rubbing the no-lens eyes compared to
respectively [456]. A useful clinical tool for estimating the magnitude of their contralateral controls in the final part of their experiment. These
corneal swelling was devised using the correlation between the number findings led them to hypothesise that “the mechanical stimulation of the
of striae or folds to the level of the observed corneal swelling in the study corneal surface, due to the physical presence of a contact lens, induces

206
P.B. Morgan et al. Contact Lens and Anterior Eye 44 (2021) 192–219

the release of inflammatory mediators that cause keratocyte dysgenesis The swelling rapidly decreases upon eye opening. In contact lens wear,
or apoptosis” [468]. this physiological effect can be artificially-induced with both rigid
A 12-month study of 30 days of overnight wear [469] found no corneal and soft contact lenses of low Dk/t, producing central corneal
significant differences in corneal thickness and in overall stromal kera­ oedema [482–485]. The low Dk/t also creates hypoxic stress to the
tocyte density among the SiHy group, the rigid corneal group and endothelial cells, promoting increased cell death as a result of the
non-wearers. However, some inflammatory markers in the tear film of chronic reduction in oxygen supply to the endothelium, in combination
the lens-wearing groups were higher than in the control group; specif­ with the effect of contact lens-induced pH changes [486]. The cell death
ically, higher concentrations of epidermal growth factor were found in leads to a reduced endothelial cell density and increased polymegethism
wearers of both SiHy and rigid corneal lens wearers compared to and pleomorphism [487,488]. An increased effect is observed with
non-wearing controls whereas interleukin-8 was shown to be increased increasing lens wear experience [489]; a short lens wear experience does
in rigid corneal lens wearers only. not produce a significant effect.
A more recent 6-month daily wear study found a greater decrease in With modern lens materials of higher Dk/t, these effects are no
the overall keratocyte density in hydrogel than in rigid corneal lens longer a common feature of contact lens wear [490–493]. However,
wearers [470]. Also, the decrease found in each group was higher than when low Dk/t lenses must be used, careful monitoring of the corneal
their neophyte counterparts. They attributed the higher loss of kerato­ endothelium should be made.
cytes in the hydrogel group to a contribution from higher stromal hyp­
oxic stress in hydrogel compared to rigid corneal lens wear. However, 4.3.2. Bedewing
they found no changes in the corneal thickness of the neophyte Clusters of oedematous droplets or leucocytes deposited on the sur­
lens-wearing groups during the 6- month study period. face of the corneal endothelium were first reported in chronically
Current knowledge on the aetiology and mechanisms of the stromal intolerant PMMA contact lens wearers in 1979 [494]. They proposed
thinning is still inconclusive and requires further research. Contact lens- that the droplets were composed of inflammatory cells whose release
induced stromal thinning may affect the integrity of the cornea in was stimulated by hypoxic stress caused by over-wear of low Dk/t len­
extreme cases. Also, progressive loss of corneal thickness in long-term ses. The ‘precipitates’ were observed on the endothelium in the area of
contact lens wearers could be a point of concern for suitability for the inferior pupil margin at high magnification and the phenomenon
future refractive surgery. was termed ‘endothelial bedewing’. The authors noted that management
with low Dk/t soft lenses had mixed results. However, endothelial
4.3. Endothelium bedewing has been reported in 20 % of 70 non-contact lens wearers,
which supports much earlier work that bedewing is a physiological
4.3.1. Polymegethism and pleomorphism feature [495]. The authors argued that the need to use slit-lamp mar­
The endothelium is the most posterior layer of the cornea and is ginal retro-illumination to view the bedewing (indicating that the
formed by a monolayer of differentiated epithelial cells. The primary droplets had a higher refractive index than the surrounding aqueous
role of the endothelium is as part of the homeostatic mechanism of humour) supported the theory that the droplets contained inflammatory
maintaining a balanced water content in the corneal stroma, which cells.
ensures corneal transparency [205]. Metabolic ion pumps within the Bedewing is certainly a physiological phenomenon in a non-lens
cells promote a shift in solute concentration between the corneal stroma wearing eye and may be associated with hypoxic stress in low Dk/t
and the anterior chamber and water moves along the concentration lens wear. The main associated symptom is fogging of vision, which may
gradient in response and out of the stroma [471]. be sufficient to reduce lens wear time or cease lens wear. Best treatment
At birth, the cells are evenly distributed across the posterior corneal when observed in low Dk/t lens wearers is to re-fit with a high Dk/t
surface and take a polyhedral shape that is most efficient in achieving contact lens material to avoid hypoxic stress, should that be the causa­
full coverage [205]. This produces a typical hexagonal cell shape, which tive factor. However, even so, symptoms and endothelial bedewing may
is found in 70–80 % of cells. Endothelial cell density averages 3100 continue.
(2700–3500) cells/mm2 in children and reduces with age to approxi­ The presence of inflammatory cells in the anterior chamber is always
mately 2200 (1000–3000) cells/mm2 by the age of 80 years (a reduction a sign of possible inflammation, which may be of much more severe
rate of c.0.25 % / year) [472–475]. A minimum endothelial cell density aetiology and consequence. Contact lens ECPs should take care to
of 400–700 cells/mm2 is thought to be necessary to maintain adequate investigate and exclude other causes for the presence of inflammatory
endothelial function and corneal transparency, but an endothelial cell cells [496].
density between 1000–2000 cells/mm2 is susceptible to corneal
decompensation [476]. The gradual reduction in endothelial cell density 4.3.3. Blebs
is due to physiological attrition in the cell numbers and to the absence of The term endothelial blebs is used to describe apparent holes
cell replication, an effect thought to be due to contact inhibition be­ observed in the regular mosaic of the corneal endothelium. They were
tween the cells [477,478]. This contact inhibition also encourages cells first described in 1977 [497] and further explained by the confocal
to spread to maintain contact with neighbouring cells, especially microscopy work of Efron. The authors proposed that the apparent
following death of a neighbouring cell. It is important to compare any absence of endothelial cells was due to oedematous swelling of the cell
changes observed from contact lens wear with these physiological causing light, reflected from the ‘bulging’ posterior surface of the
changes that occur with age. affected endothelial cell, to be directed along a divergent path and away
As the cells decrease in number, the remaining cells spread out to from the axis of observation [498]. They can occur on any area of the
cover the gaps. Polymegethism describes the appearance of the corneal cornea covered by the contact lens [499].
endothelium where the usual consistency of cell size is lost. At the same Blebs are visible within minutes of contact lens application, with
time, the cells lose their hexagonal regularity. This is called pleomor­ their numbers peaking after 30 min and diminishing over the following
phism, which describes the increase in cell shape variation [479]. Both hours [487,497,500,501]. The phenomenon has been linked to localised
phenomena have been shown to increase with age [480]. areas of endothelial swelling from acidosis caused by hypercapnia
As a metabolically-active cell layer, the endothelium is affected by (increased carbon dioxide) and hypoxia (reduced oxygen supply),
any reduction in available oxygen supplied through the cornea from the resulting from altered gas diffusion through the cornea [502]. This
atmosphere [415,481]. This happens physiologically with eye closure impaired diffusion is particularly a feature of low Dk/t contact lens wear
during sleep, resulting in a small amount of overnight corneal swelling [500,501] and can be provoked by exposing the corneal surface to a
(oedema) due to the impaired oxygen supply to the endothelium [429]. reduced O2 partial pressure to simulate hypercapnia or anoxia [502].

207
P.B. Morgan et al. Contact Lens and Anterior Eye 44 (2021) 192–219

Scleral lens wearers fitted with a large post-lens tear reservoir (400 μm measured by time-domain optical coherence tomography of both the
vs 200 μm) also develop blebs, due to hypoxia at the corneal surface central and paracentral epithelium after a single night of hyperopic
[503]. Even with moderate to high Dk/t SiHy lenses, endothelial blebs ortho-k lens wear, with greater central thickening with higher targeted
can form after one hour of eye closure [504]. In a closed eye situation, refractive change (+3.50D vs +1.50D). Instead, another study [518]
the bleb response has been reported as being greater in Asian vs. used optical pachymetry after 1 and 4 nights of ortho-k lens wear to
non-Asian eyes when wearing a low Dk/t soft contact lens [505]. reveal central and paracentral stromal thickening consistent with the
Endothelial blebs should not be confused with endothelial guttae, expected overnight wear oedema response. However, when measure­
which are generally larger, are found at the central cornea and can be ments were repeated 8 h later to allow overnight oedema to subside,
associated with corneal disease, such as Fuchs’ dystrophy [478,506, only paracentral epithelial thinning was observed, leading the authors to
507]. Nor should they be confused with Hassall-Henle Bodies, which, conclude that the anterior corneal steepening profile induced by hy­
although benign, are found in the corneal periphery and are associated peropic ortho-k was limited to change to the paracentral corneal
with aging [205,507]. epithelium thickness alone. While it is not known whether changes to
Endothelial bleb formation may be occurring more frequently than corneal thickness induced by longer term wear of hyperopic ortho-k will
expected, particularly in new contact lens wearers. Experienced lens return to baseline after cessation of lens wear, it is reasonable to expect
wearers do not show the same incidence level, but with additional that this would occur as in myopic ortho-k [522], given that it is only
interference in oxygen supply, perhaps with prolonged eyelid closure, alterations in posterior ortho-k lens curvature that differentiate the two
bleb formation may be promoted. However, there is no known clinical approaches and that corneal topography changes induced by 7 nights of
significance and no lasting negative consequence to corneal health overnight hyperopic ortho-k lens wear in presbyopes return to baseline
[508]. The best clinical treatment is to use higher Dk/t materials and to within 1 week of cessation [525].
encourage lens wearers to avoid sleeping in their lenses.
5. Ocular growth modification with contact lenses
4.4. Intentional changes to corneal thickness with orthokeratology
5.1. Changes to axial length
In modern ortho-k, rigid contact lenses are fitted to deliberately alter
corneal curvature by a targeted amount during sleep to correct ame­ Contact lens wear can have a marked impact on the axial growth of
tropia on lens removal. The anterior corneal surface flattens in response the eye and there is increasing interest in slowing ocular growth in
to wear of ortho-k lenses designed to correct myopia [509–511] and children with contact lenses [526]. This is because there is mounting
steepens in response to wear of ortho-k lenses designed to correct hy­ evidence for the increasing frequency of myopia in many populations
peropia [512,513]. While small change to posterior corneal curvature around the world and across ethnicities. By 2050, it is predicted that
[514] and asphericity [515] have been reported over the short term in almost half of the world’s population - five billion people - will be
response to ortho-k for correcting myopia, over the longer term posterior myopic, with nearly one billion high myopes at serious risk of
corneal curvature has been shown to remain unchanged [516,517]. myopia-related ocular pathology [527]. While high myopia is strongly
Instead the corneal thickness profile alters to compensate the change to linked to higher risk of cataract, retinal detachment and myopic mac­
anterior corneal curvature, with the central cornea thinning and para­ ulopathy, even lower levels of myopia are associated with increased
central cornea thickening in myopic ortho-k and the opposite profile of life-long risk of pathology compared to emmetropia [528]. Already,
central thickening and paracentral thinning in hyperopic ortho-k [518]. increasing rates of vision impairment and blindness in the working age
The changes to corneal thickness profile from myopic ortho-k, when population, due to myopic maculopathy, are evident in Asian countries
applied to Munnerlyn’s formula [519] used to calculate ablation depth [529,530].
in laser refractive surgery, accounts for the degree of refractive change With recent understanding that even reducing final myopia by 1D
that is achieved [510,520] and closely to that reported for hyperopic over a lifetime reduces risk of myopic maculopathy by 40 % [531], there
ortho-k [518]. This affirms evidence that ortho-k lens wear only in­ is much scientific interest in slowing the progression of myopia in
fluences anterior corneal curvature rather than bending the overall children. Recently analyses have indicated that axial length bears a
cornea. strong relationship to risk of life-long vision impairment, with an axial
Using the optical pachometer, one study [521] reported an increase length of greater than 26 mm associated with a likely risk of vision
to central stromal thickness immediately after lens removal following impairment across a lifetime of at least 25 % and over 90 % for eyes
overnight wear of both standard rigid corneal lenses and ortho-k, longer than 30 mm [532]. Axial length is consistently correlated with
consistent with that expected from closed eye wear contact lens myopic refraction, although the refraction-to-axial-length ratio isn’t
induced hypoxia [433]. Less central thickening was reported in the consistent across age groups or with increasing axial length [533].
ortho-k lens compared to standard rigid corneal lens wearing eyes, Despite this, due to the repeatability and incremental measurement
however, leading the authors to suggest that myopic ortho-k suppresses possible with axial length compared to myopic refraction, the former has
the central corneal oedema response from overnight rigid corneal lens become the gold standard in assessing treatment efficacy for myopia
wear. Once overnight oedema has subsided there is general agreement control in scientific studies [534]. Contact lens influence on axial length
that the central corneal thinning response from myopic ortho-k is pre­ is therefore the focus in this review.
dominantly due to thinning of the central corneal epithelium [520,522, ‘Myopia control’ is the terminology for any intervention which re­
523], however there is disagreement on which structure influences the duces the axial and refractive progression of childhood myopia, by
reported paracentral thickening from myopic ortho-k lens wear, possibly whatever mechanism and level of efficacy compared to a control [534].
due to limitations of the instrument being used. Whilst one study [520] Optical interventions for myopia control include both rigid corneal and
reported paracentral stromal thickening alone using an optical pach­ soft contact lens modalities. The appeal of an optical intervention is its
ometer, another report [522] instead paracentral epithelial thickening ability to both correct myopic ametropia as well as potentially slow its
using time-domain optical coherence tomography. Another study [523] progression as a monotherapy and thus far contact lens interventions
reported thickening of both the paracentral stroma and epithelium using have performed with more consistent and statistically significant effi­
spectral-domain optical coherence tomography. Central and paracentral cacy than spectacle lens interventions [535].
corneal thickness returns to baseline values within 3 days of dis­
continuing myopic ortho-k lens wear [522]. 5.2. Single-vision contact lenses and myopia control
Only short-term changes to corneal thickness have been reported for
hyperopic ortho-k. One study [524] reported increased thickening Studies investigating the effects of single-vision, full correction rigid

208
P.B. Morgan et al. Contact Lens and Anterior Eye 44 (2021) 192–219

corneal lenses and soft contact lenses on the progression of myopia have one eye and overnight ortho-k worn on the contralateral eye demon­
demonstrated no statistically significant efficacy [536–539]. In deter­ strated a complete halt in myopic progression in the ortho-k treated eye
mining this lack of effect, rigid corneal lenses and soft contact lenses for both six-month phases [542]. Two meta-analyses of ortho-k studies
have been compared to each other and to single-vision spectacle concurred in examining seven controlled trials over two years duration,
corrections. concluding that ortho-k reduced axial elongation by 0.27 mm over two
Standard-design rigid corneal lenses were first purported to slow years (95 % CI 0.22 to 0.32 mm) [547,548].
myopia progression in the mid-1970’s [540] with subsequent studies in The longest ortho-k intervention data reported is a retrospective
the last 20 years both supporting [541] and refuting [537] this pre­ study of 203 eyes from 66 ortho-k wearers and 36 spectacle wearing
sumption. These studies suffered methodological limitations, most controls aged 7–17 years at baseline and with a mean follow up time of
notably very high drop-out rates of almost 50 %. Managing this with a 6.32 ± 0.15 years. Across the first two years, the ortho-k group pro­
run-in adaptation period to rigid corneal lenses, the axial and refractive gressed 0.17 ± 0.02D while the control group progressed 0.52 ± 0.03D.
myopia progression in 116 single-vision distance-corrected children, This statistically significant difference was maintained from year-2-to-4
aged 8–11 years, randomised to wear either rigid corneal lenses or soft and year-4-to-6 of follow up, albeit reducing from a 0.35D difference in
contact lenses was compared. Over the three year study, the rigid the first two years to a 0.19D difference in year-4-to-6. From year-6-to-8
corneal lens wearers’ myopia progressed by around 0.50D less, but the of follow up, there was no statistically significant difference between
soft contact lens wearers showed steepening of the steep corneal me­ groups [549].
ridian by 0.88 ± 0.57D compared to 0.62 ± 0.60D in the rigid corneal
lens wearers. With this different corneal steepening accounting for about 5.3.1. Orthokeratology influence on ocular component growth other than
half of the refractive effect and no statistically significant difference in axial length
axial elongation, both single-vision contact lens correction types were Vitreous chamber depth changes in step with axial length changes in
indicated as similar from a myopia control perspective [538]. ortho-k intervention studies, with a similar significant reduction in
A comparison of single vision distance soft contact lenses with growth compared to historical controls [545,546]. Very similar vitreous
spectacles for their differential myopia control effect, in 484 children chamber depth and axial length changes were also found in another
aged 8–11 years, debunked the concern of ‘myopic creep’ (increasing study, however no comment was made on the contribution of vitreous
myopia) in soft contact lens wearers. Three years of full-time childhood chamber depth to axial length changes [545]. Vitreous chamber depth
wear revealed no difference in either axial or refractive myopia pro­ changes accounted for around 60 % of axial length changes and by
gression between the groups. There was no measured change in steep eliminating the effects of change in anterior chamber depth through
corneal meridian over time or difference between groups [539]. vitreous chamber depth quantification, this confirmed the slowing of
Subsequent contact lens myopia control studies have typically eye growth with ortho-k wear [546]. The latter authors measured
employed single-vision distance soft contact lenses or spectacles as the additional ocular component changes in their study, finding that change
control group. Only one ortho-k study has utilised a daily wear rigid in anterior chamber depth over time was not statistically significant in
corneal lens control, in a crossover design [542]. It is now readily ortho-k wearers, but a small increase of 0.06 mm per year in soft contact
accepted by both scientific and clinical consensus that single-vision lens wearers was noted. Crystalline lens thickness changes were not
distance corrections, of any form, do not reduce axial or refractive different between the groups [546]. Ortho-k can also impact corneal
myopia progression in children [543,544]. nerves as detailed above in Section 4.1.6.
The sum of the available literature indicates that ortho-k consistently
5.3. Impact of rigid corneal lenses on axial length and ocular growth in reduces myopia progression by around 45 % compared to single vision
myopia control spectacle and contact lens corrections over several two-year studies,
with a statistically significant myopia control effect likely maintained
The evidence for overnight ortho-k rigid corneal lens wear to reduce over several years of wear.
the excessive axial elongation seen in childhood myopia is relatively
new. Reports from 2005 [545] and 2009 [546] reported 47 % and 55 % 5.4. Impact of soft contact lenses on axial length and ocular growth in
less axial elongation over 2 years of wear when compared to historical myopia control
controls. Several other two-year controlled trials have taken place since,
the results of which are summarised in Table 3. A novel cross-over study While ortho-k rigid corneal lenses currently enjoy the largest scien­
design involving a daily wear, alignment fit rigid corneal lenses worn in tific volume of evidence for myopia control efficacy [547,548], the

Table 3
Summary of results for key myopia control intervention studies evaluating ortho-k. Adjusted means are presented with (standard deviations) as detailed in each paper.
N = total participants at final analysis visit; Ortho-k = orthokeratology; SV = single vision; SCL = soft contact lens. Studies 1 and 2 measured axial length using A-scan
ultrasound; studies 3-7 employed interferometry measurement (IOL Master in all cases).
Axial length change (mm) Efficacy for reducing axial
elongation

Intervention study Control intervention N (total) Duration (years) Ortho-k wearing group Control group Per year (mm) By%

Cho et al. 2005 [545] SV spectacles (historical) 70 2 0.29 (0.27) 0.54 (0.27) 0.13 46
Walline et al. 2009 [546] SV SCLs (historical) 56 2 0.25 (0.19) 0.57 (0.23) 0.16 56
Kakita et al. 2011 [550] SV spectacles 92 2 0.39 (0.27) 0.61 (0.24) 0.11 36
Cho & Cheung 2012 [551] SV spectacles 78 2 0.36 (0.24) 0.63 (0.26) 0.14 43
Santodomingo 2012 [552] SV spectacles 53 2 0.47 (0.23) 0.69 (0.27) 0.11 32
Charm 2013 [553] SV spectacles 28 2 0.19 (0.21) 0.51 (0.32) 63
Chen 2013 [554] SV spectacles 58 2 0.31 (0.27) 0.64 (0.31) 52

META-ANALYSES Absolute difference in progression between treatment


and control groups (mm)

Sun et al. 2015 [547] 7 studies 2 0.27 (95 % CI 0.22, 0.32) 0.14 45
Si et al. 2015 [548] 7 studies 2 0.26 (95 % CI 0.21, 0.31) 0.13 45

209
P.B. Morgan et al. Contact Lens and Anterior Eye 44 (2021) 192–219

largest volume of recent innovations in contact lens myopia control are Lens thickness did not change; anterior chamber depth increased in the
in various designs of soft multifocal designs. The terminology ‘multi­ multifocal soft contact lenses group, but they were statistically different
focal’ is here used to denote any ‘simultaneous image’ optical design at baseline and similar at the end of the study [558]. A novel design
where dioptric power varies either smoothly or discontinuously with multifocal soft contact lenses did not produce a statistically significant
zonal radius [555]. alteration in anterior chamber depth, crystalline lens thickness or vit­
Studies investigating bifocal and multifocal spectacles for myopia reous chamber depth [564]. Another study echoed this result with no
control in children showed limited success in the late 20th and early 21 statistically significant changes in corneal curvature or anterior chamber
st century [535]. The first publications investigating multifocal soft depth in their study [565]. Taken in sum, these appear similar to ortho-k
contact lenses for myopia control employed designs originally designed results in that generally vitreous chamber depth and axial length are the
for presbyopia. An abstract [556] and a twin study case report [557] key components altered in myopia control.
investigated a commercially available multi-zone simultaneous vision
design, being distance centred with alternating near and distance zones.
5.5. Potential mechanisms of contact lens myopia control
The results indicated a halting of myopia progression in the twin case
report and a reduction of progression by around 2/3rds in the abstract of
The mechanism of how ortho-k reduces the rate of axial elongation in
a one-year randomised clinical trial. Another commercially-available,
childhood progressive myopia is not known. The main theory involves
distance-centred, continuous aspheric design was demonstrated to
peripheral retinal input, involving differential optical stimulus between
reduce refractive progression by 50 % and axial elongation by 29 %
localised peripheral and central retinal signals creating a ‘slow down’
compared to single vision soft contact lenses in a two year study [558]. A
signal for retinal growth, as has been shown in numerous animal models.
prospective, randomized controlled trial on this same lens design
However there is conjecture that shifts in relative peripheral refraction
revealed a 36 % reduction of axial elongation in wear of a +2.50 Add,
correlate with myopia progression in humans [570,571]. Ortho-k and
with no significant effect of the +1.50 Add, both in comparison to a
multifocal soft contact lenses both shift relative peripheral refraction
single vision soft contact lens control [559].
towards more myopia – although not equivalently [572] – and both
Clinical trial results for the two first myopia-control specific designed
show statistically significant myopia control efficacy. It has also been
soft contact lenses were reported in 2011. The first, a dual-focus alter­
demonstrated that when viewing a near target in multifocal soft contact
nating design, showed promise in a cross-over trial of two 10-month
lenses wear and foveal hyperopic defocus is experienced, peripheral
phases in 2011 [560]. The second [561] is reported in Table 4, along
defocus instead tended towards emmetropia [573].
with several more controlled trials of at least one-year duration which
Similarly, on-axis simultaneous defocus occurs through the depth of
quantified axial length. Most of these studies employed novel multifocal
focus created by ortho-k or multifocal soft contact lenses, due to pro­
soft contact lenses designs rather than commercially-available presby­
nounced shifts in spherical aberration [574,575]. Changing spherical
opic designs.
aberration in turn changes accommodative demand at near [576,577]
and a single study for each of ortho-k [578] and multifocal soft contact
5.4.1. Multifocal contact lens influence on ocular component growth other
lenses [579] have indicated a relationship between improved accom­
than axial length
modative response and a better myopia control effect in myopia control
In studies where other ocular biometry measures were taken, there
contact lens wear.
are variable outcomes. A reduction in vitreous chamber depth growth
Future research in myopia controlling contact lenses will likely
was observed in multifocal soft contact lenses wearers, compared to a
further explore these mechanisms and their specific influence in indi­
control group, that was similar in magnitude to the axial length change.
vidual myopes, ideally leading to improved efficacy across all wearers.

Table 4
Summary of results for myopia control intervention studies evaluating multifocal soft contact lens designs. Adjusted means are presented with (standard deviations) as
detailed in each paper. N = total participants at final analysis visit; MFCL = multifocal soft contact lens; SV = single vision; SCL = soft contact lens. Studies 2 and 4
measured axial length using A-scan ultrasound; the rest employed interferometry measurement (IOL Master in all cases except study 9 which employed the LenStar
900). Studies 2, 6 and 10 employed a commercially-available presbyopic MFCL design, while the rest employed myopia control specific designs.
Axial length change (mm) Efficacy for reducing
axial elongation

Intervention study Control intervention N Duration MFCL wearing group Control Per year By%
(total) (years) group (mm)

1 Sankaridurg et al. 2011 [561] SV spectacles (non- 82 1 0.24 (0.17) 0.39 (0.19) 0.15 38
concurrent)
2 Walline et al. 2013 [558] SV SCLs (historical) 54 2 0.29 (0.03) 0.41 (0.03) 0.06 29
3 Lam et al. 2014 [562] SV SCLs 128 2 0.25 (0.23) 0.37 (0.24) 0.06 32
4 Fujikado et al. 2014 [563] SV SCLs 24 1 0.09 (0.08) 0.17 (0.08) 0.08 47
5 Paune et al. 2015 [564] SV spectacles 40 2 0.38 (0.21) 0.52 (0.22) 0.07 27
6 Aller et al. 2016 [565] SV SCLs 78 1 0.05 (0.14) 0.24 (0.17) 0.19 79
7 Cheng et al. 2016 [566] SV SCLs 106 1 0.23 (0.15) 0.37 (0.16) 0.14 38
8 Chamberlain et al. 2019 [567] SV SCLs 109 3 0.30 (0.27) 0.62 (0.30) 0.11 52
9 Sankaridurg et al. 2019 [568] (4 designs tested) SV SCLs 234 2 0.41 to 0.46 (across 4 0.58 (0.27) 0.06 to 22− 32
designs) 0.09
10 Walline et al. 2020 [559] (results given for high add SV SCLs 292 3 0.42 (95 % CI 0.38, 0.66 0.08 36
power MFCL + 2.50) 0.47)

META-ANALYSIS (does not include 2019 & 2020 papers) Absolute difference in progression
between treatment and control
groups (mm)

Li et al. 2017 [569] 7 1 0.27 (95 % CI 0.22, 0.32) 0.135 30− 50


studies

210
P.B. Morgan et al. Contact Lens and Anterior Eye 44 (2021) 192–219

6. Summary and future directions [6] Agostino R, Bologna M, Dinapoli L, Gregori B, Fabbrini G, Accornero N, et al.
Voluntary, spontaneous, and reflex blinking in Parkinson’s disease. Mov Disord
2008;23:669–75.
This report has provided a contemporary overview on the various [7] Jansen ME, Begley CG, Himebaugh NH, Port NL. Effect of contact lens wear and a
changes to ocular physiology and anatomy which are encountered near task on tear film break-up. Optom Vis Sci 2010;87:350–7.
during contact lens wear. Some consequences of contact lens wear can [8] Su Y, Liang Q, Su G, Wang N, Baudouin C, Labbe A. Spontaneous eye blink
patterns in dry eye: clinical correlations. Invest Ophthalmol Vis Sci 2018;59:
now be considered as largely of historic interest in many countries only 5149–56.
as modern lens designs and materials mean such changes are rarely seen. [9] McMonnies CW. The clinical and experimental significance of blinking behavior.
These include the signs of corneal hypoxia (including epithelial micro­ J Optom 2020;13:74–80.
[10] Navascues-Cornago M, Morgan PB, Maldonado-Codina C, Read ML.
cysts, stromal neovascularisation and endothelial polymegethism) – the Characterisation of blink dynamics using a high-speed infrared imaging system.
presence of which largely guided advances in contact lens materials over Ophthalmic Physiol Opt 2020;40:519–28.
the past 50 years – and changes to the palpebral conjunctiva which can [11] York M, Ong J, Robbins JC. Variation in blink rate associated with contact lens
wear and task difficulty. Am J Optom Arch Am Acad Optom 1971;48:461–7.
be readily improved with more frequent contact lens replacement as is [12] Cho P, Sheng C, Chan C, Lee R, Tam J. Baseline blink rates and the effect of visual
now commonplace. Although, overall only 23 % of fits are soft hydrogel task difficulty and position of gaze, Vol. 20; 2000.
lenses, countries such as Denmark, Japan and China are fitting [13] Doughty MJ. Consideration of three types of spontaneous eyeblink activity in
normal humans: during reading and video display terminal use, in primary gaze,
approximately equal numbers of soft hydrogel and SiHy lenses and and while in conversation. Optom Vis Sci 2001;78:712–25.
Taiwan fits more soft hydrogel lenses than SiHy [580]. [14] Bentivoglio AR, Bressman SB, Cassetta E, Carretta D, Tonali P, Albanese A.
Some changes of the eye to contact lens wear have remained stub­ Analysis of blink rate patterns in normal subjects. Mov Disord 1997;12:1028–34.
[15] Nakamori K, Odawara M, Nakajima T, Mizutani T, Tsubota K. Blinking is
bornly intransigent to improvements in both lens materials and designs.
controlled primarily by ocular surface conditions. Am J Ophthalmol 1997;124:
Bulbar conjunctival hyperaemia and cellular changes at the ocular sur­ 24–30.
face visualised by the use of sodium fluorescein and lissamine green are [16] Doughty MJ. Spontaneous eyeblink activity under different conditions of gaze
sensitive and somewhat non-specific indicators of the physiological (eye position) and visual glare. Graefes Arch Clin Exp Ophthalmol 2014;252:
1147–53.
impact of lens wear. Monitoring such changes during contact lens wear [17] Stern JA, Walrath LC, Goldstein R. The endogenous eyeblink. Psychophysiology
is likely to be a requirement for ECPs into the foreseeable future. 1984;21:22–33.
A number of identified changes have received relatively little [18] Doughty MJ. Influence of mouth and jaw movements on dynamics of spontaneous
eye blink activity assessed during slitlamp biomicroscopy. Clin Exp Optom 2018;
attention in recent times and their significance and/or clinical man­ 101:345–53.
agement is not well understood. These include blinking (which has [19] Doughty MJ, Naase T. Further analysis of the human spontaneous eye blink rate
received very limited attention in the literature) and LIPCOF which is by a cluster analysis-based approach to categorize individuals with “normal”
versus “frequent” eye blink activity. Eye Contact Lens 2006;32:294–9.
certainly a potentially important observation and may help further un­ [20] García-Montero M, Rico-Del-Viejo L, Martínez-Alberquilla I, Hernández-
derstanding of the mechanical interaction between contact lens and Verdejo JL, Lorente-Velázquez A, Madrid-Costa D. Effects of blink rate on tear
ocular surface. Emerging from this small group are LWE, interactions film optical quality dynamics with different soft contact lenses. J Ophthalmol
2019;2019:4921538.
between lenses and the Meibomian glands, and contact lens-induced [21] Martín-Montañez V, López-de la Rosa A, López-Miguel A, Pinto-Fraga J,
changes to ocular surface sensitivity which have received increasing González-Méijome JM, González-García MJ. End-of-day dryness, corneal
levels of attention and, given their possible relationship with contact sensitivity and blink rate in contact lens wearers. Cont Lens Anterior Eye 2015;38:
148–51.
lens discomfort, seem likely to be areas of strong research interest in the
[22] Yolton DP, Yolton RL, López R, Bogner B, Stevens R, Rao D. The effects of gender
coming years. and birth control pill use on spontaneous blink rates. J Am Optom Assoc 1994;65:
This report has highlighted three particular areas of interest which 763–70.
may transform the contact lens field in future years, in rather different [23] Doughty MJ. Further assessment of gender- and blink pattern-related differences
in the spontaneous eyeblink activity in primary gaze in young adult humans.
ways. The first two relate to the programmed alteration of ocular di­ Optom Vis Sci 2002;79:439–47.
mensions as seen in ortho-k and myopia control. These forms of [24] HIll RM, Carney LG. The effect of hard lens wear on blinking behaviour. Int
refractive manipulation provide distinct reasons for the use of contact Contact Lens Clin 1984;11:242–8.
[25] Roshani S. The effect of ocular surface conditions on blink rate and completeness.
lenses beyond the more traditional refractive correction and more niche Queensland University of Technology; 2011.
medical usage. The third area is the growing level of interest and [26] Van Der Worp E, De Brabander J, Swarbrick H, Hendrikse F. Eyeblink frequency
expertise in the assessment of the sub-clinical inflammatory response to and type in relation to 3- and 9-o’clock staining and gas permeable contact lens
variables. Optom Vis Sci 2008;85:E857–66.
contact lens wear. Insights provided by techniques such as in vivo [27] Carney LG, Hill RM. Variations in blinking bahaviour during soft lens wear. Int
confocal microscopy have been able to demonstrate features of the Contact Lens Clin 1984;11:250–3.
immunological response to contact lens wear and these is reason to [28] Ishak B, Thye JJY, Ali BM. Blinking characteristics and corneal staining in
different soft lens materials. Proc World Acad of Sci Eng Technol 2012.
believe that such research may be key to revealing the mechanisms [29] Collins MJ, Iskander DR, Saunders A, Hook S, Anthony E, Gillon R. Blinking
behind various physiological responses, adverse events and that cause of patterns and corneal staining. Eye Contact Lens 2006;32:287–93.
many drop-outs: contact lens discomfort. [30] Schulze M, Wong A, Haider S, Ebare K, Fadli Z, Coles-Brennan C, et al. Blink rate
in silicone hydrogel contact lens wearers during digital device use. Optom Vis Sci
2016;93. E – abstract:165122.
Acknowledgement [31] Kojima T, Matsumoto Y, Ibrahim OMA, Wakamatsu TH, Uchino M, Fukagawa K,
et al. Effect of controlled adverse chamber environment exposure on tear
functions in silicon hydrogel and hydrogel soft contact lens wearers. Invest
The CLEAR initiative was facilitated by the BCLA, with financial Ophthalmol Vis Sci 2011;52:8811–7.
support by way of Educational Grants for collaboration, publication and [32] López-de la Rosa A, Martín-Montañez V, López-Miguel A, Fernández I, Calonge M,
dissemination provided by Alcon and CooperVision. González-Méijome JM, et al. Ocular response to environmental variations in
contact lens wearers. Ophthalmic Physiol Opt 2017;37:60–70.
[33] González-Méijome JM. Qualitative and quantitative characterization of the in
References vitro dehydration process of hydrogel contact lenses. J Biomed Mater Res B Appl
Biomater 2007.
[34] Morgan PB, Efron N. In vivo dehydration of silicone hydrogel contact lenses. Eye
[1] Stapleton F, Bakkar M, Carnt N, Chalmers R, Kumar A, Marasini S, et al. CLEAR -
Contact Lens 2003;29:173–6.
Contact lens complications. Cont Lens Anterior Eye 2021;44. In press.
[35] Craig JP, Lim J, Han A, Tien L, Xue AL, Wang MTM. Ethnic differences between
[2] Doane MG. Interaction of eyelids and tears in corneal wetting and the dynamics
the Asian and Caucasian ocular surface: a co-located adult migrant population
of the normal human eyeblink. Am J Ophthalmol 1980;89:507–16.
cohort study. Ocul Surf 2019;17:83–8.
[3] Efron N. Contact Lens complications. Elsevier Health Sciences; 2019.
[36] Robinson DA. A method of measuring eye movemnent using a scieral search coil
[4] Doughty MJ. Consideration of three types of spontaneous eyeblink activity in
in a magnetic field. IEEE Trans Bio-Medical Electron 1963;10:137–45.
normal humans: during reading and video display terminal use, in primary gaze,
[37] Pult H, Riede-Pult BH, Murphy PJ. A new perspective on spontaneous blinks.
and while in conversation. Optom Vis Sci 2001;78:712–25.
Ophthalmology 2013;120:1086–91.
[5] Casse G, Sauvage J-P, Adenis J-P, Robert P-Y. Videonystagmography to assess
blinking. Graefes Arch Clin Exp Ophthalmol 2007;245:1789–96.

211
P.B. Morgan et al. Contact Lens and Anterior Eye 44 (2021) 192–219

[38] Mak FH, Harker A, Kwon KA, Edirisinghe M, Rose GE, Murta F, et al. Analysis of [72] Uçakhan Ö, Arslanturk-Eren M. The role of soft contact lens wear on meibomian
blink dynamics in patients with blepharoptosis. J R Soc Interface 2016;13. gland morphology and function. Eye Contact Lens 2019;45:292–300.
[39] Godfrey KJ, Wilsen C, Satterfield K, Korn BS, Kikkawa DO. Analysis of [73] Llorens-Quintana C, Garaszczuk IK, Szczesna-Iskander DH. Meibomian glands
spontaneous eyelid blink dynamics using a 240 frames per second smartphone structure in daily disposable soft contact lens wearers: a one-year follow-up study.
camera. Ophthal Plast Reconstr Surg 2019. Ophthalmic Physiol Opt 2020;40:607–16.
[40] Cline D, Hofstetter HW, Griffin JR. Dictionary of visual science. Butterworth- [74] Pucker AD, Jones-Jordan LA, Kunnen CME, Marx S, Powell DR, Kwan JT, et al.
Heinemann; 1997. Impact of meibomian gland width on successful contact lens use. Cont Lens
[41] Fonn D, Holden BA. Rigid gas-permeable vs. hydrogel contact lenses for extended Anterior Eye 2019;42:646–51.
wear. Am J Optom Physiol Opt 1988;65:536–44. [75] Covita A, Chen MH, Leahy C. Correlation between meibomian gland appearance
[42] Hwang K, Kim JH. The risk of blepharoptosis in contact lens wearers. J Craniofac and tear breakup time using a slit scanning ophthalmoscope. Invest Ophthalmol
Surg 2015;26:e373–4. Vis Sci 2019;60:6793.
[43] Epstein G, Putterman AM. Acquired blepharoptosis secondary to contact-lens [76] Kastelan S, Lukenda A, Salopek-Rabatić J, Pavan J, Gotovac M. Dry eye symptoms
wear. Am J Ophthalmol 1981;91:634–9. and signs in long-term contact lens wearers. Coll Antropol 2013;37(Suppl 1):
[44] Fonn D, Holden BA. Extended wear of hard gas permeable contact lenses can 199–203.
induce ptosis. CLAO J 1986;12:93–4. [77] Cho P, Yap M. Effect of contact lens wear on the tears of Hong Kong-Chinese. J Br
[45] van den Bosch WA, Lemij HG. Blepharoptosis induced by prolonged hard contact Contact Lens Assoc 1995;18:87–94.
lens wear. Ophthalmology 1992;99:1759–65. [78] Chui WS, Cho P, Brown B. Soft contact lens wear in Hong Kong - Chinese:
[46] Kersten RC, de Conciliis C, Kulwin DR. Acquired ptosis in the young and middle- predicting success. Ophthalmic Physiol Opt 2000;20:480–6.
aged adult population. Ophthalmology 1995;102:924–8. [79] Du Toit R, Situ P, Simpson T, Fonn D. The effects of six months of contact lens
[47] de Silva DJ, Collin JRO. Outcome following surgery for contact lens-induced wear on the tear film, ocular surfaces, and symptoms of presbyopes. Optom Vis
ptosis. Ophthal Plast Reconstr Surg 2011;27:186–9. Sci 2001;78.
[48] Thean JHJ, McNab AA. Blepharoptosis in RGP and PMMA hard contact lens [80] Best N, Drury L, Wolffsohn JS. Predicting success with silicone-hydrogel contact
wearers. Clin Exp Optom 2004;87:11–4. lenses in new wearers. Cont Lens Anterior Eye 2013;36:232–7.
[49] Bleyen I, Hiemstra CA, Devogelaere T, van den Bosch WA, Wubbels RJ, [81] Guillon M, Styles E, Guillon JP, Maïssa C. Preocular tear film characteristics of
Paridaens DA. Not only hard contact lens wear but also soft contact lens wear may nonwearers and soft contact lens wearers. Optom Vis Sci 1997;74:273–9.
be associated with blepharoptosis. Can J Ophthalmol 2011;46:333–6. [82] Pult H, Purslow C, Berry M, Murphy PJ. Clinical tests for successful contact lens
[50] Kitazawa T. Hard contact lens wear and the risk of acquired blepharoptosis: a wear: relationship and predictive potential. Optom Vis Sci 2008;85:E924–9.
case-control study. Eplasty 2013. [83] Wang X, Li J, Zhang R, Li N, Pang Y, Zhang Y, et al. The influence of overnight
[51] van den Bosch WA, Lemij HG. Blepharoptosis induced by prolonged hard contact orthokeratology on ocular surface and meibomian gland dysfunction in teenagers
lens wear. Ophthalmology 1992;99:1759–65. with myopia. J Ophthalmol 2019;2019:5142628.
[52] Reddy AK, Foroozan R, Arat YO, Edmond JC, Yen MT. Ptosis in young soft contact [84] Yang L, Zhang L, Jian Hu R, Yu PP, Jin X. The influence of overnight
lens wearers. Ophthalmology 2007;114:2370. orthokeratology on ocular surface and dry eye-related cytokines IL-17A, IL-6, and
[53] Fonn D, Pritchard N, Garnett B, Davids L. Palpebral aperture sizes of rigid and soft PGE2 in children. Cont Lens Anterior Eye 2021;44:81–8.
contact lens wearers compared with nonwearers. Optom Vis Sci 1996;73:211–4. [85] Korb DR, Henriquez AS. Meibomian gland dysfunction and contact lens
[54] Bron AJ, Tiffany JM, Gouveia SM, Yokoi N, Voon LW. Functional aspects of the intolerance. J Am Optom Assoc 1980;51:243–51.
tear film lipid layer. Exp Eye Res 2004;78:347–60. [86] Arita R, Fukuoka S, Morishige N. Meibomian gland dysfunction and contact lens
[55] Knop E, Knop N, Millar T, Obata H, Sullivan DA. The international workshop on discomfort. Eye Contact Lens 2017;43:17–22.
meibomian gland dysfunction: report of the subcommittee on anatomy, [87] Abelson MB, Lane K, Maffei C. Code red: the key features of hyperemia. Rev
physiology, and pathophysiology of the meibomian gland. Invest Ophthalmol Vis Ophthalmol 2010;17:92–4.
Sci 2011;52:1938–78. [88] McMonnies CW, Ho A. Conjunctival hyperaemia in non-contact lens wearers.
[56] Pucker AD, Tichenor AA. A review of contact lens dropout. Clin Optom (Auckl) Acta Ophthalmol 1991;69:799–801.
2020;12:85–94. [89] Murphy PJ, Lau JSC, Sim MML, Woods RL. How red is a white eye? Clinical
[57] Nichols JJ, Sinnott LT. Tear film, contact lens, and patient-related factors grading of normal conjunctival hyperaemia. Eye 2007;21:633–8.
associated with contact lens-related dry eye. Invest Ophthalmol Vis Sci 2006;47: [90] Pult H, Murphy PJ, Purslow C, Nyman J, Woods RL. Limbal and bulbar
1319–28. hyperaemia in normal eyes. Ophthalmic Physiol Opt 2008;28:13–20.
[58] Arita R, Itoh K, Maeda S, Maeda K, Tomidokoro A, Amano S. Association of [91] Bliss MR. Hyperaemia. J Tissue Viability 1998;8:4–13.
contact lens-related allergic conjunctivitis with changes in the morphology of [92] Oppenheimer DR, Palmer E, Weddell G. Nerve endings in the conjunctiva. J Anat
meibomian glands. Jpn J Ophthalmol 2012;56:14–9. 1958;92:321–52.
[59] Machalińska A, Zakrzewska A, Adamek B, Safranow K, Wiszniewska B, [93] McMonnies CW, Chapman-Davies A, Holden BA. The vascular response to contact
Parafiniuk M, et al. Comparison of morphological and functional meibomian lens wear. Am J Optom Physiol Opt 1982;59:795–9.
gland characteristics between daily contact lens wearers and nonwearers. Cornea [94] Efron N. Rethinking contact lens discomfort. Clin Exp Optom 2018;101:1–3.
2015;34:1098–104. [95] Efron N, editor. Chapter 11: conjunctival redness. Contact lens complications. 3rd
[60] Pucker AD, Jones-Jordan LA, Li W, Kwan JT, Lin MC, Sickenberger W, et al. ed. Edinburgh: Elsevier Sounders; 2012. p. 113–21.
Associations with meibomian gland atrophy in daily contact lens wearers. Optom [96] Efron N, editor. Chapter 13: limbal redness. Contact lens complications. 3rd ed.
Vis Sci 2015;92:e206–13. Edinburgh: Elsevier Sounders; 2012. p. 133–9.
[61] Na K-S, Yoo Y-S, Hwang HS, Mok JW, Kim HS, Joo C-K. The influence of [97] Chao C, Richdale K, Jalbert I, Doung K, Gokhale M. Non-invasive objective and
overnight orthokeratology on ocular surface and meibomian glands in children contemporary methods for measuring ocular surface inflammation in soft contact
and adolescents. Eye Contact Lens 2016;42:68–73. lens wearers - A review. Cont Lens Anterior Eye 2017;40:273–82.
[62] Ong BL, Larke JR. Meibomian gland dysfunction: some clinical, biochemical and [98] Larke JR, Humphreys JA, Holmes R. Apparent corneal neovascularisation in soft
physical observations. Ophthalmic Physiol Opt 1990;10:144–8. lens wearers. J Br Contact Lens Ass 1981;4:105–6.
[63] Arita R, Itoh K, Inoue K, Kuchiba A, Yamaguchi T, Amano S. Contact lens wear is [99] Holden BA, Sweeney DF, Swarbrick HA, Vannas A, Nilsson KT, Efron N. The
associated with decrease of meibomian glands. Ophthalmology 2009;116: vascular response to long-term extended contact lens wear. Clin Exp Optom 1986;
379–84. 69:112–9.
[64] Arita R, Itoh K, Inoue K, Amano S. Noncontact infrared meibography to document [100] Maldonado-Codina C, Morgan PB, Schnider CM, Efron N. Short-term physiologic
age-related changes of the meibomian glands in a normal population. response in neophyte subjects fitted with hydrogel and silicone hydrogel contact
Ophthalmology 2008;115:911–5. lenses. Optom Vis Sci 2004;81:911–21.
[65] Gu T, Zhao L, Liu Z, Zhao S, Nian H, Wei R. Evaluation of tear film and the [101] Efron N, Jones L, Bron AJ, Knop E, Arita R, Barabino S, et al. The TFOS
morphological changes of meibomian glands in young Asian soft contact lens International Workshop on Contact Lens Discomfort: report of the contact lens
wearers and non-wearers. BMC Ophthalmol 2020;20:84. interactions with the ocular surface and adnexa subcommittee. Invest Ophthalmol
[66] Villani E, Ceresara G, Beretta S, Magnani F, Viola F, Ratiglia R. In vivo confocal Vis Sci 2013;54:TFOS98–122.
microscopy of meibomian glands in contact lens wearers. Invest Ophthalmol Vis [102] Chen W, Xu Z, Jiang H, Zhou J, Wang L, Wang J. Altered bulbar conjunctival
Sci 2011;52:5215–9. microcirculation in response to contact lens wear. Eye Contact Lens 2017;43:
[67] Siddireddy JS, Vijay AK, Tan J, Willcox M. The eyelids and tear film in contact 95–9.
lens discomfort. Cont Lens Anterior Eye 2018;41:144–53. [103] Hu L, Shi C, Jiang H, Shi Y, Sethi Z, Wang J. Factors affecting microvascular
[68] Zhou S, Robertson DM. Wide-field in vivo confocal microscopy of meibomian responses in the bulbar conjunctiva in habitual contact lens wearers. Invest
gland acini and rete ridges in the eyelid margin. Invest Ophthalmol Vis Sci 2018; Ophthalmol Vis Sci 2018;59:4108–14.
59:4249–57. [104] Wolffsohn JS, Naroo SA, Christie C, Morris J, Conway R, Maldonado-Codina C,
[69] Zhou N, Edwards K, Colorado LH, Schmid KL. Development of feasible methods to et al. History and symptom taking in contact lens fitting and aftercare. Cont Lens
image the eyelid margin using in vivo confocal microscopy. Cornea 2020;39: Anterior Eye 2015;38:258–65.
1325–33. [105] Efron N, editor. Chapter 2: grading scales. Contact lens complications. 3rd ed.
[70] Pucker AD, Jones-Jordan LA, Marx S, Powell DR, Kwan JT, Srinivasan S, et al. Edinburgh: Elsevier Saunders; 2012. p. 21–31.
Clinical factors associated with contact lens dropout. Cont Lens Anterior Eye [106] Papas EB. Key factors in the subjective and objective assessment of conjunctival
2019;42:318–24. erythema. Invest Ophthalmol Vis Sci 2000;41:687–91.
[71] Alghamdi WM, Markoulli M, Holden BA, Papas EB. Impact of duration of contact [107] Peterson RC, Wolffsohn JS. Sensitivity and reliability of objective image analysis
lens wear on the structure and function of the meibomian glands. Ophthalmic compared to subjective grading of bulbar hyperaemia. Br J Ophthalmol 2007;91:
Physiol Opt 2016;36:120–31. 1464–6.

212
P.B. Morgan et al. Contact Lens and Anterior Eye 44 (2021) 192–219

[108] Schulze MM, Jones DA, Simpson TL. The development of validated bulbar redness [143] Begley CG, Chalmers RL, Abetz L, Venkataraman K, Mertzanis P, Caffery BA, et al.
grading scales. Optom Vis Sci 2007;84:976–83. The relationship between habitual patient-reported symptoms and clinical signs
[109] Schulze MM, Hutchings N, Simpson TL. Grading bulbar redness using cross- among patients with dry eye of varying severity. Invest Ophthalmol Vis Sci 2003;
calibrated clinical grading scales. Invest Ophthalmol Vis Sci 2011;52:5812–7. 44:4753–61.
[110] Amparo F, Wang H, Emami-Naeini P, Karimian P, Dana R. The ocular redness [144] Kim J, Foulks GN. Evaluation of the effect of lissamine green and rose bengal on
index: a novel automated method for measuring ocular injection. Invest human corneal epithelial cells. Cornea 1999;18:328–32.
Ophthalmol Vis Sci 2013;54:4821–6. [145] Lakkis C, Brennan NA. Bulbar conjunctival fluorescein staining in hydrogel
[111] Macchi I, Bunya VY, Massaro-Giordano M, Stone RA, Maguire MG, Zheng Y, et al. contact lens wearers. CLAO J 1996;22:189–94.
A new scale for the assessment of conjunctival bulbar redness. Ocul Surf 2018;16: [146] Guillon M, Maissa C. Bulbar conjunctival staining in contact lens wearers and non
436–40. lens wearers and its association with symptomatology. Cont Lens Anterior Eye
[112] Huntjens B, Basi M, Nagra M. Evaluating a new objective grading software for 2005;28:67–73.
conjunctival hyperaemia. Cont Lens Anterior Eye 2020;43:137–43. [147] Ozkan J, Ehrmann K, Meadows D, Lally J, Holden B, de la Jara PL. Lens parameter
[113] Cheung ATW, Hu BS, Wong SA, Chow J, Chan MS, To WJ, et al. Microvascular changes under in vitro and ex vivo conditions and their effect on the conjunctiva.
abnormalities in the bulbar conjunctiva of contact lens users. Clin Hemorheol Cont Lens Anterior Eye 2013;36:171–5.
Microcirc 2012;51:77–86. [148] Turhan SA, Toker E. Optical coherence tomography to evaluate the interaction of
[114] Jones L, Dumbleton K. Chapter 10: soft contact Lens fitting. In: Phillips AJSJ, different edge designs of four different silicone hydrogel lenses with the ocular
editor. Contact lenses. 6th ed. Edinburgh: Elsevier; 2018. p. 207–22. surface. Clin Ophthalmol 2015;9:935–42.
[115] Sorbara L, Maram J, Simpson T, Hutchings N. Corneal, conjunctival effects and [149] Sorbara L, Simpson TL, Maram J, Song ES, Bizheva K, Hutchings N. Optical edge
blood flow changes related to silicone hydrogel lens wear and their correlations effects create conjunctival indentation thickness artefacts. Ophthalmic Physiol
with end of day comfort. Cont Lens Anterior Eye 2018;41:193–200. Opt 2015;35:283–92.
[116] Stapleton F, Rodriguez L, Cengiz A. Ocular surface sensitivity in contact lens [150] Wolffsohn JS, Drew T, Dhallu S, Sheppard A, Hofmann GJ, Prince M. Impact of
wear. Optom Vis Sci 1999;76:108. soft contact lens edge design and midperipheral lens shape on the epithelium and
[117] Maïssa C, Guillon M, Garofalo RJ. Contact lens-induced circumlimbal staining in its indentation with lens mobility. Invest Ophthalmol Vis Sci 2013;54:6190–7.
silicone hydrogel contact lenses worn on a daily wear basis. Eye Contact Lens [151] Bandlitz S, Ruland T, Schwarz D, Jandl A, Wolffsohn J, Pult H. Clinical
2012;38:16–26. significance of contact lens related changes of ocular surface tissue observed on
[118] Stapleton F, Tan J. Impact of contact lens material, design, and fitting on optical coherence images. Cont Lens Anterior Eye 2018;41:S84.
discomfort. Eye Contact Lens 2017;43:32–9. [152] Stahl U, Willcox MDP, Naduvilath T, Stapleton F. Influence of tear film and
[119] Rabiah NI, Scales CW, Fuller GG. The influence of protein deposition on contact contact lens osmolality on ocular comfort in contact lens wear. Optom Vis Sci
lens tear film stability. Colloids Surf B Biointerfaces 2019;180:229–36. 2009;86:857–67.
[120] Allansmith MR, Korb DR, Greiner JV, Henriquez AS, Simon MA, Finnemore VM. [153] Bandlitz S, Purslow C, Murphy PJ, Pult H. Lid-parallel conjunctival fold (LIPCOF)
Giant papillary conjunctivitis in contact lens wearers. Am J Ophthalmol 1977;83: morphology imaged by optical coherence tomography and its relationship to
697–708. LIPCOF grade. Cont Lens Anterior Eye 2019;42:299–303.
[121] McGill JI, Holgate ST, Church MK, Anderson DF, Bacon A. Allergic eye disease [154] Höh H, Schirra F, Kienecker C. Lid-parallel conjunctival folds are a sure diagnostic
mechanisms. Br J Ophthalmol 1998;82:1203–14. sign of dry eye. Ophthalmologe 1995;92:802–8.
[122] Papas EB. The role of hypoxia in the limbal vascular response to soft contact lens [155] Tapasztó B, Veres A, Kosina-Hagyó K, Somfai GM, Németh J. OCT Imaging of lid-
wear. Eye Contact Lens 2003;29:S72–4. discussion S83–4, S192–4. parallel conjunctival folds in soft contact lens wearers. Optom Vis Sci 2011;88:
[123] Papas EB, Vajdic CM, Austen R, Holden BA. High-oxygen-transmissibility soft 1206–13.
contact lenses do not induce limbal hyperaemia. Curr Eye Res 1997;16:942–8. [156] Pult H, Purslow C, Murphy PJ. The relationship between clinical signs and dry eye
[124] Covey M, Sweeney DF, Terry R, Sankaridurg PR, Holden BA. Hypoxic effects on symptoms. Eye 2011;25:502–10.
the anterior eye of high-Dk soft contact lens wearers are negligible. Optom Vis Sci [157] Bron AJ, de Paiva CS, Chauhan SK, Bonini S, Gabison EE, Jain S, et al. TFOS
2001;78:95–9. DEWS II pathophysiology report. Ocul Surf 2017;15:438–510.
[125] Dumbleton KA, Chalmers RL, Richter DB, Fonn D. Vascular response to extended [158] Pult H, Bandlitz S. Lid-parallel conjunctival folds and their ability to predict dry
wear of hydrogel lenses with high and low oxygen permeability. Optom Vis Sci eye. Eye Contact Lens 2018;44(Suppl 2):S113–9.
2001;78:147–51. [159] Németh J, Fodor E, Lang Z, Kosina-Hagyó K, Berta A, Komár T, et al. Lid-parallel
[126] Chalmers RL, Dillehay S, Long B, Barr JT, Bergenske P, Donshik P, et al. Impact of conjunctival folds (LIPCOF) and dry eye: a multicentre study. Br J Ophthalmol
previous extended and daily wear schedules on signs and symptoms with high Dk 2012;96:1380–5.
lotrafilcon A lenses. Optom Vis Sci 2005;82:549–54. [160] Pult H, Murphy PJ, Purslow C. A novel method to predict the dry eye symptoms in
[127] Dhallu SK, Huarte ST, Bilkhu PS, Boychev N, Wolffsohn JS. Effect of scleral lens new contact lens wearers. Optom Vis Sci 2009;86:E1042–50.
oxygen permeability on corneal physiology. Optom Vis Sci 2020;97:669–75. [161] Sickenberger W, Pult H, Sickenberger B. LIPCOF and contact lens wearers: a new
[128] Fisher D, Collins MJ, Vincent SJ. Fluid reservoir thickness and corneal edema tool to forecast subjective dryness and degree of comfort of contact lens wearers.
during open-eye scleral lens wear. Optom Vis Sci 2020;97:683–9. Contactologia 2000;22:74–9.
[129] Thai LC, Tomlinson A, Doane MG. Effect of contact lens materials on tear [162] Wolffsohn JS, Arita R, Chalmers R, Djalilian A, Dogru M, Dumbleton K, et al.
physiology. Optom Vis Sci 2004;81:194–204. TFOS DEWS II diagnostic methodology report. Ocul Surf 2017;15:539–74.
[130] Guillon M, Maissa C. Contact lens wear affects tear film evaporation. Eye Contact [163] Bandlitz S, Purslow C, Murphy PJ, Pult H. The usefulness of optical coherence
Lens 2008;34:326–30. tomography in LIPCOF grading. Contact Lens Anterior Eye 2018;41:S67.
[131] McMonnies CW. An amplifying cascade of contact lens-related end-of-day [164] Veres A, Tapasztó B, Kosina-Hagyó K, Somfai GM, Németh J. Imaging lid-parallel
hyperaemia and dryness symptoms. Curr Eye Res 2018;43:839–47. conjunctival folds with OCT and comparing its grading with the slit lamp
[132] González-Méijome JM, López-Alemany A, Almeida JB, Parafita MA. Dynamic in classification in dry eye patients and normal subjects. Invest Ophthalmol Vis Sci
vitro dehydration patterns of unworn and worn silicone hydrogel contact lenses. 2011;52:2945–51.
J Biomed Mater Res B Appl Biomater 2009;90:250–8. [165] Bandlitz S, Purslow C, Murphy PJ, Pult H. The relationship between tear meniscus
[133] Ramamoorthy P, Sinnott LT, Nichols JJ. Contact lens material characteristics regularity and conjunctival folds. Optom Vis Sci 2014;91:1037–44.
associated with hydrogel lens dehydration. Ophthalmic Physiol Opt 2010;30: [166] Pult H, Riede-Pult BH. Impact of conjunctival folds on central tear meniscus
160–6. height. Invest Ophthalmol Vis Sci 2015;56:1459–66.
[134] Efron N, Brennan NA, Morgan PB, Wilson T. Lid wiper epitheliopathy. Prog Retin [167] Bandlitz S, Purslow C, Murphy PJ, Pult H. Influence of conjunctival folds on
Eye Res 2016;53:140–74. calculated lower tear meniscus volume. Contact Lens Anterior Eye 2018;41:
[135] Mondino BJ, Salamon SM, Zaidman GW. Allergic and toxic reactions of soft S9–10.
contact lens wearers. Surv Ophthalmol 1982;26:337–44. [168] Bandlitz S, Purslow C, Murphy PJ, Pult H. Influence of conjunctival folds on
[136] Young G, Keir N, Hunt C, Woods CA. Clinical evaluation of long-term users of two calculated tear Meniscus volume along the lower eyelid. Ocul Surf 2016;14:
contact lens care preservative systems. Eye Contact Lens 2009;35:50–8. 377–84.
[137] Jones L, Brennan NA, González-Méijome J, Lally J, Maldonado-Codina C, [169] Pult H, Korb DR, Murphy PJ, Riede-Pult BH, Blackie C. A new model of central lid
Schmidt TA, et al. The TFOS International Workshop on Contact Lens Discomfort: margin apposition and tear film mixing in spontaneous blinking. Cont Lens
report of the contact lens materials, design, and care subcommittee. Invest Anterior Eye 2015;38:173–80.
Ophthalmol Vis Sci 2013;54:TFOS37–70. [170] Pult H, Tosatti SGP, Spencer ND, Asfour J-M, Ebenhoch M, Murphy PJ.
[138] Beshtawi IM, Qaddomi J, Khuffash H, El-Titi S, Ghannam M, Otaibi R. Ocular Spontaneous blinking from a tribological viewpoint. Ocul Surf 2015;13:236–49.
surface response and subjective symptoms associated to lens care solutions in [171] Berry M, Pult H, Purslow C, Murphy PJ. Mucins and ocular signs in symptomatic
Palestine. J Optom 2019;12:248–55. and asymptomatic contact lens wear. Optom Vis Sci 2008;85:E930–8.
[139] Bernardes TF, Bonfioli AA. Blepharitis. Semin Ophthalmol 2010;25:79–83. [172] Wolffsohn Js. BCLA pioneers lecture - evidence basis for patient selection: how to
[140] Bron AJ, Evans VE, Smith JA. Grading of corneal and conjunctival staining in the predict contact lens success. Cont Lens Anterior Eye 2014;37:63–4.
context of other dry eye tests. Cornea 2003;22:640–50. [173] Pult H, Riede-Pult BH. Impact of soft contact lenses on lid- parallel conjunctival
[141] Bakkar MM, Hardaker L, March P, Morgan PB, Maldonado-Codina C, Dobson CB. folds. Cont Lens Anterior Eye 2019;42:415–9.
The cellular basis for biocide-induced fluorescein hyperfluorescence in [174] Pult H, Purslow C, Murphy PJ, Berry M. Lid wiper epitheliopathy, ocular surface
mammalian cell culture. PLoS One 2014;9:e84427. and tear film in symptomatic contact lens wearers. Acta Ophthalmol 2008;86:0.
[142] Khan TF, Price BL, Morgan PB, Maldonado-Codina C, Dobson CB. Cellular [175] Mimura T, Usui T, Yamamoto H, Yamagami S, Funatsu H, Noma H, et al.
fluorescein hyperfluorescence is dynamin-dependent and increased by Tetronic Conjunctivochalasis and contact lenses. Am J Ophthalmol 2009;148:20–25.e1.
1107 treatment. Int J Biochem Cell Biol 2018;101:54–63.

213
P.B. Morgan et al. Contact Lens and Anterior Eye 44 (2021) 192–219

[176] Veliksar TA, Gaidamaka TB, Drozhzhina GI. Ocular surface changes in mild and [209] Navascues-Cornago M, Morgan PB, Maldonado-Codina C. Lid margin sensitivity
moderate myopes differing in duration of soft contact lens wear. J ophthalmol and staining in contact lens wear versus no lens wear. Cornea 2015;34:808–16.
(Ukraine) 2018;6:3–9. [210] Efron N, editor. Chapter 10: conjunctival staining. Contact lens compications. 3rd
[177] Pult H. The repeatability of lid parallel conjunctival folds and lid wiper ed. Edinburgh: Elsevier Sounders; 2012. p. 102–12.
epitheliopathy. Contact Lens Anterior Eye 2018;41:S91. [211] Stapleton F, Tan ME, Papas EB, Ehrmann K, Golebiowski B, Vega J, et al. Corneal
[178] Pult H, Riede-Pult B. Grading of lid-parallel conjunctival folds by novice and and conjunctival sensitivity to air stimuli. Br J Ophthalmol 2004;88:1547–51.
experienced observers. Invest Ophthalmol Vis Sci 2011;52:3739. [212] Golebiowski B, Papas EB, Stapleton F. Corneal and conjunctival sensory function:
[179] Evans KS, Pult H. How important are surface properties for successful contact lens the impact on ocular surface sensitivity of change from low to high oxygen
wear? Optician 2012;5:14–8. transmissibility contact lenses. Invest Ophthalmol Vis Sci 2012;53:1177–81.
[180] Keir N, Woods J, Sickenberger W. The conjunctival response to soft contact lens [213] Golebiowski B, Chao C, Stapleton F, Jalbert I. Corneal nerve morphology,
wear: a practical guide. Optom Pract 2010;11:123–34. sensitivity, and tear neuropeptides in contact lens wear. Optom Vis Sci 2017;94:
[181] Pult H, Riede-Pult BH, Murphy PJ. The relation between blinking and 534–42.
conjunctival folds and dry eye symptoms. Optom Vis Sci 2013;90:1034–9. [214] Stapleton F, Chao C, Golebiowski B. Topical review: effects of contact lens wear
[182] Chhadva P, Alexander A, McClellan AL, McManus KT, Seiden B, Galor A. The on corneal, conjunctival, and lid margin sensitivity. Optom Vis Sci 2019;96:
impact of conjunctivochalasis on dry eye symptoms and signs. Invest Ophthalmol 790–801.
Vis Sci 2015;56:2867–71. [215] Golebiowski B, Papas E, Stapleton F. Assessing the sensory function of the ocular
[183] Gumus K, Pflugfelder SC. Increasing prevalence and severity of surface: implications of use of a non-contact air jet aesthesiometer versus the
conjunctivochalasis with aging detected by anterior segment optical coherence Cochet-Bonnet aesthesiometer. Exp Eye Res 2011;92:408–13.
tomography. Am J Ophthalmol 2013;155:238–42.e2. [216] Wolter JR. Double nerve supply of krause CORPUSCLES at the human corneal
[184] Markoulli M, Carnt N, Jalbert I, Keay L, Naduvilath T, Papas E. Resolution and limbus. Arch Ophthalmol 1964;71:409–11.
clinical characteristics of conjunctival flaps. Invest Ophthalmol Vis Sci 2007;48: [217] Lawrenson JG, Ruskell GL. The structure of corpuscular nerve endings in the
5391. limbal conjunctiva of the human eye. J Anat 1991;177:75–84.
[185] Graham AD, Truong TN, Lin MC. Conjunctival epithelial flap in continuous [218] Nichols JJ, Willcox MDP, Bron AJ, Belmonte C, Ciolino JB, Craig JP, et al. The
contact lens wear. Optom Vis Sci 2009;86:e324–31. TFOS international workshop on contact lens discomfort: executive summary.
[186] Santodomingo-Rubido J, Wolffsohn J, Gilmartin B. Conjunctival epithelial flaps Invest Ophthalmol Vis Sci 2013;54:TFOS7–13.
with 18 months of silicone hydrogel contact lens wear. Eye Contact Lens 2008;34: [219] Hübner T. Edge profiles of hydrogel contact lenses and their effect on fitting and
35–8. wearing characteristics. Wohlk Wissen 2010;2.
[187] Bergmanson JPG, Tukler J, Leach NE, Alabdelmoneam M, Miller WL. Morphology [220] Bonanno JA, Polse KA. Corneal acidosis during contact lens wear: effects of
of contact lens-induced conjunctival epithelial flaps: a pilot study. Cont Lens hypoxia and CO2. Invest Ophthalmol Vis Sci 1987;28:1514–20.
Anterior Eye 2012;35:185–8. [221] Remington LA, editor. Chapter 9: ocular adnexa and lacrimal system. Clinical
[188] Willcox M, Keir N, Maseedupally V, Masoudi S, McDermott A, Mobeen R, et al. anatomy and physiology of the visual system. 3rd ed. St. Louis, Mo: Elsevier/
CLEAR - Contact lenses wettability, cleaning, disinfection and interactions with Butterworth-Heinemann; 2012. p. 159–81.
tears. Cont Lens Anterior Eye 2021;44. In press. [222] Shumway CL, Motlagh M, Wade M. Anatomy, head and neck, eye conjunctiva.
[189] Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature StatPearls, Treasure Island (FL): StatPearls Publishing; 2019.
1998;392:245–52. [223] MacKinven J, McGuinness CL, Pascal E, Woods RL. Clinical grading of the upper
[190] Golebiowski B, Chao C, Bui KA, Lam WYW, Richdale K, Stapleton F. Effect of age palpebral conjunctiva of non-contact lens wearers. Optom Vis Sci 2001;78:13–8.
and contact lens wear on corneal epithelial dendritic cell distribution, density, [224] Efron N, editor. Chapter 12: papillary conjunctivitis. Contact lens complications.
and morphology. Cont Lens Anterior Eye 2020;43:84–90. 3rd ed. Edinburgh: Elsevier Sounders; 2012. p. 122–32.
[191] Mobeen R, Stapleton F, Chao C, Madigan MC, Briggs N, Golebiowski B. Corneal [225] Roba M, Duncan EG, Hill GA, Spencer ND, Tosatti SGP. Friction measurements on
epithelial dendritic cell density in the healthy human cornea: a meta-analysis of contact lenses in their operating environment. Tribol Lett 2011;44:387.
in-vivo confocal microscopy data. Ocul Surf 2019;17:753–62. [226] Spring TF. Reaction to hydrophilic lenses. Med J Aust 1974;1:449–50.
[192] Saliman NH, Morgan PB, MacDonald AS, Maldonado-Codina C. Subclinical [227] Karpecki PM. Contact lens wear and ocular allergy. Contact Lens Spectrum 2012;
inflammation of the ocular surface in soft contact lens wear. Cornea 2020;39: 27:26–32.
146–54. [228] Solomon A. Allergic manifestations of contact lens wearing. Curr Opin Allergy
[193] Alzahrani Y, Colorado LH, Pritchard N, Efron N. Longitudinal changes in Clin Immunol 2016;16:492–7.
Langerhans cell density of the cornea and conjunctiva in contact lens-induced dry [229] Grant T, Holden BA, Rechberger JS, Chong MS. Contact lens related papillary
eye. Clin Exp Optom 2017;100:33–40. conjunctivitis (CLPC): influence of protein accumulation and replacement
[194] Efron N, Al-Dossari M, Pritchard N. Confocal microscopy of the bulbar frequency. Invest Ophthalmol Vis Sci 1989;30:S166.
conjunctiva in contact lens wear. Cornea 2010;29:43–52. [230] Poggio EC, Abelson M. Complications and symptoms in disposable extended wear
[195] Wright EA, Payne KAP, Jowitt TA, Howard M, Morgan PB, Maldonado-Codina C, lenses compared with conventional soft daily wear and soft extended wear lenses.
et al. Preservation of human tear protein structure and function by a novel contact CLAO J 1993;19:31–9.
lens multipurpose solution containing protein-stabilizing agents. Eye Contact [231] Nason RJ, Boshnick EL, Cannon WM, Dubow BW, Freeman MI, Kame RT, et al.
Lens 2012;38:36–42. Multisite comparison of contact lens modalities. Daily disposable wear vs.
[196] Saban DR. The chemokine receptor CCR7 expressed by dendritic cells: a key conventional daily wear in successful contact lens wearers. J Am Optom Assoc
player in corneal and ocular surface inflammation. Ocul Surf 2014;12:87–99. 1994;65:774–80.
[197] Lužnik Z, Kopitar AN, Lapajne L, Pižem J, Ferrari S, Ihan A, et al. Identification [232] Rao GN, Naduvilath TJ, Sankaridurg PR, Ramachandaran L, Gora R, Kalikivayi V,
and characterization of dendritic cell subtypes in preserved and cultured et al. Contact lens related papillary conjunctivitis in a prospective randomised
cadaveric human corneolimbal tissue on amniotic membrane. Acta Ophthalmol clinical trial using disposable hydrogels. In: Investigative ophthalmology & visual
2019;97:e184–93. science, Vol. 37, lippincott-raven Publ 227 East Washington SQ; 1996. p. 5180.
[198] Müller LJ, Pels L, Vrensen GF. Ultrastructural organization of human corneal [233] Sankaridurg PR, Sweeney DF, Sharma S, Gora R, Naduvilath T, Ramachandran L,
nerves. Invest Ophthalmol Vis Sci 1996;37:476–88. et al. Adverse events with extended wear of disposable hydrogels: results for the
[199] Müller LJ, Vrensen GF, Pels L, Cardozo BN, Willekens B. Architecture of human first 13 months of lens wear. Ophthalmology 1999;106:1671–80.
corneal nerves. Invest Ophthalmol Vis Sci 1997;38:985–94. [234] Santodomingo-Rubido J, Wolffsohn JS, Gilmartin B. Adverse events and
[200] Müller LJ, Marfurt CF, Kruse F, Tervo TMT. Corneal nerves: structure, contents discontinuations during 18 months of silicone hydrogel contact lens wear. Eye
and function. Exp Eye Res 2003;76:521–42. Contact Lens 2007;33:288–92.
[201] Stapleton F, Marfurt C, Golebiowski B, Rosenblatt M, Bereiter D, Begley C, et al. [235] Tagliaferri A, Love TE, Szczotka-Flynn LB. Risk factors for contact lens-induced
The TFOS international workshop on contact lens discomfort: report of the papillary conjunctivitis associated with silicone hydrogel contact lens wear. Eye
subcommittee on neurobiology. Invest Ophthalmol Vis Sci 2013;54:TFOS71–97. Contact Lens 2014;40:117–22.
[202] Rózsa AJ, Beuerman RW. Density and organization of free nerve endings in the [236] Solomon OD, Freeman MI, Boshnick EL, Cannon WM, Dubow BW, Kame RT, et al.
corneal epithelium of the rabbit. Pain 1982;14:105–20. A 3-year prospective study of the clinical performance of daily disposable contact
[203] Acosta MC, Belmonte C, Gallar J. Sensory experiences in humans and single-unit lenses compared with frequent replacement and conventional daily wear contact
activity in cats evoked by polymodal stimulation of the cornea. J Physiol 2001; lenses. CLAO J 1996;22:250–7.
534:511–25. [237] Korb DR, Greiner JV, Herman JP, Hebert E, Finnemore VM, Exford JM, et al. Lid-
[204] Murphy PJ, Armstrong E, Woods LA. A comparison of corneal and conjunctival wiper epitheliopathy and dry-eye symptoms in contact lens wearers. CLAO J
sensitivity to a thermally cooling stimulus. Adv Exp Med Biol 2002;506:719–22. 2002;28:211–6.
[205] Remington LA, editor. Chapter 2: cornea and sclera. Clinical anatomy of the visual [238] Doughty MJ, Naase T, Donald C, Hamilton L, Button NF. Visualisation of “Marx’s
system. 3rd ed. St. Louis, Mo: St. Louis, Mo.: Elsevier/Butterworth-Heinemann; line” along the marginal eyelid conjunctiva of human subjects with lissamine
2012. p. 10–39. green dye. Ophthalmic Physiol Opt 2004;24:1–7.
[206] Norn MS. Conjunctival sensitivity in pathological cases, with simultaneous [239] Shiraishi A, Yamaguchi M, Ohashi Y. Prevalence of upper- and lower-lid-wiper
measurement of corneal and lid margin sensitivity. Acta Ophthalmol 1975;53: epitheliopathy in contact lens wearers and non-wearers. Eye Contact Lens 2014;
450–7. 40:220–4.
[207] Igarashi T, Ono M, Fujimoto C, Suzuki H, Takahashi H. The conjunctival [240] Yeniad B, Beginoglu M, Bilgin LK. Lid-wiper epitheliopathy in contact lens users
sensitivity in soft contact lens wearers. Int Ophthalmol 2015;35:569–73. and patients with dry eye. Eye Contact Lens 2010;36:140–3.
[208] Situ P, Simpson TL, Jones LW, Fonn D. Effects of silicone hydrogel contact lens [241] Li W, Yeh TN, Leung T, Yuen T, Lerma M, Lin MC. The relationship of lid wiper
wear on ocular surface sensitivity to tactile, pneumatic mechanical, and chemical epitheliopathy to ocular surface signs and symptoms. Invest Ophthalmol Vis Sci
stimulation. Invest Ophthalmol Vis Sci 2010;51:6111–7. 2018;59:1878–87.

214
P.B. Morgan et al. Contact Lens and Anterior Eye 44 (2021) 192–219

[242] Yan X-M, Liu S, Li H-L. Preliminary observation the correlation between lid-wiper [275] Alzahrani Y, Colorado L, Pritchard N, Efron N. Inflammatory cell upregulation of
epitheliopathy and dry eye. Zhonghua Yan Ke Za Zhi 2008;44:436–41. the lid wiper in contact Lens dry eye. Optom Vis Sci 2016;93:917–24.
[243] Maissa C, Guillon M, Varsani S, Wong S. Lid margins staining with lissamine green [276] Efron N, Al-Dossari M, Pritchard N. In vivo confocal microscopy of the palpebral
- contact lens wear and dry eye symptomatology. Optom Vis Sci 2009. E – abstract conjunctiva and tarsal plate. Optom Vis Sci 2009;86:E1303–8.
95679. [277] Le Q, Hong J, Zhu W, Sun X, Xu J. In vivo laser scanning confocal microscopy of
[244] Read M, Morgan P, Smith S, Maldonado-Codina C. Development of an imaging vernal keratoconjunctivitis. Clin Exp Ophthalmol 2011;39:53–60.
system to detect changes in redness of the eyelid margin. Optom Vis Sci 2014;90. [278] Hu VH, Holland MJ, Cree IA, Pullin J, Weiss HA, Massae P, et al. In vivo confocal
E – abstract 140083. microscopy and histopathology of the conjunctiva in trachomatous scarring and
[245] Jalbert I, Madigan MC, Shao M, Ng J, Cheng J, Wong D, et al. Assessing the normal tissue: a systematic comparison. Br J Ophthalmol 2013;97:1333–7.
human lid margin epithelium using impression cytology. Acta Ophthalmol [279] Abelson MB, Greiner JV, Chin TLN, Mooshian M. Adaptive changes in the
(Copenh) 2012;90:e547–52. threshold sensitivity of the tarsus as measured with Cochet-Bonnet
[246] Alghamdi WM, Markoulli M, Papas EB. The effect of contact lens wear on the aesthesiometer. Invest Ophthalmol Vis Sci 1993;34:1006.
cellular morphology of the lid wiper area. Optom Vis Sci 2018;95:491–7. [280] Stapleton F, Golebiowski B, Skotnitsky C, Tan ME, Holden BA. Corneal and
[247] Schulze M-M, Srinivasan S, Hickson-Curran SB, Berntsen DA, Howarth GF, conjunctival sensitivity in intolerant contact lens wearers. J Optom 2015;8:62–3.
Toubouti Y, et al. Lid wiper epitheliopathy in Soft contact lens wearers. Optom [281] Hiscox R, Evans K, North R, Allen L, Purslow C. Examining the effect of soft
Vis Sci 2016;93:943–54. contact lens wear on lid margin sensitivity. Cont Lens Anterior Eye 2015;38:e36.
[248] Korb DR, Herman JP, Greiner JV, Scaffidi RC, Finnemore VM, Exford JM, et al. [282] Lowther GE, Hill RM. Sensitivity threshold of the lower lid margin in the course of
Lid wiper epitheliopathy and dry eye symptoms. Eye Contact Lens 2005;31:2–8. adaptation to contact lenses. Am J Optom Arch Am Acad Optom 1968;45:587–94.
[249] Kunnen C, Lazon De La Jara P, Holden BA, Papas EB. Automated assessment of lid [283] McGowan DP, Lawrenson JG, Ruskell GL. Touch sensitivity of the eyelid margin
margin lissamine green staining. Invest Ophthalmol Vis Sci 2014;55:1976. and palpebral conjunctiva. Acta Ophthalmol 1994;72:57–60.
[250] Varikooty J, Keir N, Jones L. Optimization of assessment and grading for lid wiper [284] Navascues-Cornago M, Maldonado-Codina C, Morgan PB. Mechanical sensitivity
epitheliopathy. Optom Vis Sci 2012;89. E – abstract 120241. of the human conjunctiva. Cornea 2014;33:855–9.
[251] Knop E, Korb DR, Blackie CA, Knop N. The lid margin is an underestimated [285] Lawrenson JG, Ruskell GL. Investigation of limbal touch sensitivity using a
structure for preservation of ocular surface health and development of dry eye Cochet-Bonnet aesthesiometer. Br J Ophthalmol 1993;77:339–43.
disease. Dev Ophthalmol 2010;45:108–22. [286] Nosch DS, Joos RE, Müller D, Matt SM. General pain perception sensitivity, lid
[252] Golebiowski B, Chim K, So J, Jalbert I. Lid margins: sensitivity, staining, margin sensitivity and gas permeable contact lens comfort. Clin Exp Optom 2020;
meibomian gland dysfunction, and symptoms. Optom Vis Sci 2012;89:1443–9. 103:766–71.
[253] McMonnies CW. An examination of the relationship between ocular surface tear [287] Morgan PB, Maldonado-Codina C. Corneal staining: do we really understand what
osmolarity compartments and epitheliopathy. Ocul Surf 2015;13:110–7. we are seeing? Cont Lens Anterior Eye 2009;32:48–54.
[254] Stahl U, Jalbert I. Exploring the links between contact lens comfort, osmolarity [288] Bron AJ, Argüeso P, Irkec M, Bright FV. Clinical staining of the ocular surface:
and lid wiper staining. Cont Lens Anterior Eye 2018;41:110–6. mechanisms and interpretations. Prog Retin Eye Res 2015;44:36–61.
[255] McMonnies CW. Incomplete blinking: exposure keratopathy, lid wiper [289] Bandamwar KL, Garrett Q, Papas EB. Mechanisms of superficial micropunctate
epitheliopathy, dry eye, refractive surgery, and dry contact lenses. Cont Lens corneal staining with sodium fluorescein: the contribution of pooling. Cont Lens
Anterior Eye 2007;30:37–51. Anterior Eye 2012;35:81–4.
[256] Yamamoto Y, Shiraishi A, Sakane Y, Ohta K, Yamaguchi M, Ohashi Y. [290] Wilson G, Ren H, Laurent J. Corneal epithelial fluorescein staining. J Am Optom
Involvement of eyelid pressure in lid-wiper epitheliopathy. Curr Eye Res 2016;41: Assoc 1995;66:435–41.
171–8. [291] Bandamwar KL, Papas EB, Garrett Q. Fluorescein staining and physiological state
[257] Shaw AJ, Collins MJ, Davis BA, Carney LG. Eyelid pressure and contact with the of corneal epithelial cells. Cont Lens Anterior Eye 2014;37:213–23.
ocular surface. Invest Ophthalmol Vis Sci 2010;51:1911–7. [292] Back A. Corneal staining with contact lens wear. Cont Lens Anterior Eye 1988;11:
[258] Liu S, Dong H, Huang X-H, Tang S-H. Analysis of factors leading to lid wiper 16–8.
epitheliopathy. Eur Rev Med Pharmacol Sci 2020;24:1593–601. [293] Nichols Jj, Sinnott Lt. Tear film, contact lens, and patient factors associated with
[259] Guillon M, Maissa C. Assessment of upper and lower lid margin with lissamine corneal staining. Invest Ophthalmol Vis Sci 2011;52:1127–37.
green. Optom Vis Sci 2008;84. E – abstract 80088. [294] Schwallie JD, Mckenney CD, Long Jr WD, Mcneil A. Corneal staining patterns in
[260] Nguyen A, Hu C, Redfern R, Ritchey E. The effect of contact lens coefficient of normal non-contact lens wearers. Optom Vis Sci 1997;74:92–8.
friction on the development of lid wiper epitheliopathy. Optom Vis Sci 2018;95. E [295] Papas EB, Ciolino JB, Jacobs D, Miller WL, Pult H, Sahin A, et al. The TFOS
– abstract 185372. International Workshop on Contact Lens discomfort: report of the management
[261] Ritchey E, Powell D, Kunnen C. Diurnal variation in lid wiper epitheliopathy in and therapy subcommittee. Invest Ophthalmol Vis Sci 2013;54:TFOS183–203.
contact lens and non-contact lens wearers. Cont Lens Anterior Eye 2018;41:S74. [296] van der Worp E, De Brabander J, Swarbrick H, Nuijts R, Hendrikse F. Corneal
[262] Muntz A, Subbaraman LN, Craig JP, Jones L. Cytomorphological assessment of desiccation in rigid contact lens wear: 3- and 9-o’clock staining. Optom Vis Sci
the lid margin in relation to symptoms, contact lens wear and lid wiper 2003;80:280–90.
epitheliopathy. Ocul Surf 2020;18:214–20. [297] Chung JH. Correlation between epithelial healing rate and initial wound size in
[263] Nichols JJ, Lievens CW, Bloomenstein MR, Liu H, Simmons P, Vehige J. Dual- contact lens-induced central epithelial abrasion. Ophthalmologica 1998;212:
polymer drops, contact lens comfort, and lid wiper epitheliopathy. Optom Vis Sci 46–9.
2016;93:979–86. [298] Young G, Mirejovsky D. A hypothesis for the aetiology of soft contact lens-induced
[264] Varikooty J, Srinivasan S, Subbaraman L, Woods CA, Fonn D, Simpson TL, et al. superior arcuate keratopathy. Int Contact Lens Clin 1993;20:177–80.
Variations in observable lid wiper epitheliopathy (LWE) staining patterns in [299] Young G. Mathematical model for evaluating soft contact lens fit. Optom Vis Sci
wearers of silicone hydrogel lenses. Cont Lens Anterior Eye 2015;38:471–6. 2014;91:e167–76.
[265] Schulze MM, Srinivasan S, Hickson-Curran SB, Berntsen DA, Howarth G, [300] Diec J, Evans VE, Tilia D, Naduvilath T, Holden BA, Lazon de la Jara P.
Nichols JJ, et al. Upper lid margin staining with different soft contact lenses and Comparison of ocular comfort, vision, and SICS during silicone hydrogel contact
lens care solution combinations. Cont Lens Anterior Eye 2015;38:e15. lens daily wear. Eye Contact Lens 2012;38:2–6.
[266] Deng Z, Wang J, Jiang H, Fadli Z, Liu C, Tan J, et al. Lid wiper microvascular [301] Carnt N, Willcox MD, Evans V, Naduvilath TJ, Tilia D, Papas EB, et al. Corneal
responses as an Indicator of contact lens discomfort. Am J Ophthalmol 2016;170: staining: the IER matrix study. Contact Lens Spectrum 2007;22:38–43.
197–205. [302] Jones L, Jones D, Houlford M. Clinical comparison of three polyhexanide-
[267] Read ML, Navascues-Cornago M, Keir N, Maldonado-Codina C, Morgan PB. The preserved multi-purpose contact lens solutions. Cont Lens Anterior Eye 1997;20:
impact of contact lens wear on ocular surface mucins using a novel clinical 23–30.
fluorescence imaging system. Cont Lens Anterior Eye 2020;43:378–88. [303] Carnt N, Jalbert I, Stretton S, Naduvilath T, Papas E. Solution toxicity in soft
[268] Delaveris A, Stahl U, Madigan M, Jalbert I. Comparative performance of lissamine contact lens daily wear is associated with corneal inflammation. Optom Vis Sci
green stains. Cont Lens Anterior Eye 2018;41:23–7. 2007;84:309–15.
[269] Shaw A, Collins M, Huang J, Nguyen HMP, Kim Z, Lee G, et al. Lid wiper [304] Andrasko G, Ryen K. Corneal staining and comfort observed with traditional and
epitheliopathy: the influence of multiple lid eversions and exposure time. Cont silicone hydrogel lenses and multipurpose solution combinations. Optometry
Lens Anterior Eye 2019;42:304–10. 2008;79:444–54.
[270] Lievens CW, Norgett Y, Briggs N, Allen PM, Vianya-Estopa M. Optimal [305] Garofalo RJ, Dassanayake N, Carey C, Stein J, Stone R, David R. Corneal staining
methodology for lid wiper epitheliopathy identification. Cont Lens Anterior Eye and subjective symptoms with multipurpose solutions as a function of time. Eye
2020. Contact Lens 2005;31:166–74.
[271] Kunnen CME, Wolffsohn JS, Ritchey ER. Comparison of subjective grading of lid [306] Gorbet M, Peterson R, McCanna D, Woods C, Jones L, Fonn D. Human corneal
wiper epitheliopathy with a semi-objective method. Cont Lens Anterior Eye 2018; epithelial cell shedding and fluorescein staining in response to silicone hydrogel
41:28–33. lenses and contact lens disinfecting solutions. Curr Eye Res 2014;39:245–56.
[272] Delaveris A, Stahl U, Madigan M, Jalbert I. Comparative performance of lissamine [307] Liesegang TJ. Physiologic changes of the cornea with contact lens wear. CLAO J
green stains. Cont Lens Anterior Eye 2018. 2002;28:12–27.
[273] Guthrie SE, Jones L, Blackie CA, Korb DR. A comparative study between an oil-in- [308] Jalbert I, Stapleton F. The corneal stroma during contact lens wear. Cont Lens
water emulsion and nonlipid eye drops used for rewetting contact lenses. Eye Anterior Eye 2005;28:3–12.
Contact Lens 2015;41:373–7. [309] Holden BA, Sweeney DF, Vannas A, Nilsson KT, Efron N. Effects of long-term
[274] Elder K, Mickles C, Azhar J, Belloli A, Hans S. The effect of artificial tear extended contact lens wear on the human cornea. Invest Ophthalmol Vis Sci
treatment on lid wiper epitheliopathy and Ocular surface staining in contact lens 1985;26:1489–501.
wearers. Optom Vis Sci 2015;92. E – abstract 155246. [310] Carney LG, Brennan NA. Time course of corneal oxygen uptake during contact
lens wear. CLAO J 1988;14:151–4.

215
P.B. Morgan et al. Contact Lens and Anterior Eye 44 (2021) 192–219

[311] Hamano H, Hori M. Effect of contact lens wear on the mitoses of corneal epithelial [343] Hamrah P, Zhang Q, Liu Y, Dana MR. Novel characterization of MHC class II-
cells: preliminary report. CLAO J 1983;9:133–6. negative population of resident corneal langerhans cell-type dendritic cells. Invest
[312] Hamano H, Hori M, Hamano T, Kawabe H, Mikami M, Mitsunaga S, et al. Effects Ophthalmol Vis Sci 2002;43:639–46.
of contact lens wear on mitosis of corneal epithelium and lactate content in [344] Zhivov A, Stave J, Vollmar B, Guthoff R. In vivo confocal microscopic evaluation
aqueous humor of rabbit. Jpn J Ophthalmol 1983;27:451–8. of langerhans cell density and distribution in the corneal epithelium of healthy
[313] Lemp MA, Gold JB. The effects of extended-wear hydrophilic contact lenses on the volunteers and contact lens wearers. Cornea 2007;26:47–54.
human corneal epithelium. Am J Ophthalmol 1986;101:274–7. [345] Sindt CW, Grout TK, Critser DB, Kern JR, Meadows DL. Dendritic immune cell
[314] Cavanagh HD, Ladage PM, Li SL, Yamamoto K, Molai M, Ren DH, et al. Effects of densities in the central cornea associated with soft contact lens types and lens care
daily and overnight wear of a novel hyper oxygen-transmissible soft contact lens solution types: a pilot study. Clin Ophthalmol 2012;6:511–9.
on bacterial binding and corneal epithelium: a 13-month clinical trial. [346] López-De La Rosa A, Arroyo-Del Arroyo C, Cañadas P, López-Miguel A,
Ophthalmology 2002;109:1957–69. Calonge M, Enríquez-De-Salamanca A, et al. Are contact lens discomfort or soft
[315] Pérez JG, Méijome JMG, Jalbert I, Sweeney DF, Erickson P. Corneal epithelial contact lens material properties associated with alterations in the corneal sub-
thinning profile induced by long-term wear of hydrogel lenses. Cornea 2003;22: basal nerve plexus? Curr Eye Res 2018;43:487–92.
304–7. [347] Liu Q, Xu Z, Xu Y, Zhang J, Li Y, Xia J, et al. Changes in corneal dendritic cell and
[316] Ren DH, Petroll WM, Jester JV, Ho-Fan J, Cavanagh HD. The relationship sub-basal nerve in long-term contact lens wearers with dry eye. Eye Contact Lens
between contact lens oxygen permeability and binding of Pseudomonas 2020;46:238–44.
aeruginosa to human corneal epithelial cells after overnight and extended wear. [348] Sankaridurg PR, Rao GN, Rao HN, Sweeney DF, Holden BA. ATPase-positive
CLAO J 1999;25:80–100. dendritic cells in the limbal and corneal epithelium of guinea pigs after extended
[317] Ladage PM, Yamamoto K, Ren DH, Li L, Jester JV, Petroll WM, et al. Effects of wear of hydrogel lenses. Cornea 2000;19:374–7.
rigid and soft contact lens daily wear on corneal epithelium, tear lactate [349] Hazlett LD, McClellan SM, Hume EB, Dajcs JJ, O’Callaghan RJ, Willcox MD.
dehydrogenase, and bacterial binding to exfoliated epithelial cells. Extended wear contact lens usage induces langerhans cell migration into cornea.
Ophthalmology 2001;108:1279–88. Exp Eye Res 1999;69:575–7.
[318] Ren DH, Petroll WM, Jester JV, Ho-Fan J, Cavanagh HD. Short-term hypoxia [350] Alzahrani Y, Pritchard N, Efron N. Changes in corneal langerhans cell density
downregulates epithelial cell desquamation in vivo, but does not increase during the first few hours of contact lens wear. Cont Lens Anterior Eye 2016;39:
Pseudomonas aeruginosa adherence to exfoliated human corneal epithelial cells. 307–10.
CLAO J 1999;25:73–9. [351] Jamali A, Harris DL, Blanco T, Lopez MJ, Hamrah P. Resident plasmacytoid
[319] Ladage PM, Yamamoto K, Ren DH, Li L, Jester JV, Petroll WM, et al. Proliferation dendritic cells patrol vessels in the naïve limbus and conjunctiva. Ocul Surf 2020;
rate of rabbit corneal epithelium during overnight rigid contact lens wear. Invest 18:277–85.
Ophthalmol Vis Sci 2001;42:2804–12. [352] Boberg-Ans J. Experience in clinical examination of corneal sensitivity; corneal
[320] O’Leary DJ, Madgewick R, Wallace J, Ang J. Size and number of epithelial cells sensitivity and the naso-lacrimal reflex after retrobulbar anaesthesia. Br J
washed from the cornea after contact lens wear. Optom Vis Sci 1998;75:692–6. Ophthalmol 1955;39:705–26.
[321] Ren H, Petroll WM, Jester JV, Cavanagh HD, Mathers WD, Bonanno JA, et al. [353] Schirmer KE. Corneal sensitivity and contact lenses. Br J Ophthalmol 1963;47:
Adherence of Pseudomonas aeruginosa to shed rabbit corneal epithelial cells after 493–5.
overnight wear of contact lenses. CLAO J 1997;23:63–8. [354] Fujita H, Sano K, Sasaki S, Ohno-Matsui K, Tanaka T, Baba T, et al. Ocular
[322] Ren DH, Yamamoto K, Ladage PM, Molai M, Li L, Petroll WM, et al. Adaptive discomfort at the initial wearing of rigid gas permeable contact lenses. Jpn J
effects of 30-night wear of hyper-O(2) transmissible contact lenses on bacterial Ophthalmol 2004;48:376–9.
binding and corneal epithelium: a 1-year clinical trial. Ophthalmology 2002;109: [355] Millodot M. Effect of hard contact lenses on corneal sensitivity and thickness. Acta
27–39. discussion 39–40. Ophthalmol 1975;53:576–84.
[323] Wilson G, Fatt I. Thickness of the corneal epithelium during anoxia. Am J Optom [356] Millodot M. Does the long term wear of contact lenses produce a loss of corneal
Physiol Opt 1980;57:409–12. sensitivity? Experientia 1977;33:1475–6.
[324] O’Leary DJ, Wilson G, Henson DB. The effect of anoxia on the human corneal [357] Millodot M. Effect of long-term wear of hard contact lenses on corneal sensitivity.
epithelium. Am J Optom Physiol Opt 1981;58:472–6. Arch Ophthalmol 1978;96:1225–7.
[325] Wang J, Fonn D, Simpson TL, Jones L. The measurement of corneal epithelial [358] Polse KA. Etiology of corneal sensitivity changes accompanying contact lens wear.
thickness in response to hypoxia using optical coherence tomography. Am J Invest Ophthalmol Vis Sci 1978;17:1202–6.
Ophthalmol 2002;133:315–9. [359] Millodot M. Long-term wear of hard contact lenses and corneal integrity.
[326] Wang J, Fonn D, Simpson TL. Topographical thickness of the epithelium and total Contactologia 1978;22:8–12.
cornea after hydrogel and PMMA contact lens wear with eye closure. Invest [360] Millodot M, Henson DB, O’Leary DJ. Measurement of corneal sensitivity and
Ophthalmol Vis Sci 2003;44:1070–4. thickness with PMMA and gas-permeable contact lenses. Am J Optom Physiol Opt
[327] Lambert SR, Klyce SD. The origins of Sattler’s veil. Am J Ophthalmol 1981;91: 1979;56:628–32.
51–6. [361] Sanaty M, Temel A. Corneal sensitivity changes in long-term wearing of hard
[328] Zantos SG. Cystic formations in the corneal epithelium during extended wear of polymethylmethacrylate contact lenses. Ophthalmologica 1998;212:328–30.
contact lenses. Int Contact Lens Clin 1983;10:128. [362] Murphy PJ, Patel S, Marshall J. The effect of long-term, daily contact lens wear on
[329] Holden BA, Sweeney DF. The significance of the microcyst response: a review. corneal sensitivity. Cornea 2001;20:264–9.
Optom Vis Sci 1991;68:703–7. [363] Lum E, Golebiowski B, Gunn R, Babhoota M, Swarbrick H. Corneal sensitivity
[330] Tripathi RC, Bron AJ. Ultrastructural study of non-traumatic recurrent corneal with contact lenses of different mechanical properties. Optom Vis Sci 2013;90:
erosion. Br J Ophthalmol 1972;56:73–85. 954–60.
[331] Lowe R, Brennan NA. Corneal wrinkling caused by a thin medium water content [364] Douthwaite WA, Connelly AT. The effect of hard and gas permeable contact lenses
lens. Int Contact Lens Clin 1987;14:403–6. on refractive error, corneal curvature, thickness and sensitivity. Cont Lens
[332] Giese MJ. Corneal wrinkling in a hydrogel contact lens wearer with Marfan Anterior Eye 1986;9:14–20.
syndrome. J Am Optom Assoc 1997;68:50–4. [365] Tanelian DL, Beuerman RW. Recovery of corneal sensation following hard contact
[333] Efron N, Morgan PB. Rethinking contact lens aftercare. Clin Exp Optom 2017;100: lens wear and the implication for adaptation. Invest Ophthalmol Vis Sci 1980;19:
411–31. 1391–4.
[334] Bruce AS, Brennan NA. Corneal pathophysiology with contact lens wear. Surv [366] Alipour F, Soleimanzade M, Latifi G, Aghaie SH, Kasiri M, Dehghani S. Effects of
Ophthalmol 1990;35:25–58. soft toric, rigid gas-permeable, and mini-scleral lenses on corneal microstructure
[335] Phillips CI. Contact lenses and corneal deformation: cause, correlate or co- using confocal microscopy. Eye Contact Lens 2020;46:74–81.
incidence? Acta Ophthalmol 1990;68:661–8. [367] Lum E, Golebiowski B, Gunn R, Babhoota M, Swarbrick H. Corneal sensitivity
[336] Wilson SE, Lin DT, Klyce SD, Reidy JJ, Insler MS. Rigid contact lens decentration: with contact lenses of different mechanical properties. Optom Vis Sci 2013;90:
a risk factor for corneal warpage. CLAO J 1990;16:177–82. 954–60.
[337] Bucci Jr FA, Evans RE, Moody KJ, Tanner JB, Capozza RC, Klyce SD. The annular [368] Lum E, Golebiowski B, Swarbrick HA. Changes in corneal subbasal nerve
tinted contact lens syndrome: corneal topographic analysis of ring-shaped morphology and sensitivity during orthokeratology: onset of change. Ocul Surf
irregular astigmatism caused by annular tinted contact lenses. CLAO J 1997;23: 2017;15:227–35.
161–7. [369] Lum E, Golebiowski B, Swarbrick HA. Changes in corneal subbasal nerve
[338] Schanzer MC, Mehta RS, Arnold TP, Zuckerbrod SL, Koch DD. Irregular morphology and sensitivity during orthokeratology: recovery of change. Ocul Surf
astigmatism induced by annular tinted contact lenses. CLAO J 1989;15:207–11. 2017;15:236–41.
[339] Wang X, McCulley JP, Bowman RW, Cavanagh HD. Time to resolution of contact [370] Nieto-Bona A, González-Mesa A, Nieto-Bona MP, Villa-Collar C, Lorente-
lens-induced corneal warpage prior to refractive surgery. CLAO J 2002;28: Velázquez A. Long-term changes in corneal morphology induced by overnight
169–71. orthokeratology. Curr Eye Res 2011;36:895–904.
[340] Tseng SS-Y, Hsiao JC-J, Chang DC-K. Mistaken diagnosis of keratoconus because [371] Lum E, Golebiowski B, Swarbrick HA. Reduced corneal sensitivity and sub-basal
of corneal warpage induced by hydrogel lens wear. Cornea 2007;26:1153–5. nerve density in long-term orthokeratology lens wear. Eye Contact Lens 2017;43:
[341] Tang M, Li Y, Chamberlain W, Louie DJ, Schallhorn JM, Huang D. Differentiating 218–24.
keratoconus and corneal warpage by analyzing focal change patterns in corneal [372] Nombela-Palomo M, Felipe-Marquez G, Teus MA, Hernandez-Verdejo JL, Nieto-
topography, pachymetry, and epithelial thickness maps. Invest Ophthalmol Vis Bona A. Long-term impacts of orthokeratology treatment on sub-basal nerve
Sci 2016;57:OCT544–9. plexus and corneal sensitivity responses and their reversibility. Eye Contact Lens
[342] Stapleton F, Keay L, Jalbert I, Cole N. The epidemiology of contact lens related 2018;44:91–6.
infiltrates. Optom Vis Sci 2007;84:257–72.

216
P.B. Morgan et al. Contact Lens and Anterior Eye 44 (2021) 192–219

[373] Wang Y, Kornberg DL, St Clair RM, Lee M, Muhic I, Ciralsky JB, et al. Corneal [409] Ozkan J, Mandathara P, Krishna P, Sankaridurg P, Naduvilath T, Willcox MDP,
nerve structure and function after long-term wear of fluid-filled scleral lens. et al. Risk factors for corneal inflammatory and mechanical events with extended
Cornea 2015;34:427–32. wear silicone hydrogel contact lenses. Optom Vis Sci 2010;87:847–53.
[374] Compañ V, Oliveira C, Aguilella-Arzo M, Mollá S, Peixoto-de-Matos SC, González- [410] Moezzi AM, Fonn D, Varikooty J, Simpson TL. Overnight corneal swelling with
Méijome JM. Oxygen diffusion and edema with modern scleral rigid gas high and low powered silicone hydrogel lenses. J Optom 2015;8:19–26.
permeable contact lenses. Invest Ophthalmol Vis Sci 2014;55:6421–9. [411] Beer F, Wartak A, Pircher N, Holzer S, Lammer J, Schmidinger G, et al. Mapping
[375] Millodot M. Effect of soft lenses on corneal sensitivity. Acta Ophthalmol 1974;52: of corneal layer thicknesses with polarization-sensitive optical coherence
603–8. tomography using a conical scan pattern. Invest Ophthalmol Vis Sci 2018;59:
[376] Velasco MJ, Bermúdez FJ, Romero J, Hita E. Variations in corneal sensitivity with 5579–88.
hydrogel contact lenses. Acta Ophthalmol 1994;72:53–6. [412] Hart RW, Farrell RA. Light scattering in the cornea. J Opt Soc Am 1969;59:
[377] Larke JR, Hirji NK. Some clinically observed phenomena in extended contact lens 766–74.
wear. Br J Ophthalmol 1979;63:475–7. [413] Piatigorsky J. Gene sharing in lens and cornea: facts and implications. Prog Retin
[378] du Toit R, Situ P, Simpson T, Fonn D. The effects of six months of contact lens Eye Res 1998;17:145–74.
wear on the tear film, ocular surfaces, and symptoms of presbyopes. Optom Vis [414] Müller LJ, Pels L, Vrensen GF. Novel aspects of the ultrastructural organization of
Sci 2001;78:455–62. human corneal keratocytes. Invest Ophthalmol Vis Sci 1995;36:2557–67.
[379] Basuthkar Sundar Rao S, Simpson TL. Measurement of difference thresholds on [415] Fatt I, Bieber MT. The steady-state distribution of oxygen and carbon dioxide in
the ocular surface. Invest Ophthalmol Vis Sci 2014;55:1095–100. the in vivo cornea. I. The open eye in air and the closed eye. Exp Eye Res 1968;7:
[380] Beuerman RW, Rozsa AJ. Threshold and signal detection measurements of the 103–12.
effect of soft contact lenses on corneal sensitivity. Curr Eye Res 1985;4:742–4. [416] Riley MV. Glucose and oxygen utilization by the rabbit cornea. Exp Eye Res 1969;
[381] Patel SV, McLaren JW, Hodge DO, Bourne WM. Confocal microscopy in vivo in 8:193–200.
corneas of long-term contact lens wearers. Invest Ophthalmol Vis Sci 2002;43: [417] Freeman RD. Oxygen consumption by the component layers of the cornea.
995–1003. J Physiol 1972;225:15–32.
[382] Oliveira-Soto L, Efron N. Morphology of corneal nerves in soft contact lens wear. [418] Klyce SD. Stromal lactate accumulation can account for corneal oedema
A comparative study using confocal microscopy. Ophthalmic Physiol Opt 2003; osmotically following epithelial hypoxia in the rabbit. J Physiol 1981;321:49–64.
23:163–74. [419] Rohde MD, Huff JW. Contact lens-induced edema in vitro–amelioration by lactate
[383] Hu L, Chen J, Zhang L, Sun X, Huang J, Xie W, et al. Effects of long-term soft dehydrogenase inhibitors. Curr Eye Res 1986;5:751–8.
contact lenses on tear menisci and corneal nerve density. Eye Contact Lens 2016; [420] Huff JW. Effects of sodium lactate on isolated rabbit corneas. Invest Ophthalmol
42:196–201. Vis Sci 1990;31:942–7.
[384] Millodot M. A review of research on the sensitivity of the cornea. Ophthalmic [421] Huff JW. Contact lens-induced edema in vitro. Ion transport and metabolic
Physiol Opt 1984;4:305–18. considerations. Invest Ophthalmol Vis Sci 1990;31:1288–93.
[385] Ntola A, Murphy PJ. The effect of contact lens wear on corneal sensation. Minerva [422] Mertz GW. Overnight swelling of the living human cornea. J Am Optom Assoc
Oftalmol 2002;44:31–8. 1980;51:211–4.
[386] Millodot M, O’Leary DJ. Loss of corneal sensitivity with lid closure in humans. [423] Holden BA, Mertz GW, McNally JJ. Corneal swelling response to contact lenses
Exp Eye Res 1979;29:417–21. worn under extended wear conditions. Invest Ophthalmol Vis Sci 1983;24:
[387] Millodot M, O’Leary DJ. Effect of oxygen deprivation on corneal sensitivity. Acta 218–26.
Ophthalmol 1980;58:434–9. [424] Cox I, Zantos SG, Orsborn GN. The overnight corneal swelling response of non-
[388] Yamada M, Ogata M, Kawai M, Mashima Y. Decreased substance P concentrations wear, daily wear, and extended wear soft lens patients. Int Contact Lens Clin
in tears from patients with corneal hypesthesia. Am J Ophthalmol 2000;129: 1990;17:134–8.
671–2. [425] Mandell RB, Fatt I. Thinning of the human cornea on awakening. Nature 1965;
[389] Liu Q, McDermott AM, Miller WL. Elevated nerve growth factor in dry eye 208:292–3.
associated with established contact lens wear. Eye Contact Lens 2009;35:232–7. [426] O’Neal MR, Polse KA. In vivo assessment of mechanisms controlling corneal
[390] Fitzgerald GG, Cooper JR. Acetylcholine as a possible sensory mediator in rabbit hydration. Invest Ophthalmol Vis Sci 1985;26:849–56.
corneal epithelium. Biochem Pharmacol 1971;20:2741–8. [427] Sakamoto R, Miyanaga Y, Hamano H. Soft and RGP lens corneal swelling and
[391] Mindel JS, Mittag TW. Suppression of corneal epithelial choline acetyltransferase deswelling with overnight wear. Int Contact Lens Clin 1991;18:214–8.
activity by lid closure. Exp Eye Res 1978;27:359–64. [428] Fonn D, du Toit R, Simpson TL, Vega JA, Situ P, Chalmers RL. Sympathetic
[392] Bergenske PD, Polse KA. The effect of rigid gas permeable lenses on corneal swelling response of the control eye to soft lenses in the other eye. Invest
sensitivity. J Am Optom Assoc 1987;58:212–5. Ophthalmol Vis Sci 1999;40:3116–21.
[393] Chen J, Feng Y, Simpson TL. Human corneal adaptation to mechanical, cooling, [429] du Toit R, Vega JA, Fonn D, Simpson T. Diurnal variation of corneal sensitivity
and chemical stimuli. Invest Ophthalmol Vis Sci 2010;51:876–81. and thickness. Cornea 2003;22:205–9.
[394] Langham M. Utilization of oxygen by the component layers of the living cornea. [430] Hedbys BO, Mishima S. Flow of water in the corneal stroma. Exp Eye Res 1962;1:
J Physiol 1952;117:461–70. 262–75.
[395] Turss R, Friend J, Dohlman CH. Effect of a corneal fluid barrier on the nutrition of [431] Hedbys BO, Mishima S. The thickness-hydration relationship of the cornea. Exp
the epithelium. Exp Eye Res 1970;9:254–9. Eye Res 1966;5:221–8.
[396] Bonanno JA. Identity and regulation of ion transport mechanisms in the corneal [432] Holden BA. The Glenn A. Fry Award lecture 1988: the ocular response to contact
endothelium. Prog Retin Eye Res 2003;22:69–94. lens wear. Optom Vis Sci 1989;66:717–33.
[397] Papas E. On the relationship between soft contact lens oxygen transmissibility and [433] Holden BA, Mertz GW. Critical oxygen levels to avoid corneal edema for daily and
induced limbal hyperaemia. Exp Eye Res 1998;67:125–31. extended wear contact lenses. Invest Ophthalmol Vis Sci 1984;25:1161–7.
[398] Chang JH, Gabison EE, Kato T, Azar DT. Corneal neovascularization. Curr Opin [434] Fonn D, Sweeney D, Holden BA, Cavanagh D. Corneal oxygen deficiency. Eye
Ophthalmol 2001;12:242–9. Contact Lens 2005;31:23–7.
[399] Azar DT. Corneal angiogenic privilege: angiogenic and antiangiogenic factors in [435] Morgan PB, Brennan NA, Maldonado-Codina C, Quhill W, Rashid K, Efron N.
corneal avascularity, vasculogenesis, and wound healing (an American Central and peripheral oxygen transmissibility thresholds to avoid corneal
Ophthalmological Society thesis). Trans Am Ophthalmol Soc 2006;104:264–302. swelling during open eye soft contact lens wear. J Biomed Mater Res B Appl
[400] Dawson DW, Volpert OV, Gillis P, Crawford SE, Xu H, Benedict W, et al. Pigment Biomater 2010;92:361–5.
epithelium-derived factor: a potent inhibitor of angiogenesis. Science 1999;285: [436] Moezzi AM, Varikooty J, Schulze M, Ngo W, Lorenz KO, Boree D, et al. Corneal
245–8. swelling with cosmetic etafilcon a lenses versus no lens wear. Optom Vis Sci 2016;
[401] Roshandel D, Eslani M, Baradaran-Rafii A, Cheung AY, Kurji K, Jabbehdari S, 93:619–28.
et al. Current and emerging therapies for corneal neovascularization. Ocul Surf [437] Moezzi AM, Varikooty J, Luensmann D, Schulze M-M, Ng A, Karkkainen T, et al.
2018;16:398–414. The short-term physiological impact of switching reusable silicone hydrogel
[402] Donnenfeld ED, Ingraham H, Perry HD, Imundo M, Goldberg LP. Contact lens- wearers into a hydrogel daily disposable multifocal. Clin Ophthalmol 2019;13:
related deep stromal intracorneal hemorrhage. Ophthalmology 1991;98:1793–6. 1193–202.
[403] Keech PM, Ichikawa L, Barlow W. A prospective study of contact lens [438] Edelhauser HF. The balance between corneal transparency and edema: the
complications in a managed care setting. Optom Vis Sci 1996;73:653–8. proctor lecture. Invest Ophthalmol Vis Sci 2006;47:1754–67.
[404] Dumbleton K, Keir N, Moezzi A, Feng Y, Jones L, Fonn D. Objective and subjective [439] Cristol SM, Edelhauser HF, Lynn MJ. A comparison of corneal stromal edema
responses in patients refitted to daily-wear silicone hydrogel contact lenses. induced from the anterior or the posterior surface. Refract Corneal Surg 1992;8:
Optom Vis Sci 2006;83:758–68. 224–9.
[405] Yeung KK, Yang HJ, Nguyen AL, Weissman BA. Critical contact lens oxygen [440] Van Horn DL, Doughman DJ, Harris JE, Miller GE, Lindstrom R, Good RA.
transmissibility and tear lens oxygen tension to preclude corneal Ultrastructure of human organ-cultured cornea. II. Stroma and epithelium. Arch
neovascularization. Eye Contact Lens 2018;44(Suppl 1):S291–5. Ophthalmol 1975;93:275–7.
[406] Moezzi AM, Fonn D, Varikooty J, Richter D. Distribution of overnight corneal [441] Patel S, Marshall J, Fitzke 3rd FW. Refractive index of the human corneal
swelling across subjects with 4 different silicone hydrogel lenses. Eye Contact epithelium and stroma. J Refract Surg 1995;11:100–5.
Lens 2011;37:61–5. [442] Kikkawa Y, Hirayama K. Uneven swelling of the corneal stroma. Invest
[407] Sweeney DF. Have silicone hydrogel lenses eliminated hypoxia? Eye Contact Lens Ophthalmol 1970;9:735–41.
2013;39:53–60. [443] Lee D, Wilson G. Non-uniform swelling properties of the corneal stroma. Curr Eye
[408] Moezzi AM, Hutchings N, Fonn D, Simpson TL. Mixed model analysis of between- Res 1981;1:457–61.
subject variability in overnight corneal swelling and deswelling with silicone [444] Komai Y, Ushiki T. The three-dimensional organization of collagen fibrils in the
hydrogel lenses. Invest Ophthalmol Vis Sci 2018;59:2576–85. human cornea and sclera. Invest Ophthalmol Vis Sci 1991;32:2244–58.

217
P.B. Morgan et al. Contact Lens and Anterior Eye 44 (2021) 192–219

[445] Müller LJ, Pels E, Vrensen GF. The specific architecture of the anterior stroma [482] Carlson KH, Bourne WM, Brubaker RF. Effect of long-term contact lens wear on
accounts for maintenance of corneal curvature. Br J Ophthalmol 2001;85:437–43. corneal endothelial cell morphology and function. Invest Ophthalmol Vis Sci
[446] Müller LJ, Pels E, Schurmans LRHM, Vrensen GFJM. A new three-dimensional 1988;29:185–93.
model of the organization of proteoglycans and collagen fibrils in the human [483] Nieuwendaal CP, Odenthal MT, Kok JH, Venema HW, Oosting J, Riemslag FC,
corneal stroma. Exp Eye Res 2004;78:493–501. et al. Morphology and function of the corneal endothelium after long-term
[447] Turss R, Friend J, Reim M, Dohlman CH. Glucose concentration and hydration of contact lens wear. Invest Ophthalmol Vis Sci 1994;35:3071–7.
the corneal stroma. Ophthalmic Res 1971;2:253–60. [484] Harvitt DM, Bonanno JA. Re-evaluation of the oxygen diffusion model for
[448] Castoro JA, Bettelheim AA, Bettelheim FA. Water gradients across bovine cornea. predicting minimum contact lens Dk/t values needed to avoid corneal anoxia.
Invest Ophthalmol Vis Sci 1988;29:963–8. Optom Vis Sci 1999;76:712–9.
[449] Bettelheim FA, Plessy B. The hydration of proteoglycans of bovine cornea. [485] Sharifipour F, Idani E, Zamani M, Helmi T, Cheraghian B. Oxygen tension in the
Biochim Biophys Acta 1975;381:203–14. aqueous humor of human eyes under different oxygenation conditions.
[450] Meek KM, Boote C. The organization of collagen in the corneal stroma. Exp Eye J Ophthalmic Vis Res 2013;8:119–25.
Res 2004;78:503–12. [486] Bonanno JA. Effects of contact lens-induced hypoxia on the physiology of the
[451] Maurice DM. The structure and transparency of the cornea. J Physiol 1957;136: corneal endothelium. Optom Vis Sci 2001;78:783–90.
263–86. [487] MacRae SM, Matsuda M, Shellans S. Corneal endothelial changes associated with
[452] Maurice DM. The location of the fluid pump in the cornea. J Physiol 1972;221: contact lens wear. CLAO J 1989;15:82–7.
43–54. [488] Chang SW, Hu FR, Lin LL. Effects of contact lenses on corneal endothelium - a
[453] McLaren JW, Wacker K, Kane KM, Patel SV. Measuring corneal haze by using morphological and functional study. Ophthalmologica 2001;215:197–203.
scheimpflug photography and confocal microscopy. Invest Ophthalmol Vis Sci [489] Lee JS, Park WS, Lee SH, Oum BS, Cho BM. A comparative study of corneal
2016;57:227–35. endothelial changes induced by different durations of soft contact lens wear.
[454] Wechsler S. Striate corneal lines. Am J Optom Physiol Opt 1974;51:852–6. Graefes Arch Clin Exp Ophthalmol 2001;239:1–4.
[455] Polse KA, Mandell RB. Etiology of corneal striae accompanying hydrogel lens [490] Doughty MJ, Aakre BM, Ystenaes AE, Svarverud E. Short-term adaptation of the
wear. Invest Ophthalmol 1976;15:553–6. human corneal endothelium to continuous wear of silicone hydrogel (lotrafilcon
[456] Hine N, Holden BA. Performance of low versus high water content plus lenses for A) contact lenses after daily hydrogel lens wear. Optom Vis Sci 2005;82:473–80.
presbyopes. Int Contact Lens Clin 1992;19:257–63. [491] Mohidin N, Hoon NL, Ishak B, Mohd-Ali B. Cornea endothelial cell morphology in
[457] La Hood D, Grant T. Striae and folds as indicators of corneal edema. Optom Vis Sci short-term silicone hydrogel soft contact lens wearers – asian context. n.d.. 2021.
1990;67(S):196. [492] Papas EB. The significance of oxygen during contact lens wear. Cont Lens Anterior
[458] Efron N, Mutalib HA, Perez-Gomez I, Koh HH. Confocal microscopic observations Eye 2014;37:394–404.
of the human cornea following overnight contact lens wear. Clin Exp Optom [493] Ahmad A, Mohd-Ali B, Ishak B. Changes in the morphology of corneal endothelial
2002;85:149–55. cells in young myopic adults after 6 months of wearing soft contact lenses: a
[459] Martin R, Izquierdo M, Saber A. Investigation of posterior corneal curvature in Malaysian perspective. Clin Optom (Auckl) 2018;10:109–13.
CL-induced corneal swelling. Cont Lens Anterior Eye 2009;32:288–93. [494] McMonnies CW, Zantos SG. Endothelial bedewing of the cornea in association
[460] Moezzi AM, Fonn D, Simpson TL, Sorbara L. Contact lens-induced corneal with contact lens wear. Br J Ophthalmol 1979;63:478–81.
swelling and surface changes measured with the Orbscan II corneal topographer. [495] Hickson S, Papas E. Prevalence of idiopathic corneal anomalies in a non contact
Optom Vis Sci 2004;81:189–93. lens-wearing population. Optom Vis Sci 1997;74:293–7.
[461] Tyagi G, Collins M, Read S, Davis B. Regional changes in corneal thickness and [496] Efron N, editor. Chapter 27: endothelial bedewing. Contact lens complications.
shape with soft contact lenses. Optom Vis Sci 2010;87:567–75. 3rd ed. Edinburgh: Elsevier Saunders; 2012. p. 272–7.
[462] Brennan NA, Coles MLC, Comstock TL, Levy B. A 1-year prospective clinical trial [497] Zantos SG, Holden BA. Transient endothelial changes soon after wearing soft
of balafilcon a (PureVision) silicone-hydrogel contact lenses used on a 30-day contact lenses. Optom Vis Sci 1977;54:856–8.
continuous wear schedule. Ophthalmology 2002;109:1172–7. [498] Efron N. Contact lens-induced changes in the anterior eye as observed in vivo
[463] Hirji NK, Larke JR. Corneal thickness in extended wear of soft contact lenses. Br J with the confocal microscope. Prog Retin Eye Res 2007;26:398–436.
Ophthalmol 1979;63:274–6. [499] Ohya S, Nishimaki K, Nakayasu K, Kanai A. Non-contact specular microscopic
[464] Schoessler JP, Barr JT. Corneal thickness changes with extended contact lens observation for early response of corneal endothelium after contact lens wear.
wear. Am J Optom Physiol Opt 1980;57:729–33. CLAO J 1996;22:122–6.
[465] González-Méijome JM, González-Pérez J, Cerviño A, Yebra-Pimentel E, [500] Efron N, Kotow M, Martin DK, Holden BA. Physiological response of the
Parafita MA. Changes in corneal structure with continuous wear of high-Dk soft contralateral cornea to monocular hydrogel contact lens wear. Optom Vis Sci
contact lenses: a pilot study. Optom Vis Sci 2003;80:440–6. 1984;61:517–22.
[466] Gürdal C, Aydin S, Kirimlioğlu H, Toprak E, Sengör T. Effects of extended-wear [501] Schoessler JP, Woloschak MJ, Mauger TF. Transient endothelial changes
soft contact lenses on the ocular surface and central corneal thickness. produced by hydrophilic contact lenses. Am J Optom Physiol Opt 1982;59:764–5.
Ophthalmologica 2003;217:329–36. [502] Holden BA, Williams L, Zantos SG. The etiology of transient endothelial changes
[467] Efron N, Perez-Gomez I, Morgan PB. Confocal microscopic observations of in the human cornea. Invest Ophthalmol Vis Sci 1985;26:1354–9.
stromal keratocytes during extended contact lens wear. Clin Exp Optom 2002;85: [503] Giasson CJ, Rancourt J, Robillard J, Melillo M, Michaud L. Corneal endothelial
156–60. blebs induced in scleral lens wearers. Optom Vis Sci 2019;96:810–7.
[468] Kallinikos P, Efron N. On the etiology of keratocyte loss during contact lens wear. [504] Hamano H, Maeda N, Hamano T, Mitsunaga S, Kotani S. Corneal thickness change
Invest Ophthalmol Vis Sci 2004;45:3011–20. induced by dozing while wearing hydrogel and silicone hydrogel lenses. Eye
[469] Kallinikos P, Morgan P, Efron N. Assessment of stromal keratocytes and tear film Contact Lens 2008;34:56–60.
inflammatory mediators during extended wear of contact lenses. Cornea 2006;25: [505] Hamano H, Jacob JT, Senft CJ, Hamano T, Hamano T, Mitsunaga S, et al.
1–10. Differences in contact lens-induced responses in the corneas of Asian and non-
[470] Ohta K, Shimamura I, Shiraishi A, Ohashi Y. Confocal microscopic observations of Asian subjects. CLAO J 2002;28:101–4.
stromal keratocytes in soft and rigid contact lens wearers. Cornea 2012;31:66–73. [506] Brooks AMV, Grant G, et al. Differentiation and assessment of corneal endothelial
[471] Mergler S, Pleyer U. The human corneal endothelium: new insights into changes associated with diseases of the anterior segment of the eye. Aust N Z J
electrophysiology and ion channels. Prog Retin Eye Res 2007;26:359–78. Ophthalmol 1987;15:65–70.
[472] Laing RA, Sandstrom MM, Berrospi AR, Leibowitz HM. Changes in the corneal [507] Bergmanson JPG, Sheldon TM, Goosey JD. Fuchs’ endothelial dystrophy: a fresh
endothelium as a function of age. Exp Eye Res 1976;22:587–94. look at an aging disease. Ophthalmic Physiol Opt 1999;19:210–22.
[473] Abib FC, Barreto Junior J. Behavior of corneal endothelial density over a lifetime. [508] Efron N, editor. Chapter 28: endothelial blebs. Contact lens complications. 3rd ed.
J Cataract Refract Surg 2001;27:1574–8. Edinburgh: Elsevier Saunders; 2012. p. 278–84.
[474] Roszkowska AM, Colosi P, D’Angelo P, Ferreri G. Age-related modifications of the [509] Mountford J, Sc DA. An analysis of the changes in Cornea1 shape and refractive
corneal endothelium in adults. Int Ophthalmol 2004;25:163–6. error induced by accelerated orthokeratology. ICLC 1997;24.
[475] Niederer RL, Perumal D, Sherwin T, McGhee CNJ. Age-related differences in the [510] Swarbrick HA, Wong G, O’Leary DJ. Corneal response to orthokeratology. Optom
normal human cornea: a laser scanning in vivo confocal microscopy study. Br J Vis Sci 1998;75:791–9.
Ophthalmol 2007;91:1165–9. [511] Nichols JJ, Marsich MM, Nguyen M, Barr JT, Bullimore MA. Overnight
[476] American Academy of Ophthalmology. Corneal endothelial photography. orthokeratology. Optom Vis Sci 2000;77:252–9.
Ophthalmology 1991;98:1464–8. [512] Lu F, Sorbara L, Simpson T, Fonn D. Corneal shape and optical performance after
[477] Senoo T, Joyce NC. Cell cycle kinetics in corneal endothelium from old and young one night of corneal refractive therapy for hyperopia. Optom Vis Sci 2007;84:
donors. Invest Ophthalmol Vis Sci 2000;41:660–7. 357–64.
[478] Feizi S. Corneal endothelial cell dysfunction: etiologies and management. Ther [513] Gifford P, Swarbrick HA. Time course of corneal topographic changes in the first
Adv Ophthalmol 2018;10. 2515841418815802. week of overnight hyperopic orthokeratology. Optom Vis Sci 2008;85:1165–71.
[479] Bergmanson JP. Histopathological analysis of corneal endothelial polymegethism. [514] Owens H, Garner LF, Craig JP, Gamble G. Posterior corneal changes with
Cornea 1992;11:133–42. orthokeratology. Optom Vis Sci 2004;81:421–6.
[480] González-Méijome JM, Jorge J, Queirós A, Peixoto-de-Matos SC, Parafita MA. [515] Yoon JH, Swarbrick HA. Posterior corneal shape changes in myopic overnight
Two single descriptors of endothelial polymegethism and pleomorphism. Graefes orthokeratology. Optom Vis Sci 2013;90:196–204.
Arch Clin Exp Ophthalmol 2010;248:1159–66. [516] Tsukiyama J, Miyamoto Y, Higaki S, Fukuda M, Shimomura Y. Changes in the
[481] Fatt I, Freeman RD, Lin D. Oxygen tension distributions in the cornea: a re- anterior and posterior radii of the corneal curvature and anterior chamber depth
examination. Exp Eye Res 1974;18:357–65. by orthokeratology. Eye Contact Lens 2008;34:17–20.

218
P.B. Morgan et al. Contact Lens and Anterior Eye 44 (2021) 192–219

[517] Chen D, Lam AKC, Cho P. Posterior corneal curvature change and recovery after 6 [549] Lee YC, Wang JH, Chiu CJ. Effect of Orthokeratology on myopia progression:
months of overnight orthokeratology treatment. Ophthalmic Physiol Opt 2010; twelve-year results of a retrospective cohort study. BMC Ophthalmol 2017;17:
30:274–80. 243.
[518] Gifford P, Alharbi A, Swarbrick HA. Corneal thickness changes in hyperopic [550] Kakita T, Hiraoka T, Oshika T. Influence of overnight orthokeratology on axial
orthokeratology measured by optical pachometry. Invest Ophthalmol Vis Sci elongation in childhood myopia. Invest Ophthalmol Vis Sci 2011;52:2170–4.
2011;52:3648–53. [551] Cho P, Cheung SW. Retardation of myopia in Orthokeratology (ROMIO) study: a
[519] Munnerlyn CR, Koons SJ, Marshall J. Photorefractive keratectomy: a technique 2-year randomized clinical trial. Invest Ophthalmol Vis Sci 2012;53:7077–85.
for laser refractive surgery. J Cataract Refract Surg 1988;14:46–52. [552] Santodomingo-Rubido J, Villa-Collar C, Gilmartin B, Gutierrez-Ortega R. Myopia
[520] Alharbi A, Swarbrick HA. The effects of overnight orthokeratology lens wear on control with orthokeratology contact lenses in Spain: refractive and biometric
corneal thickness. Invest Ophthalmol Vis Sci 2003;44:2518–23. changes. Invest Ophthalmol Vis Sci 2012;53:5060–5.
[521] Alharbi A, La Hood D, Swarbrick HA. Overnight orthokeratology lens wear can [553] Charm J, Cho P. High myopia-partial reduction ortho-k: a 2-year randomized
inhibit the central stromal edema response. Invest Ophthalmol Vis Sci 2005;46: study. Optom Vis Sci 2013;90:530–9.
2334–40. [554] Chen C, Cheung SW, Cho P. Myopia control using toric orthokeratology (TO-SEE
[522] Haque S, Fonn D, Simpson T, Jones L. Corneal and epithelial thickness changes study). Invest Ophthalmol Vis Sci 2013;54:6510–7.
after 4 weeks of overnight corneal refractive therapy lens wear, measured with [555] Plainis S, Atchison DA, Charman WN. Power profiles of multifocal contact lenses
optical coherence tomography. Eye Contact Lens 2004;30:189–93. discussion and their interpretation. Optom Vis Sci 2013;90:1066–77.
205–206. [556] Aller TA. Results of a one-year prospective clinical trial (CONTROL) of the use of
[523] Kim WK, Kim BJ, Ryu I-H, Kim BJ, Kim SW. Corneal epithelial and stromal bifocal soft contact lenses to control myopia progression [abstract]. Ophthalmic
thickness changes in myopic orthokeratology and their relationship with Physiol Opt 2006:8–9.
refractive change. PLoS One 2018;13:e0203652. [557] Aller TA, Wildsoet C. Bifocal soft contact lenses as a possible myopia control
[524] Haque S, Fonn D, Simpson T, Jones L. Epithelial thickness changes from the treatment: a case report involving identical twins. Clin Exp Optom 2008;91:
induction of myopia with CRTH RGP contact lenses. Invest Ophthalmol Vis Sci 394–9.
2008;49:3345–50. [558] Walline JJ, Greiner KL, McVey ME, Jones-Jordan LA. Multifocal contact lens
[525] Gifford P, Swarbrick HA. Refractive changes from hyperopic orthokeratology myopia control. Optom Vis Sci 2013;90:1207–14.
monovision in presbyopes. Optom Vis Sci 2013;90:306–13. [559] Walline JJ, Walker MK, Mutti DO, Jones-Jordan LA, Sinnott LT, Giannoni AG,
[526] Efron N, Morgan PB, Woods CA, Santodomingo-Rubido J, Nichols JJ. et al. Effect of high add power, medium add power, or single-vision contact lenses
International contact lens prescribing survey consortium. International survey of on myopia progression in children: the BLINK randomized clinical trial. JAMA
contact lens fitting for myopia control in children. Cont Lens Anterior Eye 2020; 2020;324:571–80.
43:4–8. [560] Anstice NS, Phillips JR. Effect of dual-focus soft contact lens wear on axial myopia
[527] Holden BA, Jong M, Davis S, Wilson D, Fricke T, Resnikoff S. Nearly 1 billion progression in children. Ophthalmol 2011;118:1152–61.
myopes at risk of myopia-related sight-threatening conditions by 2050 - time to [561] Sankaridurg P, Holden B, Smith 3rd E, Naduvilath T, Chen X, de la Jara PL, et al.
act now. Clin Exp Optom 2015;98:491–3. Decrease in rate of myopia progression with a contact lens designed to reduce
[528] Flitcroft DI. The complex interactions of retinal, optical and environmental relative peripheral hyperopia: one-year results. Invest Ophthalmol Vis Sci 2011;
factors in myopia aetiology. Prog Retin Eye Res 2012;31:622–60. 52:9362–7.
[529] Iwase A, Araie M, Tomidokoro A, Yamamoto T, Shimizu H, Kitazawa Y, et al. [562] Lam CS, Tang WC, Tse DY, Tang YY, To CH. Defocus Incorporated Soft contact
Prevalence and causes of low vision and blindness in a Japanese adult population: (DISC) lens slows myopia progression in Hong Kong Chinese schoolchildren: a 2-
the Tajimi Study. Ophthalmology 2006;113:1354–62. year randomised clinical trial. Br J Ophthalmol 2014;98:40–5.
[530] Wu L, Sun X, Zhou X, Weng C. Causes and 3-year-incidence of blindness in Jing- [563] Fujikado T, Ninomiya S, Kobayashi T, Suzaki A, Nakada M, Nishida K. Effect of
An District, Shanghai, China 2001-2009. BMC Ophthalmol 2011;11:10. low-addition soft contact lenses with decentered optical design on myopia
[531] Bullimore MA, Brennan NA. Myopia control: why each diopter matters. Optom progression in children: a pilot study. Clin Ophthalmol 2014;8:1947–56.
Vis Sci 2019;96:463–5. [564] Paune J, Morales H, Armengol J, Quevedo L, Faria-Ribeiro M, Gonzalez-
[532] Tideman JW, Snabel MC, Tedja MS, van Rijn GA, Wong KT, Kuijpers RW, et al. Meijome JM. Myopia control with a novel peripheral gradient soft lens and
Association of axial length with risk of uncorrectable visual impairment for orthokeratology: a 2-year clinical trial. Biomed Res Int 2015;2015:507572.
europeans with myopia. JAMA Ophthalmol 2016;134:1355–63. [565] Aller TA, Liu M, Wildsoet CF. Myopia control with bifocal contact lenses: a
[533] Cruickshank FE, Logan NS. Optical “dampening” of the refractive error to axial randomized clinical trial. Optom Vis Sci 2016;93:344–52.
length ratio: implications for outcome measures in myopia control studies. [566] Cheng X, Xu J, Chehab K, Exford J, Brennan N. Soft contact lenses with positive
Ophthalmic Physiol Opt 2018;38:290–7. spherical aberration for myopia control. Optom Vis Sci 2016;93:353–66.
[534] Wolffsohn JS, Kollbaum PS, Berntsen DA, Atchison DA, Benavente A, Bradley A, [567] Chamberlain P, Peixoto-de-Matos SC, Logan NS, Ngo C, Jones D, Young G. A 3-
et al. IMI - clinical myopia control trials and instrumentation report. Invest year randomized clinical trial of MiSight lenses for myopia control. Optom Vis Sci
Ophthalmol Vis Sci 2019;60:M132–60. 2019;96:556–67.
[535] Walline JJ, Lindsley KB, Vedula SS, Cotter SA, Mutti DO, Ng SM, et al. [568] Sankaridurg P, Bakaraju RC, Naduvilath T, Chen X, Weng R, Tilia D, et al. Myopia
Interventions to slow progression of myopia in children. Cochrane Database Syst control with novel central and peripheral plus contact lenses and extended depth
Rev 2020;1:CD004916. of focus contact lenses: 2 year results from a randomised clinical trial. Ophthalmic
[536] Horner DG, Soni PS, Salmon TO, Swartz TS. Myopia progression in adolescent Physiol Opt 2019;39:294–307.
wearers of soft contact lenses and spectacles. Optom Vis Sci 1999;76:474–9. [569] Li SM, Kang MT, Wu SS, Meng B, Sun YY, Wei SF, et al. Studies using concentric
[537] Katz J, Schein OD, Levy B, Cruiscullo T, Saw S-M, Rajan U, et al. A randomized ring bifocal and peripheral add multifocal contact lenses to slow myopia
trial of rigid gas permeable contact lenses to reduce progression of children’s progression in school-aged children: a meta-analysis. Ophthalmic Physiol Opt
myopia. Am J Ophthalmol 2003;136:82–90. 2017;37:51–9.
[538] Walline JJ, Jones LA, Mutti DO, Zadnik K. A randomized trial of the effects of [570] Smith 3rd EL, Campbell MCW, Irving E. Does peripheral retinal input explain the
rigid contact lenses on myopia progression. Arch Ophthalmol 2004;122:1760–6. promising myopia control effects of corneal reshaping therapy (CRT or ortho-K) &
[539] Walline JJ, Jones LA, Sinnott L, Manny RE, Gaume A, Rah MJ, et al. multifocal soft contact lenses? Ophthalmic Physiol Opt 2013;33:379–84.
A randomized trial of the effect of soft contact lenses on myopia progression in [571] Atchison DA, Li SM, Li H, Li SY, Liu LR, Kang MT, et al. Relative peripheral
children. Invest Ophthalmol Vis Sci 2008;49:4702–6. hyperopia does not predict development and progression of myopia in children.
[540] Stone J. The possible influence of contact lenses on myopia. Br J Physiol Opt Invest Ophthalmol Vis Sci 2015;56:6162–70.
1976;31:89–114. [572] Ticak A, Walline JJ. Peripheral optics with bifocal Soft and corneal reshaping
[541] Khoo CY, Chong J, Rajan U. A 3-year study on the effect of RGP contact lenses on contact lenses. Optom Vis Sci 2013;90:3–8.
myopic children. Singapore Med J 1999;40:230–7. [573] Berntsen DA, Kramer CE. Peripheral defocus with spherical and multifocal soft
[542] Swarbrick HA, Alharbi A, Watt K, Lum E, Kang P. Myopia control during contact lenses. Optom Vis Sci 2013;90:1215–24.
orthokeratology lens wear in children using a novel study design. Ophthalmology [574] Ozkan J, Fedtke C, Chung J, Thomas V, Bakaraju RC. Short-term adaptation of
2015;122:620–30. accommodative responses in myopes fitted with multifocal contact lenses. Eye
[543] Gifford KL, Richdale K, Kang P, Aller TA, Lam CS, Liu YM, et al. IMI - clinical Contact Lens 2018;44(Suppl 1):S30–7.
management guidelines report. Invest Ophthalmol Vis Sci 2019;60:M184–203. [575] Gifford P, Li M, Lu H, Miu J, Panjaya M, Swarbrick HA. Corneal versus ocular
[544] Wildsoet CF, Chia A, Cho P, Guggenheim JA, Polling JR, Read S, et al. IMI - aberrations after overnight orthokeratology. Optom Vis Sci 2013;90:439–47.
interventions for controlling myopia onset and progression report. Invest [576] Tarrant J, Liu Y, Wildsoet CF. Orthokeratology can decrease the accommodative
Ophthalmol Vis Sci 2019;60:M106–31. lag in myopes. Invest Ophthalmol Vis Sci 2009;50:4294.
[545] Cho P, Cheung SW, Edwards M. The longitudinal orthokeratology research in [577] Gifford P, Cannon T, Lee C, Lee D, Lee HF, Swarbrick HA. Ocular aberrations and
children (LORIC) in Hong Kong: a pilot study on refractive changes and myopic visual function with multifocal versus single vision soft contact lenses. Cont Lens
control. Curr Eye Res 2005;30:71–80. Anterior Eye 2013;36:66–73.
[546] Walline JJ, Jones LA, Sinnott LT. Corneal reshaping and myopia progression. Br J [578] Zhu M, Feng H, Zhu J, Qu X. The impact of amplitude of accommodation on
Ophthalmol 2009;93:1181–5. controlling the development of myopia in orthokeratology. Chinese J Ophthalmol
[547] Sun Y, Xu F, Zhang T, Liu M, Wang D, Chen Y, et al. Orthokeratology to control 2014;50:14–9.
myopia progression: a meta-analysis. PLoS One 2015;10:e0124535. [579] Cheng X, Xu J, Brennan NA. Accommodation and its role in myopia progression
[548] Si JK, Tang K, Bi HS, Guo DD, Guo JG, Wang XR. Orthokeratology for myopia and control with soft contact lenses. Ophthalmic Physiol Opt 2019;39:162–71.
control: a meta-analysis. Optom Vis Sci 2015;92:252–7. [580] Morgan PB, Woods CA, Tranoudis IG, Efron N, Jones L, Grupcheva C, et al.
International contact lens prescribing in 2020. Contact Lens Spectrum 2021;2021
(January):32–8.

219

View publication stats

You might also like