NIH Public Access Author Manuscript Ophthalmology. Author manuscript; available in PMC 2012 December 01.

Size: px
Start display at page:

Download "NIH Public Access Author Manuscript Ophthalmology. Author manuscript; available in PMC 2012 December 01."

Transcription

1 NIH Public Access Author Manuscript Published in final edited form as: Ophthalmology December ; 118(12): doi: /j.ophtha Dynamics of Human Foveal Development after Premature Birth Ramiro S. Maldonado, MD 1, Rachelle V. O Connell, BS 1, Neeru Sarin, MD 1, Sharon F. Freedman, MD 1,2, David K. Wallace, MD, MPH 1,2, C. Michael Cotten, MD 2, Katrina P. Winter, BS 1, Sandra Stinnett, DrPH 1, Stephanie J. Chiu 3, Joseph A. Izatt, PhD 3, Sina Farsiu, PhD 1,3, and Cynthia A. Toth, MD 1,3 1 Department of Ophthalmology, Duke University Eye Center, Erwin Rd, Durham, NC Box 3802, 27710, USA 2 Department of Pediatrics, Duke University School of Medicine, Durham, NC, Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, Abstract Purpose To determine the dynamic morphological development of the human fovea in-vivo utilizing portable spectral domain optical coherence tomography (SDOCT). Design Prospective, observational case series. Paticipants 31 prematurely born neonates, nine children and nine adults. Methods Sixty-two neonates were enrolled in this study. SDOCT imaging was performed after examination for retinopathy of prematurity (ROP) at the bedside in non-sedated infants ages weeks post-menstrual-age PMA (PMA=gestational age in weeks + chronological age) and at outpatient follow-up ophthalmic examinations. Thirty-one neonates met eligibility criteria. Nine children and nine adults without ocular pathology served as control groups. Semi-automatic retinal layer segmentation was performed. Central foveal thickness (CFT), foveal to parafoveal (FP) ratio (CFT divided by thickness 1000 μm from the foveal center), and 3D thickness maps were analyzed. Main Outcomes Measures In-vivo determination of foveal morphology, layer segmentation, analysis of sub-cellular changes, spatio-temporal layer shifting. Results In contrast to the adult fovea, we observed several signs of immaturity in the neonates: a shallow foveal pit, persistence of inner retinal layers (IRL), and a thin photoreceptor layer (PRL) that was thinnest at the foveal center. Three-dimensional mapping showed displacement of retinal layers out of the foveal center as the fovea matured and the progressive formation of the inner/ outer segment band in the opposite direction. The FP-IRL ratios decreased as IRL migrated prior to term and minimally after that, while FP-PRL ratios increased as PRL subcellular elements 2011 American Academy of Ophthalmology, Inc. Published by Elsevier Inc. All rights reserved. Address for reprints: Cynthia A. Toth, MD: Cynthia.toth@duke.edu Duke University Eye Center 2351 Erwin Rd. DARSI Lab. Suite 103 Durham, NC, Conflict of Interest: Proprietary or commercial disclosure may be found after the references. Financial disclosure(s): Proprietary or commercial disclosure may be found after the references. Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

2 Maldonado et al. Page 2 formed closer to term and into childhood. A surprising finding was the presence of cystoid macular edema in 58% of premature neonates which appeared to affect inner foveal maturation. Conclusions This study provides the first view into development of living cellular layers of the human retina and of subcellular specialization at the fovea in premature infant eyes using portable spectral domain optical coherence tomography. Our work establishes a framework of the timeline of human foveal development, allowing us to identify unexpected retinal abnormalities that may provide new keys to disease activity, and provide a method for mapping of foveal structures from infancy to adulthood that may be integral in future studies of vision and visual cortex development. The human fovea is responsible for high spatial resolution and for central and color vision. 1 It plays an important role in human visual cortex development, regulating calcarine fissure symmetry 2 and cortical maps. 3 Abnormal development of the fovea has been related to a decrease in gray matter volume at the occipital cortex 4 and might be related to disruptions in visual pathways at the level of the optic chiasm. 5 The neurosensory retina may be divided into the photoreceptor layer (outer retina) and the inner processing and transmitting layers (inner retina) (Fig. 1). Morphologically, the adult fovea is marked by a pit that is dense with photoreceptors, and devoid of overlying inner retinal layers (IRL), and vasculature. This facilitates light transmission to the photoreceptors without interference. 6 The fovea is immature at birth. 7,8 Histologic studies, originating with those of Mann in 1964, 9 have reported: 1) persistence of IRL at the foveal center and absence of the normal foveal curvature in the fetal eye and 2) changes in the width and length of photoreceptor substructures over time. The process of foveal maturation has been described as a centrifugal migration of the IRL and a centripetal migration of the cone cells. 7,8,9 Numerous factors may influence foveal development: prematurity, 10 development of vasculature, 11 foveal tissue elasticity, intraocular pressure, retinal stretching. 12 Some of these may act synergistically. Also, eye growth throughout the neonatal period 13,14 may stretch malleable foveal tissue to form the foveal pit. 15 Studies of vascular endothelial growth factor (VEGF) expressed in the area of the future fovea suggest that hypoxia is present prior to the thinning of the retinal layers, although this does not explain the process of foveal formation. 16 Investigations of foveal pit development have been based on postmortem fixed tissue from non-human primates 17,18 or from rare human specimens, 7,8,9 with advantages of sub-micron resolution imaging and immunocytochemical labeling. However, serial sectioning is timeconsuming, specimens are obtained at one timepoint, the same tissue cannot be processed for a flat mount and serial sectioning, retinal detachment is a common fixation artifact and more severe deformation of structures occurs in fixed sections from the youngest human eyes. Measurements rely on rates of relative tissue shrinkage during processing and may not represent the configuration in-vivo. All of these factors limit study of the three-dimensional growth of the human macula, especially photoreceptors. A reliance on postmortem data precludes inputting the anatomic stage of foveal development for an individual when assessing relationships between ophthalmic maturity, retinal signaling and posterior visual pathways. Data on microanatomy of the living human fovea in premature infants in a neonatal intensive care unit (NICU) are limited to color photographs, descriptions, drawings, 19,20 and intravenous fluorescein angiography. 21 Because these lack cross-sectional information regarding cellular development, human studies could not address the interrelationship between changing foveal structures or between these and later visual function, which is of particular importance in the premature infant eye.

3 Maldonado et al. Page 3 By introducing the study of high-resolution ocular images from the NICU, we propose to define, in premature infants in-vivo, a baseline frame of reference for development of the first component of the human visual system, the retina. We also begin to define variations and pathologies involved in this development. METHODS In this study, we have utilized spectral domain optical coherence tomography (SDOCT), a rapid, noninvasive, and noncontact imaging technique which uses a broadband infrared wavelength light (840 nm) to detect backscattered light from the retina using interferometric techniques to provide cross-sectional images of the retina (Fig. 1). This high-resolution, invivo imaging modality has improved understanding of adult retinal diseases and is currently an important tool in the diagnosis and management of retinal pathology. 22,23 Imaging with OCT is based on the intrinsic reflectance of a tissue or the interface between tissue layers. It is particularly useful for retinal evaluation due to contrast between alternating layers of lower reflective cell nuclei and higher reflective axons, dendrites and melanosomes. 24 Cross-sectional images can be readily oriented within a three-dimensional stack of captured scans to define unique retinal microanatomy in-vivo. 25 We developed methods to image young children and infants during examination under anesthesia 26,27 using a hand-held portable SDOCT system (Bioptigen Inc., Research Triangle Park, North Carolina). We subsequently analyzed the optical properties of the premature infant eye to determine methods to allow SDOCT imaging of these infants without sedation. 28 For this study, we examine retinas from very young premature infants to examine the dynamic shift in substructures during a critical time in the early development of retinal signaling and visual processing and compare the organization of these substructures to that of children and adults. All adults and the parents or guardians of all infants and children consented to the participation in this Institutional Review Board approved observational study of SDOCT imaging. Because this research was performed on subjects in a NICU, research imaging could not be performed at predetermined timepoints. Instead, imaging was performed only when the NICU staff approved the status of the infant for research imaging and at times when the infant eyes were already dilated for the conventional examination. Follow-up ended if infants were transferred to another hospital or discharged, unless there was a follow-up visit on site with study imaging in clinic. SDOCT imaging was performed after ophthalmoscopic retinopathy of prematurity (ROP) screening at the bedside following the Maldonado et. al. methodology for optimizing SDOCT in neonates. 28 and using a portable hand-held SDOCT unit (Bioptigen Inc., Research Triangle Park, North Carolina). Sixty-two premature infants were enrolled as part of a prospective study comparing SDOCT findings to those from conventional ophthalmoscopic examination by a pediatric ophthalmologist for ROP. Only subjects with SDOCT imaging sessions including the fovea before 42 weeks post-menstrual-age PMA (PMA = gestational age in weeks + chronological age) and an ophthalmic ROP exam up to 42 weeks PMA or equivalent information (parents of two subjects who had outside ophthalmic follow-up were contacted by phone to identify whether laser treatment or ROP progression occurred after being discharged) were included. Nine subjects were excluded: two were discharged before imaging occurred, two were examined for systemic conditions with ocular pathology (neonatal hemochromatosis and cytomegalovirus infection), three did not have ROP screening up to 42 weeks PMA to determine maximum level of disease and two did not have imaging prior to 42 weeks. From the remaining 53 neonates, 21 progressed to advanced ROP (excluded if clinical diagnosis identified any stage 3 ROP, type 1 ROP or if the infant received laser treatment) and were excluded from the study. One additional subject who met all other inclusion criteria was excluded because no study images contained the fovea. Thus there were 31 premature

4 Maldonado et al. Page 4 infants included in this study. For information on comparative foveal characteristics with increasing age, a single imaging session was obtained in nine full-term born children (4 infants ages 1, 2, 6 and 9 months old; and 5 children ages 2, 3, 6, 6 and 15 years old) and 9 adults. Cystoid macular edema (CME) was a surprising finding in 18 of the 31 premature infants. The CME distorted retinal layers and affected retinal thickness interfering in analysis of retinal development. Thus we divided our analysis into a primary analysis of retinal development in infants without any CME (Group 1, n=13) and separately analyzed the findings of the infants with CME (Group 2, n=18). This division into Groups 1 and 2 was by subject and not by eye, as CME was bilateral in all subjects. SDOCT images were converted to a DICOM format and qualitatively graded by four experienced graders in OSIRIX medical imaging software (OSIRIX Foundation, Geneva, Switzerland). Graders were masked to other clinical data. The best three SDOCT volumes containing the fovea were evaluated for presence of pathology, and the grader selected the scan with best resolution and best foveal centration for segmentation and quantitative analysis. The foveal scan was segmented semi-automatically using a custom program, DOCTRAP (Duke OCT Retinal Analysis Program) v2.1-sf, based in MATLAB (Mathworks, Natick, MA, USA). The foveal pit was selected manually. MATLAB was used to compute thickness values from segmentation at each A-scan. Axial pixel size in retina was 3.26 um/pixel, as reported by Bioptigen Inc. Lateral pixel size was determined by dividing the scan length on retina by the number of A-scans. Scan length on retina for the neonatal eye was calculated based on the axial length for age. 13,14 Scan length ranged from mm, and number of A-scans ranged from 512 to 1260 to maintain a consistent A-scan density. To validate our assignment of retinal layers in premature infants, we followed the layers through their evolution from infancy through childhood and compared them to published light micrographs of premature infant and children s eyes at corresponding ages. 7-9 For this, we reviewed the literature searching Medline for the terms fovea, foveal development, ocular development, visual development, foveal pit formation, from 1950 to the 2010 and based assignment of layers on correlation between OCT retinal layers in adult primate and human histology. 23,24,25 SDOCT graders evaluated qualitatively the scans for the presence or absence of retinal pathology, and retinal layers at the foveal center. To determine the reproducibility of this grading an inter-reader agreement analysis was performed. A random number generator was used to select an eye from the first imaging session for each subject that was used for primary analysis as reported of Table 1 and 2. Central foveal thickness was defined as the thickness of the entire retina from the inner aspect of the inner limiting membrane (ILM) to the inner aspect of the retinal pigment epithelium (RPE) at the foveal center. The inner retinal layers (IRL) included all retinal tissue from the inner aspect of the ILM to the outer border of the inner nuclear layer (INL). The outer retinal layers (Table 1) extended from the inner aspect of the outer plexiform layer (OPL) to the inner border of the RPE. Retinal layers from top to bottom (inner to outer) are labeled in Figure 1. The PRL in this manuscript extended from the outer aspect of OPL to the inner border of RPE. Three-dimensional thickness maps were created using a custom, automated graph theorybased segmentation software (DOCTRAP Pediatric v2.1) based on Chiu et al. 29 Automated segmentation was successful at identifying the inner border of the ILM, inner border of the IS/OS, and the inner border of the RPE. Then, semi-automated segmentation was used to correct automated segmentation errors and to delineate the inner border of the OPL.

5 Maldonado et al. Page 5 A ratio of the thickness at the foveal center versus thickness 1000 μm peripheral to the fovea was computed for IRL and PRL. Ratios were compared for all study groups using measurement for one eye on the first imaging session using the Wilcoxon rank sum test (Table 2). ImageJ (U. S. National Institutes of Health, Bethesda) software was used to sum SDOCT b- scans to improve visualization of images presented for this publication (Fig. 1, Fig. 2a, and 2b top row). No post-processing was needed to segment or grade images. RESULTS For statistical analysis and to avoid intra-eye variation, only one imaging session per subject per age group was used in the statistical analysis. We determined statistical differences in the median foveal thickness measurements of premature neonates versus adults using a Wilcoxon rank-sum test and the statistical differences between all age groups using a Kruskal-Wallis test of difference among medians, where p<0.05 was considered statistically significant. Retinal imaging was possible from the earliest eye examination at 31 weeks PMA until time of discharge from the nursery in all 31 premature infants in this study (age range at imaging weeks PMA). Half were female, 61% African-American, 32% White, and <6% Hispanic-Latino. The median gestational age at birth was 26 weeks PMA (range 24 to 29 weeks). Median birth weight was 825 g ( ). A total of 106 imaging sessions were performed in the 31 neonates and there were 1 to 8 imaging sessions per infant between 31 through 42 weeks (median = 3, with 53% imaged three or more times). Five of the infants remained in the nursery or returned for follow up imaging as late as 65 weeks PMA. The older comparison groups had a single imaging session and consisted of nine children ages 2 weeks to 15 years (33% female; 56% African-American, 33% White, 11% Hispanic-Latino) and 9 adults (ages years; 62% females; 46% African-American, 38% White, 16% Hispanic-Latino). Inter-grader agreement for detection of foveal features was 92% among pairs of SDOCT certified graders. Highest agreement was found on the presence of INL and IS/OS at the fovea which was 98% and 95%, respectively, and the presence of many other common retinal pathologies, including cystoid macular edema (91%). Lowest agreement was found on the presence of GCL at the fovea (62%) and qualitative scoring of presence of pre-cystic thickening of the INL (59%). In review of SDOCT scans, we found cystoid macular edema (CME) distorting the fovea in 18 of the 31 subjects (58%) at one or more imaging sessions. CME was bilateral in the majority of cases except in only 4 imaging sessions of 4 subjects. The CME was an unexpected finding in 58% of subjects and varied in severity from a single rounded hyporeflective lesion (Fig. 3a) to more extensive edema that thickened inner layers and disturbed the foveal contour (Fig 3b) or produced an upward bulge at the foveal center (13 of 18 eyes, Fig 3c). CME was detected as early as 31 weeks PMA and was more prevalent after 37 weeks PMA (Fig. 3d). Because this was not a long term study, we did not have follow up to identify whether or when there was CME resolution in all subjects. At 42 weeks PMA, we observed persisting CME in both eyes of two subjects who also had CME documented at some point between 32 and 42 weeks. In both eyes of 6 other subjects, we observed CME resolution at 40 to 65 weeks; these infants had previously documented CME on SDOCT imaging at some timepoint between 35 and 43 weeks PMA. Because the presence of CME distorted retinal morphology and grossly changed thickness measurements, we grouped premature infants into Group 1 (n=13, no CME at any visit) and

6 Maldonado et al. Page 6 Group 2 (n=18, CME at any visit) based on the SDOCT images. Although the infants with edema (Group 2) had both, lower gestational age and weight at birth (Table 1), these differences were not statistically significant. Half of the subjects in Group 1 and 72% of subjects in Group 2 reached stage 2 ROP (by study design, all were Type 2 ROP and none reached any stage 3 disease or required laser). The non-edematous premature infant retina versus the adult The foveal center was readily identified in both eyes of all 13 neonates in Group 1 as the site of deepest central depression in the stack of serial SDOCT scans. Retinal layers and central foveal thickness at the central foveal scan in all premature infant eyes were grossly different from those of children and adults, and these differences were most pronounced at the earliest ages (Fig. 1). When compared to the adult eye, the premature eye had a visibly shallower foveal depression, presence of many IRL at the foveal center, thinner retinal layers overall, and attenuation of PRL with absence of photoreceptor sub-layers relative to the adult (Fig. 1) The median central foveal thickness in Group 1 was significantly less than the thickness in adults, and it increased progressively from infancy through adulthood (Tables 1 and 2). Three-dimensional thickness mapping created from volumetric stacks of OCT scans in these subjects demonstrated the pronounced regional variation in thickness in total retina, IRL, and photoreceptor layer (PRL), and how both regions and layers develop distinctly over time (Fig. 2a and 2b). Persistence of inner retinal layers in non-edematous premature infant eyes Median IRL thickness at the foveal center in neonates (Group 1) was significantly greater than IRL thickness in adults (Table 1,2 and Fig. 1). Horizontal neuronal elements such as nerve fiber layer and plexiform layers stood out clearly as hyperreflective layers in contrast to the hyporeflective nuclei, even at this early age. The most immature foveas (31-33 weeks) were characterized by presence of the ganglion cell layer (GCL), inner plexiform layer (IPL), and inner nuclear layer (INL) as distinct measurable layers at the foveal center (Fig. 1 a-c). While the IPL and INL persisted in all foveas from the neonatal group, these condensed into a single thin hyperreflective band in children and adults (Fig. 1 b,d). Inner retinal cell migration The thickness and number of inner retinal layers at the foveal center decreased over time as the premature infant eye matured and the foveal pit deepened. In four neonates from Group 1 with multiple imaging sessions, thickness of the IRL at the foveal center progressively decreased from 31 to 45 weeks PMA (Fig. 4a, available at Median central foveal thickness and median IRL thickness were significantly greater in Group 2 than in Group 1 (Table 2). Seven of seven Group 2 subjects that had an abnormal INL thickening without visible cystoid structures developed CME on later sessions. As central foveal IRL thickness decreased, parafoveal IRL thickness increased, resulting in a decrease in the foveal to parafoveal IRL ratio (FP-IRL thickness ratio) with increasing age (Fig. 5a, available at The median FP-IRL thickness ratio in Group 1 neonates was significantly greater than that in adults (Tables 1 and 2). 3D maps highlight regional ontogenetic changes in the retina over time. Both the FP-IRL ratios and the 3D maps of the IRL from subjects at 31 weeks PMA through 23 years of age are consistent with centrifugal cell migration of the IRL to form the foveal pit. These included data from the same eye of one premature infant from 34 to 43 weeks PMA (Figs. 2a and 2b). With increasing age there is a progressive increase in height of a parafoveal annulus of the retina in contrast to central and more peripheral retina, and the IRL is the major contributor to this increase (Fig. 2a: TL and IL) in contrast to the PRL which

7 Maldonado et al. Page 7 increases to a lesser height and over a longer period (Fig. 2a). The majority of the IRL migration occurred between weeks with minimal refinement following. The IRL were also thicker closer to the optic nerve, consistent with nerve fiber layer (NFL) distribution. 30 Although the majority of Group 1 followed a similar pattern of maturation, there was individual variation. For example, although the GCL was absent on most examinations from weeks, there were 5 different subjects where GCL was present at either 33, 35 or 37 weeks PMA. IPL and INL were present in 12 of 13 Group 1. Edema appeared to be associated with a possible delay in maturation of inner retinal layers, but did not appear to impact photoreceptor development. At weeks PMA, the GCL persisted at the fovea in 15 of 18 (83%) of Group 2 where IPL and INL were present in all foveas from weeks. FP- IRL thickness ratio was significantly higher in Group 2 than in Group 1 (Figure 5c, available at and Table 2) and, this was consistent with and without cystoid structures present which might suggest thicker and likely less mature IRL at the foveal center. Photoreceptor development and migration The in-vivo PRL was notably thin across the infant retina with the thinnest portion located at the foveal center with a median of 29 microns (range 13-62) (Fig. 1). Median PRL thickness at the foveal center in Group 1 was significantly thinner than the in the adult group (Tables 1 and 2). PRL at the foveal center increased in thickness progressively from infancy to adulthood (Table 1, Fig. 5b, available at and PRL thickness was significantly different across age groups (Group 1, Infants, Children, Adults; p< 0.001, Kruskal-Wallis test). Median PRL thickness in Group 2 was similar to PRL thickness in Group 1 (Tables 1 and 2). Repeat imaging of four neonates without macular edema revealed progressive increase in PRL thickness at the foveal center with increasing age at almost every interval (Fig. 4c, available at Nevertheless, PRL had a more rapid increase in height after 38 weeks (Figs. 2b and 4c, available at and unlike the IRL, PRL axial growth was pronounced after birth in all regions and particularly in the cone-dense fovea (Fig. 2b). The PRL was thinnest at the foveal center in Group 1 which was the opposite of the central bulging PRL configuration in the adult (Fig. 1). This resulted in a significantly lower median FP-PRL thickness ratio for Group 1 versus the adults (Tables 1 and 2, Fig. 5d, available at FP-PRL ratio was not different between groups despite a greater range of ratios for Group 2. Thus although there may be a delay in centrifugal migration of the inner retina in premature infant eyes with macular edema, we did not identify a definite delay in maturation of photoreceptor morphology when assessed at this early period of their life. Development of photoreceptor sub-elements Photoreceptor subcellular structures were absent at the foveal center on SDOCT imaging in the premature group. These structures include external limiting membrane (ELM), inner segment to outer segment (IS/OS) junction and photoreceptor outer segments (POS) (Figure 1). The time course of development of these structures appeared to vary across infants. For instance, both, the ELM, and the IS/OS junction were absent in all eyes from 31 to 42 weeks PMA, and in both eyes of a term 2 month-old infant (~46 weeks PMA) but the IS/OS junction was visible at the foveal center in three of five premature infants between weeks PMA and in one full-term infant at 45 weeks demonstrating variability in layer development between subjects. Also, another full-term infant, did not show IS/OS at 46 weeks PMA, but did show this band at 4 months of age (~54 weeks PMA).

8 Maldonado et al. Page 8 The IS/OS band, first appeared as a poorly defined reflective band barely elevated from the RPE outside the fovea at 33 weeks PMA. The ring of the IS/OS and POS formation slowly increased in thickness as the outer segments appeared both peripheral and central to this zone, reaching the foveal center by 43 weeks (Fig. 2a). The distance from the foveal center to the edge of visible IS/OS band was comparable at any point when measured nasal versus temporal, and superior versus inferior; this was corroborated by the circular zone of OS absence on the 3D maps (Fig. 2a). From 37 to 42 weeks, the foveal center to IS/OS border mean distance was approximately 1380 microns (range ) for Group 1, while in Group 2 this mean was 1654 (range ) microns which was not significantly different from Group 1. DISCUSSION The main RPE reflex, a prominent well-defined hyperreflective band, was present in all subjects from 31 weeks PMA through adulthood (Fig. 1). A second subtle hyperreflective RPE band between the IS/OS junction and the main RPE reflex is believed to be the interface between photoreceptor outer segments and retinal pigment epithelium microvilli (OS/RPE). 31 (Fig. 1). The OS/RPE band appeared to differentiate at the apical side of the RPE layer late in childhood, consistent with growth of apical microvilli of the RPE or a change in the interface with photoreceptor outer segments. It was not visible in any infants or children under age 10, and was visible with variable contrast only in mid-teens and adults. Our results show that human foveal development continues after premature birth. This is the first report documenting the development of the human fovea in-vivo (PubMed Mesh search terms: optical coherence tomography, foveal development, prematurity, retinopathy of prematurity) without postmortem artifacts, illustrating the distinct differences in development between inner retinal layers and the photoreceptors, and showing the evolution of cellular structures in three dimensions over time. In contrast to previous studies, of nonhuman primate samples 18 and postmortem human foveal specimens 7,8,9, we could monitor retinal maturation in the same individual over time and could document the variation in the timing of foveal development across individuals (Figs 2a, 2b and 4, available at aaojournal.org). Semi-automated segmentation of retinal layers was essential for three-dimensional mapping of the developing retina (Fig. 2). Utilizing this in-vivo mapping and quantifying the changing relationship between the fovea and peripheral retina (Fig. 5, available at aaojournal.org), we displayed the spatio-temporal shift in the cellular elements and structures during early development. The segmentation allowed quantitative characterization of foveal maturation from prematurity to adulthood with a reproducible imaging method (92% agreement between certified SDOCT graders). Neuronal cell generation and differentiation are known to be complete at the fovea prior 31 weeks PMA as demonstrated in human retinal whole-mounts by mitosis cessation, 32 thus cell migration and maturation are the primary events in the period of our imaging. A dynamic evaluation of cellular migration in-vivo was achieved by measuring the foveal to parafoveal ratio for the IRL and PRL thicknesses. IRL cell migration was in the opposite direction of PRL cell migration and elongation, with a relative time lag in the PRL growth (Fig. 5b, available at Timing of development of the PRL in premature infants was not always linked to the timing of development of IRL; therefore, some infants demonstrated highly developed IRL with delayed maturation of the PRL and others demonstrated the opposite. Imaging these stages of individual foveal microanatomy is likely to be useful for in-vivo studies of visual development, since the progress of retinal

9 Maldonado et al. Page 9 development in each infant may vary and may affect signaling and upstream visual pathways. 33,34 Although the change in morphology of SDOCT retinal layers with age and in FP-IRL with age in the non-edematous eyes was generally consistent with timepoints of cellular redistribution reported on histologic studies, 7,8,9,35 in-vivo analysis allowed a better sampling of the events of foveal development over time and across numerous individuals. However, none of the published postmortem specimens of infants less than one year of age were from infants who had survived for more than two days after birth, and this may explain why none of those studies describe macular edema during this period of development. Although we had longitudinal information, one weakness in our study was the inability to complete individual follow-up in all subjects because of frequent transfer to another hospital. All retinal layers reported in histologic studies were observed on SDOCT except for the transient acellular layer of Chievitz. 36 GCL and INL thickness reported on Table 1 appeared comparable to the 2-3 cells thickness of GCL and INL reported by Hendrickson et al at weeks PMA. 7 The short photoreceptor cells and absence of formed outer segments reported on histologic studies at weeks, matches well with our finding of absent IS/OS band at weeks PMA and gradual increase in height of both the photoreceptor nuclear layer and the outer segments during prematurity. In at least one case in-vivo foveal development varied from published histology: in the postmortem specimen, IRL were present at the foveal center and a there was a shallow pit in a 5 day old term infant, while in our study, many premature infants at weeks already had a well-formed foveal pit with pronounced thinning or absence of IRL. Although histology studies 7,8,9 report that foveal development appeared complete by age 15 months, we found evidence of progressive development after that timepoint. The additional band that is thought to be the interface between outer segments and RPE was not visible in any young children while it was visible in a child at 15 years of age and in adults. To identify the exact timeline of development of this reflectivity, we will need more extensive study in young children and young adults. We theorize that the timing of development of this subtle interface is visible with in-vivo SDOCT imaging but not with light microscopy of sectioned tissue due to common photoreceptor-rpe disruption or separation with fixation of ocular tissue. An unexpected finding in premature infants was the presence of CME (58% of premature subjects) at one or more timepoints from 32 to 43 weeks PMA. While visible on SDOCT images, CME was not detected on the clinical examination in our NICU. Macular thickening, edema or schisis have not been found previously in studies of neonatal ocular development, or in clinical trials of retinopathy of prematurity, except in a few cases of ROP with tractional retinal detachment extending at or near the macula 37,38,39 and a report of cystoid maculopathy in three infants from a review of ocular histopathology of the infant for a period of 15 years. 40 In the latter report, cystoid structures involved outer retinal layers in an infant who died at 11 days of life, the inner nuclear layer in one stillborn infant and splitting of NFL in another. Both stillborn infants had serious central nervous system abnormalities. The lack of previous reports of CME may be due to the previous lack of technology capable of capturing this information in earlier studies, and to the lack of human eye histopathology from subjects who lived several weeks after birth in a NICU. Although the CME bore some similarities to retinoschisis from traction which has been reported in ROP, 30,41 the cystoid spaces were located only at the fovea and in the INL, with a rare cystoid structure in the PRL (Fig 3c) and were not associated with any traction. Although infants in this study were very premature, they did not develop any advanced ROP (see

10 Maldonado et al. Page 10 methods). We have imaged similar CME in the maculas of a term infant with liver failure and ascites, and documented resolution of the cystoid structures after liver transplant. 42 Macular edema in premature infant eyes might reflect variability in remodeling of foveal architecture during development; however, we propose that this might relate to elevated VEGF levels in the eye analagous to adult CME that is responsive to anti-vegf therapy. Although no macular edema was found in foveal development histology studies, 7,8,9 increased VEGF mrna expression had been found in the early developing fovea, 16 and elevated intraocular VEGF levels have been identified in eyes undergoing surgery for advanced ROP. 43,44 More importantly, VEGF elevation was found in the second phase of ROP progression from weeks PMA. 45 Thus it should not be surprising to find macular edema in premature infant eyes with areas of peripheral avascular retina, even in without advanced ROP. The high frequency of the CME among premature infants with nonadvanced ROP points to the importance of understanding the cause of the edema, its predictive value, and whether this could impact visual development or acuity later in life. Inner retinal thickening in premature infants with CME might be the first evidence of lifelong thickening of inner retinal layers at the fovea as reported in older children and adults with a history of ROP. 10,46,47 However, studies of children with a history of premature birth have shown normal results on extensive visual function and MRI testing 48 or even advanced chromatic contrast sensitivity when compared to term birth subjects. 49 Experts have disagreed on the presence of and extent of any difference in visual pathways after premature birth. 50 Many studies were performed on surviving older populations to obtain imaging or visual function testing 10,36,37,47. Considerable discussion has centered on isolating the contributions of visual experience and of ophthalmic disorders distinct from neuromotor or other impairments on adult visual function Retinal signaling delivered to the cortex may affect development of visual pathway and cortical mapping, 3 and SDOCT imaging of the fovea would likely provide useful anatomic information on this actively changing site of sensory input and initial visual processing. In conclusion, migration, redistribution and growth of subcellular structures occur in premature infants ex-utero. The development of inner versus outer retinal layers at the fovea may progress independently in some eyes. This dynamic process of foveal maturation and specialization can be studied in-vivo in premature infants with SDOCT imaging in nonsedated infants. Many premature infants have CME, an unanticipated finding that could be an ocular biomarker linked to additional systemic or ocular disease activities but which may have little impact on foveal development. An unanswered question is whether the morphology on SDOCT could be a useful predictor of future visual acuity. In order to link structure with function, future longer term studies of macular imaging of premature infants would be useful in conjunction age-appropriate testing of visual function. Supplementary Material Refer to Web version on PubMed Central for supplementary material. Acknowledgments Michelle McCall provided critical feedback and management for this study. Financial Support: This research was made possible by the following grants: Angelica and Euan Baird; The Hartwell Foundation; Grant Number 1UL1 RR from the National Center for Research Resources (NCRR), a component of the National Institutes of Health (NIH), and NIH Roadmap for Medical Research. It s contents are solely the responsibility of the authors and do not necessarily represent the official view of NCRR or NIH.

11 Maldonado et al. Page 11 References 1. Rossi EA, Roorda A. The relationship between visual resolution and cone spacing in the human fovea. Nat Neurosci. 2010; 13: [PubMed: ] 2. Neveu MM, Von dem Hagen E, Morland AB, Jeffery G. The fovea regulates symmetrical development of the visual cortex. J Comp Neurol. 2008; 506: [PubMed: ] 3. Baseler HA, Brewer AA, Sharpe LT, et al. Reorganization of human cortical maps caused by inherited photoreceptor abnormalities. Nat Neurosci. 2002; 5: [PubMed: ] 4. Free SL, Mitchell TN, Williamson KA, et al. Quantitative MR image analysis in subjects with defects in the PAX6 gene. Neuroimage. 2003; 20: [PubMed: ] 5. Jeffery G. The albino retina: an abnormality that provides insight into normal retinal development. Trends Neurosci. 1997; 20: [PubMed: ] 6. O Brien, KM. Development of the foveal specialization. In: Tombran-Tink, J.; Barnstable, CJ., editors. Visual Transduction and Non-Visual Light Perception. Humana Press; Totowa, NJ: p Hendrickson AE, Yuodelis C. The morphological development of the human fovea. Ophthalmology. 1984; 91: [PubMed: ] 8. Yuodelis C, Hendrickson AE. A qualitative and quantitative analysis of the human fovea during development. Vision Res. 1986; 26: [PubMed: ] 9. Mann, I. The Development of the Human Eye. 3rd ed. Grune and Stratton; New York: p Ecsedy M, Szamosi A, Karko C, et al. A comparison of macular structure imaged by optical coherence tomography in preterm and full-term children. Invest Ophthalmol Vis Sci. 2007; 48: [PubMed: ] 11. Kozulin P, Natoli R, O Brien KM, et al. Differential expression of anti-angiogenic factors and guidance genes in the developing macula. Mol Vis [serial online]. 2009; 15: Available at: Springer AD. New role for the primate fovea: a retinal excavation determines photoreceptor deployment and shape. Vis Neurosci. 1999; 16: [PubMed: ] 13. Cook A, White S, Batterbury M, Clark D. Ocular growth and refractive error development in premature infants with or without retinopathy of prematurity. Invest Ophthalmol Vis Sci. 2008; 49: [PubMed: ] 14. Gordon RA, Donzis PB. Refractive development of the human eye. Arch Ophthalmol. 1985; 103: [PubMed: ] 15. Springer AD, Hendrickson AE. Development of the primate area of high acuity. 1. Use of finite element analysis models to identify mechanical variables affecting pit formation. Vis Neurosci. 2004; 21: [PubMed: ] 16. Young TL, Anthony DC, Pierce E, et al. Histopathology and vascular endothelial growth factor in untreated and diode laser-treated retinopathy of prematurity. J AAPOS. 1997; 1: [PubMed: ] 17. Provis JM, Sandercoe T, Hendrickson AE. Astrocytes and blood vessels define the foveal rim during primate retinal development. Invest Ophthalmol Vis Sci. 2000; 41: [PubMed: ] 18. Hendrickson, AE. The morphologic development of human and monkey retina. In: Albert, DM.; Jakobiec, FA., editors. Principles and Practice of Ophthalmology: Basic Sciences. Saunders; Philadelphia, PA: p Isenberg SJ. Macular development in the premature infant. Am J Ophthalmol. 1986; 101: [PubMed: ] 20. Abramov I, Gordon J, Hendrickson A, et al. The retina of the newborn human infant. Science. 1982; 217: [PubMed: ] 21. Lepore D, Molle F, Pagliara MM, et al. Atlas of fluorescein angiographic findings in eyes undergoing laser for retinopathy of prematurity. Ophthalmology. 2011; 118: [PubMed: ]

12 Maldonado et al. Page Drexler W, Morgner U, Ghanta RK, et al. Ultrahigh-resolution ophthalmic optical coherence tomography. Nat Med. 2001; 7: [PubMed: ] 23. Voo I, Mavrofrides EC, Puliafito CA. Clinical applications of optical coherence tomography for the diagnosis and management of macular diseases. Ophthalmol Clin North Am. 2004; 17: [PubMed: ] 24. Toth CA, Narayan DG, Boppart SA, et al. A comparison of retinal morphology viewed by optical coherence tomography and by light microscopy. Arch Ophthalmol. 1997; 115: [PubMed: ] 25. Anger EM, Unterhuber A, Hermann B, et al. Ultrahigh resolution optical coherence tomography of the monkey fovea: identification of retinal sublayers by correlation with semithin histology sections. Exp Eye Res. 2004; 78: [PubMed: ] 26. Scott AW, Farsiu S, Enyedi LB, et al. Imaging the infant retina with a hand-held spectral-domain optical coherence tomography device. Am J Ophthalmol. 2009; 147: [PubMed: ] 27. Chavala SH, Farsiu S, Maldonado R, et al. Insights into advanced retinopathy of prematurity using handheld spectral domain optical coherence tomography imaging. Ophthalmology. 2009; 116: [PubMed: ] 28. Maldonado RS, Izatt JA, Sarin N, et al. Optimizing hand-held spectral domain optical coherence tomography imaging for neonates, infants, and children. Invest Ophthalmol Vis Sci. 2010; 51: [PubMed: ] 29. Chiu S, Li XT, Nicholas P, et al. Automatic segmentation of seven retinal layers in SDOCT images congruent with expert manual segmentation. Opt Express [serial online]. 2010; 18: Available at: Ishikawa H, Stein DM, Wollstein G, et al. Macular segmentation with optical coherence tomography. Invest Ophthalmol Vis Sci. 2005; 46: [PubMed: ] 31. Zinn, KM.; Benjamin-Henkind, JV. Anatomy of the human retinal pigment epithelium. In: Zinn, KM.; Marmor, MF., editors. The Retinal Pigment Epithelium. Harvard University Press; Cambridge, MA: p Provis JM, van Driel D, Billson FA, Russell P. Development of the human retina: patterns of cell distribution and redistribution in the ganglion cell layer. J Comp Neurol. 1985; 233: [PubMed: ] 33. Colonnese MT, Kaminska A, Minlebaev M, et al. A conserved switch in sensory processing prepares developing neocortex for vision. Neuron. 2010; 67: [PubMed: ] 34. Feller MB. Visual system plasticity begins in the retina. Neuron. 2003; 39:3 4. [PubMed: ] 35. Hendrickson AE. Primate foveal development: a microcosm of current questions in neurobiology. Invest Ophthalmol Vis Sci. 1994; 35: [PubMed: ] 36. Smelser GK, Ozanics V, Rayborn M, Sagun D. The fine structure of the retinal transient layer of Chievitz. Invest Ophthalmol. Jul : [PubMed: ] 37. Joshi MM, Trese MT, Capone A Jr. Optical coherence tomography findings in stage 4A retinopathy of prematurity: a theory for visual variability. Ophthalmology. 2006; 113: [PubMed: ] 38. Patel CK. Optical coherence tomography in the management of acute retinopathy of prematurity. Am J Ophthalmol. 2006; 141: [PubMed: ] 39. Muni RH, Kohly RP, Sohn EH, Lee TC. Hand-held spectral domain optical coherence tomography finding in shaken-baby syndrome. Retina. 2010; 30(suppl):S [PubMed: ] 40. Trese MT, Foos RY. Infantile cystoid maculopathy. Br J Ophthalmol. 1980; 64: [PubMed: ] 41. Yu J, Ni Y, Keane PA, et al. Foveomacular schisis in juvenile X-linked retinoschisis: an optical coherence tomography study. Am J Ophthalmol. 2010; 149: [PubMed: ] 42. Maldonado RS, Freedman SF, Cotten CM, et al. Reversible retinal edema in an infant with neonatal hemochromatosis and liver failure. J AAPOS. 2011; 15:91 3. [PubMed: ] 43. Sato T, Kusaka S, Shimojo H, Fujikado T. Simultaneous analyses of vitreous levels of 27 cytokines in eyes with retinopathy of prematurity. Ophthalmology. 2009; 116: [PubMed: ]

13 Maldonado et al. Page Sonmez K, Drenser KA, Capone A Jr, Trese MT. Vitreous levels of stromal cell-derived factor 1 and vascular endothelial growth factor in patients with retinopathy of prematurity. Ophthalmology. 2008; 115: [PubMed: ] 45. Chen J, Smith LE. Retinopathy of prematurity. Angiogenesis. 2007; 10: [PubMed: ] 46. Hammer DX, Iftimia NV, Ferguson RD, et al. Foveal fine structure in retinopathy of prematurity: an adaptive optics Fourier domain optical coherence tomography study. Invest Ophthalmol Vis Sci. 2008; 49: [PubMed: ] 47. Recchia FM, Recchia CC. Foveal dysplasia evident by optical coherence tomography in patients with a history of retinopathy of prematurity. Retina. 2007; 27: [PubMed: ] 48. O Reilly M, Vollmer B, Vargha-Khadem F, et al. Ophthalmological, cognitive, electrophysiological and MRI assessment of visual processing in preterm children without major neuromotor impairment. Dev Sci. 2010; 13: [PubMed: ] 49. Bosworth RG, Dobkins KR. Chromatic and luminance contrast sensitivity in fullterm and preterm infants. J Vis [serial online]. 2009; 14:9 15. Available at: 9/13/15.long. 50. Ricci D, Cesarini L, Romeo DM, et al. Visual function at 35 and 40 weeks postmenstrual age in low-risk preterm infants [report online]. Pediatrics. 2008; 122:e Available at: pediatrics.aappublications.org/cgi/content/full/122/6 /e1193. [PubMed: ] 51. Hooks BM, Chen C. Vision triggers an experience-dependent sensitive period at the retinogeniculate synapse. J Neurosci. 2008; 28: [PubMed: ]

14 Maldonado et al. Page 14 Figure 1. Comparison of the immature versus mature retina imaged with SDOCT Summed SDOCT cross-sectional images of the retina of a 31 week-old post-menstrual-age (PMA) neonate (born at 27 weeks PMA, birth weight 1205 grams, retinopathy of prematurity zone II, Stage 2) (A,C) and a 23 year old adult born at term (B,D). Scans (C) and (D) are magnified images of A and B to reveal retinal layers in better detail. From inner (top) to outer (bottom) the layers are: nerve fiber layer (NFL), ganglion cell layer (GCL), inner plexiform layer (IPL), inner nuclear layer (INL), outer plexiform layer (OPL), outer nuclear layer (ONL), external limiting membrane (ELM), inner segment/outer segment junction (IS/OS), photoreceptor outer segments and retinal pigment epithelium microvilli (OS/RPE), retinal pigment epithelium (RPE). Note that the thick inner retinal layers (IRL) of the neonate (blue upper vertical line, layers listed in blue) correspond to thin condensed corresponding layers in the adult and that the thin outer retinal layers of the neonate (orange lower vertical line) became thicker layers in the adult eye (listed in orange, note ELM, IS/ OS and OS/RPE were not present in the premature fovea).

15 Maldonado et al. Page 15

16 Maldonado et al. Page 16 Figure 2. Map of regional changes in human foveal development by age from 31 weeks post menstrual age (PMA) until adulthood 3D maps of human foveas from (A) 31 to 43 weeks PMA and (B) after term birth. The 3D maps are constructed from vertical scans through the macula for 31 and 34 weeks PMA and from horizontal scans for the other timepoints. The central foveal SDOCT scan from each set is displayed in the top row. SDOCT scans demonstrate a progressive deepening of the foveal pit before 40 weeks PMA and subsequent photoreceptor layer (PRL) thickening in (B). TR (total retina) thickens gradually in all regions predominantly in the parafoveal area. IRL (inner retinal layer) demonstrates a centrifugal displacement of layers by a central pit size expansion and a parafoveal ring thickening. In contrast, the PRL grew centripetally from a thin layer at 31 weeks with the thinnest area around foveal center (magenta asterisk) to the fully developed PRL in adulthood with the thickest point at the center. Interestingly PRL growth occurs largely after term birth (B). Growth of the outer segment layer (OS) proceeds from complete absence at 31 weeks PMA (peripheral non-blue area represents flipping artifact at the edge of the scan) through adulthood in a centripetal pattern. Note that OS reaches the foveal center at 43 weeks PMA and continues with significant growth after term birth and well into childhood. The 31 week PMA subject was born at 27 weeks PMA, birth weight 1205 grams, and was retinopathy of prematurity-rop zone II, Stage 2. The scans from weeks correspond to an infant born at gestational age 27 weeks PMA and birth weight 1205 grams, ROP was zone II stage 2 for all timepoints,

17 Maldonado et al. Page 17 Figure 3. Cystoid macular edema (CME), frequently found in the inner nuclear layer of premature infants. Foveal center is noted by asterisk in (A) (processed summed scan),(b) and (C) We detected a range of CME in premature infant eyes, from an infrequent single cystoid lesion in the central fovea (A), to mild CME with round to moderately elongated cystoid spaces across the macula with some loss of foveal depression and elevation of photoreceptor layer (PRL) (B), to more severe CME creating a bulging fovea with cystoid structures even in the PRL (C). Postmenstrual age, retinopathy of prematurity-rop zone and stage corresponding to the date of the imaging session are noted on each scan. In (D), we summarized the number of eyes with CME (gray area) by week in 46 eyes from all 23 neonates

A spectral-domain OCT study of formerly premature children. Prat Itharat MD May 30, 2008 Vanderbilt Eye Institute Preceptor: Dr.

A spectral-domain OCT study of formerly premature children. Prat Itharat MD May 30, 2008 Vanderbilt Eye Institute Preceptor: Dr. A spectral-domain OCT study of formerly premature children. Prat Itharat MD May 30, 2008 Vanderbilt Eye Institute Preceptor: Dr. Recchia Background: Optical coherence tomography (OCT) OCT analogous to

More information

IN NICU OCT UTILIZES A CONCEPT KNOWN AS INTERFEROMETRY APPLICATIONS FOR OCT THE PRIMARY USE IN THE EYE - RETINA

IN NICU OCT UTILIZES A CONCEPT KNOWN AS INTERFEROMETRY APPLICATIONS FOR OCT THE PRIMARY USE IN THE EYE - RETINA 2016 25 YEARS OF OPTICAL COHERENCE TOMOGRAPHY OPTICAL COHERENCE TOMOGRAPHY IN NICU Marcin Stopa, MD, PhD, FEBO Department of Ophthalmology, Chair of Ophthalmology and Optometry. Poznan University of Medical

More information

In Vivo Foveal Development Using Optical Coherence Tomography

In Vivo Foveal Development Using Optical Coherence Tomography Retina In Vivo Foveal Development Using Optical Coherence Tomography Helena Lee, Ravi Purohit, Aarti Patel, Eleni Papageorgiou, Viral Sheth, Gail Maconachie, Anastasia Pilat, Rebecca J. McLean, Frank A.

More information

Cirrus TM HD-OCT. Details defi ne your decisions

Cirrus TM HD-OCT. Details defi ne your decisions Cirrus TM HD-OCT Details defi ne your decisions 2 With high-defi nition OCT Carl Zeiss Meditec takes you beyond standard spectral domain Built on 10 years experience at the vanguard of innovation, Carl

More information

Cirrus TM HD-OCT. Details define your decisions

Cirrus TM HD-OCT. Details define your decisions Cirrus TM HD-OCT Details define your decisions 2 With high-definition OCT Carl Zeiss Meditec takes you beyond standard spectral domain Built on 10 years experience at the vanguard of innovation, Carl Zeiss

More information

ROP and Imaging. Deborah M Costakos MD, MS August 12, 2016

ROP and Imaging. Deborah M Costakos MD, MS August 12, 2016 ROP and Imaging Deborah M Costakos MD, MS August 12, 2016 Acknowledgements Adam Dubis, Ph.D. Joseph Carroll, Ph.D. Ryan Vogel, MD Clinton Warren, MD C Devika Subramaniam, M.D. Fouad Zakla, M.D. Alana Trotter,

More information

Advances in OCT Murray Fingeret, OD

Advances in OCT Murray Fingeret, OD Disclosures Advances in OCT Murray Fingeret, OD Consultant Alcon, Allergan, Bausch & Lomb, Carl Zeiss Meditec, Diopsys, Heidelberg Engineering, Reichert, Topcon Currently Approved OCT Devices OCT Devices

More information

Review Article Retinal Imaging of Infants on Spectral Domain Optical Coherence Tomography

Review Article Retinal Imaging of Infants on Spectral Domain Optical Coherence Tomography BioMed Research International Volume 15, Article ID 7824, 12 pages http://dx.doi.org/10.1155/15/7824 Review Article Retinal Imaging of Infants on Spectral Domain Optical Coherence Tomography Anand Vinekar,

More information

OCT Image Analysis System for Grading and Diagnosis of Retinal Diseases and its Integration in i-hospital

OCT Image Analysis System for Grading and Diagnosis of Retinal Diseases and its Integration in i-hospital Progress Report for1 st Quarter, May-July 2017 OCT Image Analysis System for Grading and Diagnosis of Retinal Diseases and its Integration in i-hospital Milestone 1: Designing Annotation tool extraction

More information

NIH Public Access Author Manuscript Retin Cases Brief Rep. Author manuscript; available in PMC 2012 January 1.

NIH Public Access Author Manuscript Retin Cases Brief Rep. Author manuscript; available in PMC 2012 January 1. NIH Public Access Author Manuscript Published in final edited form as: Retin Cases Brief Rep. 2011 ; 5(1): 46 48. doi:10.1097/icb.0b013e3181cafc49. Intact Retinal Tissue and Retinal Pigment Epithelium

More information

Ganglion cell analysis by optical coherence tomography (OCT) Jonathan A. Micieli, MD Valérie Biousse, MD

Ganglion cell analysis by optical coherence tomography (OCT) Jonathan A. Micieli, MD Valérie Biousse, MD Ganglion cell analysis by optical coherence tomography (OCT) Jonathan A. Micieli, MD Valérie Biousse, MD Figure 1. Normal OCT of the macula (cross section through the line indicated on the fundus photo)

More information

Optical coherence tomography angiography of foveal hypoplasia

Optical coherence tomography angiography of foveal hypoplasia Optical coherence tomography angiography of foveal hypoplasia Kaivon Pakzad-Vaezi, MD 1, Pearse Keane, MD 1,2, João Nobre Cardoso, MD 1, Catherine Egan, MD 1, Adnan Tufail, MD 1. Moorfields Eye Hospital

More information

CLINICAL SCIENCES. Rajeev H. Muni, MD, FRCSC; Radha P. Kohly, MD, FRCSC, PhD; Alexander C. Charonis, MD; Thomas C. Lee, MD

CLINICAL SCIENCES. Rajeev H. Muni, MD, FRCSC; Radha P. Kohly, MD, FRCSC, PhD; Alexander C. Charonis, MD; Thomas C. Lee, MD CLINICL SCIENCES Retinoschisis Detected With Handheld Spectral-Domain Optical Coherence Tomography in Neonates With dvanced Retinopathy of Prematurity Rajeev H. Muni, MD, FRCSC; Radha P. Kohly, MD, FRCSC,

More information

Optical Coherence Tomography (OCT) in Uveitis Piergiorgio Neri, BMedSc, MD, PhD Head Ocular Immunology Unit

Optical Coherence Tomography (OCT) in Uveitis Piergiorgio Neri, BMedSc, MD, PhD Head Ocular Immunology Unit The Eye Clinic Polytechnic University of Marche Head: Prof Alfonso Giovannini November, 1991 Optical Coherence Tomography (OCT) in Uveitis Piergiorgio Neri, BMedSc, MD, PhD Head Ocular Immunology Unit

More information

Optical Coherence Tomography: Pearls for the Anterior Segment Surgeon Basic Science Michael Stewart, M.D.

Optical Coherence Tomography: Pearls for the Anterior Segment Surgeon Basic Science Michael Stewart, M.D. Optical Coherence Tomography: Pearls for the Anterior Segment Surgeon Basic Science Michael Stewart, M.D. Disclosure OCT Optical Coherence Tomography No relevant financial relationships I will refer to

More information

Optical Coherence Tomography in Diabetic Retinopathy. Mrs Samantha Mann Consultant Ophthalmologist Clinical Lead of SEL-DESP

Optical Coherence Tomography in Diabetic Retinopathy. Mrs Samantha Mann Consultant Ophthalmologist Clinical Lead of SEL-DESP Optical Coherence Tomography in Diabetic Retinopathy Mrs Samantha Mann Consultant Ophthalmologist Clinical Lead of SEL-DESP Content OCT imaging Retinal layers OCT features in Diabetes Some NON DR features

More information

Insights into Advanced Retinopathy of Prematurity Using Handheld Spectral Domain Optical Coherence Tomography Imaging

Insights into Advanced Retinopathy of Prematurity Using Handheld Spectral Domain Optical Coherence Tomography Imaging Insights into Advanced Retinopathy of Prematurity Using Handheld Spectral Domain Optical Coherence Tomography Imaging Sai H. Chavala, MD, Sina Farsiu, PhD, Ramiro Maldonado, MD, David K. Wallace, MD, Sharon

More information

OCT Angiography in Primary Eye Care

OCT Angiography in Primary Eye Care OCT Angiography in Primary Eye Care An Image Interpretation Primer Julie Rodman, OD, MS, FAAO and Nadia Waheed, MD, MPH Table of Contents Diabetic Retinopathy 3-6 Choroidal Neovascularization 7-9 Central

More information

Measuring of the fovea and foveola using line scans and 3D Macular scans obtained with spectral domain optical coherent tomography.

Measuring of the fovea and foveola using line scans and 3D Macular scans obtained with spectral domain optical coherent tomography. Measuring of the fovea and foveola using line scans and 3D Macular scans obtained with spectral domain optical coherent tomography. Vakhrameeva O.A. 1, Moiseenko G.A. 1, Maltsev D.S. 2, Sukhinin M.V. 2,

More information

The Quick Guide to OCT Mastery 50 Real Cases with Expert Analysis

The Quick Guide to OCT Mastery 50 Real Cases with Expert Analysis OPTICAL COHERENCE TOMOGRAPHY The Quick Guide to OCT Mastery 50 Real Cases with Expert Analysis VOL 1 Sanjay Sharma, MD, FRCS, MSc (Epid), MBA Ophthalmologist, Epidemiologist Queen s University, Canada

More information

Ultrahigh Speed Imaging of the Rat Retina Using Ultrahigh Resolution Spectral/Fourier Domain OCT

Ultrahigh Speed Imaging of the Rat Retina Using Ultrahigh Resolution Spectral/Fourier Domain OCT Ultrahigh Speed Imaging of the Rat Retina Using Ultrahigh Resolution Spectral/Fourier Domain OCT The MIT Faculty has made this article openly available. Please share how this access benefits you. Your

More information

Moving forward with a different perspective

Moving forward with a different perspective Moving forward with a different perspective The Leader In Vision Diagnostics Offers A New Perspective Marco has served the eyecare community by offering exceptional lane products and automated high tech

More information

Pearls, Pitfalls and Advances in Neuro-Ophthalmology

Pearls, Pitfalls and Advances in Neuro-Ophthalmology Pearls, Pitfalls and Advances in Neuro-Ophthalmology Nancy J. Newman, MD Emory University Atlanta, GA Consultant for Gensight Biologics, Santhera Data Safety Monitoring Board for Quark AION Study Medical-legal

More information

Diagnosis in AMD. Managing your AMD Patients

Diagnosis in AMD. Managing your AMD Patients Managing your AMD Patients Robert W. Dunphy, O.D., F.A.A.O. Diagnosis in AMD Have suspicion Identify relative risk Conduct surveillance Biometry Utilize technology to facilitate detection of change / stability

More information

What You Should Know About Acute Macular Neuroretinopathy

What You Should Know About Acute Macular Neuroretinopathy What You Should Know About Acute Macular Neuroretinopathy David J. Browning MD, PhD Chong Lee BS Acute macular neuroretinopathy is a condition characterized by the sudden, painless onset of paracentral

More information

HHS Public Access Author manuscript Ophthalmic Surg Lasers Imaging Retina. Author manuscript; available in PMC 2016 January 14.

HHS Public Access Author manuscript Ophthalmic Surg Lasers Imaging Retina. Author manuscript; available in PMC 2016 January 14. High-Speed Ultrahigh-Resolution OCT of Bruch s Membrane in Membranoproliferative Glomerulonephritis Type 2 Mehreen Adhi, MD, Sarah P. Read, MD, PhD, Jonathan J. Liu, PhD, James G. Fujimoto, PhD, and Jay

More information

Il contributo dell'angio-oct: valutazione integrata della componente nervosa e vascolare della malattia glaucomatosa

Il contributo dell'angio-oct: valutazione integrata della componente nervosa e vascolare della malattia glaucomatosa SIMPOSIO G.O.A.L. - LE NUOVE FRONTIERE DIAGNOSTICHE E LE LINEE DI INDIRIZZO AMBULATORIALI DEL GLAUCOMA Coordinatore e moderatore: D. Mazzacane Presidente: L. Rossetti Il contributo dell'angio-oct: valutazione

More information

Longitudinal Validation Study: Streptozotocin-Induced Diabetes as a Model of Diabetic Retinopathy in Brown Norway Rats

Longitudinal Validation Study: Streptozotocin-Induced Diabetes as a Model of Diabetic Retinopathy in Brown Norway Rats Longitudinal Validation Study: Streptozotocin-Induced Diabetes as a Model of Diabetic Retinopathy in Brown Norway Rats Robin Dean, Robert Sukhu, Leslie Nemeth, Qin Zhang, Isaac Hakim, Ali Ebramhimnejad,

More information

New Concepts in Glaucoma Ben Gaddie, OD Moderator Murray Fingeret, OD Louis Pasquale, MD

New Concepts in Glaucoma Ben Gaddie, OD Moderator Murray Fingeret, OD Louis Pasquale, MD New Concepts in Glaucoma Ben Gaddie, OD Moderator Murray Fingeret, OD Louis Pasquale, MD New Concepts in Glaucoma Optical Coherence Tomography: Is it necessary and needed to diagnose and monitor glaucoma?

More information

Optical Coherence Tomograpic Features in Idiopathic Retinitis, Vasculitis, Aneurysms and Neuroretinitis (IRVAN)

Optical Coherence Tomograpic Features in Idiopathic Retinitis, Vasculitis, Aneurysms and Neuroretinitis (IRVAN) Columbia International Publishing Journal of Ophthalmic Research (2014) Research Article Optical Coherence Tomograpic Features in Idiopathic Retinitis, Vasculitis, Aneurysms and Neuroretinitis (IRVAN)

More information

INFANTS WITH birth weights less

INFANTS WITH birth weights less CLINICAL SCIENCES of Retinopathy of Prematurity Michael X. Repka, MD; Earl A. Palmer, MD; Betty Tung, MS; for the Cryotherapy for Retinopathy of Prematurity Cooperative Group Objective: To report the timing

More information

Method for comparing visual field defects to local RNFL and RGC damage seen on frequency domain OCT in patients with glaucoma.

Method for comparing visual field defects to local RNFL and RGC damage seen on frequency domain OCT in patients with glaucoma. Method for comparing visual field defects to local RNFL and RGC damage seen on frequency domain OCT in patients with glaucoma. Donald C. Hood 1,2,* and Ali S. Raza 1 1 Department of Psychology, Columbia

More information

Optical coherence tomography of the vitreoretinal interface in macular hole formation

Optical coherence tomography of the vitreoretinal interface in macular hole formation 1092 St Thomas s Hospital, London V Tanner D S Chauhan T L Jackson T H Williamson Correspondence to: Mr V Tanner, Royal Berkshire Hospital, London Road, Reading RG1 5AN, UK tannerone@aol.com Accepted for

More information

Analysis of Peripapillary Atrophy Using Spectral Domain Optical Coherence Tomography

Analysis of Peripapillary Atrophy Using Spectral Domain Optical Coherence Tomography Analysis of Peripapillary Atrophy Using Spectral Domain Optical Coherence Tomography The MIT Faculty has made this article openly available. Please share how this access benefits you. Your story matters.

More information

OCT Angiography The Next Frontier

OCT Angiography The Next Frontier Choroid Retina avascular 5/13/2017 OCT Angiography The Next Frontier Pierce Kenworthy OD, FAAO June 9, 2017 OCT Angiography (OCTA) 2016 Non-invasive, motion contrast imaging Represents erythrocyte movement

More information

Swept-Source OCT Angiography: SS OCT Angio TM

Swept-Source OCT Angiography: SS OCT Angio TM Swept-Source OCT Angiography: SS OCT Angio TM Not available in all countries, please check with your distributor. 2015.09 Swept-Source OCT Angiography: SS OCT Angio TM Introduction Optical coherence tomography

More information

PRIMUS 200 from ZEISS The essential OCT

PRIMUS 200 from ZEISS The essential OCT PRIMUS 200 from ZEISS The essential OCT Seeing beyond the surface. ZEISS PRIMUS 200 // INNOVATION MADE BY ZEISS Clear Visualization. Advanced Technology. Reliability. Essential elements of your first OCT.

More information

OCT Interpretation in Retinal Disease

OCT Interpretation in Retinal Disease OCT Interpretation in Retinal Disease Jay M. Haynie, OD, FAAO Financial Disclosure I have received honoraria or am on the advisory board for the following companies: Carl Zeiss Meditec Advanced Ocular

More information

Parallel pathways in the retina

Parallel pathways in the retina Retinal origins of parallel pathways in the primate visual system Wednesday, September 23, 2015 Sherry, 2002 1 Parallel pathways in the retina Several different images of the outside world are sent simultaneously

More information

Ganglion cell complex scan in the early prediction of glaucoma

Ganglion cell complex scan in the early prediction of glaucoma Original article in the early prediction of glaucoma Ganekal S Nayana Super Specialty Eye Hospital and Research Center, Davangere, Karnataka, India Abstract Objective: To compare the macular ganglion cell

More information

THE EYE: RETINA AND GLOBE

THE EYE: RETINA AND GLOBE Neuroanatomy Suzanne Stensaas February 24, 2011, 10:00-12:00 p.m. Reading: Waxman Ch. 15. Your histology and gross anatomy books should be useful. Reading: Histology of the Eye from any histology book

More information

Study of clinical significance of optical coherence tomography in diagnosis & management of diabetic macular edema

Study of clinical significance of optical coherence tomography in diagnosis & management of diabetic macular edema Original Research Article Study of clinical significance of optical coherence tomography in diagnosis & management of diabetic macular edema Neha Kantilal Desai 1,*, Somesh Vedprakash Aggarwal 2, Sonali

More information

ZEISS AngioPlex OCT Angiography. Clinical Case Reports

ZEISS AngioPlex OCT Angiography. Clinical Case Reports Clinical Case Reports Proliferative Diabetic Retinopathy (PDR) Case Report 969 PROLIFERATIVE DIABETIC RETINOPATHY 1 1-year-old diabetic female presents for follow-up of proliferative diabetic retinopathy

More information

Automated Detection of Vascular Abnormalities in Diabetic Retinopathy using Morphological Entropic Thresholding with Preprocessing Median Fitter

Automated Detection of Vascular Abnormalities in Diabetic Retinopathy using Morphological Entropic Thresholding with Preprocessing Median Fitter IJSTE - International Journal of Science Technology & Engineering Volume 1 Issue 3 September 2014 ISSN(online) : 2349-784X Automated Detection of Vascular Abnormalities in Diabetic Retinopathy using Morphological

More information

Macular Ganglion Cell Complex Measurement Using Spectral Domain Optical Coherence Tomography in Glaucoma

Macular Ganglion Cell Complex Measurement Using Spectral Domain Optical Coherence Tomography in Glaucoma Med. J. Cairo Univ., Vol. 83, No. 2, September: 67-72, 2015 www.medicaljournalofcairouniversity.net Macular Ganglion Cell Complex Measurement Using Spectral Domain Optical Coherence Tomography in Glaucoma

More information

Flore De Bats, 1 Benjamin Wolff, 2,3 Martine Mauget-Faÿsse, 2 Claire Scemama, 2 and Laurent Kodjikian Introduction

Flore De Bats, 1 Benjamin Wolff, 2,3 Martine Mauget-Faÿsse, 2 Claire Scemama, 2 and Laurent Kodjikian Introduction Case Reports in Medicine Volume 2013, Article ID 260237, 7 pages http://dx.doi.org/10.1155/2013/260237 Case Report B-Scan and En-Face Spectral-Domain Optical Coherence Tomography Imaging for the Diagnosis

More information

Envisu TM C-Class SD-OCT System

Envisu TM C-Class SD-OCT System Bioptigen SDOIS is intended to acquire, process, display and save depth-resolved images of Ocular tissue microstructure using Spectral Domain Optical Coherence Tomography (SDOCT). The SDOIS is indicated

More information

Classification of ROP

Classification of ROP Classification of ROP Thomas Lee 2 Keywords Retinopathy of prematurity (ROP) Threshold ROP Plus disease Neovascularization Avascular retina Retinopathy of prematurity (ROP) is an iatrogenic disease. Prior

More information

OPTIC DISC PIT Pathogenesis and Management OPTIC DISC PIT

OPTIC DISC PIT Pathogenesis and Management OPTIC DISC PIT OPTIC DISC PIT Pathogenesis and Management Abdel-Latif Siam Ain Shams University Cairo Egypt OPTIC DISC PIT Congenital pit is an atypical coloboma usually located on the temporal edge of the disc, associated

More information

Why Is Imaging Critical in My Uveitis Practice?

Why Is Imaging Critical in My Uveitis Practice? Why Is Imaging Critical in My Uveitis Practice? Dilraj S. Grewal, MD Developed in collaboration Imaging Is the Backbone of Uveitis Workup and Monitoring Treatment Response FP FAF B- scan Multimodal Imaging

More information

Noninvasive cross-sectional imaging of the retina

Noninvasive cross-sectional imaging of the retina IDENTIFICATION OF FLUID ON OPTICAL COHERENCE TOMOGRAPHY BY TREATING OPHTHALMOLOGISTS VERSUS A READING CENTER IN THE COMPARISON OF AGE-RELATED MACULAR DEGENERATION TREATMENTS TRIALS CYNTHIA A. TOTH, MD,*

More information

8/6/17. Disclosures Aerie Pharmaceuticals Alcon BioTissue Diopsys Optovue Shire

8/6/17. Disclosures Aerie Pharmaceuticals Alcon BioTissue Diopsys Optovue Shire Nathan Lighthizer, O.D., F.A.A.O. Associate Professor Assistant Dean for Clinical Care Director of Continuing Education Chief of Specialty Care Clinics Oklahoma College of Optometry Tahlequah, OK lighthiz@nsuok.edu

More information

Yasser R. Serag, MD Tamer Wasfi, MD El- Saied El-Dessoukey, MD Magdi S. Moussa, MD Anselm Kampik, MD

Yasser R. Serag, MD Tamer Wasfi, MD El- Saied El-Dessoukey, MD Magdi S. Moussa, MD Anselm Kampik, MD Microperimetric Evaluation of Brilliant Blue G- assisted Internal Limiting Membrane Peeling By Yasser R. Serag, MD Tamer Wasfi, MD El- Saied El-Dessoukey, MD Magdi S. Moussa, MD Anselm Kampik, MD The internal

More information

Infantile cystoid maculopathy

Infantile cystoid maculopathy British Journal of Ophthalmology, 1980, 64, 206-210 Infantile cystoid maculopathy MICHAEL T. TRESE AND ROBERT Y. FOOS From the Department of Ophthalmology and the Department of Pathology, Jules Stein Eye

More information

PRIMUS 200 from ZEISS The essential OCT

PRIMUS 200 from ZEISS The essential OCT EN 00_00I The contents of the brochure may differ from the current status of approval of the product in your country. Please contact your regional representative for more information. Subject to change

More information

Optical Coherence Tomography of the Retina. New Technology - New Insights

Optical Coherence Tomography of the Retina. New Technology - New Insights Bahrain Medical Bulletin, Vol. 27, No. 3, September 2005 Optical Coherence Tomography of the Retina. New Technology - New Insights Mohinder Singh, FRCS,FRCOphth* Optical imaging is an emerging new technology,

More information

Neuroanatomy, Text and Atlas (J. H. Martin), 3 rd Edition Chapter 7, The Visual System, pp ,

Neuroanatomy, Text and Atlas (J. H. Martin), 3 rd Edition Chapter 7, The Visual System, pp , Normal CNS, Special Senses, Head and Neck TOPIC: FACULTY: LECTURE: READING: RETINA and CENTRAL VISUAL PATHWAYS P. Hitchcock, Ph.D. Department Cell and Developmental Biology Kellogg Eye Center Friday, 20

More information

Clinical Study Macular Development in Aggressive Posterior Retinopathy of Prematurity

Clinical Study Macular Development in Aggressive Posterior Retinopathy of Prematurity BioMed Research International Volume 2015, Article ID 808639, 5 pages http://dx.doi.org/10.1155/2015/808639 Clinical Study Macular Development in Aggressive Posterior Retinopathy of Prematurity Hemang

More information

When optical coherence tomography (OCT)

When optical coherence tomography (OCT) Macular Imaging: SD-OCT in nterior Segment Surgical Practice Many pathologic processes of the macula can be visualized or quantified only with this modality. y Steven G. Safran, MD When optical coherence

More information

Clinical Trial Endpoints for Macular Diseases

Clinical Trial Endpoints for Macular Diseases Clinical Trial Endpoints for Macular Diseases Developed in collaboration Learning Objective Upon completion, participants should be able to: Summarize types of biomarkers of progression and treatment response

More information

Reports. Macular Thickness as a Potential Biomarker of Mild Alzheimer s Disease

Reports. Macular Thickness as a Potential Biomarker of Mild Alzheimer s Disease Reports Macular Thickness as a Potential Biomarker of Mild Alzheimer s Disease Although several postmortem findings in the retina of patients with Alzheimer s disease (AD) are available, 1 new biomarkers

More information

Reproducibility of Choroidal Thickness Measurements Across Three Spectral Domain Optical Coherence Tomography Systems

Reproducibility of Choroidal Thickness Measurements Across Three Spectral Domain Optical Coherence Tomography Systems Reproducibility of Choroidal Thickness Measurements Across Three Spectral Domain Optical Coherence Tomography Systems The MIT Faculty has made this article openly available. Please share how this access

More information

History/principles of the OCT What does the normal retinal OCT look like Vitreal disorders Retinal/RPE disorders Choroidal disorders

History/principles of the OCT What does the normal retinal OCT look like Vitreal disorders Retinal/RPE disorders Choroidal disorders Nathan Lighthizer, O.D., F.A.A.O. Assistant Professor Assistant Dean for Clinical Care Director of Continuing Education Chief of Specialty Care Clinics Chief of Electrodiagnostics Clinic Oklahoma College

More information

SOUTH-EAST EUROPEAN JOURNAL of OPHTHALMOLOGY 2015; 1 (1) 34 40

SOUTH-EAST EUROPEAN JOURNAL of OPHTHALMOLOGY 2015; 1 (1) 34 40 Review article SOUTH-EAST EUROPEAN JOURNAL of OPHTHALMOLOGY 2015; 1 (1) 34 40 Retinal nerve fiber layer versus peripapillary capillary density assessment A powerful tool for detecting optic nerve head

More information

Advances in assessing and managing vision impairment

Advances in assessing and managing vision impairment Advances in assessing and managing vision impairment John Grigg Associate Professor and Head Discipline of Ophthalmology Consultant Ophthalmologist Sydney Eye Hospital and The Children s Hospital at Westmead

More information

OCT in Diabetic Macular Edema and its Correlation with Flourescein Angiography

OCT in Diabetic Macular Edema and its Correlation with Flourescein Angiography Uvea OCT in Diabetic Macular Edema and its Correlation with Flourescein Angiography Kirti Jaisingh MS Kirti Jaisingh MS, Yashpal Goel* MS, Kshitij Aditya** DO * Guru Nanak Eye Centre, New Delhi ** Baba

More information

ASSUMPTION OF COGNITIVE UNIFORMITY

ASSUMPTION OF COGNITIVE UNIFORMITY The Human Brain cerebral hemispheres: two most important divisions of the brain, separated by the longitudinal fissure corpus callosum: a large bundle of axons that constitutes the major connection between

More information

The Visual System. Retinal Anatomy Dr. Casagrande February 2, Phone: Office: T2302 MCN

The Visual System. Retinal Anatomy Dr. Casagrande February 2, Phone: Office: T2302 MCN The Visual System Retinal Anatomy Dr. Casagrande February 2, 2004 Phone: 343-4538 Email: vivien.casagrande@mcmail.vanderbilt.edu Office: T2302 MCN Reading assignments and Good Web Sites Chapter 2 in Tovée,

More information

Dehiscence of detached internal limiting membrane in eyes with myopic traction maculopathy with spontaneous resolution

Dehiscence of detached internal limiting membrane in eyes with myopic traction maculopathy with spontaneous resolution Hirota et al. BMC Ophthalmology 2014, 14:39 RESEARCH ARTICLE Open Access Dehiscence of detached internal limiting membrane in eyes with myopic traction maculopathy with spontaneous resolution Kazunari

More information

Plasticity of Cerebral Cortex in Development

Plasticity of Cerebral Cortex in Development Plasticity of Cerebral Cortex in Development Jessica R. Newton and Mriganka Sur Department of Brain & Cognitive Sciences Picower Center for Learning & Memory Massachusetts Institute of Technology Cambridge,

More information

RETINAL CONDITIONS RETINAL CONDITIONS

RETINAL CONDITIONS RETINAL CONDITIONS GENERAL INFORMATION RETINAL CONDITIONS RETINAL CONDITIONS WHAT ARE RETINAL CONDITIONS? Retinal conditions affect the light-sensitive tissue at the back of eye known as the retina. They include diseases

More information

Measurement of Choroidal Thickness in Normal Eyes Using 3D OCT-1000 Spectral Domain Optical Coherence Tomography

Measurement of Choroidal Thickness in Normal Eyes Using 3D OCT-1000 Spectral Domain Optical Coherence Tomography pissn: 111-8942 eissn: 292-9382 Korean J Ophthalmol 212;26(4):255-259 http://dx.doi.org/1.3341/kjo.212.26.4.255 Original Article Measurement of Choroidal Thickness in Normal Eyes Using 3D OCT-1 Spectral

More information

Retinal Nerve Fiber Layer Measurements in Myopia Using Optical Coherence Tomography

Retinal Nerve Fiber Layer Measurements in Myopia Using Optical Coherence Tomography Original Article Philippine Journal of OPHTHALMOLOGY Retinal Nerve Fiber Layer Measurements in Myopia Using Optical Coherence Tomography Dennis L. del Rosario, MD and Mario M. Yatco, MD University of Santo

More information

Corporate Medical Policy

Corporate Medical Policy Corporate Medical Policy Optical Coherence Tomography (OCT) Anterior Segment of the Eye File Name: Origination: Last CAP Review: Next CAP Review: Last Review: optical_coherence_tomography_(oct)_anterior_segment_of_the_eye

More information

Citation. As Published Publisher. Version

Citation. As Published Publisher. Version Effect of Intravitreous Anti Vascular Endothelial Growth Factor Therapy on Choroidal Thickness in Neovascular Age-Related Macular Degeneration Using Spectral-Domain The MIT Faculty has made this article

More information

Neuroscience - Problem Drill 13: The Eye and Visual Processing

Neuroscience - Problem Drill 13: The Eye and Visual Processing Neuroscience - Problem Drill 13: The Eye and Visual Processing Question No. 1 of 10 needed, (3) Pick the answer, and (4) Review the core concept tutorial as needed. 1. Which of the following statements

More information

Parafoveal Scanning Laser Polarimetry for Early Glaucoma Detection

Parafoveal Scanning Laser Polarimetry for Early Glaucoma Detection Yamanashi Med. J. 18(1), 15~ 20, 2003 Original Article Parafoveal Scanning Laser Polarimetry for Early Glaucoma Detection Satoshi KOGURE, Yoshiki TODA, Hiroyuki IIJIMA and Shigeo TSUKAHARA Department of

More information

The Natural History of Diabetic Retinopathy and How Primary Care Makes A Difference

The Natural History of Diabetic Retinopathy and How Primary Care Makes A Difference The Natural History of Diabetic Retinopathy and How Primary Care Makes A Difference We will discuss - How exactly does blood sugar control affect retinopathy? - What are other factors that we measure in

More information

NIH Public Access Author Manuscript JAMA Ophthalmol. Author manuscript; available in PMC 2013 September 10.

NIH Public Access Author Manuscript JAMA Ophthalmol. Author manuscript; available in PMC 2013 September 10. NIH Public Access Author Manuscript Published in final edited form as: JAMA Ophthalmol. 2013 May ; 131(5): 693 694. doi:10.1001/jamaophthalmol.2013.692. Effect of Intravitreous Anti Vascular Endothelial

More information

VMA at the macula resulting in VMT

VMA at the macula resulting in VMT Ocriplasmina for pharmacologic treatment in VMT Teresio Avitabile 1 Introduction PVD is a normal, physiologic process that occurs with aging; however, in some cases, PVD is incomplete Incomplete PVD localized

More information

1. AGENCY USE ONLY (Leave blank) 2. REPORT DATE 3. REPORT TYPE AND DATES COVERED FINAL 01 May 93 TO 30 Apr 95. mwwin i ii ^.. IIM«-»- "«'» ' ' i» 1

1. AGENCY USE ONLY (Leave blank) 2. REPORT DATE 3. REPORT TYPE AND DATES COVERED FINAL 01 May 93 TO 30 Apr 95. mwwin i ii ^.. IIM«-»- «'» ' ' i» 1 REPORT DOCUMENTATION PAGI AFOSR-TR- OiOl Public reoortma burden for this colleaion of information is estimated to average i nour per respor aathenna and maintaining the data needed, and completing and

More information

OCT Assessment of the Vitreoretinal Relationship in CSME

OCT Assessment of the Vitreoretinal Relationship in CSME December 2007 Sonia Rani John et al. - IFIS 375 ORIGINAL ARTICLE OCT Assessment of the Vitreoretinal Relationship in CSME Dr. Manoj S. DNB FRCS, Dr. Unnikrishnan Nair MS DO FRCS, Dr. Gargi Sathish MS Introduction

More information

A Comparative Study of Age Related Macular Degeneration In Relation To SD-OCTand Fundus Photography.

A Comparative Study of Age Related Macular Degeneration In Relation To SD-OCTand Fundus Photography. IOSR Journal of Dental and Medical Sciences (IOSR-JDMS) e-issn: 2279-0853, p-issn: 2279-0861.Volume 14, Issue 11 Ver. III (Nov. 2015), PP 33-37 www.iosrjournals.org A Comparative Study of Age Related Macular

More information

Carlson (7e) PowerPoint Lecture Outline Chapter 6: Vision

Carlson (7e) PowerPoint Lecture Outline Chapter 6: Vision Carlson (7e) PowerPoint Lecture Outline Chapter 6: Vision This multimedia product and its contents are protected under copyright law. The following are prohibited by law: any public performance or display,

More information

Diabetic Retinopathy. Barry Emara MD FRCS(C) Giovanni Caboto Club October 3, 2012

Diabetic Retinopathy. Barry Emara MD FRCS(C) Giovanni Caboto Club October 3, 2012 Diabetic Retinopathy Barry Emara MD FRCS(C) Giovanni Caboto Club October 3, 2012 Outline Statistics Anatomy Categories Assessment Management Risk factors What do you need to do? Objectives Summarize the

More information

DOME SHAPED MACULOPATHY. Ιωάννης Ν. Βαγγελόπουλος Χειρ. Οφθαλμίατρος - Βόλος

DOME SHAPED MACULOPATHY. Ιωάννης Ν. Βαγγελόπουλος Χειρ. Οφθαλμίατρος - Βόλος DOME SHAPED MACULOPATHY Ιωάννης Ν. Βαγγελόπουλος Χειρ. Οφθαλμίατρος - Βόλος DOME SHAPED MACULOPATHY-DEFINITIONS The entity Dome Shaped Macula ( DSM ) was first described by Gaucher and associates in 2008

More information

EPIRETINAL MEMBRANE & VITREOMACULAR TRACTION

EPIRETINAL MEMBRANE & VITREOMACULAR TRACTION EPIRETINAL MEMBRANE & VITREOMACULAR TRACTION Management of ERM and VMT K.V.Chalam,MD,PhD,MBA,FACS Professor and Director of Retina Loma Linda Eye Institute Los Angeles, USA REVIEW ANATOMY The vitreous

More information

Histology of the Eye

Histology of the Eye Histology of the Eye Objectives By the end of this lecture, the student should be able to describe: The general structure of the eye. The microscopic structure of:»cornea.»retina. EYE BULB Three coats

More information

Clinically Significant Macular Edema (CSME)

Clinically Significant Macular Edema (CSME) Clinically Significant Macular Edema (CSME) 1 Clinically Significant Macular Edema (CSME) Sadrina T. Shaw OMT I Student July 26, 2014 Advisor: Dr. Uwaydat Clinically Significant Macular Edema (CSME) 2

More information

Soluble Lutein (Lutemax2020 ) Prevents Retinal Damage in Streptozotocin (STZ)- induced Diabetic Rats

Soluble Lutein (Lutemax2020 ) Prevents Retinal Damage in Streptozotocin (STZ)- induced Diabetic Rats Soluble Lutein (Lutemax2020 ) Prevents Retinal Damage in Streptozotocin (STZ)- induced Diabetic Rats Vijaya Juturu, Ph.D., F.A.C.N. OmniActive Health Technologies, Inc. Morristown, NJ Acknowledgements

More information

eye as a camera Kandel, Schwartz & Jessel (KSJ), Fig 27-3

eye as a camera Kandel, Schwartz & Jessel (KSJ), Fig 27-3 eye as a camera Kandel, Schwartz & Jessel (KSJ), Fig 27-3 retinal specialization fovea: highest density of photoreceptors, aimed at where you are looking -> highest acuity optic disk: cell-free area, where

More information

Acquired vitelliform detachment in patients with subretinal drusenoid deposits (reticular pseudodrusen)

Acquired vitelliform detachment in patients with subretinal drusenoid deposits (reticular pseudodrusen) Zurich Open Repository and Archive University of Zurich Main Library Strickhofstrasse 39 CH-8057 Zurich www.zora.uzh.ch Year: 2011 Acquired vitelliform detachment in patients with subretinal drusenoid

More information

Novel imaging techniques for diabetic macular edema

Novel imaging techniques for diabetic macular edema Documenta Ophthalmologica 97: 341 347, 1999. T.J. Wolfensberger (ed.), Conference Proceedings of the 2 nd International Symposium on Macular Edema, 137 143. 2000 Kluwer Academic Publishers. Printed in

More information

Retinitis pigmentosa (RP) primarily affects the photoreceptor/pigment

Retinitis pigmentosa (RP) primarily affects the photoreceptor/pigment Thickness of Receptor and Post-receptor Retinal Layers in Patients with Retinitis Pigmentosa Measured with Frequency-Domain Optical Coherence Tomography Donald C. Hood, 1,2 Christine E. Lin, 1 Margot A.

More information

Glaucoma Clinical Update. Barry Emara MD FRCS(C) Giovanni Caboto Club October 3, 2012

Glaucoma Clinical Update. Barry Emara MD FRCS(C) Giovanni Caboto Club October 3, 2012 Glaucoma Clinical Update Barry Emara MD FRCS(C) Giovanni Caboto Club October 3, 2012 Objectives Understand the different categories of glaucoma Recognize the symptoms and signs of open angle and angle-closure

More information

Image Formation and Phototransduction. By Dr. Abdelaziz Hussein Lecturer of Physiology

Image Formation and Phototransduction. By Dr. Abdelaziz Hussein Lecturer of Physiology Image Formation and Phototransduction By Dr. Abdelaziz Hussein Lecturer of Physiology Vision Vision is a complex process through which an image of the external environment is formed on the photosensitive

More information

Structural examina.on: Imaging

Structural examina.on: Imaging ManaMa: Glaucoma Structural examina.on: Imaging Luís Abegão Pinto, MD, PhD Department of Ophthalmology CHLC Lisbon Faculty of Medicine, Lisbon University 1 11-10- 2013 Structural changes Qualitative changes

More information

Mark Dunbar: Disclosure

Mark Dunbar: Disclosure Important Things to Understand About OCT Mark T. Dunbar, O.D., F.A.A.O. Bascom Palmer Eye Institute University of Miami, School of Medicine Mark Dunbar: Disclosure Optometry Advisory Board for: Allergan

More information

Vitreomacular interface disorders. Ghanbari MD 1393:10:25

Vitreomacular interface disorders. Ghanbari MD 1393:10:25 Vitreomacular interface disorders Ghanbari MD 1393:10:25 Human vitreous after dissection of the sclera, choroid, and retina. Lamellar structure of the posterior vitreous cortex (PVC) in the monkey. V =

More information