Download ARVO 2015 Annual Meeting Abstracts 477 Early AMD Wednesday

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Image-guided radiation therapy wikipedia , lookup

Neutron capture therapy of cancer wikipedia , lookup

Medical imaging wikipedia , lookup

Transcript
ARVO 2015 Annual Meeting Abstracts
477 Early AMD
Wednesday, May 06, 2015 3:45 PM–5:30 PM
Exhibit Hall Poster Session
Program #/Board # Range: 5127–5166/C0191–C0230
Organizing Section: Retina
Contributing Section(s): Clinical/Epidemiologic Research,
Genetics, Multidisciplinary Ophthalmic Imaging, Visual
Psychophysics/Physiological Optics
Program Number: 5127 Poster Board Number: C0191
Presentation Time: 3:45 PM–5:30 PM
The effect of A2E and zeaxanthin on visual function: Modelling
AMD pathogenesis and treatment in zebrafish
D Joshua Cameron1, Eric A. Saidi1, 2, Pinakin G. Davey1. 1Optometry,
Western Univ of Hlth Sciences, Pomona, CA; 2Graduate College of
Biomedical Sciences, Western Univ of Hlth Sciences, Pomona, CA.
Purpose: Age-related macular degeneration is a devastating eye
disorder that is characterized by the progressive loss of macular or
central vision as patients age. One aspect of AMD’s pathogenesis
involves the bisretinoid A2E, which accumulates with age and has
been found in greater abundance in AMD eyes. Despite years of
study, A2E is still somewhat of an enigma. Carotenoids have been
used to delay the progression of AMD, but most studies have focused
on combinations of carotenoids rather than individual compounds.
We set out to test two hypotheses: 1) intracameral injections of A2E
will negatively affect visual function in zebrafish. 2) Injection of
zeaxanthin into the zebrafish eye would improve/protect the visual
acuity of the zebrafish.
Methods: Visual acuity, calculated in cycles per degree, was
measured in adult zebrafish to establish a consistent baseline using
the optokinetic response. Either Zeaxanthin or A2E were dissolved
into phosphate buffered saline (PBS) and injected into the anterior
chamber of the right and left eyes. PBS was injected as a control into
the anterior chamber of the right and left eyes of a separate group of
fish. Visual acuities were measured at 1 week and 3, 8 and 12 weeks
post-injection to compare to baseline values. A paired t-test was used
to compare visual acuities between subjects injected with zeaxanthin
or A2E and PBS.
Results: Intraocular injections of 50mM A2E impair visual function
in adult zebrafish. 2 weeks after injection visual acuity is significantly
reduced (p=0.01 n= 9 for each group). Zeaxanthin injections on the
other hand showed a significant improvement in visual acuity, 22%
better than before the injection (p=0.02 n=11 for zexanthin and 10 for
PBS). This improvement peaked at more than 30% for some fish a
few weeks after the injection. The enhanced visual function returned
to normal 3 months after the initial injection.
Conclusions: A2E caused a significant reduction in visual function
while zeaxanthin actually improved visual function in the adult
zebrafish. Further studies looking at the histology and other aspects
of visual function will help in characterizing the mechanism behind
these observed visual acuity changes and aid in understanding the
pathogenesis of AMD. Additionally, this platform may serve as a
model for studying the etiology of AMD and possible treatments.
Commercial Relationships: D Joshua Cameron, None; Eric A.
Saidi, None; Pinakin G. Davey, None
Support: WesternU Intramural Grant
Program Number: 5128 Poster Board Number: C0192
Presentation Time: 3:45 PM–5:30 PM
Age-Related Macular Degeneration and Aspirin Use
Fadi Shaya, Kent W. Small. Macule and Retina Institute, Glendale,
CA.
Purpose: To resolve the conflict of Aspirin’s cardiovascular
benefits versus its possible adverse effects on age-related macular
degeneration (AMD). Aspirin’s overall beneficial effects in reducing
heart attacks, stroke and sudden death have long been documented
in many large, randomized, controlled trials. Recent data from two
smaller AMD epidemiology studies have suggested a deleterious
effect on vision in patients with age-related macular degeneration.
A balanced analysis of these competing data sets is needed to
resolve these issues and aid cardiologists and ophthalmologist in
appropriately counseling their patients.
Methods: A meta-analysis was performed of six large randomized,
controlled trials using aspirin as a preventive for cardiovascular
events compared to smaller AMD epidemiological trials evaluating
the effects of aspirin on age-related macular degeneration. The
cardiovascular prevention studies consisted of a total of 95,000
individuals. The AMD epidemiological studies consisted of 13,062
participants collectively.
Results: The combined meta-analyses showed aspirin caused a
32 % reduction in the risk of non-fatal stroke (OR:0.68, 95% CI,
0.59-.79) with regular aspirin users. The study also documented
that aspirin users decreased the risk of vascular events by 15 %
(OR:0.85, 95 % CI, 0.79-0.93). In the AMD epidemiological studies,
Aspirin was associated with the increase in neovascular AMD from
1.9 % in five years, 7 % in ten years, and 9.3 % in fifteen years in
regular aspirin users (OR: 2.46, 95 % CI: 1.25-4.83). It is important
to note that the odd ratios are significantly smaller in the larger
cardiovascular prevention trials than the odd ratios in the smaller
AMD epidemiological studies.
Conclusions: Overall, the size and quality (very small confidence
intervals) of the cardiovascular studies documenting the overall
health benefit of aspirin use is much greater than the few smaller
AMD epidemiological studies suggesting possible adverse vision
effects of aspirin use in age-related macular degeneration. The
benefits of aspirin usage include preserving the duration and quality
of life by decreasing stroke and heart attack risk. These benefits seem
to far outweigh the theoretical risks of possibly exacerbating wet
AMD that can be reasonably controlled with anti-VEGF therapy. We
strongly recommend that AMD patients should be on aspirin if it is
recommended by their primary physician.
Commercial Relationships: Fadi Shaya, None; Kent W. Small,
None
©2015, Copyright by the Association for Research in Vision and Ophthalmology, Inc., all rights reserved. Go to iovs.org to access the version of record. For permission
to reproduce any abstract, contact the ARVO Office at [email protected].
ARVO 2015 Annual Meeting Abstracts
Program Number: 5129 Poster Board Number: C0193
Presentation Time: 3:45 PM–5:30 PM
Alpha-linolenic acid and risk of age-related macular
degeneration: a prospective study over more than two decades of
follow-up
Juan Wu1, Eunyoung Cho2, 3, Debra A. Schaumberg4, 3, Srinivas
Sastry5, Walter Willett1, 3. 1Department of Nutrition, Harvard School
of Public Health, Boston, MA; 2Department of Dermatology, Warren
Alpert Medical School of Brown University, Providence, RI;
3
Channing Division of Network Medicine, Brigham and Women’s
Hospital and Harvard Medical School, Boston, MA; 4Department
of Ophthalmology & Visual Sciences, University of Utah School of
Medicine, Salt Lake City, UT; 5Bethesda Retina, Bethesda, MD.
Purpose: Marine-sourced long-chain omega-3 fatty acids (n-3)
have been associated with lower risk of AMD in many observational
studies. Alpha-linolenic acid (ALA) is the primary n-3 consumed in
typical western diet due to its greater availability from plant sources
and a small amount is endogenously converted to long-chain n-3.
Thus, we aimed to investigate the association between long-term
intake of ALA and risk of age-related macular degeneration (AMD).
Methods: This prospective study included 81,847 female participants
in the Nurses’ Health Study who were ≥ 50 years old. We calculated
the intake of ALA from validated food frequency questionnaires
collected every four years beginning from 1984. By 2012, we
confirmed 1,224 intermediate and 1,023 advanced AMD cases (96%
wet AMD). Multivariate Cox proportional hazards model was used to
estimate the relative risks (RR’s) and 95% confidence intervals (CI’s).
Results: After adjusting for age, BMI, smoking and other suspected
risk factors of AMD, cumulative averaged intake of ALA was
positively associated with intermediate AMD (comparing extreme
quintiles, multivariate RR = 1.32, 95% CI = 1.10 to 1.58; p trend <
.001); the RR persisted after further adjustment for correlated dietary
risk factors including DHA, linoleic acid, saturated fat and trans fat
(RR = 1.30, 95% CI = 1.04 to 1.64; p trend = 0.02). Mayonnaise-type
salad dressing, the leading contributor to both the intake and plasma
level of ALA in this cohort, was associated with a 28% (95% CI =
1.06 to 1.55) increased risk of intermediate AMD comparing the
highest quintile of intake to the lowest. We detected a small amount
of understudied trans ALA isomers (0.05% of total fatty acids) in red
blood cells among a subsample of women (n = 408) and mayonnaise
also significantly predicted one of the trans ALA isomers (p < .001).
Intake of ALA was not significantly associated with advanced AMD
(RR = 1.09, 95% CI = 0.89 to 1.32; p trend = 0.61).
Conclusions: ALA does not appear to protect against AMD and
may increase the risk of intermediate AMD. Whether trans ALA is
responsible for the positive association deserves further investigation.
Commercial Relationships: Juan Wu, None; Eunyoung Cho,
None; Debra A. Schaumberg, None; Srinivas Sastry, None; Walter
Willett, None
Program Number: 5130 Poster Board Number: C0194
Presentation Time: 3:45 PM–5:30 PM
Drusen in the Asian pediatric population aged 6-18 yrs
Victor Chen1, Don B. Kim2, Gloria Wu3. 1Biology, UC San
Diego, Saratoga, CA; 2Biology, UC Berkeley, Berkeley, CA;
3
Ophthalmology, UC San Francisco School of Medicine, San
Francisco, CA.
Purpose: Non-exudative age related macular degeneration has been
reported to be increased in Asians in previous studies Drusen have
been reported in adults <55yrs, but is less studied in pediatric patients
(pts). This study assesses the appearance of drusen in Asian pediatric
patients in a retina practice in San Jose, CA, where 50% of the
population is of Asian descent.
Methods: Using the EHR, eClinicalWorks, from a retina practice
from 1/2013-11/2014, pts were identified who were aged 6-18 years,
with CPT code 92250 and non-retina ICD diagnosis codes with
Va=20/15-20/30. TRC-50EX Topcon camera, OIS winstation-XP,
zoom 3x feature were used to examine color and red-free photos.
The OIS winstation AREDS retinal grid was used to identify drusen
and measure drusen size. The grid was superimposed on the fundus
photo, centered at the fovea. Using the OIS Topcon software 3x zoom
feature, all quadrants, para and perifoveal regions were inspected for
drusen. This was done in all the fundus photographs with the above
criteria. Inclusion criteria: no macular disease, best corrected visual
acuity 20/15-20/30. Age range 6-18 years old. Exclusion criteria:
poor quality fundus photos, macular disease lesions or macular
diagnoses.
Results: A total of 54 patients met our inclusion criteria. 35 Asian/
South Asian, 8 Caucasian, 3 Hispanic, 1 African American, and
7 “Other” race. Of those 54, we examined the 35 Asian patients.
Average age = 12.14, sd=4.19, males n=18, avg male age=11.61,
sd=4.38, females n=17, avg female age=12.71, sd=4.04.
70 photographs were examined in total. 4/70 photos were excluded
due to poor vision in the photographed eye. No drusen in 24/70 eyes.
The remaining 42/70 eyes had drusen from 25 patients. 17 pts had
drusen in both eyes.
Patients with drusen ranged from 6-18 yrs in age. Average number of
drusen per eye OD = 3.7 ± 2.8. Average number of drusen per eye OS
= 3.0 ± 2.3. In total, 135 drusen were counted, all fewer than 65um in
diameter (C0 on the AREDS grid). 115 of 135 drusen appeared in the
outer quadrants vs. 20 found in the inner quadrants. 74 drusen (55%)
appeared in right eyes vs 61 drusen (45%) in left eyes.
Conclusions: Drusen can be found in Asian children as early as age
6. A majority of drusen were found in the perifoveal quadrants.
Commercial Relationships: Victor Chen, None; Don B. Kim,
None; Gloria Wu, None
Program Number: 5131 Poster Board Number: C0195
Presentation Time: 3:45 PM–5:30 PM
Multifunctional OCT imaging of macular degeneration for
vasculature, RPE and choroid investigation
Young-Joo Hong1, 4, Masahiro Miura2, 4, Shuichi Makita1, 4,
Deepa Kasaragod1, 4, Satoshi Sugiyama1, 3, Yoshiaki Yasuno1, 4.
1
Computational Optics Group, University of Tsukuba, Tsukuba,
Japan; 2Ibaraki Medical Center, Department of Ophthalmology,
Tokyo Medical University, Ami, Japan; 3New Development,
Tomey Corporation, Nagoya, Japan; 4Computational Optics and
Ophthalmology Group, Tsukuba, Japan.
Purpose: In order to investigate eyes of age-related macular
degeneration (AMD) comprehensively, 1-μm Jones-matrix
multifunctional optical coherence tomography (JM-OCT) imaging
was performed. This study aims at evaluating clinical utility of JMOCT imaging, based on a descriptive case series of AMD.
Methods: Eight eyes of 4 AMD cases, including 4 end stage AMD,
1 early stage AMD, 1 drusen, and 2 drusenoid pigment epithelial
detachment were imaged with JM-OCT.6 mm × 6 mm areas around
the diseased region were scanned with 512 × 1024 A-lines in 6.6
seconds. Intensity OCT, complex decorrelation OCT, birefringence
tomography, and degree-of-polarization-uniformity (DOPU)
tomography were calculated from a single scan and each tomography
represents the structure, vasculature, sub-resolution microstructural
property of the tissue, and melanin pigmentation. DOPU and complex
decorrelation signals were combined to provide comprehensive
angiography.
Results: Anormality of retinal pigment epithelium (RPE) was imaged
with high DOPU (less pigment) in 4 of 4 end stage AMD cases, and
©2015, Copyright by the Association for Research in Vision and Ophthalmology, Inc., all rights reserved. Go to iovs.org to access the version of record. For permission
to reproduce any abstract, contact the ARVO Office at [email protected].
ARVO 2015 Annual Meeting Abstracts
it was corresponding to the hypo-fluorescence of short-wavelength
autofluorescence (SW-AF) image. In another 4 eyes, SW-AF did
not show hypo-fluorescence and their RPE showed low DOPU
(pigmented). High birefringence band structure was observed in
sclera with less pigment (high DOPU signal) in 3 eyes of end stage
AMD.
Figure 1 shows a representative case of AMD. High contrast
choroidal vasculature and hypo-fluorescence were observed in the
abnormal RPE region from near infrared (NIR) AF (a) and SW-AF
(b) images, respectively. And the en face projection (c) of complex
decorrelation (g) showed similar appearance with NIR-AF, in
addition DOPU B-scan (h) showed depth resolved melanin pigment
distribution (arrowheads indicate abnormal RPE). In this region,
intensity OCT B-scan (d) showed higher penetration into sclera.
Birefringence B-scan (f) showed layered structure in the sclera
(dashed circle) as high birefringence at anterior sclera (green) and
low birefringence (blue) at posterior. The sclera was appeared with
high DOPU (less pigment). DOPU encoded complex decorrelation
angiography (d) showed distinguished choroidal vasculature at the
region of abnormal RPE and less pigmented choroid.
Conclusions: The multifunctional imaging with JM-OCT gives
comprehensive information about the vasculature, RPE, and choroid
with depth resolved imaging.
autofluorescence (FAF). On optical coherence tomography (OCT),
they appear as hyperreflective lesions in the subretinal space.
Over the course of greater than six months, some of these lesions
have been described as gradually decreasing in size and resorbing,
leaving areas of atrophy and hypoautoflourescence on FAF. This
imaging study reviews a series of two pseudovitelliform lesions that
underwent sudden and rapid collapse over the course of one month.
This represents a new clinical presentation of a known clinical entity.
Methods: This is a retrospective case review of a series of two
patients with pseudovitelliform lesions that underwent sudden
collapse over the course of one month.
Results: These two cases both had single large pseudovitelliform
subfoveal lesions with typical FAF and OCT characteristics. Visual
acuities were stable at that stage at 20/30 and 20/60. There were no
signs of subretinal or intraretinal fluid. Both patients were taking
prophylactic AREDS formula antioxidant vitamin suppelmentation.
Each underwent sudden collapse, with urgent presentation to the
clinic, as noted by significant decline in visual acuity, both to 20/100.
Visual acuity stabilized after the initial drop, the final level of acuity
seemingly corresponding with the degree of subretinal tissue loss.
One eye returned to the visual actuity baseline of 20/30 over the
course of one month. The other eye further declined and remained
at 20/200, also over the course of one month. Both cases developed
pigmentary changes on fundus exam and corresponding foveal
atrophy on OCT. The eye with persistent low vision appeared to
have greater atrophic changes on OCT than the eye that returned to
baseline visual acuity.
Conclusions: This imaging study describes a new clinical
presentation of pseudovitelliform lesions that has not been described
in the literature previously. These reported cases underwent sudden
collapse of their lesions with rapid subjective decline in visual
acuity. Vision then stabilized over the course of one month with the
final outcome appearing to correspond with the degree of subretinal
atrophy noted on OCT.
OCT of pseudovitelliform lesion prior to collapse
Commercial Relationships: Young-Joo Hong, TOMEY
CORPORATION (F), TOPCON CORPORATION (F); Masahiro
Miura, Norvatis (R); Shuichi Makita, TOMEY CORPORATION
(F), TOMEY CORPORATION (P), TOPCON CORPORATION
(F); Deepa Kasaragod, TOMEY CORPORATION (F), TOPCON
CORPORATION (F); Satoshi Sugiyama, TOMEY CORPORATION
(E); Yoshiaki Yasuno, TOMEY CORPORATION (F), TOMEY
CORPORATION (P), TOPCON CORPORATION (F)
Support: Japan Society for the Promotion of Science KAKENHI
2503053
Program Number: 5132 Poster Board Number: C0196
Presentation Time: 3:45 PM–5:30 PM
Collapse of Pseudovitelliform Lesions: A New Clinical
Presentation
Kelly Rue, Lisa C. Olmos. Ophthalmology, USC, Los Angeles, CA.
Purpose: Pseudovitelliform lesions appear in the macula as
round yellowish deposits that are hyperautoflourescent on fundus
OCT 1 month after pseudovitelliform collapse
Commercial Relationships: Kelly Rue, None; Lisa C. Olmos,
None
Support: An unrestricted grant from Research to Prevent Blindness,
New York, NY 10022
©2015, Copyright by the Association for Research in Vision and Ophthalmology, Inc., all rights reserved. Go to iovs.org to access the version of record. For permission
to reproduce any abstract, contact the ARVO Office at [email protected].
ARVO 2015 Annual Meeting Abstracts
Program Number: 5133 Poster Board Number: C0197
Presentation Time: 3:45 PM–5:30 PM
Evidence of subretinal migration of melanin-loaded cells during
dry age-related macular degeneration
Concetta Li Calzi1, 2, Michel Paques3, Florian Sennlaub4, Kiyoko
Gocho5, Jose A. Sahel3, 4, mustapha benchaboune3. 1Ophtalmologie,
ILC INSTITUT- private cabinet-, Cannes, France; 2HOPITAL
QUINZE-VINGT, Paris, France; 3Clinical Investigation Center
1423, Quinze-Vingts Hospital, Paris, France; 4Vision Institute, Paris,
France; 5Chiba Hospital, Chiba, Japan.
Purpose: During dry age-related macular degeneration (ARMD),
adaptative optics (AO) en face flood imaging improves the resolution
of pigmentary changes (Gocho et al, IOVS 2013). This led to the
discovery of the presence of numerous migrating hyporeflective
clumps within and around atrophic areas. The most likely explanation
of these images is taht they are due to the migration of melaninloaded cells (MLCs).
Here, we explored in more details the kinetics of MLCs.
Methods: Observational clinical study.4°x4° AO fundus images
were obtained with a commercially available flood imaging AO
camera (rtx1; Imagine Eyes, Orsay, France) within an IRB-approved
clinical study in eyes with dry AMD (n=4). Care was taken to capture
the progression front. The migration of MLCs was documented by
time-lapse imaging (mean interval between sessions 26 days; median
follow-up 5 months).
Results: Multiple MLCs were seen to migrate within as well at
outside of atrophic areas. Approximately 65% of hypereflective
clumps were seen to migrate during the observation period. Their
mean diameter was 19mm. The density of MLCs ranged from 300 to
800 per mm2. The linear velocity of MLCs peaked at 1um/day. The
behaviour of migrating MLCs was somewhat random, with in many
adjacent MLCs migrating in opposite directions; however, focal
agregations of MLCs were present. Agregation and disaggration of
MLCs accounted for the evolution of large pigmentary clumps. The
cone mosaic was seen over MLCs in 48% of cases.
Conclusions: During dry ARMD, extensive subretinal migration
of MLCs throughout the posterior pole is commonly observed, in
atrophic as well as in nonatrophic areas. To our knowledge, this
is the first demonstration of such subretinal migration. It is likely
that these MLCs correspond to the hypereflective dots reported by
optical coherence tomography (Coscas et al, 2013). The nature of
these cells and their role in the process of ARMD remains to be
clarified; likely candidates are sloughed RPE cells (Curcio et al,
IOVS 1998) and macrophages. The subretinal space appears thus as a
permissive milieu for the transmission of cellular inputs. The random
migration is reminiscent of the behaviour of patrolling, non activated
inflammatory cells. We therefore believe that this finding may have
important consequences for the understanding and monitoring of
dry ARMD, as well as for the validation of experimental models of
ARMD.
Commercial Relationships: Concetta Li Calzi, None; Michel
Paques, ImagineEye (C); Florian Sennlaub, None; Kiyoko Gocho,
None; Jose A. Sahel, ImagineEye (S); mustapha benchaboune,
None
Support: ANR-07-LVIE-001 and ANR-09-TECS-009-01
Clinical Trial: NCT01546181
Program Number: 5134 Poster Board Number: C0198
Presentation Time: 3:45 PM–5:30 PM
Genetic and clinical factors associated with choroidal vascular
hyperpermeability and subfoveal choroidal thickness in
polypoidal choroidal vasculopathy
Seigo Yoneyama, Yoichi Sakurada, Wataru Kikushima, Fumihiko
Mabuchi, Hiroyuki Iijima. Ophthalmology, University of Yamanashi,
Chuo, Japan.
Purpose: To investigate genetic and clinical factors associated with
choroidal vascular hyperpermeability (CVH) and subfoveal choroidal
thickness (SCT) in eyes with treatment-naïve polypoidal choroidal
vasculopathy (PCV).
Methods: We studied the consecutive 149 patients with PCV.
Presence or absence of CVH was evaluated on indocyanine green
angiography (ICGA). Subfoveal choroidal thickness (SCT) and
axial length was measured by spectral domain optical coherence
tomography and optical biometry, respectively. Variants of ARMS2
(rs10490924) and CFH (rs800292 and rs1329428) were genotyped
using TaqMan technology.
Results: Subfoveal choroidal thickness was correlated with axial
length and age (p=0.001 and p=0.02, respectively).Though there was
not a significant difference in SCT among CFH(rs800292) genotypes,
there was a statistical difference in SCT among CFH(rs1329428)
and ARMS2(rs10490924) genotypes. (p=0.002 and p=0.006,
stepwise regression analysis, respectively).Among 149 eyes with
PCV, 35 (23.5%) eyes exhibited CVH on ICGA. There was a
significant lower frequency of risk variants in CFH(rs1329428)
and ARMS2(rs10490924) in patients with CVH than patients
without CVH (p=0.029 and p=0.005, stepwise regression analysis,
respectively).
Conclusions: In addition to axial length and age, SCT in eyes with
PCV were associated with both genetic variants in CFH and ARMS2
gene. Patients with CVH might have a weaker effect on both genetic
variants than patients without CVH.
Commercial Relationships: Seigo Yoneyama, None; Yoichi
Sakurada, None; Wataru Kikushima, None; Fumihiko Mabuchi,
None; Hiroyuki Iijima, None
Program Number: 5135 Poster Board Number: C0199
Presentation Time: 3:45 PM–5:30 PM
Consuming a buttermilk drink containing lutein-enriched eggyolk daily for 1 year improves macular pigment optical density
and visual acuity in subjects with early signs of age-related
macular degeneration
Tos TJM Berendschot1, Sanne van der Made2, Elton R Kelly1,
Jogchum Plat2, Aize Kijlstra2. 1University Eye Clinic Maastricht,
Maastricht, Netherlands; 2Department of Human Biology, Maastricht
University Medical Centre, Maastricht, Netherlands.
Purpose: Lutein has been shown to be beneficial in maintaining
visual function in age-related macular degeneration (AMD). A dairy
beverage has been developed based on lutein-enriched egg yolks
and buttermilk. The eggs have been enriched in lutein via the feed of
laying hens. The primary aim of this study was to assess the effects
of 1-year daily consumption of this dairy drink containing luteinenriched egg-yolks on macular pigment optical density (MPOD)
and visual scores in subjects with early AMD. Also, plasma lutein
concentrations, and serum lipids and lipoproteins were measured.
Methods: One hundred and one subjects were recruited to participate
in this one-year, double blind, placebo-controlled intervention trial
in subjects with early signs of AMD. Subjects in the lutein group
consumed the dairy drink containing 1.4 mg lutein daily. MPOD
using heterochromatic flicker photometry, best corrected visual acuity
(BCVA), and dark adaptation were measured at the start of the study,
©2015, Copyright by the Association for Research in Vision and Ophthalmology, Inc., all rights reserved. Go to iovs.org to access the version of record. For permission
to reproduce any abstract, contact the ARVO Office at [email protected].
ARVO 2015 Annual Meeting Abstracts
after 6 months and after 12 months of consuming either the placebo
or lutein drink. Plasma lutein concentration was assessed at baseline
and at the end of the study.
Results: In the lutein group plasma lutein concentration increased
significantly from 205 ng/ml at baseline to 399 ng/ml at study end
(p<0.001). MPOD increased significantly from 0.45 to 0.52 (p<0.001)
and BCVA increased significantly from -0.04 to -0.09 (LogMar,
p=0.004). We found no changes in the placebo group. Differences in
dark adaptation between both groups were not significant.
Conclusions: Daily consumption of a dairy drink containing
lutein-enriched egg-yolks for one year improves plasma lutein
concentration, MPOD and visual acuity in subjects with early signs
of age-related macular degeneration.
Commercial Relationships: Tos TJM Berendschot,
WO2009078716, US2011015277 (P); Sanne van der Made,
Newtricious R&D B.V. (E); Elton R Kelly, None; Jogchum Plat,
WO2009078716, US2011015277 (P); Aize Kijlstra, None
Support: The project was supported by Europees Fonds voor
Regionale Ontwikkeling (OP-Zuid), Dutch Ministry of Economic
Affairs, the Province of Limburg and Newtricious R&D B.V.
Clinical Trial: NCT00902408
Program Number: 5136 Poster Board Number: C0200
Presentation Time: 3:45 PM–5:30 PM
Ultrahigh speed OCT angiography in age-related macular
degeneration using swept source optical coherence tomography
Eric Moult1, 2, Nadia K. Waheed3, Woo Jhon Choi2, Mehreen Adhi3,
Talisa de Carlo3, Benjamin Potsaid4, Vijaysekhar Jayaraman5,
Philip J. Rosenfeld6, Jay S. Duker3, James G. Fujimoto2. 1Health
Sciences and Technology, Harvard-MIT, Cambridge, MA; 2Electrical
Engineering and Computer Science, Massachusetts Institute of
Technology, Cambridge, MA; 3New England Eye Center, Tufts
University Medical Center, Boston, MA; 4Advanced Imaging Group,
Thorlabs, Inc, Newtown, MA; 5Praevium Research Inc., Santa
Barbara, CA; 6Department of Ophthalmology, University of Miami
Miller School of Medicine, Bascom Palmer Eye Institute, Miami, FL.
Purpose: To investigate the use of ultrahigh speed swept source
optical coherence tomography (SSOCT) angiography for
characterizing microvascular changes in patients with age-related
macular degeneration (AMD).
Methods: A 1050nm wavelength, 400 kHz A-scan rate SSOCT
system was used to perform volumetric optical coherence
tomography angiography (OCTA) of the retinal and choriocapillaris
vasculature in normal volunteers and AMD patients. The normal
group consisted of 63 eyes (32 subjects; 40.7 ± 14.1 years; 13 males;
19 females). The AMD group included 17 eyes with wet AMD (15
patients; 79.7 ± 8.3 years; 6 males; 9 females), 35 eyes with dry
AMD without geographic atrophy (GA) (26 patients; 77.5 ± 9.0
years; 11 males; 15 females) and 12 eyes with dry AMD with GA
(7 subjects; 75.9 ± 6.1 years; 6 males; 1 female). A technique called
variable interscan time analysis was developed to differentiate
flow impairment from atrophy; the technique works by varying the
effective interscan time between repeated B-scans.
Results: Choroidal neovascularization (CNV) was visualized in 16
of the 19 eyes with wet AMD. In these 16 eyes the CNV was found
above regions of choriocapillaris atrophy. In 14 of these eyes the
CNV was surrounded by choriocapillaris alteration. Eyes with dry
AMD without GA showed varying severities of choriocapillaris
alterations, some of which were associated with drusen and/ or retinal
pigment epithelium/ photoreceptor alteration. In all 12 eyes with GA,
OCTA showed choriocapillaris atrophy within the GA region. In 10
of these eyes OCTA showed choriocapillaris flow impairment beyond
the GA margin.
Conclusions: OCTA appears to be a promising modality for noninvasive imaging of retinal and choriocapillaris vasculature in eyes
with AMD.
CNV located adjacent to GA in a 75 year old AMD patient. (A) En
face OCT projection. (B) OCTA of choriocapillaris. (C) En face
OCTA projection. (D) Fundus autofluorescence. (E) OCT and (F)
OCTA B-scans through the dashed line in (A). Scale bars: 1mm.
75 year old GA patient. (A) Fundus autofluorescence. (B) En face
OCT projection. (C) OCTA of retinal microvasculature. (D) OCTA of
choriocapillaris. (E) OCTA of choriocapillaris as in (D) but generated
using variable interscan time analysis. (F-I) Magnified views. OCT
intensity (top) and OCTA (bottom) B-scans through dashed lines in
(D) are shown in (J-L). Scale bars: 1mm.
Commercial Relationships: Eric Moult, None; Nadia K. Waheed,
None; Woo Jhon Choi, None; Mehreen Adhi, None; Talisa de
Carlo, None; Benjamin Potsaid, Thorlabs, Inc. (E); Vijaysekhar
Jayaraman, Praevium Research Inc., (E); Philip J. Rosenfeld, Carl
Zeiss Meditec Inc. (F); Jay S. Duker, Carl Zeiss Meditec Inc. (C),
Carl Zeiss Meditec Inc. (F), EyeNetra (I), Hemera Biosciences Inc.
©2015, Copyright by the Association for Research in Vision and Ophthalmology, Inc., all rights reserved. Go to iovs.org to access the version of record. For permission
to reproduce any abstract, contact the ARVO Office at [email protected].
ARVO 2015 Annual Meeting Abstracts
(I), Ophthotech Corp (I), Optovue Inc. (C), Optovue Inc. (F); James
G. Fujimoto, Carl Zeiss Meditec Inc. (P), Optovue Inc. (I), Optovue
Inc. (P)
Support: This work was supported by the National Institute
of Health (NIH R01-EY011289-27, R44-EY022864-01, R44EY022864-02, R01-CA075289-16), Air Force Office of Scientific
Research (AFOSR FA9550-10-1-0551 and FA9550-12-1-0499), a
Samsung Scholarship, and by a Natural Sciences and Engineering
Research Council of Canada Scholarship
Program Number: 5137 Poster Board Number: C0201
Presentation Time: 3:45 PM–5:30 PM
The effect of small hard drusen on local retinal structure in
healthy adults
Hilde R. Pedersen1, Inger C. Munch4, 3, Stuart J. Gilson1, Michael
Larsen2, 3, Rigmor C. Baraas1. 1Department of Optometry and
Visual Science, Faculty of Health Sciences, Buskerud and
Vestfold University College, Kongsberg, Norway; 2Department
of Ophthalmology, Glostrup Hospital, Copenhagen, Denmark;
3
Department of Ophthalmology, University of Copenhagen,
Copenhagen, Denmark; 4Department of Ophthalmology, Roskilde
Hospital, Roskilde, Denmark.
Purpose: To investigate how small hard drusen affect local retinal
structure in healthy adults by high-resolution in-vivo imaging of the
retina.
Methods: Ninety-seven healthy males and females aged 19–36
yrs, with normal logMAR letter acuity and no observed ocular
abnormalities, were included in the study. Color fundus photographs
were obtained of the central 45 degrees in both eyes. Any bright
element smaller than 63 mm in diameter whose shape, color, or
proximity to adjacent abnormality indicated it was not hard exudate,
was classified as a small, hard druse. Eyes with drusen were imaged
with the Heidelberg Spectralis OCT and the Kongsberg Adaptive
Optics Ophthalmoscope II (AO). Retinal layers were analyzed by
calculating longitudinal reflectivity profiles (LRP). Cone density and
mosaic regularity analysis was performed over and around drusen.
Results: One or more small hard drusen were found within the
central 20 degrees in 27 eyes in 21 of the 97 subjects, indicating a
population prevalence of 21,6 % in this age group. The number of
drusen per eye ranged from 1 to 8. Eight drusen per eye were found
in one subject, all in a cluster. OCT revealed a granulated retinal
pigment epithelium (RPE) complex in the area with multiple drusen.
Discrete OCT-visible drusen (33 %) were moderately reflective and
associated with localized internal thickening (4.11–7.52 mm) of
the RPE complex or elevation of the RPE with slight displacement
of the inner/outer segment (IS/OS) layer. One druse was located
in Verhoeff’s membrane (VM). The IS/OS layer and the external
limiting membrane (ELM) were intact over all drusen, but ELM had
reduced reflectivity above the drusen in two eyes. The RPE tended
to be more reflective and prominent than VM above drusen. In AO
images, drusen were hyperreflective circular or oval elements with
blurred edges. The size of discrete drusen ranged from 20 to 56 mm.
The cone density and regularity over drusen was not significantly
abnormal. The VM druse was hyperreflective and surrounded by
discontinuous hyporeflectivity.
Conclusions: In this study only one-third of small hard drusen were
visible on OCT, all found within or under the RPE. All drusen were
visible on AO showing an intact overlying photoreceptor mosaic with
moderate alteration of the reflectivity of the adjacent components
of the retina. The results may be relevant for the study of very early
precursor stages of age-related macular degeneration.
Commercial Relationships: Hilde R. Pedersen, None; Inger C.
Munch, None; Stuart J. Gilson, None; Michael Larsen, None;
Rigmor C. Baraas, None
Program Number: 5138 Poster Board Number: C0202
Presentation Time: 3:45 PM–5:30 PM
Reticular Macular Disease (RMD): a Two-Compartment
Disorder
Colleen Cunningham1, Patrick A. Kaszubski1, Hao Cheng1, 3,
Hua Hao2, Celine Saade1, K Bailey Freund1, 4, Theodore Smith1.
1
Ophthalmology, New York University School of Medicine, New
York, NY; 2Rollins School of Public Health, Emory University,
Atlanta, GA; 3Ophthalmology, The First Affiliated Hospital of
Guangzhou University, Guangzhou, China; 4Ophthalmology, Vitreous
Retina Macula Consultants, New York, NY.
Purpose: This study demonstrates spatial and temporal relationships
between the 2 processes of RMD, subretinal drusenoid deposits
(SDD) and choroidal alterations, in early Age-Related Macular
Degeneration (AMD).
Methods: 33 eyes (26 subjects) with early AMD/no SDD (RMD) and 18 eyes (16 subjects) with early AMD/SDD (RMD+)
underwent enhanced depth imaging spectral domain optical
coherence tomography (EDI SD-OCT) in the macula for choroidal
thickness (CTh) measurements at 11 points per scan, 475 μm apart,
in 5 horizontal B scans, 1 mm apart: a grid of 55 points. These 55
data points were treated as a cluster, to control for within-subject
variation, in a generalized estimating equation model of CTh as a
function of SDD status, age, gender, OD/OS, and fundus region.
Subdividing the sample by total group median age (<=82 and >82
years) allowed analysis of the annual rate of CTh change in 4 groups
(RMD+, <=82: 7/18; RMD+, >82: 11/18; RMD-, <=82: 21/33;
RMD-, >82: 12/33).
Results: CTh was significantly reduced in the subfoveal region in
RMD+ eyes compared to RMD- eyes (mean difference= - 12 μm,
P=0.02), but not in the nasal or temporal regions. Among subjects
<=82 years old, the overall mean CTh was significantly reduced in
RMD+ eyes compared to RMD- eyes (mean difference= - 54 μm,
P=0.012). Conversely, there was no significant difference in CTh in
the older group (>=82 years) between RMD+ and RMD-. In the older
age group, with each additional year of age, the choroids of RMD+
eyes got thicker (trend, 4.8 mm/year, p=0.33), while choroids of
RMD- eyes got thinner (trend, -0.3 mm/year, p=0.39).
Conclusions: Choroidal alterations in RMD vary with age and
fundus region. The choroid was thinner in RMD+ than RMDeyes in the subfoveal region. Among subjects <=82 years old, the
choroid was significantly thinner overall in RMD+ eyes, but not in
older subjects, highlighting the role of age. In RMD+ eyes, CTh
dynamics showed a trend to thinning before age 82, followed by
thickening after this age, consistent with RMD-associated choroidal
stromal fibrosis and thickening as a late development. RMD is a
dynamic disease in 2 compartments, SDD in the subretinal space and
pathology in the choroid, which must be studied together.
©2015, Copyright by the Association for Research in Vision and Ophthalmology, Inc., all rights reserved. Go to iovs.org to access the version of record. For permission
to reproduce any abstract, contact the ARVO Office at [email protected].
ARVO 2015 Annual Meeting Abstracts
12) vs. 18% of eyes without type C (41 of 227), p<0.001). Drusen
substructures at baseline were not associated with total GA area or
VA at baseline or year 2 (all p>0.05).
Conclusions: Intermediate AMD eyes with drusen substructures
at baseline were more likely to have non-central GA at year 2. The
presence of drusen cores and/or debris on SDOCT may be associated
with development of early GA.
IR image on left: The 55 point grid for CTh measurements. Regions:
red=nasal, yellow=subfoveal, blue=temporal. EDI-OCT image on
right: Example of a CTh measurement (blue line). Yellow lines point
to SDD.
Commercial Relationships: Colleen Cunningham, None; Patrick
A. Kaszubski, None; Hao Cheng, None; Hua Hao, None; Celine
Saade, None; K Bailey Freund, Bayer HealthCare (C), Genetech
(C), Heidelberg Engineering (C), Regeneron (C), Thrombogenics (C);
Theodore Smith, Advanced Cell Technologies (C)
Support: NIH/NEI Grant R01 EY015520; NIH/NEI Grant R01
EY06109
Program Number: 5139 Poster Board Number: C0203
Presentation Time: 3:45 PM–5:30 PM
Classification and Correlation of SDOCT-visualized Drusen
Substructures with Drusen Progression in Non-neovascular AMD
Malini Veerappan1, Wai T. Wong2, Francisco A. Folgar3, Vincent Tai3,
Michelle Michelson4, Katrina Winter3, Sandra Stinnett3, Cynthia A.
Toth3, 5. 1Department of Ophthalmology, Duke University School of
Medicine, Durham, NC; 2Unit on Neuron-Glia Interactions in Retinal
Disease, National Eye Institute, NIH, Bethesda, MD; 3Department
of Ophthalmology, Duke University Medical Center, Durham, NC;
4
Trinity College of Arts & Sciences, Duke University, Durham, NC;
5
Department of Biomedical Engineering, Duke University, Durham,
NC.
Purpose: Intermediate age-related macular degeneration (AMD)
is characterized by drusen in the retinal pigment epithelial (RPE)
layer. Multiple studies suggest that drusen contain a variety of
immumodulatory molecules, including those in the complement
cascade, which may manifest as distinct drusen substructures on
spectral domain optical coherence tomography (SDOCT). We
performed a prospective, observational clinical study to learn
how SDOCT-reflective drusen substructures at baseline predict
progression of non-neovascular AMD.
Methods: Study participants from the multicenter Age-Related
Eye Disease Study 2 (AREDS2) Ancillary SDOCT Study with
intermediate AMD in one eye at baseline (n=314) were analyzed
for presence of drusen substructures. Through review of SDOCT
volumes across the macula, substructures at baseline were divided
into 4 categories (see Figure 1). Presence of geographic atrophy
(GA), total GA area, and visual acuity (VA) were assessed at baseline
and year 2. These outcomes were compared between eyes with
and without cores/debris at baseline using Fischer exact test and
Wilcoxon rank sum test.
Results: Of 239 study eyes with sufficient image quality and followup, 58 (24%) had at least one drusen substructure (min 0, median
0, max 8). Of these, 29 (50%) had type L, 34 (59%) had type H,
11 (19%) had type S, and 12 (21%) had type C. At year 2, 49 eyes
had GA. 33% of eyes with substructures (19 of 58) vs. 17% of eyes
without substructures (30 of 181) progressed to GA at year 2 (p=
0.014). GA at year 2 may be specifically driven by presence of type
H (32% of eyes with type H (11 of 34) vs. 19% of eyes without type
H (38 of 205), p=0.071) and type C (67% of eyes with type C (8 of
Figure 1. Low-reflective Core (L). High-reflective Core (H). Split
Low and High Reflective Core (S). Conical Debris (C).
Commercial Relationships: Malini Veerappan, None; Wai T.
Wong, None; Francisco A. Folgar, None; Vincent Tai, None;
Michelle Michelson, None; Katrina Winter, None; Sandra
Stinnett, None; Cynthia A. Toth, Alcon (F), Bioptigen (F),
Genentech (F), NIH 1R01EY023039 (F)
Support: Genentech FVF44005
Clinical Trial: NCT00734487
©2015, Copyright by the Association for Research in Vision and Ophthalmology, Inc., all rights reserved. Go to iovs.org to access the version of record. For permission
to reproduce any abstract, contact the ARVO Office at [email protected].
ARVO 2015 Annual Meeting Abstracts
Program Number: 5140 Poster Board Number: C0204
Presentation Time: 3:45 PM–5:30 PM
Estimating the prevalence of subretinal drusenoid deposits (SDD)
among older adults with healthy macula and age-related macular
degeneration (AMD) with multimodal imaging
Anna V. Zarubina, David Neely, Mark E. Clark, Carrie E. Huisingh,
Brian C. Samuels, Yuhua Zhang, Gerald McGwin, Cynthia Owsley,
Christine A. Curcio. Ophthalmology, University of Alabama at
Birmingham, Birmingham, AL.
Purpose: To assess SDD prevalence among older adults with healthy
macula and early and intermediate AMD using multimodal imaging.
Methods: 651 subjects (1302 eyes) ≥60 years old enrolled in
the Alabama Study of Early Age-Related Macular Degeneration
(ALSTAR) were assessed using SD-OCT, IR and FAF images,
and color fundus photographs. Our strict criteria of SDD required
identification on ≥1 en face modality and OCT or on ≥2 en face
modalities in the absence of OCT findings. Our expanded criteria of
SDD required detection on any imaging modality. Eyes with gradable
OCT images were assessed for presence and severity of AMD using
the AREDS 9-step scale. The number of subjects excluded from
the analysis because of incomplete imaging data was specific to the
SDD criteria used. SDD was considered present at the person-level if
present even in one eye.
Results: Using the strict criteria, overall sample prevalence of SDD
was 32% (200/616); of those with SDD 123/200 (62%) were affected
in both eyes. Persons with SDD were older than those without
SDD (70.1 vs 68.7 yrs, p<0.0003). When stratified by AMD status,
SDD prevalence was 53% in AMD subjects and 23% in subjects
without AMD (p<0.0001). Among those with early and intermediate
AMD, SDD prevalence was 49% and 80% respectively. After age
adjustment, those with SDD were 3.5X more likely to have AMD
than those without SDD (95% CI 2.4-5.0). Using the expanded
criteria, SDD prevalence was 74% (462/622) with 326/462 (70%)
affected in both eyes. There was no age difference between subjects
with and without SDD, but SDD prevalence was higher among those
with AMD compared to those without (85% vs 69%, p<0.0001) and
increased with disease severity. After age adjustment, persons with
SDD were 2.6X more likely to have AMD compared to those without
SDD (95% CI 1.7-4.1).
Conclusions: SDD is prevalent in over 1/2 of persons with early to
intermediate AMD, and is present in about 1/4 of older adults with
healthy maculae, even by stringent SDD criteria. A broader definition
of SDD increases its prevalence but may lead to overestimation. SDD
prevalence is strongly associated with AMD presence and severity
and increases with age. Consensus on SDD detection methods is
recommended to advance our knowledge of this lesion and its clinical
and biologic significance.
Commercial Relationships: Anna V. Zarubina, None; David
Neely, None; Mark E. Clark, None; Carrie E. Huisingh, None;
Brian C. Samuels, None; Yuhua Zhang, None; Gerald McGwin,
None; Cynthia Owsley, None; Christine A. Curcio, None
Support: R01AG04212, R01EY06109; EyeSight Foundation of
Alabama; Alfreda J. Schueler Trust; Research to Prevent Blindness.
Purpose: Choriocapillaris thickness is associated with the
progression of drusen in dry age-related macular degeneration. Local
quantification of choriocapillaris thickness has so far been difficult.
We report an automated method to segment choriocapillaris and to
quantify the local/regional choriocapillaris thickness near drusen in
dry-AMD using spectral-domain optical coherence tomography (SDOCT).
Methods: Sixty-nine patients with dry AMD underwent OCT
imaging using Heidelberg Spectralis SD-OCT with enhanced depth
imaging. We segmented choriocapillaris and drusen simultaneously
using a graph-based method. The boundary of retinal pigment
epithelium (l-RPE), Bruch’s membrane (BM) and the transition
surface between choriocapillaris and choroidal vasculature (CC-CV)
were thus segmented (see Figure 1). Mean and standard deviation
(std.) of regional sub-RPE virtual space (sub-RPE-VS) thickness
were then calculated for each patient. A drusen was defined as a
group of neighboring A-scans where sub-RPE-VS thickness was
greater than mean+2*std. Local average choriocapillaris thicknesses
was averaged over the drusen region as well as circular regions of 20,
40, to 160 microns near drusen.
Results: Choriocapillaris is significantly thinner (8.46μm) under
the drusen (see Figure 2) and it is thicker (9.19μm) just outside the
boundary of the drusen.
Conclusions: We have developed an automated method to quantify
the presence, distribution and size of drusen and measure regional
choriocapillaris thickness. Our preliminary results confirm thinner
choriocapillaris under drusen that is known from histology studies.
Whether the intriguing finding of slightly thickened choriocapillaris
just outside of drusen areas can be confirmed is the subject of an
ongoing study. Our approach has the potential to better understand
the relationships between different structures in the outer retina in
dry-AMD.
Program Number: 5141 Poster Board Number: C0205
Presentation Time: 3:45 PM–5:30 PM
Automated quantification of choriocapillaris thickness near
drusen
Li Zhang1, Elliott H. Sohn2, Robert F. Mullins2, Milan Sonka1, 2,
Michael D. Abramoff1, 2. 1Electrical and Computer Engineering,
University of Iowa, Iowa City, IA; 2Ophthalmology and Visual
Sciences, University of Iowa, Iowa City, IA.
©2015, Copyright by the Association for Research in Vision and Ophthalmology, Inc., all rights reserved. Go to iovs.org to access the version of record. For permission
to reproduce any abstract, contact the ARVO Office at [email protected].
ARVO 2015 Annual Meeting Abstracts
Figure 1. Automated segmentation of choriocapillaris and drusen (a)
Original B-scan; (b) Same B-scan with segmented surfaces.
Figure 2. Choriocapillaris thickness averaged over the drusen region
as well as circular regions of 20, 40, to 160μm near drusen.
Commercial Relationships: Li Zhang, None; Elliott H. Sohn,
None; Robert F. Mullins, None; Milan Sonka, University of Iowa
(P); Michael D. Abramoff, IDx LLC (C), IDx LLC (I), University of
Iowa (P)
Support: NIH Grant R01 EY018853, R01 EY019112, R01
EB004640
©2015, Copyright by the Association for Research in Vision and Ophthalmology, Inc., all rights reserved. Go to iovs.org to access the version of record. For permission
to reproduce any abstract, contact the ARVO Office at [email protected].
ARVO 2015 Annual Meeting Abstracts
Program Number: 5142 Poster Board Number: C0206
Presentation Time: 3:45 PM–5:30 PM
The role of age-related macular degeneration associated variants
in drusen progression
Joshua D. Hoffman3, Mark J. Van Grinsven1, Jessica Cooke Bailey2,
Mariusz Butkiewcz2, Milam A. Brantley4, Chun Li2, William K.
Scott5, Margaret A. Pericak-Vance5, Clara I. Sanchez1, Jonathan L.
Haines2. 1Diagnostic Image Analysis Group, Radboud University
Nijmegen Medical Centre, Nijmegen, Netherlands; 2Epidemiology
and Biostatistics, Case Western Reserve University, Cleveland,
OH; 3Center for Human Genetics Research, Vanderbilt University
Medical Center, Nashville, TN; 4Vanderbilt Eye Institute, Vanderbilt
University Medical Center, Nashville, TN; 5John P. Hussman Institute
for Human Genomics, University of Miami School of Medicine,
Miami, FL.
Purpose: Age-related macular degeneration (AMD) is one of the
most common causes of visual impairment in the United States (US).
Although a multitude of studies have shown that both genetic and
environmental factors contribute to the pathogenesis of AMD, little
is known on how genetics impacts the disease’s rate of progression.
In this analysis we performed a quantitative analysis of drusen
progression during the intermediate stage of the disease to understand
the influence of AMD genetic variation on this phenotype.
Methods: Color fundus photographs deposited by the Age-related
Eye Diseases Study (AREDS) to the database of Genotypes and
Phenotypes (dbGAP) were retrieved on subjects that showed absence
of geographic atrophy (GA) and choroidal neovascularization (CNV)
and a baseline diagnosis of intermediate to large drusen. Automated
drusen detection and quantification was carried out on all subject
eyes and visits that met these criteria using a previously developed
algorithm. Drusen progression was tested against 19 previously
identified genetic variants using both a cumulative genetic risk score
and single variant analyses. Pathway analysis using results from
a genome-wide quantitative association analysis of progression
rates was carried out using Pathway Analysis by Randomization
Incorporating Structure (PARIS).
Results: In the 246 subjects AREDS subjects that met our selection
criteria we observe significant correlation in bilateral drusen
progression (rho = 0.30, p-value = 0.004). We do not observe
significant correlation between the 19-variant cumulative genetic risk
score and drusen progression (rho = 0.039; p = 0.543). We do not
observe a significant association with rs10490924 in ARMS2 (p =
0.161), rs10737680 in CFH (p = 0.156), rs116503776 in C2/CFB (p
= 0.713), or rs2230199 in C3 (p = 0.728). Single marker tests of the
remaining 15 variants show a nominally significant association with
rs943080 in VEGFA (p = 0.028). The most highly associated pathway
in our pathway analysis is the cell adhesion molecule pathway (p <
0.0001).
Conclusions: In this analysis we attempt to isolate the role of
known AMD genetic variation to one aspect of this phenotypically
heterogeneous disease. The 19 common variants examined
cumulatively and individually do not associate with progression in
our dataset, although pathway analysis suggests unidentified loci may
be important for drusen progression.
Commercial Relationships: Joshua D. Hoffman, None; Mark
J. Van Grinsven, None; Jessica Cooke Bailey, None; Mariusz
Butkiewcz, None; Milam A. Brantley, None; Chun Li, None;
William K. Scott, None; Margaret A. Pericak-Vance, None; Clara
I. Sanchez, None; Jonathan L. Haines, None
Support: A6019085, EY012118, AG019726, AG044089
Program Number: 5143 Poster Board Number: C0207
Presentation Time: 3:45 PM–5:30 PM
Comparison of retinal and choriocapillaris thicknesses following
sitting to supine transition in normal and age-related macular
degeneration subjects
David R. Almeida, Li Zhang, Eric K. Chin, Robert F. Mullins, Murat
Kucukevcilioglu, Milan Sonka, Edwin M. Stone, James C. Folk,
Michael D. Abramoff, Stephen R. Russell. Ophthalmology, University
of Iowa, Iowa City, IA.
Purpose: Effects of position on retinal and choroidal structure are
absent from the literature yet may provide insights into disease states
like age-related macular degeneration (AMD). We set out to evaluate
the effect of postural change on retinal and choroidal structures in
normal volunteers and subjects with non-neovascular AMD.
Methods: Prospective observational case series. Four unaffected
volunteers (8 eyes) and 7 subjects (8 eyes) with intermediate
AMD. Normal volunteers were selected that had no evidence of
ocular disease. Subjects with AMD were required to have at least
10 intermediate sized drusen. Spectral domain optical coherence
tomography (OCT) with enhanced depth imaging in upright (sitting)
and supine positions. Stable imaging was achieved using a rotating
adjustable mechanical arm that we constructed to allow the OCT
transducer to rotate 90 degrees. The Iowa Reference Algorithms
were used to quantify choroid and choriocapillaris thickness. Main
outcome measures included changes in sitting and supine position
central macular thickness (CMT, μm), total macular volume (TMV,
mm3), choroidal thickness (CT, μm), and choriocapillaris equivalent
thickness (CCET, μm).
Results: CCET was thinner in normal subjects (9.89 mm) compared
to patients with intermediate AMD (16.73 mm) (p = 0.02); there
was no difference in overall CT between the two groups (p = 0.38).
There was a 15% CCET reduction among normal subjects when
transitioning from sitting (9.89 mm) to supine position (8.40 μm) (p
= 0.02) versus a CCET reduction of 11.1% from sitting (16.73 μm) to
supine (14.88 μm) (p = 0.10) in patients with intermediate AMD.
Conclusions: Intermediate AMD appears to be associated with
an increase in CCET and with a lack of positional responses that
are observed in the CCET of normal eyes. Our results suggest that
although outer portions of the choroid do not appear to be responsive
to modest positional or hydrostatic pressure, the choriocapillaris
capacity is, and this is measurable in vivo. Whether this physiologic
deviation that occurs in AMD is related to atrophy, inflammation, or
changes in autoregulatory factors or growth factors, remains to be
determined.
Commercial Relationships: David R. Almeida, None; Li Zhang,
None; Eric K. Chin, None; Robert F. Mullins, None; Murat
Kucukevcilioglu, None; Milan Sonka, Imaging (P); Edwin M.
Stone, None; James C. Folk, None; Michael D. Abramoff, Imaging
(P); Stephen R. Russell, None
Program Number: 5144 Poster Board Number: C0208
Presentation Time: 3:45 PM–5:30 PM
Characteristics of Pseudodrusen investigated using Multi-Modal
Imaging including PS-OCT
Magdalena Baratsits1, Ferdinand G. Schlanitz1, Katharina
Eibenberger1, Bernhard Baumann2, Urike Scheschy1, Alessio
Montuoro1, Stefan Zotter2, Michael Pircher2, Christoph K.
Hitzenberger2, Ursula Schmidt-Erfurth1. 1Department of
Ophthalmology, Medical University of Vienna, Vienna, Austria;
2
Center for Medical Physics and Biomedical Engineering, Medical
University of Vienna, Vienna, Austria.
Purpose: The aim of this study was to investigate the characteristics
of pseudodrusen (PD) and their organization within the retina in
©2015, Copyright by the Association for Research in Vision and Ophthalmology, Inc., all rights reserved. Go to iovs.org to access the version of record. For permission
to reproduce any abstract, contact the ARVO Office at [email protected].
ARVO 2015 Annual Meeting Abstracts
patients with early age-related macular degeneration (AMD) by
evaluating and cross-linking different imaging techniques with threedimensional delineation of polarization- sensitive optical coherence
tomography (PS-OCT) volume scans.
Methods: 109 patients with early AMD were examined using
SD-OCT, IR-reflectance imaging, and autofluorescence imaging.
Furthermore, patients were scanned with a wide-field PS-OCT
(scanning area 30°x30°, volume scan 1024x250). The RPE-layer and
drusen were segmented by an automated computer algorithm utilizing
the polarization-senstive information. Afterwards, the PS-OCT
datasets were delineated manually for pseudodrusen. The 3D-map of
pseudodrusen was compared with the 3D-map of the RPE layer and
drusen, and furthermore to the autofluorescence and IR-reflectance
images in order to investigate the structural characteristics at the level
of RPE in early AMD.
Results: 14 eyes were segmented manually for PD and the imaging
characteristics were evaluated by cross-linking the three-dimensional
map to autofluorescence and IR-reflectance images. In the threedimensional images, PD did not show distinct borders but rather
a plaque-like spreading between the RPE and the photoreceptor
layers. Therefore, distinction between certain types of PD was
difficult, and most PD seemed to be influenced in their form by other
microstructural alterations at the level of the RPE, such as drusen.
The combination of drusen- and pseudodrusenmap could explain
some imaging characteristics seen in the en-face autofluorescence and
reflectance images.
Conclusions: Due to the ability of the PS-OCT to detect and
segment the RPE layer, grading of pseudodrusen could be done in
a more reliable way than before. Certain PD-types were evaluated
in their three-dimensional nature for the first time. By segmenting
the hyperreflective structures located above RPE a plaque-like
deposition found. This matches findings by histologic investigations,
and may be caused by the loose-fitting structures between RPE and
photoreceptors. The combination of the en-face images extracted
from OCT volumes, i.e. RPE layer, drusen- and pseudodrusen maps,
could explain the imaging characteristics of early AMD in en-face
images such as autofluorescence and IR-reflectance in more detail.
Commercial Relationships: Magdalena Baratsits, None;
Ferdinand G. Schlanitz, None; Katharina Eibenberger, None;
Bernhard Baumann, Canon (F); Urike Scheschy, None; Alessio
Montuoro, None; Stefan Zotter, Canon (F); Michael Pircher,
Canon (C), Canon (F); Christoph K. Hitzenberger, Canon (F);
Ursula Schmidt-Erfurth, Alcon (F), Boehringer (F), Novartis (F),
Novartis (F)
Program Number: 5145 Poster Board Number: C0209
Presentation Time: 3:45 PM–5:30 PM
Reticular pseudodrusen on infrared reflectance are
topographically distinct from subretinal drusenoid deposits on en
face optical coherence tomography
Michael Heiferman, Joshua K. Fernandes, Marion R. Munk,
Rukhsana Mirza, Lee M. Jampol, Amani A. Fawzi. Department
of Ophthalmology, Northwestern University Feinberg School of
Medicine, Chicago, IL.
Purpose: Reticular pseudodrusen (RPD), most prominent on infrared
reflectance (IR), are a phenotypic finding of age-related macular
degeneration (AMD) and a risk factor for increased incidence of
progression to late stage AMD. The relationship between RPD and
subretinal drusenoid deposits (SDD) has not been systematically
evaluated. The goal of this study is to evaluate the quantitative and
topographic relationship between RPD on IR and SDD on en-face
volumetric spectral-domain optical coherence tomography (SDOCT).
Methods: RPD were marked on IR images by a masked observer.
SDD were visualized on en-face sections of SD-OCT immediately
below the external limiting membrane and identified by a semiautomated technique using NIS-Elements Ar (Nikon Instruments
Inc., Tokyo, Japan). Control RPD lesions were generated in a random
distribution for each IR image using ImageJ (NIH, Bethesda, MD).
Binary maps of control and experimental RPD and SDD were merged
and topographically and quantitatively analyzed using ImageJ.
Results: 54 eyes of 41 patients diagnosed with RPD were included
in this study. The average number of RPD lesions on IR images
was 320±44.62 compared to 127±26.02 SDD lesions on en-face
(P<0.001). The percentage of total SDD lesions overlapping RPD
was 2.91±0.87% compared to 1.73±0.68% overlapping control RPD
lesions (P<0.05). The percentage of total SDD lesions between
1-3 pixels of the nearest RPD lesion was 5.08±1.40% compared to
3.33±1.07% between 1-3 pixels of the nearest control RPD lesion
(P<0.05).
Conclusions: This study identified significantly more RPD lesions on
IR compared with SDD lesions on en-face SD-OCT and found that a
large majority of SDD (>90% of lesions) were greater than 3 pixels
away from the nearest RPD indicating that RPD and SDD are two
distinct lesions. The minority of SDD lesions (7.99±2.22%) that were
either overlapping or between 1-3 pixels from RPD were more likely
to be in that location than predicted by randomly distributed control
RPD lesions. Together, our findings indicate that RPD and SDD are
two entities that are only occasionally topographically associated,
suggesting that at some stage of their development, they could be
pathologically related.
Distribution of SDD in relation to RPD and randomly located control
RPD lesions.
Commercial Relationships: Michael Heiferman, None; Joshua
K. Fernandes, None; Marion R. Munk, None; Rukhsana Mirza,
None; Lee M. Jampol, None; Amani A. Fawzi, None
Support: R01EY021470 (AAF), Research to Prevent Blindness, NY
(Department of Ophthalmology, Northwestern University)
©2015, Copyright by the Association for Research in Vision and Ophthalmology, Inc., all rights reserved. Go to iovs.org to access the version of record. For permission
to reproduce any abstract, contact the ARVO Office at [email protected].
ARVO 2015 Annual Meeting Abstracts
Program Number: 5146 Poster Board Number: C0210
Presentation Time: 3:45 PM–5:30 PM
Association of drusen substructure findings on SD-OCT
with abnormal drusen volumes and progression to choroidal
neovascularization in the AREDS2 Ancillary Study
Randall C. Gunther1, Cynthia A. Toth1, 3, Malini Veerappan1,
Michelle Michelson1, Vincent Tai1, Katrina Winter1, Francisco
A. Folgar1, Emily Y. Chew2, Sina Farsiu1. 1Ophthalmology, Duke
University Medical Center, Durham, NC; 2Epidemiology and
Clinical Applications, National Eye Institute, NIH, Bethesda, MD;
3
Department of Biomedical Engineering, Duke University, Durham,
NC.
Purpose: To evaluate spectral domain optical coherence tomography
(SDOCT) drusen substructures for association with drusen volume
and progression to choroidal neovascularization (CNV)
Methods: Eyes with intermediate age related macular degeneration
(AMD) from the multi-center prospective Age-Related Eye Disease
Study 2 (AREDS2) Ancillary SDOCT Study were analyzed for
presence of drusen substructures at baseline. Through qualitative
SDOCT grading, substructures were divided into four types: low
reflectivity core (L), high reflectivity core (H), split low and high
reflectivity core (S), and conical debris (C). After semi-automated
SDOCT segmentation, drusen volume within a 5-mm macular field
was measured at baseline and year 2. Presence of CNV was assessed
at year 2. Outcomes were compared using Wilcoxon rank sum and
Fisher exact test.
Results: Of 314 baseline study eyes, 273 had SDOCT grading
and CNV outcomes available. 226 had both baseline and year 2
drusen volume measurements. Eyes with any substructures were
associated with greater drusen volume at baseline (0.094±0.115 mm3
vs. 0.067±0.168 mm3, p<0.001) and year 2. L-type, H-type, and
S-type were correlated with higher drusen volume at baseline (L:
0.104±0.119 mm3 vs. 0.070±0.160 mm3 (p<0.001); H: 0.094±0.104
mm3 vs. 0.070±0.164 mm3 (p<0.001); S: 0.148±0.155 mm3vs.
0.070±0.155 (p<0.001)) and year 2. C-type did not correlate with
drusen volume. At year 2, 75 eyes had significant increase in drusen
volume. 44% of eyes with any drusen substructures (28 of 63)
vs. 29% of eyes without substructures (47 of 163) had significant
increase in drusen volume over 2 years (p=0.028). No individual
substructure types were associated with drusen volume change. 36
of 273 developed CNV by year 2, but there was no difference in
frequency of CNV between eyes with any substructures (14%) and
eyes without (13%).
Conclusions: Drusen substructure presence may be associated with
larger drusen volumes in patients with intermediate AMD. L and
W type substructures may indicate a risk for both increased drusen
volume and progression from intermediate AMD to CNV.
Commercial Relationships: Randall C. Gunther, None;
Cynthia A. Toth, Alcon (F), Bioptigen (F), Genentech (F),
NIH 1RO1EY023039 (F); Malini Veerappan, None; Michelle
Michelson, None; Vincent Tai, None; Katrina Winter, None;
Francisco A. Folgar, None; Emily Y. Chew, None; Sina Farsiu,
Duke University (P), NIH R01EY022691 (F)
Support: Grenentech FVF44005
Clinical Trial: NCT00734487
Program Number: 5147 Poster Board Number: C0211
Presentation Time: 3:45 PM–5:30 PM
Standardized Assessment of Drusen Measurements from Spectral
Domain Optical Coherence Tomography (SDOCT) Scans in Dry
Age Related Macular Degeneration (AMD)
Amitha Domalpally, Yijun Huang, Dawn Myers, Taylor Starnes, Zhe
Liu, Ronald P. Danis, Barbara A. Blodi. Ophthalmology, Fundus
Photograph Reading Center, Madison, WI.
Purpose: To compare retinal pigment epithelium (RPE) and retinal
thickness assessments made with SDOCTin eyes with dry AMD and
normal eyes.
Methods: Eyes with intermediate AMD or worse in participants
between 50 – 85 years of age were evaluated in custom SDOCT
segmentation software. After converting to a standardized Digital
Imaging and Communications in Medicine (DICOM) format,
macular volume scan files from 4 different SDOCT machines were
displayed in the same software and segmented for inner limiting
membrane (ILM), RPE-drusen complex and Bruch’s membrane (BM)
in a display and analysis platform using an algorithm developed at
the University of Wisconsin’s Fundus Photograph Reading Center
(Huang, et al, PLoS One. 2013 Dec 26;8(12):e82922) (Fig 1 top).
Reading center graders reviewed each volume scan and edited
boundary line placement if required. Similar segmentation (ILM,
RPE and BM – Fig 1 bottom) was performed in SDOCT scans
from participants of same age range in whom dry AMD and other
confounding abnormalities had been ruled out.
Results: The mean central subfield (CSF) retinal thickness was
significantly different between eyes with dry AMD (n = 72) and
normal eyes (n= 435); 225.8m (SD 39.8) and 245.5m (SD 27.8)
respectively. Total retinal volume was also significantly different
between the two groups; 7.4mm3(0.5) and 7.6 mm3 (0.5) respectively.
In the central subfield, thickness of the RPE-drusen complex was
70.8 m (SD 31.6) and RPE thickness in normal eyes was 33.3 m (SD
4.6) (P<0.001). The RPE-drusen complex volume and RPE volume
was also significantly different between the two groups; 1.1 mm3 (SD
0.2) and 0.8 mm3 (SD 0.1).
Conclusions: Drusen segmentation can be effectively performed in
eyes with dry AMD and can be used as an outcome for clinical trials.
Regression in drusen volume towards normal can potentially be
identified using custom segmentation.
Commercial Relationships: Amitha Domalpally, None; Yijun
Huang, EyeKor LLC (I); Dawn Myers, None; Taylor Starnes,
None; Zhe Liu, None; Ronald P. Danis, Merck (F); Barbara A.
Blodi, None
Program Number: 5148 Poster Board Number: C0212
Presentation Time: 3:45 PM–5:30 PM
Photoreceptor disorder around subretinal drusenoid deposits
and drusen revealed by adaptive optics scanning laser
ophthalmoscopy
Xiaolin Wang1, Tianjiao Zhang1, 2, Pooja Gordara1, Alexander
Meadway1, Mark E. Clark1, Clark D. Witherspoon1, Christopher
A. Girkin1, Cynthia Owsley1, Christine A. Curcio1, Yuhua
Zhang1. 1Ophthalmology, University of Alabama at Birmingham,
Birmingham, AL; 2Biomedical Engineering, University of Alabama at
Birmingham, Birmingham, AL.
Purpose: To describe the microscopic structure of photoreceptors
impacted by extracellular lesions residing in the subretinal space
and the sub-RPE (retinal pigment epithelium) space in patients with
age-related macular degeneration (AMD), using multimodal imaging
including a new-generation research adaptive optics scanning laser
ophthalmoscope (AOSLO).
©2015, Copyright by the Association for Research in Vision and Ophthalmology, Inc., all rights reserved. Go to iovs.org to access the version of record. For permission
to reproduce any abstract, contact the ARVO Office at [email protected].
ARVO 2015 Annual Meeting Abstracts
Methods: Twenty-nine patients (n=29) with early to intermediate
AMD (grade 2-8 on the AREDS 9-step severity scale) were classified
into 3 groups. Group 1 (9 eyes of 7 subjects) have subretinal
drusenoid deposits (SDD) only in the macula, Group 2 (13 eyes of 7
subjects) have abundant (more than 20) densely packed small hard
drusen (consistent with the clinic appearance of cuticular drusen,
50 – 75 μm diameter) only, and Group 3 (17 eyes of 15 subjects)
have medium-large soft drusen (63 - 1000 μm diameter). These
lesions were ascertained by presence in color fundus photographs,
infrared reflectance, blue reflectance, autofluorescence images, and
spectral domain optical coherence tomography (SD-OCT). SDD
were classified with a 3-stage OCT-based grading system. The
photoreceptor mosaic was assessed with AOSLO.
Results: AOSLO disclosed characteristic photoreceptor reflectivity
perturbation over different lesion types. For SDD, photoreceptor
reflectivity was reduced over stage 1 and stage 2 lesions with an
indiscernible mosaic. For stage 3 SDD, AOSLO revealed a distinctive
structure showing a hyporeflective annulus surrounding a core
that is formed by the lesion material itself but bears a reflectivity
superficially resembling photoreceptors (Fig.B1). Around the densely
packed small hard drusen, the photoreceptor mosaic was largely
visible and contiguous and with reduced, patchy reflectivity (Fig.B2).
Around large soft drusen, the photoreceptor mosaic was invisible
at the edge and visible on the top (Fig.B3). The en-face appearance
of the photoreceptor mosaic was consistent with its cross-sectional
structure as rendered by SD-OCT (Fig.C1-C3).
Conclusions: The dramatic photoreceptor reflectivity perturbation
associated with SDD indicates that SDD may impose more direct and
severe impact on overlying photoreceptors than drusen, suggesting
that the retinal function in eyes with SDD may be more severely
impired.
Figure. AOSLO revealed characteristic photoreceptor reflectivity
perturbation by different lesion types
Commercial Relationships: Xiaolin Wang, None; Tianjiao Zhang,
None; Pooja Gordara, None; Alexander Meadway, None; Mark
©2015, Copyright by the Association for Research in Vision and Ophthalmology, Inc., all rights reserved. Go to iovs.org to access the version of record. For permission
to reproduce any abstract, contact the ARVO Office at [email protected].
ARVO 2015 Annual Meeting Abstracts
E. Clark, None; Clark D. Witherspoon, None; Christopher A.
Girkin, None; Cynthia Owsley, None; Christine A. Curcio, None;
Yuhua Zhang, None
Support: This project is funded by EyeSight Foundation of
Alabama (YZ), International Retina Research Foundation (YZ),
5R21EY021903 (YZ), Songs for Sight (CAG), Buck Trust of
Alabama (CAG), R01AG04212 (CO), and R01EY06109 (CC) and
institutional support from Research to Prevent Blindness, EyeSight
Foundation of Alabama, Buck Trust of Alabama, and NIH P30
EY003039.
Program Number: 5149 Poster Board Number: C0213
Presentation Time: 3:45 PM–5:30 PM
Longitudinal Quantitative OCT Analysis of Drusen in the Fellow
Eye of Patients with Unilateral Neovascular Age-Related Macular
Degeneration
Hongyang Zhang1, 2, Nizar S. Abdelfattah1, David S. Boyer3,
Srinivas R. Sadda1. 1Doheny Image Reading Center, Doheny Eye
Institute, University of California Los Angeles, Los Angles, CA;
2
Ophthalmology, Guangdong General Hospital, Guangzhou, China;
3
Retina Vitreous Associates Medical Group, Los angles, CA.
Purpose: The fellow eyes of patients with late AMD in one eye may
be at the highest risk of developing late AMD. Reduction of drusen
volume without the development of late AMD has been suggested
as a potential therapeutic endpoint in early intervention trials. To
determine the relationship between drusen volume and late AMD,
we evaluated longitudinal changes in the fellow eye of patients with
unilateral neovascular AMD using optical coherence tomography
(OCT).
Methods: In this retrospective analysis, we analyzed the fellow
eye of 42 patients (age range, 69-93 years) who had advanced
neovascular AMD in only 1 eye. All patients were treated with
intravitreal ranibizumab, pegaptanib, or/and bevacizumab and
followed up for two years. In this analysis, we focused on the fellow
eye with only evidence of drusen secondary to the non-exudative
AMD. All eyes were scanned with the Cirrus HD-OCT (Carl Zeiss
Meditec, Inc., Dublin, CA) using a 512 ×128 scan pattern. OCT
data at baseline, Month 12, and Month 24 was evaluated using the
advanced RPE analysis tool to quantify drusen volume within 3mm
and 5mm diameter circles. OCT scans were also evaluated for the
development of atrophy or CNV.
Results: Drusen without evidence of late AMD were evident in 18
participants (42.9%). After 12 months, none of the eyes developed
neovascular disease but two eyes (11.1%) developed geographic
atrophy (GA). By 24 months of follow-up, 4 eyes (22.2%) developed
GA and no eyes developed CNV. Once the eye progressed to GA
it was censored from the quantitative drusen study. After two
years, there were no statistically significant changes in drusen area
or volume for the cohort overall. There was also no statistically
significant (p = 0.38, in 3mm; p=0.10, in 5mm) difference in baseline
drusen volume between eyes which developed atrophy compared
with those that did not. However, in eyes which developed atrophy by
Month 24, drusen volume had decreased at Month 12 by 0.06 ± 0.06
mm3 compared with 0.01 ± 0.04 mm3 in eyes without atrophy.
Conclusions: Changes in drusen volume and development of GA can
be tracked using SDOCT. Although baseline drusen volume did not
appear to predict atrophy development, a decrease in drusen volume
was associated with new atrophy. This suggests the potential for
drusen volume changes to be used as biomarkers to identify eyes at
high risk for progression to atrophy.
Commercial Relationships: Hongyang Zhang, None; Nizar
S. Abdelfattah, None; David S. Boyer, Aerpio (C), Alcon (C),
Alcon (R), Allergan (C), Allergan (R), Bayer (C), Genentech (C),
Genentech (R), GS (C), KalVista (C), Neurotech (C), Nicox, Inc
(C), Novartis (C), Novartis (R), Ohr (C), Regeneron (R), Regeneron
Pharmaceuticals, Inc (C), Santaris (C), Santen (C), ThromboGenics
(C); Srinivas R. Sadda, Alcon (C), Allergan (C), Allergan (F),
Allergan (R), Carl Zeiss Meditec (C), Carl Zeiss Meditec (F), Carl
Zeiss Meditec (R), Genentech (C), Genentech (F), Novartis (C),
Optos (C), Optos (F), Optos (R), Roche (C)
Program Number: 5150 Poster Board Number: C0214
Presentation Time: 3:45 PM–5:30 PM
Reticular Pseudodrusen, Enlarging Geographic Atrophy,
and Decreased Choroidal Thickness in Age-Related Macular
Degeneration
Mariana Rossi Thorell1, Raquel Goldhardt1, Renata Portella Nunes2,
Carlos Alexandre de Amorim Garcia Filho2, Giovanni Gregori1,
Zohar Yehoshua1, William J. Feuer1, Srinivas R. Sadda3, Philip J.
Rosenfeld1. 1Bascom Palmer Eye Institute, Miami, FL; 2Federal
University of Sao Paulo - UNIFESP, Sao Paulo, Brazil; 3Doheny Eye
Institute, Los Angeles, CA.
Purpose: To compare subfoveal choroidal thickness (CT)
measurements in eyes with non-exudative age-related macular
degeneration (AMD) with and without reticular pseudodrusen (RPD)
and eyes with no known ocular disease.
Methods: Retrospective study of AMD patients enrolled in the
COMPLETE study (30 drusen-only eyes and 30 eyes with geographic
atrophy [GA]) compared to an age-distributed normal control group
(155 eyes of normal subjects). CT was measured in the subfoveal
region using enhanced-depth SD-OCT imaging. Multimodal images
were evaluated to detect the presence of RPD in eyes with nonexudative AMD.
Results: Three drusen eyes (10%) were diagnosed with RPD. In
the GA group, 20 eyes (66.7%) were diagnosed with RPD. After
controlling CT for age and axial length, the mean subfoveal CT
of the GA group was significantly thinner than the drusen group
(p=0.007), but the drusen group was not significantly different than
the normal controls (p=0.090). After separating the GA group into
eyes with and without RPD, the GA group without RPD had a mean
CT (335.3±123.2μm) that was not significantly different from normal
eyes (286.1±84.5;p=0.076) or eyes with drusen (315.6±86.2; p=0.45),
while the GA group with RPD (213.7±53.1μm) was significantly
thinner than the GA group without RPD (p=0.001). In eyes without
RPD, increasing area of GA was strongly correlated with decreasing
CT (p=0.001).
Conclusions: Subfoveal CT measurements in eyes with nonexudative AMD and without RPD were similar to normal ageadjusted control eyes. Choroidal thinning in eyes with non-exudative
AMD was associated with the presence of RPD or an increasing area
of GA.
Commercial Relationships: Mariana Rossi Thorell, Carl Zeiss
Meditec (F); Raquel Goldhardt, None; Renata Portella Nunes,
©2015, Copyright by the Association for Research in Vision and Ophthalmology, Inc., all rights reserved. Go to iovs.org to access the version of record. For permission
to reproduce any abstract, contact the ARVO Office at [email protected].
ARVO 2015 Annual Meeting Abstracts
None; Carlos Alexandre de Amorim Garcia Filho, None; Giovanni
Gregori, Carl Zeiss Meditec (F), Carl Zeiss Meditec (P); Zohar
Yehoshua, None; William J. Feuer, None; Srinivas R. Sadda, Carl
Zeiss Meditec (F); Philip J. Rosenfeld, Carl Zeiss Meditec (F)
Support: Grant from Alexion Pharmaceuticals, Carl Zeiss Meditec,
Inc. (Dublin, CA), the Macula Vision Research Foundation, an
unrestricted grant from Research to Prevent Blindness, the Feig
Family Foundation, the Emma Clyde Hodge Memorial Foundation,
and the National Eye Institute (R01EY024158, P30EY014801).
Program Number: 5151 Poster Board Number: C0215
Presentation Time: 3:45 PM–5:30 PM
Changes in Subretinal Drusenoid Deposits (SDD) During
Progression to Geographic Atrophy (GA) and Choroidal
Neovascularization (CNV) in Age-Related Macular Degeneration
(AMD)
Vivek Kumar1, Patrick A. Kaszubski1, Tal Ben Ami1, Celine Saade1,
Ana Rita Santos2, Rufino Silva2, 3, Maria Luz Cachulo2, 3, Jose G.
Cunha-Vaz2, Theodore Smith1. 1Department of Ophthalmology, New
York University School of Medicine, New York, NY; 2Association for
Innovation and Biomedical Research on Light and Image (AIBILI),
Coimbra, Portugal; 3Department of Ophthalmology, Coimbra
University Hospital, Coimbra, Portugal.
Purpose: This retrospective study utilized 3 imaging modalities to
quantitatively analyze SDD evolution in eyes that progressed from
early to late AMD over a 2-year follow-up period. We analyzed
the change in SDD area within the fundus, both in subjects who
progressed to GA and in those who progressed to CNV, with the goal
of better understanding the dynamic spatiotemporal nature of SDD
and their role in AMD progression.
Methods: 25 patients with AMD and unilateral CNV were included.
The non-CNV eyes (the study eyes) all underwent indocyanine
green angiography (ICG), short-wavelength autofluorescence (AF),
and near infrared reflectance (NIR) imaging at baseline and at
2-year follow-up. Study eyes were analyzed for SDD and for the
development of late AMD—both CNV and geographic atrophy (GA).
SDD area measurements were obtained by two graders utilizing
ImageJ software and are expressed as a percentage of total fundus
area; binary logistic regression and the Wilcoxon nonparametric test
were used for statistical analysis.
Results: 8 study eyes developed GA and 17 study eyes developed
CNV during the follow-up period. In the eyes that developed CNV,
there was a statistically significant decrease in SDD area as seen on
AF (P=0.004) and NIR (P=0.027), and the decrease in SDD area
approached statistical significance on ICG (P=0.078). By contrast, in
the eyes that developed GA, there was no significant change in SDD
area as seen on all 3 imaging modalities (ICG: P=0.75, AF: P=0.275,
and NIR: P=1).
Conclusions: SDD undergo dynamic spatiotemporal changes in eyes
with AMD. These lesions can be seen in eyes that progress to late
AMD, both GA and CNV, but a decrease in SDD area is seen only
during progression to CNV, likely as a result of worse ischemia and
increased VEGF in CNV subjects. These results suggest that the SDD
lesions migrate, are absorbed by surrounding cells, or are no longer
visible due to imaging difficulties secondary to changes present in
CNV.
All images are from the same subject; A-C at initial imaging, D-F at
final follow-up. A&D=ICG, B&E=AF, C&F=NIR
Commercial Relationships: Vivek Kumar, None; Patrick A.
Kaszubski, None; Tal Ben Ami, None; Celine Saade, None; Ana
Rita Santos, None; Rufino Silva, None; Maria Luz Cachulo, None;
Jose G. Cunha-Vaz, None; Theodore Smith, None
Support: This work was supported by an individual investigator
research award from the Foundation Fighting Blindness (RTS),
National Institutes of Health/National Eye Institute grant R01
EY015520 (RTS), and unrestricted funds from Research to Prevent
Blindness (RTS).
Program Number: 5152 Poster Board Number: C0216
Presentation Time: 3:45 PM–5:30 PM
Structural correlations of the choriocapillaris, choroid, retina,
and RPE using automated OCT-based measurements in AMD
Johanna D. Beebe1, Elliott H. Sohn1, Li Zhang2, 3, Robert F. Mullins1,
4
, Michael D. Abramoff4, 3. 1Department of Ophthalmology and
Visual Sciences, University of Iowa, Iowa City, IA; 2Department of
Electrical and Computer Engineering, University of Iowa, Iowa City,
IA; 3Iowa Institute for Biomedical Imaging, Iowa City, IA; 4Stephen
A Wynn Institute for Vision Research, Iowa City, IA.
Purpose: Alterations in the choroid play an important role in the
pathogenesis of age-related macular degeneration (AMD). Manual
and semi-automated methods of segmentation have shown good
discrimination of choroidal detail on optical coherence tomography
(OCT) but fully automated protocols to detect outer retinal structures
in AMD are lacking. This was a pilot study, in which we applied a
newly developed method of automated OCT segmentation analysis
to study the correlations of choroid, choriocapillaris, retina, and RPE
thickness in eyes with AMD.
Methods: 38 subjects with AMD had 61 line scan protocol with
enhanced depth imaging performed on the Heidelberg Spectralis SDOCT device. Automated segmentation using a graph-based method
was used to determine thickness of photoreceptor outer segment
length, RPE, choriocapillaris, and choroid. Scans of eyes with CNV
were excluded from analysis. These measurements were plotted
and a linear regression analysis was used to assess for statistical
significance.
Results: The mean thickness (μ) + standard deviation (SD) of the
choriocapillaris, RPE, OSL, and choroid was 8.73+0.65; 24.2+1.00;
39.02+6.42; 158.21+52.21 respectively. The strongest correlation
(r2=0.200) was found between the thickness of the choroid and the
OSL. There was also weak correlation between the RPE and the OSL
(r2=0.121). All other relationships had an r2 < 0.100.
©2015, Copyright by the Association for Research in Vision and Ophthalmology, Inc., all rights reserved. Go to iovs.org to access the version of record. For permission
to reproduce any abstract, contact the ARVO Office at [email protected].
ARVO 2015 Annual Meeting Abstracts
Conclusions: Automated segmentation of SD-OCT images can
be successfully performed to determine thickness of outer retinal
structures of eyes with AMD. There is a plausible correlation
between photoreceptor outer segment length and choroid that needs
confirmation in a larger data set.
Commercial Relationships: Johanna D. Beebe, None; Elliott H.
Sohn, None; Li Zhang, None; Robert F. Mullins, None; Michael D.
Abramoff, IDx LLC (C), IDx LLC (I), University of Iowa (P)
Support: NIH Grant R01 EY018853, R01 EY019112, R01
Program Number: 5153 Poster Board Number: C0217
Presentation Time: 3:45 PM–5:30 PM
Factors affecting the conversion rate of the fellow eye of patients
with neovascular age-related macular degeneration over 5 years
Elizabeth Pearce2, Sobha Sivaprasad1, Victor Chong3. 1Moorfields
Eye Hospital, London, United Kingdom; 2King’s College Hospital,
London, United Kingdom; 3Oxford Eye Hospital, Oxford University
Hospitals, Oxford, United Kingdom.
Purpose: To determine whether baseline functional and anatomical
features can predict the conversion to neovascular age-related
macular degeneration (AMD) in the fellow eye of patients receiving
anti-VEGF treatment for neovascular AMD.
Methods: In total, 50 patients with neovascular AMD in one eye
receiving anti-VEGF treatment were included in the study. The fellow
eye has to have good vision (better than 20/40) and was able to
perform all the functional and anatomical tests. Best corrected visual
acuity (BCVA), retinal sensitivity and fixation stability measured
by microperimetry (Nidek MP1), fundus photography, the central
subfield thickness (CST) and infra-red imaging obtained by the
Heidelberg Spectralis were performed. The drusen area in the central
retina was measured by customised software, and the presence of
reticular pseudo-drusen was assessed on infra-red imaging. Statistical
analysis was performed with Student t-test and Fisher Exact test.The
study was approved by local ethical committee.
Results: In total, 43 patients has completed the 5 year follow up,
14 (32%) of the study eye has developed neovascular AMD during
the follow up. The average age on entry to the study was 74.7
years. There were no significant difference in the age, sex, baseline
CST, BCVA, drusen area and fixation stability between those who
had developed neovascular AMD as compared those who did not.
However, there is a trend that those who converted was more likely
but not statistical significant to have poorer retinal sensitivity,10.5
db vs 8.9 db (p=0.1) and more likely to have reticular pseudodrusen
20.7% vs 42.9% (p=0.1).
Conclusions: In this pilot study, the conversion rate was comparable
to previously reported. Retinal sensitivity and the presence of
reticular pseudodrusen might worth to be studied in a larger cohort.
Commercial Relationships: Elizabeth Pearce, None; Sobha
Sivaprasad, Alimeria Sciences (C), Allergan (C), Bayer (C),
Novartis (C); Victor Chong, Allergan (C), Bayer (C), Novartis (C),
Quantel Medical (C)
Program Number: 5154 Poster Board Number: C0218
Presentation Time: 3:45 PM–5:30 PM
Comparing retinal sensitivity and SDOCT volumes in the AMD
Phenotype and Genotype study
Vincent Tai1, Monica B. Sevilla1, Traci E. Clemons2, Emily Y. Chew3,
Frederick L. Ferris3, Sina Farsiu1, Cynthia A. Toth1. 1Ophthalmology,
Duke University Medical Center, Durham, NC; 2Statistics,
EMMES Corporation, Rockville, MD; 3Epidemiology and Clinical
Applications, National Eye Institute, Bethesda, MD.
Purpose: To study how drusen volume and retinal pigment
epithelium (RPE) abnormal thinning (RAT) on spectral domain
optical coherence tomography (SDOCT) are associated with visual
function as assessed by best corrected visual acuity (BCVA) and
microperimetry retinal sensitivity (MPRS).
Methods: SDOCT results were compared with BCVA and macular
pattern of microperimetry (CenterVue, Inc, San Jose, CA) in patients
with various degrees of age-related macular degeneration (AMD).
After semi-automated SDOCT segmentation of the RPE-drusen
complex (RPEDC), macular volumes for drusen and RAT were
calculated from a SDOCT database of 115 healthy aged eyes1. The
area of evaluation was a 2.5mm radius centered on the fovea. For
general macular MPRS analysis, a median sensitivity score (MSS)
was calculated for all 45 eyes of 42 subjects with MPRS. In a
subgroup of 18 eyes (18 subjects) that each had an identical MPRS
grid pattern, MPRS from each pattern site was paired with SDOCT
volume within a 0.5o radius centered at that site. Volumes were
correlated with BCVA by Spearman rank correlation (ρ), and with
MPRS score by Kruskal-Wallis analysis.
1. Farsiu S et al. Quantitative Classification of Eyes with & without
Intermediate AMD Using OCT. Ophthalmol. 2014
Results: In 53 eyes without neovascular AMD or geographic atrophy,
with a BCVA score and with good quality SDOCT volume, mean
(±standard deviation) drusen volume was 0.020±0.030 mm3, and
the mean RAT volume was 3.48×10-5±7.8×10-5 mm3. Mean BCVA
score was 74.34±14.65. BCVA was worse with an increase in drusen
volume (p=0.0001).
For the 45 eyes with MAIA microperimetry, the mean MSS was
25.76±2.72 dB. Mean drusen volume was 0.021±0.036 mm3 and the
mean RAT volume was 4.93×10-5±11.29×10-5 mm3. MSS did not
correlate with overall drusen volume (p=0.45).
In the sub-group of 18 eyes that used the identical MAIA grid pattern
and that could be directly mapped and compared with OCT results,
better localized MPRS scores correlated with decreasing drusen
volume (p<0.0001) at the MPRS sites.
Conclusions: SDOCT measurements of RPEDC and drusen volumes
correlated with BCVA. In addition, localized MPRS correlates with
the underlying SDOCT drusen volume, while this association is lost
when retinal sensitivity across all of the macular sites is compared to
general macular volumes.
Commercial Relationships: Vincent Tai, None; Monica B. Sevilla,
None; Traci E. Clemons, None; Emily Y. Chew, None; Frederick
L. Ferris, None; Sina Farsiu, Duke University (P); Cynthia A.
Toth, Alcon (F), Bioptigen (F), Genetech (F), NIH 1R01EY023039
(F)
Clinical Trial: NCT01778491
Program Number: 5155 Poster Board Number: C0219
Presentation Time: 3:45 PM–5:30 PM
Widefield Choroidal Thickness Maps in Dry Age Related
Macular Degeneration with and without Reticular Pseudodrusen
Giovanni Gregori, Andrew D. Legarreta, Karen Schaal, John E.
Legarreta, Brian E. Goldhagen, Emeline R. Ramenaden, Zohar
Yehoshua, Philip J. Rosenfeld. Ophthalmology, Bascom Palmer Eye
Institute, Miami, FL.
Purpose: To obtain and compare choroidal thickness maps of eyes
with dry Age Related Macular Degeneration (AMD) over extended
retinal regions.
Methods: Twenty patients with a diagnosis of dry AMD were
imaged using a prototype swept source OCT system from Carl Zeiss
Meditec (Dublin, CA). The OCT system had a central wavelength
of 1050nm and supported acquisition of 12mmx12mmx3mm raster
scans. Each scan used a 512x512 homogeneous sampling scheme.
A novel algorithm was developed to segment the anterior a posterior
boundaries of the choroid over the full OCT datasets.
©2015, Copyright by the Association for Research in Vision and Ophthalmology, Inc., all rights reserved. Go to iovs.org to access the version of record. For permission
to reproduce any abstract, contact the ARVO Office at [email protected].
ARVO 2015 Annual Meeting Abstracts
Results: Pointwise thickness maps of the choroid were obtained
over the scan area using the automated algorithm. Using the
associated OCT fundus images, as well as other en face OCT
images, choroidal thickness information can be correlated to retinal
landmarks. In particular we examined the relationship between the
choroidal thickness and features like geographic atrophy, reticular
pseudodrusen, and drusen. The choroid is in general thinner in
eyes with reticular pseudodrusen than in eyes without reticular
pdeudodrusen, but the precise patterns of choroidal thickness vary
significantly across different eyes.
Conclusions: There can be considerable variability in choroidal
thickness at different retinal locations, in eyes with dry AMD.
Also the relationship between local choroidal thickness and other
features of dry AMD can assume different patterns in different eyes.
The detailed quantitative descriptions of choroidal thickness over
extended regions have the potential to improve our understanding of
retinal pathology.
Commercial Relationships: Giovanni Gregori, Carl Zeiss Meditec
(F); Andrew D. Legarreta, None; Karen Schaal, None; John
E. Legarreta, None; Brian E. Goldhagen, None; Emeline R.
Ramenaden, None; Zohar Yehoshua, None; Philip J. Rosenfeld,
Carl Zeiss Meditec (F)
Support: NIH Center Core Grant P30EY014801, RPB Unrestricted
Award, DOD- Grant# W81XWH-13-1-0048
Program Number: 5156 Poster Board Number: C0220
Presentation Time: 3:45 PM–5:30 PM
Automated Segmentation of Reticular Pseudodrusen and Regular
Drusen in Eyes with Non-neovascular AMD
Zhihong Hu1, Jun Xiao1, 2, Kiran Nandanan1, Srinivas R. Sadda1.
1
Doheny Eye Institute, UCLA, Los Angeles, CA; 2The Second
Hospital of Jilin University, Jilin, China.
Purpose: To quantify regular drusen (RD) and reticular pseudodrusen
(RPD) in optical coherence tomography (OCT) images from eyes
with non-neovascular age-related macular degeneration (AMD) using
an automated analysis algorithm.
Methods: In this IRB-approved study, three groups of subjects
underwent spectral domain (SD) OCT imaging in one eye using a
macular cube protocol (Spectralis OCT). Group 1 consisted of 17
healthy subjects (referred as “Normal”); group 2 had 6 AMD subjects
with only RD (referred as “Non-reticular”); and group 3 included
16 AMD subjects with both RD and RPD (referred as “Reticular”).
A graph-based approach was applied to automatically segment 5
sub-retinal boundaries: ellipsoid zone (EZ) outer, interdigitation zone
(IZ), retinal pigment epithelium (RPE) inner, RPE outer, and choroid
inner (CI). The segmentation was manually corrected as needed by a
certified OCT grader. Using these segmented boundaries, three layers
were generated for quantitative thickness analysis: (1) RPD layer
bounded by IZ and RPE inner, (2) RD layer bounded by RPE outer
and CI, and (3) RPE-drusen complex (RPEDC) bounded by EZ and
CI. Thicknesses of these layers were computed in a 4mm*4mm grid
(including 4 sub-quadrants,) as well as a circular grid (r = 1.44 mm)
centered on the fovea. Values were compared among the 3 groups
using t-test.
Results: Fig. 1 shows the mean regional thicknesses in RPD and
RD layers and Fig. 2 shows these values in RPEDC layer. The mean
thickness of RPD layer in 4mm*4mm grid was significantly greater
(p < 0.01) than in the circular grid (r = 1.44mm) with the highest
mean thickness in the superior-nasal quadrant. Conversely, the
mean thicknesses of RD and RPEDC layers in 4mm*4mm grid was
significantly smaller than that in the circular region for both “Nonreticular” (p < 0.01) and “Reticular” groups (p < 0.01). When RPD
and RD were both present, the mean RD layer thickness was greater
than that in the “Non-reticular” group in all the regions, but not
statistically significant.
Conclusions: Automated segmentation analysis suggests that RPD
are most extensive/thicker in the superior-nasal quadrant of the
macula whereas RD are thicker centrally, highlighting differences in
regional distribution of these lesions. The combined RPEDC layer
shows a similar regional distribution to RD suggesting that this
measure is more driven by the presence of RD.
Fig.1
Fig. 2
Commercial Relationships: Zhihong Hu, None; Jun Xiao, None;
Kiran Nandanan, None; Srinivas R. Sadda, Allergan (C), Carl
Zeiss Meditec (C), Carl Zeiss Meditec (F), Optos (C), Optos (F),
Optovue, Inc. (F), Regeneron (C)
Support: This work was supported in part by the Beckman Macular
Degeneration Research Center and a Research to Prevent Blindness
Physician Scientist Award.
©2015, Copyright by the Association for Research in Vision and Ophthalmology, Inc., all rights reserved. Go to iovs.org to access the version of record. For permission
to reproduce any abstract, contact the ARVO Office at [email protected].
ARVO 2015 Annual Meeting Abstracts
Program Number: 5157 Poster Board Number: C0221
Presentation Time: 3:45 PM–5:30 PM
Drusen morphology changes in nonexudative age-related
degeneration using spectral domain optical coherence
tomography after oral antioxidants supplementation: one-year
results.
Xavier Valldeperas1, Pau Romera1, Rafael Abos-Herrandiz2, Antoni
Sabala1. 1Ophthalmology, Hospital Universitari Germans Trias,
Badalona, Spain; 2Primary Health Care Division. Institut Català de la
Salut. Barcelona (Spain), Barcelona, Spain.
Purpose: To determine drusen morphology (volume and area)
changes in nonexudative age-related macular degeneration (AMD)
after one year of oral supplementation with AREDS-like formulation.
Methods: Patients with AREDS category 2 and 3 AMD were
prospectively enrolled in this study, and were randomized to receive
daily oral supplementation with lutein (12mg) + zeaxanthin (2mg)
+ astaxanthin (8mg) + omega-3 fatty acids (docosahexaenoic acid
[DHA] 540mg + eicosapentaenoic acid [EPA] 360mg) + vitamin
C (40mg) + vitamin E (20mg) + zinc (16mg) + copper (2mg),
or observation during one year. ETDRS vision, biomicroscopy,
intraocular pressure (IOP), color fundus photography and automatic
measurement of drusen with Topcon 3D-OCT 2000 (Topcon, Tokyo,
Japan) using the 6 x 6 mm 3D cube scan protocol, were performed in
all patients, at baseline and 12 months after. Automated delineation
of macular drusen was modified by the investigators when evident
segmentation errors occurred.
Results: Seventy eyes of 35 patients were included: 18 patients
received oral supplementation and the rest were observed. Visual
acuity and IOP did not significantly change in either group after 12
months. In the treatment group, drusen count did not significantly
change (p=0.715) and increase in drusen area and volume was not
statistically significant (p=0.304 and p=0.085, respectively). In the
observed group, drusen count significantly increased after 12 months
from 13.9±14.1 to 15.5±15.1 (p=0.045), as well as drusen volume
(p=0.038). Drusen area in this group remained unchanged during the
study period.
Conclusions: Patients with oral supplementation with the AREDSlike formulation show a tendency to slow drusen increase, measured
with SD-OCT. The clinical relevance of these findings is still unclear
and need further comparison with natural history in larger population
and longer duration.
Commercial Relationships: Xavier Valldeperas, None; Pau
Romera, None; Rafael Abos-Herrandiz, None; Antoni Sabala,
None
Clinical Trial: AC-11-112 (NCT02264938)
Program Number: 5158 Poster Board Number: C0222
Presentation Time: 3:45 PM–5:30 PM
Analysis of choroidal maps and fundus autofluorescence
correlates in non-exudative age-related macular degeneration
using swept source optical coherence tomography
Vittorio Capuano, Alexandra Miere, Oudy Semoun, Pietro Frascio,
Giuseppe Querques, Eric H. Souied. Department of Ophthalmology,
Centre hospitalier Intercommunal de Crïteil, Creteil, France.
Purpose: To analyze choroidal thickness maps (CMs) and fundus
autofluorescence (FAF) in patients with non-exudative age-related
macular degeneration (AMD) using swept source optical coherence
tomography (Swept-OCT).
Methods: CMs imaging were obtained using Swept-OCT (Topcon
Medical Systems, Oakland, NJ). A standardized imaging protocol
was performed in all patients: radial diameter 9.0 mm scans through
the foveal center with automated ETDRS choroidal thickness map
(9 sectors). FAF images obtained using Spectralis HRA+OCT
(Heidelberg Engineering, Heidelberg, Germany) were overlay with
Swept-OCT. Eyes were divided in: Group 1, early non-exudative
AMD with at least one large soft drusen (>125 mm); Group 2, early
non-exudative AMD with intermediate distribution of reticular
pseudodrusen; Group 3, late non-exudative AMD / Geographic
atrophy (GA); and Group 4, control subjects with no ocular diseases
The mean thickness was automatically measured in the “center”
sector within 1 mm from the center of the fovea, in 4 “inner ring”
sectors (superior, inferior, nasal, and temporal; 1 to 2 mm from the
center of the fovea), and in 4 “outer ring” sectors (superior, inferior,
temporal, and nasal; 2 to 3 mm from the center of the fovea).
Results: A total of 72 eyes of 72 consecutive patients with nonexudative AMD (56 females; mean age 79.1±8.1 years) were
included in the analysis. The mean whole choroidal thickness were
157 ± 55 mm, 126 ± 52 mm, 114 ± 38 mm et 18 8± 74 mm (in group
1,2,3 and 4 respectively). The CMs were significantly reduced in
Group 2, and 3 compared with group 1 and 4 (p <0.05). A similar
reduction in CMs were found in Group 3, compared with group 2. (p
0.001).
On the basis of FAF features in Group 3, a total of 70 ETDRS
sectors were categorized as hypo-FAF (sectors characterized by >
50% absence of FAF), and 83 ETDRS sectors were categorized as
hyper/iso-FAF (sectors characterized by ≤ 50% absence of FAF). No
statistical differences in CMs were found among ETDRS sectors with
>50% and <50% atrophic (HypoFAF) or preserved retina (Hyper/
IsoFAF) in GA group (p 0.07).
Conclusions: CMs reveal a thinner choroid in eyes with GA and
Pseudodrusen AMD patients compared with drusen and control
subjects on swept OCT. CM in the different ETDRS sectors was not
associated with FAF features in eyes with GA.
Commercial Relationships: Vittorio Capuano, None; Alexandra
Miere, None; Oudy Semoun, None; Pietro Frascio, None;
Giuseppe Querques, None; Eric H. Souied, None
Program Number: 5159 Poster Board Number: C0223
Presentation Time: 3:45 PM–5:30 PM
Microperimetry and Multifocal Electroretinography in
Reticular Pseudodrusen and Intermediate Age-Related Macular
Degeneration
Chi D. Luu1, 2, Zhichao Wu1, Lauren N. Ayton1, 2, Galina Makeyeva1,
Robyn H. Guymer1, 2. 1Centre for Eye Research Australia, East
Melbourne, VIC, Australia; 2Department of Ophthalmology,
University of Melbourne, Melbourne, VIC, Australia.
Purpose: Reticular pseudodrusen (RPD) has been increasingly
recognized as a risk factor for the development of advanced agerelated macular degeneration (AMD). Although RPD are often
present in eyes with features of the early stages of AMD, studies
of the impact of RPD on retinal function to date have only been
performed in eyes with RPD only. Thus, the purpose of this study
was to investigate the influence of RPD on retinal function in
eyes with intermediate AMD using microperimetry and multifocal
electroretinography (mfERG).
Methods: In this prospective cross-sectional study, mfERG,
microperimetry, colour fundus photography, near-infrared
reflectance (NIR) imaging and spectral-domain optical coherence
tomography (SD-OCT) scans were performed in participants with
bilateral intermediate AMD (drusen >125 μm). The presence of
RPD was defined as groups of hyporeflective lesions against a
mildly hyperreflective background on NIR, with corresponding
hyperreflective signals above the RPE band on SD-OCT. The
presence and extent of pigmentary changes and RPD within the
central 3 mm diameter was examined using an Early Treatment of
Diabetic Retinopathy Study (ETDRS) grid. Drusen volume within the
©2015, Copyright by the Association for Research in Vision and Ophthalmology, Inc., all rights reserved. Go to iovs.org to access the version of record. For permission
to reproduce any abstract, contact the ARVO Office at [email protected].
ARVO 2015 Annual Meeting Abstracts
central 3 mm diameter was also determined. The influence of these
pathological features on microperimetry and mfERG within the same
region were examined.
Results: A total of 120 eyes of 60 participants (mean age was 70.5 ±
7.2 years, range 51 to 84) were included in this study. There were 39
eyes with RPD and 81 eyes without RPD. Microperimetric sensitivity
was not significantly associated with the presence and extent of
RPD (β Coefficient = -0.18, p = 0.068), but with drusen volume (β =
-5.65, p < 0.001) and extent of pigmentary changes (β = -0.48, p <
0.001). The mfERG implicit time was independently and significantly
associated with the presence and extent of RPD (β = 0.38, p = 0.001),
drusen volume (β = 3.23, P = 0.023) and the extent of pigmentary
changes (β = 0.55, p < 0.001). However, mfERG response amplitude
was not significantly associated with the presence and extent of RPD
(β = -0.75, p = 0.130).
Conclusions: In eyes with intermediate AMD, the presence and
extent of RPD had a significant impact on cone-mediated retinal
function as measured by mfERG. Microperimetric sensitivity was
influenced by AMD features but not by the presence or extent of
RPD.
Commercial Relationships: Chi D. Luu, None; Zhichao Wu,
None; Lauren N. Ayton, None; Galina Makeyeva, None; Robyn H.
Guymer, None
Support: National Health and Medical Research Council
(NH&MRC) Project Grant (#1027624), Macular Disease Foundation
Australia (MDFA) Research Grant, Bupa Health Foundation
(Australia) and the Menzies Foundation.
Program Number: 5160 Poster Board Number: C0224
Presentation Time: 3:45 PM–5:30 PM
Choroidal thickness in eyes with central geographic atrophy
secondary to Stargardt disease and age-related macular
degeneration
Renata Portella Nunes1, 2, Potyra R. Rosa1, Raquel Goldhardt1,
Mariana Rossi Thorell1, Giovanni Gregori1, William J. Feuer1, Byron
L. Lam1, Giovanni Staurenghi3, Philip J. Rosenfeld1. 1Bascom Palmer
Eye Institute, Florianopolis, Brazil; 2Ophthalmology and Visual
Sciences department, Paulista School of Medicine, São Paulo, Brazil;
3
Deptartment of Clinical Science - Luigi Sacco, University of Milan,
Milan, Italy.
Purpose: Geographic atrophy (GA) in Stargardt disease (STGD)
results from the abnormal accumulation of A2E in photoreceptors,
which subsequently causes loss of the retinal pigment epithelium
(RPE), photoreceptors, and choriocapillaris. In age-related macular
degeneration (AMD), the primary cause of GA is not known. If the
presence of GA affects the underlying choroidal thickness (CT),
then we might expect eyes with similar amounts of GA, regardless
of etiology, to have similar CT measurements. If there’s a difference
in the CT measurements underlying GA in AMD and STGD,
then this difference might reflect a difference in the underlying
pathophysiology leading to GA. To determine if different diseases
causing GA have a similar or different affect on subfoveal CT
measurements, we compared measurements from eyes with similar
areas of GA secondary to AMD and STGD.
Methods: Patients with the diagnosis of central GA secondary to
STGD and AMD were enrolled in a prospective spectral domain
optical coherence tomography imaging study. Subfoveal CT
was measured from the central B-scan using an enhanced depth
imaging protocol. The area of GA was measured using fundus
autofluorescence imaging. Two independent graders measured the
subfoveal CT and areas of GA. AMD eyes were divided into those
with and without reticular pseudodrusen. CT from all group were
compared.
Results: A total of 22 eyes of 22 patients were included in the STGD
group and in the AMD group, and these eyes were matched with
respect to the area of GA. The mean age of the STGD patients was
48.9 (SD 17.1) and 81.8 (SD 6.2) for the AMD patients. Mean area
measurements of GA for the STGD and AMD groups were 5.4mm2
(SD 4.1) and 5.1 mm2 (SD 4.0), respectively (P=0.83). After adjusting
for age, eyes with STGD had a mean CT measurement greater
than the AMD eyes (336.1 vs.198.1mm respectively; p=0.039). But
this difference seemed to be driven by AMD eyes with reticular
pseudodrusen (RPD) and a single Stargardt case with a very thick
choroid. Eyes with RPD had statistically thinner choroids when
compared to all other groups.
Conclusions: No clinically meaningful difference was observed
between the CT in normal eyes and eyes with STGD and AMD
without RPD containing similar areas of GA. However, eyes with GA
and RPD had significantly thinner subfoveal CT.
Commercial Relationships: Renata Portella Nunes, None; Potyra
R. Rosa, None; Raquel Goldhardt, None; Mariana Rossi Thorell,
None; Giovanni Gregori, Carl Zeiss (F), Carl Zeiss (P); William
J. Feuer, None; Byron L. Lam, None; Giovanni Staurenghi,
HEIDELBERG ENGINEERING (C), OPTOS, INC. (C), OPTOVUE
(S), ZEISS (C); Philip J. Rosenfeld, Alexion Pharmaceuticals (F),
Carl Zeiss (F)
Program Number: 5161 Poster Board Number: C0225
Presentation Time: 3:45 PM–5:30 PM
Stabilization of Visual Acuity with Bilateral Macular Drusen a 10 year prospective study of uniocular peripheral retina argon
laser therapy
Danny H.-Kauffmann Jokl1, Sankha Amarakoon2, Celine Saade3,
Robert Post3, Ansh Johri3, Sander Kesting2, Suzanne Yzer2, Theodore
Smith3, Rando Allikmets1, Jan C. van Meurs2. 1Ophthalmology,
Columbia University, Bronxville, NY; 2Rotterdam Eye Hospital,
Rotterdam, Netherlands; 3New York University, New York, NY.
Purpose: Anti VEGF therapy can reduce the neovascular
complications of AMD, only AREDS supplements
can decrease such progression over a similar time period. Natural
history of drusen shows 71% with bilateral medium soft drusen
progressing to large drusen and, in 14%, to AMD over 10 years,
though, if AMD did not develop, with preservation of good vision.
(Chew JAMA Ophthalmol 2014;132:272). Following a serendipitous
observation that a retinal laser treatment for an incidental retinal tear
in an eye with medium soft drusen, with AMD in the other eye, was
followed by diminution of drusen and stabilization of vision over 10
years, we investigated the effect of such treatment on patients with
bilateral soft drusen.
Methods: Fourteen patients from the Rotterdam Eye Hospital with
bilateral macular drusen and visual acuity in each eye from 20/2020/40 consented to a single uniform treatment of 200 argon laser
applications (200 microns, 200 mw each) to the anterior superior
temporal retina. Eleven subjects (5 M, 6F; ages 58-83) were followed
for 10 years (3 subjects died during this period) with clinical
examinations and digitalized fundus photos. Drusen areas in each eye
were calculated and DNA of all subjects was analyzed for selected
AMD-associated SNPs in ARMS2 and CFH, the two major genetic
loci for AMD. The masked clinical and genetic data were then
correlated.
Results: Uniocular visual loss (≤20/100) to AMD in the 10 year
follow up period occurred in three (3/11) patients, in each case in the
non-treated eye. Bilateral visual loss to AMD occurred in one patient.
Two patients with high genetic risk showed a decrease in the drusen
area (Fig.1). The same 2 out of eleven (2/11) patients with high
genetic risk did not progress to late AMD in 10 years (Fig. 2). There
©2015, Copyright by the Association for Research in Vision and Ophthalmology, Inc., all rights reserved. Go to iovs.org to access the version of record. For permission
to reproduce any abstract, contact the ARVO Office at [email protected].
ARVO 2015 Annual Meeting Abstracts
was no association between the progression of drusen area and the
visual outcome (Fig. 3).
Conclusions: Due to the small cohort size, this 10 year prospective
pilot study can only suggest that a focal peripheral laser treatment in
one eye may provide a mechanism, e.g., an inflammatory response
that preserves binocular vision in some subjects with bilateral
macular drusen relative to the natural history and does not correlate
with the extent of macular soft drusen. A much larger cohort study
would be necessary to draw conclusions as to the genetic correlations,
if any, with this laser mode of therapy.
Results: In a cohort of 30 male and 17 female subjects with CAD,
the mean age was 63.5 years (range 44-75). 9 subjects (19.15%) had
evidence of SDD on EDI SD-OCT, significantly greater than the
known population prevalence by CP of 0.7% (P=<0.001). Allowing
for the decreased sensitivity of CP (about 35%) for RMD compared
to EDI SD-OCT, significance is still maintained (P=<0.001).
Examples of EDI-OCT imaging from a subject with CAD, and a
normal control, are demonstrated in Figure 1.
Conclusions: While previous studies have shown an overlap between
AMD and CAD risk factors, the choroidal alterations of RMD are
more strongly indicative of a vascular etiology and thus a potential
link to CAD. Our results clearly demonstrate an increased prevalence
of SDD among our CAD sample when compared to the prevalence
in the general population established by the BDES. These results
suggest that RMD and CAD may share a common pathophysiology.
Commercial Relationships: Danny H.-Kauffmann Jokl, None;
Sankha Amarakoon, None; Celine Saade, None; Robert Post,
None; Ansh Johri, None; Sander Kesting, None; Suzanne Yzer,
None; Theodore Smith, None; Rando Allikmets, None; Jan C. van
Meurs, None
Clinical Trial: AAAD5359
Program Number: 5162 Poster Board Number: C0226
Presentation Time: 3:45 PM–5:30 PM
The Association Between Reticular Macular Disease (RMD) and
Coronary Artery Disease (CAD)
Patrick A. Kaszubski, Colleen Cunningham, Vivek Kumar, Camellia
Nabati, Rachel M. Cymerman, Theodore Smith. Ophthalmology, New
York University School of Medicine, New York, NY.
Purpose: Reticular Macular Disease (RMD), a leading risk factor for
disease progression in Age-Related Macular Degeneration (AMD),
demonstrates both subretinal drusenoid deposits (SDD) and choroidal
alterations (vascular involvement). The goal of this study was to
compare the prevalence of SDD in subjects aged 30-75 years who
have documented cardiovascular disease, specifically CAD, to the
established prevalence of RMD by color fundus photography (CP)
in the general population (Beaver Dam Eye Study (BDES)) [Klein,
R. Am J Ophthalmology 2008]. CAD is a condition that can strike
in middle age, while AMD affects the elderly. A middle-aged CAD
cohort allows a search for early SDD that could link CAD and AMD/
RMD.
Methods: 47 consecutive subjects with documented CAD (ages 40 to
75 years), were enrolled from a cardiology clinic at a large city public
hospital. Before undergoing enhanced depth imaging spectral domain
optical coherence tomography (EDI SD-OCT) imaging, all subjects
completed a questionnaire regarding medical and ocular history. A
retina specialist evaluated the images for evidence of SDD and other
retinal findings. Chi-square goodness of fit was used for statistical
analysis.
Figure 1:
Top: Normal 75 year old female. Bottom: 59 year old male with
CAD. The subretinal space is marked by yellow asterisks. Note SDD
in bottom panel.
Commercial Relationships: Patrick A. Kaszubski, None; Colleen
Cunningham, None; Vivek Kumar, None; Camellia Nabati, None;
Rachel M. Cymerman, None; Theodore Smith, Advanced Cell
Technologies (C)
Support: This work was supported by an individual investigator
research award from the Foundation Fighting Blindness (RTS),
National Institutes of Health/National Eye Institute grant R01
EY015520 (RTS), and unrestricted funds from Research to Prevent
Blindness.
©2015, Copyright by the Association for Research in Vision and Ophthalmology, Inc., all rights reserved. Go to iovs.org to access the version of record. For permission
to reproduce any abstract, contact the ARVO Office at [email protected].
ARVO 2015 Annual Meeting Abstracts
Program Number: 5163 Poster Board Number: C0227
Presentation Time: 3:45 PM–5:30 PM
Retinal Layer Thickness in Drusen and Drusen-Free Retinal
Areas in Age-Related Macular Degeneration
Lisa Nivison-Smith1, 4, James Rogala2, Barbara Zangerl4, 1, Nagi
Assaad3, 4, Erica L. Fletcher5, Michael Kalloniatis4, 1. 1School of
Optometry and Vision Science, University of New South Wales,
Kensington, NSW, Australia; 2Western University, Pomona, CA;
3
Ophthalmology Department, Prince of Wales Hospital, Randwick,
NSW, Australia; 4Centre For Eye Health, University of New South
Wales, Kensington, NSW, Australia; 5Department of Anatomy and
Neuroscience, University of Melbourne, Parkville, NSW, Australia.
Purpose: Drusen are a hallmark of age-related macular degeneration
(AMD). However how drusen affect retinal structure at and beyond
their borders is still unclear. This study examines changes in retinal
thickness which occurs above drusen and compares this to drusenfree areas in the same patient and a normative population.
Methods: Spectral domain optical coherence tomography scans
through drusen in early to intermediate AMD patients (n=122) or
patients with no ocular disease (n=30) seen at the Centre for Eye
Health were reviewed and the thickness of individual retinal layers
was measured above the druse and in a drusen-free area, 150mm from
the drusen edge. Patient written consent was obtained in accordance
with the Declaration of Helsinki and approved by the Biomedical
Human Research Ethics Advisory Panel of the University of New
South Wales.
Results: Retinal thickness above drusen was significantly less (p <
0.001) than drusen-free areas (16±1%). Thinning occurred almost
exclusively in the outer retinal layers with almost no thinning in the
inner retina for single isolated drusen. Inner retinal thickness was
however reduced over large confluent drusen (5±1%). Interestingly,
the overall retinal thickness of both drusen and drusen-free areas was
significantly less to matching eccentricities in the normal population.
Conclusions: Drusen caused retinal thinning, mostly by affecting the
outer retina. Drusen-free areas in AMD patients were also thinned
suggesting the effects of drusen extend beyond the lesion borders.
This has implications in the use of presumably “normal” areas of
retina in AMD patients for disease assessment.
Commercial Relationships: Lisa Nivison-Smith, None; James
Rogala, None; Barbara Zangerl, None; Nagi Assaad, None; Erica
L. Fletcher, None; Michael Kalloniatis, None
Support: NHMRC Grant 1033224, UNSW ECR Grant PS35430
Program Number: 5164 Poster Board Number: C0228
Presentation Time: 3:45 PM–5:30 PM
Thickness map of outer retinal layer and choroid in reticular
pseudodrusen
Reiko Izumi, Ichiro Maruko, Machiko Kimura, Hideki Koizumi,
Takahiko Izumi, Tomohiro Iida. Tokyo Women’s Medical University,
Tokyo, Japan.
Purpose: Although reticular pseudodrusen (RPD) are often first
observed in the upper part of the macula, the reason remains unclear.
Our purpose in the current study is to evaluate the relationship
between the outer retinal layer and the choroid using divided
thickness map in Japanese patients with RPD.
Methods: Eighteen eyes of 14 patients (average 83 years old)
in RPD, determined by color fundus photograph, were included.
The eyes with exudative age-related macular degeneration were
excluded because of indetermination. 3D-scan was taken using
swept source optical coherence tomography (DRI-OCT, Topcon,
Japan). Segmentation lines of outer retinal layer (superior border
of outer plexiform layer to retinal pigment epithelium; RPE) and
choroidal layer (RPE to chorioscleral interface; CSI) were drawn
manually within 6mm at the macula. All areas were divided by 9
sectors according to ETDRS grid map style. The eyes in age-matched
patients (18 eyes of 17 patients) without retinal and choroidal
disorder were also examined as control.
Results: Mean outer retinal layer thickness within ETDRS map was
significantly thinner in RPD than control (133±12mm vs 143±8mm,
P<0.01). Mean choroidal thickness within ETDRS map was also
significantly thinner in RPD than control (125±39mm vs 216±77mm,
P<0.01). The thicknesses of outer retinal layer and choroid in all
areas of ETDRS map were correlated with each other (r=0.22,
P<0.01). Especially, the significant correlation was reached at upper
areas (inner and outer quadrant) of ETDRS map (r=0.37, P=0.03).
Conclusions: Thinning of not only choroid but also outer retinal
layer indicates the impairing of the photoreceptors in RPD. The
correlations of thicknesses between outer retinal layer and choroid at
upper quadrant area may elucidate the origin of RPD development.
Commercial Relationships: Reiko Izumi, None; Ichiro Maruko,
None; Machiko Kimura, None; Hideki Koizumi, None; Takahiko
Izumi, None; Tomohiro Iida, None
Program Number: 5165 Poster Board Number: C0229
Presentation Time: 3:45 PM–5:30 PM
Change in Outer Retinal Layer Thickness in Subjects with
Reticular Pseudodrusen
Muneeswar Gupta Nittala1, Yan Luo1, Zhihong Hu1, Ruth E. Hogg2,
Rufino Silva3, Giovanni Staurenghi4, Usha Chakravarthy2, Srinivas
R. Sadda1, 5. 1Ophthalmology, Doheny Eye Institute, Los Angeles,
CA; 2Center for Experimental Medicine, Queen’s University Belfast,
Belfast, United Kingdom; 3Ophthalmology Unit, Centro Hospitalare
Universitário Coimbra, Coimbra, Portugal; 4Department of Clinical
Science Luigi Sacco’ University of Milan, Milan, Italy; 5Department
of Ophthalmology, David Geffen School of Medicine at UCLA, Los
Angeles, CA.
Purpose: To evaluate the thickness of the outer retinal layers in
subjects with reticular pseudodrusen.
Methods: Thirty four eyes of 34 subjects with reticular pseudodrusen
(RPD) and 39 eyes of 39 subjects with early non-neovascular agerelated macular degeneration without RPD were enrolled in this
prospective study at three clinical sites (Belfast, Coimbra, Milan).
These subjects been fallowed up for one year. Spectral domain
optical coherence tomography (SD-OCT) volume scans was collected
in 3 clinical sites from Belfast, Coimbra and Milan using Spectralis
HRA+OCT (Heidelberg Engineering, Heidelberg Germany). Detailed
manual segmentation of outer retinal layers was performed using
custom designed and validated grading software 3D OCTOR.
Thickness measurements were derived for retina, photoreceptor outer
segments, RPE+Drusen complex and Choroid layers in both the study
cohorts.
Results: was no significant difference in retinal thickness and
photoreceptor outer segments thickness between study groups. There
was significant (p=0.004) difference in RPE+Drusen complex layer
thickness in RPD subjects (30.6 ± 3.69 (±SD) mm) compared to
those without RPD (28.21 ± 2.35 mm). In contrast, the choroid was
significantly thinner (p = 0.04) in RPD subjects with a thickness
of149.78 ± 33.56 compared with 162.42 ± 36.24 mm in subjects
without RPD. Longitudinal analysis, revealed a small but significant
(p = 0.04) thinning (25.53 ± 3.71 mm at baseline vs 24.03 ± 4.32 mm
at month 12) in RPD subjects, compared to no significant change
over time in subjects with no RPD.
Conclusions: The RPE+ Drusen complex layer is significantly
thicker and the choroid is significantly thinner in subjects with
reticular pseudodrusen. The photoreceptor outer segments appear to
thin over time in eyes with RPD.
©2015, Copyright by the Association for Research in Vision and Ophthalmology, Inc., all rights reserved. Go to iovs.org to access the version of record. For permission
to reproduce any abstract, contact the ARVO Office at [email protected].
ARVO 2015 Annual Meeting Abstracts
Commercial Relationships: Muneeswar Gupta Nittala, None; Yan
Luo, None; Zhihong Hu, None; Ruth E. Hogg, None; Rufino Silva,
Alcon, Alimera, Allergan, Bayer, Novartis, THEA (C); Giovanni
Staurenghi, None; Usha Chakravarthy, None; Srinivas R. Sadda,
Carl Zeiss Meditec, Optos, Allergan (R), Carl Zeiss Meditec, Optos,
Allergan, Genentech (F), Carl Zeiss Meditec, Optos, Allergan,
Genentech, Alcon, Novartis, Roche (C)
Clinical Trial: NA
Program Number: 5166 Poster Board Number: C0230
Presentation Time: 3:45 PM–5:30 PM
Peripapillary choroidal thickness in early age related macular
degeneration patients with reticular pseudodrusen
Jong-Hyun Oh1, Jaeryung Oh2, Cheolmin Yun2, Young Ho Kim2,
Seong-Woo Kim2, Kuhl Huh2. 1Ophthalmology, Dongguk University
Ilsan Hospital, Goyang, Korea (the Republic of); 2Ophthalmology,
Korea University College of Medicine, Seoul, Korea (the Republic
of).
Purpose: To investigate peripapillary and macular choroidal
thickness (CT) of patients with early age-related macular
degeneration (AMD) with or without reticular pseudodrusens (RPD).
Methods: We investigated the medical records of 89 eyes of 89
patients with early AMD. Eyes with early AMD were categorized into
three groups according to the extent of RPD; no RPD, localized RPD,
and diffuse RPD. Peripapillary and macular CT were measured with
images obtained by spectral domain optical coherence tomography
(SD-OCT). CT in peripapillary and macular area was compared
among groups.
Results: Both RPD groups were older and female-predominant than
non-RPD group (P = 0.007 and P = 0.030, respectively). Macular
and peripapillary CT were different among three groups (all, P <
0.001) and both RPD group showed thinner choroid at all areas than
those of non-RPD group after adjustment of age and sex (all, P ≤
0.016). Temporal peripapillary and nasal macular CT at 500 mm and
1500 mm from the fovea in eyes with diffuse RPD were significantly
thinner than those in eyes with localized RPD (P = 0.008, P < 0.001
and P = 0.018 respectively).
Conclusions: In addition to macular area, peripapillary CT including
outside of macula was thinner in eyes with RPD than those without
RPD. The papillomacular choroid showed significant changes
according to the distribution type of RPD. This result may suggest
that choroidal thinning is associated with RPD progression.
Commercial Relationships: Jong-Hyun Oh, None; Jaeryung Oh,
None; Cheolmin Yun, None; Young Ho Kim, None; Seong-Woo
Kim, None; Kuhl Huh, None
Support: Korean Ministry of Environment (2012001350010)
©2015, Copyright by the Association for Research in Vision and Ophthalmology, Inc., all rights reserved. Go to iovs.org to access the version of record. For permission
to reproduce any abstract, contact the ARVO Office at [email protected].