ARVO April 26-27, 2019 Vancouver Convention Centre East Ballrooms Vancouver, B.C.

Program Organizers James G. Fujimoto, PhD Massachusetts Institute of Technology Cambridge, Mass.

Wolfgang Drexler, PhD Medical University Vienna Vienna, Austria

Joel S. Schuman, MD NYU Langone Health, NYU School of Medicine New York, N.Y.

Table of Contents

Topic Page #

Cover…………………………………………………………………………………………………………………... 1 Sponsors………………………………………………………………………………………………………………. 4 Friday Agenda………………………………………………………………………………………………………. 5 Invited Speaker……………………………………………………………………………………………………. 6 Saturday Agenda………………………………………………………………………………………………….. 7 Program Information……………………………………………………………………………………………. 28 Thank you to our sponsors…………….…………………………………………………………………… 30

Paper Session (PDFs)

OCT Angiography………………………………………………………………………………………………….. 32 Cellular and Molecular Imaging……………………………………………………………………………. 50 New and Enhanced Technologies…………………………………………………………………………. 66 Artificial Intelligence, Image Processing, and Analysis………………………………………….. 82

Poster Session (PDFs)

New Technologies………………………………………………………………………………………………... 101 OCT Angiography…………………………………………………………………………………………………. 141 Artificial Intelligence…………………………………………………………………………………………….. 214 Clinical Imaging……………………………………………………………………………………………………. 287 MRI………………………………………………………………………………………………………………………. 375 Ultrasound…………………………………………………………………………………………………………… 385 Image Guided Surgery………………………………………………………………………………………….. 391 Image Processing and Image Analysis………………………………………………………………….. 399 Biomechanics………………………………………………………………………………………………………. 415 Fluorescence……………………………………………………………………………………………………….. 424 Basic Science………………………………………………………………………………………………………… 431 2019 Imaging in the Eye Conference

Friday, April 26, 2019 6:00 – 8:30 PM and Saturday, April 27, 2019 8:00 AM – 5:30 PM

Agenda and Program Information

Vancouver Convention Centre 999 Canada Place Vancouver, BC V6C 3E1, Canada

Please note that the conference syllabus will be available online only to registered attendees. A link will be sent prior to the conference to those that are registered A QRcode linking to the syllabus also will be printed on the registration receipt. ARVO gratefully acknowledges our Imaging in the Eye Conference sponsors. 2019 Imaging in the Eye Conference Agenda April 26 – 27, 2019 Vancouver Convention Centre (East) Ballroom A, B, and C (Conference Level) East 1, 2 and 3 (Meeting Level) Friday, April 26, 2019

4:00 – 9:00 pm Registration Ballroom A Foyer

Welcome and Opening Remarks Ballroom A & B Invited Speaker

James G. Fujimoto, PhD 6:00 – 6:15 pm Welcome/thank you to the sponsors and opening remarks Joel S. Schuman, MD, FARVO Wolfgang Drexler, PhD

Thank you to our sponsors Carl Zeiss Meditec, Wasatch Photonics, Topcon and Optos, Inc.

6:15 – 6:30 pm Introduction of the Invited Speaker James G. Fujimoto, PhD Joel S. Schuman, MD, FARVO Wolfgang Drexler, PhD

6:30 – 7:15 pm OCT in the Diagnosis and Management of Philip J. Rosenfeld, MD, PhD Age-Related Macular Degeneration Bascom Palmer Eye Institute University of Miami Miller School of Medicine

7:15 – 7:25 pm Q&As

7:25 – 7:30 pm Closing remarks James G. Fujimoto, PhD Joel S. Schuman, MD, FARVO Wolfgang Drexler, PhD

7:30 – 8:30 pm Networking Reception Ballroom C We are pleased to welcome Dr. Philip J. Rosenfeld as Opening Invited Speaker at ARVO - 2019 Imaging in the Eye Conference Friday, April 26, 2019 6:30 – 7:30 pm

Dr. Rosenfeld is currently Director and Professor of ophthalmology at the Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine. His primary clinical and research interests are in age-related macular degeneration (AMD). He is at the forefront of medical science having spent decades seeking effective therapies for AMD as the principal investigator and study chairman for numerous AMD clinical trials. He was instrumental in the clinical development of anti-VEGF therapies, the use of optical coherence tomography (OCT) guided treatment regimens using anti-VEGF therapy, the clinical development of OCT instruments, and the development and use of novel OCT algorithms for the diagnosis and management of macular diseases.

OCT in the Diagnosis and Management of Age-Related Macular Degeneration.

OCT is the gold-standard imaging technique for the diagnosis and management of both non-exudative and exudative age-related macular degeneration (AMD). The clinical convergence of OCT and vascular endothelial growth factor inhibitors (anti-VEGF therapy) led to a revolution in the treatment of neovascular and exudative ocular diseases. OCT-guided treatment with anti-VEGF therapy has become the global standard and has resulted in billions of dollars being saved by reducing the need for retreatment. OCT structural imaging combined with OCT angiographic imaging have eliminated the clinical need for dye-based angiography in managing AMD, while the use of novel algorithms has provided all the tools a clinician needs to diagnose and manage AMD. Most recently, swept-source OCT angiography has allowed us to study the choriocapillaris, a vital vascular layer that previously couldn’t be routinely imaged, yet is thought to play a pivotal role in the development and progression of AMD. 2019 Imaging in the Eye Conference Agenda April 26 – 27, 2019 Vancouver Convention Centre (East) Ballroom A, B, and C Conference Level East 1, 2 and 3 Meeting Level

Saturday, April 27, 2019

7:00 am – 2:00 pm Registration Ballroom A Foyer

7:00 am – 8:00 am Morning Coffee Meeting Room Foyer

8:00 – 8:15 am Opening remarks/Thank you to the James G. Fujimoto, PhD sponsors Joel S. Schuman, MD, FARVO Wolfgang Drexler, PhD

OCT Angiography Ballroom A & B Moderators: Yali Jia and Ruikang Wang

8:15 – 8:30 am Optical coherence tomography angiography Qinqin Zhang, R. Wang based capillary velocimetry on the

8:30 – 8:45 am Single-volume wide-field optical coherence Xiang Wei, T. Hormel, Y. Guo, S. tomographic angiography enabled by 400- Pi, Y. Jian, Y. Jia kHz swept source laser and bidirectional scanning protocol

8:45 – 9:00 am Reference-Based OCT Angiography Richard Rosen, J.S. Andrade Romo, Perfusion Density Mapping for Identifying M.V. Castanos Toral, D.B. Zhou, P. Acute and Chronic Changes in Eyes with Garcia, A. Barash, R. Weitz, T.Y. Chui Retinopathy over Time

9:00 – 9:15 am Ultrahigh-speed OCTA imaging of the Justin Migacz, R. Zawadzki, R. Jonnal, J. choriocapillaris around drusen at 1.6 MHz Werner 9:15 – 9:30 am 3D shape representation of OCT Jiong Zhang, Qiao, Yuchuan; Sharifi Angiography images Sarabi, mona; Khansari, Maziyar M.; Gahm, Jin K.; Kashani, Amir H.; Shi, Yonggang

9:30 – 9:45 am Automatic 3D Vessel analysis framework for Mona Sharifi Sarabi, Zhang, Jiong; Gahm, Optical Coherence Tomography Jin K.; Kashani, Amir; shi, Yonggang Angiography (OCTA)

9:45 – 10:00 am Rodent retinal circulation organization and Shaohua Pi, Wei, Xiang; Cepurna, location revealed by visible light OCT William; You, Qisheng; Huang, David; Morrison, John; Jia, Yali

10:00 – 10:30 AM - Morning break and Poster Presentations East Room 1, 2, and 3

Cellular and Molecular Imaging Ballroom A & B Moderators: Kostandinka Bizheva and Bin Yang

10:30 – 10:45 am Non-contact cell-detail real-time full-field Viacheslav Mazlin, Xiao, Peng; Scholler, OCT: capabilities and potential clinical Jules; Mecê, Pedro; Grieve, Kate; Irsch, applications of the novel anterior eye Kristina; Fink, Mathias1; Boccara, Claude. imaging tool.

10:45 – 11:00 am Label-free retinal cell imaging with dynamic Jules Scholler, Groux, Kassandra; Fink, full-field OCT. Mathias; Boccara, Claude; Grieve, Kate.

11:00 – 11:15 am Label-Free Imaging of Bipolar Cell Axons in Hyungsik Lim, Bucinca-Cupallari, Festa. Fresh Retina by Second-Harmonic Generation.

11:15 – 11:30 am Molecular Landscape of the Human Macula Kevin Schey; Anderson, David M.; by High-Resolution Imaging Mass McKinney, Kate H.; Messinger, Jeffrey D.; Spectrometry. Patterson, N. H.; Spraggins, Jeffrey M.; Curcio, Christine.

11:30 - 11:45 am Real-time measurement of lamina cribrosa Ian Sigal; Lee, PoYi; Brazile, Bryn; Lam, and sclera collagen architecture and Po; Zhu, Ziyi; Hua, Yi; Yang, Bin. mechanics at sub-micron resolution.

11:45 am – 12 Extended focus, 250 kHz SD-OCT for in-vivo Zohreh Hosseinaee; HAN, LE; Marchand, noon imaging of the cellular structure of the Paul J.; Bizheva, Kostadinka. human cornea. 12:00 – 12:15 pm Non-contact, laser-based confocal Michele Pascolini, Carraro, Federico; microscope for corneal imaging. Codogno, Nicola; Minozzi, Mattia; Pajaro, Ronnye; Tiso, Christian; Menin, Giulia; Pajaro, Simone; Tanassi, Cesare.

12:15 – 1:30 PM - Lunch break and Poster Presentations East Room 1, 2, and 3

New and Enhanced Technologies Ballroom A & B Moderators: Ian Sigal and Philip Rosenfeld

1:30 – 1:45 pm Towards a handheld and low-cost full-field Helge Sudkamp; Koch, Peter; Hillmann, OCT system for aberration corrected Dierck; Münst, Michael; vom Endt, imaging. Malte; von der Burchard, claus3; Roider, Johann; Birngruber, Reginald; Hüttmann, Gereon

1:45 – 2:00 pm 250 kHz, 1.6 µm axial resolution SD-OCT for Kostadinka Bizheva; Hosseinaee, Zohreh; in-vivo imaging of the human retina. HAN, LE

2:00 – 2:15 pm Spectral reflectivity of the ex vivo mouse Danielle Harper; Glösmann, Martin; retina revealed by hyperspectral confocal Gröger, Marion; Baumann, Bernhard microscopy

2:15 – 2:30 pm Waveform analysis and vessel type Leo Puyo; Paques, Michel; Fink, identification in the retina with laser Mathias; Sahel, Jose; Atlan, Michael Doppler holography

2:30 – 2:45 pm Adaptive optics imaging of retinal glia cells Zhoulin Liu; Agrawal, Anant; Saeedi, in patients with primary open angle Osamah; Hammer, Daniel. glaucoma

2:45 – 3:00 pm Axial eye length measurement through Muzammi Arain; Bello, Simon A.; coherence revival and extended imaging Kubach, Sophie; Straub, Jochen. depth using swept-source OCT

3:00 – 3:15 pm Functional retinal imaging with full-field Dierck Hillmann; Pfäffle, Clara; Spahr, swept-source OCT using enhanced Hendrik; Burhan, Sazan1; Kutzner, Lisa; processing algorithms Hilge, Felix1; Hüttmann, Gereon.

3:15 – 3:45 PM Afternoon break and Poster Presentations East Room 1, 2, and 3 Artificial Intelligence, Image Processing and Analysis Ballroom A & B Moderators: Hiroshi Ishikawa and Jennifer Lim

3:45 – 4:00 pm Real-time axial retinal motion tracking and Pedro Mecê; Mazlin, Viacheslav; correction for consistent high-resolution Scholler, Jules; Xiao, Peng; Sahel, Jose; retinal imaging with Full-Field Time-Domain Grieve, Kate; Fink, Mathias; Boccara, Optical Coherence Tomography (FFOCT). Claude.

4:00 – 4:15 pm Automated three-dimensional Hao Zhou; Chu, Zhongdi; Zhang, Qinqin; segmentation, visualization and Dai, Yining; Gregori, Giovanni; quantification of choroidal vasculature with Rosenfeld, Philip J.; Wang, Ruikang swept-source OCT.

4:15 – 4:30 pm Segmentation of inner microlayers of Amr Elsawy; Eleiwa, Taher K.; Raheem, abnormal cornea in Optical Coherence Mariam; Gregori, Giovanni; Abdel- Tomography images using graph Mottaleb, Mohamed; Abou Shousha, segmentation. Mohamed.

4:30 – 4:45 pm A universal three-dimensional registration Yuxuan Cheng; Zhang, Qinqin; Song, algorithm on OCT/OCTA for speckle Shaozhen; Chu, Zhongdi; Wang, Ruikang. reduction and visualization.

4:45 – 5:00 pm Automated volumetric segmentation of Yukun Guo; Xiong, Honglian; Hormel, retinal fluid on optical coherence Tristan; Wang, Jie; Hwang, Thomas1; Jia, tomography using deep learning. Yali.

5:00 – 5:15 pm Keeping it Clean: Artificial Intelligence Based Sripad Krishna Devalla; Pham, Tan Hung; Denoising Improves Segmentation of Zhang, Liang; Tin, Tun; Mohan, Rajan; Optical Coherence Tomography Images. Mohan, Sujatha; Aung, Tin; Schmetterer, Leopold; Thiéry, Alexandre H.; Girard, Michael.

Jennifer Lim; Bhaskaranand, Malavika; 5:15 – 5:30 pm Artificial Intelligence (AI) Screening for Ramachandra, Chaithanya; Bhat, Diabetic Retinopathy: Analysis from a Sandeep; Solanki, Kaushal; Sadda, Pivotal Multi-center Prospective Clinical Srinivas. Trial.

Poster Presentations - Posters will be on display from 8:00 am – 5:30 pm (below are the mandatory presentation times) 10:00 – 10:30 am; 12:15 – 1:30 pm; 3:15 – 3:45 pm East Room - 1, 2 and 3

New Technologies

Visible-light sensor less adaptive optics optical coherence tomography of Ryne Watterson; Wahl, Daniel; retinal response to laser exposure. Ju, Myeong Jin; Sarunic, Marinko V.

Transcranial versus transpupil illumination for fundus imaging. Timothy Weber; Mertz, Jerome

Line-field SD-OCT with 1.8 μm axial resolution and 2.5 kHz frame rate for Le Han; Hosseinaee, Zohreh; imaging the human cornea. Bizheva, Kostadinka.

High-throughput Retinal Whole Mount Imaging Using a Nikon STORM Cathryn Formichella; McGrady, Super-resolution Microscope. Nolan; Risner, Michael; Lambert, Wendi; Calkins, David.

A novel tool for the retinopathy of prematurity evaluation, associated Rodrigo Torres; Monteoliva, with a digital indirect ophthalmoscope hands-free device (DIO-HF). Guillermo A.; Saidman, Gabriela; Salvatelli, Adrián; Alazard, Gerónimo; Ortíz-Basso, Tomás; Ghersinich, Fernando; Ghersinich, Alberto.

Handheld and non-contact retinal camera for ROP monitoring: towards Guillem Carles; Preciado, Miguel; 80-degree retinal views. Zammit, Paul; Drysdale, Ross; Harvey, Andrew R.

Understanding the variability of handheld spectral-domain optical Kira Wang; Chen, Xi; Stinnett, coherence tomography measurements in supine infants. Sandra; Tai, Vincent; Tran-Viet, Du; Toth, Cynthia.

Visualizing Retinal Hemorrhage Thresholds for Q-Switched Nd: YAG Lasers Adam Boretsky; Wang, Heuy- in a Novel Porcine Model. Ching; Noojin, Gary; Elliot, William; Edsall, Peter; Shingledecker, Aurora; Rockwell, Benjamin.

Fundus imaging through lenticular media opacities using broad line Angelina Covita; Chen, Michael; fundus imaging. Leahy, Conor; Jung, Jesse J.; Lee, Scott.

Comparison of manual versus automated anchor point finding on Ashwini Tamhankar; Meng, montaging in ultra-widefield fundus imaging. Kevin; Manivannan, Niranchana; Makedonsky, Katherine; Durbin, Mary K.

Improving autofocus performance in a widefield fundus imaging system, Conor Leahy; Nolan, David; using peripheral defocus measurements. O'Hara, Keith E.; Chen, Michael; Brock, Keith; Hunder, Wilson G.; Straub, Jochen.

Repeatability of retinal curvature estimation on wide field OCT systems. Jonathan Bumstead; Steidle, Manuel; Leahy, Conor; Straub, Jochen.

Automatic pupil detection using off-axis iris images for alignment Poojan Dave; Wei, Andrew1; guidance in fundus cameras. Nolan, David; Stock, Simon; Guo, Jing; Covita, Angelina; Chen, Michael; Straub, Jochen; Durbin, Mary; Manivannan, Niranchana.

Enhanced Visualization of the Inner Retinal Layers Using Adaptive Optics Ryosuke Tamiya; Muraoka, Yuki; Optical Coherence Tomography. Kadomoto, Shin; Uji, Akihito; Nozato, Koji; Tsujikawa, Akitaka.

Meta-Analysis of Handheld Fundus Camera Validation Studies. Samantha D'Amico; Kim, Brian; Brady, Christopher J.

Moiré-pattern imaging-based IOP sensing contact lens in a Glaucoma Se-Hee Lee; Shin, Kyung-Sik; rabbit model. Cheong, Byung-Hak; Kang, Ji- Yoon; Kim, Jong-Ki.

Performance evaluation of ganglion cell analysis in normal and glaucoma Sophia Yu; Bagherinia, population using multi-retinal layer segmentation. Homayoun; Fard, Ali; Durbin, Mary; Knighton, Robert W.

Fully automatic localization of the optic disc using YOLO in colour fundus Yalin Zheng; hao, Yitian; Chen, X; photographs. Gao, Dongx; Bridge, Joshu; Zhu, Wenyu; Williams, Bryan

Quantity and Quality of Image Artifacts and Segmentation Errors in Ying Cui; Zhu, Ying3; Katz, Raviv; Diabetic Retinopathy using Wide Field Swept Source Optical Tomography Wang, Jay; Lu, Yifan; Kasetty, Angiography. Megan; Miller, John B.

OCT Angiography

Changes in Macular Retinal and Choriocapillaris Perfusion in Yining Dai; Zhang, Qinqin; Chu, without Retinopathy assessed by Swept-Source OCT Zhongdi; Zhou, Hao; Wang, Angiography. Ruikang.

OCTA flow signal enhancement by reducing residual structural signal. Tilman Schmoll; Bagherinia, Homayoun; Ren, Hugang.

Quantifying Peripapillary Microvascular Changes with Increasing Severity Zihan Sun; Tang, Fangyao; Tang, of Diabetic Retinopathy: An Optical Coherence Tomography Angiography Ziqi; Lam, Ka Ngai Alex; Wong, (OCTA) Study. Raymond; Lok, Jerry; Szeto, Simon; Ng, Siu Chun Danny; CHEUNG, Carol, Yim Lui.

Relationship between intraocular pressure and peripapillary vessel Grace Richter; Nelson, Andrew; density in treatment-naïve glaucoma patients from the African American Chu, Zhongdi; Chang, Ryuna1; Eye Disease Study. Burkemper, Bruce; Xu, Benjamin; Kashani, Amir; Varma, Rohit; Wang, Ruikang.

Preferred OCTA Scanning Protocol for Glaucoma Discrimination. Ravneet Rai; Lucy, Katie; Tracer, Nathaniel; Wu, Mengfei; de los Angeles Ramos Cadena, Maria; Kokroo, Aushim; Rathi, Siddarth; Madu, Assumpta; Jiménez- Román, Jesus; Lazcano-Gómez, Gabriel; Wong Shin, Joong; Kyung Rim, Sung; Ishikawa, Hirosh; Schuman, Joel S.; Wollstein, Gadi.

The superficial peripapillary vascular layer is the most informative for Danilo Andrade de Jesus; glaucoma diagnosis – A multi-layer study based on OCT Angiography. Sanchez Brea, Luisa; Barbosa Breda, João; Abegão Pinto, Luis; Stalmans, Ingeborg; Klein, Stefan; van Walsum, Theo.

Retinal Venous Occlusion Leads to Multiple Changes in Retinal Vessel Bill Morgan; Khoo, Ying; Rahman, Pulse Amplitude Maps. Anmar; Chandrakumar, Balaratnasingam; McAllister, Ian; Chen, Fred; Yu, Dao-Yi.

Quantification of choriocapillaris with OCTA: lessons learned. Zhongdi Chu; Gregori, Giovanni; Rosenfeld, Philip J.; Wang, Ruikang.

Automated detection of shadow artifacts in OCTA. Acner Camino; Jia, Yali; Wang, Jie; You, Qisheng; Wei, Xiang; Liu, Liang; Huang, David.

Variables Affecting Ocular Vessel Density Measurements. Maria de los Angeles Ramos Cadena; Wollstein, Gadi; Schuman, Joel S.; Lucy, Katie; Wu, Mengfei; Liu, Mengling; Rai, Ravneet S.; Jiménez-Román, Jesus; Lazcano-Gómez, Gabriel; Hernández-Monroy, Mariana; Wong Shin, Joong, 5; Kyung Rim, Sung; Ishikawa, Hiroshi.

Volumetric registration and averaging of OCTA data for enhanced image Luis De Sisternes; Lewis, Warren; quality. Callan, Thomas; Kubach, Sophie; Goldberg, Roger A.; Durbin, Mary.

Increased vessel length index in deep retinal layer angiography en face Warren Lewis; Bhattacharya, slabs via convolutional neural networks. Arindam; de Sisternes, Luis.

Analysis of Vascular Changes after Proton Irradiation using OCT-A in Gary Lamoureux; Wang, Jay; Patients with Uveal Melanoma. Lane, Anne Marie; Oxenreiter, Monica; Trofimov, Alexei V.; Miller, John B.; Shih, Helen A.; Aronow, Mary B.; Kim, Ivana K.; Gragoudas, Evangelos S.

Optical Coherence Tomography Angiography for Evaluation of Jodhbir Mehta; Devarajan, Kavya; Reperfusion following Pterygium Surgery. Ang, Marcus; Tan, Tien-En; Liu, Yu Chi.

Spatial correlation of microaneurysms detected by fluorescein Ayman Elnahry; Amsey, David J. angiography aligned with microdomains of macular ischemia delineated by optical coherence tomography angiography in patients with diabetic retinopathy.

Retinal Peripapillary Perfusion Changes in Thyroid-Related Orbitopathy Alexander Pinhas; Andrade Romo, Jorge with Asymmetric Proptosis without Compressive Optic Neuropathy S.; Lynch, Giselle; Zhou, Davis B.; Detected Using Optical Coherence Tomography Angiography. Castanos Toral, Maria V.; Tenzel, Phillip A.; Barash, Alexander; Della Rocca, David; Della Rocca, Robert; Chui, Toco Y.; Rosen, Richard B.; Reddy, Harsha S.

Microvascular reactivation in the glaucoma post-operative period. Ana Miguel; Herman, Aude; legeai, jérémy; Martin, Chloe; Silva, André.

Multi-acquisition averaging optical coherence tomography angiography Arman Athwal; Heisler, Morgan; Ju, for diabetic retinopathy. Myeong Jin; Karst, Sonja; Beg, Mirza Faisal; Navajas, Eduardo V.; Sarunic, Marinko V.

Hypertension Control Screen Using OCT-Angiography. Rebecca Zeng; Wang, Jay; Silverman, Rebecca; Diaz, Jose D.; Miller, John B.

Macular Vessel Density following Retinal Detachment using Swept Source Mohammad Dahrouj; Diaz, Danny; OCT-Angiography. Marmalidou, Anna; Katz, Raviv; Miller, John B.

Hybrid three-dimensional models reconstructed from swept-source OCT Tetsuju Sekiryu; Okamoto, Masahiro; angiography. Eifuku, Satoshi; Sugano, Yukinori.

Enhanced OCT angiography visualizations for choroidal neovascularization Jenwei Kuo; Huang, Wei Chieh; Reisman, with improved Bruch’s membrane segmentation. Charles A.

OCT angiography: Measurement of retinal macular microvasculature with Sami Hosari; Mardin, Christian; Spectral is II OCT angiography – reliability and reproducibility. Hohberger, Bettina.

Spiral Scanning OCT Angiography. Michael Niederleithner; Salas, Matthias; Leitgeb, Rainer A.; Drexler, Wolfgang; Schmoll, Tilman.

Imaging of vascular involvement in the intraocular tumor using OCT and Xiao Zhou; Zhou, Hao; Chu, Zhongdi; OCT angiography. Cheng, Yuxuan; Zhang, Qinqin; Wang, Ruikang.

Improved visualization of retinal vasculature using multi-layer Mary Durbin; Bagherinia, Homayoun; segmentation of optical coherence tomography angiography (OCTA) Fard, Ali; Santos, Torcato; Soares, Mário; images. Neves, Catarina; Lopes, Marta2; Cunha- Vaz, José.

Structure-function correlation in uveitic cystoid macular edema and Sophia Zagora; Grewal, Dilraj; Tomkins- treatment response using non-invasive Optical Coherence Tomography Netzer, Oren; Lightman, Sue. Angiography.

An algorithm to enhance OCT Angiography images. Ting Luo; Bagherinia, Homayoun; Fard, Ali.

Choroidal Changes after Photodynamic Therapy in Chronic Central Serous Ruwan Silva; Shields, Ryan Chorioretinopathy.

Analysis of Choroidal Neovascular Flow Lesions in Subretinal Sabia Handa; Arora, Atul; Agarwal, Hyperreflective Material using Optical Coherence Tomography Aniruddha; Gupta, Vishali. Angiography.

Longitudinal Follow-up of Tubercular Serpiginous-Like Choroiditis using Rohan B. Singh; Agarwal, Aniruddha; Optical Coherence Tomography Angiography. Aggarwal, Kanika; Grewal, Dilraj; Gupta, Vishali.

OCTA cube averaging for identification of vascular abnormalities in Thomas Callan; De Sisternes, Luis; Kubach, diabetic retinopathy using swept-source OCT. Sophie; Lewis, Warren; Wu, Charles; Durbin, Mary; Goldberg, Roger A.

Quantitative analysis of microvascular densities in retina and choroid Ying Zhu; Cui, Ying; Wang, Jay; Lu, Yifan; using wide-field swept source optical tomography angiography. Katz, Raviv; Miller, John B.

Optical Coherence Tomography Angiography of Acute-Onset Vogt- Grayson Armstrong; Smith, Meghan J.; Koyanagi-Harada Disease in a Young Female. Papaliodis, George N.; Miller, John B.

Optical Coherence Tomography Angiography: A useful tool for Atul Arora; Garwal, Aniruddha; Longitudinal Follow-up of Tubercular Serpiginous-Like Choroiditis. Aggarwal, Kanika; Mandadi, Spoorti K.; Grewal, Dilraj; Gupta, Vishali.

A Retrospective Study of Hypertension-induced Retinal Vascular Changes Yifan Lu; Zeng, Rebecca; Park, Jea Young; Using OCT-Angiography. Wang, Jay; Diaz, Jose Daniel; Miller, John B.

Comparison between multimodal imaging and OCT-A in a patient with Douglas, Vivian Paraskevi; Douglas, juxtapapillary choroidal neovascular membrane as a complication of optic Konstantinos; Cestari, Dean; Rizzo III, disc drusen. Joseph; Miller, John B.

Optical Coherence Tomography Angiography Imaging of Subretinal Carolyn Majcher; Trevino, Richard. Neovascularization in Macular Telangiectasia Type 2: A Case Report.

Artificial Intelligence

Improvement and validation of high precision ocular oximetry using a Damon DePaoli; Lapointe, Nicolas; convolutional neural network algorithm and a phantom eye. Tossou, Prudencio; Desroches, Joannie; Saugaveau, Patrick; Côté, Daniel C.; Sauvageau, Dominic.

Deep learning for automatic diabetic retinopathy detection under Mhd Hasan Sarhan; akedonsky, multiple image quality levels. Katherine; Mack, Meike; Durbin, Mary; Yigitsoy, Mehmet; Eslami, Abouzar.

Self-Segmentation Strategies for Unsupervised Clustering and Jeremy Benson; Nemeth, Sheila; Estrada, Visualization of Retinal Images. Trilce; Soliz, Peter.

The success rate of a vision screening program using a handheld fundus Katherine Makedonsky; Mack, Meike; camera and a deep learning image quality and diabetic retinopathy Durbin, Mary screening algorithm.

Abnormality prediction from fundus images using Deep Learning and Kunal Kumar Ramanbhai Patel; Freytag, large amounts of data. Alexander; Ranipa, Keyur; Spier, Nathalia; Urich, Alexander.

AI Quantification of OCTA en face image quality. Charles Wu

Deep learning algorithm to predict diabetic retinopathy (DR) progression Filippo Arcadu; Benmansour, Fethallah; on the individual patient level. Maunz, Andreas; Willis, Jeffrey; Prunotto, Marco; Haskova, Zdenka.

Image quality assessment of ultra-widefield fundus images using deep Sandipan Chakraborty; Ranipa, Keyur R.; convolutional neural networks. Makedonsky, Katherine; Sha, Patty; Chen, Michael; Brock, Keith; Durbin, Mary K.

Early Stage Glaucoma Diagnosis by Artificial Intelligence Assisted Subrata Batabyal; Kim, Sanghoon; Multifractal Functional OCT. Mustafi, Sourajit; Wright, Weldon; Mohanty, Samarendra.

Deep learning based robust fovea localization using OCT Angiography. Homayoun Bagherinia; Durbin, Mary

Automatic detection of optic nerve head in widefield OCT using deep Ali Fard; Agherinia, Homayoun learning.

Deep Learning-Based Method for Retinal Layer Segmentation in Optical Ivana Zadro; Lončarić, Sven; Radmilovic, Coherence Tomography Images. Marin; Vatavuk, Zoran

I am predicting laterality in external eye images using deep learning. Gary Lee; Allan, Thomas; Wu, Charles; Tamhankar, Ashwini; Covita, Angelina; Durbin, Mary.

Deep Neural Network Segmentation of Optical Coherence Tomography Julian Lo; Heisler, Morgan1; Lu, Angiography for Diabetic Retinopathy. Donghuan1; Karst, Sonja2; Vanzan, Vinicius2; Navajas, Eduardo V.2; Sarunic, Marinko V.

Method for Prototyping Convolutional Neural Networks with Fewer Paul Lee; Moinfar, Nader; Patients. Metzinger, Rebecca.

Eliminating retinal vessel shadows in en face choroidal OCT via generative Jianlong Yang; Zhang, Huihong; Zhou, adversarial networks. Kang; Li, Fei; Zheng, Ce; Hu, Yan; Zhang, Xiulan; Liu, Jiang.

Real-time Scene Understanding in Ophthalmic Anterior Segment OCT Hessam Roodaki; Grimm, Matthias; Images. Navab, Nassir; Eslami, Abouzar

Malavika Bhaskaranand; Lim, Jennifer I.; Artificial Intelligence Screening for Diabetic Retinopathy: Analysis from a Solanki, Kaushal; Ramachandra, Pivotal Multi-center Prospective Clinical Trial. Chaithanya; Bhat, Sandeep; Sadda, Srinivas.

Artificial intelligence-based automated segmentation of subretinal fluid and subretinal pigment epithelial fluid in patients with chronic central Mustafa Safi; Goldberg, Roger. serous chorioretinopathy (CSC).

Michael Chen; Durbin, Mary; Advantages and disadvantages of using deep learning-based image Manivannan, Niranchana; Brock, Keith; deconvolution on fundus images. Omlor, Lars.

Robust Nonperfusion Area Detection in Three Retinal Plexuses using Jie Wang; Hormel, Tristan; You, Qisheng; Convolutional Neural Network in OCT Angiography. Guo, Yukun; Wang, Xiaogang; Chen, Liu; Huang, David; Hwang, Thomas; Jia, Yali.

OCT image noise reduction using deep learning without additional priors. Arindam Bhattacharya; Durbin, Mary K.

A machine learning approach to predict response to anti-VEGF treatment Jayashree Nair Sahni; Maunz, Andreas; in patients with neovascular age-related macular degeneration using SD- Arcadu, Filippo; Zhang_Schaerer, Yan- OCT. Ping; Li, Yvonna; Albrecht, Thomas; Thalhammer, Andreas; Benmansour, Fethallah.

An Artificial Intelligence Deep Learning System for Discriminating Anran Ran; NGAI, Amanda, Kwan Yu; Ungradable Optical Coherence Tomography Three-Dimension Volumetric CHAN, Vivian, Wai Yin; Shi, Jian; THAM, Scans. Clement, Chee Yung; CHEUNG, Carol, Yim Lui.

Large Retinal Arterial Microaneurysm detection with Optical Coherence Konstantinos A.A. Douglas; Diaz, Jose Tomography-Angiography. Daniel; Oellers, Patrick; Miller, John B.

I have automated choroid segmentation of SD-OCT volumes using deep learning. Jonathan Oakley; Russakoff, Daniel B.

Comparison of manual and fully automatic cell hexagonality measure in Naomi Joseph; Kolluru, Chaitanya; corneal endothelium images in transplanted corneas post Descemet Menegay, Harry; Burke, Stephanie; Lass, Stripping Automated Endothelial Keratoplasty (DSAEK). Jonathan; Benetz, Beth Ann; Wilson, David.

Identification of clinically relevant glaucoma biomarkers on fundus Mohammad Norouzifard; Nemati, Ali; images using deep learning. Klette, Reinhard; Gholam Hossieni, Hamid; Nouri-Mahdavi, Kouros; Yousefi, Siamak.

Automated Detection of Retinal Fluid Using a Convolutional Neural Tristan Hormel; WANG, JIE; You, Network. Qisheng; Huang, David; Hwang, Thomas; Jia, Yali.

Clinical Imaging

Changes in Photoreceptor layer thickness in Optic Neuritis Follow up. Masoud Fard; Golizade, Alireza; Ghahvechian, Hossein; Yadegari, Samira; Ritch, Robert.

Diagnostic performance of corneal microlayer tomography in the Taher Eleiwa; Elsawy, Amr; diagnosis of Fuchs endothelial dystrophy. Roongpoovapatr, Vatookarn; Wen, Dan; Syed, Zeba1; Gameiro, Gustavo; Abou Shousha, Mohamed

Alterations of the foveal avascular zone area in glaucoma patients after Takuhei Shoji; Yoshikawa, Yuji; Kanno, glaucoma surgery. Junji; Ishii, Hirokazu; Ibuki, Hisashi; Shinoda, Kei.

Comparison of corneal ablation depth in small incision lenticule Jing Zhang extraction (SMILE) and femtosecond laser-assisted LASIK by FD-OCT.

CONGENITAL LEBER AMAUROSIS ASSOCIATED WITH MUTATION IN CBR1 Aristofanes Canamary Jr.; Louise, GENE. Fabiana; Valim, Mariana; Sallum, Juliana.

Choriocapillaris imaging in patients with Behcet′s disease using optical Yu Kawashima; Uji, Akihito; Miyata, coherence tomography angiography and image averaging. Manabu; Morooka, Satoshi; Muraoka, Yuki; Akagi, Tadamichi; Tsujikawa, Akitaka.

Multimodal imaging of experimental choroidal neovascularization. Xuan Liu; Inkernagel, Martin; Schwarzer, Petra; Kokona, Despina.

Spencer Cleland; Konda, Sri Meghana; Spectral Domain-Optical Coherence Tomography in Tracking Progression Danis, Ronald; Myers, Dawn; Trane, of Geographic Atrophy in Individuals with Late-Stage Age-Related Macular Ralph; Blodi, Barbara; Domalpally, Degeneration. Amitha.

Chemical cross-linking: a new choice for the bullous keratopathy Mengmeng Wang; Zhang, Minglian treatment.

Drusen Dynamics using Optical Coherence Tomography (OCT). Jeong Pak; Domalpally, Amitha; Huang, Yijun; Myers, Dawn; Blodi, Barbara.

Sub-clinical Diabetic Macular Edema in Chinese Diabetes Patients: A Pilot Xia Gong; Wang, Lanhua; Huang, Study. Wenyong; Li, Wangting.

Deeply pigmented clumps at fovea: A novel clinical find in Familial Chinmayi Vyas; Kadri, Dr. exudative vitreoretinopathy (FEVR). Venkatesh

The Effect of Digital Media in Age-Related Macular Degeneration: Bela Parekh; Parekh, Jai Improving Patient Awareness & Compliance.

Fernanda Mari Fujihara; Mello, Paulo Measurement of the Optic Nerve Head Descending Fibers at Bruch’s Augusto D.; Benfica, Camila Z; Castoldi, Membrane Opening Level with Spectral Domain Optical Coherence Nedio; Mendes, Fernanda M.; Tomography in Normal and Glaucoma Eyes. Finkelstein, Alessandro; Lindenmeyer, Rodrigo L.; Lavinsky, Daniel; Pakter, Helena M.; Lavinsky, Fabio.

Corneal Volume of the Normal Human Corneas calculated by Pentacam. Fernando Abib; Martins dos Santos, Renata.

Comparison of clinical outcome of anti- VEGF treatment in Diabetic macular edema combined with and without Epiretinal membranes. Mahmut Cankurtaran; Altintas, Aysegul

Personalized Pointwise Circumpapillary Retinal Nerve Fiber Layer Tobias Elze; Peschel, Thomas; Wang, Thickness (cpRNFLT) Norms. Mengyu; Li, Dian; Wirkner, Kerstin; Engel, Christoph Loeffler, Markus; Rauscher, Franziska G.

Safety and Efficacy of 1.5% Trypan Blue Assisted Vitreomacular Surgery. Subhan Tabba; Hooten, Claudia; Bourgeois, Keith; Davis, Garvin.

Corneal nerve structure in patients with primary Sjögren's syndrome in Fangting Li; Zhang, Qin; Ying, Xin; He, China. Jing; Jin, Yuebo; Cheng, Yaobin; Zhao, Mingwei.

Characterization of macular edema in the initial stages of diabetic Torcato Santos; Santos, Ana R.; Alves, retinopathy. Dalila; Marques, Inês; Lobo, Conceição; Cunha-Vaz, José.

Automatic quantification of focal capillary dropout for identification of Luis Mendes; Bhattacharya, Arindam; ischemia in patients with DR. Schwartz, Christian; Alves, Dalila; Santos, Torcato; Marques, Inês; Durbin, Mary; Cunha-Vaz, José

In vivo imaging of subretinal grafts of retinal progenitor cells derived from Xiufeng Zhong; Gao, Guanjie; HE, Liwen; human pluripotent stem cells by SD-OCT. Song, Xiaojing; Guan, Yuanyuan; Xie, Bingbing

Corneal Strain after UV-Riboflavin Cross-Linking Measured by Optical Sabine Kling; Khodadadi, Hossein Coherence Elastography.

Test-retest reliability of peripheral refraction measurements using a Katharina Foote; Leahy, Conor; widefield slit-scanning ophthalmoscope. Everett, Matt1; Straub, Jochen.

Predicting the likelihood of future keratoplasty from imaging corneal Siamak Yousefi; takahashi, hidenori2; parameters using manifold learning. Hayashi, Takahiko3; Tampo, Hironobu2; Inoda, Satoru2; Arai, Yusuke2; Asbell, Penny

Comparison of pterygium head quantification methodologies based on Olivia Lee; Maram, Jyotsna color photography of primary pterygia.

Temporal Retinal Nerve Fiber Bundle Changes in Glaucoma Imaged with Maria Castanos Toral; Zhou, Davis B.; OCT-Reflectance - Correlation with Circumpapillary OCT Retinal Nerve Jacobs, Erica B.; Andrade Romo, Jorge Fiber Layer Thickness. S.; Hood, Donald C; Ritch, Robert; Rosen, Richard B.; Chui, Toco Y.

Automated Quantification of Glaucomatous RNFL Reflectance Change Davis Zhou; Castanos Toral, Maria V.; using en-face OCT-Reflectance. Andrade Romo, Jorge S.; Eguia, Melvi; Jacobs, Erica B.; Hood, Donald C.; Ritch, Robert; Rosen, Richard B.; Chui, Toco Y.

Quantification of diabetic retinopathy-associated non-perfusion on ultra- Michael Aaberg; Patel, Tapan P.; wide-field fluorescein angiography and assessment of the impact of age, Iyengar, Rahul S.; Gilson, Marta M.; sex, race, and type of diabetes mellitus. Tran, Annie; Miranda, Caitlin; Young, Emma; Demetriou, Katarina; Devisetty, Laxmi; Musch, David C.; Paulus, Yannis M.

Morphology of trabeculectomy filtering blebs using anterior segment Rita Proença; Cunha, Joao Paulo; optical coherence tomography: a comparison of two methods. Ferreira, Joana; Santos, Arnaldo.

FINDINGS FROM THE FELLOW EYES OF LAMELLAR MACULAR HOLE: A Ismael Chehaibou; Manoharan, BILATERAL DEGENERATIVE CONDITION? Niranjan1; Govetto, Andrea3; Sarraf, David1; Hubschman, Jean-Pierre.

Optimizing the three-dimensional assessment of corneal lesions using Gibran Butt; Menduni, Francesco; Rauz, anterior segment optical coherence tomography. Saaeha; Wallace, Graham.

Scheimpflug imaging of posterior polar . Ravi Patel; Beddall, Daniel; Devabattula, Jemima; Virdee, Simrun; Nitiahpapand, Rynda; Balal, Shafi; Sharma, Anant.

High-resolution adaptive optics imaging analysis of Enhanced S-Cone Kiyoko Gocho; Kuniyoshi, Kazuki; syndrome with NR2E3 mutation. Hayashi, Takaaki; Katagiri, Satoshi; Kubota, Daiki; Takahashi, Hiroshi; Kameya, Shuhei.

Normative Corneal Optical Density Data Set: Creation and Analysis. Andrew Davis; Cheung, Albert

Adhered Posterior Hyaloid Influence on Glaucoma Structural Parameters Fabio Lavinsky; Fujihara, Fernanda Mari Evaluated with Spectral Domain Optical Coherence Tomography. F.; Lavinsky, Jaco; Castoldi, Nedio; Lindenmeyer, Rodrigo L.; Benfica, Camila Z.; Lavinsky, Daniel; Pakter, Helena M.; Mello, Paulo Augusto D.

High definition optical coherence tomography of the aqueous outflow Simon Bello; Straub, Jochen; Chen, system in glaucomatous and normal subjects. Michael.

Automatic analysis to detect the area of corneal nerve fibers on images with in vivo confocal microscopy. Koichiro Shinji

Association between optic disc parameters and refractive error. Dian Li; Elze, Tobias; Wang, Mengyu; Leutloff, Alexander; Kuckert, Carla; Wirkner, Kerstin; Engel, Christoph; Loeffler, Markus; Rauscher, Franziska G.

Application of New Imaging Technologies in High Myopic Eyes. Jochen Straub; Leahy, Conor; Foote, Katharina G.; Bagherinia, Homayoun.

Multifocal Electroretinographically Change of Laser-induced Choroidal Ming Mei Neovascularization Model in Non-human Primates.

Anterior Segment Optical Coherence Tomography imaging to study Sonal Dangda; Do, Anna; Mavrommatis, evolution of blebs after a new technique of ab interno collagen stent Maria; Panarelli, Joseph. implantation with conjunctival peritomy.

Monitoring the effect of topical steroid administration for multiple Taiichiro Chikama; Shinji, Koichiro; subepithelial corneal infiltrates following epidemic keratoconjunctivitis. Kadohiro, Yuko; Kiuchi, Yoshiaki

Kidney Function is Associated with Drusen Characteristics. Mengyu Wang; Ebert, Thomas; Tönjes, Anke; Wirkner, Kerstin; Engel, Christoph; Loeffler, Markus; Thiery, Joachim; Elze, Tobias; Rauscher, Franziska G.

OCT assessment of anterior chamber inflammation in children: a pilot Ameenat Lola Solebo; Pattani, study. Reshma; Akrabali, Saira; Rahi, Jugnoo; Edelsten, Clive.

Non-contact ultra-widefield imaging follow-up of patients with Abel Ramón; Romo-García, Efraín; Sital- retinopathy of prematurity treated with anti-VEGF, laser, scleral buckling Gastelum, Sergio; Paz-Camacho, Silvia; or cryotherapy using Optomap scanning laser. Meza-Anguiano, Alonso; Gutierrez-Ruiz, Gilberto Noe; Romero Talia.

Multispot Laser Therapy VS Conventional Laser and ultra-wide-field Talia Romero; Romo-García, fraín; Sital- photographs for Diabetic Retinopathy. Gastelum, Sergio; Paz-Camacho, Silvia; Meza-Anguiano, Alonso; Gutierrez-Ruiz, Gilberto Noe; Ramón, Abel.

MRI

Preoperative magnetic resonance imaging of superior oblique muscle: its Jingyuan Zhu; Jiang, Jingjing; Li, Ningdong role on surgical selection in children. I am measuring the Fluid Viscosity of Vitreous Body using MRI. Xingzheng Pan; Thakur, Sachin S.; Pontré, Beau; Kumarasinghe, Gamith L.; Rupenthal, Ilva D.; Donaldson, Paul; Vaghefi, Ehsan

Personalized geometrical models of the human eye developed from MRI Patrick Merz; Chacon-Caldera, Jorge; scans at 9.4 Tesla. Dörsam, Simon; Zirjacks, Patrick; Litau, Shanna; Wängler, Björn; Schad, Lothar R.; Auffarth, Gerd U.; Friedmann, Elfriede.

Magnetic resonance imaging of anti-VEGF drug diffusion in the human Simon Dörsam; Litau, Shanna; Merz, eye. Patrick R.; Wängler, Björn; Schad, Lothar R.; Auffarth, Gerd U.; Friedmann, Elfriede; Chacon-Caldera, Jorge.

Ultrasound

3D ultrasound biomicroscopy (3D-UBM) imaging and automated whole David Wilson; Wu, Hao; Helms, eye assessment of the iridocorneal angle. Richard2; Yu, Taocheng; Sevgi, Duriye D.2; Orge, Faruk; Minhaz, Ahmed T.

Scleral Strain Artefacts due to Spatiotemporal Distortion in High- Sunny Kwok; Sandwisch, Thomas; Resolution, High-Frame Rate Ultrasound Imaging. Clayson, Keyton1; Ma, Yanhui1; Liu, Jun.

Image Guided Surgery

Primary results of clinical use of the Beyeonics digitally enhanced surgical visualization system in and vitreoretinal surgery: a prospective Adiel Barak; Loewenstein, Anat1; study. Schneider, Ron; Bakalash, Sharon H.

Multimodal image-guide laser-induced retinal vein occlusion in living Yanxiu Li; Nguyen, Van Phuc; Zhang, Wei; rabbits. Xia, Xiaobo; Wang, Xueding; Paulus, Yannis M.

Yixin Yu; Zhang, Wei; Qin, Yu; Xie, Xinyi1; Photoacoustic signal-guided photo-mediated ultrasound therapy as a Yang, Xinmai; Wang, Xueding; Paulus, novel method to remove microvasculature. Yannis M.

Microscope integrated optical coherence tomography-guided autologous Ankur Singh; Dogra, Mohit; Tigari, full thickness neurosensory retinal autograft for large macular hole Basavraj; singh, Ramandeep. related total retinal Detachment.

Image processing and Image Analysis

Motion correction in 3D-OCT data by intensity-based image registration: Luisa Sanchez Brea; Andrade de Jesus, an evaluation study. Danilo; van Walsum, Theo; Klein, Stefan.

Grid deformation analysis of the macula and postoperative Ji Eun Lee; Park, Sun Ho; Lee, Jae Jung; metamorphopsia after macular hole surgery. Kwon, Han Jo; Park, Sung Who.

Automated segmentation of geographic atrophy using U-Net on custom- Niranchana Manivannan; de generated SD-OCT en face images. Sisternes, Luis; Gregori, Giovanni; Rosenfeld, Philip J.; Durbin, Mary.

Enhanced visualization of choroidal vasculature map using structural OCT. Areg Noshadi; Bagherinia, Homayoun; Callan, Tom; Durbin, Mary.

Vendor-neutral multimodal registration for en face OCT and fundus images. Reza Jafari; Reisman, Charles A.

Method for artificially degrading the signal from Optical Coherence Sophie Kubach; De Sisternes, Luis; Lewis, Tomography cube to assess variability in retinal pigment epithelial Warren; Gregori, Giovanni. detachment volumetric measurement.

Quantified fundus autofluorescence (QAF) imaging – the use of age- Thomas Ach; Kleefeldt, Nikolai; Pröbster, related standard and improved methods for analysis. Carla; Tarau, Ioana S.; Bermond, Katharina; Reichel, Clara; Sloan, Kenneth R.

Biomechanics

Morphological changes in Bruch’s membrane opening during intraocular Yanhui Ma; Kwok, Sunny; Clayson, pressure elevation. Keyton; Pan, Xueliang; Liu, Jun.

Shear strains in porcine and keratoconic human cornea. Keyton Clayson; Ma, Yanhui; Kwok, Sunny; Liu, Jun.

Reproducibility of Lamina Cribrosa Microstructure Measurements in Katie Lucy; Rai, Ravneet S.; Glidai, Yoav; Varying Intraocular and Intracranial Pressure Settings. Wu, Mengfei; Wang, Bo; Sigal, Ian A.; Smith, Matthew; Ishikawa, Hiroshi; Schuman, Joel S.; Wollstein, Gadi

Fluorescence

Designing a Hyperspectral Autofluorescence (AF) Camera for Early R. Theodore Smith; Dey, Neel; Ach, Detection of Age-related Macular Degeneration (AMD). Thomas; Curcio, Christine.

Near-infrared fluorescence detection and evaluation in an adaptive optics Tao Liu; Cornelissen, Steven; Dubra, scanning laser ophthalmoscope. Alfredo; Tam, Johnny.

Visualizing near-infrared autofluorescence from retinal pigment epithelial Kari Vienola; Zhang, Min; Sahel, Jose; cells in AMD using multi-wavelength excitation. Rossi, Ethan A.

Flavoprotein Fluorescence Changes in the Retina with Cataract Surgery. Jorge Andrade Romo; Castanos Toral, Maria V.; Zhou, Davis B.; Buxton, Douglas F.; Rosen, Richard

Basic Science

Aryl hydrocarbon receptor deficiency causes a diabetes-related vascular Meei-Ling Sheu pathology.

Protective Effects of Huoxue-Tongluo-Lishui-Fang (HTLF) on Retinal Minglian Zhang; Wang, Mengmeng Ischemia-Reperfusion Injury.

Tmem30a Deficiency in the retinal endothelial cells impairs cell Xianjun Zhu proliferation and angiogenesis.

Characterizing spatial distribution of neuronal cell types in visual areas of Kannan U V; Palaniswamy, Ramesh; mice brain imaged using Serial 2-photon tomography. Cain, Nicholas; Harris, Julie; Osten, Pavel.

5:30 pm Closing Remarks/Adjourn James G. Fujimoto, PhD Joel S. Schuman, MD, FARVO Wolfgang Drexler, PhD Program information

Statement of need The clinical community can benefit from information about the latest advances in ophthalmic imaging research. The field of imaging in the eye is rapidly changing with fundamental research advances, development of new technologies, imaging methods, clinical study and trial results. Imaging is impacting all sub-specialties in ophthalmology including glaucoma, retina, cornea and neuro-ophthalmology. New techniques are becoming available which promise to improve early detection and diagnosis, monitoring progression and treatment response, as well as improving understanding of pathogenesis and accelerating fundamental research. Target audience The educational design of this activity is intended to address the needs of basic researchers, engineers, clinician-scientists, ophthalmologists, ophthalmology residents, optometrists, PhD candidates, post- doctoral fellows, as well as medical students and fellows involved in ophthalmology or vision research. Educational objectives After completing this activity, participants should be to: • Discuss current biomedical ophthalmic imaging techniques • Describe the role of imaging in experimental and clinical ophthalmology • Critically read and understand literature involving ophthalmic imaging research Disclaimer The ARVO Imaging in the Eye Conference provides a forum for the open exchange and discussion of research results and scientific advancements in the field of ophthalmology; however, ARVO makes no representation or warranty as to the truth, originality, or accuracy of the information presented at the courses original materials distributed in connection with them. Nor are the views expressed by the individual speakers necessarily the views of ARVO. ARVO supports the ACCME’s policy on evidence- based content and encourages faculty to adhere to these standards when planning a presentation.

Participants have an implied responsibility to use the newly acquired information for scientific purposes or to enhance patient outcomes and their own professional development. The information presented in this activity is not meant to serve as a guideline for patient management. Any procedures, medications, or other courses of diagnosis or treatment discussed in this activity should not be used by clinicians without evaluation of patient conditions and possible contraindications on dangers in use, review of any applicable manufacturer’s product information, and comparison with recommendations of other authorities. Disclaimer off-label use This educational activity may contain discussion of published and/or investigational uses of agents that are not indicated by the FDA. ARVO does not recommend the use of any agent outside of the labeled indications. The opinions expressed in the educational activity are those of the faculty and do not necessarily represent the views of any organization associated with this activity. Please refer to the official prescribing information for each product for discussion of approved indications, contraindications and warnings. Audio and video recording

Recording by any means (photographing, audiotaping or videotaping) of any presentations or sessions at any ARVO Meeting is prohibited, except by an ARVO-authorized agent for official purposes. Violators risk confiscation of their equipment and/or dismissal from the meeting as deemed appropriate by ARVO.

Certificate of attendance Certificates of Attendance may be obtained by contacting ARVO Imaging Conference at [email protected].

Disclosure of conflicts of interest ARVO requires instructors, planners, managers and other individuals and their spouse/ partner who are in a position to control the content of this activity to disclose any real or apparent conflict(s) of interest they may have as related to the content of this activity. All identified conflicts of interest are thoroughly vetted by ARVO for fair balance, scientific objectivity of studies mentioned in the materials or used as the basis for content, and appropriateness of patient care recommendations. Disclosures will be displayed on a slide prior to each presentation. Conference organizing committee: James G. Fujimoto, PhD Massachusetts Institute of Technology Joel Schuman, MD NYU Langone Health, NYU School of Medicine Wolfgang Drexler, PhD Medical University Vienna Bruce Berkowitz, PhD, FARVO Wayne State University School of Medicine Christine Curcio, PhD, FARVO University of Alabama at Birmingham Alfredo Dubra, PhD Stanford University Sina Farsiu, PhD Duke University Brad Fortune, OD, PhD Devers Eye Institute Giovanni Gregori, PhD Bascom Palmer Eye Institute Joseph Izatt, PhD Duke University Yali Jia, PhD Oregon Health & Science University Jost Jonas, MD, FARVO Heidelberg University Fabrice Manns, PhD Bascom Palmer Eye Institute Richard Rosen, MD New York Eye & Ear Philip Rosenfeld, MD, PhD University of Miami Ronald Silverman, PhD Columbia University Medical Center Richard Spaide, MD Viterous Retinal Macula Consultants NY David Sarraf, MD Stein Eyes Institute, UCLA Susan Vitale, PhD, FARVO National Eye Institute/NIH Nadia Waheed, MD New England Eye Center/Tufts Medical Center Ruikang Wang, PhD University of Washington Yoshiaki Yasuno, PhD University of Tsukuba Linda Zangwill, PhD, FARVO University of California - San Diego Robert Zawadzki, PhD University of California Davis The Association for Research in Vision and Ophthalmology gratefully acknowledges our Imaging in the Eye Conference supporters:

Visit us: Booth 1037 Enabling OCT with Innovative Solutions

Spectrometers with high speed, resolution & sensitivity

Compact sample arm probes

Powerful image processing software & libraries

Custom drop-in OEM modules & subsystems

See how we are leading the Expedition Into the Eye

+1 919-544-7785 | [email protected]

See the Whole Picture with Triton™ Swept Source OCT

ARVO Booth #803

DRI OCT Triton™ Series A Multimodal Swept Source OCT

Full 360o image captured with posterior/anterior imaging capabilities of the Triton. Composite image made with 3rd party imaging software. ©2019 Topcon Medical Systems, Inc. MCA #3695 Session Abstracts Paper PDFs OCT Angiography CONTROL ID: 3195842 SESSION ABSTRACT START TIME: 8:15 AM SESSION ABSTRACT END TIME: 8:30 AM FINAL ID: 001

TITLE: Optical coherence tomography angiography based capillary velocimetry on human retina

FIRST AUTHOR: Qinqin Zhang

AUTHORS/INSTITUTIONS: Q. Zhang, R. Wang, Department of Bioengineering , University of Washington, Seattle, Washington, UNITED STATES|R. Wang, Department of Ophthalmology, University of Washington, Seattle, Washington, UNITED STATES| Purpose: To investigate the capillary velocimetry on human retina based on optical coherence tomography angiography (OCTA) scanning protocol (B-M mode) Methods: A 1060 nm swept source OCTA engine (PLEX® Elite 9000 (ZEISS, Dublin, CA)) running at 100 kHz A-line rate with motion tracking mechanism was utilized in our study. A new scanning protocol with 110 A-lines x 110 B- scans covering ~ 1mm x 1mm with 10 repetitions was designed and implemented on the device for velocimetry calculation. The frame rate is up to 500 frames per second with a distinguishable velocity from 0 mm/s to ~0.5mm/s according to the theorical analysis. Eigen decomposition (ED) based capillary velocimetry was used to extract the blood flow speed information of the capillary in human retina. Retinal and choroidal layer were segmented to investigate different pathologies, including the whole retinal layer extending from ILM to outer border of OPL for retinal diseases and the ORCC layer for choroidal neovascularization (CNV) cases. All the patients were imaged with a 3x3mm OCTA scan and then focused on the diseased region by using the velocimetry scanning protocol with a small field of view. Color mapping was used to display the speed variations of blood flow Results: Normal eyes, diabetic retinopathy (DR) eyes and CNV eyes were enrolled to evaluate the algorithm. Overall all eyes showed blood flow speed variations. In pathological eyes, abnormal blood flow speeds were observed compared to normal controls. A faster speed associated with the trunk of the CNV lesion and slower speeds associated with the lesion extremities was observed in CNV eyes. In DR patients, slower speeds were observed in capillary loops that associated with microaneurysms and as well as the suspended scattering particle in motion (SSPiM) Conclusions: A novel method for capillary velocimetry was proposed to visualize the relative blood flow speed in capillary level on human retina. The modified scanning protocol ensured a sufficient scanning speed based on a commercially available SSOCTA to differentiate the blood flow speed in a certain range, particularly in capillary level. This velocimetry method may play a role in not only studying the ocular disease but also in developing endpoints for therapeutic trials DR

CNV

Figure 1 The velocimetry results of pathological eyes. A and C: the 3x3mm OCTA scan of DR and CNV eyes, respectively; Band D: the corresponding blood flow speed color maps calculated by ED based capillary velocimetry in the red rectangle region of A and B. Red indicates fast blood flow; blue indicates slow blood flow. CONTROL ID: 3195512 SESSION ABSTRACT START TIME: 8:30 AM SESSION ABSTRACT END TIME: 8:45 AM FINAL ID: 002

TITLE: Single-volume wide-field optical coherence tomographic angiography enabled by 400-kHz swept source laser and bidirectional scanning protocol

FIRST AUTHOR: Xiang Wei

AUTHORS/INSTITUTIONS: X. Wei, T. Hormel, Y. Guo, S. Pi, Y. Jian, Y. Jia, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, UNITED STATES| Purpose: Imaging the peripheral retina using optical coherence tomographic angiography (OCTA) has generated serious interest in evaluating retinal vascular diseases, particularly in early stages of diabetic retinopathy. However, the field of view (FOV) of current OCTA is severely limited by A-line rates and inefficient scanning protocols with unnecessary fly-back A-lines. Here, we report a wide-field OCTA system that can acquire a 60-degree FOV in 6 seconds without resorting to montaging. Methods: The OCT system utilizes an AXSUN 400kHz dual swept source laser system with a center wavelength of 1060nm and 100nm bandwidth. The scanning lens system consists of two 2-inch 200mm and two 2-inch 100 mm achromatic doublets lenses. We designed the sample arm using computer-aided design (CAD) software and built it using 3D printed optical mounts. We achieve axial and transverse resolutions of 5.5 and 19 µm, respectively, with a total imaging depth of 4.2mm. The laser power on the pupil was set to be 2mW using a variable attenuator. A bidirectional scanning protocol with zero fly-back was applied during image acquisition to maximize the scanning efficiency (Fig. 1). Split-spectrum amplitude-decorrelation angiography (SSADA) was used to construct the OCTA volume. En face reflectance maps of the inner retina are generated by mean projection of structural OCT signals within the segmented inner retinal slab; en face angiograms of the inner retina are produced by mean decorrelation (flow) projection within the same slab. Results: The sensitivity of this OCT system was measured as 120dB. To test the system, we imaged the retina of seven healthy human eyes. Each scan volume contains 768 B-scans, and each B-scan contains 1536 A-lines, with two repeated B-scans acquired at each location to generate OCTA signals. We show OCT and OCTA images with a 60 degree of FOV (18 × 18 mm) can provide clinically acceptable image quality (Fig. 2). Details of the inner retinal microvasculature can be clearly appreciated by this one-shot single-volume wide-field angiogram. Due to the short scanning time, the OCTA image shows few motion artifacts even with such a large FOV. Conclusions: We successfully acquired a single-volume wide-field OCTA image with 60-degree FOV. We achieved a total scan time of 6 seconds using a 400-kHz swept source laser and an efficient bidirectional scanning protocol. Cell #1 B

_ 4 ::J i' re - 3 Q) • "O ::J .".:: 2 0. Cell#m E <{ l - Fast axis

0 - Slowaxis

o 500 iooo noo 2000 2500 3000 3500 A-line index

Figure 1. (A) Diagram of the bidirectional scan pattern; (B) voltage applied to the different scanning motors. Figure 2. (A) En face mean projection of 60-degree wide-field structural OCT of the inner retina in a healthy eye; (B) En face mean projection of 60-degree wide-field OCTA of the inner retina in the same eye. (A) and (B) were generated from the same scan volume. CONTROL ID: 3195602 SESSION ABSTRACT START TIME: 8:45 AM SESSION ABSTRACT END TIME: 9:00 AM FINAL ID: 003

TITLE: Reference-Based OCT Angiography Perfusion Density Mapping for Identifying Acute and Chronic Changes in Eyes with Retinopathy over Time

FIRST AUTHOR: Richard Rosen

AUTHORS/INSTITUTIONS: R. Rosen, J.S. Andrade Romo, M.V. Castanos Toral, D.B. Zhou, P. Garcia, A. Barash, R. Weitz, T.Y. Chui, Ophthalmology, New York Eye and Ear Infirmary of Mount Sinai Eye Faculty Practice, New York, New York, UNITED STATES|R. Rosen, M.V. Castanos Toral, D.B. Zhou, P. Garcia, A. Barash, T.Y. Chui, Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, New York, UNITED STATES| Purpose: Assessing non-perfusion and re-perfusion using perfusion density change over time can be challenging given the variability even within a single subject. Using a reference-based perfusion density change mapping approach we evaluated a variety of acute and chronic retinopathies. Methods: 28 patients (5 age-related macular degeneration-AMD, 10 diabetic retinopathy-DR, 9 retinal vein occlusion- RVO, 2 sickle cell retinopathy-SCR, and 2 radiation retinopathy-RR) and 20 controls were imaged using SDOCT (Optovue). Two 3x3mm parafoveal OCT-A scans were obtained for controls. Acute perfusion changes were assessed in 19 patients before and immediately after intravitreal injections (Fig. Top row). Chronic perfusion changes were assessed in 9 patients with follow up intervals ranging from 6 months to 4 years (Fig. bottom row). Subjects with signal strength index difference between each pair of OCT-As <10 were included for image processing. Image registration was performed for each pair of OCT-As using ImageJ. OCT-A image processing and perfusion density computation were performed using MATLAB as previously described ( ARVO 2018 E-Abstract 4944). Within subject variability of perfusion densities were computed between scans in all controls. Identification of non-perfusion and re-perfusion over time was performed in patients (Fig. Right column). Non-perfusion and re-perfusion area in patients were defined as difference in perfusion density between scans with lower than 0.1% or higher than 99.9% of the normal distribution based on the within-subject variability in controls, respectively. Results: In patients with acute perfusion changes, mean±SD of non-perfusion and re-perfusion area were 13.20±11.12% and 3.13±5.03%, respectively. Highest acute non-perfusion and re-perfusion area changes were found in patients with RR and RVO, respectively. In patients with chronic perfusion changes, mean±SD of non-perfusion and re-perfusion area were 4.39±6.92% and 1.77±2.38%, respectively. Highest chronic non-perfusion and re-perfusion area changes were found in patients with RVO and DR, respectively. Conclusions: OCT-A non-perfusion and re-perfused areas were identified in eyes with acute and chronic changes. We have demonstrated a reference-based mapping of non-perfusion and re-perfusion which allows qualitative and quantitative analysis of perfusion density change in OCT-A scans. Pre-lntravitreal Post-lntravitreal Perfusion Density Changes

Baseline 7months

0 flo•Perf1o1slon CONTROL ID: 3195920 SESSION ABSTRACT START TIME: 9:00 AM SESSION ABSTRACT END TIME: 9:15 AM FINAL ID: 004

TITLE: Ultrahigh-speed OCTA imaging of the choriocapillaris around drusen at 1.6 MHz

FIRST AUTHOR: Justin Migacz

AUTHORS/INSTITUTIONS: J. Migacz, Ophthalmology, New York Eye and Ear Infirmary, New York, New York, UNITED STATES|J. Migacz, R. Zawadzki, R. Jonnal, J. Werner, Ophthalmology, University of California Davis, Sacramento, California, UNITED STATES| Purpose: The choriocapillaris (CC) vasculature may play significant role in the progression of age-related macular degeneration (AMD). Our recently-developed ultrahigh-speed OCT angiography (OCTA) system enables the clear in- vivo visualization of CC vasculature throughout the whole macula. To gain a better understanding of the role of the CC, we imaged healthy and diseased subjects and characterized the appearance of the CC in areas around and far from drusen. Methods: We performed OCTA imaging on 13 eyes of 10 subjects with AMD. The average age was 68.6 ± 9.5 years. We also imaged 7 eyes of 7 young subjects (34 ± 15 years), and 7 eyes of 5 age-matched normals (74 ± 3.7 years). OCT imaging was performed with a custom swept-source OCT system operating at 1.64 MHz. Between 9 and 16 volumetric scans, lasting 2 seconds each, were performed on all subjects at locations ranging from the fovea to 12 degrees away. The field of view was either 1.2 or 1.5 mm. The Fourier-transform of each CC image was computed, and the dominant frequency was recorded as indicating the average vessel period in the image, a proxy for vessel density. Results: CC vessel spacing was slightly smaller (38.4 ± 3.6 μm) in normal than in those with AMD (39.5 ± 4.5 μm) and increased slightly with age (0.4 μm per decade). Fig. 1 shows the spatial frequency data as a function of age and foveal eccentricity, where the size of the bubble indicates the vessel density (#/mm). Fig. 2 shows the angiography of one normal (top two panels) and one representative AMD subject (remaining panels). CC vessels near the fovea of the normal indicate a large gap in the vasculature, which may be a so-called “flow void.” In the subject with AMD, an OCT B-scan of drusen is shown (left middle). The adjacent panel (right middle) of the OCT projection of the RPE layer indicates where drusen are present. Below (bottom right) the OCTA angiogram of the CC layer indicates that there are many areas of reduced flow (dropout) including in areas away from drusen. The corresponding reflectance OCT (left bottom) CC image is shown. There is some shadowing in this layer. Shadowing does not account for the reduced OCTA signal, so dropout areas are likely caused by lower flow. Conclusions: The ultrahigh speed system is effective for imaging the CC layer in subjects with drusen. Dropout exists where drusen are absent, suggesting that dropout may precede the appearance of drusen in these subjects.

CONTROL ID: 3195840 SESSION ABSTRACT START TIME: 9:15 AM SESSION ABSTRACT END TIME: 9:30 AM FINAL ID: 005

TITLE: 3D shape representation of OCT Angiography images

FIRST AUTHOR: Jiong Zhang

AUTHORS/INSTITUTIONS: J. Zhang, Y. Qiao, M. Sharifi Sarabi, M.M. Khansari, J.K. Gahm, Y. Shi, Laboratory of Neuro Imaging (LONI), Keck School of Medicine, University of Southern California, Los Angeles, California, UNITED STATES|J. Zhang, M.M. Khansari, A.H. Kashani, Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, California, UNITED STATES| Purpose: The studies of microvascular structures in 2D enface projection of 3D OCTA images recently show great promise in retinal image analysis field. However, direct analysis of 3D OCTA images which have rich depth-resolved microvascular information are rather limited due to low vessel visibility and strong projection artifacts. We aim to establish a 3D data representation of OCTA images via advanced shape modeling techniques to provide a high- quality microvascular visualization and to preserve 3D geometric and topological information for further analysis. Methods: We propose a novel framework including denoising, vessel enhancement, segmentation and triangular mesh representation to process 3D microvasculature in OCTA images. 3D curvelets denoising is employed to suppress speckle noise and preserve elongated structures for better continuity. The optimally oriented flux filter is applied to enhance vessels with best adaption to local scales. A binary vessel network is obtained by thresholding the enhanced OCTA images. Then, we take advantage of the established shape modeling technique to reconstruct a 3D vessel triangular mesh representation. A dataset of 200 repeated scans from 20 eyes was available for validation. Results: The proposed pipeline has been evaluated on the 3D OCTA repeated scans to verify its repeatability. Fig. 1 shows a typical example of 3D reconstructed vessel triangular meshes of a candidate scan and its overlapping with the template mesh surface. The average distance calculated from the 200 scans is 0.0056(mm), much smaller than the a voxel diagonal length 0.0174(mm). A high mean similarity ratio of 0.9324 has been obtained between scans of each subject. The high performance demonstrates an effective shape representation framework with strong repeatability. Conclusions: The proposed framework provides an intuitive 3D mesh representation of OCTA images. Geometrical and topological analysis on 3D vessel meshes can be further exploited for describing disease related pathology. Foveal eccentricity [degrees] ...... • • • •

YJcyc./mm .. • • 24 r;y£./mm • • • •• l85X,.c./mm $ • Norm•I • AMD ~ • • ...VI l1J IS Q) > ~ •• Q) tlO <( g • • • • • • • •

•• • • • • • (nun) I 0 09 0-08 0,07 0 06 0 OS 0_04 ()',03 ll'.02 (Jo.01 (a) Rc-gis:1crcd 1rlungular me:sh [b} Overl•y the candidate l)o representation of one candidate scan mesh with the template mesh Fig. 1 Example of similarity evaluation on 3D microvascular triangular mesh representation of repeated scans (RS) from the same subject. (a) The 30 triangular mesh representation of one registered candidate scan of a typical subject from the RS data.set. (b) The overlapping between the registered candidate (in yellow color) and the template (in red color) mesh surfaces. (c) The Haussdorf distance map between the mesh of the candidate scan and the mesh of the template scan is presented to show the perfect structure overlapping between two repeated scaru. Note that the repeated scan dataset was set up for the repeatability evaluation of our proposed framework. We collected 200 scans from 20 human eyes (with 10 scaru; for each eye) to validate the effectiveness and the repeatability of our method. Registration techniques are wed to align local structures of repeated scaru before validation. Prior to examination, written informed consents were obtained from subjects in accordance to the tenets of Declaration of Helsinki. CONTROL ID: 3195592 SESSION ABSTRACT START TIME: 9:30 AM SESSION ABSTRACT END TIME: 9:45 AM FINAL ID: 006

TITLE: Automatic 3D Vessel analysis framework for Optical Coherence Tomography Angiography (OCTA)

FIRST AUTHOR: mona Sharifi Sarabi

AUTHORS/INSTITUTIONS: M. Sharifi Sarabi, J. Zhang, J.K. Gahm, Y. shi, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of University of Southern California, Los Angeles, California, UNITED STATES|J. Zhang, A. Kashani, Department of Ophthalmology, Keck School of Medicine of University of Southern California, USC Roski Eye Institute, Los Angeles, California, UNITED STATES| Purpose: OCTA analysis typically involves 2D quantification based on the enface representation of retinal layers. 2D quantitative metrics can obscure the geometric and morphological information in 3D vasculature. There is a need for comprehensive 3D quantitative tools to more precisely model the complex dimensions of retinal vasculature available in OCTA scans. The aim of this study is to provide a robust and flexible 3D framework for the analysis of OCTA data. Methods: We propose an automated 3D framework for an end-to-end analysis of OCTA images. Initially, OCTA retina region extracted using OCT explorer software. In the second step, 3D Fast Discrete Curvelet Transforms was used as a denoising and enhancement tool. Optimally Oriented Flux (OOF) was applied in a hybrid setting with curvelet to generate a clear vessel map. Binary vessel segmentation of OCTA volumes were then obtained by thresholding on OOF response. Ultimately, a 3D vessel skeleton was calculated by incorporating the Hamilton-Jacobian method. The proposed method is very flexible with respect to the selection of OCT layers and is not limited to only superficial or deep retina layers. Moreover, this method provides 3D analysis and quantification of all retina vessels and small capillaries that has the potential to be valuable in early diagnoses of DR. Results: We validated our method on the OCTA images of a large-scale (n=338) DR study. Fig.1 demonstrates one representative eye selected from each group: Normal, Mild, Moderate, Severe, and PDR. For the quantitative assessment, 3D small vessel skeleton density (SVSD) of all eyes was extracted. The result of statistical analysis of healthy control compared to each stage of DR shows significant difference of SVSD in superficial and deep layers (p< 0.05). The test also reveals more significant difference (p< 0.001) in superficial layers for for earlier stage of DR (Mild), and our sublayer analysis localize the superficial layer alternation mainly in RNFL-GCL layer. Conclusions: In this work we provide a robust, flexible and automatic 3D framework for analysis of retinal vasculature available in OCTA scans. The proposed method was tested on a large-scale Diabetic Retinopathy dataset using small vessel skeleton density (SVSD) feature. The quantitative assessment shows significant difference of SVSD in superficial and deep layers of each stage of DR compared to healthy control. Fig.1. Qualitative results CONTROL ID: 3190134 SESSION ABSTRACT START TIME: 9:45 AM SESSION ABSTRACT END TIME: 10:00 AM FINAL ID: 007

TITLE: Rodent retinal circulation organization and location revealed by visible light OCT

FIRST AUTHOR: Shaohua Pi

AUTHORS/INSTITUTIONS: S. Pi, X. Wei, W. Cepurna, Q. You, D. Huang, J. Morrison, Y. Jia, Oregon Health and Science University, Portland, Oregon, UNITED STATES| Purpose: To investigate the retinal circulation organization in rats using visible-light optical coherence tomography (vis-OCT). Methods: Eight Brown Norway rat retinas were imaged with a 1.7-µm axial resolution, 50 kHz sampling rate, fiber- based vis-OCT system centered at 560 nm. Rats were anesthetized with 2.5 % isoflurane and 100% oxygen. Volumetric scans consisting of 512×3×512 A-lines covering a 2×2 mm2 area were acquired adjacent to the optic disc and in the periphery. OCT angiography was obtained by split-spectrum amplitude-decorrelation angiography (SSADA). Oxygen saturation in major retinal vessels was measured by automated spectroscopic retinal oximetry, taking advantage of the high absorption contrast of oxy- and deoxy- hemoglobin in the visible light band. Retinal layers (Fig. 1A) were segmented for projecting the en face images of the three vascular/capillary plexuses (SVP: superficial vascular plexus, ICP: intermediate capillary plexus, DCP: deep capillary plexus. Fig. 1B-D), as well as structures for the whole retina (Fig. 1E) and nerve fiber layer (NFL, Fig. 1G). Arteriovenous identification of all vessels were confirmed by measured oxygen saturation (Fig. 1E). Results: Oxygen saturation of 93% was used as a threshold to differentiate arteries from veins, based on prior observations. Arterioles were found predominantly in the SVP (Fig. 1B), whereas veins tended to drain blood from the DCP (Fig. 1D). Since inter-plexus capillaries are perpendicular to retinal layers and have greater absorption length along the light illumination direction, they appeared as dark spots on structural en face OCT (Fig. 1E), in contrast to neighboring capillaries. Inter-plexus capillaries were further validated by examining corresponding positions through the presence of capillaries in more than two plexuses in the overlaid en face angiograms of three vascular/capillary plexuses (Fig. 1F). The inter-plexus capillaries were found at capillaries end in SVP, as well as at some bifurcations,. Moreover, the capillary beds of the SVP were clearly visualized anterior to the nerve fiber bundles in the NFL slab en face image (Fig. 1G), with inter-plexus capillaries penetrating the NFL and connecting with the ICP and DCP. Conclusions: The detailed retinal circulation organization, including the spatial distribution, inter-plexus capillary identification, and the relative position to the nerve fiber layer can be revealed by vis-OCT. Figure 1 {A) D·•u" im11p of 11 brovm No,..,w-y nt retitf• h-y vhib,., lifht OCT. ll.M; itfncir limitin,i- mGmbi-en"'., Nf'l: "'°"""' fiber r.y..,-. Ga.. t;•n(lion ""'' l11yor., IPC.; innu plVi:tform leyet. INL: lnne..- nudear lctyEr, OPl: outer pleiltfonn laye..-, BM: Brudft memtnne. (B)-(0) En fttu Images of vatculer/c:eptllary pie.us.et. SVP: superflual vascular pfc.~•.u P1"1:1i1C1;.tcd in1bc HFL ~nil GCL d::i~. ICP: intennediCJh: c:apill~rv gfr:xu proie~d in the ~lab wnt~irin..l the inner bo1dcr of mL OCP: dc:iep wpillorv gfr:xv:s pri;ijC'l:.ted in the slob cont~INftl1he ou~r border of 11'1.. (E) En/oC(lmue of rtructurc prote~dfrom ILM to eM. ovult!d With me~s,urd M'!en seturotton (s02) vtfues In meJorveu:d~ ti) differentiate en art41f)' from a win in animal braetttn: 100% O,. lntt1r·P'-1ts (4pil!aries appur es dert 9pob due \(I f'•IW• ab'°tption then rif:lCttborini (4pihries and the1ane lebeled In the e11latged 11negaw1th whtte arrows. (f) overlaid en{oc:e angtograms of three vncular/.:eptllary ple•us.es to demonstrate detailed organlistlon of the retlnal drculatlon. hampks at lnter-p!Vi:us ceplllarle:t Uabeled

Room 1, 2 and 3 (Meeting Level) Cellular and Molecular Imaging CONTROL ID: 3195465 SESSION ABSTRACT START TIME: 10:30 AM SESSION ABSTRACT END TIME: 10:45 AM FINAL ID: 008

TITLE: Non-contact cell-detail real-time full-field OCT: capabilities and potential clinical applications of the novel anterior eye imaging tool

FIRST AUTHOR: Viacheslav Mazlin AUTHORS/INSTITUTIONS: V. Mazlin, J. Scholler, P. Mecê, M. Fink, C. Boccara, Institute Langevin, ESPCI, PSL Research University, CNRS, Paris, FRANCE|P. Xiao, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, CHINA|K. Grieve, K. Irsch, CHNO des Quinze Vingts, Institut de la Vision, INSERM-DHOS CIC 503, Sorbonne Universités, UPMC Univ Paris 06, CNRS, Paris, FRANCE| Purpose: We developed an improved time-domain full-field optical coherence tomography (FFOCT) system that can reliably in real-time acquire non-contact cellular resolution en face images of anterior human eye. We demonstrate instrument’s abilities to image not only the central corneal region, but also peripheral and limbal corneal zones, limbus with palisades of Vogt and vasculature. Potential applications of the new device are discussed. Methods: A customized combined FFOCT/Spectral-domain OCT (SDOCT) system was developed. In real-time it tracks the axial position of the eye and performs defocus correction, which results in instruments ability to consistently acquire and display FFOCT images in real-time. Even single FFOCT images in this new device show high signal-to- noise ratio, therefore eliminating the need for the post-processing steps, of image registration and image averaging. Fast acquisition speed (275 en face frames/sec.) was sufficient to acquire images clear from eye movements artifacts and visualize the blood flow propagation in the limbal region of the anterior eye. Results: Real-time FFOCT revealed structures in central, peripheral cornea and limbus. In cornea tear-film, superficial epithelial cells with nuclei, sub-basal nerve plexus, stromal keratocytes and nerves, endothelium with nuclei were visible. Images of limbus showed palisades of Vogt and blood vessels. Blood flow was directly visible and measured (0.3 mm/sec.). All the images had high lateral and axial resolutions (1.7 µm and 7.7 µm, respectively), had relatively large field-of-view of 1.25 mm2 and were acquired and displayed in real-time. Imaging procedure was non-contact and did not require introduction of medication into the eye. Conclusions: FFOCT can display in real-time images from corneal and limbal parts of the anterior eye. Blood flow in limbus and conjunctiva can be directly viewed and quantified. Contactless operation presents advantages over confocal microscopy in terms of comfort for the patient. Larger field of view, comparing to confocal microscopy, allows to perform more precise measurements of nerve and cell-densities, which may lead to more precise diagnosis and monitoring of the diseases, such as diabetes (linked to nerves density) and endothelial dystrophy (linked to endothelial cell density). In vivo human corneal and limbus images, captured with real-time FFOCT. CONTROL ID: 3194673 SESSION ABSTRACT START TIME: 10:45 AM SESSION ABSTRACT END TIME: 11:00 AM FINAL ID: 009

TITLE: Label free retinal cell imaging with dynamic full-field OCT

FIRST AUTHOR: Jules Scholler AUTHORS/INSTITUTIONS: J. Scholler, K. Groux, M. Fink, C. Boccara, Institut Langevin, ESPCI Paris, PSL Research University, Paris, FRANCE|K. Grieve, Quinze Vingt National Ophthalmology Hospital, Paris, FRANCE|K. Grieve, Vision Institute, Paris, FRANCE| Purpose: Microscopic imaging of 3D live tissue samples is important in disease modeling applications and development of new therapies, and non-invasive methods such as dynamic full-field OCT (D-FFOCT) hold promise for removing the need for complex and invasive fluorescent labeling. However in order for non-invasive methods to offer an effective alternative to fluorescent labeling, identification of specific cell types and behaviors is an essential step. Our purpose was to perform real time imaging with D-FFOCT of the local fluctuations in 3D samples, to use the fluctuations as features to classify cells, label-free, and to validate against ground truth data. Methods: We used D-FFOCT to measure subcellular organelle dynamics at up to 150Hz in living cells at 3D sub- micrometer resolution. We used a combination of i) ground truth fluorescent labeling and inhibitor drugs, imaged with simultaneous coincident fluorescence and real-time D-FFOCT acquisitions, to establish true cell identities and behaviors; ii) a machine learning approach to classify retinal cells using only the dynamic dimension of the signal, disregarding any morphological information. Results: We tested our methods on 3D primate retinal explants and human induced pluripotent stem cell (iPS)-derived retinal organoids at various stages of development. Comparison with fluorescence ground truth allowed us to identify live and dead cells, and use of inhibitors allowed discrimination of which subcellular organelles (e.g. mitochondria, vesicles) were responsible for the D-FFOCT signal. The machine learning framework was successful in classifying different retinal cell types with 95% accuracy. Conclusions: We demonstrated real-time quantitative D-FFOCT which allowed 3D reconstruction and time-lapse acquisitions without artifacts, and were able to validate our machine learning-based cell classification against ground- truth data. D-FFOCT with label-free cell classification offers the possibility to follow and control the evolution of the same sample, such as an in vitro retinal explant or a growing organoid for disease modeling or cell therapy applications, at different stages of its development. a) D-FFOCT image of a retinal organoid.
b) Retinal organoid with dead cells labelled : D-FFOCT image (left), overlay (right) of D-FFOCT and fluorescence (green) images.
c) First two images: segmentation on D-FFOCT images of ex vivo retina (ONL and INL), last two images: classification of cells. CONTROL ID: 3195764 SESSION ABSTRACT START TIME: 11:45 AM SESSION ABSTRACT END TIME: 12:00 PM FINAL ID: 013

TITLE: Label-Free Imaging of Bipolar Cell Axons in Fresh Retina by Second-Harmonic Generation

FIRST AUTHOR: Hyungsik Lim AUTHORS/INSTITUTIONS: H. Lim, F. BUcinca-Cupallari, CUNY, Hunter College, New York, New York, UNITED STATES| Purpose: The bipolar cells (BCs) are the primary neuron in the retina relaying visual information from the photoreceptors to the retinal ganglion cells (RGCs), and their axons comprise the inner plexiform layer (IPL) connecting the inner and outer retinas. Here a novel, label-free method is presented for visualization of the BC axons in the fresh retina ex vivo. Its utility is demonstrated for testing the persistence of BC axons in the course of glaucoma, despite the synaptic loss and dendritic atrophy of the RGCs known to occur in the process. Methods: Second-harmonic generation (SHG) microscopy was performed for imaging the BC axons in the fresh mouse retinas. A SHG setup was employed similarly as we have previously used for imaging the RGC axons. The fresh retinal flatmounts were prepared for imaging. Exploiting the broadband excitability of SHG, wavelengths of 800 and 1200 nm were compared. To confirm that the origin of non-RGC SHG signal was indeed the BC axons, transgenic Thy1-YFP mice expressing yellow fluorescent protein (YFP) in BCs were imaged by simultaneous two-photon excited fluorescence (TPEF) and SHG microscopy. To test a hypothesis relevant to glaucoma, i.e., that the BC axons persist during glaucomatous pathogenesis, DBA/2J mice were used as an experimental model of glaucoma and DBA/2J- Gpnmb+ as a non-glaucomatous control. Results: SHG signals arising from uniformly polarized microtubules (MTs) were acquired from the BC axons in the IPL as well as the RGC axons. The length of BC axons was approximately 30 µm. Due to the axial orientation, the BC axons did not respond much differently for two orthogonal polarizations of the excitation beam, unlike the RGC axons. The near-IR (NIR) excitation at 1200 nm for SHG imaging was safer to the retina and the quality of SHG images obtained at 1200 nm was comparable to or exceeded those at 800 nm. The co-localization of SHG signal with Thy1- YFP showed the spatial relationship of the BC axons with other cellular features in the IPL. Finally, SHG images of the retinas of DBA/2J mice (>12 months old) showed the BC axons persistent in regions where the RGC axons were lost. Conclusions: SHG microscopy is a novel label-free technique for visualizing the BC axons, by which the roles in pathology and visual processing can be investigated. RNFL

IPL

INL OPL CONTROL ID: 3195879 SESSION ABSTRACT START TIME: 11:00 AM SESSION ABSTRACT END TIME: 11:15 AM FINAL ID: 010

TITLE: Molecular Landscape of the Human Macula by High Resolution Imaging Mass Spectrometry

FIRST AUTHOR: Kevin Schey AUTHORS/INSTITUTIONS: K. Schey, D.M. Anderson, K.H. McKinney, N.H. Patterson, J.M. Spraggins, Biochemistry, Vanderbilt University, Nashville, Tennessee, UNITED STATES|J.D. Messinger, C. Curcio, University of Alabama- Birmingham, Birmingham, Alabama, UNITED STATES| Purpose: Imaging mass spectrometry (IMS) is a powerful tool for elucidating spatial distributions of biomolecules ranging from lipids to metabolites to proteins. With low micrometer spatial resolution, molecular distributions can be distinguished between cell and synaptic layers of human retina. The purpose of this work is to map the distribution of lipids and molecular pigments of normal and age-related macular degeneration (AMD) human macula. Methods: Paraformaldehyde-preserved eyes from human donors 80-93 years of age were analyzed in both macula and peripheral regions. Adjacent serial sections were stained with Periodic acid-Schiff and Hematoxylin stains for morphological analysis. IMS was performed at 5-15 µm spatial resolution in positive and negative ion modes on a Bruker Solarix 9.4T FTICR instrument with a modified MALDI source designed for high spatial resolution imaging experiments. Tissue autofluorescence images were also acquired from sections, before and after IMS experiments, to enable precise registration of IMS and optical signals, allowing high-confidence localization of IMS signals to individual layers and extracellular deposits. Results: Specific lipid signals observed in IMS images correlate with tissue regions, i.e. the fovea and tissue layers, i. e., retinal pigment epithelium (RPE), photoreceptor cells, and Henle fiber layer. High spatial resolution IMS combined with registration to autofluorescence and H&E images, enabled definitive localization of signals to the layers of the RPE cell bodies and apical processes as well as vertically aligned compartments of photoreceptor cells. An abundant signal observed at (m/z 799.676) was identified as PE-Cer-NMe2(42:1) and in a normal eye, localized to RPE apical processes/ outer segments below the fovea and extending out to the periphery. This lipid is also observed with very high abundance in a basal laminar deposit present in a 93 year old eye with AMD. Detection of distinct lipid signals present in drusen and basal laminar deposits were also observed. Signals attributed to lutein/zeaxanthin were detected in the foveal center. Molecular profiles of macular RPE cells differed from peripheral RPE cells. IMS allowed mapping of specific lipid and molecular pigment signals in retinal regions and layers. Unique molecular signatures were detected for specific retina cell layers and extracellular deposits. (No Image Selected)

Conclusions: CONTROL ID: 3195838 SESSION ABSTRACT START TIME: 11:15 AM SESSION ABSTRACT END TIME: 11:30 AM FINAL ID: 011

TITLE: Real-time measurement of lamina cribrosa and sclera collagen architecture and mechanics at sub-micron resolution

FIRST AUTHOR: Ian A Sigal AUTHORS/INSTITUTIONS: I.A. Sigal, P. Lee, B. Brazile, P. Lam, Z. Zhu, Y. Hua, B. Yang, Ophthalmology, University of Pittsburgh, Pittsburgh, Pennsylvania, UNITED STATES|I.A. Sigal, P. Lee, Z. Zhu, Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, UNITED STATES| Purpose: The tools available to visualize and measure eye tissue architecture and mechanics, such as polarized light microscopy, optical coherence tomography and second harmonic imaging, often lack the necessary temporal and/or spatial resolution to study crucial dynamic events. We demonstrate a new technique, snapshot polarized light microscopy (sPLM), for imaging ocular connective tissue microarchitecture and mechanics in real-time Methods: Fresh sheep optic nerve heads were trephined and cut into 16-µm thick sections. The sections were mounted on a custom biaxial-stretching device for dynamic testing while imaged at 30 frames per second using sPLM. Spectral analysis at each pixel revealed the local collagen fiber orientation, which was then used to quantify tissue architecture and load-induced changes in anisotropy. Image tracking techniques were used to determine collagen fiber displacements and deformations Results: The high spatial and temporal resolution of sPLM revealed load-induced pore deformations and beam thinning in the lamina cribrosa (Fig. 1a). Load straightened the crimped collagen fibers in the sclera (Fig. 2c), such that their fiber orientations changed from bimodal to unimodal (Fig. 2d). Tracking revealed complex deformation patterns in the sclera when under load, including significant shear, stretch, and compression (Fig. 2b), with local tissue deformations reaching 60% (Fig. 2c) Conclusions: We demonstrated that sPLM allows visualization and quantification of eye tissue architecture. The high temporal and spatial resolution of sPLM revealed sub-bundle details of tissue mechanics previously unreported. These imaging and analysis techniques can be applied in other parts of the eye and other collagenous tissue Example sPLM images. Pixel color represents local collagen fiber orientation, whereas brightness is proportional to collagen density or presence of pigment. a) Optic nerve head. b) Lamina cribrosa deformations. c) Effect of load in the sclera. d) fiber uncrimping Sclera a) before and b) after loading. Example of complex resultant c) displacements and d) stretch CONTROL ID: 3195883 SESSION ABSTRACT START TIME: 11:30 AM SESSION ABSTRACT END TIME: 11:45 AM FINAL ID: 012

TITLE: Extended focus, 250 kHz SD-OCT for in-vivo imaging of the cellular structure of the human cornea

FIRST AUTHOR: Zohreh Hosseinaee AUTHORS/INSTITUTIONS: Z. Hosseinaee, K. Bizheva, System Design Engineering, University of Waterloo, Waterloo, Ontario, CANADA|Z. Hosseinaee, L. HAN, K. Bizheva, Physics and Astronomy, University of Waterloo, Waterloo, Ontario, CANADA|P.J. Marchand, Electrical engineering, University of Polytechnique Montreal, Montreal, Quebec, CANADA| Purpose: To develop a high speed, extended-focus SD-OCT system with ~2 µm isotropic resolution sustained over ~500 µm depth-of-focus, for in-vivo, non-contact, cellular resolution imaging of the human Methods: A free-space SD-OCT system that utilizes Bessel illumination of the sample and Gaussian detection, was built in order to achieve an extended depth of focus while preserving high lateral resolution in biological tissue. The system is based on a Mach-Zender interferometer and powered by a femtosecond laser with emission spectrum centered at ~790 nm and spectral bandwidth of ~140 nm (measured at the detection end of the OCT system). The system’s axial resolution was measured to be ~2.2 µm in free space. An axicon lens with apex angle in combination with a 10x microscope objective was used to achieve a lateral resolution of ~ 2 µm over an extended depth of focus of ~ 500 µm in biological tissue. At the detection end of the system, the light was coupled into a single mode fiber, connected to a high-resolution spectrometer (Cobra, Wasatch Photonics). The spectrometer was interfaced with a linear array CMOS camera with tall pixel design, 2048 pixels, and 250 kHz data acquisition rate. The performance of the extended-focus SD-OCT system was tested by imaging healthy human cornea in-vivo. Results: The axial OCT resolution was measured to be ~ 2.2 µm in free space, corresponding to ~ 1.6 µm in biological tissue assuming a refractive index of 1.38. The lateral resolution was evaluated at various depths along the extended focus by imaging microspheres imbedded in weakly scattering agarose. The measurements showed that lateral resolution of ~2 µm is sustained over a depth-of-focus > 500 µm. Volumetric images of the anterior cornea, acquired in-vivo from healthy subjects with the extended focus SD-OCT system showed clearly the cellular structure of the corneal epithelium, sub-basal and stromal corneal nerves, as well as the collagen structure and keratocytes in the anterior stroma. Conclusions: The extended-focus UHR-OCT system developed in this study can be utilized for the diagnostics and monitoring the treatment of various corneal pathologies. (No Image Selected) CONTROL ID: 3193515 SESSION ABSTRACT START TIME: 12:00 PM SESSION ABSTRACT END TIME: 12:15 PM FINAL ID: 014

TITLE: Non-contact, laser-based confocal microscope for corneal imaging

FIRST AUTHOR: Michele Pascolini AUTHORS/INSTITUTIONS: M. Pascolini, F. Carraro, N. Codogno, M. Minozzi, R. Pajaro, C. Tiso, G. Menin, S. Pajaro, C. Tanassi, NIDEK Technologies Srl, Albignasego, ITALY| Purpose: Confocal microscopy is an effective technique for the direct observation of corneal pathologies. In this work a non-contact laser confocal microscope (CS-5) is presented, whose images are enhanced by an image fusion technique to obtain sharp details and a uniform field of view for any layer from epithelium to endothelium. Methods: CS-5 takes 1-Mpixel images at 18 fps with a Field of View of 400x400 µm2 at a distance of 12 mm from the patient’s corneal apex. Provided that in such conditions corneal images can be acquired only imposing a high confocality to carefully suppress aberrations and reflexes, the design is based on two micrometric apertures and a 2D scanning head. A red laser is used as a source and an Avalanche Photo-Diode (APD) as a detector. The confocality is so narrow that some details might fall out of focus in a given frame. Sequences of images are thus recorded by means of a micrometric z-scan and combined in stacks of about 5 images for endothelium, stroma and epithelium, respectively. After a pre-processing to reduce the noise, the possible motion is compensated by frame registration. Each resulting image is successively filtered with a moving-average spatial filter. The stack of registered images is processed with a majority filter to produce a depth map containing, for each pixel, the index of the slice having the best focus at that point. Finally, a composite image is created from the stack by selecting pixels as indicated by the depth map. Results: We tested the CS-5 on healthy patients. Acquired images show the nuclei of the endothelial cells, posterior and anterior stroma, sub-basal nerve plexus, basal cells and tear film. Raw images present good resolution but are crispy and lack the depth of focus. We evaluated the image fusion on stacks of 5-6 images of the epithelium, stroma and endothelium of enucleated pig eyes. Composite images showed that the processing algorithm enhances the overall focus and the anatomical details. Conclusions: The CS-5 is a novel corneal confocal microscope that operates in non-contact conditions minimizing the invasiveness of the examination. Image quality is comparable with contact microscopes whilst the limited depth of focus has been extended by means of image fusion, applying image registration and a majority filter. Corneal layers from the endothelium to the epithelium on healthy patients Above: sequence of 8 pig endothelium images. Below: depth map and image fusion result Poster Presentations & Lunch break Room 1, 2 and 3 (Meeting Level) New and Enhanced Technologies CONTROL ID: 3195880 SESSION ABSTRACT START TIME: 1:30 PM SESSION ABSTRACT END TIME: 1:45 PM FINAL ID: 015

TITLE: Towards a handheld and low-cost full-field OCT system for aberration corrected imaging

FIRST AUTHOR: Helge Martin Sudkamp AUTHORS/INSTITUTIONS: H.M. Sudkamp, P. Koch, M. Münst, M. vom Endt, R. Birngruber, G. Hüttmann, Medizinisches Laserzentrum Lübeck GmbH, Lübeck, GERMANY|H.M. Sudkamp, D. Hillmann, R. Birngruber, G. Hüttmann, Institut für Biomedizinische Optik, Universität zu Lübeck, GERMANY|C. von der Burchard, J. Roider, Augenklinik Kiel, UKSH, GERMANY| Purpose: Reliably imaging the retinal cone mosaic requires the correction of aberrations caused by imperfections of the eye. This correction is possible by adaptive optics, which is technically challenging and requires complex and expensive devices. Recent progress on numerical approaches for correcting aberrations promises to aberration correction without adaptive optics. These algorithms, however, require the phase information of the OCT-signal not to be disturbed by motion of the sample and, thus, high acquisition rates, which cannot be retrieved with current commercially available OCT systems. Here, we use a compact and potentially low cost full-field OCT approach, for numerically aberration corrected imaging. Methods: We imaged the human retina using off-axis full-field time-domain (OA-FF-TD) OCT. The system used consists of only a few relatively inexpensive optical components and allows to build a handheld OCT device capable of imaging the human retina in vivo. Images were recorded from healthy volunteers. An iterative computational algorithm was applied to the data to determine and correct for low order aberrations like defocus and astigmatism as well as higher order aberrations. Results: We acquired volumetric images at a field of view of 1.6 mm x 0.7 mm x 1.4 mm from various retinal regions of healthy subjects. We were able to correct a defocus of -5 dpt numerically, which renders ametropia correction unnecessary (Fig. 1). We also corrected higher order aberrations and were able to resolve the cone mosaic above an eccentricity of 3° temporal to the fovea (Fig. 2). Conclusions: OA-FF-TD-OCT is a promising new technology, for a simple and robust retina scanner system. In contrast to commercially available OCT systems, it allows for aberration free imaging of the human retina at large pupil sizes and thus is able to resolve single cells of the cone mosaic. Our technology allows to build compact devices, which could be used for home care. Area close to the optic nerve head before (left) and after numerical ametropia correction. The field of view is 1,6 mm x 1,4 mm Cone mosaic 8° temporal of to the fovea. The field of view is 5° x 2° CONTROL ID: 3195908 SESSION ABSTRACT START TIME: 1:45 PM SESSION ABSTRACT END TIME: 2:00 PM FINAL ID: 016

TITLE: 250 kHz, 1.6 µm axial resolution SD-OCT for in-vivo imaging of the human retina

FIRST AUTHOR: Kostadinka Bizheva AUTHORS/INSTITUTIONS: K. Bizheva, Z. Hosseinaee, L. HAN, Physics and Astronomy, University of Waterloo, Waterloo, Ontario, CANADA|K. Bizheva, Z. Hosseinaee, Systems Design engineering, University of Waterloo, Waterloo, Ontario, CANADA| Purpose: To develop a high speed, high resolutoion SD-OCT system for in-vivo volumetric imaging of the structure and blood vasculature of the human retina. Methods: A compact, fiberoptic SD-OCT system that conbines high axial resolution with rapid image acquisition rate, was build for in-vivo volumetric imaging of the human retina. The system is powered by a femtosecond laser (Integral, Femntolasers) with emission spectrum centered at ~790 nm and spectral bandwidth of ~140 nm (measured at the detection end of the OCT system). The retinal imaging probe consisted of 3 achromat lenses (f = 10 mm, f = f = 60 1 2 3 mm) and a pair of galvanometric scanners (6210, Cambridge Technologies). Both a 25 mm water cell and a pair of BK7 prisms were used to compensate for dispersion mismatch within the OCT system and the eye. The detection end of the SD-OCT system was comprised of a customized, high resolution spectrometer (Cobra, Wasatch Photonics), interfaced with a linear array CMOS camera (OCTOPLUS CL, e2v, Teledyne Dalsa). The camera has a tall pixel design, 2048 pixels and 250 kHz data acquisition rate. A high speed framegrabber (Teledyne Dalsa) was used to acquired the raw data. Custom Matlab-based software was used to generate imnages from the raw data and compensate for eye motion induced image artefacts in the retina OCT images. Results: The axial OCT resolution was measured to be ~ 2.2 µm in free space, corresponding to ~ 1.6 µm in retinal tissue assuming an average refractive index of 1.38. The system’s sensitivity was ~98 dB, measured at ~ 100 µm away from the zero delay line for 850 µW incident optical power. SNR roll-off was ~10 dB over a scanning range of ~ 1.4 mm. Volumetric images of the human retina, acquired in-vivo from healthy subjects, show the detailed structure of all retinal layers. Conclusions: We have developed a SD-OCT system for in-vivo human retinal imaging that combines high axial resolution with rapid image acquisition rate. (No Image Selected) CONTROL ID: 3176418 SESSION ABSTRACT START TIME: 2:00 PM SESSION ABSTRACT END TIME: 2:15 PM FINAL ID: 017

TITLE: Spectral reflectivity of the ex vivo mouse retina revealed by hyperspectral confocal microscopy

FIRST AUTHOR: Danielle J. Harper AUTHORS/INSTITUTIONS: D.J. Harper, B. Baumann, Medical University of Vienna, Vienna, AUSTRIA|M. Glösmann, Core Facility for Research and Technology, University of Veterinary Medicine Vienna, AUSTRIA|M. Gröger, Core Facility Imaging, Medical University of Vienna, AUSTRIA| Purpose: To investigate the reflectivity of the ex vivo C57BL/6 mouse retinal layers as a function of wavelength. Methods: The cornea and lens of a C57BL/6 mouse were removed from the eye and radial cuts were made to flatten the eyecup. The neural retina was lifted off from the retinal pigment epithelium (RPE), which was still attached to the underlying choroid and sclera, and both tissues were whole mounted in 70% glycerol and coverslipped. Both neural retina samples and RPE/choroid samples were imaged using a white light laser confocal microscope (Leica TCS SP8) in reflection mode with a 20x multi immersion objective. The samples were illuminated consecutively with light from 470 – 670 nm in 10 nm intervals throughout the whole sample at 5 μm intervals in depth. The hyperspectral spectra were then analyzed pixel by pixel in order to be able to characterize the wavelength dependent spectra in each retinal layer. Each image was color-corrected to account for the scattering/absorption which took place in the previous layers, using the dataset above as a reference. For visualization purposes (Fig. 1), the data were combined into an RGB image, with R = 610 – 670 nm, G = 540 - 600 nm and B = 470 - 530 nm. Results: The retinal nerve fiber layer/ganglion cell layer (RNFL/GCL) shows a heightened reflectivity in the shorter wavelengths. From the inner plexiform layer to the outer nuclear layer, the neural mouse retina displays a relatively constant reflectivity profile across the investigated wavelength range. In the RPE, the hexagonal-shaped cell walls also do not show a wavelength dependent reflectivity. The RPE melanin, however, has a very low reflectivity in the shorter wavelengths, resulting in a brownish appearance in the color representation (Fig. 1G). Conclusions: The RNFL and the melanin in the RPE are the two regions in the ex vivo mouse retina which show largely wavelength dependent reflectivity. Since in vivo imaging methods are often later correlated to histology, this could give an insight into the accuracy of some in vivo spectroscopic imaging techniques (e.g. spectroscopic optical coherence tomography), and allow for corrections to be made in order to reconstruct the true in vivo reflectivity profiles. .--s .__F~ED

_J Figure 1. A) Reconstructed depth projection of images obtained by confocal microscopy with positions of (B-H) indicated. Orange line indicates end of neural retina sample and beginning of RPE/choroid sample. B-H) Hyperspectral confocal images. B) Retinal nerve fiber layer/Ganglion cell layer. C) Inner plexiform layer. D) Inner nuclear layer. E) Outer plexiform layer. F) Outer nuclear layer. G-H) Retinal pigment epithelium melanin particles (G) and cell walls (H). Scale bars = 50 µm (B-H) CONTROL ID: 3176533 SESSION ABSTRACT START TIME: 2:15 PM SESSION ABSTRACT END TIME: 2:30 PM FINAL ID: 018

TITLE: Waveform analysis and vessel type identification in the retina with laser Doppler holography

FIRST AUTHOR: Leo Puyo AUTHORS/INSTITUTIONS: L. Puyo, M. Fink, M. Atlan, Institut Langevin, Paris, FRANCE|M. Paques, J. Sahel, Centre d'investigation clinique des Quinze-Vingts, Paris, FRANCE|M. Paques, J. Sahel, Institut de la Vision, Paris, FRANCE| Purpose: Laser Doppler holography (LDH) is a full-field blood flow imaging technique that was recently used in the human eye. The power Doppler signal measured in LDH involves several contributions such as blood flow but also ocular movements comprising fundus pulsations. We here investigate these different contributions through data processing in order to extract the retinal blood flow contribution. Methods: This study was done in accordance with the Declaration of Helsinki and all subjects gave informed consent. We analyzed the reconstructed power Doppler waveforms using different Doppler frequency ranges. We used low frequency shifts to reveal slow ocular movements and high frequency shifts to reveal flow in vessels. Additionally, we transposed for LDH some metrics that were developed for functional Doppler ultrasound such as resistivity index that measures systolodiastolic variations in blood vessels. Results: The segmentation of the power Doppler spectral density revealed that for frequency shifts from 1 to 4 kHz, the power Doppler signal is dominated by ocular fundus pulsations, while the frequency shifts for 10 to 37 kHz reveal the cardiac cycle waveform. The high frequency power Doppler signal is strongly correlated between all regions of the image, but subtracting the spatially averaged value allows to reveal waveforms typical of the probed structure (i.e. arteries or veins). Finally, we also found that calculating resistivity maps from these waveforms, we were able to propose a clear arteriovenous differentiation in the retina. Conclusions: We have shown that the power Doppler signal as measured in LDH can be decomposed to measure the contributions of fundus pulsation and blood flow in retinal capillaries and large vessels. Moreover, the distinct waveforms of blood flow in arteries and veins allows for a robust differentiation of these vessels. Power Doppler throughout cardiac cycles measured in ROI (B) shown in Fig. 2. (a) Low frequency shifts (1-4 kHz). (b) High Frequency shifts (10-37 kHz). Power Doppler measurements for high frequency shifts (10-37 kHz). (a) Power Doppler throughout cardiac cycle in a vein (V), an artery (A) and the background (B). (b) Associated power Doppler image with the ROIs drawn. (c) Power Doppler in the 3 same ROIs when subtracting average signal. (d) Coefficient of variation of the power Doppler leading to an arteriovenous differentiation. CONTROL ID: 3195895 SESSION ABSTRACT START TIME: 2:30 PM SESSION ABSTRACT END TIME: 2:45 PM FINAL ID: 019

TITLE: Adaptive optics imaging of retinal glia cells in patients with primary open angle glaucoma

FIRST AUTHOR: Zhuolin Liu AUTHORS/INSTITUTIONS: Z. Liu, A. Agrawal, D. Hammer, US Food and Drug Administration, Silver Spring, Maryland, UNITED STATES|O. Saeedi, University of Maryland Medical Center, Maryland, UNITED STATES|Purpose: Early treatment is essential to manage glaucoma disease progression which manifests as damage to ganglion cell (GC) somata and axons. Activated microglia appear early in the glaucomatous process, suggesting that retinal glia cells may be a sensitive indicator of early glaucoma. Most knowledge about glia cell (i.e., microglia and astrocyte) function has come from animal studies, but how this knowledge applies to human retina remains elusive. In this study, we used adaptive optics - optical coherence tomography (AO-OCT) to investigate glia cell density changes in glaucoma. Methods: Two eyes in two glaucoma subjects (P1: advanced glaucoma, 54 yrs and P2: mild glaucoma, 57 yrs) with hemifield defect and one age matched control eye (C1: control, 49 yrs) were imaged with the FDA multimodal AO imager. In the glaucoma subjects, three locations were imaged with the AO focus set to the inner retina: one above and one below the raphe in the approximate location of maximum and minimum retinal thickness as determined from

clinical2) OCT collected prior to AO imaging (within 3-4° of the fovea); and a third location on the raphe at 12° temporal. In the healthy control subject, AO-OCT volumes were acquired from the fovea to 12° temporal retina at an interval of 1.5°. Up to 30 volumes (lateral dimension: 1.5×1.5°) were collected and registered in three dimensions. Glia cells present at the inner limiting membrane were counted from the en face projections at all imaged locations. Results: There was a large variation in the glia cell density distribution among the three subjects imaged in this study. In C1, there were no glia cells within 7.5° from the fovea, and the cell density increased from 17 cells/mm2 at 9° to 46 cells/mm2 at 12°. The results agree with previous observation. In P1, glia cells were not evident in any of the three locations examined. In P2, more glia cells were found at the location where the retina is relatively healthy (75 cells/mm

2 2 than the inferior area affected by glaucoma (35 cells/mm ) and at 12° (52 cells/mm ).

Conclusions: The results indicate that the number of glia cells may depend on retinal location and disease stage. How glia density differs between glaucoma and healthy subjects may predict retinal health, given that the main function of those cells is to respond to pathological changes and keep the retina free of cell debris and remnants of dying cells. (No Image Selected) CONTROL ID: 3195611 SESSION ABSTRACT START TIME: 2:45 PM SESSION ABSTRACT END TIME: 3:00 PM FINAL ID: 020

TITLE: Axial eye length measurement through coherence revival and extended imaging depth using swept-source OCT

FIRST AUTHOR: Muzammi A Arain AUTHORS/INSTITUTIONS: M.A. Arain, S.A. Bello, S. Kubach, J. Straub, Carl Zeiss Meditc, Inc., Dublin, California, UNITED STATES| Purpose: Measuring and tracking eye length during regular eye exams is highly desirable, especially due to the increase in myopic eyes around the world. Here we report on using the coherence revival (CR) property of swept- source external cavity tunable laser (SS-ECTL) and extended depth imaging to measure axial eye length. Methods: A prototype PLEX® Elite 9000 (ZEISS, Dublin, CA) is used to image a human subject with 6 mm axial depth and 16 mm lateral field-of-view. The system uses a dual mode (100/200 kHz) SS-ECTL (Axsun Technologies, Billerica, MA). Due to the CR property of the source, we obtain ghost images of any object at a distance which is an integer multiple of the cavity round-trip length (L ). Fig. 1 (a) shows the conceptual idea of CR based eye length c measurement where the CR image of cornea will show up at different locations based upon relationship L = (L - e c (corneal offset + retinal offset))/n where n is the refractive index of eye and L is the geometrical eye length. We e aligned the subject to ensure that the CR of corneal reflex is within the imaging window of the system. For comparison, axial length was also measured using a commercial system IOL Master 700 (Zeiss, Jena, Germany). To increase the range of eye lengths that can be imaged for axial eye length measurement, we modified the data acquisition system to capture a 32 mm (including mirror image) axial depth image. We used a custom built human retina mimicking test eye to combine extended depth measurement capability with the CR based axial length measurement. Results: Fig. 1(b) shows 6 mm axial depth OCT scan of a human subject showing corneal reflex in a retinal scan. Knowing the corneal offset, retinal offset, and cavity length, the eye length is calculated to be 24.35 mm compared to 23.94 measured on the IOL Master 700. The small offset could be due to an approximation used in our method regarding refractive index. Fig. 2 shows a 32 mm axial depth image of a test eye where the axial length is measured to be 26.1 mm compared to 25.8 mm from data sheet. Conclusions: We have shown the feasibility of measuring axial eye length using CR feature of SS-ECTL. We have expanded the idea with extended depth imaging to include all possible eye lengths. The proposed method will enable axial eye length measurement during a normal OCT scan. /~-~------r...... ·

Le Le < n.Le ~3

t Le= n.Le t ll.D . + LC > n.Le

8 10 12 14 16 FOV(mm) (a) {b) Fig. 1: Axial length estimation using OCT.(a) Conceptual diagram for axial length measurement using CR in SS-ECTL. Corneal surface images can show up above, aligned, or below the retina depending on whether the geometrical eye length multiplied by the refractive index (n.L) is less, equal to, or greater than the cavity round-trip length (Le)· (b) B-scan of a human subject showing corneal surface and retina in the same B-scan with 6 mm axial depth and using the relationship Le=(Lc­ (corneal offset + retinal ojfset))/n 0

5

£ 15 c.. Q) '"C CCI ·- 20 ~

25

30

0 5 10 15 FOV (mm)

Fig. 2: 32 mm axial depth scan of test eye showing full 32 mm axial depth including the mirror image. CONTROL ID: 3189530 SESSION ABSTRACT START TIME: 3:00 PM SESSION ABSTRACT END TIME: 3:15 PM FINAL ID: 021

TITLE: Functional retinal imaging with full-field swept-source OCT using enhanced processing algorithms

FIRST AUTHOR: Dierck Hillmann AUTHORS/INSTITUTIONS: D. Hillmann, C. Pfäffle, H. Spahr, S. Burhan, L. Kutzner, F. Hilge, G. Hüttmann, Institute of Biomedical Optics, University of Lübeck, Lübeck, GERMANY|D. Hillmann, Thorlabs GmbH, Lübeck, GERMANY|C. Pfäffle, H. Spahr, L. Kutzner, F. Hilge, G. Hüttmann, Medical Laser Center Lübeck GmbH, Lübeck, GERMANY| Purpose: Full-field swept-source optical coherence tomography (FF-SS-OCT) acquires 3D images of the living human retina carrying meaningful phase information on the backscattered light. Here, we show that these phases can be used to enhance imaging by providing additional contrast, e.g., for imaging the function of photoreceptors and ganglion cells. We present an algorithm to extract functional data even in adverse conditions when noise and statistical sample fluctuations impede a direct phase evaluation. Methods: For FF-SS-OCT imaging we used a high speed area camera (60,000 frames/s) which acquired typically 512 interferograms during a wavelength sweep from which we reconstructed volumetric OCT data. This imaging is equivalent to an A-scan rates of about 40 MHz, largely exceeding the speed of most conventional OCT systems. We acquired series with 70 volumes during which we stimulated the retina with white light to provoke responses from photoreceptors and ganglion cells. After data reconstruction and residual motion correction, we computationally corrected for ocular aberrations if needed. Finally, the extended Knox-Thompson (KT) method, an algorithm that originated in astronomic speckle interferometry to image through the turbulent atmosphere, was adapted to achieve functional contrast. Results: The applied processing technique to visualize retinal function showed clear signals even in demanding situations, where fluctuating scatterers or uncertainties in image registration affect the imaging; situations, in which phase signals degraded over time. The KT algorithm had the biggest impact on functional ganglion cell imaging for which it significantly increased the signal-to-noise ratio. Even in image series, where standard phase differences hardly extracted meaningful functional contrast, robust signals were obtained. Conclusions: Combining phase stable data acquisition of FF-SS-OCT with suitable processing techniques is a powerful tool for structural and functional imaging in the living retina. Despite the considerably lower signal-to-noise ratio compared to conventional OCT, our phase processing techniques can extract functional signals from layers which currently cannot be imaged by scanning OCT. The combination of phase stable imaging with advanced algorithms thus brings us closer to functional imaging of the entire neural retina on a cellular level. (No Image Selected) Poster Presentations & Afternoon break Room 1, 2 and 3 (Meeting Level) Artificial Intelligence, Image Processing and Analysis CONTROL ID: 3194512 SESSION ABSTRACT START TIME: 3:45 PM SESSION ABSTRACT END TIME: 4:00 PM FINAL ID: 022

TITLE: Real-time axial retinal motion tracking and correction for consistent high-resolution retinal imaging with Full- Field Time-Domain Optical Coherence Tomography (FFOCT)

FIRST AUTHOR: Pedro Mecê AUTHORS/INSTITUTIONS: P. Mecê, V. Mazlin, J. Scholler, M. Fink, C. Boccara, Institute Langevin, Paris, FRANCE| J. Sahel, K. Grieve, Vision Institute, Quinze-Vingts Ophthalmology Hospital, Paris, FRANCE|J. Sahel, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, UNITED STATES|P. Xiao, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, CHINA| Purpose: We previously presented the very first full-field OCT (FFOCT) images of in vivo human retina. To obtain these images, it was necessary to equalize the optical path difference (OPD) of the reference arm and of the retinal layer of interest. However, as the retina moves constantly in the axial direction due to pulsation and fixation, this is a challenging task and several images have to be acquired to increase the chances of a matched OPD. In this work, we implement a real-time axial retinal motion tracker in the FFOCT, which we use to cancel the OPD and consistently obtain diffraction-limited high-resolution en-face retinal images without using adaptive optics. Methods: A customized FFOCT system was developed and combined with a spectral-domain OCT (SDOCT) system. The SDOCT B-scan is used to estimate the retina axial position using centroid algorithm. A fast voice-coil motor is installed in the reference arm giving the precise position of the reference mirror. For each iteration, the OPD is computed and a new command is sent to the voice coil to equalize the OPD. With OPD-locked, FFOCT images were acquired at 350Hz with a high-speed camera for various healthy subjects. Image stacks acquired were registered and averaged to improve signal to noise ratio (SNR). Results: The real-time OPD-locking system presents an axial range of 2.5 mm, a loop rate of 25Hz and a precision of 5µm rms. These specifications are more than sufficient to lock the OPD during image acquisition, as the FFOCT axial resolution is 8µm. In this way, we were able to consistently image the inner/outer segment (IS/OS) junction at the foveal and perifoveal regions for multiple subjects. Figure 1 shows images of the photoreceptor mosaic resolved without using adaptive optics for a 5mm diameter pupil, where the signal spatial variation is mainly due to the alignment of the subject. Conclusions: We have successfully demonstrated retinal axial motion tracking and correction enabling consistent high-resolution FFOCT retinal imaging without adaptive optics. In this way, the photoreceptor mosaic was resolved for a 5mm diameter pupil for multiple subjects. This paves the way for a straightforward implementation of a compact FFOCT ultrahigh resolution system with a transmissive adaptive lens for low order aberration correction in order to increase SNR in clinical studies. Examples of resolved photoreceptors As it comes close to the fovea, photoreceptors are mosaic for a 5mm diameter pupil no longer resolved {here 5mm diameter pupil) CONTROL ID: 3195881 SESSION ABSTRACT START TIME: 4:00 PM SESSION ABSTRACT END TIME: 4:15 PM FINAL ID: 023

TITLE: Automated three-dimensional segmentation, visualization and quantification of choroidal vasculature with swept-source OCT

FIRST AUTHOR: Hao Zhou AUTHORS/INSTITUTIONS: H. Zhou, Z. Chu, Q. Zhang, Y. Dai, R. Wang, Bioengineering, University of Washington, Seattle, Washington, UNITED STATES|G. Gregori, P.J. Rosenfeld, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, UNITED STATES| Purpose: Automated methods for assessing choroidal vasculature in the whole 3D scanning volume are surprisingly limited. This work aims to develop an automated method that accurately segments choroidal vessels from 3D OCT scans, without the necessity of OCT angiography. We propose a depth-resolved mapping for choroid vessels and 3D metrics for choroidal vasculature quantification: choroid vessel volume (CVV), choroid stroma volume (CSV), choroidal vessel volume density (VVD) and choroidal stroma-to-vessel volume ratio (SVR). Methods: SS-OCT (PlexElite, Carl Zeiss Meditec Inc.) was used to collect 12x12mm macular scans from subjects. The steps for automated segmentation of choroid slab include: 1) attenuation correction; 2) excluding optic disc; 3) graph search for Bruch’s membrane and the outer border of choroidal vessels. The 3D choroidal vessels were segmented on each B-scan of the segmented slab. From the binarized 3D choroidal vessels, en face sum projections were generated with color map indicating the depth of the vessel from Bruch’s membrane and montaged to large field- of-view. Metrics of mean choroid thickness (MCT), CVV, CSV, VVD and SVR were investigated in 3D (Fig. 1). Results: 144 normal subjects were recruited. The automatic choroid segmentations assisted by attenuation correction were validated with manual segmentations and showed good agreement on all metrics (all P<0.0001). Depth-resolved mapping of segmented 3D choroidal vessels presented vessels at different depth with different color in ultra-large field- of-view (15x20mm) (Fig.2). CVV and CSV showed significant correlations with choroid thickness (all P<0.0001). VVD and SVR were constant with small variations among all subjects (68.9±1.3% and 0.45±0.03). All choroid-related metrics were significantly correlated with age (all P<0.0001) except VVD and SVR. Conclusions: The proposed automated choroid assessment on thickness and vasculature was successful in 3D OCT scans, which will be clinically useful in quantitative assessment of a myriad of ocular diseases involving the choroid such as AMD, central serous chorioretinopathy, polypoidal choroidal vasculopathy and pathologic myopia. Fig. 1. Flow chart for automated 3D segmentation and visualization of choroidal vasculature. Fig. 2. Depth-resolved mapping of choroidal vasculature (15x20mm ultra-large field-of-view). CONTROL ID: 3195620 SESSION ABSTRACT START TIME: 4:15 PM SESSION ABSTRACT END TIME: 4:30 PM FINAL ID: 024

TITLE: Segmentation of inner microlayers of abnormal cornea in Optical Coherence Tomography images using graph segmentation

FIRST AUTHOR: Amr Elsawy AUTHORS/INSTITUTIONS: A. Elsawy, M. Abdel-Mottaleb, M. Abou Shousha, Electrical and Computer Engineering, University of Miami, Coral Gables, Florida, UNITED STATES|A. Elsawy, T.K. Eleiwa, M. Raheem, G. Gregori, M. Abou Shousha, Ophthalmology, Bascom Palmer Eye Institute, Miami, Florida, UNITED STATES| Purpose: To report on a graph-based automated segmentation method to segment the epithelium, Bowmans (BW), Stroma (ST) and Descemets (DM) microlayers of abnormal cornea using high definition optical coherence tomography (HD-OCT) images. Methods: Fifteen eyes were imaged using HD-OCT (Envisu R2210, Bioptigen, Buffalo Grove, IL, USA). These eyes had different diagnoses: Dry eye (2 eyes), Keratoconus (5 eyes), Fuchs Dystrophy (3 eyes), and controls (5 eyes). The 15 images were manually segmented by two expert operators. The automated segmentation algorithm segmented the Epithelium (EP) and the Endothelium (EN) microlayers and used them to segment the inner microlayers. A flat-EP image was obtained, and its vertical gradient was computed. A directed graph was used to segment the BW and ST as the two hyper-reflective microlayers after the EP. A flat-EN image was obtained and its vertical gradient was computed. A directed graph was used to segment the DM as the hyper-reflective microlayer before the EN. In both graphs, the edge energy included gradient energy and directional energy. Some of the segmentation results are shown in Fig. 1. Results: The epithelium, Bowmans, Stroma and Descemets microlayers of those abnormal corneas were successfully segmented using the graph method in all images. The inter-operator error between the two manual operators was 1.75 ±1.54 pixels for the epithelium, Bowmans, 1.73±1.43 for the Stroma and 3.10±2.28 for the Descemets (mean 2.17 ±1.88 pixels). The mean segmentation difference between the graph-based segmentation and the manual operators was 1.69±1.90 pixels for the epithelium, Bowmans, 2.47±3.15 pixels for the Stroma and 4.36±3.21 for the Descemets (mean 2.77±2.99 pixels). The segmentation results were comparable to the manual segmentation (see Fig. 2). The mean segmentation time for all microlayers in one image was 4.27±0.65 seconds and the segmentation had zero intra- operator error. Conclusions: The proposed graph-based segmentation method, for segmenting corneal microlayers using HD-OCT images, was objectively comparable to the manual segmentation for abnormal and healthy corneas and it is significantly faster and repeatable. Fig. 1 Examples of segmentation of inner corneal microlayers
Fig. 2 Segmentation difference comparison
CONTROL ID: 3195940 SESSION ABSTRACT START TIME: 4:30 PM SESSION ABSTRACT END TIME: 4:45 PM FINAL ID: 025

TITLE: A universal three-dimensional registration algorithm on OCT/OCTA for speckle reduction and visualization

FIRST AUTHOR: Yuxuan Cheng AUTHORS/INSTITUTIONS: Y. Cheng, Q. Zhang, S. Song, Z. Chu, R. Wang, Bioengineering, University of Washington, Seattle, Washington, UNITED STATES| Purpose: OCT images often suffer from speckle noises and motion-induced artifacts that affect its accurate, reproducible, and reliable assessment of the samples To mitigate this challenge, we propose a method based on a fast, robust and three-dimensional registration method to enhance the contrast of both OCT and OCT- Angiography(OCT-A) images without pre-processing. Methods: All data were collected by a commercial SSOCTA device (PLEX® Elite 9000 (ZEISS, Dublin, CA)). 6x6 mm 2 scans were utilized to test our method with a dimension 500x500x1560 (x, y, z). 10 repeated volumes were acquired on all participants. The 3D affine transformation was first implemented to low resolution images to correct the bulk motion between different OCT structural volumes and then use the non-ridge B-spline registration to refine the local alignment between the reference and moving volumes on both OCT structural and flow images. Finally, the corrected images were weight-averaged with the previous results and are assigned as the reference images for the next registration. The registration program was developed based on Matlab platform and it costs about 30 minutes for 10 volumes averaging on workstation configured with the Intel Xeon 2630-v3 CPU and 128G RAM. Results: Five subjects have been enrolled in this study with 10 repeated volume scans either with 3x3 mm2 scan or 6x6 mm2 scan. The deformation of sequential images can be rapidly corrected and the motion artifacts can be effectively suppressed. The average peak signal-to-noise ratio (PSNR) measures the misalignment of images, which increased from 25 dB to 72dB. Conclusions: The averaged results from both B-scan and the en-face projection of retinal layer and choriocapillaris showed an enhanced SNR and vascular connectivity comparing to the single scan image. This approach provides better visualization of microstructures and microvasculature that could be helpful in monitoring the disease progression and further assistant the therapeutic intervention. Figure 1.OCTA images of original single and averaged images. (a-c) retinal layer; (d-f) choriocapillaris layer; (g-i) the cross-sectional image located at the yellow dashed line.
Figure 2 (a) The relationship between SNR of OCT structure images and the number of averaging. The dashed line represents the fitting curve by the square of the number of repeated volumes. CONTROL ID: 3195629 SESSION ABSTRACT START TIME: 4:45 PM SESSION ABSTRACT END TIME: 5:00 PM FINAL ID: 026

TITLE: Automated volumetric segmentation of retinal fluid on optical coherence tomography using deep learning

FIRST AUTHOR: Yukun Guo AUTHORS/INSTITUTIONS: Y. Guo, H. Xiong, T. Hormel, J. Wang, T. Hwang, Y. Jia, Casey Eye Institute, Portland, Oregon, UNITED STATES| Purpose: In diabetic macular edema (DME), thickening from intraretinal fluid and thinning from neural degeneration can occur concurrently and confound the understanding of disease progression and treatment response. Current quantitative methods only evaluate overall retinal thickness. We propose a deep learning-based method to automatically detect volumetric retinal fluid on optical coherence tomography (OCT). Methods: 3 × 3-mm2 OCT scans were acquired on one eye by a 70-kHZ OCT commercial AngioVue system (RTVue- XR; Optove, Inc) from 51 participants in a clinical diabetic retinopathy (DR) study (45 with retinal edema and 6 healthy controls). A deep convolutional network with U-net-like architecture (Fig. 1) was constructed to detect and segment the retinal fluid volume. The network contains two types of input B-scans: structural OCT (Fig. 2A) and the corresponding OCTA (Fig. 2B). OCTA scans are used to enhance differentiation between retinal fluid and normal tissue. To improve the quality of OCT B-scans, two B-scans from adjacent positions were averaged. Experts manually delineated retinal fluid (red in Fig. 2C), non-fluid (black in Fig. 2C), and background area (green in Fig. 2C) on each B- scan to establish the ground truth. Data augmentation methods (horizontal flipping and Gaussian noise addition) enlarged the training data set. Six-fold cross-validation was used to evaluate the algorithm on the entire data set. Results: Compared to manual delineation, the algorithm detected retinal fluid on B-scans (Fig. 2D) with an accuracy of (Dice coefficient) 89.1 ± 2.4% (mean ± standard deviation) on cross-validation data. Compiling adjacent B-scans generates 3-dimensional volumes of the retinal fluid with a smooth profile (Fig. 2E). Conclusions: Our deep-learning method accurately segments retinal fluid volumetrically on OCT scans, providing a more complete picture of the anatomic changes in DME. Fig. 1. Retinal Fluid detection algoritlun flow chart. OCT strnctnral B-scans and corresponding: OCTA images (A) as used for the input of network (B). (C) Network output, showing: retinal fluid (blue) overlaid on OCT structural B-scans. Fig. 2. Representative data used for training and segmentation results by our network. (A) Structural OCT B-scan image. (B) OCT angiogram B-scan image. (C) Ground tJ.uth of background (green), non-fluid (black), and retinal fluid (red). (D) Segmentation result of retinal fluid (blue) overlaid on struchll"al OCT B-scan. (E) Volumetric representation of the retinal fluid segmentation results generated by our network. CONTROL ID: 3195696 SESSION ABSTRACT START TIME: 5:00 PM SESSION ABSTRACT END TIME: 5:15 PM FINAL ID: 027

TITLE: Keeping it Clean: Artificial Intelligence Based Denoising Improves Segmentation of Optical Coherence Tomography Images

FIRST AUTHOR: Sripad Krishna Devalla AUTHORS/INSTITUTIONS: S. Devalla, T. Pham, L. Zhang, T. Tin, M. Girard, Biomedical Engineering, National University of Singapore, Singapore, SINGAPORE|T. Pham, T. Tin, T. Aung, L. Schmetterer, M. Girard, Singapore Eye Research Institute, Singapore, Singapore, SINGAPORE|R. Mohan, S. Mohan, Rajan Eye Care Hospital, Chennai, Tamil Nadu, INDIA|L. Schmetterer, Lee Kong Chian School of Medicine,NTU, Singapore, SINGAPORE|A.H. Thiéry, Statistics and Applied Probability, National University of Singapore, Singapore, SINGAPORE| Purpose: To develop an artificial intelligence (deep learning) denoising framework to improve segmentation of optical coherence tomography (OCT) images of the optic nerve head (ONH), independently of the device being used. Methods: Volume scans were acquired through the centre of the ONH using two commercial OCT devices (Spectralis [Heidelberg]:97 B-Scans; Cirrus [Zeiss]: 200 B-Scans) for both the eyes of 1000 subjects. From each volume, noisy (raw) and clean (average of 5 consecutive slices) images were obtained (38,800 Spectralis and 80,000 Cirrus images of each kind in total). A multi-device deep learning network to denoise the OCT images and visually harmonize the device specific characteristics (speckle noise, intensity, contrast, etc.) was developed. The network was trained with 30,000 images (noisy/clean) from each device (one network for each device; shared weights between each network) and tested on the remaining noisy images. The denoising performance was assessed qualitatively and quantitatively using signal-to-noise ratio (SNR) on the unseen images from both the devices. Further, a custom 3D segmentation network was trained on 6 OCT volumes (Spectralis only) to segment 6 ONH tissues. Segmentation quality was then assessed on unseen volumes (for each device) qualitatively (200 volumes) and quantitatively (200 images) using the Dice coefficient (DC; between 0-1; mean of all tissues). To assess the benefits of denoising on segmentation performance, the entire process (training and testing) was repeated on the denoised OCT volumes. Results: With the proposed multi-device denoising network, we were able to successfully denoise (an increase in the SNR greater than 123% for all cases) the noisy OCT images from both devices (Figure 1 (a)). When trained on the noisy Spectralis volumes, the 3D segmentation network was able to isolate 6 ONH tissues with a DC of 0.87/0.66 (Spectralis/Cirrus) on unseen volumes. Upon denoising, a significant improvement (p<0.05) in the DC (0.93/0.89) was observed (Figure 1 (b)). Conclusions: We have developed a custom deep learning approach to denoise OCT images from multiple devices simultaneously. Through this process, the network was able to visually harmonize the images from both the devices. We believe this could facilitate the development of clinically robust device-independent deep learning tools for segmentation & diagnosis applications. a Denoising Performance Baseline (Noisy) De noised Legend

-r (GCL) Inn« Plcxltorm U>ycr (IPL) lnne< Nuclear Layer (INLI Outer "lexiform Layer (OPL) Outer 'luclear L•yer (ONI.) ~etinat Pigment Epithelium (RPE) (.;noro10

b Effect of Denoising on Segmentation OCT Images Segmentations

Baseline Denoised Manual Baseline De noised

Figure 1 (a): Baseline (I) B-scan obtained from a Cirrus OCT device, and its corresponding denoised image (II) are shown.

Figure 1(b): A custom 30 deep learning segmentation network is trained on 6 OCT volumes (Sµectri1lis) and their corresµonding manui1I segmentations. The trained network is tested on OCT volumes trom Cirrus device.(1" row) Baseline (I) and its corresponding denoised (ii) image obtained from a Orrus device is shown. The manual and the deep learning segmentalion obtained on lhe baseline OCT image are shown in (iii) and (iv) respeciively. The entire proce3s (training and testing) i3 repeated on lhe denoised volumes. The deep learning sP.gmentAtion obl-'lined on lhe mmisponding denoisP.<'I imBgP. is shown in (v). The im>1gP.S in the 2•0 row repre~nt lhe AAmP. for Another Cirrus B-Scan. The retinal nerve fiber layer and the prelamina in red, ganglion cell complex in green, all other retinal layers in blue, retinal pigment epithelium in pink, choroid in yellow, lamina cribrosa in cyan. noise in grey and the vitreous humor in black. CONTROL ID: 3195847 SESSION ABSTRACT START TIME: 5:15 PM SESSION ABSTRACT END TIME: 5:30 PM FINAL ID: 028

TITLE: Artificial Intelligence (AI) Screening for Diabetic Retinopathy: Analysis from a Pivotal Multi-center Prospective Clinical Trial

FIRST AUTHOR: Jennifer I Lim AUTHORS/INSTITUTIONS: J.I. Lim, Ophthalmology, University of Illinois at Chicago, Chicago, Illinois, UNITED STATES|M. Bhaskaranand, C. Ramachandra, S. Bhat, K. Solanki, Eyenuk, California, UNITED STATES|S. Sadda, Doheny Eye Institue, California, UNITED STATES|S. Sadda, Ophthalmology, UCLA, Los Angeles, California, UNITED STATES| Purpose: Evaluate an artificial intelligence (AI) system to screen people with diabetes at point-of-care for diabetic retinopathy (DR) including diabetic macular edema (DME). Methods: We conducted a prospective multi-center study (NCT03112005) in which patients with diabetes were enrolled consecutively initially and later preferentially based on enrichment criteria. The study subjects underwent undilated 2-field (macula centered and disk centered images) for the EyeArt AI eye screening system and dilated 4-wide field stereoscopic fundus photography. The EyeArt system provided eye-level results for referable DR (rDR), which is defined as moderate non-proliferative DR (NPDR) or higher (International Clinical DR (ICDR) severity scale) or clinically significant DME. The EyeArt system was evaluated against the clinical reference standard based on adjudicated grading of the 4-wide field photographs by expert graders at the Wisconsin Fundus Photograph Reading Center using the Early Treatment Diabetic Retinopathy Study (ETDRS) Severity Scale. Statistical analyses were performed with variance adjustment to account for the correlation between eyes of the same patient. Results: 1822 eyes from 911 subjects were included. Of these, 1718 eyes were gradable using the clinical reference standard and 326 of these were positive for rDR (290 moderate NPDR, 4 severe NPDR, 31 proliferative DR, and 85 clinically significant DME) and 1392 eyes were negative for rDR (1134 no apparent DR and 258 mild NPDR). Sensitivity of the EyeArt system using undilated images was 95.5% [95% CI: 93.0% - 97.9%], specificity was 86.0% [95% CI: 83.8% - 88.3%, and gradability rate was 87.5% [95% CI: 85.4% - 89.7%]. With a dilate-if-needed photography protocol (where dilated images were used for subjects with ungradable EyeArt results on undilated images), the gradability rate of the EyeArt system improved to 97.4% [95% CI: 96.4% - 98.5%], sensitivity was 95.5% [95% CI: 93.1% - 97.8%], and specificity was 86.5% [95% CI: 84.4% - 88.6%]. Conclusions: The EyeArt AI system compared favorably with the clinical reference standard from 4-wide field stereoscopic images and met the predetermined sensitivity and specificity endpoints for the detection of referable DR in people with diabetes. (No Image Selected) Poster Presentations

10:00- 10:30 am; 12:15 - 1:30 pm; 3:15 - 3:45 pm Room 1, 2, and 3 Session Abstracts Poster PDFs New Technologies Found 161 Records

CONTROL ID: 3195593 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB028 TITLE: Visible-light sensorless adaptive optics optical coherence tomography of retinal response to laser exposure FIRST AUTHOR: Ryne Watterson AUTHORS/INSTITUTIONS: R. Watterson, D. Wahl, M. Ju, M.V. Sarunic, Engineering Science, Simon Fraser University, Burnaby, British Columbia, CANADA| Purpose: In vivo visualization of cellular-level retinal structure with adaptive optics optical coherence tomography enables studies of retinal changes in mouse models of diseases causing blindness. We present our work on visible- light sensorless adaptive optics (VIS-SAO) OCT for structural and fluorescence imaging in the small eyes of mice. The response of the retina to laser exposures is studied in mice with the VIS-SAO OCT. Methods: The VIS-SAO OCT system shown in Fig 1 was constructed based on our previous reports using a supercontinuum laser and tunable filter. Red light with a central wavelength of 635 nm and a bandwidth of 35 nm was delivered at 100 µW into the mouse eye. The system also provided GFP excitation for fluorescence imaging. The system used two deformable elements including a Variable Focus Lens (VFL), and segmented Deformable Mirror (DM). The Adaptive Optics was performed using image-based optimization algorithm, driven by the en face VIS-SAO OCT images. A dichroic mirror was used to co-align a retinal exposure laser with the OCT light, such that higher power light could be focused onto the retina while simultaneously recording images. Results: VIS-SAO OCT images were acquired in pigmented mice. Representative images of mouse retina are shown in Fig 2 demonstrating the high-resolution imaging capability of our system. The B-scan in Fig 2a demonstrates VIS- SAO OCT imaging with the red light. The image in Fig 2b shows the ability to visualize the GFP emission and the effect of SAO optimization from the same system. We have done experiments to investigate retinal response with VIS- SAO OCT during and after laser exposure. The retinal response to different exposures will be presented. Conclusions: We present a VIS-SAO OCT system for small animal retinal imaging, which enables wavefront sensorless aberrations correction for user-selected layers. In vivo retinal imaging of pigmented mice is presented, and the image quality improvement resulting from AO correction is demonstrated. Fig. 1: Multimodal VIS-SAO OCT and fluorescence imaging system. DM, deformable mirror; GM, galvanometer scanner; VFL, variable focus lens; PMT, photo multiplier tube; FC, fiber coupler; MEB, multi-edge beam splitter; DCM, dichroic mirror. Fig. 2: (a) VIS-SAO OCT B-scan of mouse retina with 635nm center wavelength. (b) Comparison of before and after SAO is applied to GFP fluorescence images of microglia. CONTROL ID: 3193378 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB029 TITLE: Transcranial versus transpupil illumination for fundus imaging FIRST AUTHOR: Timothy D. Weber AUTHORS/INSTITUTIONS: T.D. Weber, J. Mertz, Department of Biomedical Engineering, Boston University, Brookline, Massachusetts, UNITED STATES| Purpose: We recently introduced a method to obtain single-pass transmission images of the fundus. The method exploits the relatively deep penetration of near-infrared (NIR) light in tissue to transcranially illuminate the fundus. In this paper, we compare images obtained with the transcranial method to those obtained quasi-simultaneously with conventional transpupil fundus reflection photography. Methods: For transcranial fundus illumination, we coupled NIR light from a high-power LED into a fiber bundle and placed the distal end on the subject’s temple. For reflection imaging, we routed light from a separate LED (at the same wavelength) into the illumination path of a commercial non-mydriatic fundus camera. The fundus camera was equipped with a sCMOS camera. By toggling the two LED channels, in synchrony with the free running camera, we obtained simultaneous reflection and transmission fundus images. 64 frames, corresponding to 2-3 cardiac cycles, were registered, averaged, and flattened for display to improve contrast to noise. Results: Example images of fundus reflection (left column) and transmission (right column) are shown in the attached figure. Notably, in the transmission image, the optic disc is bright compared with its surroundings, likely due to the lack of pigmented epithelium at this location. Also in transmission, the large retinal vessels (shown in the second row zoomed areas) are uniformly dark across vessel lumen, in contrast with the reflection images, which exhibit varying levels of central reflex. Conclusions: NIR light penetrates deeply enough through the skull and head to enable diffuse transmission fundus imaging. This illumination geometry has advantages, including overall simplicity: the entire naturally-dilated pupil may be used to collect images. The transmission geometry is also advantageous for fundus spectroscopy, in contrast with fundus reflectometry where the presence of several light paths (from multiple reflections) adds significant complexity to light transport models. Reflection (left) versus transmission (right) fundus images over a 20° field of view in a healthy volunteer. The illumination for both is an 850-nm LED. To accentuate vessel detail, the second row of images is a zoomed view of the same area in the images above . All images are scaled by their minimum and maximum values. CONTROL ID: 3194305 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB030 TITLE: Line-field SD-OCT with 1.8 μm axial resolution and 2.5 kHz frame rate for imaging the human cornea FIRST AUTHOR: LE HAN AUTHORS/INSTITUTIONS: L. HAN, Z. Hosseinaee, K. Bizheva, department of physics and astronomy, University of Waterloo, Waterloo, Ontario, CANADA|Z. Hosseinaee, K. Bizheva, department of system design engineering, University of Waterloo, Waterloo, Ontario, CANADA| Purpose: To develop and test the performance of a line-field SD-OCT (LF-SD-OCT) with 2.5 kHz frame rate and ~ 2.5 µm isotropic resolution in biological tissue, for in-vivo, non-contact imaging of the cellular structure of the human cornea. Methods: We have developed a LF-SD-OCT system for in-vivo, non-contact imaging of the cellular structure of the human cornea. The system is powered by a femtolaser with emission spectrum centered at 790 nm and spectral bandwidth of 130 nm, resulting in 1.8 µm axial OCT resolution. A cylindrical lens is used to project a line-shaped imaging beam at the corneal surface. By using 5x and 10x microscope objectives, we were able to achieve FOV of 2.6 mm x 1.3 mm and 1.3 mm x 1.3 mm respectively, and lateral resolution of ~5 µm to ~ 2.5 µm respectively. The detection end of the LF-SD-OCT is comprised of a transmission grating spectrometer and a fast 2D camera (1920 x 1080 pixels). The line scan rate of the system was ~2,500 B-scans/s. The system's SNR was measured to be ~92 dB at ~ 100 µm away from the zero-delay line for 2.8 mW imaging power. The system's SNR roll-off was ~17 dB over a scanning range of 1 mm. Volumetric (1080 x 400 x 960) images of the healthy human corneas were acquired both ex- and in-vivo from locations slightly inferior relative to the corneal apex in order to avoid the strong back reflections at the apex. Results: The cross-sectional corneal image in Fig. 1A shows very clear delineation of the boundaries of the Bowman's membrane with the epithelial layer and the anterior stroma. The collagen structure of the stroma is also visible. Figures 1B-1F show the same volumetric data with enface projections at various depths inside the cornea. Stomal keratocytes can be clearly resolved in both the volumetric image (Fig. 1F) and the corresponding enface image (Fig. 1G). Conclusions: Currently, LS-SD-OCT is able to map the morphology of the human cornea. The next step is to combine the LS-SD-OCT with numerical adaptive optics in order to visualize the cellular and sub-cellular structure of the corneal tissue from OCT images acquired in-vivo. A Bscan of human cornea acquired ex-vivo (A). Flattened 3D corneal image with the top layer corresponding to the anterior epithelium (B), midway through the epithelial layer (C), posterior end of the epithelial layer (D), Bowman’s membrane (E) and the anterior stroma (F). An enface view of the corneal stroma is shown in (G) CONTROL ID: 3195852 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB031 TITLE: High-throughput Retinal Whole Mount Imaging Using a Nikon STORM Super-resolution Microscope FIRST AUTHOR: Cathryn Formichella AUTHORS/INSTITUTIONS: C. Formichella, N. McGrady, M. Risner, W. Lambert, D. Calkins, Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, UNITED STATES| Purpose: To develop a high-throughput method for retinal whole-mount imaging and analysis that would allow for capture of images throughout the entire depth of the retina at a resolution that accommodates tracing of retinal ganglion cell dendrites, identification of individual filled cells, and determination of retinal orientation based on a marker protein. Methods: Mouse retinal whole-mounts that were used for intracellular retinal ganglion cell electrophysiology were o fixed in 2% paraformaldehyde at 4 C for 24 hrs. Retinas were immunolabeled for nonphosphorylated neurofilament H (SMI-32, 1:1,000; BioLegend), choline acetyltransferase (ChAT, 1:500; Millipore), and S-cone opsin (1:500; Millipore). o Retinas were first blocked in 5% normal donkey serum for 2 hours, incubated in primary antibodies for 3 days at 4 C, incubated with appropriate secondary antibodies (1:200; Jackson ImmunoResearch Laboratories, Inc.) and coverslipped with Fluoromount G (Southern Biotech, Birmingham, AL). A Nikon STORM super-resolution fluorescent microscope was used to obtain micrographs of whole retinal flat-mounts en montage via the Vanderbilt University Medical Center Cell Imaging Shared Resource. Images were obtained every 2.5 microns over a total z-stack depth of 50 microns to ensure RGC dendritic arbors and S-cone opsin (used for retinal orientation) were captured. Image z- stacks were combined to create an all-in-focus image using the Extended Depth of Focus (EDF) plug-in in Nikon Elements. Results: Average time to image a mouse whole-mount retina using the STORM microscope was 50.9 ± 2.7 minutes. Image processing (EDF and montage generation) took an additional 20 to 30 minutes. The same imaging on a FV- 1000 inverted confocal microscope takes 56.0 ± 6.9 minutes. Image processing for the confocal files takes an additional hour as montages are generated manually. Images from the STORM microscope can be used to trace retinal ganglion cell dendrites by hand or using a user-defined macro. Conclusions: The benefits of STORM imaging for retinal whole-mounts include shorter image-capture and processing times, image processing steps that are automated, and higher resolution images that allow for outcome measurements such as dendritic length, dendritic area, distance from optic disc. (No Image Selected) CONTROL ID: 3195775 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB032 TITLE: A novel tool for the retinopathy of prematurity evaluation, associated with a digital indirect ophthalmoscope hands-free device (DIO-HF) FIRST AUTHOR: Rodrigo M. Torres AUTHORS/INSTITUTIONS: R.M. Torres, Ocular Surface & Immunology. Basic and clinical Research, Centro de Ojos Dr. Lódolo, Colonia Avellaneda, Entre Rios, ARGENTINA|A. Salvatelli, The Information Systems Laboratory, Faculty of Engineering, University of Entre Rios (FiUNER), Oro Verde, Entre Ríos, ARGENTINA|G.A. Monteoliva, G. Saidman, Ophthalmology section, Department of Neonatology, Evita Pueblo Hospital, South Zone ROP net, Berazategui, Buenos Aires, ARGENTINA|G. Alazard, Director of Information Technology, Argentinian Council of Ophthalmology (CAO), Ciudad Autónoma de Buenos Aires, Buenos Aires, ARGENTINA|G. Alazard, Santa Lucía Centro Oftalmológico, Gualeguaychú, Entre Ríos, ARGENTINA|T. Ortíz-Basso, Department of Ophthalmology, Hospital Italiano de Buenos Aires, Ciudad Autónoma de Buenos Aires, Buenos Aires, ARGENTINA|T. Ortíz-Basso, Centro Oftalmológico Santa Rosa, Santa Rosa, La Pampa, ARGENTINA|F. Ghersinich, A. Ghersinich, Clinica Veterinaria Ranelagh, Ranelagh, Buenos Aires, ARGENTINA| Purpose: The digital indirect ophthalmoscope is a headband-mounted device to be used with smartphones, developed to performing indirect hands-free technique for Retinopathy of Prematurity (ROP) diagnosis and follow-up. The purpose of this work is to describe and evaluate the efficiency of a scleral rotator led plus fiber-optic transillumination system, for the ocular fundus evaluation. Methods:

An experimental study was performed to evaluate a prototype, developed with a LED source connected to a 1 mm plastic fiber optic. It is linked to an oval round shape extreme of a scleral depressor similar to “lens ansa”, to delivery the cold light at the other side. (Figure 1) To evaluate the function of the prototype, under general anesthesia, 2 dogs fundoscopy was performed, after a lid speculum was placed. Firstly only with the DIO-HF with smartphone set in flash mode “ON” (group A) and then with the aid of the illumination prototype (group B). Pictures and video frames were taken for both procedures. In the experimental set-up, both eyes were carefully explored. After that, images were processed to be subjectively analyzed. The subjective evaluation was realized to obtain the opinion of 10 ophthalmologists specialized in ROP, to know which ones of them they will prefer. Groups A and B have been masked and ophthalmologist has asked three possible options about their preference: 1- Group A; 2- Group B or 3: There is no difference between both groups. All of the pictures and frame videos were taken for only one ophthalmologist. Results: Images and videos from both groups let to perform a good exploratory exam. The quality of the images and videos obtained in group B have ba etter aspect, less light artifacts. The subjective evaluation shows that all the ophthalmologists have preferred group B. Conclusions: This new prototype seems to be useful to improve the quality image obtained with the DIO-HF, with potential usefulness in smartphone telemedicine networks for ROP, and ROP training.

CONTROL ID: 3193631 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB033 TITLE: Handheld and non-contact retinal camera for ROP monitoring: towards 80-degree retinal views FIRST AUTHOR: Guillem Carles AUTHORS/INSTITUTIONS: G. Carles, M. Preciado, P. Zammit, R. Drysdale, A.R. Harvey, University of Glasgow, Glasgow, UNITED KINGDOM| Purpose: Diagnosing and monitoring the progression of Retinopathy of Prematurity (ROP) is mainly performed using indirect ophthalmoscopy, and treatment is decided based on in situ exploration of the retinas of neonates, missing the benefit of objective assessment and long-term monitoring offered by recorded images. Diagnosing and assessing the severity of ROP requires visualizing the retinal periphery, however glare severely affects image quality if wide field-of- view (WFOV) is attempted, especially using a handheld and non-contact device. We propose an innovative technique for a handheld ophthalmoscope to provide images with sufficiently WFOV and quality to enable ROP diagnosis and monitoring. Methods: A handheld ophthalmoscope was designed and prototyped to acquire WFOV images of the retina, as shown in Figure 1. The system is based on a computational combination of multiple images that are recorded in rapid sequence. The acquisition is electronically synchronised with an illumination system designed to promote non- redundancy of information between images. This computational-imaging approach enables the reconstruction of a glare-suppressed and WFOV images of the retina. The illumination system is sourced by LED illumination and was designed to illuminate a WFOV area of the retina. Results: Images of retinas from volunteers were recorded and processed. Example results are shown in Figure 1 and demonstrate glare-free retinal imaging with a WFOV of 80°. Synchronization of illumination and image acquisition was electronically controlled and achieved the recording of the required multiple images in less than 200ms. Conclusions: A non-contact and handheld ophthalmoscope was designed and prototyped, capable of recording retinal views of up to 80°, enabling the assessment and monitoring the progression of ROP. Being non-contact and achieving WFOV provides a benefit over existing devices for ROP as corneal contact induces undesirable additional stress to premature infants. Multi-spectral information may also be extracted from the images, for example oximetry over tracked vessels may be possible. In particular, two-wavelength oximetry provides arterial/vein discrimination which benefits the diagnosis of Plus disease. The camera is sufficiently compact to operate within an incubator. Design and prototype of the ROP ophthalmoscope, and example retrieved image reaching 80° field of view. CONTROL ID: 3194518 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB046 TITLE: Understanding the variability of handheld spectral-domain optical coherence tomography measurements in supine infants FIRST AUTHOR: Kira Wang AUTHORS/INSTITUTIONS: K. Wang, X. Chen, S. Stinnett, V. Tai, D. Tran-Viet, C. Toth, Duke University, Durham, North Carolina, UNITED STATES| Purpose: Central foveal thickness (CFT) measurements are critically important in the assessment of foveal development and retinal diseases in infants and children. The repeatability and reproducibility of CFT measurements in non-sedated infants is not known. The aim of this retrospective study is to evaluate the variation and reproducibility of supine handheld SDOCT (hh-SDOCT) imaging in infants compared to conventional tabletop imaging of fixating adults. Methods: Two groups were selected for this retrospective study. Group 1 included 25 imaging sessions from 21 preterm infants (mean age at imaging 39 weeks) without macular edema imaged supine in the nursery using hh- SDOCT (Bioptigen, RTP, NC). Group 2 included 25 imaging sessions from 25 adults without eye disease, imaged using tabletop SDOCT (Farsiu et. al, Ophthalmology 2014). For each imaging session, three macular, zero-degree volume scans from one eye were selected. CFTs (distance between the internal limiting membrane and the Bruch’s membrane) for all 150 scans were measured using a customized script for automatic segmentation with manual correction. The coefficient of variations (CV) were calculated over all 25 imaging sessions for both handheld and tabletop systems. The intraclass correlation coefficients (ICC) were calculated with a 95% confidence interval to assess the reproducibility of both systems. Results: The mean CFT was 157.1 ± 35.6 μm for Group 1 and 244.5 ± 18.4 μm for Group 2. CFT measurements from hh-SDOCT imaging of supine infants and tabletop imaging of fixating adults were reproducible. The mean CV was 3.8 for Group 1 (infants) and 2.1 for Group 2 (adults). The ICC (95% confidence interval) was 0.94 (0.90, 0.97) for Group 1 and 0.91 (0.83, 0.96) for Group 2. Conclusions: CFT measurements using both handheld and tabletop SDOCT systems were reproducible. In the developing infant eye, these reproducible measures can be useful in the application to neonatal studies in the nursery. (No Image Selected) CONTROL ID: 3195892 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB043 TITLE: Visualizing Retinal Hemorrhage Thresholds for Q-Switched Nd:YAG Lasers in a Novel Porcine Model FIRST AUTHOR: Adam Boretsky AUTHORS/INSTITUTIONS: A. Boretsky, G. Noojin, SAIC, San Antonio, Texas, UNITED STATES|H. Wang, W. Elliot, P. Edsall, Ocular and Sensory Trauma, US Army Institute of Surgical Research, Ft. Sam Houston, Texas, UNITED STATES|A. Shingledecker, Bioeffects Division/Veterinary Services Branch, Air Force Research Laboratory, Ft. Sam Houston, Texas, UNITED STATES|B. Rockwell, Bioeffects Division/Optical Radiation Bioeffects Branch, Air Force Research Laboratory, Ft. Sam Houston, Texas, UNITED STATES| Purpose: Neodymium-doped yttrium aluminum garnet (Nd:YAG) lasers are among the most commonly used lasers with a wide variety of applications from biomedicine to manufacturing. The ubiquity of these lasers increases the likelihood of accidental ocular injury resulting in permanent visual impairment. We performed dosimetry studies to determine retinal damage thresholds in the porcine eye with nanosecond Q-switched Nd:YAG lasers. Methods: A mini Yucantan pig model exhibited similarities in ocular size and retinal anatomy to human eyes. The Nd:YAG laser (Continuum, Minilite II), tuned to 1064 nm with a pulse width of five nanoseconds, delivered laser energy to precise locations on the porcine retina via an articulating arm attached to a modified Topcon fundus camera. Retinal imaging modalities including fundus photography, real-time video, confocal scanning laser ophthalmoscopy (cSLO), and spectral domain optical coherence tomography (SD-OCT) provided complementary high-resolution visualization of retinal morphology at multiple time points ranging from immediately after laser exposure up to 10 days post exposure. Results: The retinal damage thresholds were grouped into three categories (1) minimum visible lesion (MVL), (2) contained hemorrhagic lesion (CHL), and (3) vitreal hemorrhagic lesion (VHL). Probit analysis determined the effective dose for 50% probability of damage (ED ) for each lesion category. The threshold to produce a hemorrhagic lesion 50 was less than 500 μJ and the threshold to produce a MVL was below 100 μJ in the porcine model, which closely correlates with previously published results in other animal models. Conclusions: The determination of ocular damage thresholds for common lasers such as the Nd:YAG is important because of the inherent risks, particularly for near infrared wavelengths that are not visible to the unaided eye. The hemorrhagic lesions represent an laser-induced form of retinal injury with the potential for extensive visual impairment. The porcine model provided a close approximation in the anatomical size and optical properties of the human eye making it a suitable model for retinal laser-tissue interaction studies. Additional immunohistochemical analysis may provide further insights into the differences in the inflammatory response and wound healing dynamics for each lesion category. (No Image Selected) CONTROL ID: 3195568 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB042 TITLE: Fundus imaging through lenticular media opacities using broad line fundus imaging FIRST AUTHOR: Angelina Covita AUTHORS/INSTITUTIONS: A. Covita, M. Chen, C. Leahy, J. Jung, S. Lee, Carl Zeiss Meditec, Inc, California, UNITED STATES| Purpose: Broad line fundus imaging (BLFI) is a technique that aims to reduce artifacts in wide field fundus images. The illumination and imaging paths are separated in the patient pupil. By illuminating only a narrow strip of the retina at any instant, the illumination is separated from the imaging path, removing haze and fluorescence from the anterior segment out of the retinal image (refer to Figure 1). The purpose of this study is to characterize the performance of a commercially available BLFI system for imaging through varying levels of media opacity. Methods: Eyes with varying types and severities of cataracts were recruited for this study. The pupils were pharmacologically dilated, and the cataracts were graded at the slit lamp by a licensed eye care provider. Each subject was then imaged on a CLARUS™ 500 (ZEISS, Dublin, CA). A wide field color image with central fixation was obtained for each eye. Images were exported and graded by an ophthalmic technician. Each image was graded on a 1-5 scale, where 5 exhibit an image that a clinician can assess and diagnose with complete confidence. These images present exceptional sharpness and contrast (Figure 2). 1 represents zero visibility and a non-diagnostic image. A passing image is graded 3 or above. Results: 61 eyes of 32 subjects with an age range of 55-95, (mean 68, SD=19), were imaged as part of this study. 61 images were acquired and graded. Types of cataract included cortical sclerotic (CS), nuclear sclerotic (NS) and posterior subcapsular cataract is (PSC). Figure 3 displays the number of passing images out of the total image count of each cataract type and grade. The percentage indicates the passing grade (>Grade 3). The majority of images had a passing grade or better. Conclusions: In this group of subjects, BLFI produced clinically usable images in the presence of a variety of media opacities. Further studies would be useful to compare the performance of BLFI vs traditional fundus cameras or scanning LASER ophthalmoscopes. Figure 1: Figure2: Figure 3:

cataract cs NS PSC grade

~ 41...,,1 /a, 11/ a (lOOS<':I 1_ 6/E 8/8 J/j_ (100~ 1 (100%\ llOCo/cl 2 E/15 20/20 5/ 5 (100~ 1 (100%) (ltl0%l .3 If~ !t/5 11/ 3 (100~1 (el0%) 4 1 ~ 1/4 11/ 3 (50% ) CONTROL ID: 3194199 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB045 TITLE: Comparison of manual versus automated anchor point finding on montaging in ultra-widefield fundus imaging FIRST AUTHOR: Ashwini Tamhankar AUTHORS/INSTITUTIONS: A. Tamhankar, K. Meng, N. Manivannan, K. Makedonsky, M.K. Durbin, Carl Zeiss Meditec, Inc, California, UNITED STATES| Purpose: Early indications of some of the retinal diseases like ocular ischemic syndrome, retinal detachment and sickle cell retinopathy are apparent in the periphery of retina. Montaging fundus images gives us the ability to view the extreme periphery of the retina. Before montaging, the anchor point defined at the fovea, can be manually chosen by the user or automatically detected using an algorithm. The purpose of this study was to compare the image quality of ultra-widefield (UWF) montaged images generated using manual and automated anchor point finding. Methods: TrueColor widefield (WF) fundus images (50) were captured on 25 subjects per eye (19 healthy and 6 diseased eyes) using CLARUS™ 500 (ZEISS, Dublin, CA). The montaging workflow involved anchor point finding (APF), distortion correction, feature finding and montaging. Anchor point (fovea) selection was performed manually and automatically for comparison. The user executed the manual and automated APF on WF images and generated montaged ultra WF (UWF) images. For qualitative evaluation, montage images were graded by the clinical expert for image quality considering vessel breakage, uniformness of both halves, artifacts, and color balance on the scale of 1 to 5 (1: unsuccessful, 2: poor, 3: fair, 4: good, 5: excellent). The image quality grade results of montaged images for manual and automated process were compared. Statistical analysis was performed to assess differences in the image quality. Results: Both, manual and automated montage workflow received image quality grade average of 4.7 indicating good to excellent image quality. Paired t test (p=1) results do not show statistically significant difference with 95% confidence interval in the manual versus automated image quality. Conclusions: In this study, the image quality of the ultra-widefield montaged images generated using manual and automated anchor point finding were comparable. This shows that the performance of automated anchor point finding process is reliable in generating montaged images with clinically acceptable image quality. Manual and Automated Montage UWF Images (Healthy Eye Images) Manual and Automated Montage UWF Images (Diseased Eye Images) CONTROL ID: 3195099 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB039 TITLE: Improving autofocus performance in a widefield fundus imaging system, using peripheral defocus measurements FIRST AUTHOR: Conor Leahy AUTHORS/INSTITUTIONS: C. Leahy, D. Nolan, K.E. O'Hara, M. Chen, K. Brock, W.G. Hunder, J. Straub, Carl Zeiss Meditec, Inc., Dublin, California, UNITED STATES| Purpose: Traditionally, fundus camera designs incorporate a large collection aperture to allow for comfortable levels of illumination, which limits the depth-of-field. For widefield systems, maintaining good focus across the full field-of- view (FOV) can be challenging due to the wide range of human retinal curvatures. In this study, we propose an autofocus scheme for a widefield fundus imaging system, aimed at balancing focus quality throughout both the central and peripheral field. Methods: A widefield slit-scanning ophthalmoscope, CLARUS™ 500 (ZEISS, Dublin, CA), with prototype software was used for imaging the retina over a 90° FOV. During alignment, the displacement on the retina between two illumination beams entering the pupil at different locations was measured and used to infer the best focus setting. In the proposed approach, focus error is estimated for both central and peripheral retinal regions simultaneously (Figure 1A). An aggregated focus setting is then applied to balance focus quality across the 90° FOV. Color fundus images were captured from 12 eyes at a variety of fixation orientations, totaling 22 different imaged retinal fields. Each field was imaged using (i) autofocus based on an estimate of focus error near the center of the FOV, and (ii) the proposed approach. The captured images were graded for overall focus quality by a licensed clinician; the assigned grades were normalized to a range of 1-5 (with 5 signifying the best focus quality). Results: The mean scores for focus quality were 3.32 ± 0.84 for central autofocus, and 4.05 ± 0.79 for the proposed autofocus strategy. Using the proposed strategy, 16/22 images achieved a grade of 4 or better, compared to 7/22 using central autofocus. Conclusions: Our results suggest that an autofocus strategy during image acquisition that attempts to compensate peripheral defocus, as well as central focus error, has the potential to improve the overall quality of focus in widefield fundus images. Figure 1. (A) The displacement on the retina between two illuminating beams is used to infer focus error at different retinal locations; (B) 90° fundus image of a myopic eye. Using central autofocus, features near the FOV center appear sharp (C), but important detail in the periphery exhibits defocus (D). Using the proposed autofocus scheme, good image sharpness was maintained both centrally (E) and peripherally (F). CONTROL ID: 3195227 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB034 TITLE: Repeatability of retinal curvature estimation on wide field OCT systems FIRST AUTHOR: Jonathan Bumstead AUTHORS/INSTITUTIONS: J. Bumstead, M. Steidle, C. Leahy, J. Straub, Carl Zeiss Meditec, Inc., Dublin, California, UNITED STATES| Purpose: Wide field optical coherence tomography (OCT) enables imaging large regions of the retina with the potential to make accurate measurements of the retinal surface profile. However, the apparent curvature of the retina in conventional OCT images does not represent the actual anatomical shape. Here we present a technique for ® estimating the retinal curvature from data acquired with a Swept-Source OCT system (PLEX Elite 9000, ZEISS, Dublin, CA) and an extensive tolerance analysis demonstrating its capabilities. Methods: In a B-scan image, each line orthogonal to an A-scan corresponds to a surface in the sample with constant group delay (GD). By simulating the system imaging the Arizona eye model in Zemax (Fig. 1), we calculated the A- scan paths (for different angular orientations of the scan mirrors) and the surface of constant GD for the PLEX® Elite 9000 with 80° field-of-view add-on lens. B-scan images acquired with the system were transformed by repositioning the displayed A-scans in angle and space to match the GD surface profile calculated in the optical model. After segmentation of retinal layers, a circular fit was applied to the retinal pigment epithelium to quantify the retinal curvature. To validate the performance of the system, we conducted a tolerance analysis that determined the error in retinal curvature measurements as a function of various parameters and refractive error (Fig. 2). Results: The axial length of the eye has the biggest effect on errors in the retinal curvature estimation (Fig. 2(a)). The eye position and tilt relative to the system had a negligible effect, regardless of refractive error (Fig. 2(c)-(f)). Conclusions: The GD technique presented can potentially estimate the curvature of the retinal surface. Our simulations demonstrate that the axial length of the eye is critical for estimating the retinal curvature and the working distance should be considered for hyperopic patients (Fig. 2(b)). Figure 1. (a) Eye model displaying difference in retinal surface and surface of constant group delay (b) OCT image of ® retina collected with PLEX Elite 9000 (c) Corrected OCT image. Figure 2. (a) Percent error in estimated retinal curvature as a function of axial length and refractive error ranging from -15 to 5 Diopter. Results are from simulations conducted in Zemax. (b-f) Same as a, but for other parameters in the model. CONTROL ID: 3195347 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB040 TITLE: Automatic pupil detection using off-axis iris images for alignment guidance in fundus cameras FIRST AUTHOR: Poojan Dave AUTHORS/INSTITUTIONS: P. Dave, A. Wei, D. Nolan, S. Stock, A. Covita, M. Chen, J. Straub, M. Durbin, N. Manivannan, Carl Zeiss Meditec, Inc., Dublin, California, UNITED STATES|S. Stock, Department of Electrical Engineering, Karlsruhe Institute of Technology, GERMANY|J. Guo, Carl Zeiss (Shanghai) Co., Ltd., CHINA| Purpose: The purpose of this study is to create a pupil tracking algorithm to find the center of the pupil within 400 micrometers of ground truth (manual annotations) in non-mydriatic external eye images. Pupil detection is crucial for automation and alignment guidance, which can improve the quality of fundus image acquisitions. TM Methods: In the ultra-widefield fundus imaging system CLARUS 500 (ZEISS, Dublin, CA), two iris cameras provide an off-axis view of the patient’s eye along with the position of the pupil within the field of view. In this retrospective study, we used 654 external eye images (pixel size: 320x240) of non-mydriatic pupils (<3.5 mm pupil size) from 29 subjects. Manual annotations of pupil boundary and the center were marked by an expert grader. The dataset is divided into training (534 images from 18 subjects) and testing sets (120 images from 11 subjects). Fig 1 shows the flowchart of the proposed pupil detection algorithm. The algorithm consists of two blocks: 1) coarse region-of-interest (ROI) finder and 2) fine-tuned pupil detector. Coarse ROI finder consisted of a single-shot detector (SSD) with 7 convolutional neural networks (CNN). A bounding box with the highest confidence score is used as the starting point for the fine-tuned detection. The Shootingstar algorithm is an extension of the Starburst pupil detection algorithm. The Shootingstar implementation shoots rays at five positions (the center and on the four corners of the bounding box). The Euclidean distances between the pupil centers determined by the algorithm in the test set were compared with the manual annotations. Results: Fig 2 shows the results of correct and incorrect detections from the proposed algorithm. The algorithm achieved an accuracy of 91.5% in tracking pupils within 400 micrometers. Incorrect results are usually caused by the patient blinking or being in the middle of a blink. The execution time of the algorithm is 57.4 ± 3.8 ms using an Intel® Core™ i7-6920HQ [email protected]. Conclusions: The proposed algorithm provides a reliable solution for pupil detection for alignment guidance for fundus image capture. The algorithm can detect up to 17 frames per second and would be suitable for real-time pupil tracking. Fig 1. Workflow of the proposed pupil tracking algorithm Fig 2. Top row shows correct detection; Bottom row shows incorrect detections. (Red = Manually annotated pupil, Blue = Algorithm detected center) CONTROL ID: 3179661 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB041 TITLE: Enhanced Visualization of the Inner Retinal Layers Using Adaptive Optics Optical Coherence Tomography FIRST AUTHOR: Ryosuke Tamiya AUTHORS/INSTITUTIONS: R. Tamiya, Y. Muraoka, S. Kadomoto, A. Uji, A. Tsujikawa, Department of ophthalmology & visual sciences Kyoto university graduate school of medicine, Kyoto university, Kyoto, Kyoto, JAPAN|K. Nozato, CANON INC., Tokyo, JAPAN| Purpose: Optical coherence tomography (OCT) has aided in elucidation of physiologic or pathological changes in the human retina in vivo. However, on conventional OCT, the contrast of the ganglion cell layer (GCL) to the inner plexiform layer (IPL) is not high enough to precisely evaluate the pathological changes in the inner retina at each layer. In this study, we have developed a prototype adaptive-optics (AO)-OCT system, and investigated whether an evaluation of each inner retinal layer could be performed using the AO-OCT. Methods: Seven eyes of 7 healthy volunteers (mean age 36 ± 6 years, range 30-44 years) were imaged with a prototype AO-OCT system (Canon Inc., Tokyo, Japan). The AO-OCT system provides high resolution retinal imaging with an axial resolution of 3.4 μm and a transverse resolution of approximately 3 μm. A total of 400 B-scans were acquired and averaged to obtain the final images for analyses. The superior fovea, which was located 0.75 mm superiorly from the center of the foveola, was vertically scanned with the AO-OCT, and the contrast-to-noise ratio (CNR) of GCL to IPL was calculated. The CNR calculated on AO-OCT was compared to that of a conventional, non- AO-OCT (OCT HS-100, Canon Inc.). In addition, the average thicknesses of the retinal nerve fiber layer (RNFL), GCL, and IPL were measured with a width of 1.0 mm. Results: During OCT imaging, the developed AO system could successfully detect an ocular aberration in real time in all subjects. When the focus of AO-OCT was placed on the inner retinal layers, the contrast between the GCL and the adjacent inner retinal layers was apparently increased, and the borders between layers were more sharply demarcated than those imaged by using conventional SD-OCT. The CNR of GCL to IPL on AO-OCT (1.920±0.880) was significantly higher than that on non-AO-OCT (0.884±0.307, P=0.0365) and the ratio of CNR on AO-OCT to that on non-AO-OCT was 2.35±1.24. In AO-OCT, the average retinal thicknesses of RNFL, GCL, and IPL were 14.0 ± 2.0 μm, 56.4 ± 4.7 μm, 30.5 ± 2.6 μm, respectively. The measurement value of each retinal thickness was consistent with that of previous histological investigations (Iwasaki et al., IOVS 1986). Conclusions: In AO-OCT, contrast between GCL and IPL significantly increased compared to that of conventional OCT imaging. Enhancement of GCL contrast on AO-OCT may be useful for layer-by-layer evaluation of the inner retina. (No Image Selected) CONTROL ID: 3195562 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB044 TITLE: Meta-Analysis of Handheld Fundus Camera Validation Studies FIRST AUTHOR: Samantha D'Amico AUTHORS/INSTITUTIONS: S. D'Amico, B. Kim, C.J. Brady, Univeristy of Vermont Larner College of Medicine, Burlington, Vermont, UNITED STATES|S. D'Amico, B. Kim, C.J. Brady, University of Vermont Medical Center, Vermont, UNITED STATES| Purpose: Fundus cameras are vital tools in addressing preventable blindness, but traditional desktop cameras are often costly, bulky, and dependent on the skills of the user. Handheld fundus cameras have reduced these barriers, but data on their efficacy are limited. We reviewed existing literature to identify validation studies of handheld fundus cameras and conducted a meta-analysis of the sensitivity and specificity of the camera. Methods: We conducted a Pubmed search in October 2018 for “handheld fundus camera” and “portable fundus camera” and a Pubmed and Google Scholar search for “Horus scope”, “Visuscout”, “Pictor plus”, “Retinavue”, “Versacam”, “Kowa Genesis”, “Epicam”, “Smartscope”, and “Microclear Luna”. Papers not in English were excluded. A title and abstract review was conducted and non-validation studies were excluded. Full-text articles were assessed and excluded if the camera was not commercially available, the study was not a camera validation study, there was no comparison to the gold standard, or data was not quantified. Studies included in the meta-analysis validated a handheld fundus camera against a gold standard method. If not provided, sensitivity and specificity were calculated using true and false positive and negative values for each grader. True positives/negatives and false positives/negatives were averaged to generate mean values for each study if there was more than one grader. Mean sensitivity and specificity were calculated for each study. Any reported data on image quality/gradability was extracted. Results: Ninety-five studies were reviewed for inclusion in the review with 7 included in the meta-analysis (Figure 1). The Pictor/Pictor Plus camera was used in all but 1 study. Mean sensitivity ranged from 79.20-100% and mean specificity from 72.09-93.85% (Figure 2). The sensitivity and specificity for all studies calculated using a weighted average were 85.32% (95% CI: 80.15-89.54%) and 84.63% (95% CI: 80.30-88.34%). Analysis of image quality/gradability was not feasible due to the heterogeneity of reporting in included studies. Conclusions: We found that hand-held cameras are capable of achieving acceptable sensitivity and specificity values as screening tools. While these values may not be acceptable for all clinical programs, they offer a foundation to build upon for future development of handheld fundus cameras. Figure 1: Flow diagram of the review and meta-analysis Table 1: Summary of included studies CONTROL ID: 3189238 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB035 TITLE: Moiré-pattern imaging-based IOP sensing contact lens in a Glaucoma rabbit model FIRST AUTHOR: Se-Hee Lee AUTHORS/INSTITUTIONS: S. Lee, Vision Science and Optometry, Catholic University of Daegu, Gyungsan, KOREA (THE REPUBLIC OF)|S. Lee, J. Kim, Biomedical Engineering, Catholic University of Daegu, Daegu, KOREA (THE REPUBLIC OF)|K. Shin, B. Cheong, J. Kang, Korea Institute of Science and Technology, Seoul, KOREA (THE REPUBLIC OF)| Purpose: Continuous monitoring of intraocular pressure(IOP) is necessary for successful treatment of glaucoma since patients can have a wide variation of IOP in circadian pattern and during the medication. This study presents a new method of IOP measurement by generating Moiré imaging pattern that is proportional to the IOP change with a single contact lens(CL) and a virtual second reference image that needs no additional CL Methods: The IOP of rabbit was controlled by injection of Healon into anterior chamber and checked by tonometer and UBM. After injection of Healon, the Moire pattern on the CL was captured by conventional camera. To measure the IOP change, the Moiré effect is formed by superimposing a camera-captured image on the micro pattern of CL with computer assisted a virtual reference image. To extract IOP data from overlapped Moire patterns, the relative changes in the image pattern were converted into a waveform signals over pixel by pixel by Histograms of Gradient(HOG) algorithm using MATLAB, in which pixel intensity information were converted into the gradient information. Then waveform analysis by Fourier transform provided approach to monitor IOP change. Results: As increased the IOP in rabbit eye, the Moire pattern on the CL(Fig 1A) was changed accordingly, producing overlapped image patterns(Fig 1B) in proportion to the IOP change. We compared the Moire pattern changes with the anterior chamber angle(ACA) that was measured according to the applied pressure(0-30 mmHg)(Fig 1C). The nd extracted pattern changes were obtained from overlapping with computer-assisted 2 Moire pattern by image analysis, which were proportion to the IOP(Fig 1D) and well coincided with ACA in a number of selected IOPs(Fig 1E). Conclusions: The Moire pattern on the CL was changed by applied IOP or in glaucoma rabbit where IOP was increased. It was possible to extract the pattern change from computer-assisted image analysis. IOP increase and ACA were correlated in glaucoma rabbit model. In conclusion, IOP was predicted by analyzing the shape of the Moire pattern on the CL according to the pressure. Representative Moire pattern on CL (A), superimposed patterns with computer assisted a virtual reference image (B) and UBM images showing measurement of ACA. Moire patterns showed their proportional change with different IOPs (D) and correlation with measured ACA according to IOP in Glaucoma rabbit model (E). CONTROL ID: 3195348 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB037 TITLE: Performance evaluation of ganglion cell analysis in normal and glaucoma population using multi-retinal layer segmentation FIRST AUTHOR: Sophia Yu AUTHORS/INSTITUTIONS: S. Yu, H. Bagherinia, A. Fard, M. Durbin, Carl Zeiss Meditec, Inc., Dublin, California, UNITED STATES|R.W. Knighton, Bascom Palmer Eye Institute, University of Miami, Miami, Florida, UNITED STATES| Purpose: Optical coherence technology (OCT) provides the ability to measure ganglion cell + inner plexiform layer thickness (GCIPL) and visualize the various tissues within the retina. GCIPL thickness is measured as the difference between the inner boundary of the retinal nerve fiber layer (RNFL) and the outer boundary of the inner plexiform layer (IPL). We have developed a multi-retinal layer segmentation algorithm (MLS) that segments the RNFL and IPL as well as other layers. The purpose of this study is to evaluate the performance of the GCIPL using MLS algorithm. Methods: Subjects with no retinal disease and with glaucoma were recruited from 2 sites and scanned with the TM 200x200 and 512x218 macular cube scans over 6x6 mm using the CIRRUS HD-OCT 5000 (ZEISS, Dublin, CA). Accuracy of MLS was compared against RNFL and IPL segmentations hand-drawn by a grader trained in segmenting OCT volumetric images (gold standard). The grader drew RNFL and IPL boundaries over three specified B-scans (at 2.1mm, 3 mm, 3.89 mm) per OCT volume (512x218 macular cube scans). Bland Altman and regression plots are reported on a total of 132 B-scans. The repeatability, coefficient of variance (COV) and receiver operating characteristic (ROC) curve for each subfield of the EDTRS grid centered on the fovea is reported from 49 normal subjects (282 cube scans) and 49 glaucoma subjects (244 cube scans). Results: In the Bland Altman and regression plots (Fig 1), the overall mean difference between MLS and manual 2 segmentation for RNFL and IPL was found to be less than +/- 3 µm. R values show a very high correlation between gold standard and MLS. The 95% limits (+/- 1.96 times standard deviation) of the difference between gold standard and MLS calculated for RNFL and IPL are also reported. The area under the curve (AUC) was calculated in all subfields (Fig 2). The minimum AUC was 0.94 (subfield 3) and the maximum AUC was 0.99 (subfield 6). The repeatability of normal and glaucoma cases were calculated separately for all 6 subfields (Fig 2). The COV for normal cases was less than 2.4%. The largest COV for glaucoma cases was 6% (subfield 4). Conclusions: Our proposed segmentation algorithm achieves high accuracy, repeatability and reproducibility for GCIPL thickness analysis. It is therefore a promising tool to detect glaucoma and monitor its progression using OCT imaging. Fig 1 Fig 2 CONTROL ID: 3194430 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB038 TITLE: Fully automatic localisation of the optic disc using YOLO in colour fundus photographs FIRST AUTHOR: Yalin Zheng AUTHORS/INSTITUTIONS: Y. Zheng, Y. Zhao, X. Chen, D. Gao, J. Bridge, W. Zhu, B. Williams, Department of Eye and Vision Science, University of Liverpool, Liverpool, UNITED KINGDOM|Y. Zheng, St Paul’s Eye Unit, Royal Liverpool University Hospitals NHS Trust, Liverpool, UNITED KINGDOM|Y. Zhao, Ningbo Institute of Industrial Technology, Chinese Academy of Sciences, Ningbo, CHINA| Purpose: Detection of the optic disc (OD) is important for the management of eye disease. Knowledge of the OD is considered essential for the diagnosis and screening of many retinal diseases, with the OD centre often regarded as a reference point for locating other retinal structures in the assessment of colour fundus photographs. The purpose of this work is to develop a fully automatic method for the localisation of the OD from colour fundus photographs. Methods: We propose a fully automatic method for OD localisation in colour fundus photographs. The proposed method is based on a YOLO (you only look once) network architecture, which allows the simultaneous localisation of an object of interest and detection of the bounding box. 2708 images from 7 publically available datasets were used along with corresponding manual OD annotations from expert graders. We trained the network on 1508 colour fundus images from 6 publically available datasets (DRIONS: 110; DRISHTI: 101; ONHSD: 88; ORIGA: 650; REFUGE: 400) th and tested on the 1200 images from the 7 dataset (MESSIDOR). Localisation accuracy was evaluated in terms of ¼, ½ and one OD radius (ODR). Results: State-of-the-art detection results were achieved, demonstrating the excellent performance of the method and robustness of the detection. The detected OD centres in 1149 images out of 1200 images (95.75%) were within ¼ ODR of the annotated centre. The detected OD centres were within ½ and one ODR for 1199 images (99.9%). Importantly, these results have been achieved in the external validation of our model with data from a different population. Six localisation examples are shown in the figure. Conclusions: A reliable and accurate automation method was proposed for the localisation of the OD and validated on external data, achieving state-of-the-art results. This can save considerable amounts of time, improving disease management and diagnostic potential, and paving the way for complete, fully automated systems to be realised for diagnosing eye disease. Figure: Six randomly chosen examples demonstrating the localisation where rectangles in red highlight the detected bounding boxes of the optic disc. CONTROL ID: 3195900 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB036 TITLE: Quantity and Quality of Image Artifacts and Segmentation Errors in Diabetic Retinopathy using Wide Field Swept Source Optical Tomography Angiography. FIRST AUTHOR: Ying Cui AUTHORS/INSTITUTIONS: Y. Cui, R. Katz, J. Wang, J.B. Miller, Retina service, Mass Eye and Ear,Department of Ophthalmology, Harvard Medical School, Massachusetts, UNITED STATES|Y. Cui, Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong, CHINA|Y. Zhu, Ophthalmology, Xiangya Hospital, Central Southern University, CHINA|Y. Lu, Harvard Medical School, Massachusetts, UNITED STATES|M. Kasetty, School of Medicine, Tuft University, Massachusetts, UNITED STATES| Purpose: To analyze the quality and prevalence of image artifacts and segmentation errors in widefield swept source optic tomography angiography (SS-OCTA) in diabetic retinopathy (DR). Methods: We conducted a prospective, observational study in Mass Eye and Ear from December 2018 to February 2019. Proliferative diabetic retinopathy (PDR), non-proliferative diabetic retinopathy (NPDR), diabetic patients with no DR and healthy eyes with no ocular diseases were included. All patients were imaged with an SS-OCTA (PLEX® Elite 9000) in active eye tracking and the montage image composed of two 15mm*9mm angio-image (superior and inferior) was used for analysis. Best-corrected visual acuity (BCVA, LogMAR) and signal strength index (SSI) were recorded. Images were independently evaluated by two graders using the OCT-A motion artifact score (MAS; score I-IV) and segmentation accuracy score (SAS; Sore I-IIB) after automatically removing projection. All statistic analyses were performed using SAS ® software. Results: Seventy-two eyes (30 eyes with PDR, 19 eyes NPDR, 13 eyes of no DR and 10 healthy eyes) were included in the study. The average age of the participants was 56.08±4.06 years, and there was no significant difference among the 4 groups (P=0.68). Compared with the superior part(8.64±0.20), mean SSI in inferior (8.52±0.20) was significantly lower (P<0.05). In addition mean MAS was 0.83±0.12 and 0.97±0.16 in superior and inferior part, respectively. Segmentation was accurate in health (100%), no DR eyes (92.3%) and NPDR (78.95%). Segmentation errors were mostly observed in DR patients with neovascularization (NV), diabetic macular edema (DME) and epiretinal membrane (EM). Furthermore, the lowest MAS (1.2 ± 0.11) and highest percentage of inaccurate segmentation (60%) were found in inferior part in eyes with PDR. Media opacity in 18 eyes (24.0%), and in superior part (10 eyes, 55.56%). We also observed alignment error (22 eyes, 30.56%) and periphery artifacts (47 eyes,65.28%) in montage images. Conclusions: Motion artifacts and segmentation errors are frequent in eyes with PDR in wide-field OCTA. It is necessary to assess the motion artifacts and segmentation of montage image prior to get meaningful and reliable results, especially in DR patients with NV, DME and EM. (No Image Selected) OCT Angiography CONTROL ID: 3195614 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB047 TITLE: Changes in Macular Retinal and Choriocapillaris Blood Perfusion in Diabetes without Retinopathy assessed by Swept-Source OCT Angiography FIRST AUTHOR: Yining Dai AUTHORS/INSTITUTIONS: R. Wang, Ophthalmology, University of Washington, Seattle, Washington, UNITED STATES|Y. Dai, Retina, Shanxi Eye Hosptial, Taiyuan, Shanxi, CHINA|Y. Dai, Q. Zhang, Z. Chu, H. Zhou, R. Wang, Bioengineering, University of Washington, Washington, UNITED STATES| Purpose: To investigate the vascular perfusion of retina and choriocapillaris (CC) in diabetic patients without retinopathy using swept source optical coherence tomography angiography (SS-OCTA). Methods: SS-OCTA (PLEX® Elite 9000 (ZEISS, Dublin, CA)) running at 100 kHz A-line rate was used to collect 6 x 6 2 mm macular scans from subjects. Three retinal layers and CC layer were segmented by the semi-automatic method. Vessel area density (VAD) and vessel skeleton density (VSD) were calculated on three retinal layers in 1.5mm rim (R1.5), 2.5mm rim (R2.5) and the entire rim (R4.0) that exclude the central 1 mm circle (C1). The non-perfusion area (NPA) was also investigated in R1.5 and R2.5 in three retinal plexuses. Foveal avascular zone (FAZ) area was automatically measured in three retinal layers. For CC, the percentage of flow deficits (FD %) and the average size of FDs were measured in C1, R1.5, and R2.5, as well as the entire 5.0mm circle (C5). Results: A total of 16 eyes from 16 diabetic patients without clinically visible retinopathy and 16 control eyes from 16 age-matched nondiabetic subjects were included in this study. Mean duration of diabetes was 2.1 ± 1.20 years. There was no significant difference between the two groups in all retinal quantitative parameters (VAD, VSD, and NPA) within all regions (R1.5, R2.5, and R4, P>0.05). There was no significant difference in the FAZ area between the two groups (all P>0.05). No statistical difference in FD % or average FD size within C1 regions was found between the two groups. Mean FD % in CC was significantly increased in diabetic eyes compared to controls within R1.5 (22.75 % vs. 14.33 %; P < 0.001), R2.5 (15.77 % vs. 9.72 %; P = 0.001), and C5 regions (17.55 % vs. 11.07 %; P < 0.001). Similarly, the average size of FDs was significantly increased in diabetic eyes compared to controls within R1.5 2 2 2 2 2 (2823.5 µm vs. 1931.9 µm ; P < 0.001), R2.5 (2158.2 µm vs. 1618.8 µm ; P = 0.005), and C5 regions (2454.7 µm 2 vs. 1799.6 µm ; P = 0.002) . Conclusions: CC perfusion was found to be decreased in diabetic patients without retinopathy as compared to age- matched nondiabetic controls, which occurred before the microvascular changes in the macular retina. CC perfusion may be an early predictor for the microvascular changes in the eye at an early stage of diabetes instead of the retinal vasculature. Control

Diabetes Choriocapillaris VAD map VSD map NPA map FAZ map CONTROL ID: 3195170 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB048 TITLE: OCTA flow signal enhancement by reducing residual structural signal FIRST AUTHOR: Tilman Schmoll AUTHORS/INSTITUTIONS: T. Schmoll, H. Bagherinia, H. Ren, Carl Zeiss Meditec, Inc., Dublin, California, UNITED STATES|T. Schmoll, Medical University of Vienna, Vienna, AUSTRIA| Purpose: The image quality of OCT angiography (OCTA) strongly depends on the contrast between static tissue and moving scatterers, i.e. blood cells travelling through capillaries. Simply calculating the difference or variance between multiple acquisitions at the same location attenuates signal from static tissue and highlights perfused capillaries. However, the high contrast OCTA images clinicians are used to, undergo significant image processing before they are displayed. We present a fast, practical approach for further suppressing signal from static tissue and hence enhancing the contrast of OCTA images. Methods: We generate OCTA B-scans using the Optical Micro Angiography (OMAG) algorithm, taking the complex OCT signal as input. In such images (Fig. 1b), signal from static tissue is, in contrast to signal from the capillaries, already significantly attenuated. One can however still observe some residual signal from static tissue. We therefore assume that this image corresponds to the additive mix (I ) of a fraction of the structural intensity signal (ωI ) and the l u pure flow signal (x), i.e. I = x + ωI . Because one can assume the desired flow signal, x, with all structure removed, to l u look fundamentally different from the structure image, I , ω can be solved by minimizing the square of the normalized 2 u cross-correlation between I and I as argmin γ (I , I - ωI ). Its explicit solution is ω = cov(I , I ) / var(I ), which can u l ω u l u u l u be rapidly computed. TM Results: We have tested our method on scans from CIRRUS HD-OCT 5000 with AngioPlex OCT Angiography (ZEISS, Dublin, CA). Fig. 1a) shows the structural intensity B-scan I , b) the mixed structure-flow image I and c) the u l resulting flow image x. A clear improvement in static tissue suppression can be observed. Fig 2 shows OCTA en face projection images before (a) and after (b) the processing described above. A substantial contrast enhancement can again be appreciated. Further, a suppression of the horizontal lines in a) can be observed, which are a common OCTA artifact typically caused by eye motion. Conclusions: We present a practical and fast method to improve the contrast of retinal OCTA images. Testing on clinical data demonstrated superior signal suppression of static tissue. Fig. 1 a) structural B-scan; b) direct output of the OMAG algorithm; c) resulting flow image after proposed processing Fig 2 a) direct output of the OMAG algorithm; b) resulting flow image after proposed processing CONTROL ID: 3195664 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB049 TITLE: Quantifying Peripapillary Microvascular Changes with Increasing Severity of Diabetic Retinopathy: An Optical Coherence Tomography Angiography (OCTA) Study FIRST AUTHOR: Zihan Sun AUTHORS/INSTITUTIONS: Z. Sun, F. Tang, Z. Tang, K. Lam, R. Wong, J. Lok, S. Szeto, S. Ng, C. CHEUNG, Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Kowloon, HONG KONG| Purpose: To determine the association between vessel density (VD) of the radial peripapillary capillaries (RPC) and severity of diabetic retinopathy (DR). Methods: This prospective cross-sectional study included diabetic subjects with different stages of DR from a tertiary eye hospital in Hong Kong. DR severity was assessed by retinal specialists according to the international clinical diabetic retinopathy disease severity scale on dilated fundus examinations. Eyes with proliferative DR were excluded. All subjects underwent OCTA with a swept-source OCT (DRI-OCT Triton, Topcon, Inc, Tokyo, Japan). Slab of the RPC was extracted for image analysis. VD was measured by using an automated customized MATLAB program, with or without removal of large vessels. Linear mixed-effect model was used to determine the association bwtween VD and DR severity, adjusting for age, gender, duration of diabetes, axial length, HbA1c and inter-eye correlation. Results: A total of 298 eyes (97 without DR, 87 with mild NPDR, 87 with moderate NPDR, 27 with severe NPDR) of 173 subjects were included in the final analysis. The average VDs were 61.47%, 60.65%, 59.71%, 57.17% in groups of eyes without, with mild, moderate and severe NPDR, respectively. We found a significant correlation between DR 2 2 severity and VD in univariate (β=-0.011; P-trend<0.001; R =0.453) and multivariate (β=-0.009; P-trend=0.003; R =0.459) analysis. After removing large vessels, average VDs were 57.29%, 56.81%, 55.17% and 52.48% respectively. Associations between VD and DR severity remained statistically significant in both univariate (β=-0.013; P- 2 2 trend<0.001; R =0.483) and multivariate (β=-0.013; P-trend<0.001; R =0.494) models. Conclusions: VD of RPC is significantly associated with DR severity and the association become even stronger when large vessels are removed. A significant decline of VD of RPC with increasing DR severity implies its potential in monitoring disease progression in patient with diabetes. A-D: original images; E-F: binarization; I-L: denoising; M-P: quantification; VD_incLV: vessel density (large vessels included); VD_excLV: vessel density (large vessels removed). CONTROL ID: 3195585 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB050 TITLE: Relationship between intraocular pressure and peripapillary vessel density in treatment-naïve glaucoma patients from the African American Eye Disease Study FIRST AUTHOR: Grace Marie Richter AUTHORS/INSTITUTIONS: G.M. Richter, A. Nelson, R. Chang, B. Xu, A. Kashani, Ophthalmology, USC Roski Eye Institute, Los Angeles, California, UNITED STATES|Z. Chu, R. Wang, University of Washington, California, UNITED STATES|B. Burkemper, R. Varma, Southern California Eyecare and Vision Research Institute, California, UNITED STATES| Purpose: Perfused radial peripapillary capillaries (RPCs), as measured by optical coherence tomography angiography (OCTA), are reduced in glaucoma, and this relationship is proportional to the extent of nerve damage. We studied a group of treatment-naïve glaucoma patients from the African American Eye Study to determine the ocular factors associated with OCTA parameters. We hypothesized that higher intraocular pressure would be independently associated with reduction of the perfused RPCs. Methods: 6x6mm spectral domain OCTA scans centered on the optic nerve were obtained on participants 40 years and older from the African American Eye Disease Study (AFEDS). RPC vessel density (VD) was determined using custom software with exclusion of large vessels. Multivariate linear regression was used to identify ocular factors associated with RPC VD and flux. Candidate variables included: intraocular pressure (IOP), age, signal strength (SS), visual field mean deviation (MD), central corneal thickness (CCT), ocular perfusion pressure (2/3 mean arterial pressure minus intraocular pressure; OPP), mean retinal nerve fiber layer thickness (mRNFL), and axial length (AL). Results: 3840 eyes from 2025 participants received OCTA imaging. Of these, 1473 eyes from 1042 subjects had SS ≥ 7 out of 10 and good quality images based on a standardized image quality grading algorithm. Of these, 45 eyes from 36 patients were diagnosed with glaucoma and had never received treatment. 31 eyes from 31 patients were studied due to some missing data and inclusion of only 1 eye per participant. In this group, average RPC VD was 0.26 ± 0.079. The factors independently associated with RPC VD were: IOP (β=-.0026, p=.035), mRNFL (β=.0043, 2 p<.0001), and SS (β=.02; p=.043), with a model R of 0.83. Conclusions: From this group of treatment-naïve glaucoma patients from a population-based cohort, higher IOP was associated with reduced RPC VD, controlling for SS and mRNFL. These results support the idea that elevated IOP reduces the peripapillary microcirculation in glaucomatous eyes, and that this phenomenon may result from reduced autoregulation in glaucomatous eyes. (No Image Selected) CONTROL ID: 3193635 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB051 TITLE: Preferred OCTA Scanning Protocol for Glaucoma Discrimination FIRST AUTHOR: Ravneet Singh Rai AUTHORS/INSTITUTIONS: R.S. Rai, K. Lucy, N. Tracer, M. Wu, M. de los Angeles Ramos Cadena, A. Kokroo, S. Rathi, A. Madu, H. Ishikawa, J.S. Schuman, G. Wollstein, NYU Eye Center, NYU Langone Health, New York, New York, UNITED STATES|J. Jiménez-Román, G. Lazcano-Gómez, Glaucoma, Asociación Para Evitar la Ceguera en México I.A.P., Mexico City, CDMX, MEXICO|J. Wong Shin, Gangneung Asan Hospital, University of Ulsan, Gangneung, Gangneung, KOREA (THE REPUBLIC OF)|J. Wong Shin, S. Kyung Rim, Ophthalmology, College of Medicine, University of Ulsan, Asan Medical Center, Seoul, KOREA (THE REPUBLIC OF)| Purpose: OCT Angiography (OCTA) can be used to measure retinal vessel density (VD). These scans can be of various sizes and may be centered on the optic nerve head (ONH) or macula. In this study, we examined the glaucoma discrimination performance of VD using different scanning sizes and locations and compared it with the performance of conventional structural and functional biomarkers to identify the best glaucoma discrimination scanning protocol. Methods: 79 healthy and glaucomatous eyes (50 subjects) were included in the study. Subjects with diabetes, vascular disease, or who were using medications known to affect retinal thickness were excluded. 3x3 and 6x6mm ONH and macula OCTA images were obtained using Cirrus HD-OCT Angioplex (Zeiss, Dublin, CA). Global and sectoral VD was calculated using native software on the device. Area under the receiver operating characteristics (AUC) was used to determine the discrimination ability of VD, retinal nerve fiber layer (RNFL) thickness, rim area, cup- to-disc (C/D) ratio, ganglion cell inner plexiform layer (GCIPL) thickness, and visual field mean deviation (MD). Bootstrapping was used for comparison between the AUCs. Results: Subjects with glaucoma had statistically significantly different measurements than healthy individuals for all tested parameters except for the majority of macula VD (both 3x3 and 6x6 scanning sizes; Table). VD measurements that had the best glaucoma discrimination ability were acquired from the ONH from all sectors of the 3x3 scans and in the outer and full sectors in the 6x6 scans (Table). For these ONH parameters, no significant difference was detected from the best discriminating parameter (average RNFL and rim area). All macula VD measurements had significantly worse discrimination performance. Conclusions: Among VD scanning options, the ONH scans are the most suitable for glaucoma discrimination. However, the coarse sampling in the larger scan (6x6mm) reduces this capability inside and immediately adjacent to the ONH. Scan Location Scan Size Parameter

Age(\") 39.8 (14.2) Average RNFL Thickness 93.59 (9.07) (µm) Ave rage GCIPL thickness -80.09 (4.57) 66.33 (9.02) <0.001 0.91 0.55 (µm) Ave rage C/D ratio 0.54 0.74 <0.001 0.81 0.02 (0.39,0.65)* (0.65,0.82)* 2 Rim area (mm ) 1.30 0.83 <0.001 0.93 1.0 (1.1 5,1.38)1 (0.62,0.98)* MD(dB) -0.16 -4.46 <0.001 0.87 0.40 (-0.74,0.22)1 (-13.36,-2.08)1 Central VD (mm- 1) 5.23 (4.45) 1.23 (2.60) <0.001 0.81 0.06 Inner VD (mm-1) 19.88 (2.33) 16.01 (3.36) <0.001 Full VD (mm-1) 18.18 (2.39) 17.31 (18.04) <0.001 Central VD (mm- 1) 4.53 (4.12) 1.23 (2.36) 0.001 Inner VO (mm-1) 17.73 (2.13) 15.42 (2.77) 0.001 Outer VD (mm- 1) 18.62 (1.21) 17.06 (1.60) <0.001 Full VD (mm-1) 18.00 (1.15) 16.24 (1.56) <0.001 Central VD (mm- 1) 10.63 (4.56) 10.42 (4.52) 0.83 Inner VO (mm-1) 21.10 (2.18) 20.04 (2.27) 0.10 Full VD {mm-1) 19.60 (2.97) 18.93 (2.38) 0.20 Central VD (mm- 1) 8.08 (4.48) 8.65 (3.84) 0.56 Inner VO (mm-1) 16.96 (2.94) 16.96 (2.37) 0.46 Outer VD (mm- 1) 17.82 (1.52) 16.01 (2.32) <0.001 0.03 Full VD (mm-1) 17.36 (1.86) 16.03 (2.25) 0.005 0.71 0.006 • Median (25, 75 quartil es)

0 p value for comparison between healthy and glaucoma calculated from Wilcoxon Rank-Sum test.

••• RNFL as the reference since it has the highest AUC. CONTROL ID: 3195700 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB052 TITLE: The superficial peripapillary vascular layer is the most informative for glaucoma diagnosis – A multi-layer study based on OCT Angiography. FIRST AUTHOR: Danilo Andrade De Jesus AUTHORS/INSTITUTIONS: D. Andrade De Jesus, L. Sanchez Brea, S. Klein, T. van Walsum, Erasmus MC, Rotterdam, NETHERLANDS|J. Barbosa Breda, I. Stalmans, Department of Neurosciences, KU Leuven, Leuven, BELGIUM|L. Abegão Pinto, Visual Sciences Study Center, University of Lisbon, Lisbon, PORTUGAL|J. Barbosa Breda, Ophthalmology Department, Centro Hospitalar Universitário de S. João, Porto, PORTUGAL|I. Stalmans, Ophthalmology Department, University Hospitals Leuven, Leuven, BELGIUM| Purpose: To evaluate the ability of microvascular density obtained from OCT Angiography (OCTA) at different peripapillary layers to discriminate glaucoma from controls. Methods: OCTA 3x3mm optic disc centred scans were retrieved from 45 healthy controls and 97 glaucoma patients with the Cirrus 5000 HD OCT (Carl Zeiss, USA). Images were analysed with an in-house algorithm that measures circumpapillar microvascular density (cpmVD) after automatically removing the Optic Nerve Head (ONH) optically hollow and large retinal vessels from the superficial layer. The resulting mask was used to retrieve the cpmVD from all six layers available (superficial, deep, avascular, whole retina, choriocapillaris, and choroid – see Fig. 1). A linear support vector machine (SVM) model with 5-fold cross validation was implemented. The area under the receiver operating characteristic curve (AUC) was computed for the cpmVD at each layer and combining multiple layers. A group comparison with the Kruskal-Wallis test followed by the Dunn test with Holm-Bonferroni correction for pairwise comparison was also performed to infer the ability of each layer to discriminate different glaucoma severity levels. Results: All layers showed significantly different cpmVD values between glaucoma and controls (p<0.001), with the exception of the avascular layer. The highest AUC was observed for the superficial layer (0.88±0.03; Fig. 2a). In a multi-layer analysis, the superficial and choriocapillaris layers combined showed the highest AUC (0.92±0.02; Fig. 2b). Regarding glaucoma severity, all layers, except for the avascular, showed a significant cpmVD change between levels of severity (p<0.05). However, only the superficial layer presented a significant difference between all groups in a pairwise comparison (p<0.05). Conclusions: The OCTA superficial layer is the best at discriminating healthy subjects from glaucoma patients and between glaucoma severity levels. Moreover, a combination of superficial and choriocapillaris layers seems to yield an even higher accuracy. Automatic detection of the microvasculature area (green) for the superficial layer and use of the resulting mask in all remaining layers. The blue disc covers the ONH optically hollow area. ROC curve for the SVM model based on each layer (a) and based on the superficial cpmVD and the combination of superficial with choriocapillaris cpmVDs (b). CONTROL ID: 3195411 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB053 TITLE: Retinal Venous Occlusion Leads to Multiple Changes in Retinal Vessel Pulse Amplitude Maps. FIRST AUTHOR: Bill Morgan AUTHORS/INSTITUTIONS: B. Morgan, Y. Khoo, A. Rahman, B. Chandrakumar, I. McAllister, F. Chen, D. Yu, Lions Eye Institute, University of Western Australia, Nedlands, Western Australia, AUSTRALIA| Purpose: To use retinal and optic nerve pulse amplitude mapping in central retinal vein occlusion (CRVO) and hemiretinal vein occlusion (HVO) subjects to test whether significant differences in pulse amplitudes and their distribution may exist. Methods: Video photography of retina allied with ophthalmodynamometry was used to measure photoplethysmography (PPG) estimates of blood column pulse amplitude in time with the cardiac cycle using harmonic analysis at each 5 x 5 pixel location (approximately 30 x 30 micron loci). Maps of pulse amplitude values were constructed allowing co-localisation with vascular anatomic features. CRVO, HVO, and normal eyes were examined. The relationships between pulse amplitude, diagnosis and distance from the optic disc centre were analysed in retinal vessels using linear mixed modelling. Results: 82,685 vessel measurements from 26 eyes of 15 subjects (8 male, mean age 68 +/- 16 years) were examined, subdivided into 14 with CRVO, 4 with HVO, and 8 normal. Normal subjects had mean 8.5 units retinal venous pulse amplitude with a rapid drop (attenuation) in amplitude with distance from disc centre (-6 units/disc diameter) and a significant increase in mean amplitude of 0.031 units/mmHg as intraocular pressure rose (p=0.0000, see figure). CRVO and HVO veins had significantly lower mean pulse amplitude (2.5 units, p<0.0072) with an increase with distance from the centre unlike normals (p=0.0000, see figure) and no increase in amplitude with intraocular pressure elevation. HVO amplitudes actually fell with increased IOP (0.04 units/mmHg, p=0.0000). Pulse amplitudes in the retinal arterioles were significantly greater (p<0.0120) and attenuated more rapidly (p=0.0000) in normals compared to HVO and CRVO. Conclusions: Retinal and arterial pulse amplitudes are significantly reduced in CRVO and HVO with different amplitude relationships to location along the vessel and with intraocular pressure. These features can be seen on amplitude heat maps co-localised with vessel anatomy. Normal and CRVO retinal video frame (A, B) and pulse amplitude maps (C,D) with pules amplitude scale. CONTROL ID: 3188564 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB054 TITLE: Quantification of choriocapillaris with OCTA: lessons learned FIRST AUTHOR: Zhongdi Chu AUTHORS/INSTITUTIONS: Z. Chu, R. Wang, Department of Bioengineering, University of Washington, Seattle, Washington, UNITED STATES|G. Gregori, P.J. Rosenfeld, Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine , Miami, Florida, UNITED STATES| Purpose: To demonstrate necessary cautions when quantifying choriocapillaris (CC) using optical coherence tomography angiography (OCTA). Methods: A retrospective, observational, cross-sectional case series. 3x3 mm and 6x6 mm macular OCTA scans of 6 eyes from 6 normal subjects and 6 eyes from 6 subjects with drusen secondary to AMD were obtained. The CC slab was extracted and the CC flow deficits (FDs) were segmented with two previously published algorithms: fuzzy C- means approach (the FCM method) and Phansalkar’s local thresholding (the Phansalkar method). Five radius options were used to investigate how the radius selection has an effect on the FD segmentation when using the Phansalkar method. FD density (FDD), mean FD size (MFDS), FD number (FDN), FD area (FDA) and inter-capillary distance (ICD) were calculated for comparison. Repeatability was assessed as coefficient of variation (CV) and Pearson’s correlation analysis was conducted. Results: 3x3 mm scans resulted in higher repeatability (CV: 0.57%-11.42%) compared to 6x6 mm scans (CV: 1.19% - 41.14%). For the Phansalkar method, larger radius option resulted in higher repeatability. ANOVA tests resulted in significant differences (p<0.001) among the FCM method and the Phansalkar method with different radius options, true for all CC metrics and scan sizes investigated. In 3x3 mm scans, significant correlation was found between the FCM method and the Phansalkar method for all quantitative CC metrics other than FDN (all p<0.001, 0.90

C-G: color coded FD segmentation map (red) by the Phansalkar method overlaid with OCTA CC image

(grey) and relative large retinal vessels (yellow), with 2-pixel radius, 5-pixel radius, 12-pixel radius, 15-p ixel radius, and 30-pixel radius, respectively. H: 6x6 mm OCTA CC en face image; I: color coded FD segmentation map (red) by the FCM method overlaid with original CC image (grey) and relatively large retinal blood vessels (yellow); J-N : color coded FD segmentation map (red) by the Phansalkar method overlaid with OCTA CC image (grey) and relative large retinal vessels (yellow), with 2-pixel radius, 5-pixel radius, 12-pixel radius, 15-pixel radius, and 30-pixel radius, respectively. CONTROL ID: 3188001 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB055 TITLE: Automated detection of shadow artifacts in OCTA FIRST AUTHOR: Acner Camino AUTHORS/INSTITUTIONS: A. Camino, Y. Jia, J. Wang, Q. You, X. Wei, L. Liu, D. Huang, Casey Eye Institute, OHSU, Oregon, UNITED STATES|Y. Jia, J. Wang, Department of Biomedical Engineering, OHSU, Oregon, UNITED STATES| Purpose: Artifacts in optical coherence tomography angiography (OCTA) can be caused by motion, projections of superficial blood flow or shadows from opaque objects anterior to the retina (e.g. vitreous floaters, pupil boundary). While motion and projection artifacts have been studied extensively, shadow artifacts have been overlooked thus far. We aim at detecting shadow artifacts to exclude them from quantitative OCTA metrics. 2 Methods: 3×3 and 6×6 mm macular OCTA scans were acquired by a spectral-domain OCT system (AngioVue). Retinal layers were segmented by a directional graph search algorithm implemented in a customized image processing platform. Background signal from avascular tissue was removed by an iterative, regression based bulk motion subtraction algorithm. A machine learning ensemble method was trained with OCTA of healthy subjects where shadows were manufactured by placing a PLA filament between the cornea and the instrument. Labeling of the shadow positions used in training was based on variation of the local vessel density compared to a scan of the same subject under optimal imaging conditions. The features used for shadow classification were local flow index, low projected reflectance in both the inner and outer retina, and low standard deviation of the reflectance in the retinal slab. Software performance was evaluated in scans acquired from healthy subjects by progressively increasing signal attenuation with neutral density filters (NDF). Results: The sensitivity and specificity of the software classification were 91.6% and 86.9%, taking the manual segmentation of one expert grader as ground truth. Another expert grader performed with a sensitivity of 87.2% and a specificity of 93.3% with respect to the same reference. Shadows were detected on OCTA scans of intermediate uveitis and diabetic retinopathy (Fig.1). Vessel density of participants in the NDF experiment was independent of signal strength after shadow exclusion (Fig. 2). Conclusions: Areas of unreliable OCTA signal due to shadows were successfully detected in diabetic retinopathy and uveitis. As OCTA becomes a common imaging modality used in the ophthalmic practice, exclusion of these artifacts will help to improve the confidence with which OCTA is used to diagnose and evaluate retinal diseases. This is important as older patients often have cataracts, vitreous opacities, small pupil, dry eye – conditions in which shadows are difficult to prevent during imaging sessions. En face OCTA (SVC) OCT A-line average projection Shadow overlapped on OCTA

Uveitis

Diabetic retinopathy

Fig. 1. Automatic detection of shadow artifact examples on uveitis and diabetic retinopathy patients. SVC - Superficial vascular plexus >. .f"' (A) -;;; 60 (B) . . -~60 (C) .. ~60 Cl 40 '------>-- L--+--~-----~ > 70 80 40 60 80 40 60 80 SSI SSI SSI

Fig. 2 Evaluation o flh..: shagr.,o;si,·elv incr"3•ing opltcal signal aumualion wilb n<"UU-<11 dmsity fihers {J\UI·} {A) Scam acquilef' show iude~nd~nce of Table. Average vessel length (mm) calculated usingHessian and CNN processing

CONTROL ID: 3195768 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB059 TITLE: Analysis of Vascular Changes after Proton Irradiation using OCT-A in Patients with Uveal Melanoma FIRST AUTHOR: Gary Lamoureux AUTHORS/INSTITUTIONS: G. Lamoureux, A. Lane, M. Oxenreiter, M.B. Aronow, I.K. Kim, E.S. Gragoudas, Ocular Melanoma Center, Massachusetts Eye and Ear, Harvard Medical School, Massachusetts, UNITED STATES|J. Wang, J.B. Miller, Retina Service, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, UNITED STATES|A.V. Trofimov, H.A. Shih, Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, UNITED STATES| Purpose: To evaluate changes in macular vasculature within 2 disc diameters (DD) of the fovea using optical coherence tomography angiography (OCT-A) after proton beam irradiation (PBI) in patients with uveal melanoma. Methods: This cross-sectional study included patients with small-medium tumors (height ≤5mm and largest basal diameter ≤15mm) located within 2DD of the fovea or optic disc who completed OCT-A scans after PBI. The majority (53.8%) of post-treatment OCT-A scans were completed within 3 years (±6 months) after treatment (mean time from PBI to OCT-A: 3.5 years; range: 2.1 - 7.0 years). Patients with tumors abutting the optic disc and involving the fovea were excluded. Automated measures of vessel density (VD) in both the superficial capillary plexus (SCP) and the deep capillary plexus (DCP) in the parafoveal area (area measuring 3 mm around fovea) were obtained using the built-in angioanalytics software on the Optovue Avanti. Radiation dose to the fovea was obtained using treatment planning software (EyePlan version 3.06). Spearman correlation coefficient and Wilcoxon rank-sum test were used to evaluate relationships between tumor location, radiation dose, vascular changes and visual acuity (VA). Results: Thirteen patients (62% male; median age: 65.5 years) were included in this study; 10 (77%) had tumors located within 2 DD of the fovea and 9 (69.2%) had baseline visual acuity (VA) of 20/40 or better. The mean SCP VD in the parafoveal region for tumors located within 2DD of the fovea was 37.7% compared to 47.4% for tumors further away from the fovea (P=.01). Differences in mean DCP VD in this region were not significantly different (42.5% vs. 46.8% for tumors <2DD and > 2DD from fovea respectively, P=.50). Median radiation dose to the fovea was 46 Gy (RBE), and inversely related to both SCP VD (r=-.89, P<.001) and DCP VD (r=-.53, P=.06). There was a trend toward reduced SCP VD in those with worse VA compared to those with VA of 20/40 or better (34.4% v. 42.6%, P=.08). Conclusions: These data suggest that tumors in close proximity to the fovea and high foveal radiation dose are associated with decreased VD in both the superficial capillary plexus and deep capillary plexus in the parafoveal region of the retina. Larger studies are needed to further explore the relationship between these OCT-A metrics and vision loss after radiation therapy. (No Image Selected) CONTROL ID: 3189031 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB060 TITLE: Optical Coherence Tomography Angiography for Evaluation of Reperfusion following Pterygium Surgery FIRST AUTHOR: Jodhbir Mehta AUTHORS/INSTITUTIONS: J. Mehta, K. Devarajan, M. Ang, T. Tan, Y. Liu, Cornea and Tissue Engineering and Stem Cell, Singapore Eye Research Institute, Singapore, SINGAPORE| Purpose: To describe the use of optical coherence tomography angiography (OCTA) to quantitatively monitor the conjunctival graft revascularization after pterygium excision and conjunctival autograft (CAG) transplantation. Methods: Ten patients undergoing pterygium excision and femtosecond laser-assisted CAG transplantation were included. OCTA was performed at 1 week, 1 and 3 months postoperatively at the CAG transplantation site and harvested area. The vessel density at three different depths: conjunctival epithelium or CAG epithelium, conjunctival stroma or CAG stroma, and episclera, was evaluated and quantified. The revascularization rate was assessed and correlated with the postoperative CAG thickness. Results: No intraoperative and postoperative complications occurred. Reperfusion of the CAGs was observed at 1 week, and early reperfusion within the first month accounted for more than half of graft revascularization. The vessel regrowth density was 9.6±2.6 % and 11.1±2.8 % between 1 week and 1 month, and was 9.8±1.8 % and 11.9±1.9 % between 1 and 3 months, at the CAG and underlying episcleral levels, respectively. All the CAGs were well-perfused at 3 months. The vessel regrowth density was significantly and strongly correlated with the changes of CAG thickness in a negative relationship (γ=-0.94, P=0.019). At the harvested site, the vascular network of episclera was not affected, and the conjunctival vascularization was restored at 1 month. Conclusions: OCTA is a promising tool to evaluate the vascularization or revascularization of conjunctiva, conjunctival graft and episclera, in a quantitative and serial manner, helping in diseases diagnosis and treatment monitoring. The graft revascularization rate was predictive of postoperative graft deswelling. (No Image Selected) CONTROL ID: 3193646 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB061 TITLE: Spatial correlation of microaneurysms detected by fluorescein angiography aligned with microdomains of macular ischemia delineated by optical coherence tomography angiography in patients with diabetic retinopathy FIRST AUTHOR: Ayman G. Elnahry AUTHORS/INSTITUTIONS: A.G. Elnahry, Ophthalmology, Cairo University, Cairo, EGYPT|D.J. Ramsey, Ophthalmology, Lahey Hospital & Medical Center, Peabody, Massachusetts, UNITED STATES|D.J. Ramsey, Ophthalmology, Tufts University School of Medicine, Boston, Massachusetts, UNITED STATES| Purpose: To characterize the relation between microaneurysms (MAs) identified by fundus fluorescein angiography (FFA) and areas of diabetic macular ischemia (DMI) delineated by optical coherence tomography angiography (OCTA) using a commercially available alignment software. Methods: Patients with diabetic retinopathy were recruited from Cairo University clinic. FFA was performed with a TRC 50DX (Topcon, Tokyo, Japan) or Spectralis HRA2 (Heidelberg Engineering, Heidelberg, Germany). OCTA was performed by an Optovue RTvue XR Avanti (Optovue, Fremont, CA) using the 6x6 mm macula scan. All FFA images were automatically cropped and aligned with their respective OCTA image using the i2k Align Retina software (Dual- Align, Clifton Park, NY). All images were de-identified to mask grader. The foveal avascular zone (FAZ) and areas of DMI were manually delineated on OCTA images and MAs marked on the corresponding FFA images then images were overlaid using ImageJ (NIH, Bethesda, MD) and analyzed. Results: Nine eyes of 7 patients with DMI were included. The average number of MAs marked in FFA images was 110 ± 29. Only 32.8% of superficial capillary plexus (SCP) was ischemic, yet 61.7% of MAs were associated with such areas, while only 23.4% of the deep capillary plexus (DCP) was ischemic, yet 45.3% of MAs were associated with such areas. As the area of DMI increased so did the total number of MAs associated with both the SCP (R = 0.842, p<0.01) and DCP (R = 0.766, p<0.05) ischemia. About 75% of these MAs bordered ischemic areas (within ±50um); only a minority were localized within ischemic regions. The number of MAs surrounding the FAZ (5.27 ± 3.15 MAs/mm2) was similar to other areas of macular ischemia (5.79 ± 1.04 MAs/mm2 for SCP and 6.53 ± 2.58 MAs/mm2 for DCP, n.s.). As the FAZ increased, so did the number of associated MAs (R = 0.836, p<0.01). Conclusions: MAs seen on FFA are strongly associated with the borders of areas of DMI on OCTA including the FAZ. Although more MAs are found in relation to DMI in the SCP, the concentration of MAs associated with DMI in the DCP was greater. Overall, the rate of MA association was twice that expected based on area measurements alone. Few MAs are present inside low-flow regions likely because capillary loss would be associated with regression of MAs. (No Image Selected) CONTROL ID: 3194211 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB062 TITLE: Retinal Peripapillary Perfusion Changes in Thyroid-Related Orbitopathy with Asymmetric Proptosis without Compressive Optic Neuropathy Detected Using Optical Coherence Tomography Angiography FIRST AUTHOR: Alexander Pinhas AUTHORS/INSTITUTIONS: A. Pinhas, J.S. Andrade Romo, G. Lynch, D.B. Zhou, M.V. Castanos Toral, P.A. Tenzel, A. Barash, D. Della Rocca, R. Della Rocca, T.Y. Chui, R.B. Rosen, H.S. Reddy, Department of Ophthalmology, New York Eye and Ear Infirmary of Mount Sinai, New York, New York, UNITED STATES|A. Pinhas, Department of Ophthalmology, State University of New York Downstate Medical Center, Brooklyn, New York, UNITED STATES|G. Lynch, D.B. Zhou, Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, New York, UNITED STATES| Purpose: Thyroid-related orbitopathy (TRO) causes hemodynamic changes in the orbit from compression and congestion that may cause ischemic damage to the optic nerve. This study explores the retinal peripapillary perfusion in TRO eyes using optical coherence tomography angiography (OCTA). Methods: We reviewed 4.5x4.5mm en face “RPC layer” OCTA scans of 6 TRO patients with asymmetric proptosis without compressive optic neuropathy and a clinical assessment score of less than 3 (12 eyes, mean age 41 years, range 23-69 years), and 133 healthy controls (133 eyes, mean 41.5 years, range 11-83 years). The 6 relatively less proptotic TRO eyes were grouped into Group A, while their relatively more proptotic fellow eyes (on average 1.67mm more proptotic) were grouped into Group B. A novel algorithm from our laboratory was used to automatically segment non-capillary from capillary peripapillary blood vessels and to calculate respective perfusion densities. ANOVA and pairwise Tukey-Kramer testing were performed to detect differences in perfusion densities between Groups A and B and the control group. Results are presented as average ± standard deviation. A p value of less than 0.05 was considered statistically significant. Results: Noncapillary peripapillary perfusion density in TRO eyes was found to be significantly decreased compared to healthy controls (Group A 17.2±4.9% and Group B 13.6±3.4% vs. controls 21.5±4.7%; p=0.001). A greater decrease was noted in eyes with relatively more proptosis than in fellow eyes, however this difference did not reach statistical significance (Group A vs. Group B; p=0.12). Peripapillary capillary perfusion densities in all 3 groups (Group A 42.2±1.9% vs. Group B 42.2±2.0% vs. controls 42.5±1.5%) showed no significant difference. Clinical markers were not different between Groups A and B. Conclusions: TRO eyes with more proptosis showed a greater decrease in noncapillary peripapillary perfusion, while capillary perfusion and routine clinical markers were not significantly affected. Intraorbital compression on the central retinal blood vessels in TRO may be responsible for these findings. OCTA provides a new sensitive tool for monitoring the subclinical hemodynamic effects on the optic nerve head in TRO, and may be useful for earlier diagnosis and assessment of treatment response. (No Image Selected) CONTROL ID: 3194309 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB063 TITLE: Microvascular reactivation in the glaucoma post-operative period FIRST AUTHOR: Ana MIGUEL AUTHORS/INSTITUTIONS: A. MIGUEL, A. Herman, J. legeai, C. Martin, Ophthalmology, Polyclinique de la Baie, Avranches, France, Granville, FRANCE|A. MIGUEL, A. Silva, CINTESIS, Faculty of Medicine of Oporto, Portugal, Porto, PORTUGAL| Purpose: Vessel density (VD), as assessed by OCT Angiography (OCTA), is thought to be relevant in the prognosis of glaucoma. We intended to verify whether a rapid decrease of intra-ocular pressure (IOP) during the early post- operative period of glaucoma surgery was accompanied by an increase in VD. Methods: We present an observational prospective clinical study of patients with increased intra-ocular pressure shortly after glaucoma surgery (from day 0 to week 6), in which we performed an OCTA, before and 15 minutes after normalization of IOP (through laser suture lysis, removal of releasable sutures or goniopuncture). We included patients from April to September 2018, previously submitted to glaucoma surgery, presenting for the routine post- operative consultation with IOP of 21mmHg or higher, who accepted to participate and to sign consent (we had local Ethics Committee approval). Other ophthalmological or systemic pathologies were excluded. We performed an OCTA (Triton, Topcon®) with scans of 4.5 x 4.5mm centered in the optic disc, before consultation and after the manoeuvre to decrease the IOP; fifteen minutes later, IOP was measured and a new OCTA was performed. Results: Four patients (3 males, 1 female, 15 to 71 years old) were included in our analysis. We have observed a rapid increase in vascular density, as detected by OCTA, after normalisation of intra-ocular pressure (through goniopuncture after sclerectomy, and suture laser lysis or removal of releasable suture after trabeculectomy) in the early post-operative period (in figure 1, the VD is represented by a colour scale, in which blue and dark colours represent low VD; and yellow, orange and red colours represent high VD; in panel A before, and in panel B after these manoeuvres; in patients 1 to 4). Conclusions: In all cases, a rapid decrease in intraocular pressure after each of these manoeuvres was associated with a marked improvement in whole-image optic disc VD. These preliminary results corroborate the dynamic nature of vessel density in glaucomatous eyes. We introduce a new concept: rise-up, in which a rapid decrease in intra- ocular pressure is associated with a rapid increase in the whole-image optic nerve vessel density. To our knowledge, we are the first to identity an immediate substantial increase in the vessel density, in the early glaucoma post- operative period. Further prospective studies with a larger sample are welcomed. lA 1B 2A 2B

3A 3B 4A 4B CONTROL ID: 3195338 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB064 TITLE: Multi-acquisition averaging optical coherence tomography angiography for diabetic retinopathy FIRST AUTHOR: Arman Athwal AUTHORS/INSTITUTIONS: A. Athwal, M. Heisler, M. Ju, M. Beg, M.V. Sarunic, Faculty of Applied Science, Simon Fraser University, Delta, British Columbia, CANADA|S. Karst, E.V. Navajas, Department of Ophthalmology and Visual Sciences, University of British Columbia , British Columbia, CANADA| Purpose: The Deep Capillary Plexus (DCP) is thought to be the region of initial insult in Diabetic Retinopathy (DR). However, single frame OCT-A images of the DCP are often insufficient for robust quantification of this layer. The purpose of this study is to investigate the qualitative and quantitative improvements in OCT-A images of the DCP in DR eyes as a result of serial acquisition and averaging. Methods: Ten serially acquired OCT-A 2x2mm images centered at the fovea were acquired from twelve DR eyes (ranging in severity from mild non-proliferative DR to proliferative DR) using a swept-source 1060nm OCT clinical prototype. For each volume, the enface images of the superficial, deep, and summed layers were divided into micro- saccade free strips, which were then registered to each other and averaged. This algorithm was also applied on ten 1x1mm images acquired with our Adaptive Optics OCT-A system and compared to one 3x3mm scan acquired the same day with a Zeiss PLEX Elite OCT-A, the latter of which features hardware-based motion tracking. All imaging was performed with REB approval. Results: A comparison of the single frame and averaged OCT-A images is shown in Fig. 1. The average SNR of the superficial plexus angiograms increased from 18.81 ± 2.50 dB in the single frame images to 24.23 ± 3.32 dB in the averaged images. For the DCP the average SNR increased from 17.32 ± 1.51 dB in the single frame images to 23.91 ± 2.84 dB in the averaged images.Fig. 2 illustrates the single frame and averaged AO-OCT-A images in comparison with the corresponding region within the PLEX Elite OCT-A image. After averaging, regions containing microaneurysms gain enhanced contrast to the background vasculature and are more readily identifiable. Conclusions: Multi-acquisition and averaging of OCT-A and AO-OCT-A images results in qualitative and quantitative improvement in images of DR eyes, especially for the DCP. These methods make quantification of retinal vascular features more reliable, thereby facilitating investigation of their roles in the early pathophysiology of diabetic retinopathy. All Layers Superfic:lal Ple

Fig. 1. Single fram e images vs. averaged images (whole retinal thickness, superficial and deep pleX1Js] for a subj ect with DR. Single Frame AO Averaged AO ~ > <.-· .- i t,(;;/,""-{. : ...... ,__ .' t.,.,.. . -,"t" ;.. -I 1,' ' ,., '. 1'.,, •..,, • .' ' --.. -,. ...' ~ • I A '.1-- / .', ,1.:_ .s- • •,J.' • • ' · -r.. "' .· ,-,., .:~··~· '\ - -·;....: .. ·. - ~ . .. _,.---, . -.--·\··.. ~. " ·~ ·-\ ....• r ··~·-""'..• • • • ·- - Fig. 2. Single fram e OCT-A and AO-OCT-A images vs. averaged AO-OCT-A image for a subject with DR. CONTROL ID: 3195781 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB065 TITLE: Hypertension Control Screen Using OCT-Angiography FIRST AUTHOR: Rebecca Zeng AUTHORS/INSTITUTIONS: R. Zeng, J. Wang, R. Silverman, J.D. Diaz, J.B. Miller, Ophthalmology, Massachusetts Eye & Ear Infirmary, Boston, Massachusetts, UNITED STATES|R. Zeng, Boston University School of Medicine, Boston, Massachusetts, UNITED STATES|R. Silverman, Tufts University School of Medicine, Massachusetts, UNITED STATES| Purpose: Systemic hypertension (HTN) has been associated with retinal vasculature changes, such as focal arteriolar narrowing and microaneurysms. While optical coherence tomography-angiography (OCT-A) is a quick and non- invasive imaging device, little has been done to examine the effects of systemic hypertension on the retinal vasculature by OCT-A. Herein, we propose a study to evaluate the retinal vasculature and vasoreactivity of systemic hypertensive patients with OCT-A. Methods: Prospective, observational, IRB-approved study of patients with HTN. We excluded patients with chorioretinal diseases and systemic diseases that might affect OCT-A metrics. All eyes were imaged with the Zeiss Angioplex OCT. After a baseline OCT-A was taken, patients were asked to hold their breath for 30 seconds before capturing another OCT-A. Vessel density (VD) and skeletonized vessel density (SVD) of the superficial and deep capillary plexus and choriocapillaris in the macula and optic disc were calculated using ImageJ. A multi-level mixed model tested the association between systemic hypertension and retinal vasculature changes, adjusting for age, sex and hypertensive medications. Results: We imaged 9 eyes of 6 patients with HTN and 19 eyes of 12 aged-matched control eyes. We only identified a statistically significant increase in hypertensive patients compared to controls in the VD and SVD of the superficial capillary plexus of the optic nerve (p < 0.05). We found no other significant differences in the OCT-A metrics with or without breath-holding. Conclusions: Hypertension was associated with increases in the optic disc VD and SVD. These early results of a small study suggest that there may be OCT-A biomarkers of hypertension control. We did not find breath holding to be a sufficient stressor to bring on additional hypertensive changes. Larger studies are needed to investigate the impact of hypertension severity. (No Image Selected) CONTROL ID: 3195863 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB066 TITLE: Macular Vessel Density following Retinal Detachment using Swept Source OCT-Angiography FIRST AUTHOR: Mohammad Dahrouj AUTHORS/INSTITUTIONS: M. Dahrouj, D. Diaz, A. Marmalidou, R. Katz, J.B. Miller, Ophthalmology , Mass. Eye and Ear/Harvard Medical School , Boston, Massachusetts, UNITED STATES| Purpose: To evaluate whether optical coherence tomography angiography (OCTA) can detect any changes in superficial and deep vessel density changes at the level of the macula in patients with retinal detachment within 5 months following surgical repair. Methods: A retrospective study included 42 eyes of 21 patients with a history of retinal detachment. Patients were imaged with the Topcon Triton Swept Source Optical Coherence Tomography Angiography (OCT-A) system between 3-6 months following surgical repair. A 3×3 mm area, centered on the fovea, was scanned for all the study eyes. Using image J software, the superficial and deep retinal vessel densities were measured in the retinal detachment eyes and compared to the normal fellow eye. The vessel densities were evaluated to in terms of their impact on visual acuity. Results: Five month following surgical repair, the superficial vessel density in eyes with retinal detachment eye was 28.39 ± 3.7 % and 27.02 ± 4.05 % in normal control eyes. The deep vessel density in eyes with retinal detachment was 29.16 ± 1.86 % compared to 28.20 ± 3.17 % in normal control eyes. There was no statistical significance between the two groups. We further stratified by status on retinal detachment and compared the data of mac-on (n=5) vs. mac off (n=16). The superficial vessel density in eyes with mac-on retinal detachment eyes was 30.31 ± 2.45 % compared to 27.79 ± 3.8 % mac-off retinal detachment. There was no statistical significance between the two groups. Conclusions: Five months following surgical repair, there appear to be no statistically significant difference in the superficial and deep retinal vessel densities in eyes with retinal detachment compared to normal fellow eyes. Although there seem to be a trend of higher superficial vessel density in mac-on RD compared to mac-off RD, the difference was not statistically significant. Larger studies and longer follow up following repair are required to further assess for vessel density changes following retinal detachment. (No Image Selected) CONTROL ID: 3195730 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB067 TITLE: Hybrid three dimensional modeles recunstructed from swept-source OCT angiography FIRST AUTHOR: Tetsuju Sekiryu AUTHORS/INSTITUTIONS: T. Sekiryu, Y. Sugano, Ophthalmology, Fukushima Medical University, Fukushima, JAPAN|M. Okamoto, S. Eifuku, Systems Neuroscience, Fukushima Medical University, Fukushima, JAPAN| Purpose: 3D image analysis may be beneficial to analyze a detail of the choroidal vasculature. Although a whole vascualar structure must be delineated for this purpose, commercially aveilable OCT angiography could not detect a whole choloidal vessel. OCT angiography provide two types of the vessel images in the choroid, i.e. heigh intensity flow information in OCT angiography (OCT-A) and silhouette images of the large vessels in stractural OCT (OCT-S). We attempted to make hybrid 3D models of the choroidal vessel from two different types of vessel images and examined the characteristics of the 3D models. Methods: Design: Prospective observational case series. Subjects:20 eyes of 20 healthy individuals The images were taken by using swept source (SS)-OCT (Elite 9000, Zeiss, Carl Zeiss Meditec, Dublin, California, USA). 6 x 6 mm square images of OCT-S and OCT-A were exported separately as stack images. A OCT-S image stack was inverted to white vessel images to black background. Vessel images of OCT-A and inverted OCT-S were segmented and visualized three dimensionally by using open-source software 3D slicer, which was an open-source software platform for biomedical research (ver. 4.10; https://www.slicer.org/). Vessel volume of the choroid was measured. Similarity of segmented choroidal vessel between OCT-S and OCT-A was calculated. Results: The ages of the subject was 28 ± 4.3 (mean ± SD). The axial length was 24.6 ± 1.0 mm.The choroidal vessel was successfully segmented and visualized three dimensionally in all eyes. The vessels visualized from OCT-A arranged surround a large choroidal vessels visualized from OCT-S. Total volume of the choroidal vessel at the fovea 3 the choroid was 3.01 mm , which decreased with the axial length.The vessel ratio to the volume of the choroidal tissue was 40%. Of the total vessel volume, 73% were vessels visualized from OCT-S and 27% were vessels visualized from OCT-A. The mean of Dice similarity coefficient between the vessel volume from OCT-S and OCT-A was low (0.05 ± 0.02). Conclusions: More than 40% of the choroidal tissues at the fovea may be composed of blood vessels in the hybrid 3Dmodel in healthy subjects.The choroidal vessel volume calculated from the silhouette image of OCT-S may be 27% or more less than the actual vessel volume. (No Image Selected) CONTROL ID: 3195891 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB068 TITLE: Enhanced OCT angiography visualizations for choroidal neovascularization with improved Bruch’s membrane segmentation. FIRST AUTHOR: Jenwei Kuo AUTHORS/INSTITUTIONS: J. Kuo, W. Huang, C. Reisman, Topcon Healthcare Solutions, Oakland, New Jersey, UNITED STATES| Purpose: Visualizing optical coherence tomography (OCT) angiography (OCT-A) volumes with choroidal neovascularization (CNV) can be challenging. To enhance CNV visualization, an improved Bruch’s Membrane (BM) segmentation algorithm with different projection range and methodology is proposed. Methods: The choriocapillaris (CC) slab plays an important role in OCT-A visualizations of OCT images with CNV. Often, an OCT-A projection within a thin CC slab can be used to visualize CNV. To obtain reliable BM boundary segmentation results along the CC, an automated BM segmentation methodology is proposed. Firstly, edge detection techniques are applied to obtain candidate edge segments. Secondly, BM edge segments are determined by feature analysis, and the final BM result is obtained by 3D fitting. The obtained BM result borders the CC and can delineate that projection slab. We also have proposed to use dynamic thickness determined by the result of retinal pigment epithelium (RPE) and BM to enhance the contrast in CNV areas. Different projection techniques are also applied to further emphasize the CNV area. Results: The improved BM segmentation algorithm was tested on three 3mmx3mm (320 a-lines x 320 b-scans) and two 6x6 (512x512) macula OCT-A scans acquired by 1050nm wavelength swept-source OCT (DRI OCT Triton, Topcon Corp., Tokyo, Japan). Comparing to manual segmentation results, signed differences (in micron) in 3x3 and 6x6 images are 6.15 ± 8.12 and -1.44 ± 7.75, respectively. Figure 1(a) shows the mean projection within a slab consisting of the CC only. The vessel structure on the left is not clear. Dynamic projection range can help increase the contrast (Fig. 1(b)). Max projection can further highlight the vessel structure (Fig. 1(c)). Figure 1(d) shows the result of integration-based projection where the CNV area is emphasized. Conclusions: An automated BM segmentation routine is proposed to provide reliable BM boundary to delineate the CC. Combined with dynamic projection range, the visualization of CNV area can be further improved for clinical interpretation. OCT-A visualization with improved BM segmentation algorithm. (a) Mean projection within a slab consisting of the CC (BM to BM plus 10.4 microns). (b)(c)(d) are obtained by dynamic projection range from the middle of RPE and BM to BM plus 10.4 microns. (b) Mean projection. (c) Max projection. (d) Integration projection. CONTROL ID: 3175664 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB069 TITLE: OCT angiography: Measurement of retinal macular microvasculature with Spectralis II OCT angiography – reliability and reproducibility FIRST AUTHOR: Sami Hosari AUTHORS/INSTITUTIONS: S. Hosari, C. Mardin, B. Hohberger, Eye Hospital, Friedrich-Alexander-University Erlangen, Erlangen, GERMANY| Purpose: Analysis of chorioretinal microvasculature is important in clinical all day life and research. Different Optical Coherence Tomography Angiography (OCT-A) tools with their own analysis software generate diverse data of OCT-A scans, emphasizing the importance of reliability analysis of each OCT-A device and analysis software. The aim of the present study was to investigate the reliability of macular microvasculature characteristics measured with OCT-A of Heidelberg OCT II with a custom-made quantification software (Erlangen-Angio-Tool (EA-Tool) version 1.0). Methods: Twenty-three eyes of 23 normal subjects were measured by OCT-A (Heidelberg OCT II Spectralis, Heidelberg, Germany). Repeated scans of the macula (size: 2.9 mm x 2.9 mm) including 3 retinochoroidal layers (superficial vascular plexus (SVP), deep capillary plexus (DCP), intermediate vascular plexus (IVP)) were performed with a B-scan thickness of 5.7 µm, separated into 12 sectors (s1-s12). Vessel density (VD) and foveal avascular zone (FAZ) area were calculated for each en face vascular layer, respectively. Reliability was shown by intraclass correlation coefficients (ICC). st Results: (1) Mean VD was in 32.28 ± 3 (SVP), 22.79 ± 3 (ICP) and 25.40 ± 3 (DCP) of the 1 and 32.62 ± 2 (SVP), nd 22.89 ± 3 (ICP) and 25.51 ± 3 (DCP) of the 2 scan. (2) Sector analysis yielded no significantly different VD of s1- st nd s12 between 1 and 2 scan of SVP, ICP, and DCP, respectively (p>0.05). (3) ICC of VD were excellent (s1, s2, s4, s9, s12 (SVP); s2, s4 (ICP); s3, s4 (DCP)) and good (s5-s8, s10 (SVP); s1, s3, s5, s8, s9, s11, s12 (ICP); s1, s2, s5, st nd s9, s11, s12 (DCP)). (4) Reliability of FAZ showed excellent ICC on 1 and 2 OCT-A scan in SVP, IVP, and DCP. st nd (5) No significant differences of FAZ area in SVP, ICP and DCP were observed between 1 and 2 scan (p>0.05). Conclusions: As OCT-A data of retinochoroidal microvasculature, quantified with the Erlangen-Angio-Tool, showed a good reliability, EA-Tool can be used as quantification software for analysis of macular vessel density and FAZ metrics in clinical use and research. Region of interest (i.e. macula) marked with an annulus, subdivided into 12 sectors of 30° (s1-s12). CONTROL ID: 3195139 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB070 TITLE: Spiral Scanning OCT Angiography FIRST AUTHOR: Michael Niederleithner AUTHORS/INSTITUTIONS: M. Niederleithner, M. Salas, R.A. Leitgeb, W. Drexler, T. Schmoll, Center of Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, AUSTRIA|T. Schmoll, Carl Zeiss Meditec, Inc., Dublin, California, UNITED STATES| Purpose: Traditional raster scanning in optical coherence tomography (OCT) puts a lot of strain on the galvanometer scanners as they must continuously accelerate and deaccelerate the scan mirrors. This limits the maximum scan rate at which they can be operated. For applications where very high scan rates are essential, e.g. confocal laser scanning microscopy and 4D intra-surgical OCT, scan patterns which avoid sudden changes in scan direction are preferred. Examples of such patterns include Lissajous and spiral scan patterns. We demonstrate the use of spiral scan patterns for OCT and OCT angiography (OCTA) imaging of the human retina. Methods: We developed a point scanning ophthalmic swept-source OCT (SS-OCT), with a 100 kHz akinetic swept laser source (Insight Photonics, Longmont, CO, 1060nm central wavelength, 70nm tuning range) where we can drive the galvanometer scanners with arbitrary scan patterns. Spiral patterns with different radii and sampling densities were pre-calculated. Patterns with constant number of A-scans per “circle” were used for OCTA to maintain a constant temporal sampling. For structural OCT imaging, the number of A-scans per arc was increased in order to maintain a constant spatial sampling. For both, the acquisition time for a full volume of 250,000 A-scans was approximately 2.5s. After acquisition, the A-scans were remapped to a rectangular grid using the A-scan locations recorded during acquisition. OCTA images were then generated by calculating the speckle variance between neighboring arcs. Results: We show OCT structural en face images (Figure 1) as well as OCTA en face images (Figure 2) of the human retina, acquired with a 1.5 mm diameter spiral scan pattern. Because different scan patterns were used for OCT structural and OCTA imaging, Figure 1 and 2 are showing slightly different region of interests on the retina. Conclusions: We demonstrated OCTA imaging with spiral scan patterns. This may be valuable for applications where the maximum acquisition rate is otherwise limited by the maximum scanner frequency of a raster scan, like for example intra-surgical 4D OCT. Next steps include increasing the field of view and the investigation of motion correction and eye tracking methods for non-raster scan patterns. Figure 1 En face projection of an OCT images of a healthy retina Figure 2 En face projection of an OCTA image of a healthy retina CONTROL ID: 3195677 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB071 TITLE: Imaging of vascular involvement in intraocular tumor using OCT and OCT angiography FIRST AUTHOR: Xiao Zhou AUTHORS/INSTITUTIONS: X. Zhou, H. Zhou, Z. Chu, Y. Cheng, Q. Zhang, R. Wang, Bioengineering, University of Washington, Seattle, Washington, UNITED STATES| Purpose: To investigate the feasibility of using OCT and OCT angiography (OCTA) to image the vascular involvement in ocular choroidal tumors. Methods: Enrolled subjects were imaged using swept source OCTA (PLEX® Elite 9000 (ZEISS, Dublin, CA)) . Each subject underwent OCT imaging equipped with OCTA scanning protocol. 6x6 mm and 12x12 mm scanning patterns were utilized. OCT signal attenuation was corrected to enhance image contrast of 3D SS-OCT structural dataset, upon which an automatic segmentation software was applied to generate vascular information. Choroidal slab was automatically segmented from Bruch’s membrane to the choroidal-scleral interface using a graph search method. Minimum en face projection was used to extract the choroidal blood vessels from OCT structural image. OCTA angiogram was also generated to enhance the visualization of blood vessels within tumor region. Manual splicing of the en face projections between structural OCT image and angiogram from OCTA in the tumor region was performed on each subject. Results: 5 patients diagnosed with ocular tumor were enrolled in this study. The proposed method was useful to achieve the visualization of vascular information within tumor and its surrounding regions, from which feeding vessels to choroidal ocular tumor (Figure 1 A&B&C) may be identified. 12x12 mm scans were better to delineate vascular information surrounding the tumor than 6x6 mm, facilitating the identification of feeding vessels. However, 6x6 mm scans gave more detailed vascular information within tumor. Conclusions: This preliminary study describes a method to visualize vascular involvement in choroidal tumor and to identify possible feeding vessels. Results indicate that the proposed method has the potential to provide quantification assessment of choroidal ocular tumor. Figure 1. Representative vascular images of choroidal layer acquired from one ocular tumor patient (12x12 mm scan protocol). (A) choroidal vascular image obtained from OCTA algorithm, B) choroidal vascular image obtained from structural OCT images, and C) the spliced image between OCTA image and the structural image to show vascular information within and surrounding the tumor. CONTROL ID: 3192740 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB072 TITLE: Improved visualization of retinal vasculature using multi-layer segmentation of optical coherence tomography angiography (OCTA) images FIRST AUTHOR: Mary K Durbin AUTHORS/INSTITUTIONS: M.K. Durbin, H. Bagherinia, A. Fard, Carl Zeiss Meditec, Inc., San Francisco, California, UNITED STATES|T. Santos, M. Soares, C. Neves, M. Lopes, J. Cunha-Vaz, AIBILI - Association for Innovation and Biomedical Research on Light, Coimbra, PORTUGAL| Purpose: Visualization of vasculature from OCTA is typically done by generating en face angiography images between boundaries of interest segmented from the structural OCT data. The purpose of this study is to qualitatively and quantitatively evaluate a new multilayer segmentation algorithm in comparison to a commercial implementation that segments only the retinal pigment epithelium (RPE) and inner limiting membrane (ILM), and estimates the remaining layers based on predetermined rules. Methods: One eye from each of 34 subjects with early diabetic retinopathy were imaged on a CIRRUS™ HD-OCT ® 5000 with AngioPlex OCT Angiography (ZEISS, Dublin, CA) with the 3x3 OCT Angio scan. The commercial segmentation defines the superficial retinal layer (SRL) is as the layer between the ILM (Z ) and an approximation IPL ILM of the inner plexiform layer, Z =Z +70%*(T ), where T is the thickness between ILM and the ILM ILM-OPL ILM-OPL outer plexiform layer (OPL), which is estimated as Z = – 110 μm. The deep retinal layer (DRL) is defined OPL ZRPEfit as being between the IPL and OPL. For the multilayer method, the SRL is defined as being between the segmented ILM and the segmented outer boundary of the IPL minus 10 microns, while the DRL is defined as being between the segmented IPL minus 10 microns and the segmented outer boundary of the inner nuclear layer (INL) plus 30 microns. The density of vasculature in these angiography en face images was quantified using the commercial method to estimate the percent area of perfused vasculature (Perfusion Density). The quantitative results were compared using linear regression. The images were also reviewed qualitatively to determine which provided a visualization of the vasculature most consistent with the expected anatomy. Results: Multilayer segmentation qualitatively had better vessel continuity and visualization of the deep capillary plexus especially in the parafoveal region (See Figure 1). Regression analysis found the two methods were well correlated, with an r-squared of 0.90 for the SRL (slope of 1.09, offset of 0.05) and 0.89 for the DRL (slope of 1.1, offset of 0.01). Conclusions: Multilayer segmentation can allow layers to be created based on the expected structural location, and improves visibility of microvasculature in the deeper retinal layers. Quantitatively the results are similar to the commercial method. Superficia lSiab• CONTROL ID: 3194231 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB073 TITLE: Structure function correlation in uveitic cystoid macular edema and treatment response using non-invasive Optical Coherence Tomography Angiography FIRST AUTHOR: Sophia Zagora AUTHORS/INSTITUTIONS: S. Zagora, Ophthalmology, Save Sight Institute, Sydney Eye Hospital and University of Sydney, Sydney, New South Wales, AUSTRALIA|S. Zagora, D. Grewal, Moorfields Eye Hospital, UNITED KINGDOM|D. Grewal, Duke Eye Center, North Carolina, UNITED STATES|O. Tomkins-Netzer, S. Lightman, Ophthalmology, UCL, London, UNITED KINGDOM|O. Tomkins-Netzer, Ophthalmology, Bnai Zion Medical Center, Technion – Israel Institute of Technology, Haifa, ISRAEL| Purpose: We conducted a cross sectional cohort study to investigate whether the structural integrity of the superficial capillary plexus (SCP) and deep capillary plexus (DCP) and the size of the largest intraretinal cyst and the preserved retinal tissue on en face imaging using optical coherence tomography angiography (OCTA) correlates with vision and is predictive of visual recovery after treatment in eyes with uveitic cystoid macular edema (CME). Methods: Patients with non-infectious uveitic CME (>320 microns) who underwent local steroid treatment had baseline and follow up en face OCTA images (Zeiss Angioplex, Carl Zeiss Meditec) with a scanning area of 3×3-mm centered on the fovea. The SCP in the ganglion cell layer and the DCP beneath the inner plexiform layer (IPL) were measured. The FAZ area (3×3mm) of each vascular layer was calculated after conversion to 8-bit grayscale images using ImageJ software (NIH, Bethesda, MD). The border of the FAZ was manually drawn by two retinal specialists. The preserved retinal tissue on baseline en face OCT imaging, FAZ area in the SCP and DCP and disorganization of retinal inner layers (DRIL) in the central 1000 microns were correlated with best-corrected logMAR VA. Results: Thirty-five eyes of 30 patients (14 female, 16 male) with a mean age of 52 and vision of 38 letters were included. Mean vascular density in the SCP was 13.50±4.67% and DCP was 10.57±2.64%. Mean FAZ in the SCP was 0.28±0.12mm2 and DCP was 1.09±0.47. Correlation coefficients for baseline VA (ETDRS letters) correlated with the vessel density in the SCP (0.64, p=0.08) and DCP (r=0.78, p=0.019). There was also a negative correlation with the area of the FAZ in the SCP (r=-0.33, p=0.24) and DCP (r=-0.40, p=0.19) with vision. DRIL in the central 1000 microns correlated negatively with vision. (r=-0.37, p=0.21). Area of largest cyst at baseline on en face OCT correlated with vision (r=-0.66, p=0.1). Conclusions: Using OCTA, vessel density in the SCP and DCP, correlated with VA. Similarly there was a negative correlation of FAZ area in the SCP and DCP with vision. Structural integrity of the layers and preserved retinal tissue on en face OCTA negatively correlated with vision indicating that the vascular and structural changes in the DCP and SCP on OCTA could serve as a non invasive surrogate biomarker for VA. Figure 1.

Figure 1. Measuring the FAZ in the superficial capillary plexus (A) and deep capillary plexus (8) utilizing Image J. B scan OCT (C) showing measurement of disorganization of retinal inner layers (DRIL) in the central 1000 microns

Figure 2.

Figure 2. Measuring the vessel density in the superficial capillary plexus (A and D) and deep capillary plexus (8 and E) after converting the image to binary 8-bit image and applying thresholding algorithm using Otsu method to threshold the vessels as black. There are some issues with image artifact in eyes with CME where cysts mask the vasculature in the deep capillary plexus (8 and E). En Face image showing the intraretinal cysts as dark areas. Using thresholding the larger cysts were identified in red and the area was calculated (C and F). CONTROL ID: 3178574 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB074 TITLE: An algorithm to enhance OCT Angiography images FIRST AUTHOR: Ting Luo AUTHORS/INSTITUTIONS: T. Luo, H. Bagherinia, A. Fard, Carl Zeiss Meditec Inc. , Dublin, California, UNITED STATES| Purpose: High quality angiography images are crucial in assisting diagnosis of vasculature related retinopathy. We proposed an algorithm to enhance OCT Angiography (OCTA) images. The combination of high density scans and proposed image enhancement algorithm improves the visualization of microvasculature details for large field of view (FOV). Methods: Two prototype high-density scan patterns consisting of 490x490 (6x6mm) and 654x654 (8x8mm) A-scans with a sample spacing of 12.3 µm were implemented.Four scan patterns were used: 1) 3x3mm FOV, sample spacing 12.3 µm, 2) 6x6mm FOV, sample spacing 17µm, 3)8x8mm FOV, sample spacing 23µm, 4)12x12mm FOV, sample spacing 24 µm. Patterns 2 and 3 were used for both regular and HD scans. TM Images from normal/diseased eyes were acquired at least one of the scan patterns using CIRRUS HD-OCT 5000 and AngioPlex® OCT Angiography (Zeiss, Dublin, CA). Superficial and deep capillary plexus slabs (SCP, DCP) were generated using the standard CIRRUS software. The OCTA image enhancement algorithm is based on an anisotropic diffusion method using the diffusion tensor filtering to enhance the vessel structure connectivity with a smoothing effect that adapts to the underlying vascular structure. The computation is based on an iterative process to determine each successive image in the family and continued until a sufficient degree of smoothing is obtained. Three graders reviewed the en face images before and after enhancement using the following criteria: 1=much worse; 2=worse; 3=about the same; 4=improved; 5=much improved. Results: Images from 44 subjects with normal and diseased eyes (e.g., DR, dry AMD, BRVO, etc.) using each of the four scan patterns were processed with the algorithm. The improvements were presented on both high and low density scans (fig 1). The lower and upper 90% nonparametric confidence limit from 3 graders are being reported in table 1. Conclusions: An OCTA vasculature enhancement algorithm is presented in this abstract. The results showed a significant improvement in vessel structure connectivity and vascular granularity reduction specifically in high-density OCTA images. The enhanced OCTA images improves the visualization of microvasculature details in larger FOV. Figure 1 SRL (a) and DRL (b) slabs of the 8x8mm HD Angiography are shown before and after enhancement using an anisotropic diffusion filter. Table 1. Lower and upper 90% nonparametric confidence limit from Cirrus 5000
CONTROL ID: 3195955 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB075 TITLE: Choroidal Changes after Photodynamic Therapy in Chronic Central Serous Chorioretinopathy FIRST AUTHOR: Ruwan Silva AUTHORS/INSTITUTIONS: R. Silva, R. Shields, Stanford University, California, UNITED STATES| Purpose: To report the acute and long term choroidal findings in patients with chronic central serous chorioretinopathy (CCSC) undergoing ½ fluence photodynamic therapy (PDT).

Methods: This is a prospective, consecutive, interventional, non-comparative case series at a tertiary referral vitreoretinal practice. Twenty one eyes from 21 patients are included. Results: Among 21 eyes, all had improved vision following PDT during a mean of 5.2 +/- 4.8 months of follow-up. The incidence of choroidal neovascular membranes was 19.0% (4/21) with only one of these eyes 4.8% (1/21) requiring intravitreal anti-VEGF injections. Changes in the choriocapillaris were noted in 28.6% (6/21) eyes following PDT. Eighteen patients (85.7%) demonstrated striking regions of dilated choroidal vessels closely corresponding to regions of outer retinal changes and subretinal fluid. Twenty patients (95.2%) experienced resolution of subretinal fluid within 3 months of receiving PDT. Conclusions: OCTA may provide new insight into the pathogenesis of CSC namely in identifying a correlation between structures of the deep choroid and changes in the retinal pigment epithelium and neurosensory retina. (No Image Selected) CONTROL ID: 3195426 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB076 TITLE: Analysis of Choroidal Neovascular Flow Lesions in Subretinal Hyperreflective Material using Optical Coherence Tomography Angiography FIRST AUTHOR: SABIA HANDA AUTHORS/INSTITUTIONS: S. HANDA, A. Arora, A. Agarwal, V. Gupta, Advanced Eye Center, PGIMER, Chandigarh, INDIA| Purpose: Subretinal Hyperreflective Material (SHRM) is a morphological feature seen on OCT. This Subretinal Hyperreflective Material (SHRM) may harbour neovessels, the presence of which has important implications on the management. OCT Angiography provides a novel tool to pick up these neovessels early. We did a retrospective, observational study to describe the morphology, characteristics, bioactivity and multimodal imaging (MMI) features of choroidal neovascular (CNV) flow lesions within subretinal hyperreflective material (SHRM) using optical coherence tomography angiography (OCTA). Methods: In this study, consecutive subjects with non-age-related macular degeneration manifesting SHRM on optical coherence tomography (OCT) were included. Various etiologies included infectious and non-infectious uveitis, myopia, and angioid streaks. Multimodal imaging was performed using OCTA, swept-source OCT, color fundus photography, and fluorescein angiography (FA). Intrinsic flow signals were assessed within SHRM on OCTA and follow-up evaluation was performed after treatment with anti-VEGF agents. Results: 15 eyes of 15 patients (10 males; mean age: 41.06 ± 12.14 years) were included in the study. CNV flow lesions were observed in 10 eyes with SHRM characterized by fine loops of vessels with hair-pin loop configuration, flow signals on OCTA, extensive branching pattern and perilesional halo. On FA, the presence of CNV could not be conclusively confirmed in any eye. There was no fluid on OCT in any eye. Intravitreal ranibizumab (RNZ) was given in all the patients. All the subjects demonstrated reduction in CNV flow signals on OCTA and decreased size, branching patterns and regression of CNV. There was no change in the size, morphology, and pattern of the SHRM in any eye. Conclusions: OCTA provides high-resolution imaging of CNV activity inside SHRM lesions. After treatment with intravitreal RBZ, the CNV lesions tend to regress but there is no change in the morphology of SHRM in these eyes. (No Image Selected) CONTROL ID: 3195831 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB077 TITLE: Longitudinal Follow-up of Tubercular Serpiginous-Like Choroiditis using Optical Coherence Tomography Angiography FIRST AUTHOR: Rohan Bir Singh AUTHORS/INSTITUTIONS: R.B. Singh, Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, UNITED STATES|R.B. Singh, Ophthalmology, Government Medical College & Hospital, Chandigarh, Chandigarh, INDIA|A. Agarwal, K. Aggarwal, V. Gupta, Advanced Eye Center, Post Graduate Institute of Medical Education and Research, Chandigarh, Chandigarh, INDIA|D. Grewal, Ophthalmology, Duke Eye Center, Durham, North Carolina, UNITED STATES| Purpose: Choriocapillaris flow deficit areas on optical coherence tomography angiography (OCTA) in active tubercular serpiginous-like choroiditis (TB SLC) co-localize with hypocyanescent lesions on indocyanine green angiography. OCTA allows assessment of recovery of the inner choroid with therapy and helps in monitoring the development of choriocapillaris atrophy in larger placoid lesions. Thus, OCTA aids the management and prognostication of TB SLC non-invasively. To describe follow-up of tubercular serpiginous-like choroiditis (TB SLC) using multimodal imaging including optical coherence tomography angiography (OCTA). Methods: In a prospective cohort study, patients with active TB SLC underwent OCTA, enhanced-depth imaging OCT, and autofluorescence. Fluorescein and indocyanine green angiography (ICGA) were performed at baseline and 3 months. Results: 16 eyes (mean age: 31.5±11 years; 8 eyes with multifocal and 8 with placoid SLC) were included. At baseline, OCTA showed choriocapillaris flow deficit areas co-localizing with hypocyanescent lesions on ICGA in all eyes. Multifocal SLC showed near-complete resolution of flow deficit with minimal choriocapillaris atrophy with therapy. Placoid SLC showed extensive atrophy that agreed well with ICGA. Conclusions: OCTA reveals serial choriocapillaris changes in TB SLC that agree well with ICGA. Despite therapy, larger placoid lesions develop choriocapillaris atrophy in flow deficit areas. (No Image Selected) CONTROL ID: 3195333 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB078 TITLE: OCTA cube averaging for identification of vascular abnormalities in diabetic retinopathy using swept-source OCT FIRST AUTHOR: Thomas Callan AUTHORS/INSTITUTIONS: T. Callan, L. De Sisternes, S. Kubach, W. Lewis, C. Wu, M. Durbin, Carl Zeiss Meditec, Inc., Dublin, California, UNITED STATES|R.A. Goldberg, Bay Area Retina Associates, Walnut Creek, California, UNITED STATES| Purpose: Optical coherence tomography angiography (OCTA) can image vascular changes such as microaneurysms (MA), vessel caliber, capillary non-perfusion and ischemia in diabetic retinopathy (DR). This study investigated the use of averaging to improve image quality and compared averaged to single images in detection of these entities using swept-source OCT (SS-OCT). ® Methods: DR patients were imaged with the PLEX Elite 9000 (ZEISS, Dublin, CA) SS-OCT. Four to seven Angio 6x6 mm images were acquired on each patient. Single images were topographically analyzed using quality maps and the better quality repetition was chosen as a reference for registration. Averaged images were created by volumetrically registering the remaining frames to the reference image. Vascular abnormalities were identified in both the single and averaged images for each eye. Results: Fourteen eyes of 12 patients with DR of varying severity were included in this study. With few exceptions, the same vascular abnormalities were found in both the single and image-averaged scans (Table 1). One eye showed MAs and one had a better-defined area of non-perfusion in the image-averaged scan. The image quality of the small blood vessels was superior with the averaged scans, particularly in the deep vessel layer (Figure 1). Conclusions: In this study, most DR vascular abnormalities were recognized with both single and image-averaged scans. Image quality was found to be improved with averaging, but did not make a significant difference in identification of abnormalities. Both the superficial and deep vascular layers were enhanced with the image-averaged scans. Table 1: Comparison between single and image-averaged scans Figure 1: Superficial and deep slabs of single scan (top) and image-averaged (6x) scan (bottom). Note areas of ischemia, MAs and foveal avascular zone similarities between the two slabs with improved resolution of vessels in the image-averaged slabs in the superficial layer (left) and deep layer (right). CONTROL ID: 3195771 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB079 TITLE: Quantitative analysis of microvasular densities in retina and choroid using wide field swept source optical tomography angiography FIRST AUTHOR: Ying Zhu AUTHORS/INSTITUTIONS: Y. Zhu, Y. Cui, J. Wang, R. Katz, J.B. Miller, Retina Service, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, UNITED STATES|Y. Zhu, Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, Hunan, CHINA|Y. Cui, Department of Ophthalmology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, CHINA|Y. Lu, Harvard Medical School, Boston, Massachusetts, UNITED STATES| Purpose: Optical tomography angiography (OCTA) has been used to quantify changes in microvasculature in diabetic retinopathy, however mostly focused on the macula. The present study investigated the possible changes of both retina and choroid in a wide field by swept source OCTA (SS-OCTA). Methods: We conducted a prospective, observational study in Mass Eye and Ear from December 2018 to February 2019. Treatment naïve non-proliferative diabetic retinopathy (NPDR), diabetic patients with no diabetic retinopathy (DR) and healthy control eyes with no ocular diseases were included. All patients were imaged with a SS-OCTA (PLEX® Elite 9000; Carl Zeiss Meditec, Inc, Dublin, CA) and the montage image composed of two 15mm by 9mm angio image (superior and inferior) was used for analysis. Vessel density (VD) and skeleton density (SD) of superficial retina, deep retina and choriocapillaris were calculated using Image J (NIH). All statistic analyses were performed using SPSS software 22.0 (SPSS Inc, Chicago, IL) and one-way ANOVA was used for multiple comparisons between groups. Results: Twenty-two eyes (13 eyes with NPDR, 4 eyes of diabetic patients without DR and 5 healthy control eyes) were included in the study. Severe NPDR had significant lower VD and SD in both superficial and deep retina compared to other groups (P<0.001). Compared to mild NPDR (VD 0.496±0.015, SD 0.207±0.004), VD (0.524±0.028) and SD (0.220±0.009) of superficial retina in healthy control were significantly higher (P<0.05). VD in both superficial (0.527±0.018) and deep layer (0.614±0.017) of retina in eyes of diabetic patients without diabetic retinopathy were significant higher than those in mild NPDR (superficial VD 0.496±0.015, deep VD 0.571±0.015, P<0.05). However no statistically significant changes were detected between healthy controls and diabetic patients without diabetic retinopathy. The average choriocapillaris VD (0.527±0.027) and SD (0.266±0.012) in eyes with diabetic retinopathy was lower than those in control (VD 0.549±0.043, SD0.272±0.014), but this did not reach statistical significance (P>0.05). Conclusions: Quantitative analysis of wide filed OCTA images could indicate the severity of diabetic retinopathy. (No Image Selected) CONTROL ID: 3195301 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB080 TITLE: Optical Coherence Tomography Angiography of Acute-Onset Vogt-Koyanagi-Harada Disease in a Young Female FIRST AUTHOR: Grayson Wilkes Armstrong AUTHORS/INSTITUTIONS: G.W. Armstrong, G.N. Papaliodis, J.B. Miller, Ophthalmology, Massachusetts Eye & Ear , Boston, Massachusetts, UNITED STATES|M.J. Smith, Faculty of Medicine & Dentistry, University of Alberta, Alberta, CANADA| Purpose: Optical coherence tomography angiography (OCTA) is capable of non-invasively assessing choroidal blood flow. To date, limited knowledge exists on OCTA findings in uveitic diseases preferentially affecting the choroid, such as Vogt-Koyanagi-Harada (VKH) disease. We present unique OCTA imaging findings in acute VKH disease and show that OCTA imaging may be beneficial in early diagnosis, assessing for response to treatment, and monitoring for relapse of disease in VKH. Methods: A young female presenting to Massachusetts Eye & Ear (Boston, USA) diagnosed with acute VKH underwent complete ophthalmic exam with multimodal imaging. Optical coherence tomography angiography was performed using the Optovue Avanti, using AngioVue OCTA Software (Optovue, Inc., Fremont, CA, USA). Results: Clinical examination revealed a visual acuity of 20/40 OD and 20/25-1 OS, with normal pupils and intraocular pressures. Anterior segment revealed conjunctival injection, endothelial keratoprecipitates, and 2+ cell without hypopyon. Posterior examination demonstrated 1+ vitritis as well as serous macular detachments (Fig 1A). Fundus autofluorescence demonstrated hyperfluorescence in areas of serous retinal detachment (Fig 1B). Fluorescein angiography demonstrated delayed choroidal filling with optic disc leakage as well as macular and peripapillary focal staining resembling a ‘starry sky’ pattern in early and late frames (Fig 1C-D). Spectral-domain OCT showed bilateral serous retinal detachments with a subretinal fibrinous septae in the right eye (Fig 1E). OCTA demonstrated (1) diffuse areas of decreased choriocapillaris flow signal in areas of serious retinal detachments (Fig 2C-D, solid line), (2) numerous smaller focal areas of choriocapillaris flow void (Fig 2C-D, broken line), and (3) normal choriocapillaris flow signal beneath the right eye subretinal fibrinous septae. All clinical and imaging findings resolved at 2.5 months after treatment with systemic and topical corticosteroids (Fig 2E-F). Conclusions: We present three unique OCTA imaging findings in the acute phase of VKH. All OCTA findings resolved in our patient after use of corticosteroids. These findings support the conclusion that OCTA imaging may be beneficial in early diagnosis, assessing for response to treatment, and monitoring for relapse of disease in VKH.

CONTROL ID: 3195361 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB081 TITLE: Optical Coherence Tomography Angiography: A useful tool for Longitudinal Follow-up of Tubercular Serpiginous-Like Choroiditis. FIRST AUTHOR: Atul Arora AUTHORS/INSTITUTIONS: A. Arora, A. Agarwal, K. Aggarwal, S.K. Mandadi, V. Gupta, OPHTHALMOLOGY, PGIMER, Chandigarh, CHANDIGARH, INDIA|D. Grewal, Ophthalmology, Duke University, North Carolina, UNITED STATES| Purpose: Tubercular Serpiginous like choroiditis(SLC) is a chronic recurrent form of posterior uveitis with centrifugal spread with an active advancing edge. While ICGA remains the gold standard for assessing pathologic alterations in choroidal circulation, it is an invasive technique with a risk of dye-related serious side effects. Optical coherence tomography angiography (OCTA) is a novel non- invasive imaging tool to study the retinal and choroidal vasculature and its involvement in choroiditis. We did a prospective cohort study to describe follow-up of tubercular serpiginous-like choroiditis (TB SLC) using multimodal imaging including optical coherence tomography angiography (OCTA). Methods: Patients with active TB SLC underwent OCTA, enhanced-depth imaging OCT, and autofluorescence. Fluorescein and indocyanine green angiography (ICGA) were performed at baseline and 3 months. Results: 16 eyes (mean age: 31.5±11 years; 8 eyes with multifocal and 8 with placoid SLC) were included. At baseline, OCTA showed choriocapillaris flow deficit areas co-localizing with hypocyanescent lesions on ICGA in all eyes. Multifocal SLC showed near-complete resolution of flow deficit with minimal choriocapillaris atrophy with therapy. Placoid SLC showed extensive atrophy that agreed well with ICGA. Conclusions: OCTA reveals serial choriocapillaris changes in TB SLC that agrees well with ICGA. Despite therapy, larger placoid lesions develop choriocapillaris atrophy in flow deficit areas. Choriocapillaris flow deficit areas on optical coherence tomography angiography (OCTA) in active tubercular serpiginous-like choroiditis (TB SLC) co-localize with hypocyanescent lesions on indocyanine green angiography. OCTA allows assessment of recovery of the inner choroid with therapy and helps in monitoring the development of choriocapillaris atrophy in larger placoid lesions. Thus, OCTA aids the management and prognostication of TB SLC non-invasively. (No Image Selected) CONTROL ID: 3195639 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB082 TITLE: A Retrospective Study of Hypertension-induced Retinal Vascular Changes Using OCT-Angiography FIRST AUTHOR: Yifan Lu AUTHORS/INSTITUTIONS: Y. Lu, Harvard Medical School, Boston, Massachusetts, UNITED STATES|R. Zeng, Boston University, Boston, Massachusetts, UNITED STATES|J. Park, Yale School of Medicine , New Haven, Connecticut, UNITED STATES|J. Wang, J. Diaz, J.B. Miller, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, UNITED STATES| Purpose: Hypertension (HTN), or high blood pressure, has been shown to induce changes in the retinal vasculature, such as focal arteriolar narrowing and microaneurysm. Optical coherence tomography angiography (OCT-A), a fast and non-invasive imaging device, has emerged as a novel tool to evaluate the retinal microvasculature in many chorioretinal vascular diseases. However, there is a lack of literature on the use of OCT-A devices in systemic HTN. Herein, we evaluated the relationship between systemic HTN and microvascular changes in the retina using OCT-A. Methods: This is a retrospective review conducted at Massachusetts Eye and Ear Infirmary (MEEI). All patients were imaged using the Optovue RTVue. Patients with systemic HTN were included. Patients with history of chorioretinal diseases including diabetic retinopathy, retinal vein occlusion, ocular ischemic syndrome, age-related macular degeneration, macular telangiectasia, epiretinal membrane, as well as history of diabetes mellitus were excluded from analysis. Foveal avascular zone (FAZ) area and vessel densities (VD) of the superficial capillary plexus (SCP), deep capillary plexus (DCP) and choriocapillaris (CC) were calculated utilizing ImageJ software. A multi-level mixed model using the Statistical Analysis Software (SAS) was employed for data analysis. Results: Seven patients (n = 7 eyes) with HTN were included in the comparison group and 16 patients (n = 19 eyes) without HTN were selected for the control group. No significant difference was found in SCP FAZ (p = 0.545), DCP FAZ (p = 0.102), SCP VD (p = 0.614), DCP VD (p = 0.536), or CC VD (p = 0.497) between patients with and without HTN. Conclusions: OCT-A is a nascent imaging technology with vast clinical applications, including those related to chorioretinal diseases and potentially systemic disorders that induce retinal vascular changes. Although hypertension has been known to alter the retinal vasculature, we found no significant difference in the retinal vessel densities by OCT-A imaging in this small cohort. A larger patient sample, including patients with more severe systemic hypertensive disease, may yield better biomarkers of hypertension control. (No Image Selected) CONTROL ID: 3195798 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB083 TITLE: Comparison between multimodal imaging and OCT-A in a patient with juxtapapillary choroidal neovascular membrane as a complication of optic disc drusen FIRST AUTHOR: Vivian Paraskevi Douglas AUTHORS/INSTITUTIONS: V. Douglas, K. Douglas, D. Cestari, J. Rizzo III, J.B. Miller, Mass Eye and Ear, Massachusetts, UNITED STATES| Purpose: Optic Nerve Head Drusen (ONHD) are acellular, calcified bodies usually located anterior to the lamina cribosa and are associated with several complications including retinal vascular occlusion, anterior ischemic optic neuropathy, and choroidal neovascular membrane (CNVM). We compared optical coherence tomography angiography (OCT-A) with traditional multimodal imaging in a patient with bilateral optic nerve head drusen, severe visual field loss and central vision sparing. Methods: We present a case of a middle-aged male with longstanding visual field loss secondary to bilateral optic nerve head drusen (ONHD) since childhood. His medical history was significant for hypertension and T2DM non- insulin dependent and positive family history of ONHD (M) and DM (F). On examination, the BCVA was 20/20-2 OU with a trace rAPD OD. Biomicroscopy of the anterior segment was unremarkable and ocular pressure was within normal limits OU. Dilated fundus examination revealed extensive optic nerve disc drusen OU and retinal hemorrhages and exudates in the inferonasal macula adjacent to the disc margin. Fundus autofluoresence, fluorescein angiography, OCT and OCT-A were performed. Results: The optic disc drusen are striking on both the fundus photos (Fig. 1) and particularly the autofluorescence (Fig. 2). Traditional intravenous angiography methods showed late leakage on fluorescein angiography (inferonasal macula) consistent with a juxtapapillary CNVM OD. Swept Source OCT also nicely illustrated both the juxtapapillary subretinal fluid and optic disc drusen. OCT-A identified a small juxtapapillary choroidal neovascular membrane (CNVM) inferonasal to the macula correlating with the area of retinal hemorrhage and exudates. Conclusions: OCT-A imaging allows for precise in vivo localization of the retinal and choroidal vasculatures, including the peripapillary network. Thus, it can be a valuable and noninvasive tool for the detection of ONHD but also for its complications and prognosis.

CONTROL ID: 3195800 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB084 TITLE: Optical Coherence Tomography Angiography Imaging of Subretinal Neovascularization in Macular Telangiectasia Type 2: A Case Report FIRST AUTHOR: Carolyn Majcher AUTHORS/INSTITUTIONS: C. Majcher, R. Trevino, Optometry, University of the Incarnate Word, Fair Oaks Ranch, Texas, UNITED STATES| Purpose: Macular telangiectasia type 2 (MacTel2) is characterized by bilateral perifoveal telangiectatic capillaries th manifesting in the 5-6 decade. Subretinal neovascularization (SRN) infrequently complicates MacTel2 and is likely retinal in origin (Charbel Issa et al. Prog Retin Eye Res 2014). Few reports have described OCTA findings in proliferative MacTel2 (Villegas et al. Case Rep Ophth Med 2017, Zhang et al. Retina 2015) and is the purpose of this case report. Methods: A 56 year-old Hispanic female with a history of progressive vision decrease presented with MacTel2 complicated by SRN OS. Due to healthcare inaccessibility she was then lost to follow-up until 8 months later. 6mm and 3mm macular OCTA (AngioPlex©, Carl Zeiss Meditec) was performed at the initial and 8 month exams along with structural OCT. The superficial, deep, and avascular preset enface displays as well as a customized avascular display OS were analyzed. Results: Initial examination revealed perifoveal retinal opacity, mild crystalline deposits, and temporal pigmented plaques OU. Additionally, superior perifoveal grayish subretinal thickening was noted OS. Structural OCT imaging revealed central outer retinal atrophy with associated subfoveal atrophic clefts OU. Two hyperreflective deposits (SRN) were visible superior and nasal to the fovea OS. The superior SRN membrane was associated with minimal adjacent RPE hyperpigmentation and had no associated fluid. The nasal SRN membrane had questionable adjacent subfoveal SRF. OCTA revealed enlarged foveal avascular zones with irregular contour OU. Prominent dilated telangiectatic perifoveal capillaries, particularly temporally, were present OU. Custom avascular enface display OS revealed 2 SRN complexes superior and nasal to the fovea. Blunted venules of the retina were present overlying both SRN membranes and appeared to anastomosis with them. OCT findings 8 months later were unchanged OD. The nasal SRN membrane OS developed new, adjacent SRF although no morphologic change in the SRN membranes themselves were noted. Conclusions: OCTA is a non-invasive technology that provides depth-resolved, high resolution microvascular detail of perifoveal telangiectasia in patients with MacTel2. This revolutionary tool is useful in detecting, quantifying, and morphologically characterizing SRN membranes that may complicate MacTel2. OD OS Artificial Intelligence CONTROL ID: 3176343 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0101 TITLE: Improvement and validation of high precision ocular oximetry using a convolutional neural network algorithm and a phantom eye FIRST AUTHOR: Damon T. DePaoli AUTHORS/INSTITUTIONS: D.T. DePaoli, D.C. Côté, CERVO Brain Research Center, Quebec City, Quebec, CANADA|N. Lapointe, J. Desroches, P. Saugaveau, D. Sauvageau, Zilia, Quebec, Quebec, CANADA|D.T. DePaoli, P. Tossou, D.C. Côté, Université Laval, Qubeec, Quebec, CANADA|D. Sauvageau, University of Alberta, Edmonton, Alberta, CANADA| Purpose: Retinal oximetry is a non-invasive imaging technology that enables the measurement of oxygen saturation (SO ) in the eye fundus. The goal of this research was to validate a convolutional neural network (CNN) algorithm 2 designed to calculate and improve the precision of SO measurements from diffuse reflectance spectra (DRS) taken 2 on the optic nerve head (ONH). Methods: The ocular oximetry device developed by Zilia was used to acquire diffuse reflectance spectra (DRS) on several ONH-mimicking liquid optical phantoms (phantom eye). The oxygenation of the blood circulating in the phantom eyes was dynamically cycled from 100% to 0% oxygenation (using yeast and oxygen gas). SO 2 measurements were made simultaneously with Zilia’s device and gold standard devices for comparison throughout the experiments. The phantom eyes were made to assess variations in blood volumes and scattering coefficients, corresponding to typical ranges observed for ONH optical properties. The procedure was then repeated with several cataract-simulating contact lenses integrated to the optical path to show robustness of oximetry measurements. Finally, we apply the algorithm to spectra acquired in vivo on subjects at baseline and hyperoxic conditions. Results: We found good agreement in SO measurements between the results obtain with the Zilia device using the 2 CNN algorithm and the gold standard references in all phantom eyes. We specifically show strong robustness in precision, even when all 3 of the experimental cataract-simulating lenses were used. This is significant since cataracts has traditionally plagued oximetry measurements. Lastly, we show that ocular oximetry measurements showed reliable increases in in vivo oxygenation, consistent with results from tissue oximetry, in test-subjects provided with 100% oxygen. Conclusions: We present, here, further validation that the oximetry device and CNN algorithm developed to measure SO in the eye fundus produces reliable, precise measurements even under conditions where blood volume fractions 2 vary, optical scattering changes, and cataract-simulating contact lenses are included. Furthermore, we show that the algorithm can be applied to in vivo measurements. (No Image Selected) CONTROL ID: 3179913 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0105 TITLE: Deep learning for automatic diabetic retinopathy detection under multiple image quality levels FIRST AUTHOR: Mhd Hasan Sarhan AUTHORS/INSTITUTIONS: M. Sarhan, Technical University of Munich, Munich, GERMANY|M. Yigitsoy, A. Eslami, Carl Zeiss Meditec, AG, GERMANY|K. Makedonsky, M. Mack, M. Durbin, Carl Zeiss Meditec, Inc., Dublin, California, UNITED STATES| Purpose: Deep learning techniques are showing promising results for automatic diabetic retinopathy (DR) screening on fundus images. We analyze the effect of image quality of images collected with a handheld fundus camera on the performance of a DR screening algorithm. Methods: This retrospective study used 257 fovea-centered fundus images from 93 subjects collected using VISUSCOUT® 100 (ZEISS, Jena, Germany) handheld fundus camera. The images are annotated positive for DR if signs of DR are visible (mild or more in International Clinical Diabetic Retinopathy disease severity scale). Image quality is assessed using a subjective 1-5 scale (1-very poor; 2-poor; 3-fair; 4-good; 5-excellent). Fig 1 shows an example of each quality level. 233 images were annotated as negative for DR (healthy) and 24 were annotated as positive for DR. We used our Diabetic Retinopathy Deep Network grading model (DRDN) that is trained on ~35k publicly available fundus images for grading the severity of DR. DRDN is used to classify VISUSCOUT images for Healthy vs DR by regarding a mild or more prediction as DR. No data from the same handheld device has been used for training the model. Sensitivity (Sn), specificity (Sp) and area under the curve of receiver operating characteristics curve (AUC) were reported for all images. Sn and Sp were used to analyze the effect of image quality on DR prediction performance. Results: DRDN shows good prediction power for unseen images from handheld camera reaching AUC of 0.98 on images from all quality profiles. No false negatives were triggered, hence, Sn=1. The model gave 22 false positives with Sp=0.905, 95% Confidence Interval (CI) [0.866, 0.943]. The results are shown in Fig 2. Conclusions: A high number of false positives (Sp=0.8, 95% CI [0.656, 0.943]) occurred in very poor quality Images. For images of poor and above quality, we observe a consistent performance of the algorithm with less effect of image quality on the Sp. Using samples from fair quality and above gives the highest results with Sp=0.926, 95% CI [0.886, 0.966] which suggests that using images of above-poor quality would decrease the number of false positives in the DR screening. Examples of fundus images for each quality level Sn and Sp of 5 quality levels and all levels combined; vertical black lines show 95% CI; the amount of available images for each quality is in parentheses; red dotted horizontal line is Sp over all images CONTROL ID: 3184328 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0104 TITLE: Self-Segmentation Strategies for Unsupervised Clustering and Visualization of Retinal Images FIRST AUTHOR: Jeremy Benson AUTHORS/INSTITUTIONS: J. Benson, T. Estrada, Computer Science, University of New Mexico, Albuquerque, New Mexico, UNITED STATES|J. Benson, S. Nemeth, P. Soliz, VisionQuest Biomedical, New Mexico, UNITED STATES| Purpose: The purpose of this research is to demonstrate a data-driven feature extraction technique that highlights prominent structures in retinal images for uses in visualization, segmentation, classification, and unsupervised clustering. Data labeling and model building are both time-consuming and expensive processes, and this research aims to expedite the process of initial data triage. Methods: We introduce a novel data representation that consists of cropping, restructuring, resizing, and subtracting an image from its original form, as is depicted in Figure 1. Using this image representation, histograms are calculated, reduced via Principal Component Analysis (PCA), and then clustered via t-distributed Stochastic Neighbor Embedding, grouping images of similar characteristics, as can be seen in Figure 2. The entire process relies only on the given dataset in question and does not require any labeling or model building. Results: We validate on a set of 1,145 images from a set of diabetic healthcare clinics, graded for quality and pathology by a certified ophthalmic medical technologist. Our data representation enables individual images to be segmented without any filtering or template matching. Our visualization results produce distinct clusters (ranging from “poor” to “high” levels of quality, including other features like bright artifacts, eyelashes, and depigmentation) for real- world sets. Conclusions: Retinal images vary due to camera, photographer skill, and individual being photographed. Detecting pathology or quality is often accomplished using large datasets and building models for future inferences. As the factors that influence retinal images change, these algorithms fail to scale. We present a novel approach that simply uses the data itself to perform a self-filtering and clustering with other data in the set. This method works with any camera type and does not require model building or data labeling, a time-consuming and expensive process. Data Representation Example Clustering Example CONTROL ID: 3188630 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB098 TITLE: Success rate of a vision screening program using a handheld fundus camera and a deep learning image quality and diabetic retinopathy screening algorithm FIRST AUTHOR: Katherine Makedonsky AUTHORS/INSTITUTIONS: K. Makedonsky, M. Mack, M. Durbin, Carl Zeiss Meditec, Inc., Dublin, California, UNITED STATES| Purpose: Diabetic Retinopathy (DR) affects one-third of adults with diabetes over the age of 40 according to a CDC survey. Early detection of DR is vital; however, access to care in remote areas of the world may be difficult. A platform called VISUHEALTH enables eye care providers to access retinal images and evaluate them for eye disease. ® VISUSCOUT 100 (ZEISS, Jena, Germany) is a handheld fundus camera used to capture images of the retina that may be uploaded to the VISUHEALTH platform. The purpose of this study is to evaluate the success rate of an image quality and AUTO-DR algorithm within the VISUHEALTH platform on images acquired at a vision screening event. Methods: Central images were acquired on 89 patients using the VISUSCOUT 100 at a vision screening event (Fig 1). Images for each patient were imported into the VISUHEALTH platform where an algorithm (AUTO-DR) determined the image quality and diabetic retinopathy status of the patient. An optometrist retrospectively evaluated the images and determined the success rate of the algorithm implemented within VISUHEALTH.

Results: 89 patients (Mean Age: 53 years, Standard deviation: 14) were screened with the VISUSCOUT 100. 20 of 89 patients were diabetics, and of those 17 were over the age of 40. The AUTO-DR algorithm identified 4 patients with ungradable images and 10 patients with diabetic retinopathy. An optometrist evaluated the images and determined that there were 14 patients with ungradable images, and 5 patients who had diabetic retinopathy. All of the patients with diabetic retinopathy were diabetics over the age of 40. Of the gradable images, the sensitivity of the DR algorithm was 100% and the specificity was 94%. The image quality algorithm sensitivity was 97% and specificity was 86%. Conclusions: Based on the results from the vision screening, 29% of the diabetics over age 40 had DR. Screening for ® diabetic retinopathy remains very important in order to prevent future blindness. The VISUSCOUT 100 is an easy to use camera that can be incorporated in the vision screening setting, however it is advised that an optometrist review images flagged as having diabetic retinopathy to prevent false positives. Images may be considered ungradable in the event of a cataract or pupils under 3.5 mm, therefore a referral is advised to properly evaluate the patient. ® DR image acquired with VISUSCOUT . CONTROL ID: 3193250 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0102 TITLE: Abnormality prediction from fundus images using Deep Learning and large amounts of data FIRST AUTHOR: Krunalkumar Ramanbhai Patel AUTHORS/INSTITUTIONS: K. Patel, K. Ranipa, CARIn, Carl Zeiss India (Bangalore) Pvt. Ltd., Bangalore, INDIA|A. Freytag, Carl Zeiss AG, Jena, GERMANY|N. Spier, Carl Zeiss AG, Oberkochen, GERMANY|A. Urich, Carl Zeiss AG, Munich, GERMANY| Purpose: Recent screening solutions for eye-related diseases mostly focus on individual diseases like DR, AMD, or Glaucoma. In contrast, we aim for a generic solution that allows screening for any abnormality in patient’s eyes. Potential users of report that classifies the retina as normal or abnormal could be corporate employees who are covered by insurance agency who is paying for this screening as part of the wellness program. Methods: Due to the application requirements, we present an automated approach based on deep neural networks. To allow for reliable abnormality predictions from the model, we trained with large amounts of manually annotated data. Therefore, we conducted experiments on a 2D fundus image dataset obtained with a low-cost, hand-held fundus camera, Visuscout® 100 (ZEISS, Jena, Germany). We collected data from a non-mydriatic clinical setup using VISUHEALTH platform. VISUHEALTH is a cloud based platform, through which a referred patient by GP or Diabatoloist is managed (screened) by Retinal specialist remotely. The dataset is comprised of 135k images of normal patients and 13k images of patients with different signs of abnormality. A medical expert annotated all images. The dataset was split into 80% for training and 20% for testing by randomly assigning patients exclusively either to train or to test. For the prediction of abnormality, we trained state-of-the-art convolutional neural networks either from scratch or pre-trained from ImageNet. Training datasets were up sampled to allow for balanced class distributions. For different fractions of the dataset, we ran the training with four random initializations to allow for statistical relevance of results. Results: Fig. 1 shows the results of a trained model on a held-out test set. The model reaches an AUC of 92%. For a sensitivity of 86%, our model reaches a specificity level of 86%. Fig. 2 demonstrates the benefit of collecting large medical datasets, especially for training from scratch. Conclusions: We evaluated abnormality prediction from 2D fundus images with deep neural networks. Leveraged by large amounts of data, our trained models surpassed 92% AUC on a real-world held-out test set comprised of 14K images. Fig. 1: Results of predicting abnormality from 2D fundus images Fig. 2: Dependency of the sensitivity-equals-specificity working points (left) and the AUC (right) on the amount of training data. CONTROL ID: 3193413 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB099 TITLE: AI Quantification of OCTA en face image quality FIRST AUTHOR: Charles Wu AUTHORS/INSTITUTIONS: C. Wu, R&D, Carl Zeiss Meditec, Incorporated, Dublin, California, UNITED STATES| Purpose: Optical coherence tomography angiography (OCTA) en face scans can be subject to artifacts from media opacities, eye movement, and dry eyes that may occur only in parts of the image. This study introduces a method to generate quantitative maps that describe the quality of an OCTA acquisition at each en face location. The values recorded can be averaged to provide a score that correlates with subjective quality grades. Methods: A machine learning linear regression model is trained to recognize the relationship between the texture properties and assigned quality grades of OCTA images. Grades were assigned on a 0-5 scale by two graders, with higher values indicating higher image quality (sharpness, visibility of clinical features, reduced artifacts). The training was done on 150 spectral-domain OCT (SD-OCT) angiography images captured on CIRRUS™ HD-OCT 5000 with AngioPlex® OCT Angiography (ZEISS, Dublin, CA) (57 3x3mm, 56 6x6mm, and 37 8x8mm scans from 29 eyes), and 452 swept-source OCT (SS-OCT) images captured on PLEX® Elite 9000 (ZEISS, Dublin, CA) (378 3x3mm, 37 6x6mm, 37 12x12mm from 39 eyes). The model was tested using 133 SD-OCT images (52 3x3mm, 44 6x6mm, and 37 8x8mm scans from 29 eyes) and 243 SS-OCT images (6x6mm scans from 49 eyes). The correlation of the quality scores predicted by the model and those assigned manually as well as the visual correlation of the quality maps with regional quality in individual acquisitions were assessed. Results: The model performed well for the CIRRUS images with mean absolute score error (MASE) of 0.49 ± 0.37 in the 3x3mm scans, 0.6 ± 0.4 in 6x6 scans, and 0.6 ± 0.41 in 8x8 scans. Overall MASE of the model for CIRRUS data was 0.56 ± 0.39. The MASE for the PlexElite was 0.6 ± 0.48. For scans of differing quality acquired on the same eye, the generated maps corresponded well with visual assessment of the images (Figure 1). Conclusions: The machine learning model was able to assess the quality of OCTA scans with relative accuracy and produce regional quality maps that correlated well with visual quality of the data. This method may be useful in providing quantitative feedback to operators when acquiring OCTA data, such as prompting the operator to reacquire an image if the quality metric is below a certain threshold. Figure 1: Quality maps obtained from repeated scans of the same eyes with varying quality. Top: 3x3mm scans from CIRRUS with AngioPlex. Bottom: 6x6mm scans from PLEX Elite.Submit Caption CONTROL ID: 3193731 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB093 TITLE: Deep learning algorithm to predict diabetic retinopathy (DR) progression on the individual patient level FIRST AUTHOR: Filippo Arcadu AUTHORS/INSTITUTIONS: F. Arcadu, F. Benmansour, A. Maunz, Roche Informatics, Roche, Basel, SWITZERLAND|F. Arcadu, F. Benmansour, A. Maunz, M. Prunotto, Roche Personalized Healthcare, Roche, Basel, SWITZERLAND|J. Willis, Z. Haskova, Clinical Science Ophthalmology, Genentech, Inc, South San Francisco, California, UNITED STATES|J. Willis, Z. Haskova, Roche Personalized Healthcare, Genentech, Inc, South San Francisco, California, UNITED STATES|M. Prunotto, Immunology, Infectious Disease & Ophthalmology, Roche, Basel, SWITZERLAND| Purpose: Diabetic eye disease is a major cause of vision loss and its global burden is expected to continue to worsen over the next few decades. The insidious nature of DR progression can leave the disease undetected until it threatens vision, therefore early identification of patients at risk is important. The steps on the Early Treatment DR Study (ETDRS) DR severity scale (DRSS) indicate the risk for progression in a group of patients with similar baseline severity, but the tools to effectively triage patients into fast vs slow DR progressors at the individual level are limited. As part of Roche’s comprehensive initiative in Ophthalmology Personalized Healthcare, we developed an automated deep-learning (DL) algorithm to predict individuals that are likely to experience significant DR progression over the next 2 years. Methods: Stereoscopic 7-field color fundus photographs and ETDRS DRSS scores from sham-treated study eyes and fellow eyes (n=683, n=682, n=645 eyes at months [M] 6, 12, and 24 respectively) of DR patients with macular edema from RIDE/RISE (NCT00473382/NCT00473330), were used for training and validation of DL algorithms with the goal to predict ≥2-step DRSS worsening at M6, M12, and M24 in individual patients. The area under the curve (AUC) was calculated and 5 times repeated 5-folds cross validation was performed to compute statistics of DL model performance. Results: At baseline, DRSS in sham-treated study eyes and fellow eyes ranged from 10 (absent) to 71 (high-risk proliferative DR). The manually detected rates of ≥2-step worsening in sham study and fellow eyes at M24 were 9.6% and 11.7%, respectively. The best DL algorithm was able to predict ≥2 step ETDRS DRSS worsening at M6, M12, and M24 at an AUC of 0.68 ± 0.13 (sensitivity 66% ± 23% and specificity 77% ± 12%), 0.79 ± 0.05 (sensitivity 91% ± 8% and specificity 65% ± 12%) and 0.77 ± 0.04 (sensitivity 79% ± 12% and specificity 72% ± 14%), respectively. Conclusions: Our pilot work established the feasibility of developing an automated algorithm for predicting patients with significant DR worsening over a 2-year period. Our data generated on eyes with a broad range of DR severities suggest a possible presence of predictive signals that precede microvascular abnormalities detectable to human graders. Validation in a real world setting is needed to make the algorithm generalizable to the overall population with diabetes. (No Image Selected) CONTROL ID: 3193811 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0107 TITLE: Image quality assessment of ultra-widefield fundus images using deep convolutional neural networks. FIRST AUTHOR: SANDIPAN CHAKROBORTY AUTHORS/INSTITUTIONS: S. CHAKROBORTY, K.R. Ranipa, Carl Zeiss India (Bangalore) Pvt. Ltd., INDIA|K. Makedonsky, P. Sha, M. Chen, K. Brock, M.K. Durbin, Carl Zeiss Meditec, Inc., Dublin, CA, United States., Dublin, California, UNITED STATES| Purpose: Insufficient image quality of fundus images results in impairment of manual or automatic image grading process, resulting in loss of a data for that particular visit. Automated evaluation of image quality would help the operator by giving a quick feedback to reacquire the images when image quality is inadequate. In this paper, we propose a deep neural network based approach that predicts the quality of a high resolution fundus image immediately after acquisition. Methods: We trained a deep Residual Network (ResNet) using TensorFlow for the task of image quality prediction. TM For model training, we used 561 good & 74 bad quality images taken using CLARUS 500 (Zeiss, Dublin, CA ). The test set has 270 images of good quality and 28 of inadequate quality. Bad quality images used for training is about ~12%, which is greater than the average occurrence of bad quality images in any random image set, to address the issue of unbalanced classes in training data. The most common artifacts which obscure the clinically useful information and interfere with the ability to interpret the image are severe blurring, reflex, vignetting, striping, or rainbow discoloration as shown in Fig. 1. Given the very small dataset and unbalanced sample size, we use a data augmentation step at the time of model training by applying random flips to the images. To speed up the training and inference, we downscaled the high-resolution images and cropped each image to 224 x 224 pixel each. Results: The results are shown in Fig. 2. The algorithm achieves an Area under the Curve (AUC) of 97.20%. Out of the 9 misclassified images, 66% of them are bad quality images, which are classified as good quality images. Conclusions: We present a solution for automated image quality assessment of fundus images taken by CLARUS 500. The presented deep learning algorithm achieves a very promising result even with a handful of data. Thus, it enables the operator to obtain a feedback on image quality at acquisition time and to reacquire images when image quality is poor. Fig. 1: Shows images used in model training (a) Good Quality Images (b) Bad Quality Images
Fig. 2: Results for Automated Fundus Image Quality Assessment.


CONTROL ID: 3194347 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0112 TITLE: Early Stage Glaucoma Diagnosis by Artificial Intelligence Assisted Multifractal Functional OCT FIRST AUTHOR: Subrata Batabyal AUTHORS/INSTITUTIONS: S. Batabyal, S. Kim, S. Mustafi, W. Wright, S. Mohanty, R&D, Nanoscope Technologies LLC, Bedford, Texas, UNITED STATES| Purpose: Glaucoma is a leading eye disorder which has few symptoms and difficult to detect in early stages. The detection of disease progression remains challenging in glaucoma due to the variable and slowly progressive nature of the disease, measurement-variability in standard assessment procedure and of imaging devices, and the lack of a commonly acceptable reference standard. Herein, we describe the development of multifractal OCT with added artificial intelligence (AI) to detect early signs of glaucoma from structural and functional imaging. Methods: Superior nano/micro-structural alteration and movement associated with retinal/trabecular meshwork dysfunction is obtained by locally connected fractal dimension analysis of B and C-scan OCT image. The stimulation enabled fluctuations in retinal ganglion cell (RGC) layer and pulsatile motion of the trabecular meshwork (TM) is probed by multifractal analysis of the spatial and temporal-varying phase/intensity signal of OCT scan. Based on this method, we have developed a multifractal OCT device for optically detecting changes in RGC/TM activities for early diagnosis of glaucoma. Feature extraction from multifractal parameters is achieved by AI. A large set of multifractal images are used in training and weight calibration, which is validated with respect to ground truth. Results: Our preliminary studies based on Multifractal OCT system could differentiate between wild type (control) and mouse with RGC dysfunction. Successful identification of glaucoma in mouse model was possible by analyzing the functional and structural OCT data using multifractal algorithm. Conclusions: Taken together, the AI assisted multifractal OCT system will pave the way for a clinically translatable approach for early stage glaucoma detection. People at greater risk for developing glaucoma can greatly benefit with such early diagnostics for timely intervention. (No Image Selected) CONTROL ID: 3194600 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0108 TITLE: Deep learning based robust fovea localization using OCT Angiography FIRST AUTHOR: Homayoun Bagherinia AUTHORS/INSTITUTIONS: H. Bagherinia, M. Durbin, Carl Zeiss Meditec, Inc., Dublin, California, UNITED STATES| Purpose: Automated analyses of retinal disease use the location of fovea as a reference point. Finding the fovea location using structural OCT en face images in disease cases can be a hard problem due to the poor contrast in structural OCT enface and loss of foveal pit. In contrast, OCT-A superficial slabs provide high contrast vasculature images. This abstract proposes a robust method to find the center of the fovea in OCT-A images using a convolutional neural network (CNN) architecture. Methods: Images from normal and eyes with retinal disease such as AMD and DR were acquired with field of views TM ® (FOV) of 3x3mm, 6x6mm, 8x8mm, 9x9mm, 12x12mm, 15x9mm using CIRRUS HD-OCT 5000 with AngioPlex ® OCT Angiography, and PLEX Elite 9000 SS-OCT (ZEISS, Dublin, CA). Superficial capillary plexus vasculature images were generated and framed into a 512x512 pixel image over 12x12mm FOV to create the training and testing images. The fovea location of 1330 images (1064 and 266 for training and testing) has been marked by a human expert. The training images were augmented by rotating each image around the center between ±10 degrees to increase the training images to 7448. The target images are 3mm diameter binary discs centered at the fovea location. A U-net with 5 contracting and 5 expansive layers with rectified linear unit, max pooling, binary cross-entropy loss, and sigmoid activation in the final layer were used for an end-to-end training. The fovea center was detected by template matching using the predicted image and the 3 mm binary disc. Results: Figure 1 shows examples of fovea locations detected by the algorithm and human expert with corresponding predicted area of 3mm diameter disc centered at the fovea for normal and various disese cases such as AMD and DR. Figure 2 shows a difference plot of the fovea location detected by the algorithm and human expert. The success rate is 97% for an error smaller than 150 μm between the algorithm and human expert. Conclusions: We demonstrated a robust method to find the center of the fovea in OCT Angiography images using a U-net architecture. Robust detection of the fovea location is crucial for automated analyses of retinal disease. Fig 1: Examples of fovea location detected by the algorithm (red cross) and human expert (green cross) with corresponding predicted segmentation of 3 mm diameter disc centered at the fovea. Fig 2: Difference plot of fovea location detected by the algorithm and human expert. CONTROL ID: 3194604 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0100 TITLE: Automatic detection of optic nerve head in widefield OCT using deep learning FIRST AUTHOR: Ali Fard AUTHORS/INSTITUTIONS: A. Fard, H. Bagherinia, Carl Zeiss Meditec, Inc., Dublin, California, UNITED STATES| Purpose: Localization of the optic nerve head (ONH) in OCT and OCTA images is of crucial importance for accurate analysis of the peripapillary region. Automated detection of this landmark is particularly challenging in widefield OCT images of diseased eyes due to the presence of pathologies. Here we present a robust deep learning method for automatic ONH detection in OCT images of healthy and diseased eyes. Methods: Our method employs a convolutional neural network to segment ONH regions in OCT en face images. OCT en face images were generated from outer boundary of outer plexiform layer to 0.5mm below retinal pigment epithelium in 1064 6x6mm and 12x12mm OCT volumes acquired using PLEX® Elite 9000 SS-OCT (ZEISS, Dublin, CA). The ONH center was marked in the images (821 for training and 243 for testing) by human experts. All OS eyes were flipped along the vertical axis to match the OD eye for training purposes. A 3-channel U-net architecture was used in which 5 contracting and 5 expansive convolutional layers, ReLU activation, max pooling, binary cross entropy loss, and sigmoid activation in final layer were employed. The input channels were OCT en face, vessel enhance OCT en face, and OCT contrast map (intergral of absolute axial gradient) (Fig 1). A 4mm binary mask was created as the target image around the grader-indentified ONH center. The binary mask was shifted by 1-mm temporally due to the location of ONH at the edge of the scan. Data augmentation (rotation around the center between -9° and 9° with a step of 3°) was performed to increase the number of training images. The U-net predicted the ONH area followed by a template matching using a 4mm diameter disc to find the ONH center. Results: Figure 2 shows examples of test images with predicted ONH area, and success rate and histogram plots of the error between the ONH detected by the algorithm and human expert. As it is evident, the algorithm identified the ONH with an accuracy of 300µm and 250µm in 98% and 95% of all cases, respectively. The mean and standard deviation of the error were found to be 97µm and 76µm. Conclusions: Our results suggest that the ONH can be robustly and accurately detected in OCT en face images using a U-net architecture in presence of pathology. Fig.1: Example of training input (A-C) and target output (D) of the algorithm. Fig.2: (A1-A4): Test input and output images. (B-C) Success rate plot and histogram of error in detecting the ONH center. CONTROL ID: 3194734 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0110 TITLE: Deep Learning Based Method for Retinal Layer Segmentation In Optical Coherence Tomography Images FIRST AUTHOR: Ivana Zadro AUTHORS/INSTITUTIONS: I. Zadro, S. Lončarić, Faculty of Electrical Engineering and Computing, Zagreb, CROATIA|M. Radmilovic, Z. Vatavuk, Department of Ophthalmology, University Hospital Centre “Sestre milosrdnice“, Zagreb, CROATIA| Purpose: Manual segmentation of retinal layers in pathological optical coherence tomography (OCT) scans is time- consuming, expert-grader dependent and prone to errors. Because of this, an automatic process of retinal layer segmentation is needed. The objective of this study is to investigate the applicability of the U-Net network with a postprocessing method for automatic segmentation of retinal layers from a database of OCT images with age-related macular degeneration (AMD). Methods: The method consists of three steps: the first step is the U-Net neural network for layer segmentation. The second step is a postprocessing method which corrects the neural network outputs by utilizing the following a priori knowledge about the retinal layers: a) the layers are topologically ordered, b) the layers cannot be intertwined. The third part is a Canny edge filtering used for layer boundary extraction. The expert grader annotated four layer boundaries in a total of 1270 B-scans: inner limiting membrane (ILM), inner nuclear layer/inner plexiform layer boundary (IPL/INL), retinal pigment epithelium (RPE) and Bruch’s membrane (BM). The proposed model was trained and validated on 23 macular spectral-domain OCT volumes of eyes with age-related macular degeneration. To evaluate a model a leave-one-out volume validation was repeated 23 times. The Dice similarity index was used for layer segmentation accuracy whereas the accuracy of the layer boundary segmentation was evaluated by the average surface distance error (ASDE). Results: Average Dice similarity indices for three layers and surface ASDE metrics for four layer boundaries across all 23 validation experiments are given in Table 1. The comparison between the manual segmentation and the segmentation using the proposed method is shown in Figure 1. Figure 1 (a) shows the original OCT image with manually segmented layer boundaries, Figure 1 (b) is the corresponding U-Net network output, Figure 1 (c) is the result of the postprocessing step, and Figure 1 (d) shows the final result after applying Canny edge filtering. Conclusions: The average Dice similarity indices above 90% for all three layers and low average surface distance errors for all four layer boundaries indicate that the proposed method can be used for effective automatic segmentation of retinal layers, even in the presence of retinal pathological changes. Table 1. Figure 1. a) b) c) d) CONTROL ID: 3194976 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0111 TITLE: Predicting laterality in external eye images using deep learning FIRST AUTHOR: Gary Lee AUTHORS/INSTITUTIONS: G. Lee, T. Callan, C. Wu, A. Tamhankar, A. Covita, M. Durbin, Carl Zeiss Meditec, Inc., Dublin, California, UNITED STATES| Purpose: Ophthalmic devices often acquire ancillary eye images during clinical testing. For example, external eye photos are often used for gaze tracking in perimetry, where the visual field can be highly dependent on laterality for expedited testing and post-processing. In this study, we explored the use of deep learning (DL) approaches to predict laterality in eye images. 2 Methods: A total of 22,185 grayscale eye images (472x472 pixels, 40.7x40.7 mm ) were extracted from visual field exams previously acquired on HFA3 perimeters (ZEISS, Dublin, CA) from 134 eyes (83 subjects, age range: 23 to 72 years) in a series of ethics board-approved clinical research studies. Images were split into training (12,770 images, 94 eyes, 53 subjects), validation (3,517 images, 20 eyes, 10 subjects), and test sets (5,898 images, 20 eyes, 20 subjects) in a 70:15:15 ratio of eyes, restricting subjects to a given set. The test set contained 11 right (OD) and 9 left (OS) eyes. Two convolutional neural networks (CNNs) were trained with augmentation on adaptive histogram equalized input images in Tensorflow/Keras. VGGfrozen used a well-known CNN (VGG-16), pre-trained on ImageNet and frozen, replacing the fully connected (FCN) layers with two FCN layers (100 neurons + ReLU + dropout) and a final FCN layer (2 neurons + softmax). Images were resized to 224x224x3. VGGmini used the first three convolution/pooling blocks of VGG-16 (8-fold filter reduction) with the same final FCN layers used in VGGfrozen. Images were resized to 64x64. Performance was assessed by computing global accuracy (percent of all images correct), class accuracy (mean of per eye accuracy), and gradient-weighted class activation maps (Grad-CAM). Results: Three errors were found for each CNN, resulting in global and class accuracies of 99.95% and 99.93% for VGGfrozen compared to 99.95% and 99.94% for VGGmini (see Table 1). Errors correlated to blinks, poor contrast, or features near the decision threshold (see examples in Figure 1 for original vs. input images plus Grad-CAM overlays). Conclusions: Classic DL approaches enabled accurate prediction of laterality in eye images in a preliminary cohort. In general, DL methods combined with relevant data and labels may provide solutions for related tasks such as gaze tracking, self-alignment, and finding metrics of test quality. Table 1. Summary of Networks and Performance
Figure 1. Example CNN outputs: (a) typical, (b) blink, and (c) poor contrast
CONTROL ID: 3195344 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0113 TITLE: Deep Neural Network Segmentation of Optical Coherence Tomography Angiography for Diabetic Retinopathy FIRST AUTHOR: Julian Lo AUTHORS/INSTITUTIONS: J. Lo, M. Heisler, D. Lu, M.V. Sarunic, School of Engineering Science, Simon Fraser University, Burnaby, British Columbia, CANADA|S. Karst, V. Vanzan, E.V. Navajas, Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, CANADA| Purpose: Diabetic retinopathy (DR) is a microvascular complication of diabetes manifesting in abnormal retinal circulation. The purpose of this study is to investigate the viability of machine learning algorithms in the automatic segmentation and quantification of retinal microvasculature and inter-capillary spaces in optical coherence tomography angiography (OCTA) images in clinical diabetic populations. Methods: A convolutional neural network (CNN) was used to generate a binarized segmentation of the retinal microvasculature, followed by additional post-processing to analyze inter-capillary spaces. OCTA images of both healthy and diabetic patients across all levels of DR severity were acquired using three commercial systems (PLEX Elite 9000, Zeiss, Inc.; AngioPlex, Zeiss, Inc.; AngioVue, Optovue, Inc.), as well as one prototype swept-source OCT system. The retinal layers were segmented to separate the superficial and deep capillary complex. Manual segmentation of the microvasculature was performed on 80 images, of which 60 were used for training, and 20 for validation and evaluation. Inter-capillary spaces were identified based on two metrics: the area of the region, as well as the distance from the centroid to the nearest capillary. Results were compared to a reference set of 10 healthy subjects and labeled in standard deviation maps. Results: Across OCTA images from both healthy and diabetic patients, the CNN segmented the microvasculature with an accuracy of 86.77%, with a Dice similarity index of 83.94% to the manual segmentations. Representative performance of the automated segmentations is shown in Fig. 1 (a)-(d). Fig. 1 (a) shows the original OCTA image, (b) the microvasculature segmentation, and (c) and (d) the standard deviation map based on area and the distance to the nearest capillary, respectively. Conclusions: The CNN-generated segmentations of the microvasculature had a high accuracy and similarity to manual segmentations across all OCTA systems. Automated quantification of inter-capillary spaces allows for accurate analysis of retinal circulation which is a biomarker for DR. Although the area measurements are useful in identifying large inter-capillary spaces, the distance to the closest capillary may present the highest clinical utility due to its direct correlation to the amount of circulation received. 2-3,,9 SD >BSD Fig. 1: An original OCTA image ( 6x.6mm field-of-view. supe1ficial capillary plexus) acquired using the Plex­ Elite 9000 is displayed in (a). with corresponding microvasculature segmentation in~ b I. with inter-capillary spaces la belled based on the above color scheme by ai·ea (c I and distance to closest capillary (d I CONTROL ID: 3195345 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0106 TITLE: Method for Prototyping Convolutional Neural Networks with Fewer Patients FIRST AUTHOR: Paul Lee AUTHORS/INSTITUTIONS: P. Lee, N. Moinfar, RCWNY, Clarence, New York, UNITED STATES|R. Metzinger, Tulane University, Louisiana, UNITED STATES| Purpose: Developing Convolutional Neural Network (CNN) for image classification requires a large training dataset and creates barrier to entry. We describe a method to train CNN models with fewer patient images by combining clinical knowledge and image processing to detect retinal hemorrhages. This method multiplies the data that can be extracted from each patient image to create training data. Methods: 40 retinal fundus images were validated to contain retinal hemorrhage by retinal specialists. Each image was standardized to 600x400 pixels from which 50x50 pixel images were sampled to yield 240 subimages per full image. To reduce overfitting, random areas were sampled, similar to bagging method used in Random Forest models. Each subimage was reviewed by retinal specialists to collect 1000 images, with and without retinal hemorrhages. The dataset was divided into 70:30 train:test data ratio.

CNN model was trained with python implementation of Tensorflow using Keras API. Binary crossentropy loss function, rectified linear unit activation function and "Adam" stochastic gradient method were used with 3 perceptron layers. The trained model was validated against the test data and 10 unseen images where each input image was broken down to 50x50 pixel units similar to the training data. Patient was labeled as positive if the model indicated a retinal hemorrhage in any of the subimages. Results: Total of 2000 images were created and labeled for training and testing data. Sensitivity and specificity were measured at 77% and 65% respectively. The optimal model was obtained using 3 layers with 128 inputs and two dense layers. Conclusions: We applied clinical insight to reduce the amount of patients needed to create training data to train a CNN model to detect retinal hemorrhages. The insight was that retinal hemorrhages can take on certain morphologies and that the presence of any retinal hemorrhage is pathological. This allowed us to reframe the question from searching for retinal hemorrhages in the entire fundus photo, to instead searching in smaller subsections. In other words, we reduced the size of the hay stack in searching for the needle. We don’t claim to compete against larger, well-designed efforts with better results; rather, we offer a method for prototyping initial models that can be applied without requiring significant investment in obtaining data 50x50 pixel of retinal hemorrhage 50x50 pixel of no retinal hemorrhage CONTROL ID: 3195385 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB091 TITLE: Eliminating retinal vessel shadows in en face choroidal OCT via generative adversarial networks FIRST AUTHOR: Jianlong Yang AUTHORS/INSTITUTIONS: J. Yang, H. Zhang, K. Zhou, Y. Hu, J. Liu, Cixi Institute of Biomedical Engineering, Chinese Academy of Sciences, Ningbo, CHINA|K. Zhou, School of Information Science and Technology, ShanghaiTech University, CHINA|F. Li, X. Zhang, Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, Sun Yat-sen University, CHINA|C. Zheng, Ophthalmology of Shanghai Children’s Hospital, CHINA|J. Liu, Department of Computer Science and Engineering, Southern University of Science and Technology, CHINA| Purpose: In the OCT imaging of the eye, the morphology of the choroidal vasculature is influenced by the vessel shadows from the retinal layers. Recently, an attenuation compensation (AC) approach to enhance the B-frames was proposed for eliminating the retinal vessel shadows. From the en face OCT view, the shadow removal can be seen as an inpainting task, which refers to repairing the shadow-contaminated regions of the choroid. In this work, we propose to use a two-stage generative adversarial network (GAN) for this task and compare its performance with that of a traditional coherence transport inpainting (CTI) method. 2 Methods: The OCT data sets we used were 6×6 mm volumetric scans from a Topcon 100-kHz swept source system. We employed a two-stage GAN inpainting architecture as shown in Fig. 1. The retinal vessel mask was extracted from the RPE layer. It was fed into the network together with the input en face choroidal OCT and its Canny edge map. The first stage of the GAN was used to connect the vessel edges at the locations of the shadow mask. Then the connected edge map combining with the en face OCT image were used in the second stage of the GAN to finish the shadow removal task. Our generators used Perceptual GAN architecture and the discriminators used PatchGAN architecture. Two fine-tune layers were added at each stage to fit our gray scale images with the pre- trained RGB models. Results: As shown in Fig. 2, the AC method is capable of removing small-vessel and capillary shadows while leaves the large-vessel shadows unaffected. The inpainting-based methods can entirely eliminate the shadows, but the CTI method brings smeary and blurry patterns around the inpainting regions. The proposed GAN-based method, on the other hand, is able to avoid generating artificial patterns and preserve the shape, connectivity, and contrast of the choroidal vessels. Conclusions: The proposed GAN-based method outperforms the existing AC and CTI methods in both the vessel elimination and preserving the morphology of the choroidal vasculature. It will benefit the quantification of the OCT- based analysis and diagnoses in ophthalmology. Architecture of the two-stage generative adversarial network for the shadow removal task. Comparison of different methods for eliminating the retinal vessel shadows. The retinal vessel masks generated from the en face RPE image are presented here for indicating the locations of the shadows. CONTROL ID: 3195456 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB095 TITLE: Real-time Scene Understanding in Ophthalmic Anterior Segment OCT Images FIRST AUTHOR: Hessam Roodaki AUTHORS/INSTITUTIONS: H. Roodaki, A. Eslami, Carl Zeiss Meditec, Munich, GERMANY|H. Roodaki, M. Grimm, N. Navab, Technische Universität München, Munich, GERMANY|N. Navab, Johns Hopkins University, Baltimore, Maryland, UNITED STATES| Purpose: Machine learning algorithms are useful and efficient at interpreting medical images and segmenting anatomies. Here we present an approach that goes one step further by gaining scene understanding using cutting- edge machine learning techniques. Our method reliably detects anatomies of the anterior segment of the eye in OCT B-scans and implicitly understands the location of acquisition. Methods: The utilized neural network architecture in our work is U-net, a convolutional classifier without any fully connected layers, with multiple modifications. Batch renormalization is introduced to increase training efficiency. Squeeze and excitation layers are added to improve interdependencies between channels. Dilated convolutions are also used to increase the receptive field of the network. Our design emphasizes on scene understanding to accurately learn the correct position of anatomies relative to each other. The ADAM algorithm is used for training with cross entropy loss as cost function. The neural network classifies input image pixels as one of cornea, sclera, iris or background classes. A spectral domain OCT system is used for data acquisition. An automated method is employed to capture random OCT B-scans with varying parameters such as size, scale, location and gain from ex-vivo porcine eyes as training dataset. Multiple images are captured from each location as a form of data augmentation. Annotation of the anatomies in the acquired dataset is initialized by multiple automated algorithms and then manually refined. Results: In total 7503 training images and 1136 validation images are used. The network achieves an accuracy of 95.62% pixel classification over all classes and the entire validation dataset. Inference on a 1024×1024 OCT B-scan takes about 50 milliseconds. Conclusions: We have presented a reliable method using machine learning for real-time OCT anatomy classification with acceptable accuracy. The algorithm succeeds in segmentation independent of the input image size, scale or location. However, to train a neural network effectively for clinical use, gathering large datasets of human subjects or domain adaptation is crucial. Ophthalmic anterior segment OCT B-scans and anatomy classification results. Red represents cornea, green is sclera, and blue shows iris. CONTROL ID: 3195627 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0114 TITLE: Artificial Intelligence Screening for Diabetic Retinopathy: Analysis from a Pivotal Multi-center Prospective Clinical Trial FIRST AUTHOR: Malavika Bhaskaranand AUTHORS/INSTITUTIONS: M. Bhaskaranand, K. Solanki, C. Ramachandra, S. Bhat, EYENUK, INC., Woodland Hills, California, UNITED STATES|S. Sadda, Doheny Eye Institute, Los Angeles, California, UNITED STATES|J.I. Lim, University of Illinois Chicago, Chicago, Illinois, UNITED STATES| Purpose: Evaluate an artificial intelligence (AI) system to screen people with diabetes at point-of-care for diabetic retinopathy (DR) including diabetic macular edema (DME). Methods: We conducted a prospective multi-center study (NCT03112005) in which patients with diabetes were enrolled consecutively initially and later preferentially based on enrichment criteria. The study subjects [MB1] underwent undilated 2-field fundus photography (macula centered and disk centered images) for the EyeArt AI eye screening system and dilated 4-wide field stereoscopic fundus photography. The EyeArt system provided eye-level results for referable DR (rDR), which is defined as moderate non-proliferative DR (NPDR) or higher (International Clinical DR (ICDR) severity scale) or clinically significant DME. The EyeArt system was evaluated against the clinical reference standard based on adjudicated grading of the 4-wide field photographs by expert graders at the Wisconsin Fundus Photograph Reading Center using the Early Treatment Diabetic Retinopathy Study (ETDRS) Severity Scale. Statistical analyses were performed with variance adjustment to account for the correlation between eyes of the same patient. Results: 1822 eyes from 911 subjects were included. Of these, 1718 eyes were gradable using the clinical reference standard and 326 of these were positive for rDR (290 moderate NPDR, 4 severe NPDR, 31 proliferative DR, and 85 clinically significant DME) and 1392 eyes were negative for rDR (1134 no apparent DR and 258 mild NPDR). Sensitivity of the EyeArt system using undilated images was 95.5% [95% CI: 93.0% - 97.9%], specificity was 86.0% [95% CI: 83.8% - 88.3%, and gradability rate was 87.5% [95% CI: 85.4% - 89.7%]. With a dilate-if-needed photography protocol (where dilated images were used for subjects with ungradable EyeArt results on undilated images), the gradability rate of the EyeArt system improved to 97.4% [95% CI: 96.4% - 98.5%], sensitivity was 95.5% [95% CI: 93.1% - 97.8%], and specificity was 86.5% [95% CI: 84.4% - 88.6%]. Conclusions: The EyeArt AI system compared favorably with the clinical reference standard from 4-wide field stereoscopic images and met the predetermined sensitivity and specificity endpoints for the detection of referable DR in people with diabetes. (No Image Selected) CONTROL ID: 3195650 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB096 TITLE: Artificial intelligence-based automated segmentation of subretinal fluid and subretinal pigment epithelial fluid in patients with chronic central serous chorioretinopathy (CSC) FIRST AUTHOR: Mustafa Safi AUTHORS/INSTITUTIONS: M. Safi, R. Goldberg, Ophthalmology, California Pacific Medical Center, San Leandro, California, UNITED STATES| Purpose: Automated image analysis using artificial intelligence-powered software offers the promise of more efficient detection, identification and analysis of ocular pathology. Pegasus, an automated AI decision support platform, was used to identify both subretinal fluid (SRF) and sub-retinal pigment epithelium fluid (PED) in patients with chronic CSC (Figure 1), and the accuracy of these results was evaluated. Methods: Optical coherence tomography (OCT) volume scans of patients with CSC who participated in the Short Term Oral Mifepristone for CSC (STOMP) study were evaluated by the Pegasus system (Visulytix, London, UK). The results were then evaluated by two graders, and scored for the entire volume scan from 0 – 4, based on the accuracy of the automated detection results (0, 0-20% of fluid detected; 1: 21-40%, 2: 41-60%; 3: 61-80%; 4: 81-100%). Scores were given to both the SRF and PED volumes and averaged; qualitative notes were obtained as well. Results: 30 eyes were evaluated at 5 different time points; a total of 145 OCT volume scans were assessed. The mean score for the accuracy of SRF and PED detection were 3.5 and 1.9, respectively. The majority of scans were graded as a 4 for SRF detection (64%), while PED detection was scored a 4 by both graders only 32% of scans. Eyes with large SRF and serous PEDs were, for the most part, accurately detected, though as the fluid waned during the course of the STOMP study, very small slivers of SRF or PED went under-detected. The presence of sub-retinal hyperreflective material on OCT seemed to present the largest hurdle for the accurate quantification of SRF (Figure 2). Of note, there was less variability in the SRF scoring than the PED scoring (p<0.05). Conclusions: The Pegasus AI automated AI decision support system performed well to detect SRF and PED volume in a population of CSC patients, and may be helpful as a tool to identify and follow these patients over time. Expanded data sets may help further improve the detection algorithms, especially in a dynamic disease like CSC, where the fluid status can fluctuate significantly over short time periods.

CONTROL ID: 3195678 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0103 TITLE: Advantages and disadvantages of using deep learning-based image deconvolution on fundus images FIRST AUTHOR: Michael Chen AUTHORS/INSTITUTIONS: M. Chen, M. Durbin, N. Manivannan, K. Brock, Carl Zeiss Meditec, Inc. , California, UNITED STATES|L. Omlor, Carl Zeiss AG, GERMANY| Purpose: Due to differences in eye anatomy, local variation in image blur can be a problem in widefield fundus images. This forms a spatially varying defocus estimation problem that has recently been addressed using multi-scale deep learning (DL) methods for blind motion deblurring. This study aims to characterize the performance of a DL network to improve vessel sharpness in fluorescein angiograms. Methods: We adapted the multi-scale approach mentioned above, but replaced the residual networks blocks with (residual) U-nets. For training, sharp patches from fluorescein angiograms were blurred with point spread functions modelled with realistic aberration parameters. The network was trained using 30,000 patches of size 256x256. Each patch was deconvolved separately and recombined into the final image.

Images were graded by a clinician. Overall sharpness was graded from 1 to 5, with 5 being the sharpest. To further characterize the clinical utility of the images, a more detailed grading was done in the areas of disease, and evaluated in the context of dye transit time and disease state. The appearances of retinal and choroidal vessels were graded separately as better, same, or worse than the original. Results: Forty-one images were included in the validation set, and included images from different dye transit times and diseases. Processed images were graded as the same or better in overall image sharpness in 90.2% of images, in retina appearance in 82.9% of images, and in choroid appearance in 97.6% of images.

In 8 cases, the processed image was graded as worse. In these cases, a more detailed analysis was done. In 6 cases, small vascular details were removed. In 1 case, perivascular dye leakage appeared sharpened. In another, the vessel-like structures were generated in an area with no blood vessels. Conclusions: Caution should be taken when applying DL for image processing. Because DL relies on large amounts of annotated data for training, rare or atypical diseases may have a higher chance of producing poor results.

In this study, despite general improvements to image sharpness, some images showed undesired results. The purpose of fundus imaging is to reliably depict the anatomy of the eye, allowing clinicians to rely on the images in medical decision making. Changes that add or remove features may affect clinical utility and should be avoided. Original (left) image and DL-generated (right) image. CONTROL ID: 3195681 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB086 TITLE: Robust Nonperfusion Area Detection in Three Retinal Plexuses using Convolutional Neural Network in OCT Angiography FIRST AUTHOR: JIE WANG AUTHORS/INSTITUTIONS: J. WANG, T. Hormel, Q. You, Y. Guo, D. Huang, T. Hwang, Y. Jia, Casey Eye Institute, Oregon Health&Science University, Portland, Oregon, UNITED STATES|J. WANG, Y. Jia, Department of Biomedical Engineering, Oregon Health&Science University, Oregon, UNITED STATES|X. Wang, Shanxi Eye Hospital, Taiyuan, Shanxi, CHINA|L. Chen, Department of Computer Science & Electrical Engineering, Oregon Health&Science University, Oregon, UNITED STATES| Purpose: To develop a robust nonperfusion area (NPA) detection method in three retinal plexuses using convolutional neural network (CNN) in optical coherence tomography angiography (OCTA). 2 Methods: 3 × 3-mm OCTA scans were acquired on one eye by a 70-kHz OCT commercial AngioVue system (RTVue-XR; Optove, Inc) from 428 participants in clinical diabetic retinopathy (DR) studies. No scans were excluded due to image quality. OCTA scans from 10 healthy volunteers were degraded with manufactured artifacts from neutral density filters and defocus. En face superficial vascular complex (SVC), intermediate capillary plexus (ICP) and deep capillary plexus (DCP) angiograms were generated on projection-resolved OCTA. The angiogram and the corresponding en face reflectance map of each plexus, as well as the inner retinal reflectance comprised the input to the designed CNN. A trained grader determined the ground truth NPA by evaluating both the angiogram and the reflectance maps. We then randomly selected 59 diabetic and 5 healthy cases with manufactured signal reductions for testing. The remaining cases were used for training the CNN. To preserve the feature resolution and detect the features in multi- scales, the dilated convolution kernels replaced the pooling layers and parallelized the output layer by extending the field of view. The softmax activation function generated the probability maps of NPA output. Pixels with probability > 0.5 were classified as NPA. Results: On the healthy eye scans with manufactured signal reduction, the CNN consistently detected foveal avascular zones (FAZ) as NPA, regardless of the signal strength (Fig. 1). On the scans from clinical studies, NPAs were effectively detected in DR subjects of various disease stages with minimal interference by low signal strength and bulk motions. On all testing scans with the wide range of signal strength index (SSI: 44-88), the mean intersection-over-union (mIOU) of NPA detection was 0.81±0.11 in all layers (Fig. 2), indicating the high accuracy of the algorithm. The coefficient of variation of NPA detection on healthy eyes between scans within same visit was 0.07±0.01, suggesting the high repeatability. Conclusions: The proposed algorithm detects NPA with a high level of agreement with manual grading in all retinal capillary plexuses, regardless of manufactured signal strength attenuation or low signal quality in clinical studies. NPA on healthy eyes with signal reduction attenuated by NDFs

Optical density=O Optical density=0.2 Optical density=0.4 Optical density=0.6 SSI= 82 SSI=73 SSI=61 SS1=52

SVC

TCP

DCP

Fig. I Nonperfusion area (NPA) detection on scans of a healthy eye with manufactured signal strength reduction. T11e detected NPA regions weie labeled with blue and overlaid on angiograms. Rows show different anatomical slabs (top ~ SVC; center~ ICP; and bottom DCP) with different degrees of signal strength reduction in each colmnn. SVC: superficial vascular complex ~ which is defined by the inner 80% of the ganglion cell complex (GCC); ICP: intem1ediate capillmy plexus which is defined by the outer 20% of the GCC and inner 50% of the inner nuclear layer; DCP: deep capilla1y plexus which is defined by the outer 50% of the INL and outerplexif01m layer (OPL). NPA on severe DR Angiogram Ground Tn1th NPA Probability Outputs

SVC

ICP

DCP

Fig.2 Nonperfusion area (NPA) detection on low quality scans of severe diabetic retinopathy. Rows indicate anatomical slab. Column 1: the input angiogrnm; colmnn 2: grnund tmth of NPA ; column 3 :· associated probability map from the CNNs ; colunm4: algorithm outputoflabeled AA. CONTROL ID: 3195697 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB092 TITLE: OCT image noise reduction using deep learning without additional priors FIRST AUTHOR: Arindam Bhattacharya AUTHORS/INSTITUTIONS: A. Bhattacharya, M.K. Durbin, Carl Zeiss Meditec, Dublin, California, UNITED STATES| Purpose: Optical coherence tomography (OCT) imaging quality is often limited by various noise sources, which may hinder the ability to visualize fine tissue features. Recent advances in deep learning techniques have made it possible to perform effective image noise reduction. Such methods based on image translation require prior knowledge such as higher-resolution images or pre-defined noise characteristics. We propose a formulation of the noise reduction problem based on the U-net architecture which does not require additional prior knowledge by directly learning from the input images. Methods: A CIRRUS™5000 HD-OCT with AngioPlex® OCT Angiography (ZEISS, Dublin, CA) was used to acquire 3x3mm scans (245 B-scans, 245 A-scans/B-scan and 1024 pixels/A-scan). Each B-scan location was repeated 4 times. We trained a custom convolutional neural network on pairs of images from B-scans from the same scan. A pre- processing step registered the four B-scans within a cluster, and each cluster of registered B-scans was used to generate three pairs of training sets. Existing approaches using averaged images as priors suffer from ‘blurring’ caused by imperfect registration. Those instead using additive noise are biased towards the noise statistic added. The proposed network instead only learns to denoise the content noise characteristics. Our network is a modified five-layer U-net architecture with deep supervision. A custom loss function enforces a symmetric loss between training images. We also use a linear combination of L1 and L2 loss and deep supervision for faster convergence and accurate results. Results: Fig 1 shows the results of one of our test cases. First column shows single input slice and the corresponding output slice. The volume rendering shows the rendering of 50 slices (1024x245x50). The cropped images show closeups of regions of interest. The network learns anisotropic feature preserving smoothing. Fig 2 shows the histogram along the yellow line (one A-scan) for a single original slice in blue and network output in red. Following the curves, we see that the network decreases the noise without deteriorating the peaks representing the signal ranges. Conclusions: We present a robust feature preserving denoising method which can automatically learm the characteristic noise in OCT data without additional information. Fig. 1 Fig 2. CONTROL ID: 3195705 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB094 TITLE: A machine learning approach to predict response to anti-VEGF treatment in patients with neovascular age- related macular degeneration using SD-OCT FIRST AUTHOR: Jayashree Nair Sahni AUTHORS/INSTITUTIONS: J. Nair Sahni, A. Maunz, F. Arcadu, Y. Zhang_Schaerer, Y. Li, T. Albrecht, A. Thalhammer, F. Benmansour, Pharma Research and Early Development, F. Hoffmann-La Roche , Basel, SWITZERLAND| Purpose: The purpose of this study was to identify baseline spectral-domain optical coherence tomography (SD-OCT) features for predicting response to anti-VEGF therapy in patients with neovascular AMD, and to build a model capable of providing individual patient predictions. Methods: Clinical trial data of patients (N=793) with neovascular AMD treated with ranibizumab from all arms of the HARBOR study (NCT00891735) was used to train and validate the models to predict response. Response to anti- VEGF therapy was defined as Best Corrected Visual Acuity (BCVA) ≥20/40 at month 12 in those subjects with BCVA ≤ 20/40 at baseline. Baseline age, sex, SD-OCT, and BCVA data were included. Automated segmentation of retinal layers and fluid-filled regions over a 6 X 6mm cube of SD-OCT images centered on the fovea was used to extract 62 SD-OCT features: 44 layer related features in 2-D space, 9 layer related in a 3D space and 9 fluidic area related features in 3-D space. To gain insight into the CatBoost model trained from the data, SHAP (SHapley Additive exPlanations) method was used to interpret patient-level model predictions. Model performance was assessed in terms of area under the receiver operating curve (AUROC) using 5-fold cross validation. Results: AUROC to predict response at month 12 using only baseline data was 0.77 (95% CI 0.73 – 0.82). Baseline BCVA, central subfield thickness, central subfield volume, and intra-retinal fluidic volume were among the most impactful measurements to predict response according to SHAP analysis, driving predictions, up, down, down, and up, respectively, in favor of reaching BCVA ≥20/40. Conclusions: We proposed and evaluated a machine learning methodology to predict probability of achieving functional BCVA from SD-OCT scans taken at treatment initiation. In the new era of therapies targeting multiple pathways in the management of neovascular AMD, the results of this retrospective analysis allow identification of patients who are likely to be good responders to anti-VEGF, thus enabling selection of appropriate patient population for the novel therapies and a precision medicine approach. (No Image Selected) CONTROL ID: 3195718 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB097 TITLE: An Artificial Intelligence Deep Learning System for Discriminating Ungradable Optical Coherence Tomography Three-Dimension Volumetric Scans FIRST AUTHOR: anran ran AUTHORS/INSTITUTIONS: A. ran, A. NGAI, V. CHAN, J. Shi, C. THAM, C. CHEUNG, The Chinese University of Hong Kong, Hong Kong, CHINA| Purpose: The image quality of optical coherence tomography (OCT) scans is crucial for the accurate and reliable interpretation of retinal structures. Traditionally, signal strength is used as an index to include or exclude OCT scans for further analysis. However, it is insufficient to assess other image quality issues which require specialized knowledge in OCT for such assessment. Our study aims to develop and evaluate a deep learning system (DLS) as an automated tool for filtering out ungradable OCT volumetric scans. Methods: We proposed a two-stage method to improve the effectiveness of training and the generalization of DLS. In a total of 3205 optic disc OCT volumetric scans were extracted from Cirrus HD-OCT (Carl Zeiss Meditec, Inc., Dublin, CA, USA), in which contains 778 eyes collected from a tertiary eye hospital in Hong Kong for training (80%) and testing (20%). Each scan was labeled as gradable or ungradable by a trained grader and then by a senior grader to mitigate the dissonance. OCT volumes fulfilling either the criteria: 1) missing data, 2) motion artifacts, 3) blurry, 4) signal loss, or 5) poor centration were defined as ungradable. We developed a3D model based on RestNet18 structure to discriminate ungradable optic disc OCT scans from gradable ones. We further evaluated our system with two independent datasets of 1009 scans from 803 eyes collected from another two eye clinics. Receiver Operation Characteristics (ROC) curve, the area under the ROC curve (AUC), sensitivity and specificity were used to evaluate the performance. Results: A total of 4214 volumes were used. In the internal validation, the DLS achieved an AUC of 0.918 (95%CI, 0.895 to 0.942). The optimal sensitivity, specificity, and accuracy were 0.876, 0.846, and 0.856, respectively. In the two external validation datasets, the AUC were 0.777 (95%CI, 0.738 to 0.816) and 0.806 (95%CI, 0.769 to 0.864), with sensitivity of 0.787 and 0.718, specificity of 0.699 and 0.815, accuracy of 0.727 and 0.782, respectively. Conclusions: The presented DLS achieved a good performance to discriminate the ungradable from gradable OCT volumes in both internal and external validation. Tiibl@ 1 Char~btristia of the aII the iha:lr participants Table 2 The opllmum performance of th@ 30 de~ l@arni nB i)'5btm in both int..rnal a~ l!lh!rnal validation U'llJ'.Jd;jH Gra1fable Pllll~t AUC(MCIJ 511!ral1Mt, SpKlflclly Acuiraq- Tralni~ and Intimal ~alldation clata!.l!l 0.87e O.lilfi 0.856 l't>. cf OCT idLITlM g)g 2266 \ lnll'rniil O.!rul I'll. of p;iler1h 2(Jfi 2113 \ V;illdiitlon IOJPJ'l-0.'IU) (0,05 &!id111 ~male/llm;ijt) 11l/90 lllg,/m Eltlmll o.m O.JB7 O.ti!ll o.m i't>.Cff¥1!! E 476 1a.1J1-0.1116~ \ 't.!lldall111_1 E'f' Ir11it/ll 1t I H0/1Al w/m O.IZ Ertlm.11 11.JOfi o.ru 0.815 0.7112 ~ 1 1!!! jrlllliln t SDI u.mu 57..!W.8 O.!lO 10.~.86') Emmal ..alldlllioo dlllaset l jPWH) 'nlld;tlon_l l'll.clOChdLl'llB 202 ~ \ l't>. cf p;il@Plh 102 2119 \ Gendl!r jm;leffl!m;ij~) til/«J 118,IHl O

Agl!1~ jrnean t ~I Jl.fd:ll.ti tiUtll.J CD,O!i btmial 11Jlldali!lf1 d-'314! 2 ITl'tlEQ l't>. cf OCT idLITll!S 71 m \ I'll. cl p;ileri1 51 l4S \ GendH ~mill!/fl!rnalt) 31/20 flJ{lr. O.lD i'll.Ofl!l'BI 71 223 \ E'f' ~ r11it/ll 1t I 41/~0 lll8,t115 0.18 Af!1~ jlTINn t SDI fi1.&H5.0 fi0.m1.1 O.li:'i Cl...

Q) 0

1:- U) / ·a 0 """'r;; (!) '

N I 0 - l•nlllVlltdlliet1 - l.Jl•"*\l•k:l•b>!i. 1 0 -- c.i.m..iv.w.IUl'l_t 0

0.0 0.2 0.4 0.6 0.8 1.0 1 - Spe<:ificity

Flgurt 1. AUC of 3D deep learning S)'Stem in both internal and external validation CONTROL ID: 3195807 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB085 TITLE: Large Retinal Arterial Microaneurysm detection with Optical Coherence Tomography-Angiography FIRST AUTHOR: Konstantinos A.A. Douglas AUTHORS/INSTITUTIONS: K. Douglas, J. Diaz, P. Oellers, J.B. Miller, MASS EYE AND EAR, Massachusetts, UNITED STATES| Purpose: Retinal microaneurysms are small saccular outpouchings that arise from the retinal capillary system. The purpose of this study was to compare the ability of traditional imaging methods and Optical Coherence Tomography- Angiography (OCT-A) to detect retinal microaneurysms. Methods: This is a case of a 63-year-old healthy woman who was presented to our department in the context of a routine eye exam. She denied any vision changes. On examination, best-corrected visual acuity was 20/20 OU. Biomicroscopy of the anterior segment was unremarkable apart from mild nuclear sclerosis of both eyes. Dilated fundus examination of the left eye revealed small retinal hemorrhage with surrounding exudation, most consistent with a large retinal microaneurysm. Fluorescein angiography, OCT and OCT-A were performed. Results: FA showed correlating hyperfluorescence, consistent with a retinal microaneurysm (Fig.1).OCT-A with segmentation of the superficial retinal layers accurately depicted the exact anatomy of the microaneurysm (Fig. 2). In addition, structural en-face OCT revealed exudation nearby. OCT-A B-scan indicated blood flow through the aneurysm and a high resolution OCT image demonstrated hard exudates and focal retinal edema. The patient was referred to her internist for workup of cardiovascular risk factors. The lesion was observed with plans for laser treatment for foveal threatening complications such as progression of lipid exudates or development of intraretinal fluid. Conclusions: This case is interesting, since the aneurysm is fairly large, yet isolated without any other fundus abnormalities consistent with vascular disease. While microaneurysms can be visualized on fundus examination, further imaging modalities such as fluorescein angiography and OCT-A can be used to confirm the diagnosis, especially if they occur in the setting of hemorrhage and/or exudation. OCT-A imaging allows for efficient and noninvasive detection of a large retinal microaneurysms and provides detailed anatomical information.

CONTROL ID: 3195861 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0109 TITLE: Automated choroid segmentation of SD-OCT volumes using deep learning FIRST AUTHOR: Jonathan D Oakley AUTHORS/INSTITUTIONS: J.D. Oakley, D.B. Russakoff, Voxeleron LLC, Pleasanton, California, UNITED STATES| Purpose: To automatically segment the choroid’s posterior boundary in SD-OCT and evaluate for accuracy across different retinal pathologies. Methods: The posterior boundary of the choroid was manually delineated in 50 OCT volumes of the macula using Orion’s layer editing tool (Voxeleron, Pleasanton, CA). The volumes were chosen from a control population without retinal pathology with the following exclusions: axial motion issues, choroid boundary out of the field of view / not clearly defined. The OCT data was denoised and down-sampled to a volume of 128x128x128. Various deep convolutional neural network (CNN) U-Net architectures were explored/optimized to assign three labels to each OCT voxel: background, retina and choroid. 5-fold cross validation, where no eye from the same subject was in both training and test sets, was used to assess performance. Overall segmentation accuracy was reported using a dice coefficient to gauge the entire volume of overlap between the manual and automatic segmentations. To evaluate the method’s ability to generalize to unseen data and pathologies, the best architecture/model, trained on the previous 50 cases, was applied to a new data set comprising 30 volumes of mixed retinal pathologies acquired from a separate center. Results: During cross validation, the best segmentation network had average dice scores of 0.99 (STD=0.004), 0.99 (0.004) and 0.95 (0.019) for background, retina and choroid, respectively. On the unseen retinal pathology data, the dices scores were: 0.98 (0.009), 0.99 (0.006) and 0.91 (0.033). Example segmentations from the original data and the retinal pathology data are shown in Figs 1 and 2, respectively. Conclusions: Quantification of choroidal thickness can inform upon a variety of chorioretinal diseases. Such assessment is challenging, particularly in SD-OCT devices, but with suitable pre-processing, deep learning offers a ready solution. Comparison to ground truth shows excellent correlation and generalizability, paving the way for wider use in OCT, enhanced depth imaging, and swept source devices. Fig (1): (a) automated retinal segmentation from the control group (N=50) of the ILM (red) and RPE (blue) (Orion) and manual choroid segmentation (green); (b) the posterior boundary of the choroid is replaced with the deep-learning result. Fig (2): Example automated segmentation results based on the second data set of mixed retinal pathologies (N=30). CONTROL ID: 3195877 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB089 TITLE: Comparison of manual and fully automatic cell hexagonality measure in corneal endothelium images in transplanted corneas post Descemet Stripping Automated Endothelial Keratoplasty (DSAEK) FIRST AUTHOR: Naomi Joseph AUTHORS/INSTITUTIONS: N. Joseph, C. Kolluru, D. Wilson, Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, UNITED STATES|H. Menegay, J. Lass, B. Benetz, Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio, UNITED STATES|H. Menegay, S. Burke, J. Lass, B. Benetz, Cornea Image Analysis Reading Center, University Hospitals Eye Institute, Cleveland, Ohio, UNITED STATES|D. Wilson, Radiology, Case Western Reserve University, Cleveland, Ohio, UNITED STATES| Purpose: We developed deep-learning-based automatic cell segmentation and a hexagonality (HEX) estimation method for analysis of corneal endothelial cell (EC) images taken 6 months to 3 years post DSAEK. As manual analysis is very laborious, successful automatic estimation of HEX would allow comprehensive computational analysis of large datasets of over 1000s of EC images. Methods: We used a subset of 130 specular microscopy images from 26 eyes collected from the Cornea Preservation Time Study (CPTS). Trained readers performed traditional corner analysis using the CAS/EB software (HAI Labs Inc., Lexington, MA) to generate cell border contours by marking the corners of individual cells. The percentage of hexagonal cells (HEX) output from the software was recorded for each annotated image. In order to automatically compute the HEX metric for a given image, we first trained a deep neural network algorithm (U-Net) to segment ECs in the microscopy images. 65 images were used as the held-out test set. Next, segmentations from the network were binarized and cleaned using morphological operations. Finally, results of connected components analysis on the predicted masks were fed as inputs to a shape detection algorithm that estimates the percentage of hexagonal cells. Results: From the 65 test images, we found no statistically significant difference between the HEX measure from our fully automatic deep learning based segmentation approach and the ground truth reader annotations (p = 0.90). Over our heterogeneous test data, HEX values were 56.77 ± 0.91 and 56.91 ± 1.11 for manual and automated, respectively. Conclusions: We found that our shape detection algorithm, based on an iterative end point fit method, can closely estimate cell HEX in an automatic fashion on an initial discovery dataset containing 65 clinical quality specular microscopy images. Further work is necessary to improve the neural network predictions in order to achieve greater correlation between the measurement groups. Fig. 1: Results of algorithm. (a) Corner analysis with borders in green; (b) probabilities from the deep neural network; (c) results of algorithm on binarized network outputs with H=hex. Fig. 2: Bland Altman plot of HEX measures between the CAS/EB software manual analysis and our shape detection algorithm using the automatic segmentations. CONTROL ID: 3195887 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB090 TITLE: Identification of clinically relevant glaucoma biomarkers on fundus images using deep learning FIRST AUTHOR: Mohammad Norouzifard AUTHORS/INSTITUTIONS: R. Klette, EEE, Auckland University of Technology , Auckland, NEW ZEALAND|A. Nemati, School of Engineering and Technology, University of Washington, Tacoma, Washington, UNITED STATES| M. Norouzifard, H. GholamHossieni, School of Engineering, Computer and Mathematical Sciences, Auckland University of Technology, Auckland, NEW ZEALAND|K. Nouri-Mahdavi, Department of Ophthalmology, University of California Los Angeles, Los Angeles, California, UNITED STATES|S. Yousefi, Department of Ophthalmology, University of Tennessee Health Science Center, Tennessee, UNITED STATES| Purpose: To develop a deep learning framework that can automatically identify clinically relevant features on glaucoma fundus images. Methods: Fundus photographs from 267 normal eyes and 160 eyes with glaucoma were included. We developed a deep learning model based on the pre-trained NASNet and used heat map technique to assess parts of the fundus image that were driving the classification, thus allowing localization of clinically relevant objects on retinal fundus images. After training the model, all 427 fundus images were used as input to the proposed model (based on a deep pre-trained classifier) consisting of the region of interest on fundus photographs. The clinical diagnosis labels of fundus images were validated by a glaucoma specialist and the outcome of deep learning was assessed by experts to assure clinical relevance. Results: The accuracy of the method in discriminating normal eyes from eyes with glaucoma was 92%. The validation accuracy on an independent dataset of 455 images was 90%. Among fundus images that had been classified to glaucoma group, we observed that deep learning had identified significant features mostly in the superior/inferior peripapillary regions, within the optic nerve head, as well as in their pattern of large blood vessel structure. Conclusions: We developed a deep learning model based on pre-trained parameters that was able to detect clinically relevant glaucoma features from fundus images with high accuracy. This approach could be useful in glaucoma clinics as well as in general practice settings as an assistive tool for screening glaucoma in the absence of glaucoma clinicians. Validation of our findings in an independent cohort with larger number of fundus images is required. Figure 1. The utility of optic nerve head structure region in detecting glaucoma. Left: Optic nerve head structures identified as a significant feature for detection of glaucoma based on the proposed deep learning model, Right: A sample input fundus image overlaid on the optic nerve head structure in the left panel. Figure 2. The utility of blood vessel structure feature in detecting glaucoma. Left: Blood vessels structure identified as a significant feature for detection of glaucoma. Right: The input fundus image overlaid on the blood vessel structure in the left panel. CONTROL ID: 3195931 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB087 TITLE: Automated Detection of Retinal Fluid Using a Convolutional Neural Network FIRST AUTHOR: Tristan Hormel AUTHORS/INSTITUTIONS: T. Hormel, J. WANG, Q. You, D. Huang, T. Hwang, Y. Jia, Casey Eye, Oregon Health and Science University, Portland, Oregon, UNITED STATES| Purpose: Diabetic macular edema (DME) is a major cause of vision loss and is not amenable to screening with photographic methods. Retinal fluid (RF) on optical coherence tomography (OCT) may be an earlier sign than macular thickening to identify eyes at risk for vision loss from DME. We developed a convolutional neural network (CNN) for automated detection of RF in OCT volumes. Methods: A commercial spectral-domain OCT system (RTVue-XR, Optovue, Inc.) captured OCT macular scans 2 (304×304 A-lines covering 3×3 or 6×6 mm area) from both eyes of a total 172 patients with diabetes and generated 2150 scan volumes by including repeat visits. An expert graded each volume for the presence of fluid. We reserved 200 volumes for testing and used the remainder for training. We did not exclude any scans due to poor image quality. Our CNN architecture consists of data compression, feature extraction and decision-making steps (Fig. 1). Data compression was accomplished through successive convolution, batch normalization and rectified linear unit activation. Feature extraction was achieved by additionally including max pooling layers. Finally, fully connected layers and a softmax activation function made the prediction. Volumes with a probability over 50% of containing RF were classified as positive for its presence. Results: Compared to expert manual grading, the trained network classified the volumes for the presence of RF with an accuracy of 83.33%, a precision of 84.62%, and recall of 81.05% on the test data set. Fig. 2 shows example B- scans with the associated detection probability. Conclusions: Our CNN detects RF with high accuracy on a diverse data set regardless of scan quality or severity of 2 disease. Volumetric evaluation obviates the need for selection of specific B-scans. Inclusion of both 3×3 and 6×6 mm scans makes our results agnostic to scan dimensions. OCT dnla volume na1.n compr~sio11 Fenl.ure extrnct.ion Oecision making Cln.~sific111.ion

Retina l fluid ......

Kon-iluid containing

Figure I: ·' 'etwork arc.hitecture. The CNJ\' consists of three components: data oomprcs~ion , feature extraction, and llt:dsion making. Volume mm inpub ar<> Jirsl oornpr<>sseJ using convolulions, balch nonnalizalion, and n:clilieJ linear unit (ReUJ) activation. Features are then extracted hy additionall y including maximum pooling operations, and cla~s ifi cation is completed after flattened data is pa~sed through fully connected l ayer~ to a softmax decision. P=0.28 P=0.27 P=0.64 P=0.73

Figure 2: Examples of correctly labeled OCT volumes. True negative cases (blue borders; a and b) are predicted to contain retinal fluid (RF) witll probability less tllan 0.5; while true positive cases (red borders; c and d) are predicted to contain RF witll probability greater tllan 0.5. Examples (c) and (d) show tllat our CNNs is capable correctly identifying fluid even when tile volumes involved are quite small. Clinical Imaging CONTROL ID: 3181342 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0124 TITLE: Changes in Photoreceptor layer thickness in Optic Neuritis Follow up FIRST AUTHOR: Masoud Fard AUTHORS/INSTITUTIONS: M. Fard, A. Golizade, H. Ghahvechian, S. Yadegari, Farabi Eye Hospital, Tehran, IRAN (THE ISLAMIC REPUBLIC OF)|R. Ritch, Mount Sinai Hospital, New York, UNITED STATES| Purpose: Ganglion cell inner plexiform (GCIP) layer thinning following acute optic neuritis (AON) is well-known. However, the onset of changes in the outer retinal layers and their trend during the follow up period need further study. We examined the changes in outer retinal thickness during the first six months follow up of AON. Methods: 50 AON patients underwent optical coherence tomography (OCT) and visual function testing at baseline, one month, and 6 months after onset of AON. Longitudinal changes in retinal layer thickness were analyzed using linear mixed model. Results: Mean age at diagnosis was 31.9 ± 8.8 years. Mean visual acuity was 1.01± 0.83 logMAR that reached to 0.45 ± 0.13 logMAR at six months. Mean GCIPL thickness decreased at one month relative to baseline from 64.3 ± 7.2 µm to 55.9± 6.8 µm (P<0.001 for all macular sectors). There were no significant changes in GCIPL thickness between one and 6 months. ONL thickness in the 6 mm macular area increased from 58.6±6.8 µm to 61.2±8.1 µm at one month (P<0.001) and then decreased at six month (59.8±5.8 µm) relative to one month, reaching the baseline thickness. ONL thicknesses in the central 3 mm macular area were not significant at one and six months compared to baseline values. Outer plexiform thickness and retinal pigment epithelium were also unaffected during the course of optic neuritis. Conclusions: Following AON, transient changes occur in the photoreceptor layer and then revert to baseline. These findings help further our understanding of the pathogenesis of AON. one month six month

~ -'---~;'!~------' ~!~'!-~

-_ ~ ..... - - ,--. ~ ~ l;.v ...... -1 - r ___,..,,,~ -_,."

------~

. ~=--""''

- , _ \~ '1 _' ., . . . . '' . . - i :, 1 ~ "'-r·...., --· - ~ - .. -.0 ,..-.,.c ,. .• ~' r:i I ·1- / It: 1 . - . . I ·- . -

-

_:.._,, u!!! ~ •

:...... _ __ ,_ .. c - .. -- --- ~

I - -- .__ F _; ...... t . .- .- --.. I~-- . .

.. ::..-.. . - - · ~ - .. ---.. 1]f_ ~--' _- :-_ '' 1]1- _-

~~ I~ I_ -

~· ~ --..----1 ·.... " CONTROL ID: 3187434 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0140 TITLE: Diagnostic performance of corneal microlayer tomography in the diagnosis of Fuchs endothelial dystrophy FIRST AUTHOR: Taher Kamel Eleiwa AUTHORS/INSTITUTIONS: T.K. Eleiwa, A. Elsawy, V. Roongpoovapatr, D. Wen, Z. Syed, G. Gameiro, M. Abou Shousha, Ophthalmology, Bascom Palmer Eye Institute, Coral gables, Florida, UNITED STATES|T.K. Eleiwa, Ophthalmology, Faculty of Medicine, Benha University, EGYPT|A. Elsawy, Electrical and Computer Engineering, University of Miami, Coral gables, Florida, UNITED STATES| Purpose: To evaluate the utility of regional Endothelium/Descemet complex thickness (rDMT) obtained using corneal microlayer tomography technique versus regional total corneal thickness in the diagnosis of Fuchs endothelial dystrophy (FED) Methods: 86 eyes (36 with FED, along with 50 age-matched controls) were imaged using high-definition optical coherence tomography with 6 mm radial cuts centered on the corneal vertex. Custom-made segmentation algorithm was used to generate 6 mm color-coded maps and bulls-eye maps divided into central, paracentral, and peripheral regions [fig.1]. Independent-samples T-test was performed to compare the means of central, paracentral, and peripheral DMTs and total corneal thickness between studied groups. Receiver operating characteristic curves were used to determine the accuracy of the rDMTs and regional total corneal thickness in differentiating between both groups Results: The mean central, paracentral, and peripheral DMT was significantly thicker in FED group compared to controls with a mean difference of (7, 8, and 11µm, p<0.001) in central, paracentral, and peripheral regions, respectively. Similarly, the mean central, paracentral, and peripheral total corneal thickness was significantly thicker in FED group with a mean difference of (40, 37, and 35µm, p<0.001) in central, paracentral, and peripheral regions, respectively. The rDMTs achieved higher accuracy than total corneal thickness in differentiating Fuchs eyes from the normal ones with area under the curve (AUC) = 0.954, 0.977, and 0.990 versus 0.803, 0.807, and 0.792 for central, paracentral, and peripheral regions, respectively [fig.2]. Peripheral DMT with an optimal cut off value (OCV) of 22µm achieved the highest sensitivity 92% and specificity 96% versus 69% sensitivity and 80% specificity for central corneal thickness with an OCV of 540µm Conclusions: Peripheral, central, and paracentral DMTs are novel signs for the diagnosis of Fuchs endothelial dystrophy with excellent accuracy in differentiating it from normal corneas Fig.1: Graph-based segmentation algorithm showing the generated color-coded maps and bulls-eye maps for the total corneal thickness (left side) and endothelium/Descemet complex thickness (right side) Fig.2: Combined ROC curves of the regional corneal thickness, and Endothelium/Descemet thickness showing higher accuracy of the latter in differentiating Fuchs eyes from the normal ones CONTROL ID: 3187554 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0116 TITLE: Alterations of the foveal avascular zone area in glaucoma patients after glaucoma surgery FIRST AUTHOR: Takuhei Shoji AUTHORS/INSTITUTIONS: T. Shoji, Y. Yoshikawa, J. Kanno, H. Ishii, H. Ibuki, K. Shinoda, Ophthalmology, Saitama Medical University, Iruma, Saitama, JAPAN| Purpose: To evaluate quantitative change in the foveal avascular zone (FAZ) area following glaucoma surgery using swept-source optical coherence tomography angiography (SS-OCTA) systems. Methods: We enrolled primary open angle glaucoma patients who underwent surgery to reduce intraocular pressure from April 2018 to September 2018. Two masked examiners reviewed image quality and excluded images that did not fulfill our image criteria. After excluding seven eyes for which images before or after surgery were blurred, 29 eyes in 23 patients (nine men, 14 women) were analyzed. The number of eyes undergoing various surgeries was as follows: two eyes underwent trabeculectomy, 13 eyes underwent treatment using Ex-PRESS, seven eyes underwent microhook trabeculotomy, four eyes were treated using a trabectome, one eye underwent Ahmed valve implantation, and two eyes underwent needling. Results: Patients were aged 65.1 ± 12.0 years (mean ± standard deviation), and the intraocular pressure value significantly decreased from 21.9 ± 11.1 mmHg preoperatively to 10.3 ± 3.2 mmHg postoperatively (p<0.001, paired t- 2 2 test). The FAZ area was significantly reduced to 0.309 ± 0.122 mm (0.396 ± 0.140 mm after ocular axis correction) 2 2 from 0.338 ± 0.143 mm preoperatively (0.434 ± 0.168 mm after ocular axis correction) (p<0.001, paired t-test). A proportion of the retinal blood vessels of the macular region that could not be confirmed preoperatively was drawn postoperatively, and the FAZ area had been reduced. Conclusions: The FAZ area imaged using SS-OCTA was significantly smaller after glaucoma surgery. It was suggested that the macular capillary retinal hemodynamics change with the reduction in intraocular pressure due to glaucoma surgery. (No Image Selected) CONTROL ID: 3188392 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0135 TITLE: Comparison of corneal ablation depth in small incision lenticule extraction (SMILE) and femtosecond laser- assisted LASIK by FD-OCT FIRST AUTHOR: Jing Zhang AUTHORS/INSTITUTIONS: J. Zhang, Eye Center, Beijing Tongren Hospital, Beijing, CHINA| Purpose: To compare the predictability of ablation depth in central cornea between small incision 1enticule extraction(SMILE) and femtosecond laser-assisted LASIK. Methods: This is a retrospective, investigational,comparative case study. One hundred and four patients(201 eyes) underwent SMILE and 108 patients (202 eyes) underwent FS-LASIK were enrolled.Central corneal thickness was measured by RTVue FD-OCT before and 1 month after surgery.The differences between the actual and theoretically expected depth were defined as the central corneal cutting error and were compared between two groups. Results: In SMILE group,The actual ablation depth was significantly less than the theoretically values,(78.34±16.57μm vs 94.25±15.92μm)(t=-25.178,P<0.01), while there are no significant difference in FS- LASIK group (86.92±25.56μm vs 89.71±24.93μm (t=-0.816,P=0.489).In contrast,the central corneal cutting error was statistically higher in SMILE group than in FS-LASIK group(19.49±7.24μm vs 7.35±6.21μm)(t=13.67,P<0.01). Conclusions: The actual corneal ablation depth was significantly less than the theoretical value by SMILE procedure, while the disparities were not significant in FS-LASIK , the predictability of FS-LASIK was better. (No Image Selected) CONTROL ID: 3191451 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0154 TITLE: CONGENITAL LEBER AMAROUSIS ASSOCIATED WITH MUTATION IN CBR1 GENE FIRST AUTHOR: Aristofanes Canamary Jr AUTHORS/INSTITUTIONS: A. Canamary Jr, F. Louise, M. Valim, J. Sallum, visual science, Federal University of São Paulo, Sao Paulo, Sao Paulo, BRAZIL| Purpose: To describe funduscopic changes and imaging findings in patients with Congenital Leber Amaurosis associated with mutation in CRB1 gene. Methods: Male, 16 a, brown, with progressive low visual acuity, since childhood, worse at night, associated with involuntary vertical movements in (AO). He denies consanguinity in the family. The mother did not perform prenatal care and used alcohol and cigarettes during . Sisters without ocular and systemic alterations. At birth, the cephalic perimeter was above the 97% percentile and normal neuropsychomotor development. It presents obesity and brachydactyly. Ophthalmic examination 20/80 OD and 20/50 OE, bilateral pseudofacia. At the fundoscopy AO they present optic disk drusen, areas of yellow /whitish and blackish pigmentation in posterior pole and periphery, increase in vascular tortuosity and in extreme periphery vascular lesion (Coats). Results: FAF shows hyperautofluorescent area AO. FA shows delay of arterial filling and hyperfluorescence by extravasation in the periphery in vascular injury region AO. OCT shows attenuation of the differentiation of the internal layers of the retina, thinning of the outer plexiform, thickening of the external nuclear layer, loss of differentiation of the most hyperreflective outer layers and atrophy of the choriocapillar. ERG with absence of rods and 95% decrease in cones. Complementary audiometry and ultrasound examinations of urinary tract were normal. Genetic test identified mutation in CRB1 gene. Conclusions: There are descriptions of mutations in approximately 11 genes, among them CRB1 in Congenital Leber Amaurosis. This gene is responsible for the phototransduction and structuring of the photoreceptors. The fundoscopy and OCT findings, such as increased thickness and loss of retinal layer differentiation, increase suspicion of mutation in this gene. However, genetic test is required for confirmation.

CONTROL ID: 3192315 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0125 TITLE: Choriocapillaris imaging in patients with Behcet′s disease using optical coherence tomography angiography and image averaging FIRST AUTHOR: Yu Kawashima AUTHORS/INSTITUTIONS: Y. Kawashima, A. Uji, M. Miyata, S. Morooka, Y. Muraoka, T. Akagi, A. Tsujikawa, Kyoto university, Kyoto, JAPAN| Purpose: Imaging of the choriocapillaris (CC) in diseased eyes requires advanced imaging technology such as optical coherence tomography angiography (OCTA). In our recent study, the image averaging technique showed the CC in detail. The averaging technique made possible to compare normal eye and disease eye using on OCTA. This study was undertaken to investigate the CC in patients with Behcet′s disease (BD) using en-face image averaging on OCTA. Methods: This was a retrospective case study. OCTA imaging of the macula, covering a 3×3 mm area centered on the fovea, was performed using the commercially available HS100 HR-SD-OCT system (Canon Inc., Tokyo, Japan). Each eye underwent 10 OCTA scans, which were averaged to obtain high image quality cube data via the built-in software. The CC slab was binarized, and the number and size of flow voids were quantified. Retinal projection artifacts were removed before CC quantification. External limiting membrane (ELM), ellipsoid zone (EZ), and cone outer segment termination (COST) status were separately judged by 2 retinal specialists. Results: Forty-four eyes (26 eyes of patients with BD; 18 normal eyes) were included. ELM, EZ, and COST damage were more frequently observed in patients with BD than in those with normal eyes (27% vs 0%, P = 0.046; 27% vs 0%, P = 0.047; and 77% vs 0%, P <.0001, respectively). Flow void counts were significantly smaller in patients with BD than in those with normal eyes (1275 ± 275 vs 1488 ± 216; P = 0.001). Total area of flow voids were significantly larger in patients with BD than in those with normal eyes (61066 ± 15670 vs 49409 ± 15040; P = 0.040). Average size of flow voids were larger in patients with BD than in those with normal eyes (53 ± 28 vs 35 ± 15; P = 0.010). The percentage area of flow voids were higher in patients with BD than in those with normal eyes (28 ± 7 vs 23 ± 7; P = 0.040). In BD patients with eye symptoms versus those with no eye symptoms, counts, total area, average size, and percentage area of flow void were not significantly different (P = 0.857, 0.562, 0.833, and 0.562, respectively). Conclusions: Averaged images of CC in patients with BD showed significant damage and larger and sparser flow voids than those of normal eyes. Our results indicated that patients with BD had CC damage in the absence of ocular attacks, which could be detected using OCTA. (No Image Selected) CONTROL ID: 3192481 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0146 TITLE: Multimodal imaging of experimental choroidal neovascularization FIRST AUTHOR: xuan liu AUTHORS/INSTITUTIONS: X. liu, Xián Jiaotong University, Xian, Shaanxi, CHINA|M. Zinkernagel, P. Schwarzer, D. Kokona, Inselspital, Unviersity of Bern, SWITZERLAND| Purpose: To compare different imaging modalities for assessing experimental choroidal neovascularization in rodents. Methods: Experimental choroidal neovascularization (eCNV) was induced by rupturing Bruch’s membrane with a laser. Multimodal imaging, including fluorescein angiography (FA), cSLO multicolor imaging (including blue reflectance (BR; 488 nm), green reflectance (GR; 515 nm) and infrared reflectance (IR; 820 nm) ) as well as optical coherence tomography angiography were used to quantify the presumed area of eCNV 7 and 14 days after laser. At day 14, multimodal imaging was compared with histopathologic measurements from isolectin-stained choroidal flatmounts. Results: Choroidal neovascularization area measurements showed a decrease in all imaging modalities between day 7 and 14. Correlation between different imaging modalities at day 7 showed that FA and multicolor imaging correlated best (r=0.4, p<0.05). At day 14, there was a high correlation between area measurements in flatmounts and FA (r=0.6, p<0.001) and multicolor imaging (r=0.4, p<0.05). Area measurements of OCTA with other imaging modalities was poor both at day 7 and 14. Conclusions: FA quantification of CNV area correlated best with CNV area measured in isolectin stained choroidal flatmounts and remains the gold standard to quantify CNV area in vivo. 7Day 14 Day

OCT-A

OTC.A REP·REP FIT 150000

100000 c Flat Mount u~ 0 a c 50000 0 c

c 0 0 50000 100000 150000 200000 Flat Mounl chRPE +'

Fig 2. This figure shows the relationship between OCT-A and Flat mount. With this imagine, we can find the new vessel clearly. On OCT-A imagine day 7 and I 4, we can easily find the laser spot(the dark area). Also on the imagine of Flat mount, the laser spot is clear. The correlation of OCT-A and flat mount is very low. +' Infrared Gr"n Sh... ~ ~eflecta~ce Rcncct.;)ncc Rcilcct.:>ncc M ultI Color MC 1 2&0000

200000

! 150000 g i 100000 50000 ~- •+-~~·~·~~·~~~~~~~ 0 SOOOO 10(1000 15GOOQ :>00000 Fl.a• MoYnl chRPE

Fig 3. This figure shows the Multicolor imagine including Infrared Reflectance, Green Reflectance and Blue Reflectance. The laser spots were clearer on imagine ofInfrared Reflectance than the other model. And there is not much difference between day 7 and day I 4. The correlation between Multicolor and Flat mount was the best than OCT-A . ...., CONTROL ID: 3193472 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0152 TITLE: Spectral Domain-Optical Coherence Tomography in Tracking Progression of Geographic Atrophy in Individuals with Late-Stage Age-Related Macular Degeneration FIRST AUTHOR: Spencer Carl Cleland AUTHORS/INSTITUTIONS: R. Danis, Eyekor, Inc, Madison, Wisconsin, UNITED STATES|S.C. Cleland, S. Konda, D. Myers, R. Trane, B. Blodi, A. Domalpally, Ophthalmology and Visual Sciences, Fundus Photograph Reading Center, Madison, Wisconsin, UNITED STATES| Purpose: Geographic atrophy (GA) is diagnostic of late-stage dry age-related macular degeneration (AMD), which is a blinding disease with no treatment options. GA area and annual enlargement are common metrics to evaluate the progression of GA and the efficacy of interventions. By gold standard, GA area and annual growth rate are measured with fundus autofluorescence (FAF). However, novel software using the OCT Split Tool (Eyekor, Inc.) makes it possible to measure GA area using spectral domain-optical coherence tomography (SD-OCT). The aim of this study is to assess the reproducibility of measuring GA using SD-OCT and compare GA annual growth rate to established methods. Methods: Participants with GA underwent FAF, color fundus photography (CFP), infrared imaging (IR), and SD-OCT imaging at baseline and 12 months in the affected eye(s). Neovascular AMD was excluded using fluorescein 2 angiography. Graders followed established criteria to determine GA area in mm for FAF, CFP, and IR images. Areas were measured using a custom developed OCT split tool that allows for mapping areas of retinal layer loss on co- registered IR images. SD-OCT images were graded for GA area using two distinct criteria: 1) contiguous retinal pigment epithelium (RPE) loss and 2) hypertransmission through Bruch’s membrane (waterfall). Each image was assessed by two graders followed by senior adjudication. Results: 28 eyes were evaluated using all imaging modalities. 3 of these were excluded due to poor image quality. 2 The mean baseline area with SD-OCT using RPE loss criteria was 7.26 (SD 4.87) mm and waterfall criteria was 8.23 2 2 2 (SD 5.1) mm . The mean change in area over 12 months was 1.81 (SD 1.1) mm and 1.83 (SD 1.42) mm , respectively. GA area measurements using SD-OCT grading had no statistically significant difference compared to 2 FAF, IR, or CFP. The mean difference between the two graders was 0.21 mm (95% CI -1.34,0.96) using the loss of 2 RPE criteria and 0.36 mm (95% CI -0.75,2.28) using the waterfall criteria. Conclusions: The OCT split tool provides a novel and reproducible means of measuring GA area and provides area measurements that are comparable to other imaging modalities. (No Image Selected) CONTROL ID: 3193893 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0139 TITLE: Chemical cross-linking: a new choice for the bullous keratopathy treatment FIRST AUTHOR: Mengmeng Wang AUTHORS/INSTITUTIONS: M. Wang, M. Zhang, Hebei Provincial Eye Hospital, Xingtai, Hebei, CHINA| Purpose: To evaluate the effects of corneal CXL using chemical agents (glyceraldehyde and genipin) on the rabbit bullous keratopathy models established by descemetorhexis. Methods: 35 rabbits were randomly divided into five groups. Group A (n = 5) is the control group. The right eyes of animals in Groups B, C, D, E, F, and G (n = 5, resp.) were suffered with descemetorhexis procedures. From the 8th day to the 14th day postoperatively, the right eyes in Groups C, D and F were instilled with hyperosmolar drops, glyceraldehydedrops, and genipin drops respectively; the right eyes in Group E and G were instilled with hyperosmolar-glyceraldehyde drops and hyperosmolar-genipin dropsrespectively. Centralcorneal thickness (CCT), corneal transparency score, and histopathological analysis were applied on the eyes in each group. Results: Compared with Group A, statistically significant increase in CCT and corneal transparency score was found in Groups B, C, D, E, F and G at 7 d postoperatively (P < 0.05) and in Groups C, D, E, F and G at 14d postoperatively (P < 0.05). Conclusions: Chemical CXL improved the CCT and corneal transparency of the rabbit bullous keratopathy models. Topical instillation with hyperosmolar-glyceraldehyde/genipin solution seems to be a good choice for the bullous keratopathy treatment. (No Image Selected) CONTROL ID: 3194090 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0123 TITLE: Drusen Dynamics using Optical Coherence Tomography (OCT) FIRST AUTHOR: Jeong W Pak AUTHORS/INSTITUTIONS: J.W. Pak, A. Domalpally, D. Myers, B. Blodi, Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, Wisconsin, UNITED STATES|Y. Huang, EyeKor Inc, Madison, Wisconsin, UNITED STATES| Purpose: Increase in drusen volume as measured by OCT may be a risk factor for end stage age-related macular degeneration (AMD). Change in overall drusen volume does not adequately capture the dynamic nature of drusen progression in an eye, with some drusen growing and others fading. We assessed these uneven regional changes in drusen volume on OCT as the disease progressed. Methods: OCT was evaluated from 95 eyes with intermediate AMD at baseline and month 12. Segmentation of drusen was performed from top of the RPE layer to top of Bruch’s Membrane using semi-automated custom software to generate drusen volume in each of the 9 subfields of the OCT grid. Reproducibility was assessed by masked regrading of 50 eyes. 3 3 Results: The mean drusen volume at baseline was 1.02 mm (±0.17 mm ) and mean change at 12-month was 0.03 3 3 mm (±0.05 mm ). An increase in the mean drusen volume was seen in 72 eyes (75.8%), 15 (15.8%) showed a decrease and 8 (8.4%) showed no change over one year. In 59% of the AMD eyes, uniform change in drusen volume was observed across all subfields; 47% showed uniform increase and 12% uniform decrease in drusen volume across all subfields. In 40%, there was regional variation for change in subfields within the grid, such that within the same eye some subfields increased while others decreased. Intergrader agreement assessed using Bland Altman plots showed 3 a mean difference of 0.02 mm (confidence limit -0.08, 0.02) between the two graders in drusen volume. Conclusions: Drusen volume on OCT is a useful tool to measure change in drusen burden over time. Total volume is an insufficient metric to capture drusen progression. Mapping drusen volume by OCT subfield may provide a better understanding of the dynamic changes in AMD. Monitoring drusen dynamics may be an important outcome for clinical trials and future prevention therapy. Figure 1 A heat map showing changes in drusen volume in individual subfields of the OCT grid. Warm colors (red) represent increase, cool colors (blue) represent decrease and no color represents no change. There is regional variability from subfield to subfield in drusen change. CONTROL ID: 3194267 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0157 TITLE: Sub-clinical Diabetic Macular Edema in Chinese Diabetes Patients: A Pilot Study. FIRST AUTHOR: Xia Gong AUTHORS/INSTITUTIONS: X. Gong, L. Wang, W. Huang, W. Li, Zhongshan Ophthalmic Center, Guangzhou, CHINA| Purpose: This pilot, observational study aimed to select eyes with subclinical diabetic macular edema (SC-DME) and find out its relevant factors in Chinese diabetes patients. Methods: st 1432 diabetes patients from Yuexiu District, Guangzhou City, China, had been voluntarily recruited by Nov. 21 , 2018. SC-DME is defined as macular center point thickness (CPT) in optical coherent tomography (OCT) ranges from 225 to 299μm with no clinically apparent macular edema in fundus examination according to Diabetic Retinopathy Clinical Research Network. 7-field fundus photography was taken through pharmacologically dilated pupils for grading diabetic retinopathy. Optical biometric and systematic information was also collected. Patients with other macular diseases or unclear OCT images will be excluded. Results: 1031 diabetes patients (1995 eyes) are included. Their average age is 65.6±8.31. 182 patients (17.7%,223 eyes) have SC-DME. The present of SC-DME is significantly related to R1 diabetic retinopathy (OR=0.07,P<0.01) and longer axial length (OR=1.47,P=0.001), while age, gender, blood pressure have no significant correlation with SC- DME. Conclusions: About 1/6 of participants with diabetes have SC-DME. Diabetes patients with R1 retinopathy and longer axial length have a higher risk of developing SC-DME. (No Image Selected) CONTROL ID: 3194276 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0160 TITLE: Deeply pigmented clumps at fovea : A novel clinical find in Familial exudative vitreo-retinopathy (FEVR) FIRST AUTHOR: Chinmayi Himanshuroy Vyas AUTHORS/INSTITUTIONS: C.H. Vyas, D. Kadri, vitreo-retina , Nethradhama Superspeciality eye hospital, Bangalore, Karnataka, INDIA| Purpose: Familial exudative vitreo-retinopathy (FEVR) is a rare hereditary disease affecting retinal vascular development. We report a novel clinical finding of deeply pigmented clumps mimicking spotted cystic dystrophy findings in a case of FEVR using Fundus fluorescein angiography (FFA), Optical coherence tomography (OCT). Methods: A 33-year-old female presented to our clinic with complaints of reduced vision in right eye noticed since 6 months. She was a diagnosed case of familial exudative vitreo-retinopathy (FEVR) with myopia and no past history of prematurity or hospitalization and/or oxygenation after birth and a family history of similar complaints in her mother and maternal grandmother. Her anterior segment findings were unremarkable with best corrected visual acuity (BCVA) of LogMAR 0.5 right and LogMAR 0.3 in the left eye. Dilated fundus examination of both eyes revealed myopic tilted discs with deeply pigment clumps with surrounding hypo-pigmented haloes and yellowish sub retinal deposits at the fovea and peripheral avascular retina with exudation. Wide field fundus fluorescein angiography (FFA) revealed abnormal foveal vasculature near the pigmented spots with late staining of the foveal lesions. Peripheral retina showed classical FEVR findings of vascular non-perfusion areas in the temporal retina with late peripheral leakage suggestive of exudation. Optical coherence tomography (OCT) scan in the right eye showed altered foveal contour with presences of intraretinal schisis and foveal thinning and left eye showed blunting of the foveal dip with IS-OS junction disruption subfoveally. The scan passing through the pigment clumps showed high-reflectance with back shadowing. Results: Both eyes FFA guided peripheral scatter laser was done to the leaking areas. Patient was advised to undergo genetic testing to understand the nature of the inheritance pattern and genetic defect. At the final visits patients BCVA stabilized to LogMAR 0.3 and reduction in retinal schisis was noted on OCT in right eye. The results of genetic testings’ are awaited. Conclusions: Our case reports the presence of deeply pigmented clumps at the fovea in association with familial exudative vitreoretinopathy mimicking the pigmented clumps noted in spotted cystic dystrophy which gives new insight to this condition. (No Image Selected) CONTROL ID: 3194343 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0158 TITLE: The Effect of Digital Media in Age-Related Macular Degeneration: Improving Patient Awareness & Compliance FIRST AUTHOR: Bela Parekh AUTHORS/INSTITUTIONS: B. Parekh, J. Parekh, EyeCare Consultants of NJ, Woodland Park, New Jersey, UNITED STATES| Purpose: To examine the role of digital media in enhancing patients’ knowledge of their age-related macular degeneration condition and compliance of their follow-up and subsequent interventions. Methods: The research was conducted at EyeCare Consultants of New Jersey’s two locations, in Woodland Park (WP) and Edison (E), New Jersey.

A survey was conducted through interviews with two ophthalmologists, one optometrist, and one retinologist who each studied their next 100 patients that presented with any form of macular degeneration (50-WP, 50-E). During their visits, patients were shown videos from CheckedUp (a digital application) pertaining to age-related macular degeneration. CheckedUp is an online health technology that uses videos to create better educated and more confident patients and physicians, empowering them to make informed health decisions together. After patients watched the videos, staff posed the following question: “Did the videos help you to better understand the condition(s) affecting your eyes?” Additionally, doctors surveyed patients across multiple visits to observe whether the videos affected the patients’ understanding of macular degeneration, the need for follow-up/intervention, and the importance of overall prevention including diet, smoking cessation and multi-vitamins. Results: The poll results indicated that 92% of patients at the Woodland Park location found the videos to be helpful in their personal understanding of their macular degeneration, while 66% of patients at the Edison location answered likewise. Conclusions: Considering that 79% of patients surveyed found the CheckedUp videos to be helpful in understanding their macular degeneration, it was concluded that digital media can be an effective tool for improving patient education, especially in long-term conditions like macular degeneration. Moreover, the doctors’ observations indicate that patient education is strongly correlated to compliance, suggesting that digital media may play a role in improving overall patient compliance in macular degeneration. It should be noted that many Edison patients are newly- immigrated Asian Americans with English as a second language, while Woodland Park patients are all fluent. As the CheckedUp videos are predominantly only available in English or Spanish, it is likely that a language barrier caused the 26% disparity between locations. (No Image Selected) CONTROL ID: 3194362 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0119 TITLE: Measurement of the Optic Nerve Head Descending Fibers at Bruch’s Membrane Opening Level with Spectral Domain Optical Coherence Tomography in Normal and Glaucoma Eyes. FIRST AUTHOR: Fernanda Mari Fagundes Fujihara AUTHORS/INSTITUTIONS: F.F. Fujihara, Ophthalmology, Hospital Banco de Olhos de Porto Alegre, Porto Alegre, RS, BRAZIL|P.D. Mello, F. Lavinsky, Department of Ophthalmology, Federal University of São Paulo, Sao Paulo, SP, BRAZIL|C.Z. Benfica, N. Castoldi, F.M. Mendes, A. Finkelstein, R.L. Lindenmeyer, D. Lavinsky, H.M. Pakter, F. Lavinsky, Department of Ophthalmology, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, BRAZIL|F.M. Mendes, A. Finkelstein, R.L. Lindenmeyer, D. Lavinsky, Department of Ophthalmology, Federal University of Rio Grande do Sul, Porto Alegre, RS, BRAZIL|H.M. Pakter, Ophthalmology Department, Hospital Nossa Senhora da Conceição, Porto Alegre, RS, BRAZIL| Purpose: This study aims to evaluate a novel structural parameter: the descending fibers width at the Bruch’s membrane opening level (DF-BMO) and its association with other structural and functional parameters in normal and glaucoma patients. Methods: Subjects with glaucoma presenting typical optic nerve head (ONH) findings, high intraocular pressure with or without visual field damage and normal controls were included. Patients underwent 24-2 perimetry (Humphrey Field Analyzer) and SD-OCT (Spectralis). A vertical B-scan of the ONH with the largest cup extension was obtained for the measurements. The mean width between the BMO and the innermost portion of the descending fibers in the inferior and superior portions of the cup constituted the DF-BMO (Figure 1). Automated individual macular layers were checked for proper segmentation and plotted using the average of sectors from the ETDRS grid circle. Statistical analysis was performed using generalized estimating equations (GEE) to allow for clustered observations. Age, gender, presence of glaucoma, disc area and cup extension were accounted in the model. The area under the receiver operating characteristics curve (AUROC) to discriminate between glaucoma and normal eyes for DF-BMO was also calculated. Results: 109 eyes (67 subjects) qualified for the study. The number of eyes was 95 in the glaucoma and 14 in the control group. The mean DF-BMO was 260.04 ± 106.06 µm and 476.14 ± 124.70 µm, for glaucoma and normal eyes, respectively. Other characteristics are described in Table 1. In the GEE models, DF-BMO was significantly associated with cRNFL (P=0.001) and the inner circle of the GCIPL (P=0.006). The inferior portion of the DF-BMO was associated with the inferior cRNFL (P<0.001), whereas their superior counterparts were not (P=0.674). Age and gender weren’t significant in any of the models. The AUROC for the DF-BMO was 0.896 (P<0.001). At the cutoff value of 365 µm, the sensitivity and specificity were 83.2% and 78.6%, respectively, with a positive predictive value of 96.3%. Conclusions: The DF-BMO was significantly associated with structural glaucoma parameters (cRNFL and inner circle of GCIPL) and differentiated well between normal and glaucoma patients. This parameter may constitute an additional OCT biomarker for the structural diagnosis and monitoring of glaucoma. Figure 1 Table 1 CONTROL ID: 3194514 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0137 TITLE: Corneal Volume of the Normal Human Corneas calculated by Pentacam FIRST AUTHOR: Fernando Abib AUTHORS/INSTITUTIONS: F. Abib, Anatomy, Federal Univiersity of Parana, Curitiba, BRAZIL|F. Abib, R. Martins dos Santos, Corne, Clinica de Olhos Prof. Dr. Fernando Abib, Curitiba, BRAZIL| Purpose: The Corneal Endothelial Dysfunction (CED) has acquired importance in recent years, since it has come to be recognized as an indication of endothelial transplantation. CED causes corneal edema, and depending on its cause it may be reversible, such as in cases of acute glaucoma, panuveitis, endothelitis, among others. It may also be classified as reversible, partially reversible or irreversible, depending on the degree of endothelial mosaic injury. The authors recognize endothelial transplantation as the single form of treatment of irreversible CED. The diagnosis and classification of CED should consider the presented symptoms, visual acuity with better optic correction, pachymetric study - preferably serialized by maps and resources obtained by Pentacam or similar device, and the study of the volumetric cornea, also preferably serial. The world literature lacks in determining reference values of normality for corneal volume. The aim of this study is to present values of normal human corneal volume obtained in normal patients. Methods: Cross-sectional study with database of the 150 right eyes of the patients submitted to cornea study with Pentacam HR (Germany) at the Cornea Service of the Clinic of Eyes Prof. Dr. Fernando Abib, Curitiba, Brazil. The corneal volume 3 was calculated in a 3mm, 5mm, 7mm, and 10mm diameter zone around the corneal apex. corneal volume (mm ) results are presented the minimum, maximum average, standard deviation, and 95% confidence interval (CI). The results are presented by descriptive statistics. The used software was Microsoft Excel. Results: The volume of the all studied corneas by zone diameter (Figure 1). The results of the volume by zone 3 3 3 diameter Figure 2): Corneal volume at the 3mm zone: range 3 - 4.9mm , 3.98 ± 0.25mm and 95%CI 3.98 ± 0.4 mm 3 3 3 (3.94 - 4.02); Corneal volume at the 5mm zone: range 9 - 14.2mm , 11.63 ± 0.69mm and 95%CI 11.63 ± 0.11mm 3 3 (11.52 - 11.74); Corneal volume at the 7mm zone: range 20.8 - 30.0mm , 24.87 ± 1.41mm and 95%CI 24.87 ± 3 3 3 0.22mm (24.65 - 25.09); Corneal volume at the 10mm zone: range 49.8 - 70.1mm , 60.13 ± 3.56mm and 95%CI 3 60.13 ± 0.57 mm (59.56 - 60.70). Conclusions: The normal human corneal volumetric results for the zones of 3mm, 5mm, 7mm, and 10mm can collaborate in the diagnosis of corneal edema, contributing to the classification of Corneal Endothelial Dysfunction. Figure 1 The volume of the Studied Corneas by zone

Corneal Volume (mm3)

0

0 1 5 9 13 17 21 25 29 33 37 41 45 49 53 57 61 65 69 73 77 81 85 89 93 97 101105109113117121125129133137141145]4 Examined corneas

- Zone D 3mm: - Ione D 5mm: - Zone D 7mm: Zone D 1Omm: Figure 2 The Results of the Corneal Volume by Zone Corneal Zone D Zone D Zone 07 Zone, D Volume 3mm 5mm1 mm 1Omm Minimum 3 9.4 20.8 49.8 Maximum 4.9 14.2 30 70.1 Average 3.98 11.63 24.87 60.13 St. Deviation 0.25 0.69 1.41 3.56 CONTROL ID: 3194897 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0148 TITLE: Comparison of clinical outcome of anti- VEGF treatment in Diabetic macular edema combined with and without Epiretinal membranes FIRST AUTHOR: mahmut cankurtaran AUTHORS/INSTITUTIONS: M. cankurtaran, A. altintas, ulucanlar eye training and research hospital, Ankara, TURKEY| Purpose: To evaluate the effect of epiretinal membranes (ERMs) on intra-vitreal (IV) anti-vascular endothelial growth factor (anti-VEGF) injection for Diabetic Macular Edema (DME) treatment. Methods: Sixty eyes of 60 patients with DME were divided into two group either the DME with ERM (DME-ERM) or the DME without ERM (DME) based on optical coherence tomography (OCT) images. Each eyes treated by IV anti- VEGF according to PRN protocol. The central macular thickness (CMT) and best-corrected visual acuity (BCVA) changes before and after the treatment were compared between two groups. The results were analyzed using the Mann-Whitney U test, Wilcoxon test, Chi-square test, and Kolmogorov-Smirnov test. Results: DME-ERM consist of 37 eyes and DME had 23 eyes. The mean age was 60.43 and 64.04 years in DME and DME-ERM respectively.( p=0.231) The average follow-up time was 16.92 and 21.74 month in DME and DME-ERM respectively (p=0.507). The mean age and follow-up period were similar in two groups. The mean CMT was 401.41 and 414.74 μm before treatment and 348.08 and 330.17 μm after treatment in DME and DME-ERM respectively, which were statistically insignificant (p=0.102, p=0.927). Even the CMT decrement with the treatment was significant in each group, amount of the reduction was higher in DME-ERM (84.57 u) than DME (53.32) (p= 0.032). Even the initial BCVAs were not different (0.39 and 0.68 Log MAR in DME and DME-ERM respectively) and last BCVA was significantly lower in DME-ERM than DME (0.76 and 0.37 Log MAR respectively), the mean changes in BCVAs with the treatment (-0.027 in DME and 0.086 DME-ERM) in were not different between groups (p=0.614). Conclusions: The presence of ERM does not decrease the effect of IV anti-VEGFs in terms of reducing CMT in DME and does not significant negative effect on visual improvement. (No Image Selected) CONTROL ID: 3194927 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0115 TITLE: Personalized Pointwise Circumpapillary Retinal Nerve Fiber Layer Thickness (cpRNFLT) Norms FIRST AUTHOR: Tobias Elze AUTHORS/INSTITUTIONS: T. Elze, M. Wang, D. Li, Harvard Medical School, Boston, Massachusetts, UNITED STATES|T. Elze, K. Wirkner, C. Engel, M. Loeffler, F.G. Rauscher, Leipzig Research Centre for Civilization Diseases (LIFE), Leipzig University, GERMANY|T. Peschel, C. Engel, M. Loeffler, F.G. Rauscher, Institute for Medical Informatics, Statistics and Epidemiology, Leipzig University, Leipzig, GERMANY| Purpose: cpRNFLT measured by optical coherence tomography (OCT) aids clinicians to diagnose optic neuropathies. To date, OCT device norms are solely based on age and neglect individual eye anatomy. Here, we derive personalized normative distribution models based on age, sex, and an estimate of the true scanning radius from participants of the population-based LIFE Adult study. Methods: Among the 10,000 age and sex stratified study participants, reliable (≥50 B-scan repetitions, quality ≥20 dB, ≤5% missing A-scans) Spectralis SD-OCT cpRNFLT scans (768 A-scans, diameter: 12°) of one randomly selected eye without clinically significant findings on fundus or OCT images were included. The true scanning radius on the retina, which depends on eye anatomy, was estimated based on scanning focus. At each location, an optimal model of a Gaussian RNFLT distribution was determined by Akaike's information criterion among the 64 possible models composed of all combinations of parameters age, sex, and radius for mean and variance, respectively. Results: 5,646 eyes from 5,646 subjects (ages: 20-79 years, 54.8% females) were selected. The color bars in Fig. 1 illustrate the best models for each of the 768 locations, placed below the cpRNFLT mean curve as a spatial reference. Age, sex and radius have significant impacts on cpRNFLT mean and variance on most retinal locations. Apart from nasal areas, increasing age is associated with thinner RNFL. Larger scanning radius is related to thicker temporal RNFL and thinner RNFL on all other areas. Males have thicker superior RNFL, females thicker RNFL at most other locations. Additionally, we implemented these new cpRNFLT norms as open source software that mimics the Spectralis printout but provides a personalized normative color plot specific to age, sex, and radius. Fig. 2 A and B show cpRNFLT of two healthy eyes. While the conventional norms (top) result in seeming abnormalities (black line crossing yellow/red areas) on numerous locations, our personalized norms (bottom) only indicate few abnormalities. Conclusions: In addition to age, sex and scanning radius have significant impacts on normative cpRNFLT distributions. Based on over 5,600 subjects, we provide new, personalized cpRNFLT norms. To facilitate clinical applications, we implemented these norms in an open source software that superimposes cpRNFLT on a color plot closely resembling the current device printout. Fig.1 Fig.2 CONTROL ID: 3194989 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0153 TITLE: Safety and Efficacy of 1.5% Trypan Blue Assisted Vitreomacular Surgery FIRST AUTHOR: Subhan Tabba AUTHORS/INSTITUTIONS: S. Tabba, C. Hooten, G. Davis, Ophthalmology, University of Texas at Houston McGovern Medical School, Houston, Texas, UNITED STATES|C. Hooten, Wake Forest Baptist Medical Center, Hawaii, UNITED STATES|K. Bourgeois, Columbus Community Hospital, Texas, UNITED STATES|K. Bourgeois, Houston Methodist St. Catherine Hospital, Texas, UNITED STATES| Purpose: Trypan blue (TB) 0.15% was supplied by a compounding pharmacy to surgeons in the area for six years. After six years, the pharmacy informed the surgeons that there had been a systematic error in the production of TB 0.15% resulting in the production of TB 1.5%, a ten-fold increase in the FDA approved concentration. The goal of the study is to evaluate the outcomes, efficacy, and complication rates of the use of Trypan blue 1.5% in vitreomacular surgery. Methods: A consecutive retrospective chart review of subjects 18 years or older who had undergone Trypan blue 1.5% assisted vitreomacular surgery for macular pucker or macular hole between 2009 to 2012 was performed. Patients with a history of intraocular surgery were excluded with the exception of uncomplicated clear cornea cataract extraction. The primary outcomes were retinal anatomy based on OCT images (decreased central thickness, macular hole closure) and Best Corrected Visual Acuity (BCVA). Complication and adverse event rates were analyzed. Results: 351 patients undergoing surgery by two primary surgeons are included. The difference between preoperative and post operative visual acuity 2.39 ± 4.49 lines, p<0.0001. 84.3% of macular holes closed. There was no toxicity or complications reported. Conclusions: Visual outcomes and surgical success rate with TB 1.5% are similar to the reported rates from the use of TB 0.15%. Fig 1. Demographics Fig 2. Pre- and post- vitrectomy best corrected visual acuity. CONTROL ID: 3195097 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0138 TITLE: Corneal nerve structure in patients with primary Sjögren's syndrome in China. FIRST AUTHOR: Fangting Li AUTHORS/INSTITUTIONS: F. Li, Q. Zhang, X. Ying, J. He, Y. Jin, Y. Cheng, M. Zhao, Peking University People's Hospital, Beijing, CHINA| Purpose: To analyze the in vivo confocal microscopic (IVCM) morphology of corneal nerves in primary Sjögren's syndrome to study its relationship with the clinical evaluation in China. Methods: 22 dry eye disease (DED) patients with primary Sjögren's syndrome (pSS) and 20 control subjects with non-Sjögren dry eye disease (NSDE) were included. Each patient underwent an evaluation of ocular surface disease using the tearfilm break-up time (TBUT); non-invasive tear film break-up time (NIKBUT); non-invasive tear meniscus height (NIKTMH); corneal staining (NEI scale); schirmer test; meibography; and corneal subbasal nerve analysis with IVCM. Right eye of each subject was included in the study. Results: SS patients had significantly severer symptoms: lower TBUT (P< 0.001); lower Schirmer I test results (P< 0.001) compared with the NSDE group. But there was no significant difference in NIKBUT between two groups (P = 0.910). SS patients had more meibomian gland dropout compared with the NSDE patients, the difference was not significant (P= 0.109). The nerve density of subbasal nerves and number of nerves were significantly lower in SS group than NSDE group (P= 0.001 and P= 0.003 respectively). Conclusions: Sjögren syndrome dry eye (SSDE) patients have more severe clinical tests results than non-Sjögren dry eye disease (NSDE) patients. Compared with NSDE patients, we found the SSDE patients shows decreased corneal subbasal nerves density and numbers. (No Image Selected) CONTROL ID: 3195106 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0147 TITLE: Characterization of macular edema in the initial stages of diabetic retinopathy FIRST AUTHOR: Torcato Santos AUTHORS/INSTITUTIONS: T. Santos, A.R. Santos, D. Alves, I. Marques, C. Lobo, J. Cunha-Vaz, AIBILI - Association for Innovation and Biomedical Research on Light and Image, Coimbra, Coimbra, PORTUGAL|A.R. Santos, Department of Orthoptics, Superior School of Health, Polytechnic of Porto, Porto, Porto, PORTUGAL|C. Lobo, J. Cunha-Vaz, University of Coimbra, Coimbra, Coimbra, PORTUGAL| Purpose: To characterize intracellular and extracellular retinal edema in minimal to moderate diabetic retinopathy (DR) through OCT-Leakage. Methods: In this retrospective cross-sectional study, 142 eyes from 142 patients (28% women) aged 52-88 years were imaged by spectral domain optical coherence tomography (SD-OCT) and color fundus photography. Using the 7 field ETDRS protocol, the eyes were grouped into three DR severity groups, levels 10 to 20 minimal, 35 mild and 43 to 47 moderate DR. DRCR.net standards for Cirrus SD-OCT were used to identify eyes with subclinical (SCME) and clinical macular edema (central involved macular edema - CIME). Retinal layer thicknesses were calculated using an in-house segmentation algorithm to assess the contributions of each individual layer to the overall thickness value. OCT-Leakage, a new algorithm to detect sites of low optical reflectivity (LOR) was applied to the full retina and to each retinal layer. Extracellular fluid was measured by the LOR area ratio, which stands for the fraction of the number of A- scans with LOR and the total number of A-scans, within the considered area. Results: The 142 eyes were divided into three severity groups, with 54 eyes in group 10-20, 54 eyes in group 35 and 34 eyes in group 43-47. Macular edema, either SCME or CIME, was present in 43% of eyes in group 10-20, 41% of eyes in group 35 and 38% in group 43-47. CIME was found in 7%, 7% and 12%, respectively for each group. In eyes with macular edema, either SCME or CIME, increases in retinal thickness were significantly correlated with increases in the LOR ratios (INL: r=0.45, p<0.001; OPL: r=0.48, p<0.001). The edema was identified as predominantly intracellular (retinal thickness increase without corresponding increase in LOR ratio) in group 10-20 (65%) and extracellular (retinal thickness increase with corresponding increase in LOR ratios) in groups 35 (77%) and 43-47 (69%) (p=0.010). Conclusions: Retinal edema can occur at any time since the initial stages of diabetic retinal disease, independently of its severity, as eyes from diabetic patients in the initial stages of DR with different ETDRS retinopathy grading show similar prevalence of SCME and CIME. The INL is the more susceptible layer to increase in the occurrence of retinal edema. Retinal edema appears to be mostly extracellular except in the initial stages of the diabetic retinal disease where intracellular edema is predominant. (No Image Selected) CONTROL ID: 3195115 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0151 TITLE: Automatic quantification of focal capillary dropout for identification of ischemia in patients with DR FIRST AUTHOR: Luis Mendes AUTHORS/INSTITUTIONS: L. Mendes, C. Schwartz, D. Alves, T. Santos, I. Marques, J. Cunha-Vaz, AIBILI - Association for Innovation and Biomedical Research on Light, Coimbra, PORTUGAL|A. Bhattacharya, M. Durbin, Carl Zeiss Meditec AG, California, UNITED STATES| Purpose: Compare the performance of three novel algorithms that aim to quantify the focal capillary dropout in patients with DR with the available method of vessel density (VD). Methods: Metrics associated with focal capillary dropout (FCD) may provide major clinical value by indicating the eyes at increased risk for progression to more severe stages of retinopathy.Three new methods were developed in the scope of this study that aim to detect regions associated with FCD in the superficial retinal plexus. The first method (perfusion method) aims to detect holes in the OCTA perfusion slab using morphological filters.The second, texture method, aims to detect the changes in the texture of the OCTA slab associated with regions with FCD.The regions detected by the third method, fusion method, correspond to the regions simultaneous detected by the other two FCD methods.The associated FCD metric for each method correspond to the total of pixels of the detected areas.The results obtained with data, acquired using the CIRRUS™ HD-OCT 5000 with AngioPlex® OCT Angiography (ZEISS, Dublin, CA) (angiography 3x3), from 142 eyes from subjects with DR (68±7 years) categorized by ETDRS severity groups were compared with the results obtained with the data from 45 healthy eyes.A ROC curve was used to compare the performance of the three methods with the VD reported by the Density Exerciser software (version 10.0.0.12787). Results: From the analysis of the ROC curve (Figure 1) obtained when the task was to distinguish the DR eyes from the normal eyes the best FCD method (the perfusion method) got an AUC equal to 0.81 and for the VD the AUC was equal to 0.75. The perfusion and the fusion method show similar behavior, especially in the region with high sensitivity. When the task was to distinguish the ETDRS 35-47 group from healthy plus the ETDRS 10-20 group the best FCD method (the fusion method) got an AUC equal to 0.76 and for the VD the AUC was equal to 0.74 (Figure 2). Conclusions: The results obtained using the methods to identify zones of FCD do not appear to significantly improve the discrimination of DR eyes comparing with generalized VD suggesting that in the initial stages of diabetic retinal disease the capillary dropout is generalized. Figure 1 - ROC when the task was to distinguish DR eyes from healthy eyes. Figure 2 - ROC when the task was to distinguish ETDRS 35-47 group from healthy + ETDRS 10-20 group. CONTROL ID: 3195120 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0156 TITLE: In vivo imaging of subretinal grafts of retinal progenitor cells derived from human pluripotent stem cells by SD- OCT FIRST AUTHOR: Xiufeng Zhong AUTHORS/INSTITUTIONS: X. Zhong, G. Gao, L. HE, X. Song, Y. Guan, B. Xie, Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou, CHINA| Purpose: Many types of retinal cells have been successfully generated from pluripotent stem cells (PSCs), providing unlimited source for cell therapy of retinal degenerative diseases. This study aims to evaluate the timecourse morphological changes of both transplants and host retina of rabbit by spectral-domain optical coherence tomography (SD-OCT). Methods: The human induced PSCs were induced into 3D retinal organoids. Retinal progenitor cells (RPCs) were isolated from the organoids and transplanted into the subretinal space of rabbits. The dynamic changes of retina in rabbits with cell treatment were monitored by SD-OCT. SD-OCT were done before and after surgery at multiple intervals for up to 24 weeks. Immunostaining and histological examination were also performed. Rabbits injected with vehicle were served as control. Results: With SD-OCT, hypo-reflective subretinal blebs in rabbits with cell or vehicle delivery were observed within 1hr and disappeared one day after operation (DAO). In control group, vehicle injection caused neural retina thinning over 2-wk period from loss of ONL to whole retinal layers dependent on the volume delivered (20ul-150ul). In rabbits injected with 20ul cell suspension (5.0-10.0x10^6 cells), cell grafts could be detected as early as 1 DAO, showing an oval or band-like hyper-reflective mass in the subretinal space. The size of cell mass slightly increased in 28 DAO, then decreased and became stable afterwards (24-week followup). While the host retina located above the cell mass became thinning and disorganized, with innermost neural retina left more or less. The cells in the margin of the grafts migrated out with scattered high reflection in host retina. No layered structures were found within the grafts. Histological findings were similar to those of SD-OCT. Immunostaining demonstrated that hiPSC-derived RPCs differentiated most into Recoverin+ photoreceptor cells in the subretinal space of rabbits. Rhodopsin+ rods and L/M opsin+ cones were also appeared at 4 months after transplantation.

Conclusions: SD-OCT is a reliable tool for in vivo evaluation of cell transplants and host retina. hiPSC-derived RPCs grafts prefer forming high reflective mass rather than layered structure in subretinal space of rabbits.

(No Image Selected) CONTROL ID: 3195305 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0134 TITLE: Corneal Strain after UV-Riboflavin Cross-Linking Measured by Optical Coherence Elastography FIRST AUTHOR: Sabine Kling AUTHORS/INSTITUTIONS: S. Kling, H. Khodadadi, Information Technology and Electrical Engineeriung, Swiss Federal Institute of Technology Zurich, Zurich, Zurich, SWITZERLAND| Purpose: Optical coherence elastography (OCE) is a promising technique for high-resolution strain imaging in ocular tissues. We have recently developed an approach to capture corneal strain maps when subjecting the eye to intraocular pressure (IOP) changes in a range similar to diurnal physiological changes. The purpose of the current study was to assess local differences in corneal deformation and axial strain in ex vivo porcine corneas that have been treated with corneal cross-linking (CXL) treatment. Methods: 6 freshly-enucleated whole porcine eye globes were obtained and prepared for CXL. First, the epithelium was removed and 0.1%-riboflavin instilled for 30min. Subsequently, half of each cornea was subjected to UV 2 2 irradiation at either 3mW/cm for 30min or 9mW/cm for 10min, the non-irradiated part served as control. For strain imaging, eyes were mounted on a customized silicon mold and initial IOP was adjusted to 15mmHg by inserting a needle connected to a pressure control unit into the anterior chamber. Different levels of corneal strain were induced by first in- and then decreasing IOP by a total of 5mmHg, in steps of 1mmHg. Each IOP step was adjusted by a customized pressure system and controlled by a closed-loop routine written in LabView. 12 cross-sectional B-scans were recorded at each IOP step. The axial displacement maps were computed from magnitude and phase changes in the raw OCT signal. Strain maps were generated by computing the gradients in axial direction. Results: Differences between CXL and control tissue were not directly visible in axial strain maps, but became apparent when performing local comparisons: Mean overall elastic modulus was 1.1x higher (p=0.014) in cross-linked compared to control tissue (16.0±0.4 vs 14.5±0.4 kPa). A hysteresis between IOP increase and decrease was observed, which was lower after CXL than in controls (1.18 vs 1.47 Pa, p<0.022). Axial strain was higher in the posterior half compared to the anterior half cornea, both with CXL (factor 2.48±0.9) and in controls (factor 2.59±0.5). Conclusions: The derived corneal strain maps permit a localized comparison of biomechanical characteristics. A higher elastic modulus along with a smaller hysteresis after CXL indicates increased stiffness and reduced viscosity confirming previous literature. Axial strain map CONTROL ID: 3195328 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0162 TITLE: Test-retest reliability of peripheral refraction measurements using a widefield slit-scanning ophthalmoscope FIRST AUTHOR: Katharina G. Foote AUTHORS/INSTITUTIONS: K.G. Foote, C. Leahy, M. Everett, J. Straub, Carl Zeiss Meditec, Inc., Dublin, California, UNITED STATES|K.G. Foote, School of Optometry and Vision Science Graduate Group, University of California, Berkeley, Berkeley, California, UNITED STATES| Purpose: Reliable and repeatable measurements of peripheral refraction are important in assessing eyes for clinical study of myopia. A slit-scanning ophthalmoscope could be a comparatively inexpensive instrument for reliably measuring peripheral refraction. We performed a prospective, test-retest reliability study in order to validate the repeatability of peripheral refraction measurements on a slit-scanning ophthalmoscope system. Methods: Peripheral refraction measurements over a 90° field-of-view (FOV) were acquired for 16 eyes of 16 subjects with a range of refractive errors (-9.5 to +4.0; average: -2.91D). Each eye was dilated with tropicamide and phenylephrine fifteen minutes prior to imaging with a widefield slit-scanning ophthalmoscope (CLARUS™ 500, ZEISS, Dublin, CA) with prototype software that measures the relative vertical shift on the retina of two beams entering the eye, thus measuring the vertical component of the peripheral refraction. The same operator tested each eye 3 times, with a break between each measurement to realign the eye. Peripheral refraction was computed as relative peripheral refractive error, i.e. as the difference between the refractive error 30° temporally and the central foveal refractive error. Repeatability of the system was assessed with a Bland-Altman analysis using the last 2 measurements, and test- retest standard deviation (TRT-SD) using all 3 measurements. Results: Intra-subject repeatability was observed through a Bland-Altman analysis with 95% limit of agreement (LoA) (r = 0.95, lower LoA: -1.06, upper LoA: 1.88; Figure 1A). The TRT-SD was 0.72D. The established trend that increasing myopia is associated with relative hyperopia in the periphery is revealed for this dataset (Figure 1B). Conclusions: The repeatability of a measure of peripheral refraction, using a widefield slit-scanning ophthalmoscope, was demonstrated. Possible sources of variability could include changes in pupil alignment, or orientation of patient fixation. Figure 1. (A) Bland-Altman analysis of difference in relative peripheral refraction; blue line: upper LoA, orange line: lower LoA, green line: mean of differences; (B) Average relative peripheral refraction versus central refractive error with standard deviation error bars. CONTROL ID: 3195355 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0141 TITLE: Predicting the likelihood of future keratoplasty from imaging corneal parameters using manifold learning FIRST AUTHOR: Siamak Yousefi AUTHORS/INSTITUTIONS: S. Yousefi, P. Asbell, Ophthalmology, University of Tennessee Health Science Center, Memphis, Tennessee, UNITED STATES|H. takahashi, H. Tampo, S. Inoda, Y. Arai, Ophthalmology, Jichi Medical University, JAPAN|T. Hayashi, Ophthalmology, Yokohama Minami Kyosai Hospital, JAPAN| Purpose: To determine if machine learning can screen patients for ectatic disease and to determine if corneal shape, thickness, and elevation parameters can be used to identify patients who may be at higher risk for penetrating keratoplasty (PKP), lamellar keratoplasty (LKP), descemet’s stripping automated endothelial keratoplasty (DSAEK) or descemet’s membrane endothelial keratoplasty (DMEK) intervention Methods: We selected 3,318 corneal optical coherence tomography (OCT) images from the baseline visit of 12,242 eyes (Casia instrument, Tomey, Japan). We applied principal component analysis on 424 corneal shape, thickness, and elevation parameters followed by t-distributed stochastic neighbor embedding (tSNE) manifold learning to reduce the number of dimensions. We employed density-based unsupervised clustering. Our post hoc analysis revealed that clusters were attributed to different keratoconus stages. Assessment of 333 eyes with post-operative keratoplasty showed that the method can also predict the risk for future keratoplasty. Results: The mean age of participants was 69.7 (SD=16.1) and 59% were female. Manifold learning of 18 principal components followed by unsupervised clustering identified five non-overlapping clusters (Fig. 1). Clusters 1 and 2 composed of mainly normal eyes and eyes with small ectasia (assessed by ectasia screening index; ESI of Casia, Fig. 2). Cluster 3 corresponded to early stage anterior and moderate stage posterior ectasia. Cluster 4 attributed to early stage posterior but moderate stage anterior ectasia. Cluster 5 was corresponded to advanced stage anterior and posterior ectasia. The specificity was 96.5% and the sensitivity was 90.4%. The normalized likelihood of future surgery for eyes mapped onto clusters 1 to 5 were 3.1%, 2.9%, 45.3%, 29.5%, and 19.2%, respectively. Conclusions: Manifold learning can identify the ectasia status from corneal parameters and assist the refractive surgeon in identifying those patients who may be at higher risk for future PKP and LKP. Moreover, the method can identify patients who may be at higher risk for future DSAEK and DMEK interventions. Figure 1. Left: Corneal data on tSNE map, middle: clusters, and right: predicting the likelihood of future keratoplasty (eyes with post-operative keratoplasty marked with asterisk). Figure 2. Left: Total ESI index, Middle: Anterior ESI index, Right: posterior ESI index of eyes at each cluster.
CONTROL ID: 3195360 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0128 TITLE: Comparison of pterygium head quantification methodologies based on color photography of primary pterygia FIRST AUTHOR: Olivia L Lee AUTHORS/INSTITUTIONS: O.L. Lee, J. Maram, Doheny Eye Institute, Pasadena, California, UNITED STATES|O.L. Lee, Ophthalmology, UCLA, Los Angeles, California, UNITED STATES| Purpose: To compare and validate two grading methods for quantification of pterygium size and surface area based on color photos of primary pterygia. Methods: Color photographs of 164 eyes with primary pterygia taken in primary gaze with a modified single lens reflex camera system (Canfield Scientific Inc, Fairfield, NJ, USA) were analyzed. Utilizing ImageJ (National Institutes of Health, Bethesda, MD USA) software, three size parameters of the head of the pterygium were quantified included 2 length (mm), width (mm), and surface area (mm ) of the pterygium head. Two grading methodologies were compared using differing strategies to demarcate the head of the ptergygium. The first method defined the head of the pterygium as any fibrovascular tissue crossing the corneal limbus, including any associated corneal opacity. The second grading method defined the pterygium head as any vascularized tissue past the corneal limbus with discernable active blood flow; any avascular fibrous tissue or opacity on the cornea was excluded. Results: Paired t test was used to assess the validation of the two pterygium grading methods. The results from the first pterygium grading method yielded mean measurements for width, length and surface area of pterygia head as 2 2 follows: 3.97 ± 1.02 mm (1.65-6.22 mm), 2.41 ± 1.16 mm (0.65-5.37 mm) and 7.27 ± 4.65 mm (0.89-18.39 mm ) respectively. The results from the second pterygium grading method yielded mean measurements for width, length and surface area of pterygia head as follows: 4.49 ± 1.33 mm (0.72- 7.50 mm), 2.22 ± 1.04 mm (0.57-5.23mm) and 7.39 ± 5.12 2 2 mm (0.37-24.12 mm ) respectively. Results showed a significant difference in width of the pterygium (p=0.0001) when comparing the two methods of grading, however there was no significant difference in the length (p=0.08) and area (p=0.41) of the pterygium. Conclusions: Two grading methodologies are presented, demonstrating strategies for reproducibly quantifying the size of pterygium head based on two dimensional color photographs of the eye taken in primary gaze with a simple set focus camera. With both methods, measurements of the width, length and surface area of the corneal portion of the pterygium can be quantified using objective grading criteria. Such methodologies can be used to standardize our evaluation of pterygia in clinical research and clinical trials. (No Image Selected) CONTROL ID: 3195487 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0118 TITLE: Temporal Retinal Nerve Fiber Bundle Changes in Glaucoma Imaged with OCT-Reflectance - Correlation with Circumpapillary OCT Retinal Nerve Fiber Layer Thickness. FIRST AUTHOR: Maria V. Castanos Toral AUTHORS/INSTITUTIONS: M.V. Castanos Toral, D.B. Zhou, E.B. Jacobs, J.S. Andrade Romo, R. Ritch, R.B. Rosen, T.Y. Chui, Research, New York Eye and Ear Infirmmary, Mount Sinai, New York City, New York, UNITED STATES|D.B. Zhou, R.B. Rosen, T.Y. Chui, Icahn School of Medicine, New York City, New York, UNITED STATES|D.C. Hood, Department of Psychology, Columbia University, New York City, New York, UNITED STATES|D.C. Hood, Department of Ophthalmology, Columbia University, New York City, New York, UNITED STATES| Purpose: To evaluate the correlation between retinal nerve fiber bundle (RNFB) width along the temporal raphe and circumpapillary retinal nerve fiber layer (cpRNFL) thickness in glaucomatous eyes using OCT-reflectance (OCT-R) and OCT, respectively. Methods: 7 controls and 11 primary open angle glaucoma (POAG) patients with either superior or inferior arcuate visual field defects were imaged using a SDOCT system (Avanti RTVue-XR; Optovue). Ten 3x3mm scans centered at 9° temporal to the fovea were obtained and averaged (Mo PMID:28068370). A OCT-R slab located between the ILM and 15µm below was used for RNFB width measurement (Fig A1 & B1). On the OCT-R, six 80-100 µm RNFB segments located above and below the temporal raphe were identified. RNFB width was then measured based on the first derivative of the straightened RNFB intensity profiles. Corresponding cpRNFL thickness in the superior and inferior quadrants was measured in each participant (Heidleberg Spectralis HRA + OCT) (Fig A2 & B2). Relationship between RNFB width and cpRNFL thickness were evaluated using linear regression. Results: RNFBs along the temporal raphe were clearly visualized in all controls and POAG patients using OCT- reflectance (Fig A1 & B1). RNFB width along the temporal raphe was decreased in all POAG patients (control: 34.07±7.96µm vs POAG: 23.77±4.32µm). cpRNFL thickness thinning was found in the POAG patients (control: 125.8±15.85µm vs POAG: 69.61±19.98µm). Statistical significant correlation was found between the RNFB width and cpRNFL thickness (linear regression; r=0.56, P=0.0008). Conclusions: Glaucomatous damage on temporal RNFB width measured using OCT-reflectance is associated with cpRNFL thickness thinning measured using the OCT. For glaucomatous RNFB damage detection, RNFB imaging along the temporal raphe using OCT-reflectance may provide additional information to the cpRNFL thickness. (No Image Selected) CONTROL ID: 3195488 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0121 TITLE: Automated Quantification of Glaucomatous RNFL Reflectance Change using en-face OCT-Reflectance FIRST AUTHOR: Davis B Zhou AUTHORS/INSTITUTIONS: D.B. Zhou, M.V. Castanos Toral, J.S. Andrade Romo, R.B. Rosen, T.Y. Chui, Ophthalmology, New York Eye and Ear, New York, New York, UNITED STATES|D.B. Zhou, R.B. Rosen, T.Y. Chui, Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, New York, UNITED STATES|M. Eguia, D.C. Hood, Psychology, Columbia University, New York, New York, UNITED STATES|E.B. Jacobs, R. Ritch, Einhorn Clinical Research Center, New York Eye and Ear Infirmary, New York, New York, UNITED STATES|D.C. Hood, Ophthalmology, Columbia University, New York, New York, UNITED STATES| Purpose: To develop an automated method to quantify retinal nerve fiber bundle (RNFB) defect progression, in patients with glaucoma, captured by en-face OCT-Reflectance (OCT-R) scans. Methods: Six eyes of 5 POAG patients with arcuate visual field defects were imaged at two visits 11-29 months apart using a SD-OCT system (Avanti RTVue-XR; Optovue). Ten 4.5x4.5mm circumperipapillary OCT-R scans were taken at each imaging session. The scans were then co-registered and averaged using ImageJ (PMID:28068370). A layer between the ILM and RNFL was extracted for analysis. The difference in illumination between scans was adjusted using background image correction based on the OCT-R choroid layer (76μm anterior to the RPE and the posterior limit of the OCT-R scan). Images with motion and specular reflectance artifacts were excluded. Remaining images were polar transformed for vertical representation of arcuate RNFB defects.

For each patient, within-visit variation of en-face OCT-R at 3.4mm from the optic disk was computed using the two baseline scans with the greatest difference in pixel-intensity variance. Difference in normalized pixel intensity was calculated between scans after excluding major blood vessels, and mean±SD of this pixel-intensity difference was computed.

RNFL defect progression was determined by similarly calculating pixel-intensity difference between the first baseline and follow-up scan and identifying regions 3.5 standard deviations or more below the mean baseline pixel-intensity. Results: Thirteen regions of RNFL-defect progression were identified in five out of six eyes. Five inferior temporal progressions were revealed between 141.1° and 160.2° in three eyes, with a yearly change of 0.6°±0.8° (mean+SD) per eye in this region. Six superior temporal areas of progression were found between 196.2° and 224.3° in two eyes, with 3.7°±0.6° annual superior temporal progression per eye. Global yearly progression was 1.7°±2.2° per eye. Conclusions: Comparison of RNFL reflectance change via en-face OCT-R provides an additional automated quantitative metric for identifying glaucoma progression. The regions of RNFB alteration can be delineated and compared to local changes in visual field defects, RNFL thickness, and OCT-Angiography perfusion. (No Image Selected) CONTROL ID: 3195523 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0155 TITLE: Quantification of diabetic retinopathy-associated non-perfusion on ultra-wide-field fluorescein angiography and assessment of the impact of age, sex, race, and type of diabetes mellitus FIRST AUTHOR: Michael Aaberg AUTHORS/INSTITUTIONS: M. Aaberg, T.P. Patel, R.S. Iyengar, M.M. Gilson, A. Tran, C. Miranda, E. Young, K. Demetriou, L. Devisetty, D.C. Musch, Y.M. Paulus, Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan, UNITED STATES|D.C. Musch, Epidemiology, University of Michigan, Ann Arbor, Michigan, UNITED STATES|Y.M. Paulus, Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, UNITED STATES| Purpose: This study evaluates the association between diabetic retinopathy (DR) related non-perfusion (NP) observed using ultra-wide-field (UWF) fluorescein angiography (FA) and variables including age, sex, race, and type of diabetes mellitus (DM). Methods: A retrospective chart review of patients who had previously been diagnosed with DM, evaluated at the University of Michigan Kellogg Eye Center, Ann Arbor, and undergone Optos UWF FA imaging between January 2009 and May 2018 was conducted after approval from the IRB. Demographic and clinical data were acquired for each patient. Trained, masked graders segmented regions of NP within each presenting image. Eyes with severe media opacities or panretinal laser photocoagulation were excluded. Optos research software was used to determine the surface area (SA) of the segmented regions for a nominal eye diameter of 24mm. A linear mixed model was used to evaluate univariate associations and to account for inter-eye correlation. Results: 573 eyes from 349 patients were included. The mean age of the cohort was 57.7 years, and 42.4% of patients were female. 65.0% of patients were white, 24.4% were African American, 4.6% were Asian, and 6.0% were unknown/other. 81.7% of patients were diagnosed with type II DM, 17.8% had type I DM, and 0.6% were unknown. 2 2 Men had significantly greater NP SA than women, 74.57 mm vs. 60.02 mm respectively (p=0.01). The NP SA of 2 2 patients over age 65 (56.41 mm ) was significantly less than that of younger age groups: 18-44 years (75.92 mm ; 2 2 p=0.02), 45-54 years (71.44 mm ; p=0.04), and 55-64 years (72.57 mm ; p=0.02). African American patients had a 2 2 significantly greater NP SA (77.59 mm ) than white patients (64.38 mm ) (p=0.03). Type I and II DM did not significantly differ in NP SA. Conclusions: Age, race, and sex had significant impacts on the quantified extent of NP on univariate analysis. Multivariable linear mixed model results will be presented to better understand how these variables collectively associate with the clinical status of DR in these patients. Example segmentation of an Optos UWF FA image. Red, green, and blue indicate NP, foveal avascular zone, and neovascularization, respectively. Average NP observed within distinct population subsets. Yellow indicates the reference group and asterisks denote statistical significance (p<0.05). CONTROL ID: 3195526 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0120 TITLE: Morphology of trabeculectomy filtering blebs using anterior segment optical coherence tomography: a comparison of two methods FIRST AUTHOR: Rita Proença AUTHORS/INSTITUTIONS: R. Proença, J. Cunha, J. Ferreira, A. santos, Ophthalmology, Centro Hospitalar de Lisboa Central, Lisboa, PORTUGAL| Purpose: Anterior segment imaging optical coherence tomography (AS-OCT) can be a useful aid in glaucoma surgery. Recent studies have shown its importance in both the preoperative morphologic evaluation of glaucoma patients as well as postoperative evaluation of filtering bleb functionality. Our purpose is to evaluate post-trabeculectomy filtering and non-filtering bleb characteristics in both time-domain OCT (TD-OCT, Visante™, Carl Zeiss) and spectral-domain OCT (SD-OCT, Heidelberg Spectralis ® anterior segment module), assess the usefulness of AS-OCT in evaluating postoperative filtering bleb function and compare both methods results. Methods: Observational case series of 20 eyes of 20 patients who had undergone trabeculectomy in the last 4 years. Eyes were classified into 2 groups: failed blebs (FBs) and non-failed blebs (NFBs). Bleb structures were assessed with both TD-OCT and SD-OCT. Blebs were analyzed in terms of total height, wall thickness and reflectivity, microcysts, pattern, fluid filled cavity height, position and width of the filtration opening and correlation with IOP and slit lamp morphology. Results: Of the 20 patients, 12 were male and 8 were female, with a mean age of 74.6 years (±7.99). Average preoperative intraocular pressure was 25.6mmHg and postoperative 13.58mmHg. Functioning blebs had significantly more intraepithelial microcyst density than failed blebs. Thicker bleb walls with lower wall reflectivity correlated with lower IOP. Larger fluid filled cavity height and hyporeflective area volume also correlated with successful blebs. Failed blebs revealed more scarring processes. A good degree of concordance was observed between slit lamp morphology and AS-OCT evaluation. A trend was observed towards a more defined characterization of bleb wall morphology in SD-OCT and a better visualization of deep scleral structures in TD-OCT. Conclusions: AS-OCT is a non-contact, simple and reproducible method to analyze the morphology of trabeculectomy blebs. It may be useful in evaluating bleb functionality, aiding clinical evaluation and allowing an early identification of filtration failure. (No Image Selected) CONTROL ID: 3195537 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0145 TITLE: FINDINGS FROM THE FELLOW EYES OF LAMELLAR MACULAR HOLE: A BILATERAL DEGENERATIVE CONDITION? FIRST AUTHOR: Ismael Chehaibou AUTHORS/INSTITUTIONS: I. Chehaibou, N. Manoharan, D. Sarraf, J. Hubschman, Retina, Stein Eye Institute, UCLA, Los Angeles, California, UNITED STATES|I. Chehaibou, Retina, Hopital Lariboisiere, Paris, FRANCE|A. Govetto, Retina, Oftalmico Hospital, Milan, ITALY| Purpose: To evaluate vitreoretinal interface disorders and macular changes in the fellow eyes of patients with lamellar macular hole (LMH). Methods: Patients were divided in three groups based on their optical coherence tomography (OCT) features of the main eye at baseline: group A (LMH), group B (mixed lesions) and group C (macular pseudohole (MPH) or epiretinal membrane foveoschisis (EMF)). History of retinal conditions including rhegmatogenous retinal detachment (RRD), retinal tear (RT), vitreo-macular surgery, and OCT features at baseline and last follow-up were recorded and compared between the three groups. Results: Ninety eights patients were enrolled in this study: 58 in the group A, 20 in the group B and 20 in the group C. At baseline, a significantly higher rate of RRD in fellow eyes was noted in group A (8/58, 14%) compared to the other groups (0/40, 0%), P = 0.020. Five out of 58 other eyes in group A (9%), 4/20 (20%) in group B and 0/20 (0%) in group C had a history of laser treated retinal tear (P = 0.080). A significant higher rate of FTMH was found in fellow eyes of group A (11/58, 19%), compared to groups B (1/20, 5%) and C (1/20, 5%), P = 0.045. In the group A, three of 58 others patients (5%) showed a bilateral LMH, and one of 58 (2%) had a vitreomacular traction. A epiretinal material, which had common OCT features than the proliferation described in LMH, was present in 28/58 fellow eyes in group A (48%), 5/20 fellow eyes in group B (20%) and 1/20 in group C (5%), P = 0.001. Over the follow-up period, two fellow eyes in the group A developed a LMH and another one developed a FTMH. Conclusions: A high rate of macular and peripheral vitreoretinal diseases was noted in LMH’s fellow eyes. In addition, isolated epiretinal proliferation was detected in the macular area of a much higher number of fellow eyes in Group A versus groups B or C. This proliferation and the high rate of retinal pathologies seen on LMH’s fellow eyes suggest a potential bilateral intraretinal degenerative process in LMH.

CONTROL ID: 3195545 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0132 TITLE: Optimising the three dimensional assessment of corneal lesions using anterior segment optical coherence tomography FIRST AUTHOR: Gibran Butt AUTHORS/INSTITUTIONS: G. Butt, S. Rauz, G. Wallace, Academic Unit of Ophthalmology, Institute of Inflammatiion and Ageing, University of Birmingham, Birmingham, UNITED KINGDOM|F. Menduni, School of Life and Health Sciences, Aston University, UNITED KINGDOM| Purpose: Anterior segment optical coherence tomography (AS-OCT) is a developing technology finding increasing uses in clinical practice. Swept-source lasers machines have reduced the scanning times, making it possible to obtain high quality large volume scans of the anterior segment. The assessment and documentation of cornea-opacifying lesions such as keratitis, dystrophies and corneal scarring has typically been subjective using slit-lamp biomicroscopy or by photography. Human and technical factors can make obtaining serial photographs under standardized conditions difficult thereby making comparative analyses unreliable. Additionally, this only yields 2-dimensional (2D) information. We present the development of an AS-OCT technique to quantify corneal lesions in 3D to address this unmet need. Methods: Five female patients aged between 16 and 37 with corneal lesions received AS-OCTs using the the Casia II whilst attending the Birmingham & Midland Eye Centre. A semi-automated Matlab algorithm was developed to compute the lesion volume. Patient OCT images were deconvolved and high-pass filtered to optimise contrast and enhance edges. The region of interest was selected and lesion segmented using Matlab’s connectivity algorithm to extract the lesion. The final lesion volume in three dimensions was reconstructed in relation to the scanning resolution of the instrument. The algorithm was tested against a manual scar assessment performed by an ophthalmologist. Results: Four patients were diagnosed with acute keratitis, of which 1 was herpetic, the other 3 were presumed bacterial, and one chronic post-traumatic corneal scar. Of the 2 patients who had microbiological samples taken, no growth was detected. The lesions demonstrated varying location, morphology and intensity. The Matlab algorithm 3 successfully segmented the corneal lesion in every patient scan. An average lesion volume of 0.052 mm was found 3 3 within the 5 cases, ranging from 0.014 mm to 0.088 mm . These were compared to manually segmented reconstructions. No significant difference (p>0.05) was found between the scar volume computed by the algorithm and the volume manually reconstructed by the experienced ophthalmologist. Conclusions: Our analysis protocol enabled 3D quantitative assessment of these lesions with this pilot data set. This tool has the potential to be utilized in a variety of contexts and for corneal pathology. (No Image Selected) CONTROL ID: 3195600 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0126 TITLE: Schiempflug imaging of posterior polar cataracts FIRST AUTHOR: Ravi Patel AUTHORS/INSTITUTIONS: R. Patel, D. Beddall, J. Devabattula, S. Virdee, R. Nitiahpapand, S. Balal, A. Sharma, Moorfields Eye Centre at Bedford Hospital, Moorfields Eye Hospital NHS Foundation Trust, Bedford, UNITED KINGDOM| Purpose: Cataract surgery for posterior polar cataracts (PPCs) is associated with significant risk of posterior capsular rupture, vitreous loss, and nucleus drop. The surgical challenge is compounded pre-operatively by difficulty in diagnosis, progression monitoring, and risk stratification of PPCs. We report a case series of Scheimpflug imaging of PPCs, to evaluate the potential role of imaging in the mitigation of these pre-operative difficulties. Methods: Two male and four female patients (ten eyes), median age 53 years old (range 46-58) with suspected PPCs identified on slit lamp examination, underwent Pentacam Scheimpflug imaging (Oculus, Wetzlar, Germany). All information was collected and analysed retrospectively from electronic health records. Results: Scheimpflug imaging provided detailed anatomical characterisation in all ten PPCs. In five eyes (three patients), imaging identified significant posterior protrusion, with a simulated posterior lenticonus tear drop sign suggestive of pre-existent posterior capsule deficiency (Image 1). Some degree of posterior protrusion existed in four eyes, deemed to be surgically insignificant in extent, with nil posterior protrusion in the remainder (Image 2). Two of the six patients who had Scheimpflug imaging (three eyes) underwent cataract surgery. Hydrodilineation was used in place of hydrodissection in all procedures, with no intraoperative or postoperative complications. Four of six patients opted against surgery following risk/benefit counselling, including all with tear drop sign. Conclusions: Scheimpflug imaging facilitates diagnosis and characterises anatomy of PPCs. It can identify pre- existing posterior capsule deficiency. Serial scheimpflug imaging could provide objective monitoring of progression towards higher risk anatomy, to help optimise timing of surgery, and provide continuity between changes of clinician. By facilitating risk stratification, imaging can help personalise pre-operative counselling. Further work is needed to characterise larger cohorts and to identify and correlate new frameworks for risk stratification with clinical outcomes. To date, this represents the largest such case series we could identify. Scheimpflug imaging of a posterior polar cataract in a left eye with tear drop sign present, nil surgery performed Scheimpflug imaging of a posterior polar cataract in a right eye with tear drop sign absent, underwent surgery CONTROL ID: 3195608 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0142 TITLE: High resolution adaptive optics imaging analysis of Enhanced S-Cone syndrome with NR2E3 mutation FIRST AUTHOR: Kiyoko Gocho AUTHORS/INSTITUTIONS: K. Gocho, D. Kubota, S. Kameya, Ophthalmology, Nippon Medical School, Chiba Hokusoh, Inzai, Chiba, JAPAN|K. Kuniyoshi, Ophthalmology, Kinki University, Osaka, JAPAN|T. Hayashi, S. Katagiri, Ophthalmology, The Jikei University School of Medicine, Tokyo, JAPAN|H. Takahashi, Ophthalmology, Nippon Medical School, Tokyo, JAPAN| Purpose: Enhanced S-cone syndrome (ESCS) is a rare and unique, slowly progressive autosomal recessive retinal degeneration related to mutation in NR2E3. The histopathological report showed two-fold increased number of Short- wavelength-sensitive cones (S cones) with absence of rods. Also in vivo high-resolution retinal image of ESCS was reported using adaptive optics (AO) scanning laser ophthalmoscopy (Park et al, 2015). However, structural feature in vivo still remains unclear. This study was to characterize the in vivo high-resolution retinal images using spectacle domain optical coherence tomography (SD-OCT) and AO fundus camera of four patients with enhanced S-cone syndrome (ESCS). Methods: Eight eyes from four cases with ESCS with mutation in NR2E3 (2 males and 2 females, mean age was 52.3 y.o., ranged from 40 to 60 y.o.) underwent ophthalmological examinations including fundus autofluorescence (FAF), TM SD-OCT (Cirrus 5000TM, Zeiss)and AO fundus camera (rtx1 , Imagine eyes, France). Cone density and spacing were measured using 80x80 pixels square images obtained by AO from 1 to 6 degrees temporal and nasal. The results were compared with previously published histological normative data (Curcio et al, 1990) and normal control AO data. Results: Decimal BCVA of four cases were 20/63 to 20/20. Three cases showed mid-nummular peripheral pigmentary deposition and atrophy in RPE and one case showed subtle pigmentary changes with small hyperpigmented areas within the vascular arcades in FAF. In OCT, multiple disruptions of the interdigitation line and ellipsoid zone was observed in all cases. One eye showed the cysts macular edema. AO images showed some visible photoreceptors at the center of fovea in 7 eyes. At 1 degree from the center of fovea, the cone spacing was 8.77±0.85(μm) and the cone 4 2 density was (1.59±0.32)×10 (cone/mm ) on average in ESCS. At all eccentricity in the patients with ESCS, the cone spacing were significantly larger and the cone density were significantly lower than that of normative data. Conclusions: We observed the retinal microstructure on 4 cases with ESCS using AO camera. As all cones were only S-cone in ESCS patients, the cone packing was significantly lower than the normal retina in AO imaging analysis, which related to the OCT findings. (No Image Selected) CONTROL ID: 3195625 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0136 TITLE: Normative Corneal Optical Density Data Set: Creation and Analysis. FIRST AUTHOR: Andrew Rollin Davis AUTHORS/INSTITUTIONS: A. Cheung, Cornea, Virginia Eye Consultants, Norfolk, Virginia, UNITED STATES|A.R. Davis, A. Cheung, Ophthalmology, Eastern Virginia Medical School, Norfolk, Virginia, UNITED STATES| Purpose: To assess the normative corneal optical density values of normal eyes for Anterior Segment Optical Coherence Tomography (AS-OCT) images to serve as a control group for future study. Methods: We analyzed 52 corneal AS-OCTs (Spectralis, Heidelberg) from 16 eyes of 8 patients with ImageJ (NIH). Inclusion criteria comprised (1) the central 3 images above or below the central corneal vertex image (2) quality index >=25 and <= 34. Exclusion criteria comprised (1) presence of significant image artifact or (2) large peak air density. The central 5.5 mm were analyzed. The maximum epithelial reflectance (MER) and average stromal density (ASD) were recorded for 11 sectors, each sector was 500 um in width.

In each scan, the maximum epithelial reflectance (MER) and average stromal density (ASD) were calculated in each sector. In the aggregate data set, the average maximum epithelial reflectance (AMER) and the overall average stromal density (OASD) were calculated irrespective of horizontal position and with respect to horizontal position (AMER(x), OASD(x)). Results: Qualitatively, the MER and ASD for individual scans decreased with respect to lateral position in a linear or quadratic manner.

Quantitative results of the aggregate data set irrespective of position showed there was a high linear correlation coefficient (LCC) between AMER and OASD (r =0.86 ). The relative standard deviation of AMER (RSDAMER) and the relative standard deviation of OASD (RSDOASD) also showed a high LCC ( r = 0.82 ).

Quantitative results of the aggregate data respective of position showed a parabolic relationship between AMER(x) versus position and OASD(x) versus position. A quadratic function was fit to the data set using the standard error of the mean (SEM). For the AMER(x) fit, the reduced chi-squared (RCS) was 8, for the OASD(x), it was 6.42.

Quality versus AMER and OASD irrespective of position were analyzed and a linear relationship between AMER and OASD versus quality was found (RCS of 0.54 and 0.40 respectively). An attempt to analyze the relative SDOASD and relative SDAMER showed a complex relationship with a quality index of 29 being the nadir. Conclusions: A relationship between MER and ASD was demonstrated both respective and irrespective to position. The overall backscatter of the stroma is related to the reflectance of the epithelium. In addition, the MER and ASD may vary in a parabolic fashion with respect to position from the center of the cornea. (No Image Selected) CONTROL ID: 3195630 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0117 TITLE: Adhered Posterior Hyaloid Influence on Glaucoma Structural Parameters Evaluated with Spectral Domain Optical Coherence Tomography FIRST AUTHOR: Fabio Lavinsky AUTHORS/INSTITUTIONS: F. Lavinsky, J. Lavinsky, N. Castoldi, R.L. Lindenmeyer, C.Z. Benfica, D. Lavinsky, Ophthalmology, Hospital de Clinicas de Porto Alegre, Porto Alegre, BRAZIL|F. Lavinsky, P.D. Mello, Department of Ophthalmology, Paulista School of Medicine, São Paulo Hospital, Federal University of São Paulo., São Paulo, São Paulo, BRAZIL|J. Lavinsky, R.L. Lindenmeyer, C.Z. Benfica, D. Lavinsky, H.M. Pakter, Department of Ophthalmology, Federal University of Rio Grande do Sul, BRAZIL|F.F. Fujihara, Ophthalmology, Hospital Banco de Olhos, BRAZIL|H.M. Pakter, Ophthalmology, Hospital Nossa Senhora da Conceição, BRAZIL| Purpose: The aim of this study is to evaluate if an adhered posterior hyaloid in glaucomatous eyes appreciated with Spectral Domain Optical Coherence Tomography (SD-OCT) influences the measurements of individual inner macular layers and circumpapillary retinal nerve fiber layer (cRNFL). Methods: Subjects with glaucoma presenting typical optic nerve head (ONH) findings, high intraocular pressure with or without visual field (VF) damage were included. Patients underwent 24-2 perimetry (SITA standard; Humphrey Field Analyzer; Zeiss) and SD-OCT (Spectralis; Heidelberg Engineering). Subjects were divided into 2 groups: with and without adhered posterior hyaloid. Subjects were also divided into three stages based on the mean deviation (MD) of the VF: ≥-6 dB; between < -6 dB and ≥-12 dB; and < -12 dB (early, moderate and severe), respectively. Automated individual inner macular layers and the summation of layers were checked for proper segmentation and plotted using the average of the sectors from the ETDRS Grid circles (diameters: center 1mm, inner circle 3mm, outer circle 6mm). Statistical analysis was performed using generalized estimating equations to allow for clustered observations. Age and visual field aforementioned severity category were accounted in the model. Results: 95 eyes (60 subjects) qualified for the study. The mean age was 68.62 ± 9.64. 39 eyes were in the non-adhered posterior hyaloid group and 56 in the adhered posterior hyaloid group (Table 1). The VF severity distribution between non-adhered and adhered posterior hyaloid was: 16 and 36 for early; 9 and 9 for moderate; and 14 and 11 for severe glaucoma, respectively. The presence of adhered posterior hyaloid was not significantly associated with the cRNFL thickness (p= 0.128) and with the thickness of the following macular layers (inner and outer circles of the ETDRS grid respectively): ganglion cell layer (GCL) (p=0.082 and p=0.306); inner plexiform layer (IPL) (p=0.069 and p=0.143), ganglion cell layer/inner plexiform layer (GCIPL) (p=0.072 and p=0.223). Conclusions: The presence of an adhered posterior hyaloid wasn't significantly associated with the macular inner layers and cRNFL when accounting for age and visual field severity stage. Longitudinal studies are warranted to determine if changes in the vitreoretinal interface impact the parameters evaluated in glaucoma monitoring. Table 1- Functional and Structural Parameters in No Posterior Hyaloid Adhered and Posterior Hyaloid Adhered Groups.

Parameter No posterior hyaloid Posterior hyaloid adhered adhered (N=39) (N=56} Mean SD Mean SD MD -10.11 8.47 -6.47 7.63 VFI 71.90 27.92 83.48 22.75 GCLinner 37.22 9.99 42.31 9.28 GCLouter 27.29 5.01 29.79 4.87 IPL inner 33.14 6.22 36.55 5.30 IPL outer 24.31 3.28 26.00 2.92 GCIPL inner 70.37 16.09 78.86 14.47 GCIPL outer 51.60 8.08 55.78 7.64 cRNFL 70.49 18.81 78.64 17.52 MO=mean deviation; VFl=visual field index, GCL = ganglion eell layer; IPL = inner plexifonn layer; GCIPL = ganglion cell layer/inner plexifonn layer. Inner and ouler refer to the inner and outer circle of ETD RS grid (3mm and 6mm, respectively). cRNFL= circumpapillary retinal nerve fiber layer. CONTROL ID: 3195651 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0122 TITLE: High definition optical coherence tomography of the aqueous outflow system in glaucomatous and normal subjects FIRST AUTHOR: Simon Antonio Bello AUTHORS/INSTITUTIONS: S.A. Bello, J. Straub, M. Chen, Carl Zeiss Meditec, Inc., Dublin, California, UNITED STATES| Purpose: The aqueous inflow and outflow systems are crucial in maintaining intraocular pressure (IOP) balance, a key risk factor in glaucoma development and progression. Swept-Source optical coherence tomography (SS-OCT) can be used to image the individual anatomical components of the outflow system, which may be useful in glaucoma research as well as pre and post-surgical assessment of minimally invasive glaucoma surgeries (MIGS). We conducted a clinical study to evaluate the ability of a SS-OCT device to acquire High Definition images of these structures ® Methods: A modified PLEX Elite 9000 (ZEISS, Dublin, CA) SS-OCT system operating at 1060 nm was used to image the anterior segment of 5 healthy volunteers and one glaucoma subject. The limbal area of every subject was imaged using a 3x3mm scan pattern which captures 300 A-scans/B-scan, repeated, registered and averaged 20 times, and a total of 51 B-scans. A single-line scan containing 1024 A-scans was also used to image the ocular lens Results: A total of 10 eyes of 6 subjects were imaged in this study. Figure 1A and 1B show the ciliary body of two subjects with their eyes aligned to allow light penetration through the scleral tissue. The deep penetration of the SS- OCT wavelength together with image averaging allows for visualization of the deeper structures. Figures 1C illustrates the location of Schlemm’s canal, while 1D shows a collector channel branching out to an episcleral vein. Lastly, image 1E was acquired from a fully dilated patient. The 16mm long frame shows most of the lens capsule and its nucleus. The reduction in noise due to registration and averaging allows the user to discern the tissue layers Conclusions: We have demonstrated the ability of an SS-OCT system to obtain high definition structural images of the anterior segment structures and the aqueous outflow system. Being able to clearly asses such a tissue noninvasively could be a powerful tool to evaluate and track glaucoma surgery recipients as well as patients with uveitis and ciliary tumors Figure 1. HD-OCT of the anterior segment structures. (a-b) ciliary body, organ responsible for aqueous fluid production. (c-d) images show the outflow channel system. (e) lens imaging of a dilated patient
CONTROL ID: 3195657 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0133 TITLE: Automatic analysis to detect the area of corneal nerve fibers on images with in vivo confocal microscopy FIRST AUTHOR: Koichiro Shinji AUTHORS/INSTITUTIONS: K. Shinji, Hiroshima University, Hiroshima, JAPAN| Purpose: In vivo confocal microscopy (IVCM) imaging has been applied to evaluate structural alternation of cornea including corneal nerve fibers (CNF). Analysis methods for IVCM images are reported, but limited and not standardized so far. Our purpose is to detect the area of CNF (ACNF) with open and clear criteria, using new ® commercially available software: Quick Grain (Inotech, Hiroshima, Japan) that can analyze IVCM images automatically. Methods: Eleven healthy volunteers were included in this study. CNF were visualized by Heidelberg Retina Tomograph 3 equipped with a Rostock Cornea Module (Heidelberg Engineering, Heidelberg, Germany ). The highest-quality image was selected per 1 person to be analyzed. The selected image had a definition of 384 x 2 384 pixels over an area of 0.16 mm . At first, in order to distinguish CNF from background, some spots whose size was smaller than 5 pixels were eliminated as luminance unevenness. Following this procedure, we adjusted the threshold of brightness to detect CNF adequately. This threshold was adjusted to match manually detected shapes of CNF. In accordance with these settings, ACNF was detected automatically, and then compared with manually detected ACNF. Results: The mean automatically detected ACNF and manually detected ACNF were 4.8%/image and 4.6%/image. Automatically detected ACNF was significantly correlated with manually detected ACNF (p<0.01). Conclusions: Our results suggested that this new method can be an objective index to detect ACNF. Furthermore, threshold to detect objects can be adjusted, therefore this method would allow us to measure area of other corneal structures such as normal keratocytes and dendritic cells. (No Image Selected) CONTROL ID: 3195665 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0127 TITLE: Association between optic disc parameters and refractive error FIRST AUTHOR: Dian Li AUTHORS/INSTITUTIONS: D. Li, T. Elze, M. Wang, K. Wirkner, C. Engel, M. Loeffler, F.G. Rauscher, Leipzig Research Centre for Civilization Diseases (LIFE), Leipzig University, Leipzig, Saxony, GERMANY|D. Li, T. Elze, M. Wang, Schepens Eye Research Institute, Harvard Medical School, Boston, Massachusetts, UNITED STATES|A. Leutloff, C. Kuckert, Optometry and Ophthalmic Optics, Beuth University of Applied Sciences, Berlin, Brandenburg, GERMANY|A. Leutloff, C. Engel, M. Loeffler, F.G. Rauscher, Institute for Medical Informatics, Statistics and Epidemiology, Leipzig University, Leipzig, Saxony, GERMANY| Purpose: Myopia has previously been associated with changes in optic disc morphology and size, which itself interacts with ocular elongation. Here, we study the relationship of spherical equivalent (SE) of refractive error and optic disc area (corrected for ocular magnification vs. uncorrected), disc ovality, and disc rotation angle. Methods: From the population-based LIFE Adult study, a subgroup with extended eye examination was selected with reliable autorefractive SE (iProfiler Plus, Carl Zeiss Vision, Aalen, Germany) and peripapillary SLO fundus images (Spectralis SD-OCT, Heidelberg Engineering, Heidelberg, Germany). On fundus images, optic disc borders were marked on 16 equidistant radial locations (see Fig.1). Disc area, i.e. the area enclosed by the tracing, was additionally corrected for magnification by a method based on defocusing (Garway-Heath et al., Br. J. Ophthalmol. 82(6), 643-649, 1998). An optimal ellipse was fitted to each disc with the center, long and short axis, and rotation angle as parameters. Disc ovality was calculated as the ratio between the long and short axis. To study the relationship between angle, ovality, SE, and disc area, full Bayesian model comparisons of all parameter combinations were calculated with Bayes factor compared to the null model as the selection criterion. Results: 557 eyes of 285 subjects were selected (sex and age-stratified; age range: 25 to 85 years). Uncorrected disc area was positively correlated with SE (r=0.24, p<0.001). No significant correlation remained after correction for magnification (r=-0.04, p=0.32). The Bayesian model comparison revealed no relationships between rotation angle and any other parameters (Fig.2A). Larger ovality was associated with more negative SE (r=-0.14, p=0.001) and smaller corrected disc area (r=-0.20, p<0.001). The Bayesian model comparison revealed a combination between SE and disc area to be the best model for ovality (Fig.2B). Conclusions: The positive association between disc area and SE disappears after correction for ocular magnification, which indicates that myopic optic discs only seem to be smaller due to an increased distance between the lens and optic disc. Disc ovality, which can be used as a two-dimensional estimate of disc tilt and has previously been discussed as a risk factor for optic neuropathies, is related to myopia, smaller corrected disc area, and strongest to their combination. Fig.1 Fig.2 CONTROL ID: 3195676 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0143 TITLE: Application of New Imaging Technologies in High Myopic Eyes FIRST AUTHOR: Jochen Straub AUTHORS/INSTITUTIONS: J. Straub, C. Leahy, K.G. Foote, H. Bagherinia, Carl Zeiss Meditec, Inc., Dublin, California, UNITED STATES| Purpose: High myopia is a growing epidemic world-wide and is the subject of extensive scientific efforts. The tools available to scientists are mostly limited to refractive error, axial length, central choroidal thickness, and peripheral refraction. These tools provide mainly single measurement points, are difficult and time-consuming to administer, have limited availability, or are expensive. We demonstrate how new ophthalmic imaging technologies can provide better data for high myopic eyes over a large 90-degree field of view (FOV), including choroidal thickness, retinal curvature, and peripheral refraction maps. Methods: Retinal curvature and choroidal thickness were assessed using a prototype 200kHz swept-source optical coherence tomography system (OCT) with a 90-degree FOV and segmentation software. Retinal curvature was estimated using the method described by Steidle in Photonic Solutions for Better Health Care VI (2018). Peripheral TM refraction was measured using a CLARUS 500 fundus imager (ZEISS, Dublin, CA) with prototype software, using the method described by Everett (ARVO 2018). To validate the results, we created models of individual human eyes by customizing the Arizona Eye Model using measured axial eye length, refractive error, corneal power, and retinal curvature (Fig 2). Simulated peripheral refraction was compared to the measurement. Results: A total of 42 eyes of 21 subjects were enrolled in the study ranging in axial length from 21.92 to 29.81mm. 90-degree FOV OCT scans were acquired in 26 eyes of 13 subjects. Choroidal thickness ranged from 93 to 440 microns, retinal radius of curvature from 10.8 to 16.0mm. Peripheral refraction was measured in 42 eyes of 21 subjects and ranged from -2.15 to +9.0 diopters. Simulation and measurement of peripheral refraction were matched within +/- 3D. Conclusions: Wide-field fundus imaging and OCT enable new methods for measuring large FOV choroidal thickness, retinal curvature, and peripheral refraction. These technologies will help to further advance our knowledge of high myopia, myopia progression, and myopia treatment. Fig. 1(a) Estimation of retinal curvature and segmentation of retinal layer boundaries. (b) Relative peripheral refraction. Fig. 2: Simulation of an individual human eye, modelling refractive error and peripheral refraction. (a) layout of the optical model showing cornea, crystalline lens, and retina. (b) Respective spot diagrams on-axis and off-axis. CONTROL ID: 3195711 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0159 TITLE: Multifocal Electroretinographical Change of Laser-induced Choroidal Neovascularization Model in Non-human Primates FIRST AUTHOR: Ming Mei AUTHORS/INSTITUTIONS: M. Mei, Ophthalmology, WuXi AppTec, LTD, Suzhou, Jiangsu, CHINA| Purpose: To assess the multifocal electroretinogram (mfERG) change in laser-induced choroidal neovascularization (CNV) model in Cynomolgus monkeys treated with anti-VEGF substance. Methods: Six Cynomolgus monkeys with normal eyes were recruited. Eight spots of laser (532 nm) was shot on the retina around 1PD away surrounding the macular to break the Bruch's membrane to induce CNV. Two weeks after, examinations of mfERG, flash ERG, OCT, and FFA were performed, and the animals were randomly divided into two groups according to the degree of retinal blood vessel leakage. One group was intravitreally injected with Conbercept and the other with saline. The same ocular examinations were performed once at Week 1, 2, and 3 post-dose, respectively. Results: Two weeks post-laser shot, blood vessel leakage and retinal edema were observed at the laser spots. After grouping randomly according to the FFA leakage ranking, flash ERG showed similar signals between the two groups, but mfERG showed better signal in the planned Conbercept-treated group. After treatment, the leakage and retinal edema were significantly alleviated by Conbercept. Flash ERG showed an overall stronger signals, mfERG showed that the signal improvement was in the laser-injured area.

Conclusions: Compared to flash ERG, mfERG was a more precise and sensitive biomarker for anti-CNV drug efficacy assessment at the preclinical stage. (No Image Selected) CONTROL ID: 3195785 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0130 TITLE: Anterior Segment Optical Coherence Tomography imaging to study evolution of blebs after a new technique of ab interno collagen stent implantation with conjunctival peritomy FIRST AUTHOR: Sonal Dangda AUTHORS/INSTITUTIONS: S. Dangda, A. Do, J. Panarelli, New York Eye and Ear Infirmary, New York, New York, UNITED STATES|J. Panarelli, Ophthalmology, NYU Langone Health, New York, New York, UNITED STATES|M. Mavrommatis, Icahn School of Medicine, New York, New York, UNITED STATES| Purpose: To analyze changes in bleb morphology over time post-ab interno XEN Gel stent (Aquesys Inc., California, USA) implantation aided by conjunctival peritomy. Methods: Patients who underwent successful XEN Gel stent implant [defined as ≥20% reduction in intraocular pressure (IOP)] were included in the study. They were grouped according to their postoperative follow up duration: short-term (up to 3 months, group A), intermediate (>3 months-12 months, group B) and long-term (>12 months, group C). Bleb morphology was studied using the anterior segment feature of the Triton Swept Source Optical Coherence Tomography (AS-OCT). Twelve radial scans (16mm) centered over the bleb area were obtained. The bleb characteristics noted were: bleb wall thickness, internal height, and degree of internal reflectivity, which was classified as low, medium, high according to the color scale on AS-OCT. Bleb morphology was further characterized as uniform, subconjuctival separation, microcytic multiform and multiple internal layer. Results: Images were obtained on a total of 23 eyes. There were 9, 10 and 4 eyes each in Group A, B and C with the mean age of patients being 74.22±6.5, 69.55±8.50 and 61.5±3.41 years respectively. At a mean follow-up of 0.9±0.9 months, AS-OCT showed a bleb wall thickness of 115.67±22.3µm, bleb height of 547.56±230.77µm with the presence of a posterior episcleral fluid (PEF) lake of 179.0±106.85µm in group A. Thereafter, the bleb wall thickness and height showed a decline to 106.80±31.09µm(p=0.48) and 534.90±181.99µm(p=0.89) at a mean follow-up of 6.45±2.41 months in group B and 98.50±42.30µm(p=0.48) and 492.50±66.32µm(p=0.52) at a mean follow-up of 39.95±11.34 months in group C respectively, with a simultaneous increase in the PEF lake to 381.30±286.17µm in group B(p=0.06) and to 514.75±231.21µm in group C(p=0.05). Most of the blebs showed medium internal reflectivity (83%) and high bleb wall reflectivity (61%). The most common morphology was subconjuctival separation (67%) in the early period (Fig.1) which gradually remodeled to multiple internal layer (75%) in the late follow-up (Fig.2). Conclusions: AS-OCT can be the imaging of choice in assessing blebs with newer implants and considering variations in technique. This study characterizes features of bleb evolution in Xen implantation with conjunctival peritomy. Averaging success rate 4/ 4 .______l 0 Averaging success rate 4/4 CONTROL ID: 3195792 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0129 TITLE: Monitoring the effect of topical steroid administration for multiple subepithelial corneal infiltrates following epidemic keratoconjunctivitis FIRST AUTHOR: Taiichiro Chikama AUTHORS/INSTITUTIONS: T. Chikama, K. Shinji, Y. Kadohiro, Y. Kiuchi, Ophthalmology and Visual Science, Hiroshima University, Hiroshima, JAPAN| Purpose: The pathology of multiple subepithelial corneal infiltrates (MSI) following epidemic keratoconjunctivitis (EKC) consists of immune response by dendritic cells, which can be observed by in vivo confocal microscopy (IVCM). Formerly, we manually counted number of dendritic cells and convinced that they decreased by topical steroid administration. In this study, we have tried to establish an automated procedure to recognize dendritic cells. Methods: Six eyes of 6 cases with MSI following EKC participated in this study at Hiroshima University Hospital. All of them were treated with topical steroid administration. We manually counted number of dendritic cells at the initial visit and 1 month later. At the same time, the area occupied by dendritic cells were automatically detected with new ® software Quick Grain ( InoTech, Hiroshima, Japan). We evaluated a correlation between manually counted number of dendritic cells and automatically detected area of occupation by dendritic cells statistically. Results: At the initial visit and 1 month later, the automatically measured area was significantly correlated with 2 2 2 2 2 2 manually counted number (0.13mm /mm and 486.4/mm , p=0.03) (0.05 mm /mm and 301.1/mm , p=0.03). Compared with the initial visit, both number and area of dendritic cells significantly decreased 1month after (p=0.04 and p=0.03). Conclusions: Our result suggested that the automatically measured area occupied by dendritic cells could be a new index of MSI following EKC. This method would allow us to measure area of other corneal structures such as normal keratocytes and nerve fibers. (No Image Selected) CONTROL ID: 3195803 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0144 TITLE: Kidney Function is Associated with Drusen Characteristics FIRST AUTHOR: Mengyu Wang AUTHORS/INSTITUTIONS: M. Wang, K. Wirkner, C. Engel, M. Loeffler, J. Thiery, T. Elze, F.G. Rauscher, Leipzig Research Centre for Civilization Diseases (LIFE), Leipzig University, Leipzig, GERMANY|M. Wang, T. Elze, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, UNITED STATES|T. Ebert, A. Tönjes, Medical Department III – Endocrinology, Nephrology, Rheumatology, Leipzig University Medical Center, Leipzig, GERMANY|T. Ebert, Div. of Renal Medicine, Department of Clinical Sciences, Intervention and Technology (CLINTEC), Karolinska Institutet, Huddinge, SWEDEN|A. Tönjes, IFB AdiposityDiseases, Leipzig University Medical Center, Leipzig, GERMANY|C. Engel, M. Loeffler, F.G. Rauscher, Institute for Medical Informatics, Statistics, and Epidemiology (IMISE), Leipzig University, Leipzig, GERMANY|J. Thiery, Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, Leipzig University, Leipzig, GERMANY| Purpose: Chronic kidney disease (CKD) has been associated with age-related macular degeneration (AMD). We aim to further investigate if kidney function is related to drusen characteristics independent of known risk factors. Methods: The drusen characteristics were graded on 97 Spectralis spectral-domain optical coherence tomography (OCT) B-scans for 811 subjects from the population-based Leipzig Research Center for Civilization Diseases (LIFE) adult study. The eye with worse drusen characteristics was selected if both eyes were available. Drusen type was graded into 3 categories and ordinally coded: none (0), only hard drusen (punctate elevation under retinal pigment epithelium [RPE] with sharp margins, 1) and presence of soft drusen (dome-shaped structures under RPE with indiscrete margins, 2). Drusen size was measured in horizontal meridian within RPE and classed into 4 groups ordinally coded: none (0), small (<63 μm, 1), medium (≥63 but <125 μm, 2) and large (≥125 μm, 3). The estimated glomerular filtration rate (eGFR) as assessed by the CKD-EPI equation was compared by drusen type and size with t- test corrected for multiple comparisons. Optimal models predicting drusen characteristics were selected by stepwise regression from eGFR and known risk factors of age, sex, blood pressure, glycated hemoglobin A1c (HbA1c), antidiabetic treatment, and smoking status. Partial correlations between eGFR and drusen characteristic adjusting for risk factors remaining in the optimal models were evaluated. Results: The mean (standard deviation) of age for the 811 subjects (395 female) was 58.4 (10.8) years. Fig. 1 (a) and (b) show examples of drusen type and size graded on OCT. Fig. 2 (a) and (b) displays the eGFR distributions by drusen type and drusen size, respectively. Deteriorating kidney function with lower eGFR was significantly correlated to more severe drusen type (r = -0.10, p = 0.004) adjusting for age, sex, HbA1c, and smoking status, and significantly correlated to larger drusen size (r = -0.11, p = 0.002) adjusting for age, sex, and smoking status. Notably, Worsening kidney function significantly (p < 0.05 for both) contributed to the presence of hard/small drusen prior to early AMD compared to no drusen adjusting for known risk factors. Conclusions: Impaired kidney function is related to severe drusen type and size independent of known risk factors, and contributes to the early formation of hard/small drusen prior to early AMD. Fig. 1 Fig. 2 CONTROL ID: 3195856 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0131 TITLE: OCT assessment of anterior chamber inflammation in children: a pilot study FIRST AUTHOR: Ameenat Lola Solebo AUTHORS/INSTITUTIONS: A. Solebo, J. Rahi, PPP, UCL GOS ICH , London , UNITED KINGDOM|A. Solebo, R. Pattani, S. Akrabali, J. Rahi, C. Edelsten, Ophthalmology, Great Ormond Street Hospital, UNITED KINGDOM| Purpose: Disease activity in anterior uveitis, the most common manifestation of childhood disease, is currently assessed using the slit-lamp based SUN scale, which is open to interobserver variability, and not validated for paediatric use. We undertook a pilot study of anterior segment OCT (ASOCT) capture of active childhood anterior uveitis. Methods: Observational prospective study involving children with and without uveitis, with acquisition of Optovue Avanti anterior segment scans (8 line asterisk formation centred on corneal apex) per eye) following informed consent. Inter and intra-observer reliability of manual counting of acquired images (Bland-Altman limits of agreement, BA LoA), and sensitivity and specificity of ASOCT detection of active inflammation assessed. Results: A total of 26 children aged 3yrs to 15yrs (median 8yrs) underwent imaging, including 19 children with a known diagnosis of uveitis, of whom 12 had active chronic anterior inflammation as confirmed by two examiners at the slit lamp. Time taken to acquire images ranged from 22mins to 1.5mins per child, with patient acceptability scores for acquisition process consistently >85/100 on visual analogue scale. Mean intraobserver image count agreement - 0.3(95%CI -0.2/0.2), BA LoA -1.1(-1.4/-0.8) to 1.0(0.7/1.4). Interobserver agreement -0.5(-0.9/-0.2), BA LoA -2.5(-3.1/- 1.9) to 1.5(0.8/2.1). Sensitivity of ASOCT manual image cell count for diagnosis of active inflammation 92%(62%- 99%), specificity 86%(58%-98%), negative predictive value 92%(65%-99%). Conclusions: Non-contact, high-resolution ophthalmic imaging holds the promise of transforming paediatric practice. Further work is needed to determine the analytic validity, reproducibility, and concurrent and predictive clinical validity of manual and automated anterior segment OCT quantification of active inflammation. (No Image Selected) CONTROL ID: 3195862 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0150 TITLE: Non-contact ultra-widefield imaging follow-up of patients with retinopathy of prematurity treated with anti-vegf, laser, scleral buckling or cryotherapy using Optomap scanning laser FIRST AUTHOR: Abel Ramón AUTHORS/INSTITUTIONS: A. Ramón, E. Romo-García, S. Sital-Gastelum, S. Paz-Camacho, A. Meza-Anguiano, G. Gutierrez-Ruiz, T. ROMERO, Retina , Centro de Investigacion y Docencia en Cienias de la Salud, Culiacán , Sinaloa, MEXICO| Purpose: To describe the anatomical results in the follow-up of patients treated with anti-vegf, laser, scleral buckling or cryotherapy with retinopathy of prematurity Methods: A retrospective review of patients who attended the retinal service of Hospital Hospital Regional "Manuel Cárdenas de la Vega" with diagnosis of Retinopathy of Prematurity and treated with laser anti-vegf, cryotherapy or scleral cerclage between March 2018 and January 2019 Results: We included 14 patients from 7 patients with ROP diagnosis who needed treatment. 7 male and 7 female.

Scleral bucling: 3 eyes Laser: 8 eyes Cryotherapy: 2 eyes Antivegf: 10 eyes

The images were obtained by 3 evaluators to manipulate the patient's head, the body and one captured from photographs.

The images obtained show clear views of posterior pole, peripheral retina and are useful for evaluating scleral indentation, laser scars and cryotherapy, as well as helping in the follow-up of intra-retinal hemorrhages

We found a retinal detachment, residual intraretinal hemorrhages, most with stable retinas after treatment. Conclusions: Non-contact ultra-widefield imaging using Optos is capable of acquiring clinically useful high-quality images to evaluate anatomical results of patients treated for retinopathy of prematurity and help in the follow-up of these patients.

Optomap could be useful in documenting and monitoring patients treated for retinopathy of prematurity

CONTROL ID: 3195875 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0161 TITLE: Multispot Laser Therapy VS Conventional Laser and ultra wide-field photographs for Diabetic Retinopathy FIRST AUTHOR: TALIA ROMERO AUTHORS/INSTITUTIONS: T. ROMERO, E. Romo-García, S. Sital-Gastelum, S. Paz-Camacho, A. Meza-Anguiano, G. Gutierrez-Ruiz, A. Ramón, RETINA Y VITREO, CENTRO DE INVESTIGACIÓN Y DOCENCIA EN CIENCIAS DE LA SALUD, Sinaloa, MEXICO| Purpose: Compare the time of application of laser multispot vs conventional laser, as well as assess the pain that patients present during its application. Methods: It was a simple blind randomized clinical trial in patients with a definitive diagnosis of proliferative diabetic retinopathy and severe non-proliferative diabetic retinopathy In this study, patients were chosen for laser application in both eyes, one eye received conventional laser treatment and the other eye received the multispot laser treatment, the application time was measured with each of the lasers, and a pain scale was applied to each of the participants. Each of the patients underwent a complete ophthalmologic exploration as well as the performance of ultra wide-field photographs OPTOS before and after the laser application. Results: We studied 17 pairs of eyes to detect a difference of at least 1 minute at the time of application, assumed an average time of 4.5 ± 1 in the group of a single shot. The formula T and a level of significance of 5% were used. Conclusions: The laser multispot proved to be less painful as well as requiring less application time. As well as taking ultra-wide field photos allows us to evaluate changes in the retina due to diabetes and that the patient better understand his illness. (No Image Selected) MRI CONTROL ID: 3189098 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0165 TITLE: Preoperative magnetic resonance imaging of superior oblique muscle: its role on surgical selection in children FIRST AUTHOR: Jingyuan Zhu AUTHORS/INSTITUTIONS: J. Zhu, J. Jiang, N. Li, ophthalmology, Beijing Children's Hospital, Beijing, CHINA| Purpose: To explore the practicability of preoperative magnetic resonance (MR) imaging of superior oblique muscle on surgical selection of patients under 18 years old with superior oblique (SO) muscle palsy and to determine whether the routine use of this MR imaging successfully guides the surgical procedures. Methods: Patients under 18 years old with unilateral class I or II superior oblique muscle palsy (Knapp Classification) examined by prism, eye movement and visual function to meet clinical criteria were evaluated with T2-weighted MR imaging before surgery from January 1, 2015 through December 31, 2016. In this observational, single-center, cohort study, we studied 82 patients who underwent an ipsilateral SO tuck after MRI manifestation of SO hypoplasia at the tendon, and underwent weakening of the ipsilateral inferior oblique (IO) muscle (IO recession or IO denervation & extirpation) after the MRI manifestation of hypoplasia or absence of the SO at the belly. We followed up the long-term surgical outcomes from January 1, 2017 through December 31, 2018 (2 years postoperative or more) by comparing the change of prism degree (PD). Results: Thirty patients were manifested SO hypoplasia at the tendon through T2-weighted imaging. They were treated with ipsilateral SO tuck and the postoperative primary position deviation was not greater than 5 PD (28 of 30 patients, p<0.05). The greatest hypertropia of these patients was in opposite down oblique field, which means SO underacting, corresponding to Knapp Classification II. Fifty two patients were manifested hypoplasia or absence of the SO at the belly through T2-weighted imaging. They underwent IO recession or IO denervation & extirpation and the postoperative primary position deviation was not greater than 5 PD (50 of 52 patients, p<0.01). The greatest hypertropia of these patients was in opposite up oblique field, which means IO overacting corresponding to Knapp Classification I. Conclusions: Preoperative MR imaging of SO might partially benefits surgical selection for patients with SO palsy under 18 years old. When used before surgery, MR imaging might guide the surgical procedures also corresponding to clinical criteria. Hypoplasia of right superior oblique at tendon, demonstrating superior oblique underaction in right eye. Absent belly of left superior oblique, demonstrating possible inferior oblique overaction in left eye. CONTROL ID: 3190994 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0166 TITLE: Measuring the Fluid Viscosity of Vitreous Body using MRI FIRST AUTHOR: Xingzheng Pan AUTHORS/INSTITUTIONS: X. Pan, E. Vaghefi, School of Optometry and Vision Science, University of Auckland, Auckland, NEW ZEALAND|S.S. Thakur, B. Pontré, G.L. Kumarasinghe, I.D. Rupenthal, Department of Ophthalmology, School of Medicine, University of Auckland, NEW ZEALAND|P. Donaldson, Department of Physiology, School of Medical Science, University of Auckland, NEW ZEALAND| Purpose: It has been shown that vitreous viscosity changes with age and these changes are highly correlated with retinal detachment. However, there is no non-invasive method to measure the viscosity clinically at the moment. We propose a novel non-invasive MRI-based viscometry method. Methods: Synthetic vitreous samples with various viscosities were prepared by mixing various concentrations of hyaluronic acid and agar (1:1 weight ratio). The rheological parameters – storage modulus (G'), loss modulus (G''), complex viscosity (η*) and consistency index (K) of these samples were measured using a Discovery HR-2 rheometer (TA Instruments, USA). A 3T clinical MRI (SKYRA, Siemens, Germany) equipped with a 16 channel knee coil was used to image samples. Transverse relaxation rates (R ) were processed from MR images using custom-built 2 software. Clinical translation was attempted by measuring the same parameters of ex-vivo porcine eyes (n = 7). Results: MRI-measured of the synthetic vitreous samples correlated well with rheology-obtained parameters using exponential fits. Based on the measured MRI data of ex-vivo porcine vitreous and using the correlations obtained previously, a η*and K were predicted to be 2.37±0.40 Pa.s and 0.10±0.01, respectively. These values agreed with that of measured by rheology which were 2.34±0.59 Pa.s and 0.38±0.08, respectively. Conclusions: We have developed a non-invasive MRI-based viscometry technique, which has the potential of being implemented clinically. Our next step is to put our technique through a clinical trial to determine vitreous liquefactions involved in normal aging eyes and ocular pathologies. (No Image Selected) CONTROL ID: 3193958 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0164 TITLE: Personalized geometrical models of the human eye developed from MRI scans at 9.4 Tesla FIRST AUTHOR: Patrick R Merz AUTHORS/INSTITUTIONS: P.R. Merz, G.U. Auffarth, Department of Ophthalmology, Lions Eyebank, Heidelberg, GERMANY|J. Chacon-Caldera, L.R. Schad, Computer Assisted Clinical Medicine, Medical Faculty Mannheim, Mannheim, GERMANY|S. Dörsam, P. Zirjacks, E. Friedmann, Interdisciplinary Center for Scientific Computing, Institute of Applied Mathematics, Heidelberg, GERMANY|S. Litau, B. Wängler, Molecular Imaging and Radiochemistry, Department of Clinical Radiology and Nuclear Medicine, Mannheim, GERMANY|P.R. Merz, S. Dörsam, G.U. Auffarth, E. Friedmann, Department of Ophthalmology, David J Apple Center for Vision Research, Heidelberg, GERMANY| Purpose: We develop data based geometrical models for individual human eyes where each organ is described by suitable mathematical functions. The model parameters are calibrated to high resolution 2D multi slice MRI scans of human autopsy eyes. The resulting geometrical models can be used as 3D printouts for experimental work in the laboratory or for grid constructions to perform numerical simulation of therapeutic approaches for several eye diseases like retinal diseases and glaucoma Methods: We measured 7 autopsy eyes (Ethics votum S-134/2018) in a preclinical small animal MRI scanner (Bruker Biospec 94/20, Germany) using a Bruker quadrature volumetric transceiver coil. All procedures conformed to HIPAA regulations and the Declaration of Helsinki for research involving human subjects. Human autopsy eyes were enucleated within 24h after death and used within 48h. Intraocular pressure was adjusted to normal by injection of ophthalmic viscosurgical device (HEALON, Johnson & Johnson Vision, USA) near the optic nerve before examination. For measurements eyes were put into 50 ml centrifugation tubes (Sarstedt, Germany). Measurements were performed using a 2D RARE sequence with the following parameters: TE/TR=60/3085ms, FA=90°, Rare Factor 8, 2 Resolution=100x100µm , Slices=24 with 4mm thickness, Averages=12, TA=22m49s Results: From the generated digital imaging data (Fig. 1) we segment the geometry of the components of the eye (Fig. 2), i.e. anterior chamber, vitreous body, lens, iris, ciliary body, cornea and sclera. The mathematical models for the shape of these components are calibrated to these imaging data using parameter estimation methods (method of least squares) and are visualized in a 3D scene Conclusions: With the MRI scans at 9.4 Tesla we obtained high resolution images to segment the components of the eye. With this data we reconstruct personalized human eye models which can be used for numerical simulation to test in silico personalized therapies of retinal diseases and glaucoma Fig. 1: left: the high resolution MRI scans at 9.4 Tesla of a human autopsy eye; middle: the 2D RARE sequence no 7 from totally 12, representing data of a slice of 4 mm thickness; right: the structures are captured with all details, here the lens, ciliary body, iris and cornea Fig. 2: Geometrical representation of the single components of the human eye after segmentation and model calibration from the 3D MRI scans at 9.4 Tesla CONTROL ID: 3195742 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0163 TITLE: Magnetic resonance imaging of anti-VEGF drug diffusion in the human eye FIRST AUTHOR: Simon Dörsam AUTHORS/INSTITUTIONS: S. Dörsam, S. Litau, P.R. Merz, B. Wängler, L.R. Schad, G.U. Auffarth, E. Friedmann, J. Chacon-Caldera, University of Heidelberg, Heidelberg, GERMANY| Purpose: Today's standard therapy of the most important epidemiological retinal diseases e.g. age-related macular degeneration and macular edema in diabetic retinopathy is the intravitreal anti-VEGF (Vascular Endothelial Growth Factor) monotherapy. It consists of serial injections into the vitreous humor through pars plana. The exact amount and position of the injections relies on empirical knowledge and the treatment often fails. Here we present dynamic MR ® imaging of the anti-VEGF drug EYLEA after injection in autopsy eyes to visualize its distribution. Methods: All procedures conformed to HIPAA regulations and the Declaration of Helsinki for research involving human subjects. We performed MRI on two human autopsy eyes (Ethics votum S-134/2018) enucleated within 24h after death and scanned within 48h. Intra ocular pressure was adjusted to normal by injection of ophthalmic viscosurgical device (HEALON, Johnson & Johnson Vision, USA) near the optic nerve before examination. To make the drug visible in the MRI, the syringe was washed out with 2 mg gadolinium before use. The remaining amount was mixed with the drug and used as a stainer. The injection of 2 mg (50 µl) EYLEA® was performed following the instructions for use. For comparison in the second eye 2 mg gadolinium was injected. The eyes were scanned in 50ml centrifugation tubes (Sarstedt, Germany). MRI measurements were acquired simultaneously in a preclinical 9.4T animal scanner (Bruker Biospec 94/20, Ettlingen, Germany) using a Bruker quadrature volumetric transceiver coil 5 min after drug injection. A 2D RARE sequence was used with the parameters: TE/TR=11/618ms, FA=90°, Rare Factor 4, Resolution=200µmx200µm, Slices=24 with 4mm thickness, Averages=3, TA=22m49s. Results: We obtained whole eye data of drug distribution which diffused inhomogeneously in the vitreous and was visible around the injection site, the middle of the eye and at the bottom near the optic nerve. The interconnected shape of the drug distribution indicates the action of the gravitational force. Conclusions: The data could be used for treatment planning and outcome predictions. Moreover, vitreous consistency which differs individually with age and disease could also be assessed. Distribution of gadolinium in the first autopsy eye and of anti-VEGF stained with gadolinium in the second autopsy eyes.
Ultrasound CONTROL ID: 3194746 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0167 TITLE: 3D ultrasound biomicroscopy (3D-UBM) imaging and automated whole eye assessment of the iridocorneal angle FIRST AUTHOR: David L Wilson AUTHORS/INSTITUTIONS: D.L. Wilson, H. Wu, T. Yu, A.T. Minhaz, Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, UNITED STATES|R. Helms, D.D. Sevgi, F. Orge, Opthamology, Case Western Reserve University, Cleveland, Ohio, UNITED STATES| Purpose: We created a new high-resolution (50-MHz) 3D ultrasound biomicroscopy (3D-UBM) imaging system to enable new assessments of the anterior segment structures (e.g., cornea, anterior chamber, iris, trabecular-iris angle, and ciliary body and processes). Here, using 3D-UBM image volumes, we report automated 360-degree assessment of the trabecular-iris angle (TIA), an important biomarker for glaucoma patients. Methods: The 3D-UBM system automatically linearly scans a 2D UBM probe across the eye and collects several hundred 2D images to create a high-resolution volume. To assess TIAs, processing steps are: (1) Automatically align images to reduce effects of eye motion. (2) Reduce noise using advanced 3D algorithms. (3) Use interactive visualization software to create reformatted, rotational views about the optic-axis. (4) Use a semantic-segmentation convolutional neural network (CNN) to segment the anterior chamber. (5) Use a modified, automated TIA method to assess the angle for each of 360-degree rotational views. (6) Generate statistical reports and an en face heat map of TIAs. Results: CNN segmentations of the anterior chamber gave folded “leave-one-eye-out” excellent Dice score of 0.96±0.01. Most errors arose from ultrasound artifacts at the front of the anterior chamber rather than from the corners where errors would affect the TIA. Automated measurements tended to lie within the spread of 4 readers, giving insignificant difference to readers (p = 0.996). In normal, in vivo eyes, the mean angle was within expected clinical range. Good analysis and measurement repeatability was determined by comparing results from different operators of the software on a single eye and by repeated measurements of a cadaver eye, respectively. For example, 360 measurements at different rotational angles around a single normal eye gave 36.5±5.8 deg and COV of 0.15. Conclusions: Using 3D-UBM, one can acquire 3D data sets and measure TIA in a highly automated fashion. Clearly, the method has sufficient precision to assess a condition such as angle closure glaucoma. Potentially, the ability to assess TIA over the entire eye with heat maps (Figure) could inform a more precise assessment of drainage.
Figure. Heat map of TIA measurements.
CONTROL ID: 3195445 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0168 TITLE: Scleral Strain Artefacts due to Spatiotemporal Distortion in High-Resolution, High-Frame Rate Ultrasound Imaging FIRST AUTHOR: Sunny Kwok AUTHORS/INSTITUTIONS: S. Kwok, T. Sandwisch, K. Clayson, Y. Ma, J. Liu, Biomedical Engineering, The Ohio State University, Columbus, Ohio, UNITED STATES|J. Liu, Ophthalmology and Visual Science, The Ohio State University, Columbus, Ohio, UNITED STATES| Purpose: Ultrasound imaging of objects in motion may suffer from image distortion due to sequential A-line scanning. This study aims to characterize and reduce distortion-induced strain artefacts in ultrasound elastography of the sclera. Methods: A 50 MHz ultrasound probe (Vevo 2100, VisualSonics) was used to acquire radiofrequency B-mode frames at 128 frames per second of a porcine sclera during whole eye motion. The eye was displaced either parallel or perpendicular to the imaging axis of the ultrasound probe to simulate axial and lateral displacements, respectively (Fig 1A). The globe was cyclically displaced at a frequency of 1.5 Hz for five cycles of 10, 50, and 100 µm in both directions, simulating a potential range of whole eye motion during fixation. A previously validated ultrasound speckle tracking algorithm (Tang & Liu, JBME 2012) was used to estimate the strains for the image frames corresponding to peak and trough displacements of each cycle (Fig 1B). Frames immediately next to the peak and trough but within the same half-cycle (Fig 1C) were used to evaluate whether the strain artefacts can be reduced by selecting the frames obtained during eye motion of the same speed and direction. Results: Parallel and perpendicular motion both produced axial and lateral strain artefacts of small amplitudes (0.010% and 0.032%, respectively, Fig 2). Lateral strain artefacts are more prominent at higher displacements, and the magnitude of both axial and lateral strain artefacts appear to increase with increased speed of motion. Frames within the same half-cycle had significantly smaller strain artefacts, reducing to 0.01%, 0.02%, and 0.03% for 10, 50, and 100 µm, respectively (Fig 2). Conclusions: Ultrasound imaging modalities relying on sequential A-line scanning may create image distortion when the imaged object is in motion, and this distortion may generate measureable strain artefacts in high-resolution ultrasound elastography. Using frames within the same half cycle, i.e. frames obtained at the same velocity, could minimize strain artefacts over a range of eye motion. Fig 1: (A) Scleral motion relative to ultrasound imaging axis; (B) initial frame selection process to calculate strain; (C) improved frame selection within same half-cycle Fig 2: Strain artefacts before (black) and after (red) reduction of (A) axial and (B) lateral strain in both parallel and perpendicular probe orientations Image Guided Surgery CONTROL ID: 3193864 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0170 TITLE: Primary results of clinical use of the Beyeonics digitally enhanced surgical visualization system in cataract and vitreoretinal surgery: a prospective study. FIRST AUTHOR: Adiel Barak AUTHORS/INSTITUTIONS: A. Barak, A. Loewenstein, Ophthalmology, Tel Aviv Medical Center, Tel Aviv, ISRAEL|A. Barak, Ophrthalmology, Tel Aviv University, ISRAEL|R. Schneider, S.H. Bakalash, Beyeonics LTD., ISRAEL|A. Loewenstein, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, ISRAEL| Purpose: The Beyeonics system is a digitally enhanced surgical visualization system that was adapted from a fighter pilot technology. It consists of a head wearable display, and a high-resolution 3D imaging and software platform. The head display projects magnified images and data from multiple sources and is controlled by intuitive head gestures. Due to the use of head wearable display and head gesture usage, concerns were raised about the learning curve, fatigue and discomfort, which may rise during intraocular surgery. Thus, a clinical study was initialized aimed to compare surgeons' opinions and comfort levels using Beyeonics system, as well as performing a primary evaluation of anatomical surgical results. Methods: Five surgeons performed the total of 34 surgeries using the Beyeonics system. 9 surgeries were cataract extraction performed by anterior segment surgeon (DV). All other surgeries were primarily vitrectomies with/without cataract extraction-performed by experienced vitreoretinal surgeons. Ergonomics, educational value, image sharpness, depth perception, field of view and technical skills were analyzed through analysis of a questionnaire, comparing surgeons experiences to normal microscope usage. Results: 34 patients were enrolled in the study. Surgeries included cataract surgeries (9) and variety of vitreoretinal procedures, including vitrectomy for retinal detachment, ERM removal, vitreous hemorrhage and silicone oil removal. Depth perception was rated similar to normal microscopy. Field of view, educational values and image resolution were rated superior when using the 3-D system. Color perception and ergonomics were rated superior when using traditional microscopy. All 34 surgeries (100%) achieved anatomic success with one surgery. Conclusions: The Beyeonics system was superior from an ergonomics perspective, as compared with the microscope. It enabled the physician freedom of movement, operating while being aware of the happening in the OR. Physicians reported a short learning curve and was described by surgeons as equal or superior in most surgeries performed. The head-wearable display system is offering a novel way of operating which may become a new standard for ophthalmic surgery as ongoing improvements are applied. (No Image Selected) CONTROL ID: 3195574 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0169 TITLE: Multimodal image-guide laser induced retinal vein occlusion in living rabbits FIRST AUTHOR: Yanxiu Li AUTHORS/INSTITUTIONS: Y. Li, V. Nguyen, Y.M. Paulus, Department of Ophthalmology and Visual Science, University of Michigan, Ann Arbor, Michigan, UNITED STATES|Y. Li, X. Xia, Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, CHINA|W. Zhang, X. Wang, Y.M. Paulus, Department of Biomedical Engineering, University of Michigan, Michigan, UNITED STATES| Purpose: Retinal vein occlusion (RVO) is a leading cause of vision loss and blindness in older patients. We perform multimodal imaging with integrated photoacoustic microscopy (PAM) and spectral-domain optical coherence tomography (SD-OCT) to improve the efficiency for visualizing RVO and retinal neovascularization (RNV) in living rabbits using a Rose Bengal RVO model. Methods: The Rose Bengal laser-induced RVO model was performed on 8 New Zealand rabbit eyes. A custom- made, multimodal imaging system with a wavelength tunable pulsed laser (3-6 ns pulse duration, 1 kHz repetition rate, Ekspla NT-242, Lithuania), a scan lens, an ocular lens, and an ultrasonic transducer was integrated and coaxially aligned with a Thorlabs Ganymede-II-HR OCT. Retinal vascular dynamic changes were monitored and evaluated at 4, 28, 35, 42, and 84 days following laser-induced RVO, using the multimodal PAM and OCT imaging along with conventional color fundus photography and fluorescein angiography (FA). Results: In vivo experiments demonstrates that RNV developed in the rabbit RVO model at day 28 after photocoagulation, and the RNV was stable from 1 to 3 months (Figure 2). SD-OCT can identify the cross-sectional structure of RVO. PAM imaging characterized the location and the margins of the occluded vasculature as well as the morphology of individual RNV with high contrast and high resolution. Conclusions: The multimodality PAM-OCT system can precisely help visualize and distinguish individual retinal microvessels, their depth, and the surrounding anatomy. Thus, the proposed multimodal ocular imaging system may provide a potential imaging platform for improved visualization and characterization of retinal neovascularization diseases in a safe and efficient manner in future. Detection and stabilization of RNV in RVO model: (a-d) color fundus images of the retina acquired at 28, 35, 42, and 84 days following RVO induction. (e-h) Zoom-in view of the black rectangles from Figure a-d. (i-l) Corresponding maximum intensity projection PAM images acquired along the black rectangles from Figure a-d. The PAM images illustrate the structure of individual RNV is unchanged. (m-p) cross-sectional B-can OCT images acquired along the black lines in Figure e-h. CONTROL ID: 3195766 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0172 TITLE: Photoacoustic signal-guided photo-mediated ultrasound therapy as a novel method to remove microvasculature FIRST AUTHOR: Yixin Yu AUTHORS/INSTITUTIONS: Y. Yu, W. Zhang, Y. Qin, X. Xie, X. Wang, Y.M. Paulus, University of Michigan, Ann Arbor, Michigan, UNITED STATES|Y. Yu, Xiangya Hospital, Central South University, Changsha, Hunan, CHINA|X. Yang, University of Kansas, Kansas, UNITED STATES| Purpose: Real-time image-guided retinal laser therapy allows for automatic feedback to reliably and reproducibly treat the vasculature. We have developed a novel, controllable, signal-guided photo-mediated ultrasound therapy (PUT). PUT can selectively treat blood vessels using a combination of low intensity nanosecond pulse duration laser and ultrasound without damaging surrounding tissue under real-time guidance of photoacoustic (PA) signal. Methods: An integrated PA sensing and PUT system was developed to investigate the feasibility of using a laser system (Continuum Powerlite DLS 8010, Santa Clara, CA) and ultrasound transducer (H107, Sonic Concepts, Bothell, WA) for PA sensing and PUT treatment in real-time. Nd: YAG laser was used as the source to produce laser and the ultrasound transducer was used to deliver ultrasound bursts to the treatment area and detect PA signals in real-time. During PUT treatment, different anti-vascular effects were induced in an in vivo chicken yolk sac membrane model, 45 samples were involved. The anti-vascular effects related PA signal changes were immediately detected with PA sensing. Results: PA signals were acquired before and immediately after PUT. Statistically significant differences were found before and after treatment in shrinkage and rupture effect. A significant decrease in PA signal amplitude was observed with vessel shrinkage, whereas vessel rupture induced a significant increase in PA signal. The real-time detected PA signals were divided into three groups based on the treatment effect. When no treatment was noted on the target vessels, no notable change was shown in detected PA signal. PA signal decreased significantly with PUT vessel shrinkage. PA signal amplitude increased abruptly with PUT causing vessel rupture until the bleeding stabilized. Conclusions: With the capability to precisely control the treatment effects, PA-guided PUT holds significant promise as a novel, non-invasive, controllable method to treat the eye microvasculature with reduced side-effects and no systemic photosensitizing dye. (No Image Selected) CONTROL ID: 3195827 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0171 TITLE: Microscope integrated optical coherence tomography-guided autologous full thickness neurosensory retinal autograft for large macular hole related total retinal Detachment FIRST AUTHOR: ankur singh AUTHORS/INSTITUTIONS: A. singh, M. Dogra, B. Tigari, R. singh, Advanced Eye Centre, Post Graduate Institute of Medical Education & Research, Chandigarh, Ghaziabad, Uttar Pradesh, INDIA| Purpose: To evaluate the feasibility and utility of microscope integrated optical coherence tomography (MIOCT) in patients undergoing full thickness neurosensory retinal autograft for refractory macular hole associated retinal detachment. Methods: We analyzed two eyes of two patients who had undergone a neurosensory retinal autograft for large macular hole associated retinal detachment. Both cases had MIOCT guided placement and sizing of the retinal autograft. Time taken for obtaining MIOCT images, real-timemorphology of the retinal autograft (intraoperative and postoperative), anatomic and functional outcomes were noted. Results: The first case had optic disc pit related maculopathy with a large macular hole and total retinal detachment. She had undergone a vitrectomy with internal limiting membrane peeling elsewhere. The second patient had a treatment naive large macular hole with total retinal detachment. Both patients underwent vitrectomy with MIOCT guided autologous neurosensory retinal autograft placement and silicone oil tamponade. At 6 month and 3-month follow-up respectively, both patients had closed macular holes, attached retinas, and improvement in visual acuity. Conclusions: MIOCT provides intra-operative visualization of macular holes and provides real-time feedback regarding dimensions of the retinal autograft, thus aiding in, accurate sizing of the graft. This ensures that the autograft fits snugly in the macular hole, thereby restoring the macular structure. Intra-operative surgeons view showing large full-thickness macular hole with
associated retinal detachment and its MIOCT aided management in, case one. Intra-operative surgeons view showing large full-thickness macular hole with
associated retinal detachment and its MIOCT aided management in, case two. Image Processing and Image Analysis CONTROL ID: 3193940 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0177 TITLE: Motion correction in 3D-OCT data by intensity-based image registration: an evaluation study FIRST AUTHOR: Luisa Sanchez Brea AUTHORS/INSTITUTIONS: L. Sanchez Brea, D. Andrade de Jesus, T. van Walsum, S. Klein, Erasmus MC, Rotterdam, NETHERLANDS| Purpose: A modular open-source registration software (Elastix) is used to systematically study the performance of different registration approaches in 3D-OCT imaging. Methods: Two public datasets of fovea-centred volumes (6.7x6.7mm) collected with a Bioptigen SD-OCT (NC, USA) were used – 20 Age-related Macular Degeneration (AMD) subjects in dataset 1, and 249 AMD subjects and 115 healthy controls in dataset 2. Each volume consists of 100 B-scans and 1000 A-scans per B-scan. Dataset 1 was used to study inter-observer variability, and dataset 2, for the evaluation of the registration. Intensity-based registration with rigid transformation models (translations (T) and translations plus rotations (E)) applied between consecutive B- scans was considered (Fig. 1). Mean squared difference (MS), normalized correlation (NC), and mutual information (MI) similarity metrics were compared. Manual segmentations of the inner limiting membrane, inner retinal pigment epithelium, and outer Bruch's membrane, by two experts in dataset 1, and by one expert in dataset 2, were used for validation. Results: Inter-rater variability was calculated as the mean axial difference between the segmentations of two experts on the same B-scan, yielding 4.2±0.7, 7.4±1.4, 5.1±1.1 (μm) for each layer respectively. The average distances between layer segmentations on consecutive B-scans before registration were 16.4±5.6, 17.5±5.4, and 16.5±5.6 for the AMD and 12.2±4.2, 12.3±4.2, and 12.2±4.2 for the control group. All the approaches reduced the misalignments between B-scans observed in the original volumes. The combination of E and MI showed the best results (6.2±3.2, 8.6±3.5, 7.0±3.2 for the AMD group and 4.8±2.3, 5.2±2.4, 5.1±2.4 for the control group). Significant differences (Mann-Whitney U p<0.01) were observed between the AMD and control groups, both in the original volumes and after registration (Fig. 2), and also when comparing the results of different registration approaches in the same group, AMD or Control (Wilcoxon signed-rank p<0.01). Conclusions: Intensity-based image registration effectively reduces the misalignment between consecutive B-scans. The choice of registration parameters has a significant impact on the accuracy, so further validation and algorithm optimization studies are recommended. Registration pipeline. Distribution of the average differences in the unregistered and registered data. CONTROL ID: 3194203 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0176 TITLE: Grid deformation analysis of the macula and postoperative metamorphopsia after macular hole surgery FIRST AUTHOR: Ji Eun E Lee AUTHORS/INSTITUTIONS: J.E. Lee, S. Park, J. Lee, H. Kwon, S. Park, Ophthalmology, Pusan National University Hospital, Busan, KOREA (THE REPUBLIC OF)| Purpose: Postoperative metamorphopsia was reportedly related to deformation of the macula after macular hole surgery with the internal limiting membrane peeling. However, the mechanism is still under debate. The purpose of the present study was to investigate the correlation between postoperative metamorphopsia and macular deformation by analyzing the deformation of the grid overlaid on the fundus photograph. Methods: Consecutive eyes that underwent pars plana vitrectomy for idiopathic macular hole and achieved hole closure were analyzed retrospectively. The fundus photographs were taken before and at 6 months. Postoperative vertical and horizontal metamorphopsia was assessed using M-chart. Two photos were overlapped matching the major arcade vessels. The 6x6 mm grid having crossing lines at 1mm interval was overlaid on the preoperative photo. Each node was anchored at the photo, which was deformed by moving the node to match the retinal vasculatures to those of the postoperative photo (Figure 1). Differences in the coordinates of the nodes were calculated and analyzed to find correlation with M-score. Parafoveal deformation was defined as differences in coordinates between the center node and the first adjacent nodes, and perifoveal deformation as between the first and second nodes. Results: In 33 eyes, the average displacements of the nodes were 25.92um to the disc and 14.03um inferiorly. On the vertical lines of the grid, the average difference in X-coordinates between the adjacent nodes was 81.62 um, and on the horizontal line, the average difference in Y-coordinates was 47.01 um. Horizontal M-score was correlated with the superior perifoveal horizontal deformation of the vertical line on the fovea (p=0.013), and vertical M-score was correlated with the temporal perifoveal vertical deformation of the horizontal line on the fovea (p=0.007) Conclusions: Postoperative metamorphopsia after macular hole surgery was correlated with perifoveal deformation of the macula. Figure 1. Grid analysis of postoperative deformation of the macula after macular hole surgery. CONTROL ID: 3194216 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0173 TITLE: Automated segmentation of geographic atrophy using U-Net on custom-generated SD-OCT en face images FIRST AUTHOR: Niranchana Manivannan AUTHORS/INSTITUTIONS: N. Manivannan, L. de Sisternes, M. Durbin, Carl Zeiss Meditec, Inc., Dublin, California, UNITED STATES|G. Gregori, P.J. Rosenfeld, Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, UNITED STATES| Purpose: Geographic atrophy (GA) is a condition associated with loss of the retinal pigment epithelium (RPE) and represents the late stage of non-exudative age-related macular degeneration (AMD). This research aims to develop a fully-automated segmentation and quantification algorithm for spectral domain optical coherence tomography (SD- OCT) en face images. Methods: This retrospective study used 250 macular cubes (512x128x1024: 58, 200x200x1024: 192) obtained from 155 patients using CIRRUS™ HD-OCT 4000 and 5000 (ZEISS,Dublin,CA). Experts manually drew the GA ground truth (GT) segmentations in the en face images. For each macular cube, a 3-channel en face GA projection image was generated by combining 1) sub-volume section of choroid; 2) slab projection surrounding RPE and 3) retinal thickness between the RPE and inner limiting (ILM) layer. The training and testing sets of custom-generated en face images were comprised of 225 eyes (GA:187, drusen with no GA:19 and healthy:19) and 25 eyes (GA:11, drusen with no GA:5 and healthy:9). The contracting, bottleneck and expansive path of the U-Net consisted of 4 convolutional neural networks (CNN), 2 CNN with 0.5 dropout and 5 CNN blocks (fig. 1). Binary cross entropy and dice coefficient loss were used for training. ‘Icing on the Cake’ was used to fine-tune the model. Segmentations by the algorithm in the test set were compared with the GT using quantitative measurements (Bland-Altman, area and Pearson’s correlation). Results: Fig. 2 shows the results of the proposed algorithm, Advanced RPE Analysis and the GT. The absolute and 2 fractional area differences between GA regions generated by the proposed algorithm and the GT were 0.11±0.17mm 2 and 5.51±4.7% as opposed to 0.54±0.82mm and 25.61±42.3% for Advanced RPE Analysis. The inference time was 1183 ms per en face image using an Intel® i7CPU. Correlations of GA areas generated by the proposed algorithm and Advanced RPE Analysis with the GT were 0.9996 (p-value<0.001) and 0.9259 (p-value<0.001). The Bland- Altman plot between the GT and the segments generated using proposed algorithm showed stronger agreement than advanced RPE analysis. Conclusions: Quantitative and qualitative evaluations demonstrated that the proposed algorithm for segmenting GA in SD-OCT en face images showed very strong agreement with ground truth by manual grading. Fig 1. Flowchart of proposed algorithm Fig 2. Results of the proposed algorithm CONTROL ID: 3194596 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0174 TITLE: Enhanced visualization of choroidal vasculature map using structural OCT FIRST AUTHOR: Areg Noshadi AUTHORS/INSTITUTIONS: A. Noshadi, H. Bagherinia, T. Callan, M. Durbin, Carl Zeiss Meditec, Inc., Dublin, California, UNITED STATES| Purpose: The analysis of the choroidal layer using structural optical coherence tomography (OCT) has become an important tool for the diagnosis of ocular diseases. Through generating choroidal slabs, the vasculature in this layer can be visualized. However, artifacts due to overlying superficial retinal vasculature shadows can lead to misinterpretation of the choroidal vasculature maps. We present a method to reduce the described artifacts from choroidal layer en face images for enhanced visualization and analysis. Methods: Our method has following steps: 1) Segment retinal pigment epithelium (RPE) and choroidal-scleral boundary. 2) Create RPE retinal layer slab from 40μm above RPE to 10μm below RPE. 3) Create choroidal layer slab from 10μm below RPE to scleral-choroidal boundary. 4) Assuming that RPE retinal slab U is partially superimposed on choroidal layer slab X resulting in a choroidal layer slab with vessel shadow artifacts I, the artifact removal can be formulated as X = I – wU with w ∈[0,1] being the fraction of U superimposed on X. w is solved by minimizing the weighted average of the normalized cross-correlation 2 2 square γ of local region U and X (8x8 pixels) with the local weight of v =Var(U )Var(X ), formulated as min ∑ γ (U ,X i i 2 i i i w i i )v /∑ v . The explicit solution is w=∑ Cov(U ,I )Var(U )/∑ Var (U ). i i i i i i i i i i

Performance of the algorithm was evaluated using 96 SS-OCT volume data of 500x500 A-scans over 12x12mm acquired using PLEX® Elite 9000 (ZEISS, Dublin, CA). The volume data includes normal and diseases such as Age- related Macular Degeneration. A grader reviewed the images before and after artifact removal, and rated the level of enhancement as being (5) much improved, (4) improved, (3) about the same, (2) worse, or (1) much worse. Results: Figure 1 shows choroidal layer slabs with vessel shadow artifacts, the corresponding RPE retinal layer slabs, and the resulting choroidal slabs without artifacts. Our results showed 88 images were graded 4 or above. 8 remaining images were graded 3. The lower and upper 95% nonparametric confidence limit of the grading are 3 and 5 respectively. Conclusions: We present a method to reduce vessel shadow artifacts in choroidal layer slab images. Our approach is specifically useful for visualization and diagnosis of a variety of ocular diseases. Figure 1: RPE slab (left), choroidal slab image with vessel shadow artifacts (center), and choroidal slab after vessel shadow artifacts removal (right). CONTROL ID: 3194796 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0178 TITLE: Vendor neutral multimodal registration for en face OCT and fundus images FIRST AUTHOR: Reza Jafari AUTHORS/INSTITUTIONS: R. Jafari, C. Reisman, Research and Development, Topcon Healthcare Solutions, Oakland, New Jersey, UNITED STATES| Purpose: To present an automated, robust, and accurate method to align images from different modalities (en face optical coherence tomography (OCT) versus color fundus images and color fundus versus FAF/FA/red-free images) using a novel multimodal registration technique. Methods: OCT images and fundus photos of varying types are widely used in the diagnosis of eye diseases, and an automated registration routine helps clinicians to diagnose and monitor diseases from both imaging modalities. The proposed multimodal registration method is KAZE feature-based. After removing the background and enhancing contrast as a preprocessing for input images, KAZE features are detected and extracted. Inlier features are identified by calculating distances between corresponding features. The transformation matrix is then calculated to register the target and reference images. The proposed method was tested on 69 pairs of images captured by TRC-50DX and DRI OCT Triton (both Topcon Corp., Tokyo, Japan); images from other vendors were also used. The size of the OCT en face (macula or disc region, or both) and the fundus images (color, FAF, FA, red-free) are varied. The tested images were as follows: color to color, en face to color, FAF or FA to color, and red-free to color. The registration accuracy was evaluated using root mean square error (RMSE), which measured the degree of misalignment between feature points of reference images and corresponding feature points in target images. Results: By visual check, the registrations for all input image pairs were successful. Quantitatively, the mean accuracy was 1.35±0.52 pixel and the algorithm also performed well in the presence of artifacts, such as vignetting and media opacity. Conclusions: A multimodal registration based on KAZE features was proposed and implemented for OCT en face, color, FAF, and red-free images. The method was tested on different scan modes and different resolutions. The experimental results suggest the algorithm is robust and accurate. (No Image Selected) CONTROL ID: 3195533 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0175 TITLE: Method for artificially degrading the signal from Optical Coherence Tomography cube to assess variability in retinal pigment epithelial detachment volumetric measurement FIRST AUTHOR: Sophie Kubach AUTHORS/INSTITUTIONS: S. Kubach, L. De Sisternes, Carl Zeiss Meditec, Inc, Dublin, California, UNITED STATES|W. Lewis, Bayside Photonics, Inc, Yellow Springs, Ohio, UNITED STATES|G. Gregori, Bascom Palmer Eye Institute, Miami, Florida, UNITED STATES| Purpose: Measurement of retinal pigment epithelium detachment volume (RPED) is an important characteristic to be able to measure when designing the needed signal in an OCT system. In this study we assess the variability in the RPED volume in dry AMD eyes with signal-to-noise reduction by artificially degrading the OCT signal Methods: Repeated Angio 6x6mm scans from 5 dry AMD eyes imaged on the PLEX® Elite 9000 swept-source OCT (ZEISS, Dublin, CA) were artificially corrupted to simulate different reduction in signal. The method to reduce the OCT signal is the following: Data from an 8-bit OCT cube are unlogged after determining the pixels value above and below (deltadB/10) the noise level. The noise is then subtracted from the signal and the signal is scaled by a factor 10 where deltadB is the signal reduction factor. The noise is then added back to the signal and the log is taken to produce an 8- bit OCT cube. After segmentation of the Retinal pigment epithelial (RPE), RPED volume corresponding to the volume between the elevated RPE and RPE fit in the central 5 mm circle was computed on the original cubes and on the cubes with different reduction in signal. Results: Table 1 shows for each eye, the mean and standard deviation for RPED volume within a 5mm radius circle when considering the original dataset (i.e no signal reduction) and the same dataset degraded by 1.5 dB, 3 dB and 4.5 dB. The sensitivity in RPED volume with OCT signal degradation varies from eye to eye. The change in RPED volume with signal degradation is small, less than half the standard deviation for all eyes except for eye #2 where the change is statistically significant, with a volume reduction of 5 sigma at 4.5 dB from baseline. Figure 1 shows for each eye the RPED volume and error bar as a function of OCT function of signal. Eye 2 (blue plot) shows a statistical change in RPED volume. If we take a closer look at the RPE elevation map for all 5 eyes (figure 2), there is a clear difference in the RPE elevation profile, with a more pronounced RPE elevation for eye #1 and #3 and smaller elevations more spread out for eye #2. Conclusions: The described method for artificially degrading the OCT signal can be used to assess the sensitivity of the RPED volume independently from any system-to-system variations. Eye 1 Volume RPE elevati on Eye 2 Volume RPE elevation Eye 3 Volume RPE e levation Eye 4 Volume RPE elevation Eye 5 Volume RPE elevation

Signal M ean Stddev M ean Stddev M ean Stddev M ean Stddev M ean St ddev 3 3 3 3 3 3 3 reduction (mm ) (mm ) (mm ) (mm') (mm' ) (mm ) (mm ) (mm') (mm ) (mm )

OdB 0.0426 0.0008 0.0316 0.0011 0.0427 0.0107 0.0547 0.0053 0 .0004 0.0005 ·1.5dB 0.0425 0.0009 0.0277 0.0021 0.0431 0.0096 0.0556 0.0055 0 .0009 0.0015 -3dB 0.0425 0.0007 0.0273 0.0015 0.0422 0 .0095 0.0543 0.0027 0 .0009 0.0014 -4.5dB 0.0427 0.0006 0.0274 0.0004 0.0384 0.0121 0.0558 0.0057 0 .0006 0.0010

Table 1: Volume ofRPE elevation for each ~ve over a 5 mm circle centered on the retina For each the mean and 1 standard deviation from 4 repeated scans are reported

_ ..----V-ol_um_•_RP_ E..,•_••_•_••-·o_n_c_ha_n"'-.._.., _ ._th_s-'lg'rn-• l_,._du_ c•_·o_n_--1 0 07

0"6

00>

()_O t

-00Od1 ~------~------~------~B l Seit> 3dB .... . Signal reduction n dB

Figure 1: RPE volume elevation as a function ofOCT signal degradation for each 5 eyes Figure 2-a: Baseline RPE elevation map for all 5 eyes

Figure 2-b: RPE elevation map for all 5 eyes at 4.5 dB signal degradation CONTROL ID: 3195644 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0179 TITLE: Quantified fundus autofluorescence (QAF) imaging – the use of age-related standard retinas and improved methods for analysis FIRST AUTHOR: Thomas Ach AUTHORS/INSTITUTIONS: T. Ach, N. Kleefeldt, C. Pröbster, I.S. Tarau, K. Bermond, C. Reichel, Department of Ophthalmology, University Hospital Wuerzburg, GERMANY|K.R. Sloan, Department of Ophthalmology & Visual Sciences, University of Alabama at Birmingham, Birmingham, Alabama, UNITED STATES|K.R. Sloan, Department of Computer Science, University of Alabama at Birmingham, Birmingham, Alabama, UNITED STATES| Purpose: In vivo QAF uses an internal reference to compare retinal AF intensities between subjects and in follow-ups. For analysis, normally the mean QAF intensity (QAF8; PMID: 22016060) of a ring of certain width around the fovea is measured (PMID: 23860757, 26551331). The predefined area at a fixed location, however, limits precise analysis of subtle changes or lesions exceeding common QAF8 patterns. This study offers improved analysis patterns and provides standard retinas for each decade for detailed QAF analysis. Methods: 140 healthy subjects (5-77 years, clear lens) underwent multimodal imaging (color fundus, red free, infrared, AF (488,787nm), QAF (488nm) and SD-OCT using Spectralis and modified HRA2 (Heidelberg Engineering) and FF450 (Zeiss) cameras/devices. Custom FIJI plugins enabled: 1. Determination of the fovea and the edge of the optic disc in SD-OCT/infrared images. 2. Alignment and superimposition of multimodal images based on retinal vasculature. 3. Adjustment to age-related optical media density (for QAF). 4. Building of standard QAF retina maps for each decade and comparison of individual retinas to these standards. 5. To semi-automatically detect and edit retinal vasculature signals (if necessary for analysis). 6. Use of predefined grids (ETDRS; Delori pattern, modified grids) to manually draw regions of interest (free-hand tool) for detailed QAF analysis at specified regions. Results: For each decade, at least 9 subjects were multimodal imaged. All images were registered based on vasculature and then matched using the location of the fovea and the edge of the optic disc. QAF standard retina maps for each decade plot QAF intensities for each pixel at a certain distance and direction from the fovea. Modified, finer overlay grids (as commonly available) revealed that the hot spot of AF (temporal superior edge of the macula) slightly shifts with age. Free-hand tools enabled to accurately measure QAF values within sharply defined regions of interest. Conclusions: A pre-requisite for accurate QAF analysis is age-corrected standard retinas which then can be used for comparison with diseased eyes. Furthermore, modified analysis patterns enable a more precise evaluation of selected areas or disease-related lesions re QAF changes (e.g., in age-related macular degeneration), currently examined in ongoing studies. (No Image Selected) Biomechanics CONTROL ID: 3194850 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0181 TITLE: Morphological changes in Bruch’s membrane opening during intraocular pressure elevation FIRST AUTHOR: Yanhui Ma AUTHORS/INSTITUTIONS: Y. Ma, S. Kwok, K. Clayson, J. Liu, Department of Biomedical Engineering, The Ohio State University , Columbus, Ohio, UNITED STATES|K. Clayson, Biophysics Interdisciplinary Group, The Ohio State University, Columbus, Ohio, UNITED STATES|X. Pan, Department of Biomedical Informatics, The Ohio State University, Columbus, Ohio, UNITED STATES|J. Liu, Department of Ophthalmology and Visual Science, The Ohio State University, Columbus, Ohio, UNITED STATES| Purpose: To quantify the changes in the location and diameter of Bruch’s membrane opening (BMO) in response to acute intraocular pressure (IOP) elevation by using ultrasound imaging. Methods: Whole globe inflation tests were performed in 7 human donor eyes (age: 31-74 years old) by increasing IOP from 5 to 50mmHg with 0.5mmHg steps below 30mmHg and 2mmHg steps above. The IOP was held constant at each pressure level for 30 seconds. Cross-sectional images of the posterior eye centered on the optic nerve head (ONH) were acquired along the superior-inferior meridian at each pressure step using a 50MHz ultrasound probe (MS700, Vevo2100, VisualSonics). Two Bruch’s membrane termination points were manually delineated in all B-mode images (i.e., at pixel level) (Fig.1). Center and length of the line connecting these two points was tracked at each pressure step to investigate the morphological change of BMO during IOP elevation. Displacements and changes of BMO were calculated relative to the location and diameter of BMO at the baseline pressure (i.e., 5mmHg). Distributive displacement of the posterior eye was calculated by an ultrasound speckle tracking algorithm. Results: BMO moved posteriorly and the posterior displacement increased with increasing IOP (71.7±51.9µm at 25mmHg and 129.9±82.4µm at 50mmHg) (Fig.2A). Displacement of BMO was larger than the average within ONH at every IOP level (all p<0.001), and highly correlated with the displacement of ONH (R>0.95) (Fig.2B). A negative displacement gradient was observed in the ONH in the through-thickness direction (Fig.2C). The initial BMO at the baseline pressure ranges from 1224.7 to 1609.4µm (1470.2±133.7µm). Change of BMO diameter is minimal in response to IOP elevation (Fig.2D). Conclusions: We previously reported that there was minimal expansion of the scleral canal (on average about 20 mm) when IOP was raised from 5 to 30mmHg (Ma et al, IOVS, 2019). This study showed minimal change in BMO diameter. BMO moved more posteriorly than the ONH as a whole. Fig.1 Delineation of BMO terminations on ultrasound image
Fig.2 (A) Displacement of BMO and ONH (B) correlation of BMO and ONH displacement (C) color map of posterior displacement for a representative human donor eye (D) change of BMO diameter(n=7)
CONTROL ID: 3195578 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0182 TITLE: Shear strains in porcine and keratoconic human cornea FIRST AUTHOR: Keyton Clayson AUTHORS/INSTITUTIONS: K. Clayson, Y. Ma, S. Kwok, J. Liu, Biomedical Engineering, The Ohio State University, Columbus, Ohio, UNITED STATES|J. Liu, Ophthalmology & Visual Science, The Ohio State University, Columbus, Ohio, UNITED STATES| Purpose: Corneal collagen fiber slippage due to excessive shear is hypothesized as a biomechanical mechanism for keratoconus (Meek et al, IOVS 2015). This study aimed to characterize the distribution of corneal shear strains in normal porcine eyes and keratoconic human donor eyes using high-resolution ultrasound speckle tracking. Methods: Ten porcine whole globes were obtained from a local abattoir and tested within 72 hrs postmortem, and two donor globes from an 84 year old donor with bilateral keratoconus of different severity were obtained from Lions VisionGift (Portland, OR) and tested within 12 hrs of receipt (84 hrs postmortem). Porcine globes were pretreated in a 10% dextran solution, secured to a custom-built holder and immersed in 0.9% saline. Human globes were pretreated in a 5% poloxamer-188 solution overnight to return the corneas towards physiological hydration, and remained in this solution during testing. All globes were preconditioned with 20 pressure cycles from 5-30 mmHg and then subjected to inflation testing from 5-30 mmHg in either 0.5 mmHg (porcine) or 1.0 mmHg (human) steps. Radiofrequency data of B- mode images of the central cornea were acquired using a 50 MHz ultrasound probe (MS700, VisualSonics), and corneal strains were calculated using an ultrasound speckle-tracking technique (Tang and Liu, ASME 2012). Results: In porcine corneas, shear strain was minimal throughout the imaged corneal cross-section (Fig. 1) and had significantly smaller magnitude than the radial and tangential strains (-0.63%±1.54% vs. -8.59%±1.31% and 3.93%±1.79% at 30 mmHg, all p’s<0.05). In contrast, both the mild and advanced keratoconic corneas had significant strain variations through the corneal thickness, with shear strains similar in magnitude to the radial and tangential strains observed in both globes (Fig. 2). Conclusions: Strain patterns appear altered in keratoconic cornea as compared to normal corneas, with a more varied response throughout the cornea. Shear strain magnitudes appear to increase in keratoconic corneas, consistent with previous observations of altered collagen organization in the disease. Weakness in resisting shear deformation may be involved in corneal ectasia and warrants future investigation. Fig. 1. Average strain response for porcine corneas (n=10) with strain maps from a representative globe at 30 mmHg. Fig. 2. Strain maps at 30 mmHg from a bilateral keratoconic donor, with arrows indicating cone location. CONTROL ID: 3195843 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0180 TITLE: Reproducibility of Lamina Cribrosa Microstructure Measurements in Varying Intraocular and Intracranial Pressure Settings FIRST AUTHOR: Katie Lucy AUTHORS/INSTITUTIONS: K. Lucy, R.S. Rai, Y. Glidai, M. Wu, H. Ishikawa, J.S. Schuman, G. Wollstein, Department of Ophthalmology, NYU Eye Center, NYU Langone Health, New York, New York, UNITED STATES|M. Wu, Departments of Population Health and Environmental Medicine, New York University, New York, New York, UNITED STATES|B. Wang, I.A. Sigal, M. Smith, Department of Ophthalmology, UPMC Eye Center, Eye and Ear Institute, Pittsburgh, Pennsylvania, UNITED STATES| Purpose: To examine the effect of varying levels of intraocular (IOP) and intracranial pressure (ICP) on the reproducibility of lamina cribrosa (LC) microstructure measurements. Methods: Spectral-domain OCT scans of the optic nerve head (ONH) were obtained from adult healthy rhesus macaque monkeys while IOP and ICP were changed in a controlled environment. Gravity-based perfusion through a needle inserted into the anterior chamber controlled IOP (low, medium, high settings). Perfusion through the lateral ventricle controlled ICP (low, high settings). Scans were registered in 3D and LC microstructure measurements (beam thickness, pore diameter) were calculated from shared regions among scans acquired at each setting using a previously described segmentation algorithm. Microstructure measurement results were used to calculate the beam/pore ratio of each scan, and a 2-way ANOVA test compared the effect of different IOP and ICP settings on measurement reproducibility. Results: The results of 2 eyes were analyzed. For average beam thickness IOP had a significant effect on measurement reproducibility but ICP did not (p=0.005, p=0.66, respectively). For average pore diameter IOP also had a significant effect on measurement reproducibility but ICP did not (p=0.009, p=0.97, respectively). The effect of IOP and ICP on beam/pore ratio reproducibility was not significant (p=0.23, p=0.80, respectively). Results are summarized in Figure 1. Conclusions: Our study provides evidence that beam/pore ratio measurements are reproducible regardless of acquisition at different IOP and ICP settings. This parameter is less influenced by scanning angle and image quality than other measurements. This information supports direct comparison of beam/pore ratio measurements obtained in varying pressure settings. Figure 1. Effect of Intraocular Pressure (IOP) and Intracranial Pressure (ICP) on Lamina Cribrosa Measurements. 2- way ANOVA test was used for comparison; data from 2 eyes was aggregated. Varying IOP significantly affected beam thickness and pore diameter but did not affect beam/pore ratio. ICP did not have a significant effect on any of the measurements. Fluorescence CONTROL ID: 3193956 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0185 TITLE: Designing a Hyperspectral Autofluorescence (AF) Camera for Early Detection of Age-related Macular Degeneration (AMD) FIRST AUTHOR: R Theodore Theodore Smith AUTHORS/INSTITUTIONS: R.T. Smith, Ophthalmology, Icahn School of Medicine of Mount Sinai, New York, New York, UNITED STATES|N. Dey, Computer Science, New York University, New York, New York, UNITED STATES|T. Ach, Ophthalmology, University of Wurzburg, Wurzburg, GERMANY|C. Curcio, Ophthalmology, University of Alabama Birmingham, Birmingham, Alabama, UNITED STATES| Purpose: Optimize excitation wavelengths of a proposed clinical hyperspectral AF camera for early detection of AMD. Soft drusen and basal linear deposit (BLinD) are the lipid rich material of the Oil Spill on Bruch’s membrane (BrM) of early AMD. Drusen are focal and recognizable clinically. BLinD is thin, diffuse, and invisible clinically, even on high resolution optical coherence tomography (OCT), but is detectable ex vivo on hyperspectral AF imaging. Optimal spectral excitations, however, are yet unknown. Methods: 20 tissues, retinal pigment epithelium (RPE)/BrM flatmounts from AMD donors underwent hyperspectral AF imaging with 4 excitation wavelengths (436, 450, 480 and 505 nm), and the resulting image cubes were simultaneously decomposed with non-negative tensor factorization (NTF), an extension of non-negative matrix factorization. NTF results are essentially unique and robust to spectral initializations, up to rotations and scale factors, and also deliver the excitation spectra for the fluorophore sources of the recovered emissions, that is, the most efficient wavelengths among those tested for excitation of given fluorophores. 2 basic emission spectra were recovered for a clear separation: a total RPE spectrum (lipofuscin (LF)) and a subRPE spectrum (drusen and BLinD). Results: A composite emission spectrum for drusen and BlinD, the SDr spectrum, was consistently recovered with peak at 520 nm (Fig, upper left, blue spectrum.) The total LF spectrum was also consistent, peaking at about 570 nm, with secondary peaks or shoulders at 600 and 650 nm (Fig, upper left, red spectrum). SDr localized to drusen and subRPE deposits (Fig, lower left), the LF spectrum to the LF compartment in the RPE (Fig, lower right) with histopathologic sensitivity and specificity. Remarkably, the excitation spectra for both SDr and LF peaked at 450 nm in all 20 samples (Fig, upper right, blue and red spectra). Thus, 450 nm not only optimally excited SDr, it was also more efficient than the classic 480 nm for exciting lipofuscin AF. Dual excitations at ~450 and ~480 nm were the most efficient and reliable pair for detection of early AMD lesions. Conclusions: Classic 488 nm excitation, with additional 450 nm excitation. a hyperspectral AF detector, and suitable image analysis, should be capable of clinical detection and quantification of drusen and BLinD in early AMD. Emission Spectra Excitation Spectra ~ 0.4 ~0.8 Vl Vl , Component 1 ~ c c -- Component 2 Q.J Q.J I .µ 0.3 .µ 0.6 -- c c "C 0.2 "C 0.4 '', , Q.J Q.J N N ,, re 0.1 re 0.2 ....E ....E ' \."' --- 0 0 z 0 z 0 --- 500 600 700 440 460 480 500 Wavelength (nm) Wavelength (nm) Abundance 1 Abundance 2 CONTROL ID: 3195541 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0186 TITLE: Near infrared fluorescence detection and evaluation in an adaptive optics scanning laser ophthalmoscope FIRST AUTHOR: Tao Liu AUTHORS/INSTITUTIONS: T. Liu, J. Tam, National Eye Institute, National Institutes of Health, Bethesda, Maryland, UNITED STATES|S. Cornelissen, Boston Micromachines Corporation, Cambridge, Massachusetts, UNITED STATES|A. Dubra, Department of Ophthalmology, Stanford University, Palo Alto, California, UNITED STATES| Purpose: To evaluate and select a suitable photodetector for collecting near infrared autofluorescence (IRAF) and indocyanine green (ICG) fluorescent light in a custom-built adaptive optics scanning light ophthalmoscope and then characterize the performance of the system. Methods: One photomultiplier tube (PMT, Hamamatsu H7422-50) and one avalanche photodiode (APD, Excelitas C30659-900-R8AH with a custom amplifier circuit provided by Boston Micromachines) were inserted into the same fluorescence detection channel of the AOSLO and used to detect the weaker IRAF and stronger ICG signals, which share similar emission bands (805-840 nm) but with 2-4x difference in signal strength. Readout electronics were identical except for the amplifiers. Images from an ICG model eye consisting of ICG-stained paper and from a human subject were used to select the better-performing detector, which was further tested on human subjects to evaluate the impact of detector gain and confocal pinhole (PH) size. Signal-to-noise ratio (SNR) was used to evaluate detector performance on the ICG model eye. Images from subjects were normalized to a simultaneously acquired nonfluorescent channel. Results: Although the APD had higher quantum efficiency (QE) with the capability to detect higher near-infrared wavelengths, images of the ICG model eye acquired by the APD were grainier and noisier, with SNR 8x and 12x lower than PMT images for 2.5 and 7.5 Airy Disk Diameter (ADD) PHs, respectively. This was consistent with images from a human eye, where retinal pigment epithelial cells could be seen in IRAF and ICG images using the PMT but only faintly (for ICG) or not at all (for IRAF) using the APD. Thus, the PMT was selected. The standard deviation of IRAF and ICG PMT images increased monotonically with gain (favorable for distributing signal over a broader range of usable bits). However, near the upper gain control voltage limit, the images become very noisy. When increasing the detector PH from 1 to 7.5ADD, the throughput increased by about 10X compared to a theoretical 1.25X for a diffraction-limited system, likely due to the presence of residual aberrations. Conclusions: For near-infrared fluorescence imaging in the eye, superior performance was achieved using a PMT when compared to an APD. Improved images can be captured by increasing the internal gain up to (but not beyond) the upper limit and by increasing the size of the PH. (No Image Selected) CONTROL ID: 3195558 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0183 TITLE: Visualizing near-infrared autofluorescence from retinal pigment epithelial cells in AMD using multi-wavelength excitation FIRST AUTHOR: Kari Viljami Vienola AUTHORS/INSTITUTIONS: K.V. Vienola, M. Zhang, J. Sahel, E.A. Rossi, Ophthalmology, University of Pittsburgh, Pittsburgh, Pennsylvania, UNITED STATES|E.A. Rossi, Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, UNITED STATES| Purpose: To visualize determine the efficacy of multi-wavelength excitation for imaging retinal pigmented epithelial cells (RPEs) in vivo using near-infrared autofluorescence (NIRAF) in healthy controls and age-related macular degeneration (AMD) patients. Methods: NIRAF imaging of the RPE was performed in healthy volunteers and patients diagnosed with AMD using a multi-channel adaptive optics scanning laser ophthalmoscope (AOSLO). Two separate superluminescent diodes (centered at 663 nm and 795 nm) were used for NIRAF excitation and confocal reflectance imaging. The time- averaged optical power at pupil plane was 50 µW for 663 nm and 180 µW for 795 nm with a 1.5 deg. field of view. NIRAF signal was collected in an emission band from 814 to 870 nm. A 2.1 Airy disk diameter pinhole was placed in front of the NIRAF detector and an automated algorithm was used for pinhole positioning to maximize the NIRAF signal. The NIRAF images were co-registered using the corresponding reflectance channel(s) as the reference for eye motion. Results: Individual RPE cells were successfully imaged in the macula of healthy controls and AMD patients with either 663 nm excitation, 795 nm excitation, or simultaneous excitation with both. Shorter wavelength excitation alone produced a slightly noisier image; however, this may be due to different focusing of the two wavelengths due to chromatic aberration. RPE morphology was similar between the two excitation wavelengths with a darker center and hyper autofluorescent border, consistent with the known hexagonal mosaic of RPE cells. Conclusions: Each excitation wavelength alone was able to produce NIRAF emission capable of resolving individual RPE cells. Combined excitation resulted in a more robust signal. Ongoing work is aimed at determining whether this emission arises from the same structures within the RPE and how NIRAF excited with these different wavelengths may be altered in AMD. Simultaneous excitation of RPE fluorophores with a combination of wavelengths may improve imaging of individual RPE cells. (No Image Selected) CONTROL ID: 3195778 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0184 TITLE: Flavoprotein Fluorescence Changes in the Retina with Cataract Surgery. FIRST AUTHOR: Jorge Santiago Andrade Romo AUTHORS/INSTITUTIONS: J.S. Andrade Romo, M.V. Castanos Toral, D.B. Zhou, D.F. Buxton, R. Rosen, Ophthalmology, New York Eye and Ear Infirmary of Mount Sinai, New York, New York, UNITED STATES| Purpose: To evaluate the impact of cataract surgery on retinal metabolism using flavoprotein fluorescence (FPF) monitoring. Methods: Nine subjects (non-diabetics) were included in the study. Retinal metabolic analysis was performed pre and post cataract surgery using the OcuMet Beacon (Ocusciences, Ann Arbor, MI). It features a customized fundus camera with 467 nm excitation and 535 nm emission filters and a back-illuminated electron multiplying charge-coupled camera. The depth of focus enables capture of FPF values from all layers of the retina in a central 13° diameter circular region of interest (ROI) centered at the fovea (Fig). Patients were imaged immediately prior to surgery and post-operatively at an interval range of 31-103 days. During each imaging session, four scans were acquired to generate an average raw FPF value in gray scale units (gsu). Scans were also processed to compensate for the influence of cataracts or IOLs, providing adjusted FPF measurements (lens FPF and retinal FPF). Color-coded maps were then obtained for the ROI, with warmer colors indicating greater mitochondrial oxidative stress (Fig). Paired sample t-test was used to analyze differences between the pre-op and post-op FPF values. Results: Mean raw FPF, lens FPF, and retinal FPF values before surgery were 455, 426, and 29gsu, respectively; and following surgery were 186, 112, and 74gsu. All FPF values were significantly different before and after surgery (p<0.010). Conclusions: Flavoprotein fluorescence (FPF) analysis mirrors fluctuations in retinal metabolic stress induced by cataract surgery. A clearer picture of the complexities of blue light penetration through the lens will be necessary to better quantify the contribution of elevated mitochondrial stress in the increase of FPF before it can become a useful clinical tool. Figure: Before and after cataract surgery images from a single subject. A1, B1) SLO retinal image with ROI (green circle). A2) Pre-op raw FPF color map, warm hue is present (high fluorescence from cataract) (raw FPF: 758gsu). B2) Post-op raw FPF, a cooler hue is present (lower fluorescence from IOL) (raw FPF: 480gsu). A3) Pre-op retinal FPF color map, faint blue hue speckled pattern (retinal FPF: 36gsu). B3) Post-op retinal FPF color map, more uniform and intense blue (retinal FPF: 94gsu). SLO: scanning laser ophthalmoscopy, FPF: flavoprotein fluorescence, gsu: gray scale units. Basic Science CONTROL ID: 3175631 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0190 TITLE: Aryl hydrocarbon receptor deficiency causes a diabetes-related vascular pathology FIRST AUTHOR: Meei-Ling Sheu AUTHORS/INSTITUTIONS: M. Sheu, College of Life Sciences, National Chung Hsing University, Taichung, Taiwan, Institute of Biomedical Sciences, Taichung 40227, Taiwan, TAIWAN| Purpose: Diabetic retinopathy (DR) is typically characterized by retinal inflammation, vascular leakage, blood-retinal barrier breakdown and neovascularization. However, the molecular mechanisms underlying the inflammation signaling contributes to the progression of DR remain unclarified. Aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that has been implicated in inflammation and pathological vascular angiogenesis. Methods: As AhR contributes to vascular dysfunction; we speculate that AhR might play a role in microangiopathies, such as diabetic retinopathy (DR). Therefore, we investigated the impact of AhR on retinal vascular damage using AhR knock out mice (AhRKO) during development of DR and its possible mechanisms. ELISA detected Nε- carboxymethyllysine (CML) and inflammatory cytokines levels in the serum from human samples and several diabetic murine models, especially the latter also used LC/MS/MS analysis the whole eye tissue. Immunofluorescence staining identified CML expression on retinal pigment epithelium (RPE) layer and endothelial cells. Furthermore, intravitreal injection (IVI) of pharmacological inhibitors or agonists for diabetic rat model, and observed pathological features by retinal leukostaisis, optical coherence tomography (OCT) and H&E staining. Pericyte loss and acellular capillary formation were assessed in streptozotocin-induced diabetic AhRKO and wild-type (WT) mice. Expressions of immune cells were assessed by immunofluorescence in the diabetic retinas as well as in retinal endothelial cells depleted of AhR by siRNA and stimulated with CML. Results: Similar to diabetic WT retinas, non-diabetic AhRKO retinas showed a significant decrease in pericyte coverage in comparison with non-diabetic WT retinas. Hyperglycemia or diabetes condition further aggravates pericyte loss in diabetic AhRKO retinas. Acellular capillary formation was detected in the diabetic AhRKO retinas. In cultured primary retinal endothelial cells, stimulation with CML and AhRKO depletion comparably increased immune cells expression. Conclusions: Our data identify AhR as a protective factor in the retina, which controls immune cells expression. AhRKO-deficient mice are a suitable model for studying mechanisms underlying diabetic retinal vascular damage. (No Image Selected) CONTROL ID: 3194011 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0189 TITLE: Protective Effects of Huoxue-Tongluo-Lishui-Fang (HTLF) on Retinal Ischemia-Reperfusion Injury FIRST AUTHOR: Minglian Zhang AUTHORS/INSTITUTIONS: M. Zhang, M. Wang, Hebei Provincial Eye Hospital, Xingtai, CHINA| Purpose: Huoxue-Tongluo-Lishui-Fang (HTLF) is an ancient Chinese herbal decoction, which could be effective on multiple ocular conditions, including diabetic retinopathy, and retinal vascular occlusions according to TCM doctor's clinical experiences. This study was applied to investigate the protective mechanism of HTLF on retinal ischemia/reperfusion (I/R) injury and tissue edema in rabbit eyes. Methods: New Zealand white rabbits were randomly divided into Normal Control Group, I/R Model Group and HTLF Treatment Group. The I/R Model Group and HTLF Treatment Group were further divided into 6 sub-groups based on the time phases (6h, 12h, 1d, 2d, 3d and 5d) respectively. Acute retinal I/R injury was induced in rabbit eyes of I/R Model Group and HTLF Treatment Group through elevation of intraocular pressure (IOP) using anterior chamber perfusion. In HTLF Treatment Group, the HTLF was started from 5 days before I/R modeling, 2 times a day until death; purified water insdupt instead of HTLF was used in Normal Control Group and I/R Model Group. Using OCT scanning on rabbit fundus, the retinal thickness was measured pre- and 6h, 12h, 1d, 2d, 3d and 5d post-I/R modeling, respectively. Blood-retinal barrier (BRB) leakage was analyzed using the Evans blue (EB) method. The IL-6, IL-1β, TNF-α, malondialdehyde (MDA), nitric oxide (NO) and SOD were quantified by ELISA. AQP4 protein expression was detected using Western-blot analysis. Results: The retinal thickness was increased gradually 6h post-I/R modeling, and reached its maximum at 1d. EB leakage area in I/R Model Group increased significantly 24h post-I/R modeling, indicating the increased blood-retinal barrier permeability. Less retinal thickness and EB leakage area were found in HTLF Treatment Group compared with I/R Model Group. Compare with the increase of IL-6, IL-1β, TNF-α, MDA and NO, a significant and rapid decrease of SOD was found in I/R Model Group 1d post-I/R modeling. In HTLF Treatment Group, an increase of SOD accompanied by the decrease of IL-6, IL-1β, TNF-α, MDA and NO was found, which were all significantly different from the data in I/R Model Group. Conclusions: Our results indicated that HTLF has protective effects against retinal I/R injury and reduce retinal edema in rabbits. This protective mechanism of HRLF may be related with its antioxidative, anti-inflammatory and anti- leakage ability. (No Image Selected) CONTROL ID: 3194384 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0188 TITLE: Tmem30a Deficiency in the retinal endothelial cells impairs cell proliferation and angiogenesis FIRST AUTHOR: Xianjun Zhu AUTHORS/INSTITUTIONS: X. Zhu, Center for Human Molecular Genetics, Sichuan Provincial People's Hospital, Chengdu, Sichuan, CHINA| Purpose: Phosphatidylserine (PS) is PS asymmetry in the eukaryotic cell membrane is maintained by a group of proteins belonging to the P4-ATPase family, namely, PS flippases. The folding and transporting of P4-ATPases to their cellular destination requires a beta-subunit member of the TMEM30 protein family. Loss of Tmem30a has been shown to cause multiple disease conditions. However, its roles in vascular development have not been elucidated. Methods: We analyzed the role of Tmem30a in endothelial cells using primary human retinal endothelial cells (HREC). Tmem30a expression was analyzed by realtime-PCR. shRNA mediated knockdown of Tmem30a was performed. Tube formation was assessed using In vitro Matrigel tube formation assays. Endothelial cell specific knock out mouse model was generated by crossing a conditional allele of Tmem30a to an inducible PDGFbeta-Cre ER line. Retinal vascular development was assessed in wholemount retinas staining with isolectin, which labels the blood vessels. Blood vessel barrier integrity was evaluated by Ter119 staining of red blood cells. Cell proliferation was assessed by 5-ethynyl-2'-deoxyuridine (EDU)lableing. Transcritome analysis was performed by RNA-seq using Tmem30a knockdown HREC and knockout tissues. Results: Our data indicated that knockdown of TMEM30A in primary human retinal endothelial cells led to reduced tube formation. In mice, endothelial cell (EC)-specific deletion of Tmem30a led to retarded retinal vascular development with a hyperpruned vascular network as well as blunted-end, aneurysm-like tip endothelial cells (ECs) with fewer filopodia at the vascular front and reduced number of tip cells. Deletion of Tmem30a also impaired vessel barrier integrity. Mechanistically, deletion of TMEM30A caused reduced EC proliferation by inhibiting VEGF-induced signaling. Conclusions: Our data show that TMEM30A plays critical roles in retinal vascular angiogenesis, which is a fundamental process in vascular development. Our findings reveal essential roles of TMEM30A in angiogenesis, and providing a potential therapeutic target. In vitro studies of the function of TMEM30A in tube formation. Tmem30a is essential for sprouting angiogenesis and vessel barrier integrity in mouse retinas. Reduced tip ECs and i△EC filopodia in retinal vessels of Tmem30a and Tmem30a-iKO mice was shown. The number of tip cells and filopodia was counted in each genotypes and plotted in G. CONTROL ID: 3195873 SESSION ABSTRACT START TIME: 8:00 AM SESSION ABSTRACT END TIME: 8:00 AM FINAL ID: PB0187 TITLE: Characterizing spatial distribution of neuronal cell types in visual areas of mice brain imaged using Serial 2- photon tomography FIRST AUTHOR: Kannan U V AUTHORS/INSTITUTIONS: R. Palaniswamy, P. Osten, Neuroscience, Cold Spring Harbor Laboratory, Floral Park, New York, UNITED STATES|K. U V, Neuroscience, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, UNITED STATES|N. Cain, J. Harris, Allen Institute for Brain Sciences, Seattle, Washington, UNITED STATES| Purpose: Many molecularly defined neuronal types have been discovered in last few decades. We don’t have a comprehensive understanding on the distribution of these cell types in the whole brain, different functional pathways and within each region of these pathways. These series of experiments help in mapping the distribution of few of these cell types in different regions involved in the visual pathway of the mouse brain. Methods: Novel mice created from Allen Institute of Brain Study’s library of molecularly defined cell types have these different cell types fluorescently labeled. 5 male and 5 female 8-10-week-old mice are killed and perfused and embedded in an agarose block and crosslinked. Once fixed these brains are imaged using a serial two-photon tomography (STPT) microscope. The entire brains are imaged in multiple field of views of 800mx800m at 1m x 1m XY resolution. Each coronal section is imaged with 16x12 FOVs with 10% overlap and spaced 50m apart in Z. All the FOVs are stitched together to create a whole mouse brain image of size 13000x9000x280 voxels. The fluorescently labeled cells are automatically identified using a convolution neural network (CNN) across the whole brain. The CNN was trained using markups from 60 FOV randomly sampled from different regions across the entire brain. Further, the sample brains are registered to the Allen Brain Institute’s Common co-ordinate framework (CCF) space. The cell densities are computed for all the cell types in the CCF space. The cell density maps of the visual areas are extracted and clustered to identify the similarity in distribution. Results: We have characterized the cell distribution maps multiple cell types in the following areas of the visual pathway: Anterolateral visual area (VISal) Anteromedial visual area (VISam) Lateral visual area (VISl) Primary visual area (VISp) Posterolateral visual area (VISpl) posteromedial visual area (VISpm) Lateral geniculate nucleus (LGN)

For the following cell types: Parvalbumin Slc32 Gad2 Chat Ctgf-T2A Rbp4 Rorb Cux2 Nstr1 Tlx3 Emx1 Conclusions: This work has generated density maps of various cell types in regions of the brain involved in the visual system. These cell types have been clustered based on the spatial distribution of the cells. (No Image Selected) The Association for Research in Vision and Ophthalmology gratefully acknowledges our Imaging in the Eye Conference supporters:

Visit us: Booth 1037 Enabling OCT with Innovative Solutions

Spectrometers with high speed, resolution & sensitivity

Compact sample arm probes

Powerful image processing software & libraries

Custom drop-in OEM modules & subsystems

See how we are leading the Expedition Into the Eye

+1 919-544-7785 | [email protected]

See the Whole Picture with Triton™ Swept Source OCT

ARVO Booth #803

DRI OCT Triton™ Series A Multimodal Swept Source OCT

Full 360o image captured with posterior/anterior imaging capabilities of the Triton. Composite image made with 3rd party imaging software. ©2019 Topcon Medical Systems, Inc. MCA #3695 The Association for Research in Vision and Ophthalmology

1801 Rockville Pike, Suite 400 Rockville, MD 20852-5622

Tel: +1.240.221.2900 ARVO.org/Imaging