1
|
Carlà MM, Giannuzzi F, Boselli F, Crincoli E, Rizzo S. Extensive macular atrophy with pseudodrusen-like appearance: comprehensive review of the literature. Graefes Arch Clin Exp Ophthalmol 2024; 262:3085-3097. [PMID: 39133226 PMCID: PMC11458735 DOI: 10.1007/s00417-024-06600-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/22/2024] [Accepted: 07/25/2024] [Indexed: 08/13/2024] Open
Abstract
PURPOSE This review focuses on extensive macular atrophy with pseudodrusen-like appearance (EMAP), a recently described maculopathy presenting with pseudodrusen-like lesions and chorioretinal atrophy more pronounced in the vertical axis. METHODS Narrative review of the literature published until May 2024. RESULTS The early onset age of EMAP (50-55 years) and its distinctive natural history, which includes night blindness followed by severe vision loss, differentiate it from atrophic age-related macular degeneration (AMD). A clear pathogenesis has not been determined, but risk factors include female gender and complement system abnormalities (altered levels of C3 and CH50). Moreover, lifelong exposure to pesticides has been suggested as risk factor for direct neuronal degeneration involving rods and cones. In the early phase of the disease, reticular pseudodrusen-like lesions appear in the superior perifovea and tend to coalescence horizontally into a flat, continuous, reflective material localized between the retinal pigmented epithelium and Bruch's membrane. Over time, EMAP causes profound RPE and outer retinal atrophy in the macular area, with a recent classification reporting a 3-stages evolution pattern. Blue autofluorescence showed rapidly evolving atrophy with either hyperautofluorescent or isoautofluorescent borders. Significant similarities between the diffuse-trickling phenotype of geographic atrophy and EMAP have been reported. Macular neovascularization is a possible complication. CONCLUSION EMAP is specific form of early-onset atrophic macular degeneration with rapid evolution and no treatment. Further studies are needed to assess the best management.
Collapse
Affiliation(s)
- Matteo Mario Carlà
- Ophthalmology Department, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy.
- Catholic University "Sacro Cuore", Rome, Italy.
| | - Federico Giannuzzi
- Ophthalmology Department, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
- Catholic University "Sacro Cuore", Rome, Italy
| | - Francesco Boselli
- Ophthalmology Department, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
- Catholic University "Sacro Cuore", Rome, Italy
| | - Emanuele Crincoli
- Ophthalmology Department, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
- Catholic University "Sacro Cuore", Rome, Italy
| | - Stanislao Rizzo
- Ophthalmology Department, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
- Catholic University "Sacro Cuore", Rome, Italy
| |
Collapse
|
2
|
Menean M, Sacconi R, Tombolini B, L'abbate G, Beretta F, Bandello F, Querques G. RETICULAR PSEUDODRUSEN DISAPPEARANCE AFTER DEVELOPMENT OF MACULAR NEOVASCULARIZATION. Retina 2024; 44:1688-1695. [PMID: 39287531 DOI: 10.1097/iae.0000000000004173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
PURPOSE To explore changes in reticular pseudodrusen (RPD) number and location after the development of macular neovascularization (MNV) in eyes with prior intermediate age-related macular degeneration, focusing on different retinal regions differently affected by MNV. METHODS This retrospective longitudinal study included intermediate age-related macular degeneration eyes with RPD that developed MNV. Reticular pseudodrusen were assessed at baseline when MNV was diagnosed (MNV stage) and after anti-vascular endothelial growth factor treatment. Three regions of interest were considered: MNV area, subretinal fluid (SRF) area, and a marginal area of 1,000 µm around SRF (marginal zone). Reticular pseudodrusen counts were compared with age- and sex-matched control eyes with RPD that did not develop MNV. RESULTS Reticular pseudodrusen number exhibited a significant decrease after MNV development in the MNV area (P = 0.048) and in the area with SRF (P = 0.078). A statistically significant decrease was also disclosed in the marginal area around SRF (P = 0.002), associated with larger SRF areas. Control eyes did not show any significant change in the RPD count. CONCLUSION Reticular pseudodrusen reduction after MNV development suggests a complex interplay involving the MNV itself, the presence of SRF, and trophic changes. The results of this study highlight the role of MNV in retinal nutritional balance and provide intriguing results in the RPD life cycle.
Collapse
Affiliation(s)
- Matteo Menean
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy; and
- Department of Ophthalmology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Riccardo Sacconi
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy; and
- Department of Ophthalmology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Beatrice Tombolini
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy; and
- Department of Ophthalmology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Gaia L'abbate
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy; and
- Department of Ophthalmology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Federico Beretta
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy; and
- Department of Ophthalmology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Francesco Bandello
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy; and
- Department of Ophthalmology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Giuseppe Querques
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy; and
- Department of Ophthalmology, IRCCS San Raffaele Hospital, Milan, Italy
| |
Collapse
|
3
|
Duic C, Mukherjee S, Pfau K, Thavikulwat A, Domalpally A, Keenan TDL, Chew E, Cukras C. Local and Global Associations of Reticular Pseudodrusen in Age-Related Macular Degeneration. Ophthalmol Retina 2024; 8:646-656. [PMID: 38278174 DOI: 10.1016/j.oret.2024.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 01/03/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024]
Abstract
PURPOSE To investigate the spatial distribution of reticular pseudodrusen (RPD) in eyes with age-related macular degeneration (AMD) and their correlation with functional measures, retinal thickness, and changes over time. DESIGN Longitudinal, cohort study. PARTICIPANTS Thirty-five participants with RPD and spectrum of AMD severity (including no AMD). METHODS Multimodal imaging was graded by a reading center, including evaluation of color fundus imaging to assess AMD severity scores. Reticular pseudodrusen presence on OCT volumes was confirmed on en face imaging and the RPD extent was contoured on infrared images. One study eye per participant underwent rod-mediated dark adaptation, measuring rod intercept time (RIT) at 5° and, if needed, 12° superior to the fovea. MAIN OUTCOME MEASURES The primary outcome was RIT and OCT thickness measures which were correlated with RPD area. RESULTS A total of 51 eyes had ≥ 1 visit with RPD detected (mean follow-up, 2.19 ± 2.04 years; range, 0-5 years), totaling 169 eye-based visits with RPD. Of the 51 eyes with RPD, 5 (9.8%) developed geographic atrophy and 17 (33.3%) progressed to neovascular AMD. Larger RPD areas were detected more frequently in AMD severity scores 6-7. Reticular pseudodrusen area within an eye generally increased over time. The lesion distribution showed a predilection for the superior retina, especially the outer superior subfield of the ETDRS grid, with the central subfield having least involvement. Reticular pseudodrusen area was inversely correlated with central subfield thickness and positively correlated with RIT at 5° (P = 0.001; r2 = 0.01) and 12° (P = 0.004; r2 = 0.01). Rod-mediated dark adaptation at 5° reached the test ceiling in > 85% of visits, irrespective of RPD lesion presence/absence at the test location. Retinal thickness decreased monotonically, with the central subfield demonstrating the greatest percentage change over 5 years (Δ = -5.47%). CONCLUSIONS In AMD, RPD involve predominantly the superior retina but can involve all ETDRS subfields and evolve over time. Eyes with RPD exhibit structural and functional impairments that can be measured beyond the boundaries of the RPD lesions, suggesting changes associated with RPD are associated with both local changes and a more widespread process. FINANCIAL DISCLOSURES Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.
Collapse
Affiliation(s)
- Cameron Duic
- Division of Epidemiology and Clinical Applications, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Souvick Mukherjee
- Division of Epidemiology and Clinical Applications, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Kristina Pfau
- Department of Ophthalmology, University Hospital Bonn, Bonn, Germany; Department of Ophthalmology, University Hospital Basel, Basel, Switzerland
| | - Alisa Thavikulwat
- Division of Epidemiology and Clinical Applications, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Amitha Domalpally
- Department of Ophthalmology, University of Wisconsin, Madison, Wisconsin
| | - Tiarnan D L Keenan
- Division of Epidemiology and Clinical Applications, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Emily Chew
- Division of Epidemiology and Clinical Applications, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Catherine Cukras
- Division of Epidemiology and Clinical Applications, National Eye Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
4
|
Mauschitz MM, Hochbein BJ, Klinkhammer H, Saßmannshausen M, Terheyden JH, Krawitz P, Finger RP. Prevalence and determinants of subretinal drusenoid deposits in patients' first-degree relatives. Graefes Arch Clin Exp Ophthalmol 2024; 262:53-60. [PMID: 37672102 PMCID: PMC10805990 DOI: 10.1007/s00417-023-06221-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 08/21/2023] [Accepted: 08/26/2023] [Indexed: 09/07/2023] Open
Abstract
PURPOSE Subretinal drusenoid deposits (SDDs) are distinct extracellular alteration anterior to the retinal pigment epithelium (RPE). Given their commonly uniform phenotype, a hereditary predisposition seems likely. Hence, we aim to investigate prevalence and determinants in patients' first-degree relatives. METHODS We recruited SDD outpatients at their visits to our clinic and invited their relatives. We performed a full ophthalmic examination including spectral domain-optical coherence tomography (SD-OCT) and graded presence, disease stage of SDD as well as percentage of infrared (IR) en face area affected by SDD. Moreover, we performed genetic sequencing and calculated a polygenic risk score (PRS) for AMD. We conducted multivariable regression models to assess potential determinants of SDD and associations of SDD with PRS. RESULTS We included 195 participants, 123 patients (mean age 81.4 ± 7.2 years) and 72 relatives (mean age 52.2 ± 14.2 years), of which 7 presented SDD, resulting in a prevalence of 9.7%. We found older age to be associated with SDD presence and area in the total cohort and a borderline association of higher body mass index (BMI) with SDD presence in the relatives. Individuals with SDD tended to have a higher PRS, which, however, was not statistically significant in the multivariable regression. CONCLUSION Our study indicates a potential hereditary aspect of SDD and confirms the strong association with age. Based on our results, relatives of SDD patients ought to be closely monitored for retinal alterations, particularly at an older age. Further longitudinal studies with larger sample size and older relatives are needed to confirm or refute our findings.
Collapse
Affiliation(s)
- Matthias M Mauschitz
- Department of Ophthalmology, University Bonn, Ernst-Abbe-Straße 2, 53127, Bonn, Germany.
| | - Benedikt J Hochbein
- Department of Ophthalmology, University Bonn, Ernst-Abbe-Straße 2, 53127, Bonn, Germany
| | - Hannah Klinkhammer
- Institute for Genomic Statistics and Bioinformatics, University Bonn, Bonn, Germany
- Institute for Medical Biometry, Informatics and Epidemiology, University Bonn, Bonn, Germany
| | | | - Jan H Terheyden
- Department of Ophthalmology, University Bonn, Ernst-Abbe-Straße 2, 53127, Bonn, Germany
| | - Peter Krawitz
- Institute for Genomic Statistics and Bioinformatics, University Bonn, Bonn, Germany
| | - Robert P Finger
- Department of Ophthalmology, University Bonn, Ernst-Abbe-Straße 2, 53127, Bonn, Germany
| |
Collapse
|
5
|
Abdolrahimzadeh S, Zweifel SA, Di Pippo M, Bajka A, Scuderi G, Lotery AJ. Central macular choriocapillaris impairment as a manifestation of microvascular disease in eyes with subretinal drusenoid deposits. Eye (Lond) 2024; 38:173-178. [PMID: 37419959 PMCID: PMC10764916 DOI: 10.1038/s41433-023-02654-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 06/20/2023] [Accepted: 06/21/2023] [Indexed: 07/09/2023] Open
Abstract
BACKGROUND/OBJECTIVES Microvascular alterations and choroidal impairment are emerging as a pathologic pathway in age-related macular degeneration (AMD). This study aimed to evaluate the central macular choriocapillaris (CC) in eyes with subretinal drusenoid deposits (SDD) and the retinal microvasculature in patients with early AMD phenotypes. SUBJECTS/METHODS This was an institutional, multicentric observational cross-sectional study. Ninety-nine eyes of 99 subjects; 33 eyes with SDD only, 33 eyes with conventional drusen (CD) only, and 33 eyes of healthy age-matched subjects were included. Comprehensive ophthalmologic examination and optical coherence tomography angiography (OCTA) was performed. The central macular flow area of the CC was analysed in the SDD group and the vessel density of the retinal superficial capillary plexus (SCP) and deep capillary plexus (DCP) was analysed in the SDD and CD groups using automated OCTA output parameters. RESULTS The flow area of the CC in the SDD group was significantly reduced (p ≤ 0.001) with respect to the healthy control group. There was a trend of reduction of vessel density of the SCP and the DCP in the SDD and CD group with respect to controls, although this did not reach statistical significance. CONCLUSIONS OCTA data in the present report corroborate the role of vascular damage in early AMD with CC impairment in the central macular area in eyes with SDD.
Collapse
Affiliation(s)
- Solmaz Abdolrahimzadeh
- Ophthalmology Unit, Neurosciences, Mental Health, and Sense Organs (NESMOS) Department, Faculty of Medicine and Psychology, University of Rome Sapienza, Rome, Italy.
- St. Andrea Hospital, Via di Grottarossa 1035/1039, Rome, Italy.
| | - Sandrine Anne Zweifel
- Department of Ophthalmology, University Hospital Zurich (USZ), University of Zurich, Zurich, Switzerland
| | - Mariachiara Di Pippo
- Ophthalmology Unit, Neurosciences, Mental Health, and Sense Organs (NESMOS) Department, Faculty of Medicine and Psychology, University of Rome Sapienza, Rome, Italy
| | - Anahita Bajka
- Department of Ophthalmology, University Hospital Zurich (USZ), University of Zurich, Zurich, Switzerland
| | - Gianluca Scuderi
- Ophthalmology Unit, Neurosciences, Mental Health, and Sense Organs (NESMOS) Department, Faculty of Medicine and Psychology, University of Rome Sapienza, Rome, Italy
- St. Andrea Hospital, Via di Grottarossa 1035/1039, Rome, Italy
| | - Andrew John Lotery
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.
| |
Collapse
|
6
|
Wu Z, Schmitz-Valckenberg S, Blodi BA, Holz FG, Jaffe GJ, Liakopoulos S, Sadda SR, Bonse M, Brown T, Choong J, Clifton B, Corradetti G, Corvi F, Dieu AC, Dooling V, Pak JW, Saßmannshausen M, Skalak C, Thiele S, Guymer RH. Reticular Pseudodrusen: Interreader Agreement of Evaluation on OCT Imaging in Age-Related Macular Degeneration. OPHTHALMOLOGY SCIENCE 2023; 3:100325. [PMID: 37292179 PMCID: PMC10244688 DOI: 10.1016/j.xops.2023.100325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 06/10/2023]
Abstract
Purpose To determine the interreader agreement for reticular pseudodrusen (RPD) assessment on combined infrared reflectance (IR) and OCT imaging in the early stages of age-related macular degeneration across a range of different criteria to define their presence. Design Interreader agreement study. Participants Twelve readers from 6 reading centers. Methods All readers evaluated 100 eyes from individuals with bilateral large drusen for the following: (1) the presence of RPD across a range of different criteria and (2) the number of Stage 2 or 3 RPD lesions (from 0 to ≥ 5 lesions) on an entire OCT volume scan and on a selected OCT B-scan. Supportive information was available from the corresponding IR image. Main Outcome Measures Interreader agreement, as assessed by Gwet's first-order agreement coefficient (AC1). Results When evaluating an entire OCT volume scan, there was substantial interreader agreement for the presence of any RPD, any or ≥ 5 Stage 2 or 3 lesions, and ≥ 5 definite lesions on en face IR images corresponding to Stage 2 or 3 lesions (AC1 = 0.60-0.72). On selected OCT B-scans, there was also moderate-to-substantial agreement for the presence of any RPD, any or ≥ 5 Stage 2 or 3 lesions (AC1 = 0.58-0.65) and increasing levels of agreement with increasing RPD stage (AC1 = 0.08, 0.56, 0.78, and 0.99 for the presence of any Stage 1, 2, 3, and 4 lesions, respectively). There was substantial agreement regarding the number of Stage 2 or 3 lesions on an entire OCT volume scan (AC1 = 0.68), but only fair agreement for this evaluation on selected B-scans (AC1 = 0.30). Conclusions There was generally substantial or near-substantial-but not near-perfect-agreement for assessing the presence of RPD on entire OCT volume scans or selected B-scans across a range of differing RPD criteria. These findings underscore how interreader variability would likely contribute to the variability of findings related to the clinical associations of RPD. The low levels of agreement for assessing RPD number on OCT B-scans underscore the likely challenges of quantifying RPD extent with manual grading. Financial Disclosures Proprietary or commercial disclosure may be found after the references.
Collapse
Affiliation(s)
- Zhichao Wu
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia
- Ophthalmology, Department of Surgery, The University of Melbourne, Melbourne, Australia
| | - Steffen Schmitz-Valckenberg
- Department of Ophthalmology and GRADE Reading Center, University of Bonn, Bonn, Germany
- Utah Retinal Reading Center (UREAD) John A. Moran Eye Center, University of Utah, Salt Lake City, Utah
| | - Barbara A. Blodi
- Department of Ophthalmology and Visual Sciences, Wisconsin Reading Center (WRC), University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Frank G. Holz
- Department of Ophthalmology and GRADE Reading Center, University of Bonn, Bonn, Germany
| | - Glenn J. Jaffe
- Department of Ophthalmology, Duke University, Durham, North Carolina
| | - Sandra Liakopoulos
- Cologne Image Reading Center and Laboratory (CIRCL) and Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Department of Ophthalmology, Goethe-University Frankfurt, Germany
| | - Srinivas R. Sadda
- Doheny Imaging Reading Center (DIRC) and Doheny Eye Institute, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Mari Bonse
- Cologne Image Reading Center and Laboratory (CIRCL) and Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Tyler Brown
- Utah Retinal Reading Center (UREAD) John A. Moran Eye Center, University of Utah, Salt Lake City, Utah
| | - John Choong
- Department of Ophthalmology, Duke University, Durham, North Carolina
| | - Bailey Clifton
- Utah Retinal Reading Center (UREAD) John A. Moran Eye Center, University of Utah, Salt Lake City, Utah
| | - Giulia Corradetti
- Doheny Imaging Reading Center (DIRC) and Doheny Eye Institute, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Federico Corvi
- Doheny Imaging Reading Center (DIRC) and Doheny Eye Institute, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Andrew C. Dieu
- Department of Ophthalmology and Visual Sciences, Wisconsin Reading Center (WRC), University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Vivienne Dooling
- Cologne Image Reading Center and Laboratory (CIRCL) and Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Jeong W. Pak
- Department of Ophthalmology and Visual Sciences, Wisconsin Reading Center (WRC), University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | | | - Cindy Skalak
- Department of Ophthalmology, Duke University, Durham, North Carolina
| | - Sarah Thiele
- Department of Ophthalmology and GRADE Reading Center, University of Bonn, Bonn, Germany
| | - Robyn H. Guymer
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia
- Ophthalmology, Department of Surgery, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
7
|
Khan AH, Chowers I, Lotery AJ. Beyond the Complement Cascade: Insights into Systemic Immunosenescence and Inflammaging in Age-Related Macular Degeneration and Current Barriers to Treatment. Cells 2023; 12:1708. [PMID: 37443742 PMCID: PMC10340338 DOI: 10.3390/cells12131708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 06/22/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
Landmark genetic studies have revealed the effect of complement biology and its regulation on the pathogenesis of age-related macular degeneration (AMD). Limited phase 3 clinical trial data showing a benefit of complement inhibition in AMD raises the prospect of more complex mediators at play. Substantial evidence supports the role of para-inflammation in maintaining homeostasis in the retina and choroid. With increasing age, a decline in immune system regulation, known as immunosenescence, has been shown to alter the equilibrium maintained by para-inflammation. The altered equilibrium results in chronic, sterile inflammation with aging, termed 'inflammaging', including in the retina and choroid. The chronic inflammatory state in AMD is complex, with contributions from cells of the innate and adaptive branches of the immune system, sometimes with overlapping features, and the interaction of their secretory products with retinal cells such as microglia and retinal pigment epithelium (RPE), extracellular matrix and choroidal vascular endothelial cells. In this review, the chronic inflammatory state in AMD will be explored by immune cell type, with a discussion of factors that will need to be overcome in the development of curative therapies.
Collapse
Affiliation(s)
- Adnan H. Khan
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
- Southampton Eye Unit, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Itay Chowers
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91121, Israel
| | - Andrew J. Lotery
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
- Southampton Eye Unit, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| |
Collapse
|
8
|
Abdolrahimzadeh S, Di Pippo M, Sordi E, Cusato M, Lotery AJ. Subretinal drusenoid deposits as a biomarker of age-related macular degeneration progression via reduction of the choroidal vascularity index. Eye (Lond) 2023; 37:1365-1370. [PMID: 35739243 PMCID: PMC10169760 DOI: 10.1038/s41433-022-02134-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 05/09/2022] [Accepted: 06/09/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND/OBJECTIVES This study aimed to analyse the role of the choroid in early age-related macular degeneration (AMD) by analysing choroidal vascularity index (CVI) in pure cohorts of patients with subretinal drusenoid deposits (SDD) or conventional drusen (CD). SUBJECTS/METHODS This was an observational cross-sectional study. Comprehensive ophthalmologic examination and multimodal imaging including fundus photography, autofluorescence, near infrared reflectance, and spectral domain optical coherence tomography (SDOCT) was performed. CVI processing was performed on a foveal horizontal SDOCT scan with binarization using Image J Image software and calculated as the ratio between luminal area (LA) and total area (TA). RESULTS Sixty-nine eyes of 69 participants were included; 23 eyes with SDD alone, 22 eyes with CD alone, and 24 control eyes of healthy age-matched subjects. CVI was significantly reduced in the SDD and CD group compared to controls (p = 0.0001). Post-hoc analysis revealed a significant reduction of CVI in the SDD versus the control group (p = 0.0002), in the CD versus the control group (p = 0.001), and in the SDD versus the CD group (p = 0.006). Covariance analysis showed a significant difference of LA (p = 0.033) but no significant difference of TA (p = 0.106) between the three groups. Direct comparison between CD and SDD showed a significant reduction of LA and TA in the SDD group. CONCLUSIONS CVI may have prognostic implications in early AMD. SDD is a biomarker of AMD progression and the mechanism for this could be via reduction of the CVI.
Collapse
Affiliation(s)
- Solmaz Abdolrahimzadeh
- Ophthalmology Unit, Neurosciences, Mental Health, and Sense Organs (NESMOS) Department, Faculty of Medicine and Psychology, University of Rome Sapienza, Rome, Italy.
- St. Andrea Hospita, Via di Grottarossa 1035/1039, Rome, 00189, Italy.
| | - Mariachiara Di Pippo
- Ophthalmology Unit, Neurosciences, Mental Health, and Sense Organs (NESMOS) Department, Faculty of Medicine and Psychology, University of Rome Sapienza, Rome, Italy
| | - Edoardo Sordi
- Ophthalmology Unit, Neurosciences, Mental Health, and Sense Organs (NESMOS) Department, Faculty of Medicine and Psychology, University of Rome Sapienza, Rome, Italy
| | - Mattia Cusato
- Ophthalmology Unit, Neurosciences, Mental Health, and Sense Organs (NESMOS) Department, Faculty of Medicine and Psychology, University of Rome Sapienza, Rome, Italy
| | - Andrew John Lotery
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
| |
Collapse
|
9
|
Wang X, Sadda SR, Ip MS, Sarraf D, Zhang Y. In Vivo Longitudinal Measurement of Cone Photoreceptor Density in Intermediate Age-Related Macular Degeneration. Am J Ophthalmol 2023; 248:60-75. [PMID: 36436549 PMCID: PMC10038851 DOI: 10.1016/j.ajo.2022.11.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 11/14/2022] [Accepted: 11/16/2022] [Indexed: 11/27/2022]
Abstract
PURPOSE To evaluate cone photoreceptor density in clinically unremarkable retinal regions in patients with age-related macular degeneration (AMD) using adaptive optics scanning laser ophthalmoscopy (AOSLO). DESIGN Prospective case series with normal comparison group. METHODS Ten eyes of 7 patients with intermediate AMD were studied, including 4 with predominantly subretinal drusenoid deposits (SDD) and 3 without SDD. Macular regions with a clinical absence of AMD-associated lesions were identified by cone packing structure on AOSLO and optical coherence tomography. Cone density was measured in 1174 clinically unremarkable regions within the central subfield (CSF), the inner (IR), and outer rings (OR) of the Early Treatment Diabetic Retinopathy Study grid over 39.6 ± 3.3 months and compared with age-matched normal values obtained in 17 participants. RESULTS Cone density decreased at 98.3% of the examined locations over time in the eyes with AMD. In the CSF, IR, and OR, cones declined by -255 ± 135, -133 ± 45, and -59 ± 24 cones/degree2/year, respectively, in eyes with SDD, and by -212 ± 89, -83 ± 37, and -27 ± 18 cones/degree2/year, respectively, in eyes without SDD. The percentage of retinal loci with cone density lower than normal (Z score < -2) increased over the follow-up: from 42% at the baseline to 80% at the last visit in eyes with SDD and from 31% to 70% in eyes without SDD. CONCLUSIONS AOSLO revealed cone photoreceptor loss in regions that appear otherwise unremarkable clinically. These findings may help explain the loss of mesopic sensitivity reported in these areas in eyes with intermediate AMD.
Collapse
Affiliation(s)
- Xiaolin Wang
- From the Doheny Eye Institute (X.W., S.R.S., M.I., Y.Z.), Pasadena, California
| | - SriniVas R Sadda
- From the Doheny Eye Institute (X.W., S.R.S., M.I., Y.Z.), Pasadena, California; Department of Ophthalmology, University of California-Los Angeles (S.R.S., M.I., D.S., Y.Z.), Los Angeles, California
| | - Michael S Ip
- From the Doheny Eye Institute (X.W., S.R.S., M.I., Y.Z.), Pasadena, California; Department of Ophthalmology, University of California-Los Angeles (S.R.S., M.I., D.S., Y.Z.), Los Angeles, California
| | - David Sarraf
- Department of Ophthalmology, University of California-Los Angeles (S.R.S., M.I., D.S., Y.Z.), Los Angeles, California; Stein Eye Institute (David Sarraf), Los Angeles, California, USA
| | - Yuhua Zhang
- From the Doheny Eye Institute (X.W., S.R.S., M.I., Y.Z.), Pasadena, California; Department of Ophthalmology, University of California-Los Angeles (S.R.S., M.I., D.S., Y.Z.), Los Angeles, California.
| |
Collapse
|
10
|
Xu R, Wang Y, Du J, Salido EM. Retinal Metabolic Profile on IMPG2 Deficiency Mice with Subretinal Lesions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1415:457-463. [PMID: 37440072 DOI: 10.1007/978-3-031-27681-1_67] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
The interphotoreceptor matrix (IPM) is the extracellular matrix between the photoreceptors and the retinal pigment epithelium (RPE). The IPM has two proteoglycans: the IPM proteoglycans 1 and 2 (IMPG1 and IMPG2, respectively). Patients with mutations on IMPG2 develop subretinal vitelliform lesions that affect vision. We previously created an IMPG2 knockout (KO) mice model that generates subretinal lesions similar to those found in humans. These subretinal lesions in IMPG2 KO mice retinas are, in part, composed of mislocalized IMPG1. In addition, IMPG2 KO mice show microscopic IMPG1 material accumulation between the RPE and the photoreceptor outer segments. In this work we discuss the possibility that material accumulation on IMPG2 KO mice retinas affects photoreceptor metabolism. To further investigate this idea, we used targeted metabolomics to profile retinal metabolome on IMPG2 KO mice. The metabolite set enrichment analysis showed reduced glutamate metabolism, urea cycle, and galactose metabolism suggesting affected energy metabolism in mice retinas of IMPG2 KO mice with subretinal lesion.
Collapse
Affiliation(s)
- Rong Xu
- Departments of Biochemistry and molecular medicine, and Ophthalmology and Visual Sciences, West Virginia University, Morgantown, WV, USA
| | - Yekai Wang
- Departments of Biochemistry and molecular medicine, and Ophthalmology and Visual Sciences, West Virginia University, Morgantown, WV, USA
| | - Jianhai Du
- Departments of Biochemistry and molecular medicine, and Ophthalmology and Visual Sciences, West Virginia University, Morgantown, WV, USA
| | - Ezequiel M Salido
- Departments of Biochemistry and molecular medicine, and Ophthalmology and Visual Sciences, West Virginia University, Morgantown, WV, USA.
| |
Collapse
|
11
|
Yednock T, Fong DS, Lad EM. C1q and the classical complement cascade in geographic atrophy secondary to age-related macular degeneration. Int J Retina Vitreous 2022; 8:79. [PMID: 36348407 PMCID: PMC9641935 DOI: 10.1186/s40942-022-00431-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 10/21/2022] [Indexed: 11/10/2022] Open
Abstract
Geographic atrophy (GA) secondary to age-related macular degeneration (AMD) is a retinal neurodegenerative disorder. Human genetic data support the complement system as a key component of pathogenesis in AMD, which has been further supported by pre-clinical and recent clinical studies. However, the involvement of the different complement pathways (classical, lectin, alternative), and thus the optimal complement inhibition target, has yet to be fully defined. There is evidence that C1q, the initiating molecule of the classical pathway, is a key driver of complement activity in AMD. C1q is expressed locally by infiltrating phagocytic cells and C1q-activating ligands are present at disease onset and continue to accumulate with disease progression. The accumulation of C1q on photoreceptor synapses with age and disease is consistent with its role in synapse elimination and neurodegeneration that has been observed in other neurodegenerative disorders. Furthermore, genetic deletion of C1q, local pharmacologic inhibition within the eye, or genetic deletion of downstream C4 prevents photoreceptor cell damage in mouse models. Hence, targeting the classical pathway in GA could provide a more specific therapeutic approach with potential for favorable efficacy and safety.
Collapse
Affiliation(s)
- Ted Yednock
- Annexon Biosciences, 1400 Sierra Point Parkway Building C, 2nd Floor, Brisbane, CA, 94005, USA
| | - Donald S Fong
- Annexon Biosciences, 1400 Sierra Point Parkway Building C, 2nd Floor, Brisbane, CA, 94005, USA.
| | - Eleonora M Lad
- Department of Ophthalmology, Duke University Medical Center, 2351 Erwin Rd, Durham, NC, 27705, USA
| |
Collapse
|
12
|
Developing and validating a multivariable prediction model which predicts progression of intermediate to late age-related macular degeneration-the PINNACLE trial protocol. Eye (Lond) 2022; 37:1275-1283. [PMID: 35614343 PMCID: PMC9130980 DOI: 10.1038/s41433-022-02097-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/27/2022] [Accepted: 05/06/2022] [Indexed: 11/08/2022] Open
Abstract
AIMS Age-related macular degeneration (AMD) is characterised by a progressive loss of central vision. Intermediate AMD is a risk factor for progression to advanced stages categorised as geographic atrophy (GA) and neovascular AMD. However, rates of progression to advanced stages vary between individuals. Recent advances in imaging and computing technologies have enabled deep phenotyping of intermediate AMD. The aim of this project is to utilise machine learning (ML) and advanced statistical modelling as an innovative approach to discover novel features and accurately quantify markers of pathological retinal ageing that can individualise progression to advanced AMD. METHODS The PINNACLE study consists of both retrospective and prospective parts. In the retrospective part, more than 400,000 optical coherent tomography (OCT) images collected from four University Teaching Hospitals and the UK Biobank Population Study are being pooled, centrally stored and pre-processed. With this large dataset featuring eyes with AMD at various stages and healthy controls, we aim to identify imaging biomarkers for disease progression for intermediate AMD via supervised and unsupervised ML. The prospective study part will firstly characterise the progression of intermediate AMD in patients followed between one and three years; secondly, it will validate the utility of biomarkers identified in the retrospective cohort as predictors of progression towards late AMD. Patients aged 55-90 years old with intermediate AMD in at least one eye will be recruited across multiple sites in UK, Austria and Switzerland for visual function tests, multimodal retinal imaging and genotyping. Imaging will be repeated every four months to identify early focal signs of deterioration on spectral-domain optical coherence tomography (OCT) by human graders. A focal event triggers more frequent follow-up with visual function and imaging tests. The primary outcome is the sensitivity and specificity of the OCT imaging biomarkers. Secondary outcomes include sensitivity and specificity of novel multimodal imaging characteristics at predicting disease progression, ROC curves, time from development of imaging change to development of these endpoints, structure-function correlations, structure-genotype correlation and predictive risk models. CONCLUSIONS This is one of the first studies in intermediate AMD to combine both ML, retrospective and prospective AMD patient data with the goal of identifying biomarkers of progression and to report the natural history of progression of intermediate AMD with multimodal retinal imaging.
Collapse
|
13
|
Vilela MAP, Battaglia Parodi M. Extensive macular atrophy with pseudodrusen-like: Case series and review. Eur J Ophthalmol 2022:11206721221102269. [PMID: 35607265 DOI: 10.1177/11206721221102269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
PURPOSE To describe the clinical characteristics of two patients affected by extensive macular atrophy with pseudodrusen-like (EMAP). METHODS Two patients affected by EMAP underwent multimodal imaging, including fundus autofluorescence and optical coherence tomography. RESULTS The patients showed the typical clinical appearance with macular atrophy with larger vertical axis surrounded by pseudodrusen-like deposits involving the midperiphery, associated with paving stone lesions in the retinal periphery. CONCLUSION EMAP is a complex condition sharing clinical characteristics of age-related macular degeneration. Further studies are warranted to identify the early biomarker of the disease.
Collapse
|
14
|
Scuderi L, Gattazzo I, de Paula A, Iodice CM, Di Tizio F, Perdicchi A. Understanding the role of microperimetry in glaucoma. Int Ophthalmol 2022; 42:2289-2301. [DOI: 10.1007/s10792-021-02203-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 12/23/2021] [Indexed: 11/28/2022]
|
15
|
Wu L, Monge M, Araya A. Subretinal drusenoid deposits: An update. Taiwan J Ophthalmol 2022; 12:138-146. [PMID: 35813798 PMCID: PMC9262011 DOI: 10.4103/tjo.tjo_18_22] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 03/12/2022] [Indexed: 11/30/2022] Open
Abstract
A wide spectrum of phenotypic manifestations characterizes age-related macular degeneration (AMD). Drusen is considered the hallmark of AMD and is located underneath the retinal pigment epithelium (RPE). In contrast, subretinal drusenoid deposits (SDDs), also known as reticular pseudodrusens, are located in the subretinal space, on top of the RPE. SDDs are poorly detected by clinical examination and color fundus photography. Multimodal imaging is required for their proper diagnosis. SDDs are topographically and functionally related to rods. SDDs cause a deep impairment in retinal sensitivity and dark adaptation. SDDs are dynamic structures that may grow, fuse with each other, or regress over time. An intermediate step in some eyes is the development of an acquired vitelliform lesion. The presence of SDD confers an eye a high risk for the development of late AMD. SDD leads to macular neovascularization, particularly type 3, geographic atrophy, and outer retinal atrophy.
Collapse
|
16
|
Abdolrahimzadeh S, Di Pippo M, Sordi E, Zweifel SA. Inner Retinal Layer Thickness Alterations in Early Age Related Macular Degeneration in Eyes with Subretinal Drusenoid Deposits or Conventional Drusen. J Clin Med 2021; 10:jcm10215136. [PMID: 34768654 PMCID: PMC8584863 DOI: 10.3390/jcm10215136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/26/2021] [Accepted: 10/29/2021] [Indexed: 11/16/2022] Open
Abstract
The purpose of this study was to evaluate central and parafoveal inner retinal layer thickness in patients with subretinal drusenoid deposits (SDD) or conventional drusen (CD). Participants underwent comprehensive ophthalmoscopic examination. Evidence of SDD or CD was evaluated with near infrared reflectance and spectral domain optical coherence tomography. Quantification of subfoveal lesions was made through a qualitative analysis of vertical and horizontal SD-OCT scans centered on the fovea. Inner retinal layer macular thickness measurements were obtained for central circles with 1, 3, and 5 mm diameter. Continuous variables were compared by the analysis of covariance (ANCOVA) with post-hoc Tukey HSD correction for multiple comparison analysis. Fifty-five patients were included in the study; 18 eyes with SDD alone, 19 eyes with CD alone, and 18 eyes of healthy age-matched subjects. Eight eyes with SDD (44%) and 13 eyes with CD (68%) had subfoveal lesions. There was significant reduction in the inner retinal layer thickness in the central 1mm area and in the superior 3 mm area in the SDD and CD group compared to controls. In conclusion the inner retinal layer is thinner in the central macula and in the superior parafovea in eyes.
Collapse
Affiliation(s)
- Solmaz Abdolrahimzadeh
- Ophthalmology Unit, Neurosciences, Mental Health, and Sense Organs (NESMOS) Department, Faculty of Medicine and Psychology, University of Rome Sapienza, St. Andrea Hospital, Via di Grottarossa 1035/1039, 00189 Rome, Italy; (M.D.P.); (E.S.)
- Correspondence:
| | - Mariachiara Di Pippo
- Ophthalmology Unit, Neurosciences, Mental Health, and Sense Organs (NESMOS) Department, Faculty of Medicine and Psychology, University of Rome Sapienza, St. Andrea Hospital, Via di Grottarossa 1035/1039, 00189 Rome, Italy; (M.D.P.); (E.S.)
| | - Edoardo Sordi
- Ophthalmology Unit, Neurosciences, Mental Health, and Sense Organs (NESMOS) Department, Faculty of Medicine and Psychology, University of Rome Sapienza, St. Andrea Hospital, Via di Grottarossa 1035/1039, 00189 Rome, Italy; (M.D.P.); (E.S.)
| | - Sandrine Anne Zweifel
- Department of Ophthalmology, University Hospital Zurich, Frauenklinikstrasse 24, 8091 Zurich, Switzerland;
- University of Zurich, Rämistrasse 71, 8006 Zurich, Switzerland
| |
Collapse
|
17
|
Borooah S, Papastavrou V, Lando L, Han J, Lin JH, Ayyagari R, Dhillon B, Browning AC. Reticular Pseudodrusen in Late-Onset Retinal Degeneration. Ophthalmol Retina 2021; 5:1043-1051. [PMID: 33352318 PMCID: PMC8217414 DOI: 10.1016/j.oret.2020.12.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 12/08/2020] [Accepted: 12/15/2020] [Indexed: 02/05/2023]
Abstract
PURPOSE To characterize the association of reticular pseudodrusen (RPD) with late-onset retinal degeneration (L-ORD) using multimodal imaging. DESIGN Prospective, 2-center, longitudinal case series. PARTICIPANTS Twenty-nine patients with L-ORD. METHODS All patients were evaluated within a 3-year interval with near-infrared reflectance, fundus autofluorescence, and spectral-domain OCT. In addition, a subset of patients also underwent indocyanine green angiography, fundus fluorescein angiography, mesopic microperimetry, and multifocal electroretinography. MAIN OUTCOME MEASURES Prevalence, topographic distribution, and temporal phenotypic changes of RPD in L-ORD. RESULTS A total of 29 patients with molecularly confirmed L-ORD were included in this prospective study. Reticular pseudodrusen was detected in 18 patients (62%) at baseline, 10 of whom were men. The prevalence of RPD varied with age. The mean age of RPD patients was 57.3 ± 7.2 years. Reticular pseudodrusen was not seen in patients younger than the fifth decade of life (n = 3 patients) or in the eighth decade of life (n = 5 patients). Reticular pseudodrusen were found commonly in the macula with relative sparing of the fovea and also were identified in the peripheral retina. The morphologic features of RPD changed with follow-up. Two patients (3 eyes) demonstrated RPD regression. CONCLUSIONS Reticular pseudodrusen is found frequently in patients with L-ORD and at a younger age than in individuals with age-related macular degeneration (AMD). Reticular pseudodrusen exhibits quick formation and collapse, change in type and morphologic features with time, and relative foveal sparing and also has a peripheral retinal location in L-ORD.
Collapse
Affiliation(s)
- Shyamanga Borooah
- Centre for Clinical Brain Sciences, School of Clinical Sciences, University of Edinburgh, Edinburgh, United Kingdom; Shiley Eye Institute, University of California, San Diego, La Jolla, California.
| | | | - Leonardo Lando
- Shiley Eye Institute, University of California, San Diego, La Jolla, California; Department of Ophthalmology, Federal University of Goias, Goiania, Brazil
| | - Jonathan Han
- Shiley Eye Institute, University of California, San Diego, La Jolla, California
| | - Jonathan H Lin
- Shiley Eye Institute, University of California, San Diego, La Jolla, California; Departments of Ophthalmology and Pathology, Stanford University, Stanford, California; Veterans Affairs, Palo Alto Healthcare System, Palo Alto, California
| | - Radha Ayyagari
- Shiley Eye Institute, University of California, San Diego, La Jolla, California
| | - Baljean Dhillon
- Centre for Clinical Brain Sciences, School of Clinical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Andrew C Browning
- Newcastle Eye Centre, Royal Victoria Infirmary, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
18
|
BASELINE PREDICTORS ASSOCIATED WITH 3-YEAR CHANGES IN DARK ADAPTATION IN AGE-RELATED MACULAR DEGENERATION. Retina 2021; 41:2098-2105. [PMID: 33625114 PMCID: PMC8368068 DOI: 10.1097/iae.0000000000003152] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE To assess the relationship between baseline age-related macular degeneration (AMD) and disease stage, as well as optical coherence tomography features seen in AMD, with 3-year changes in dark adaptation (DA). METHODS Prospective longitudinal study including patients with AMD and a comparison group (n = 42 eyes, 27 patients). At baseline and 3 years, we obtained color fundus photographs, spectral-domain optical coherence tomography, and rod-mediated DA (20 minutes protocol). Multilevel mixed-effect models were used for analyses, with changes in rod intercept time at 3 years as the primary outcome. As some eyes (n = 11) reached the DA testing ceiling value at baseline, we used 3-year changes in area under the DA curve as an additional outcome. RESULTS Baseline AMD, AMD stage, and hyperreflective foci on optical coherence tomography were associated with larger changes in rod intercept time at 3 years. When change in area under the DA curve was used as an outcome, in addition to these features, the presence of retinal atrophy and drusenoid pigment epithelial detachment had significant associations. New subretinal drusenoid deposits at 3 years were also associated with more pronounced changes in rod intercept time and area under the DA curve. CONCLUSION Specific optical coherence tomography features are associated with DA impairments over time, which supports that structural changes predict functional loss over 3 years.
Collapse
|
19
|
Temel E, Batıoğlu F, Demirel S, Özcan G, Yanık Ö, Özmert E. Vascular and structural alterations of the choroid evaluated by optical coherence tomography angiography and enhanced-depth imaging optical coherence tomography in eyes with reticular pseudodrusen and soft drusen. Photodiagnosis Photodyn Ther 2021; 36:102549. [PMID: 34562645 DOI: 10.1016/j.pdpdt.2021.102549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 09/01/2021] [Accepted: 09/20/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND To assess the vascularity of choriocapillaris and structural choroidal differences in eyes with reticular pseudodrusen (RPD) and soft drusen. METHODS 21 eyes with RPD (group 1), 17 eyes with soft drusen (group 2), and 19 eyes as a control group (group 3) were included in this study. Choriocapillaris vascular density and flow area were measured by optical coherence tomography angiography. Total choroidal area, luminal area, stromal area, and lumen/stroma ratios were measured on optical coherence tomography B-scans converted to binary images. RESULTS Mean choriocapillaris vascular density was higher in group 3 than other groups (group 1 vs 3, p = 0.001; group 2 vs 3, p = 0.003). Mean flow area in choriocapillaris was higher in group 3 than other groups (group 1 vs 3, p = 0.001; group 2 vs 3, p = 0.001). Mean luminal, stromal, and total choroidal areas decreased in group 1 and group 2 compared to controls (p < 0.001, p < 0.001, and p < 0.001, respectively). The stroma ratio decreased in group 1 compared to group 3 (p = 0.013). The lumen ratio and lumen/stroma ratio increased in group 1 compared to group 3 (p = 0.012 and p = 0.008, respectively). CONCLUSIONS The choroid of eyes with RPD and soft drusen was affected in both choriocapillaris and whole choroid layer.
Collapse
Affiliation(s)
- Emine Temel
- Kırşehir Ahi Evran Training and Research Hospital, Department of Opthalmology, Kırşehir, Turkey; Ankara University Faculty of Medicine, Department of Ophthalmology, Ankara, Turkey.
| | - Figen Batıoğlu
- Ankara University Faculty of Medicine, Department of Ophthalmology, Ankara, Turkey
| | - Sibel Demirel
- Ankara University Faculty of Medicine, Department of Ophthalmology, Ankara, Turkey
| | - Gökçen Özcan
- Kırşehir Ahi Evran Training and Research Hospital, Department of Opthalmology, Kırşehir, Turkey; Ankara University Faculty of Medicine, Department of Ophthalmology, Ankara, Turkey
| | - Özge Yanık
- Ankara University Faculty of Medicine, Department of Ophthalmology, Ankara, Turkey
| | - Emin Özmert
- Ankara University Faculty of Medicine, Department of Ophthalmology, Ankara, Turkey
| |
Collapse
|
20
|
Cleland SC, Domalpally A, Liu Z, Pak JW, Blodi BA, Bailey S, Gehrs K, Wallace R, Tinker L, Mares JA. Reticular Pseudodrusen Characteristics and Associations in the Carotenoids in Age-Related Eye Disease Study 2 (CAREDS2), an Ancillary Study of the Women's Health Initiative. Ophthalmol Retina 2021; 5:721-729. [PMID: 33387684 PMCID: PMC8243566 DOI: 10.1016/j.oret.2020.12.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 12/23/2020] [Indexed: 01/09/2023]
Abstract
PURPOSE To determine the prevalence and morphologic features of reticular pseudodrusen (RPD) and their association with participant demographics and age-related macular degeneration (AMD) status in the Carotenoids in Age-Related Eye Disease Study 2 (CAREDS2) sample, an ancillary study of the Women's Health Initiative Observational Study. DESIGN Cross-sectional, multicenter, natural history study. PARTICIPANTS Nine hundred and twenty-seven eyes from 466 postmenopausal women 69 to 101 years of age. METHODS Multimodal imaging, including spectral-domain (SD) OCT and infrared reflectance (IR), were used to identify RPD characteristics, including location (within or outside the 6-mm diameter circle centered at the macula), presence of peripapillary RPD, pattern of RPD, and RPD area. Age-related macular degeneration features from SD OCT, IR, and color photographs also were assessed and AMD severity was categorized. MAIN OUTCOME MEASURES Reticular pseudodrusen prevalence using SD OCT and IR imaging and AMD status. RESULTS Reticular pseudodrusen were present in 130 eyes (14% of eyes, 16% of participants), with increasing prevalence with age: 7% in those younger than 78 years, 14% in those 78 to 83 years of age, and 30% in those older than 83 years. Using clinical classification of AMD with color photography, RPD were seen in 2.4% of eyes with no AMD or aging changes, 11.5% in early AMD, 25.1% in intermediate AMD, and 51.1% in late AMD. Mean RPD area was 17.4 mm2 (standard deviation, 14.7 mm2). Ribbon morphologic RPD (53%) was more common than dot morphologic RPD (36%). Reticular pseudodrusen mostly were located both within and outside the 6-mm circle with primarily superior retinal distribution. Reticular pseudodrusen were visualized with corresponding color fundus photography in only 38 eyes (4% of total eyes). Participants with and without RPD had a visual acuity±standard error of 77.9 ± 1.4 letters and 81.3 ± 0.4 letters, respectively (P = 0.02). CONCLUSIONS The prevalence of RPD in CAREDS2 increased with age and was associated with AMD severity. Reticular pseudodrusen were detected in eyes without other features of AMD and could represent an earlier disease state. Multimodal imaging with SD OCT and IR has significantly greater sensitivity for visualizing RPD than color fundus photography.
Collapse
Affiliation(s)
- Spencer C Cleland
- Department of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, Wisconsin
| | - Amitha Domalpally
- Department of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, Wisconsin.
| | - Zhe Liu
- Department of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, Wisconsin
| | - Jeong W Pak
- Department of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, Wisconsin
| | - Barbara A Blodi
- Department of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, Wisconsin
| | - Steven Bailey
- Department of Ophthalmology, Oregon Health & Science University, Casey Eye Institute, Portland, Oregon
| | - Karen Gehrs
- Department of Ophthalmology & Visual Sciences, University of Iowa Hospital & Clinics, Iowa City, Iowa
| | - Robert Wallace
- Department of Epidemiology, University of Iowa College of Public Health, Iowa City, Iowa
| | - Lesley Tinker
- Division of Public Health Sciences, Department of Cancer Prevention Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Julie A Mares
- Department of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
21
|
Brinks J, van Dijk EHC, Klaassen I, Schlingemann RO, Kielbasa SM, Emri E, Quax PHA, Bergen AA, Meijer OC, Boon CJF. Exploring the choroidal vascular labyrinth and its molecular and structural roles in health and disease. Prog Retin Eye Res 2021; 87:100994. [PMID: 34280556 DOI: 10.1016/j.preteyeres.2021.100994] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/04/2021] [Accepted: 07/07/2021] [Indexed: 12/14/2022]
Abstract
The choroid is a key player in maintaining ocular homeostasis and plays a role in a variety of chorioretinal diseases, many of which are poorly understood. Recent advances in the field of single-cell RNA sequencing have yielded valuable insights into the properties of choroidal endothelial cells (CECs). Here, we review the role of the choroid in various physiological and pathophysiological mechanisms, focusing on the role of CECs. We also discuss new insights regarding the phenotypic properties of CECs, CEC subpopulations, and the value of measuring transcriptomics in primary CEC cultures derived from post-mortem eyes. In addition, we discuss key phenotypic, structural, and functional differences that distinguish CECs from other endothelial cells such as retinal vascular endothelial cells. Understanding the specific clinical and molecular properties of the choroid will shed new light on the pathogenesis of the broad clinical range of chorioretinal diseases such as age-related macular degeneration, central serous chorioretinopathy and other diseases within the pachychoroid spectrum, uveitis, and diabetic choroidopathy. Although our knowledge is still relatively limited with respect to the clinical features and molecular pathways that underlie these chorioretinal diseases, we summarise new approaches and discuss future directions for gaining new insights into these sight-threatening diseases and highlight new therapeutic strategies such as pluripotent stem cell‒based technologies and gene therapy.
Collapse
Affiliation(s)
- J Brinks
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands
| | - E H C van Dijk
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands
| | - I Klaassen
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - R O Schlingemann
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands; Department of Ophthalmology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands; Department of Ophthalmology, University of Lausanne, Jules Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - S M Kielbasa
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, Leiden, the Netherlands
| | - E Emri
- Department of Clinical Genetics, Section of Ophthalmogenetics, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - P H A Quax
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - A A Bergen
- Department of Clinical Genetics, Section of Ophthalmogenetics, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - O C Meijer
- Department of Medicine, Division of Endocrinology and Metabolism, Leiden University Medical Center, Leiden, the Netherlands
| | - C J F Boon
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands; Department of Ophthalmology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
22
|
Khan AH, Sutton J, Cree AJ, Khandhadia S, De Salvo G, Tobin J, Prakash P, Arora R, Amoaku W, Charbel Issa P, MacLaren RE, Bishop PN, Peto T, Mohamed Q, Steel DH, Sivaprasad S, Bailey C, Menon G, Kavanagh D, Lotery AJ. Prevalence and phenotype associations of complement factor I mutations in geographic atrophy. Hum Mutat 2021; 42:1139-1152. [PMID: 34153144 PMCID: PMC9290714 DOI: 10.1002/humu.24242] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/09/2021] [Accepted: 06/13/2021] [Indexed: 12/20/2022]
Abstract
Rare variants in the complement factor I (CFI) gene, associated with low serum factor I (FI) levels, are strong risk factors for developing the advanced stages of age-related macular degeneration (AMD). No studies have been undertaken on the prevalence of disease-causing CFI mutations in patients with geographic atrophy (GA) secondary to AMD. A multicenter, cross-sectional, noninterventional study was undertaken to identify the prevalence of pathogenic rare CFI gene variants in an unselected cohort of patients with GA and low FI levels. A genotype-phenotype study was performed. Four hundred and sixty-eight patients with GA secondary to AMD were recruited to the study, and 19.4% (n = 91) demonstrated a low serum FI concentration (below 15.6 μg/ml). CFI gene sequencing on these patients resulted in the detection of rare CFI variants in 4.7% (n = 22) of recruited patients. The prevalence of CFI variants in patients with low serum FI levels and GA was 25%. Of the total patients recruited, 3.2% (n = 15) expressed a CFI variant classified as pathogenic or likely pathogenic. The presence of reticular pseudodrusen was detected in all patients with pathogenic CFI gene variants. Patients with pathogenic CFI gene variants and low serum FI levels might be suitable for FI supplementation in therapeutic trials.
Collapse
Affiliation(s)
- Adnan H Khan
- Division of Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,Southampton Eye Unit, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Janice Sutton
- Division of Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Angela J Cree
- Division of Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Samir Khandhadia
- Southampton Eye Unit, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Gabriella De Salvo
- Southampton Eye Unit, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - John Tobin
- Gyroscope Therapeutics Limited, Stevenage, UK
| | - Priya Prakash
- The Eye Unit, The Princess Alexandra Hospital NHS Trust, Harlow, UK
| | - Rashi Arora
- Department of Ophthalmology, Salisbury District Hospital, Salisbury NHS Foundation Trust, Salisbury, UK
| | - Winfried Amoaku
- Eye and ENT Centre, Queen's Medical Centre, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Peter Charbel Issa
- Oxford Eye Hospital and Oxford NIHR Biomedical Research Centre, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.,Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Robert E MacLaren
- Oxford Eye Hospital and Oxford NIHR Biomedical Research Centre, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.,Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Paul N Bishop
- Division of Evolution and Genomic Sciences, Faculty of Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, UK.,Manchester Royal Eye Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Tunde Peto
- Centre for Public Health, School of Medicine, Institute of Clinical Sciences, Queen's University Belfast, Belfast, UK
| | - Quresh Mohamed
- Department of Ophthalmology, Gloucestershire Royal Hospital, Gloucestershire Hospitals NHS Foundation Trust, Gloucester, UK
| | - David H Steel
- Sunderland Eye Infirmary, South Tyneside and Sunderland NHS Foundation Trust, Sunderland, UK.,Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Sobha Sivaprasad
- Institute of Ophthalmology, University College London, London, UK
| | - Clare Bailey
- Clinical Research Unit, Bristol Eye Hospital, University Hospitals Bristol NHS Foundation Trust, Bristol, UK
| | - Geeta Menon
- Department of Ophthalmology, Frimley Park Hospital, Frimley Health NHS Foundation Trust, Camberley, UK
| | - David Kavanagh
- National Renal Complement Therapeutics Centre, Royal Victoria Infirmary, Newcastle upon Tyne, UK.,Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Andrew J Lotery
- Division of Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,Southampton Eye Unit, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| |
Collapse
|
23
|
Scholl HPN, Boyer D, Giani A, Chong V. The use of neuroprotective agents in treating geographic atrophy. Ophthalmic Res 2021; 64:888-902. [PMID: 34153966 DOI: 10.1159/000517794] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 05/12/2021] [Indexed: 11/19/2022]
Affiliation(s)
- Hendrik P N Scholl
- Institute of Molecular and Clinical Ophthalmology, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - David Boyer
- Retina-Vitreous Associates Medical Group, Los Angeles, California, USA
| | - Andrea Giani
- Boehringer Ingelheim International GmbH, Ingelheim am Rhein, Germany
| | - Victor Chong
- Boehringer Ingelheim International GmbH, Ingelheim am Rhein, Germany
| |
Collapse
|
24
|
Sparrow JR, Parmann R, Tsang SH, Allikmets R, Chang S, Jauregui R. Shared Features in Retinal Disorders With Involvement of Retinal Pigment Epithelium. Invest Ophthalmol Vis Sci 2021; 62:15. [PMID: 34115091 PMCID: PMC8196415 DOI: 10.1167/iovs.62.7.15] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
When using spectral domain optical coherence tomography (SD-OCT) to inform the status of outer retina, we have noted discrete hyperreflective lesions extending through photoreceptor-attributable bands that have a similar presentation in multiple retinal diseases. These lesions present as either corrugated thickenings of interdigitation zone and ellipsoid zone bands or in later stages as rectangular or pyramidal shaped foci that extend radially through photoreceptor cell-attributable bands. In ABCA4-related and peripherin-2/RDS-disease (PRPH2/RDS), monogenic forms of retinopathy caused by mutations in proteins expressed in photoreceptor cells, these punctate lesions colocalize with fundus flecks in en face images. In fundus albipunctatus and retinitis punctata albescens, diseases caused by mutations in genes (retinol dehydrogenase 5, RDH5; and retinaldehyde-binding protein 1, RLBP1) encoding proteins of the visual cycle, these lesions manifest as white dot-like puncta. Similar aberrations in photoreceptor cell-attributable SD-OCT reflectivity layers manifest as reticular pseudodrusen (RPD) in short-wavelength fundus autofluorescence and near-infrared fundus autofluorescence fundus images and are linked to age-related macular degeneration a complex disease. Despite differences in the etiologies of retinal diseases presenting as fundus flecks, dots and RPD, underlying degenerative processes in photoreceptor cells are signified in SD-OCT scans by the loss of structural features that would otherwise define healthy photoreceptor cells at these foci.
Collapse
Affiliation(s)
- Janet R Sparrow
- Department of Ophthalmology, Harkness Eye Institute, Columbia University, New York, New York, United States.,Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York, United States
| | - Rait Parmann
- Department of Ophthalmology, Harkness Eye Institute, Columbia University, New York, New York, United States
| | - Stephen H Tsang
- Department of Ophthalmology, Harkness Eye Institute, Columbia University, New York, New York, United States.,Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York, United States
| | - Rando Allikmets
- Department of Ophthalmology, Harkness Eye Institute, Columbia University, New York, New York, United States.,Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York, United States
| | - Stanley Chang
- Department of Ophthalmology, Harkness Eye Institute, Columbia University, New York, New York, United States
| | - Ruben Jauregui
- Department of Ophthalmology, Harkness Eye Institute, Columbia University, New York, New York, United States
| |
Collapse
|
25
|
Kumawat D, Padhy SK, Kumar V. Clinical and Multimodal Imaging Features of Subretinal Drusenoid Deposits. J Ophthalmic Vis Res 2021; 16:187-194. [PMID: 34055256 PMCID: PMC8126748 DOI: 10.18502/jovr.v16i2.9082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 01/16/2021] [Indexed: 11/24/2022] Open
Abstract
Purpose To describe the multimodal imaging (MMI) features of subretinal drusenoid deposits (SDD) in Indian population. Methods Patients diagnosed to have SDD from January 2016 to December 2018 at our tertiary care center were recruited. The diagnosis of SDD was made on the basis of MMI consisting of a combination of color fundus photography (CFP), optical coherence tomography (OCT), red-free (RF) imaging, blue autofluorescence (BAF), and near-infra red reflectance (NIR) imaging. The morphological type and distribution of SDD and the associated retinal lesions were reviewed. Results Twenty-three patients with SDD were included. The mean age of the patients was 68.1 ± 12.2 years. SDD were noted in 77.8% of eyes clinically (n = 35/45) and could be detected in 100% of these eyes with OCT. The morphology of SDD was nodular in 65.7% of eyes (n = 23/35), reticular in 5.7% (n = 2/35), and mixed pattern in the remaining cases. BAF and NIR showed hyporeflective nodular lesions often with a target configuration. The location was commonly in the perifoveal area, mostly involving the superotemporal quadrant (74.3%, n = 26/35). Associated retinal lesions were type-3 neovascularization or retinal angiomatous proliferation in 17.1% (n = 6/35), disciform scar in 11.4% (n = 4/35), type-1 neovascularization in 8.5% (n = 3/35), and geographic atrophy in 5.7% (n = 2/35) of eyes. The mean subfoveal choroidal thickness was 186.2 ± 57.8 µm. Conclusion SDD commonly have a nodular morphology and their identification often requires confirmations with OCT. Advanced age-related macular degeneration features are frequently present in eyes with SDD and the fellow eyes.
Collapse
Affiliation(s)
- Devesh Kumawat
- Dr Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, India
| | - Srikanta K Padhy
- Dr Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, India
| | - Vinod Kumar
- Dr Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
26
|
Laíns I, Wang JC, Cui Y, Katz R, Vingopoulos F, Staurenghi G, Vavvas DG, Miller JW, Miller JB. Retinal applications of swept source optical coherence tomography (OCT) and optical coherence tomography angiography (OCTA). Prog Retin Eye Res 2021; 84:100951. [PMID: 33516833 DOI: 10.1016/j.preteyeres.2021.100951] [Citation(s) in RCA: 117] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 01/17/2021] [Accepted: 01/20/2021] [Indexed: 02/08/2023]
Abstract
The advent of optical coherence tomography (OCT) revolutionized both clinical assessment and research of vitreoretinal conditions. Since then, extraordinary advances have been made in this imaging technology, including the relatively recent development of swept-source OCT (SS-OCT). SS-OCT enables a fast scan rate and utilizes a tunable swept laser, thus enabling the incorporation of longer wavelengths than conventional spectral-domain devices. These features enable imaging of larger areas with reduced motion artifact, and a better visualization of the choroidal vasculature, respectively. Building on the principles of OCT, swept-source OCT has also been applied to OCT angiography (SS-OCTA), thus enabling a non-invasive in depth-resolved imaging of the retinal and choroidal microvasculature. Despite their advantages, the widespread use of SS-OCT and SS-OCTA remains relatively limited. In this review, we summarize the technical details, advantages and limitations of SS-OCT and SS-OCTA, with a particular emphasis on their relevance for the study of retinal conditions. Additionally, we comprehensively review relevant studies performed to date to the study of retinal health and disease, and highlight current gaps in knowledge and opportunities to take advantage of swept source technology to improve our current understanding of many medical and surgical chorioretinal conditions. We anticipate that SS-OCT and SS-OCTA will continue to evolve rapidly, contributing to a paradigm shift to more widespread adoption of new imaging technology to clinical practice.
Collapse
Affiliation(s)
- Inês Laíns
- Retina Service, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Harvard Retinal Imaging Lab, Boston, MA, USA
| | - Jay C Wang
- Retina Service, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Harvard Retinal Imaging Lab, Boston, MA, USA
| | - Ying Cui
- Retina Service, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Harvard Retinal Imaging Lab, Boston, MA, USA; Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Raviv Katz
- Retina Service, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Harvard Retinal Imaging Lab, Boston, MA, USA
| | - Filippos Vingopoulos
- Retina Service, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Harvard Retinal Imaging Lab, Boston, MA, USA
| | - Giovanni Staurenghi
- Eye Clinic, Department of Biomedical and Clinical Science "Luigi Sacco", University of Milan, Italy
| | - Demetrios G Vavvas
- Retina Service, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Joan W Miller
- Retina Service, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - John B Miller
- Retina Service, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Harvard Retinal Imaging Lab, Boston, MA, USA.
| |
Collapse
|
27
|
Dutheil C, Le Goff M, Cougnard-Grégoire A, Gattoussi S, Korobelnik JF, Rougier MB, Schweitzer C, Delcourt C, Delyfer MN. Incidence and Risk Factors of Reticular Pseudodrusen Using Multimodal Imaging. JAMA Ophthalmol 2021; 138:467-477. [PMID: 32163116 DOI: 10.1001/jamaophthalmol.2020.0266] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Importance Although retinal multimodal imaging is needed for diagnosing reticular pseudodrusen (RPD), the incidence of RPD in the general population typically has been assessed only using fundus photographs, which may underestimate their incidence. Objectives To describe the incidence of RPD using retinal color photographs, spectral-domain optical coherence tomography scans, fundus autofluorescence, and near-infrared reflectance images among individuals 77 years of age or older and to analyze the associated risk factors of RPD. Design, Setting, and Participants The ALIENOR (Antioxydants, Lipides Essentiels, Nutrition et Maladies Oculaires) Study is a cohort of French individuals 77 years of age or older. Data for this study were collected between February 22, 2011, and February 15, 2017, with a mean (SD) follow-up of 3.7 (1.0) years (range, 1.2-5.6 years). At baseline, 501 individuals were eligible to participate. Of 1002 eyes, 197 had prevalent RPD, advanced age-related macular degeneration, or ungradable images. Of the remaining 805 eyes, 333 were missing follow-up data; therefore, the statistical analyses included data from 472 eyes. Data management and statistical analyses were performed between March 15, 2017, and April 5, 2019. Main Outcomes and Measures Reticular pseudodrusen were considered as present if detected by at least 2 of the following imaging methods: color fundus photographs, fundus autofluorescence, near-infrared reflectance, and spectral-domain optical coherence tomography images. Results Of the 472 eyes analyzed, 263 (55.7%) were from female participants, and the mean (SD) age was 81.9 (3.2) years. Forty-three eyes developed RPD, corresponding to an annual incidence rate of 2.9% (95% CI, 1.9%-4.4%) per participant and an estimated 5-year risk of 13.5%. In multivariable analysis, 4 risk factors of incident RPD were identified: subfoveal choroidal thinning (hazard ratio [HR], 0.99; 95% CI, 0.99-1.00 per 10-μm decrease in thickness; P = .02) and the presence of the minor allelic variants rs10490924 for ARMS2 (HR, 3.57; 95% CI, 1.80-7.10; P < .001), rs1061170 for CFH (HR, 2.12; 95% CI, 1.02-4.41; P = .04), and rs10468017 for LIPC (HR, 2.57; 95% CI, 1.37-4.82; P = .003). Lipophilic statin therapy was associated with a lower incidence of RPD (HR, 0.13; 95% CI, 0.02-0.74; P = .02). Conclusions and Relevance With the use of multimodal imaging, the RPD incidence rate was higher than previously reported in other population-based studies using fundus color images. Individuals with subfoveal choroidal thinning or carrying minor allelic variants for ARMS2, CFH, or LIPC had an increased risk for RPD, whereas lipophilic statin therapy was associated with a lower incidence.
Collapse
Affiliation(s)
- Cyril Dutheil
- Lifelong Exposure Health and Aging Team, Inserm, Bordeaux Population Health Research Center, Université de Bordeaux, Bordeaux, France.,Service d'Ophtalmologie, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Mélanie Le Goff
- Lifelong Exposure Health and Aging Team, Inserm, Bordeaux Population Health Research Center, Université de Bordeaux, Bordeaux, France
| | - Audrey Cougnard-Grégoire
- Lifelong Exposure Health and Aging Team, Inserm, Bordeaux Population Health Research Center, Université de Bordeaux, Bordeaux, France
| | - Sarra Gattoussi
- Lifelong Exposure Health and Aging Team, Inserm, Bordeaux Population Health Research Center, Université de Bordeaux, Bordeaux, France.,Service d'Ophtalmologie, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Jean-François Korobelnik
- Lifelong Exposure Health and Aging Team, Inserm, Bordeaux Population Health Research Center, Université de Bordeaux, Bordeaux, France.,Service d'Ophtalmologie, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Marie-Bénédicte Rougier
- Lifelong Exposure Health and Aging Team, Inserm, Bordeaux Population Health Research Center, Université de Bordeaux, Bordeaux, France.,Service d'Ophtalmologie, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Cédric Schweitzer
- Lifelong Exposure Health and Aging Team, Inserm, Bordeaux Population Health Research Center, Université de Bordeaux, Bordeaux, France.,Service d'Ophtalmologie, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Cécile Delcourt
- Lifelong Exposure Health and Aging Team, Inserm, Bordeaux Population Health Research Center, Université de Bordeaux, Bordeaux, France
| | - Marie-Noëlle Delyfer
- Lifelong Exposure Health and Aging Team, Inserm, Bordeaux Population Health Research Center, Université de Bordeaux, Bordeaux, France.,Service d'Ophtalmologie, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| |
Collapse
|
28
|
Chiang TTK, Keenan TD, Agrón E, Liao J, Klein B, Chew EY, Cukras CA, Wong WT. Macular Thickness in Intermediate Age-Related Macular Degeneration Is Influenced by Disease Severity and Subretinal Drusenoid Deposit Presence. Invest Ophthalmol Vis Sci 2021; 61:59. [PMID: 32602904 PMCID: PMC7415904 DOI: 10.1167/iovs.61.6.59] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose To investigate how macular thickness varies with intermediate age-related macular degeneration (iAMD) severity and the presence of subretinal drusenoid deposits (SDDs). Methods A longitudinal prospective study of 143 participants >50 years of age with no to intermediate AMD who were followed with multimodal imaging and functional testing. Participants were stratified by iAMD severity according to imaging features. Macular thicknesses measurements over the central circles with 1-mm, 3-mm, and 6-mm diameters obtained from ocular coherence tomography imaging were compared across severity categories using cross-sectional (143 eyes) and longitudinal (subset of 77 eyes followed for 4 years) multivariate analyses. Results Compared with control eyes without large drusen or SDDs (Group 0), central maculas of lower risk eyes with unilateral large drusen (Group 1) were thicker (P = 0.014), whereas higher risk eyes with SDDs (Group SDD) were thinner (P = 0.02) in cross-sectional multivariate analyses. In longitudinal analyses, maculas with SDDs thinned more rapidly over 4 years relative to control eyes (P = 0.0058), which did not show significant thinning. More rapid central macular thinning was associated with worse baseline best-corrected visual acuity (BCVA) (P = 0.016) and more rapid BCVA decline (P = 0.0059). Conclusions Macular thickness in iAMD varies with disease severity, showing small increases in eyes with large drusen and decreases in eyes with SDDs. Active processes possibly related to neuroinflammation and neurodegeneration may be contributory. Longitudinal central macular thickness evaluation is an accessible outcome measure relevant to functional measures and is potentially useful for iAMD interventional studies.
Collapse
|
29
|
Age-Related Macular Degeneration: Epidemiology and Clinical Aspects. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1256:1-31. [PMID: 33847996 DOI: 10.1007/978-3-030-66014-7_1] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Age-related macular degeneration (AMD) is a degenerative disease of the human retina affecting individuals over the age of 55 years. This heterogeneous condition arises from a complex interplay between age, genetics, and environmental factors including smoking and diet. It is the leading cause of blindness in industrialized countries. Worldwide, the number of people with AMD is predicted to increase from 196 million in 2020 to 288 million by 2040. By this time, Asia is predicted to have the largest number of people with the disease. Distinct patterns of AMD prevalence and phenotype are seen between geographical areas that are not explained fully by disparities in population structures. AMD is classified into early, intermediate, and late stages. The early and intermediate stages, when visual symptoms are typically absent or mild, are characterized by macular deposits (drusen) and pigmentary abnormalities. Through risk prediction calculators, grading these features helps predict the risk of progression to late AMD. Late AMD is divided into neovascular and atrophic forms, though these can coexist. The defining lesions are macular neovascularization and geographic atrophy, respectively. At this stage, visual symptoms are often severe and irreversible, and can comprise profoundly decreased central vision in both eyes. For these reasons, the condition has major implications for individuals and society, as affected individuals may experience substantially decreased quality of life and independence. Recent advances in retinal imaging have led to the recognition of an expanded set of AMD phenotypes, including reticular pseudodrusen, nonexudative macular neovascularization, and subtypes of atrophy. These developments may lead to refinements in current classification systems.
Collapse
|
30
|
Abstract
PURPOSE To analyze static characteristics and dynamic functionality of retinal vessels in eyes with drusen and reticular pseudodrusen (RPD) using dynamic vessel analyzer. METHODS Patients with clinical diagnosis of isolated RPD or medium-large drusen and healthy controls were enrolled in the study between July 2016 and May 2018. Participants underwent complete ophthalmologic examination, including enhanced depth imaging structural optical coherence tomography, dynamic retinal vessel analysis, and static retinal vessel analysis. RESULTS Twenty-eight eyes of 23 patients with drusen (9 men, mean age 77 ± 6 years), 22 eyes of 16 patients with RPD (7 men, mean age: 76 ± 6 years), and 22 eyes of 22 control subjects (11 men, mean age of 75 ± 6 years) were enrolled. Static retinal vessel analysis did not show any significant difference between the three groups for the central retinal artery equivalent (P = 0.11), the central retinal vein equivalent (P = 0.27), and the arteriovenous ratio (P = 0.30). Dynamic vessel analysis showed significantly reduced arterial dilation in eyes with drusen (P = 0.0001) and RPD (P = 0.015) compared with control subjects. No significant difference was seen between drusen and RPD groups (P = 0.32). Dynamic vessel analysis of retinal veins showed no differences between the three groups (P = 0.10). CONCLUSION Dynamic vessel analysis in eyes with drusen and RPD revealed an impaired retinal arterial dilation in response to flicker light stimulation, which further supports the relationship between cardiovascular risk and age-related macular degeneration.
Collapse
|
31
|
Otero-Marquez O, Chung H, Lee CS, Choi EY, Ledesma-Gil G, Alauddin S, Lee M, Bhuiyan A, Smith RT. Subretinal Deposits in Pre-eclampsia and Malignant Hypertension: Implications for Age-Related Macular Degeneration. Ophthalmol Retina 2020; 5:750-760. [PMID: 33130003 DOI: 10.1016/j.oret.2020.10.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 10/14/2020] [Accepted: 10/26/2020] [Indexed: 11/18/2022]
Abstract
PURPOSE To describe the incidence of subretinal deposits that are similar in structure and stage on OCT imaging to subretinal drusenoid deposits (SDDs) in age-related macular degeneration (AMD) in patients with hypertensive choroidopathy secondary to severe pre-eclampsia and malignant hypertension (MHT) and the implications of this ischemic choroidopathy for the pathophysiologic characteristics of SDDs in AMD. DESIGN Retrospective cross-sectional study. PARTICIPANTS Thirty-three pre-eclampsia patients and 25 MHT patients with serous retinal detachment (SRD) in at least 1 eye were included. METHODS Serial multimodal images, including enhanced depth imaging spectral-domain OCT of eyes with hypertensive choroidopathy secondary to pre-eclampsia and MHT, were reviewed at 2 time points, the acute phase (within 4 weeks of initial hypertensive insult) and the recovery phase (beyond 4 weeks). MAIN OUTCOME MEASURES Incidence of SDD-like lesions in patients with hypertensive choroidopathy secondary to pre-eclampsia and MHT. RESULTS Subretinal drusenoid deposit-like lesions were observed exclusively in eyes with SRD. Serous retinal detachment occurred in 87.87% of eyes of pre-eclampsia patients and in 94% of eyes of MHT patients. Subretinal drusenoid deposit-like lesions occurred in 28.57% of all eyes with SRD, in 32.76% of eyes with SRD from the pre-eclampsia group, and in 23.40% of eyes with SRD from the MHT group. Vascular imaging suggested underlying choroidal ischemia in all patients (12 eyes) in which it was performed. CONCLUSIONS Choroidal ischemia may be the underlying mechanism of SDD-like lesions in patients with pre-eclampsia and MHT choroidopathy. These findings potentially are of utmost importance in understanding the mechanism of the reticular macular disease subtype of AMD. Reticular macular disease is characterized by the known association of choroidal insufficiency and SDD, with choroidal insufficiency postulated, but not proven, to be causative. Pre-eclampsia and MHT choroidopathy seems to be a model for lesions similar to SDD in AMD developing based on choroidal insufficiency and, as such, may offer further insights into the pathoetiologic features of SDD in AMD.
Collapse
Affiliation(s)
- Oscar Otero-Marquez
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Hyewon Chung
- Department of Ophthalmology, Konkuk University School of Medicine, Konkuk University Medical Center, Seoul, Republic of Korea
| | - Christopher Seungkyu Lee
- Department of Ophthalmology, The Institute of Vision Research, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eun Young Choi
- Department of Ophthalmology, The Institute of Vision Research, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Gerardo Ledesma-Gil
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sharmina Alauddin
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Minsub Lee
- Department of Ophthalmology, Konkuk University School of Medicine, Konkuk University Medical Center, Seoul, Republic of Korea
| | - Alauddin Bhuiyan
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - R Theodore Smith
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
32
|
Charbel Issa P, Tysoe C, Caswell R. Late-onset Pseudoxanthoma Elasticum Associated with a Hypomorphic ABCC6 Variant. Am J Ophthalmol 2020; 218:255-260. [PMID: 32442430 DOI: 10.1016/j.ajo.2020.05.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 12/18/2022]
Abstract
PURPOSE To describe patients with late-onset pseudoxanthoma elasticum (PXE) associated with a likely hypomorphic ABCC6 variant. DESIGN Retrospective observational case series. METHODS Clinical evaluation, multimodal retinal imaging, genetic testing, and molecular modeling. RESULTS Three patients, in whom vision symptoms first arose at 80 years of age or later, showed age-related macular degeneration (AMD)-like fundus changes. However, features characteristic of PXE, including discrete angioid streaks and reduced fluorescence on late-phase indocyanine green angiography, prompted genetic testing which revealed the c.1171A>G variant in combination with a large deletion in the ABCC6 gene in each case. None of the patients had obvious skin changes or cardiovascular disease atypical for their age. Comparative molecular modeling supported the hypothesis that the c.1171A>GABCC6 variant acted as a hypomorphic variant. CONCLUSIONS Late-onset PXE extends the spectrum of ectopic calcification disorders caused by mutations in ABCC6 and may clinically be limited to the eye, mimicking AMD. Patients may be identified based on specific ocular changes, whereas skin and cardiovascular changes may remain ambiguous. The study provides evidence for a role for hypomorphic ABCC6 variants in the pathogenesis of PXE.
Collapse
|
33
|
Lee SE, Lim HB, Shin YI, Ryu CK, Lee WH, Kim JY. Characteristics of the inner retinal layer in the fellow eyes of patients with unilateral exudative age-related macular degeneration. PLoS One 2020; 15:e0239555. [PMID: 32966311 PMCID: PMC7511006 DOI: 10.1371/journal.pone.0239555] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 09/08/2020] [Indexed: 11/01/2022] Open
Abstract
OBJECTIVE To investigate the thicknesses of the ganglion cell-inner plexiform layer (GC-IPL) and retinal nerve fiber layer (RNFL) of the fellow eyes of patients with unilateral exudative age-related macular degeneration (AMD). METHODS A total of 107 patients with unilateral exudative AMD [34 of typical choroidal neovascularization (tCNV), Group A; 73 of polypoidal choroidal vasculopathy (PCV), Group B] and 73 normal control eyes (Group C) were included. Drusen and subretinal drusenoid deposits were assessed in all participants using fundus photography, autofluorescence, and optical coherence tomography (OCT). The GC-IPL and RNFL thicknesses were measured using Cirrus HD-OCT and compared among groups. Linear regression analyses were used to evaluate the factors associated with GC-IPL thicknesses. RESULTS The average GC-IPL thicknesses of Groups A, B, and C were 77.09 ± 3.87, 80.10 ± 6.61, and 80.88 ± 6.50 μm, respectively (p = 0.022). Sectoral GC-IPLs and central macular thicknesses (CMTs) were significantly different among groups (all, p <0.05), whereas none of the RNFL parameters differed significantly (all, p >0.05). Multivariate linear regression analyses revealed that age (p <0.001), CMT (p <0.001), and tCNV (p = 0.013) were significantly associated with average GC-IPL thickness, and the rate of reduction of GC-IPL thickness with increasing age in the fellow eyes of tCNV patients was higher than those in the PCV and control groups. CONCLUSIONS Unilateral tCNV patients exhibited statistically significant reduction of the GC-IPL thickness in the fellow eyes, compared to values of the fellow eyes of unilateral PCV patients or control patients. RNFL values trended to be lower but did not reach statistical significance.
Collapse
Affiliation(s)
- Seong Eun Lee
- Department of Ophthalmology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Hyung Bin Lim
- Department of Ophthalmology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Yong Il Shin
- Department of Ophthalmology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Rhee’s Eye Hospital, Daejeon, Republic of Korea
| | - Cheon Kuk Ryu
- Department of Ophthalmology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Woo Hyuk Lee
- Department of Ophthalmology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Jung-Yeul Kim
- Department of Ophthalmology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- * E-mail:
| |
Collapse
|
34
|
Akyol E, Lotery A. Gene, Cell and Antibody-Based Therapies for the Treatment of Age-Related Macular Degeneration. Biologics 2020; 14:83-94. [PMID: 32982165 PMCID: PMC7494004 DOI: 10.2147/btt.s252581] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/04/2020] [Indexed: 12/30/2022]
Abstract
Here we discuss antibody, cell and gene-based therapies that are currently available and under investigation for both wet and dry age-related macular degeneration (AMD). We initially discuss ocular anatomy, AMD modelling as well as the underlying pathophysiology of AMD. The antibody-based trials which have revolutionised the management of wet AMD are reviewed. The latest concepts in antibody therapy for wet AMD such as the port delivery systems, bispecific antibodies, designed ankyrin repeat protein (DARPINs) and brolucizumab are explored. Furthermore, the antibody-based trials targeting the complement pathway to reduce progression of geographic atrophy (GA) in dry AMD are discussed. Stem cell therapy and gene therapy are novel treatment modalities with no established clinical use in wet or dry AMD. Here, we discuss their efficacy so far in clinical trials. Their benefits and risk in the treatment of both wet and dry AMD are evaluated.
Collapse
Affiliation(s)
- Engin Akyol
- Clinical Neurosciences Research Group, Faculty of Medicine, University of Southampton, Southampton General Hospital, SouthamptonSO16 6YD, UK
| | - Andrew Lotery
- Clinical Neurosciences Research Group, Faculty of Medicine, University of Southampton, Southampton General Hospital, SouthamptonSO16 6YD, UK
| |
Collapse
|
35
|
Proteoglycan IMPG2 Shapes the Interphotoreceptor Matrix and Modulates Vision. J Neurosci 2020; 40:4059-4072. [PMID: 32265257 DOI: 10.1523/jneurosci.2994-19.2020] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/29/2020] [Accepted: 03/19/2020] [Indexed: 12/13/2022] Open
Abstract
Photoreceptor neurons are surrounded by an extracellular matrix, called the interphotoreceptor matrix (IPM). Activities crucial to vision occur within the IPM, including trafficking of nutrients and metabolites, retinal attachment, and interactions needed for normal outer segment phagocytosis. The IPM includes the following two unique proteoglycans: IPM proteoglycan 1 (IMPG1) and IMPG2. Patients with mutations in IMPG1/IMPG2 develop visual deficits with subretinal material accumulation, highlighting the critical role of the IPM in vision. To determine the role of these proteoglycans in retinal physiology and the pathologic mechanisms that lead to vision loss, we generated mouse models lacking IMPG1/IMPG2. In normal retina, IMPG1 and IMPG2 occupy distinct IPM compartments, represent the main source of chondroitin sulfate and are fundamental for the constitution of the cone-specific glycocalyx stained by the PNA (peanut agglutinin) lectin marker. No evident morphologic or functional deficits were found in mice lacking IMPG1. In the absence of IMPG2, IMPG1 abnormally accumulated at the subretinal space need, likely leading to the formation of subretinal lesions and reduced visual function. Interestingly, mice lacking both IMPG1 and IMPG2, regardless of sex, showed normal retinal structure and function, demonstrating that the aberrant IMPG1 distribution is the main cause of the visual alterations observed in the absence of IMPG2. In conclusion, our results show the dependence of secreted proteoglycans such as IMPG1 on the extracellular environment to properly integrate into the matrix, demonstrate the role of IMPG2 in shaping the IPM, and shed light on the potential mechanisms leading to the development of subretinal lesions and vision loss.SIGNIFICANCE STATEMENT The photoreceptors are specialized neurons that drive phototransduction in the mammalian retina. These cells are organized and surrounded by an extracellular matrix, the interphotoreceptor matrix (IPM). Mutations in IPM proteoglycans are associated with blindness in humans. Our studies show that two specific proteoglycans of the IPM, IPM proteoglycan 1 (IMPG1) and IMPG2, form a dynamic structure with distinct localization and dependency. When IMPG2 is absent, IMPG1 cannot integrate into the IPM, leading to abnormal proteoglycan accumulation and visual deficits. This work adds a new layer of understanding to IPM physiology and describes the pathologic events following deficits in proteoglycans, providing novel possibilities for visual restoration in patients with IMPG-related pathologies.
Collapse
|
36
|
A Pilot Study Evaluating the Effects of 670 nm Photobiomodulation in Healthy Ageing and Age-Related Macular Degeneration. J Clin Med 2020; 9:jcm9041001. [PMID: 32252424 PMCID: PMC7231137 DOI: 10.3390/jcm9041001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 03/25/2020] [Accepted: 03/31/2020] [Indexed: 12/28/2022] Open
Abstract
Limited evidence suggests that the application of 670 nm of red light alters the course of aged decline. A previous report on 18 patients showed regression of drusen and improvement in visual functions in patients with intermediate age-related macular degeneration (AMD) by 12 months. We evaluated the functional and structural effects of applying 670 nm light to 31 patients with intermediate AMD and 11 people aged 55 years or above with normal retina. The study eyes were treated daily in the morning with a 670 nm hand-held light source housed in a torch-like tube that emitted energy equivalent to 40 mW/cm2 or 4.8J/ cm2 for 2 min at the viewing aperture. Visual function in terms of best-corrected visual acuity, low luminance visual acuity, scotopic thresholds and rod-intercept time were compared between baseline and 1, 3, 6 and 12 months. Structural changes on optical coherence tomography OCT and colour photographs were also assessed. Five withdrew consent voluntarily due to the intensity of the study visit assessments and two developed neovascular AMD and were excluded from further treatment and the analysis. In normal ageing, there was an improvement in scotopic thresholds in the group with no AMD by 1.77dB (p = 0.03) and no other parameters showed any clinically significant change. In eyes with intermediate AMD, there was no significant improvement in any functional or structural changes at any time point up to 12 months although the compliance was good. This pilot study shows that photobiomodulation with 670 nm has no effect in patients who have already progressed to intermediate AMD.
Collapse
|
37
|
Abdolrahimzadeh S, Fameli V, Tizio FD, Staso FD, Fenicia V, Scuderi G. The Effect of Topical Bromfenac on Intraretinal and Subretinal Fluid in Neovascular Age-Related Macular Degeneration. J Curr Ophthalmol 2020; 32:203-206. [PMID: 32671307 PMCID: PMC7337015 DOI: 10.4103/joco.joco_105_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 10/26/2019] [Accepted: 11/10/2019] [Indexed: 11/05/2022] Open
Abstract
Purpose: To report the effect of topical bromfenac, a non-steroidal anti-inflammatory drug (NSAID), in a case of neovascular age-related macular degeneration (AMD). Methods: An 85-year-old woman presented with a complaint of visual acuity reduction in the right eye. Comprehensive ophthalmological examination and retinal imaging were performed. Results: Best corrected visual acuity was 2/100. Fundus examination showed reticular pseudodrusen and a small hemorrhage in the fovea. Fluorescein angiography showed an active neovascular membrane. Spectral-domain optical coherence tomography (SD-OCT) confirmed diagnosis and revealed subretinal and intraretinal fluid. The patient refused recommended intravitreal anti-vascular endothelial growth factor treatment and received topical bromfenac 0.09% twice daily. Follow-up with SD-OCT showed subretinal followed by intraretinal fluid reduction at 16 weeks after treatment. Conclusion: Short-term reduction of subretinal and intraretinal fluid was observed with topical bromfenac monotherapy in neovascular AMD.
Collapse
Affiliation(s)
- Solmaz Abdolrahimzadeh
- Ophthalmology Unit, Neurosciences, Mental Health and Sensory Organs Department, St. Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Valeria Fameli
- Ophthalmology Unit, Department of Sense Organs, Sapienza University of Rome, Rome, Italy
| | - Federico Di Tizio
- Ophthalmology Unit, Neurosciences, Mental Health and Sensory Organs Department, St. Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Federico Di Staso
- Ophthalmology Unit, Neurosciences, Mental Health and Sensory Organs Department, St. Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Vito Fenicia
- Ophthalmology Unit, Neurosciences, Mental Health and Sensory Organs Department, St. Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Gianluca Scuderi
- Ophthalmology Unit, Neurosciences, Mental Health and Sensory Organs Department, St. Andrea Hospital, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
38
|
Gabrielle PH, Seydou A, Arnould L, Acar N, Devilliers H, Baudin F, Ben Ghezala I, Binquet C, Bron AM, Creuzot-Garcher C. Subretinal Drusenoid Deposits in the Elderly in a Population-Based Study (the Montrachet Study). Invest Ophthalmol Vis Sci 2019; 60:4838-4848. [PMID: 31747683 DOI: 10.1167/iovs.19-27283] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose The aim of this study was to investigate the prevalence of subretinal drusenoid deposits (SDD) and to identify associated factors in an elderly population. Methods The participants of the population-based Montrachet study underwent an exhaustive ophthalmologic examination, including color fundus photography and macular spectral domain-optical coherence tomography (SD-OCT), coupled with infrared reflectance imaging. The presence of SDD and other age-related macular degeneration lesions, according to the European Eye Epidemiology SD-OCT classification of macular diseases, and subfoveal choroidal thickness were recorded. Moreover, the association of SDD and both clinical and demographic factors as well as plasma levels of vitamin E and lutein/zeaxanthin (L/Z) were analyzed. Results The mean age of patients was 82.3 ± 3.8 years and 62.7% were female. The prevalence of SDD was 18.1% (n = 205) in the subjects with at least one eye interpretable (n = 1135). In multivariate analysis, SDD was positively associated with increasing age (OR, 4.6; 95% CI, 2.8-7.7; P < 0.001 for subjects aged >85 years), female sex (OR, 1.7; 95% CI, 1.2-2.4; P = 0.005), and plasma L/Z level (OR, 1.2; 95% CI, 1.0-1.5; P = 0.039), and negatively associated with lipid-lowering drugs use (OR, 0.5; 95% CI, 0.3-0.9; P = 0.014 for statin medications) and subfoveal choroidal thickness (OR, 0.8; 95% CI, 0.7-0.9; P = 0.002). Conclusions The prevalence of SDD was high in subjects older than 75 years, more frequent in women, and was associated with a thinner choroid. The association with lipid-lowering drugs deserves further investigation.
Collapse
Affiliation(s)
- Pierre-Henry Gabrielle
- Department of Ophthalmology, University Hospital, Dijon, France.,Eye and Nutrition Research Group, CSGA, UMR 1324 INRA, 6265 CNRS, Burgundy University, Dijon, France
| | - Alassane Seydou
- Eye and Nutrition Research Group, CSGA, UMR 1324 INRA, 6265 CNRS, Burgundy University, Dijon, France.,Department of Epidemiology, INSERM unit, University Hospital, Dijon, France
| | - Louis Arnould
- Department of Ophthalmology, University Hospital, Dijon, France
| | - Niyazi Acar
- Eye and Nutrition Research Group, CSGA, UMR 1324 INRA, 6265 CNRS, Burgundy University, Dijon, France
| | - Hervé Devilliers
- Department of Epidemiology, INSERM unit, University Hospital, Dijon, France
| | - Florian Baudin
- Department of Ophthalmology, University Hospital, Dijon, France
| | | | - Christine Binquet
- Department of Epidemiology, INSERM unit, University Hospital, Dijon, France
| | - Alain Marie Bron
- Department of Ophthalmology, University Hospital, Dijon, France.,Eye and Nutrition Research Group, CSGA, UMR 1324 INRA, 6265 CNRS, Burgundy University, Dijon, France
| | - Catherine Creuzot-Garcher
- Department of Ophthalmology, University Hospital, Dijon, France.,Eye and Nutrition Research Group, CSGA, UMR 1324 INRA, 6265 CNRS, Burgundy University, Dijon, France
| |
Collapse
|
39
|
Domalpally A, Agrón E, Pak JW, Keenan TD, Ferris FL, Clemons TE, Chew EY. Prevalence, Risk, and Genetic Association of Reticular Pseudodrusen in Age-related Macular Degeneration: Age-Related Eye Disease Study 2 Report 21. Ophthalmology 2019; 126:1659-1666. [PMID: 31558345 DOI: 10.1016/j.ophtha.2019.07.022] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 07/19/2019] [Accepted: 07/23/2019] [Indexed: 11/30/2022] Open
Abstract
PURPOSE To determine the prevalence of reticular pseudodrusen (RPD) in eyes with age-related macular degeneration (AMD), assess the role of RPD as an independent risk factor for late AMD development, and evaluate genetic association with RPD. DESIGN Prospective cohort study. PARTICIPANTS Participants with intermediate AMD in 1 or both eyes enrolled in the Age-Related Eye Disease Study 2 (AREDS2), a 5-year multicenter study of nutritional supplement. METHODS Fundus autofluorescence (FAF) images from a subset of AREDS2 participants were evaluated at annual visits for presence of RPD. Six single nucleotide polymorphisms-rs10490924 (ARMS2), rs1061170 (CFH), rs2230199 (C3), rs116503776 and rs114254831 (C2/CFB), and rs943080 (VEGF-A)-and the genetic risk score (GRS) were assessed for association with RPD. Development of late AMD, defined as geographic atrophy (GA) or neovascular AMD (NVAMD), was identified. MAIN OUTCOME MEASURES Prevalence of RPD, odds ratio (OR) of late AMD development, and genetic associations of RPD. RESULTS The FAF images were evaluated for 5021 eyes (2516 participants). Reticular pseudodrusen were seen in 1186 eyes (24% of eyes, 29% of participants). Prevalence of RPD varied with baseline AREDS AMD severity level: 6% in early AMD (n = 458), 26% in intermediate AMD (n = 2606), 36% in GA (n = 682), and 19% in NVAMD (n = 1246). Mean age of participants with RPD was 79 years (standard deviation [SD], 7) and 75 years (SD, 8) in those without RPD (P < 0.0001). Reticular pseudodrusen were more frequent in female participants (65% RPD vs. 53% no RPD). Odds ratio adjusted for baseline age, gender, race, educational status, smoking, and AMD severity level for 1710 eyes at risk of developing late AMD at the next annual visit was 2.42 (95% confidence interval [CI], 1.80-3.24; P < 0.001) for GA and 1.21 (95% CI, 0.87-1.7; P = 0.26) for NVAMD. Presence of RPD was significantly associated with higher GRS (P < 0.0001) and ARMS2 risk alleles (P < 0.0001) and, at a nominal level, with C3 risk alleles (P = 0.04) and CFH risk alleles (P = 0.048 for homozygotes). CONCLUSIONS Participants with RPD have an increased risk of progression to GA but not NVAMD. ARMS2 risk alleles and higher GRS were associated with the presence of RPD. This study suggests that RPD are an important risk marker and should be included in classification systems used for patient prognosis.
Collapse
Affiliation(s)
- Amitha Domalpally
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin.
| | - Elvira Agrón
- Division of Epidemiology and Clinical Applications, National Eye Institute, Bethesda, Maryland
| | - Jeong W Pak
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin
| | - Tiarnan D Keenan
- Division of Epidemiology and Clinical Applications, National Eye Institute, Bethesda, Maryland
| | | | | | - Emily Y Chew
- Division of Epidemiology and Clinical Applications, National Eye Institute, Bethesda, Maryland
| |
Collapse
|
40
|
Nomura Y, Inoue T, Asano S, Shimizu-Asano K, Azuma K, Ogawa A, Murata H, Asaoka R, Obata R. Retinal sensitivity in angioid streaks. Graefes Arch Clin Exp Ophthalmol 2019; 257:1591-1599. [PMID: 31044269 DOI: 10.1007/s00417-019-04332-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 04/15/2019] [Accepted: 04/18/2019] [Indexed: 11/28/2022] Open
Abstract
PURPOSE To investigate retinal sensitivity in eyes with angioid streaks (AS). METHODS This study was a retrospective observational case series. A total of 13 eyes from 9 patients with AS underwent ophthalmologic examination, including fundus autofluorescence (FAF), spectral domain optical tomography (SD-OCT), and microperimetry. Retinal pigment epithelium (RPE) atrophy was assessed using FAF. Outer retinal atrophy, more specifically, disruption of the ellipsoid zone, was evaluated using SD-OCT images. The association between retinal sensitivity, RPE atrophy or outer retinal atrophy, and patient demographic characteristics or ophthalmologic findings were investigated. RESULTS The mean area of outer retinal atrophy was 14.5 ± 12.9 mm2, significantly larger than the mean area of RPE atrophy (9.0 ± 9.0 mm2; P = 0.0028). The average retinal sensitivity in the area of RPE atrophy was 2.4 ± 5.6 dB, that in the area of outer retinal atrophy outside RPE atrophy was 14.6 ± 7.5 dB, and that in the area without any atrophy was 25.6 ± 5.1 dB. Reticular pseudodrusen (RPD) was observed in 5 eyes. Eyes with RPD showed a greater extent of both outer retinal atrophy and RPE atrophy than those without RPD and a greater decrease in mean retinal sensitivity. CONCLUSIONS In eyes with AS, especially in those with RPD, large areas of outer retinal atrophy and RPE atrophy were observed, and the retinal sensitivity in the areas was correspondingly decreased.
Collapse
Affiliation(s)
- Yoko Nomura
- Department of Ophthalmology, University of Tokyo School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Tatsuya Inoue
- Department of Ophthalmology, University of Tokyo School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Shotaro Asano
- Department of Ophthalmology, University of Tokyo School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Kimiko Shimizu-Asano
- Department of Ophthalmology, University of Tokyo School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Keiko Azuma
- Department of Ophthalmology, University of Tokyo School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Asako Ogawa
- Department of Ophthalmology, University of Tokyo School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Hiroshi Murata
- Department of Ophthalmology, University of Tokyo School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Ryo Asaoka
- Department of Ophthalmology, University of Tokyo School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Ryo Obata
- Department of Ophthalmology, University of Tokyo School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| |
Collapse
|
41
|
Inan S, Baysal Z, Inan UU. Long-Term Changes in Submacular Choroidal Thickness after Intravitreal Ranibizumab Therapy for Neovascular Age-Related Macular Degeneration: 14-Mo Follow-Up. Curr Eye Res 2019; 44:908-915. [PMID: 30909756 DOI: 10.1080/02713683.2019.1600195] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Purpose: To study (i) the long-term effects of intravitreal ranibizumab treatment on changes in submacular choroidal thickness and (ii) the relationship between any resulting changes in choroidal thickness and visual outcomes following such treatment in patients with neovascular age-related macular degeneration (n-AMD). Methods: Reviewed were medical records of 30 consecutive, treatment-naïve, patients with unilateral n-AMD (n = 30 eyes) and unaffected fellow eyes (n = 26 eyes) (controls). Monthly injections of 0.5 mg ranibizumab were administered until stabilization of n-AMD, with additional injections as needed over the following 14-mo. Choroidal thickness was determined using enhanced-depth imaging-optical coherence tomography (EDI-OCT) before and after initiation of ranibizumab therapy. Choroidal thickness measurements were generated via manual segmentation. Results: The mean age of patients was 71.9 ± 7.4 (56-83) years; the mean best-corrected visual acuity (BCVA) of affected eyes improved from 51.1 to 59.4 letters (p < 0.001); and the mean number of injections was 9.16 ± 1.75. Subfoveal choroidal thickness decreased from 208.3 ± 73.7 μm at baseline to 185.3 ± 70.1 μm at mo-14 (p < 0.001), with significant (p < 0.001) decreases at all measured time points. Choroidal thickness also tended to decline in fellow eyes but was only statistically significant nasally 1,000 μm (p =0.04). Mean changes in choroidal thickness did not correlate with BCVA at mo-14 (p = 0.76). Disciform scars and geographic atrophy (p = 0.017), and BCVA (p < 001) at baseline were predictive of visual outcome. Age (p = 0.001), reticular drusen (p = 0.004), and size of choroidal neovascularized area (p = 0.042) were predictive of decreases in choroidal thickness. Conclusions: Submacular choroidal thickness appeared to decrease significantly in eyes with n-AMD over a 14-mo period of ranibizumab treatment. No corresponding decrease in choroidal thickness occurred in fellow eyes.
Collapse
Affiliation(s)
- Sibel Inan
- a Department of Ophthalmology, Health Sciences University Medical School , Afyonkarahisar , Turkey
| | - Zeki Baysal
- b Department of Ophthalmology, Batman State Hospital , Batman , Turkey
| | - Umit Ubeyt Inan
- c Department of Ophthalmology, Parkhayat Hospital , Afyonkarahisar , Turkey
| |
Collapse
|
42
|
Waugh N, Loveman E, Colquitt J, Royle P, Yeong JL, Hoad G, Lois N. Treatments for dry age-related macular degeneration and Stargardt disease: a systematic review. Health Technol Assess 2019; 22:1-168. [PMID: 29846169 DOI: 10.3310/hta22270] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Age-related macular degeneration (AMD) is the leading cause of visual loss in older people. Advanced AMD takes two forms, neovascular (wet) and atrophic (dry). Stargardt disease (STGD) is the commonest form of inherited macular dystrophy. OBJECTIVE To carry out a systematic review of treatments for dry AMD and STGD, and to identify emerging treatments where future NIHR research might be commissioned. DESIGN Systematic review. METHODS We searched MEDLINE, EMBASE, Web of Science and The Cochrane Library from 2005 to 13 July 2017 for reviews, journal articles and meeting abstracts. We looked for studies of interventions that aim to preserve or restore vision in people with dry AMD or STGD. The most important outcomes are those that matter to patients: visual acuity (VA), contrast sensitivity, reading speed, ability to drive, adverse effects of treatment, quality of life, progression of disease and patient preference. However, visual loss is a late event and intermediate predictors of future decline were accepted if there was good evidence that they are strong predictors of subsequent visual outcomes. These include changes detectable by investigation, but not necessarily noticed by people with AMD or STGD. ClinicalTrials.gov, the World Health Organization search portal and the UK Clinical Trials gateway were searched for ongoing and recently completed clinical trials. RESULTS The titles and abstracts of 7948 articles were screened for inclusion. The full text of 398 articles were obtained for further screening and checking of references and 112 articles were included in the final report. Overall, there were disappointingly few good-quality studies (including of sufficient size and duration) reporting useful outcomes, particularly in STGD. However we did identify a number of promising research topics, including drug treatments, stem cells, new forms of laser treatment, and implantable intraocular lens telescopes. In many cases, research is already under way, funded by industry or governments. LIMITATIONS In AMD, the main limitation came from the poor quality of much of the evidence. Many studies used VA as their main outcome despite not having sufficient duration to observe changes. The evidence on treatments for STGD is sparse. Most studies tested interventions with no comparison group, were far too short term, and the quality of some studies was poor. FUTURE WORK We think that the topics on which the Health Technology Assessment (HTA) and Efficacy Mechanism and Evaluation (EME) programmes might consider commissioning primary research are in STGD, a HTA trial of fenretinide (ReVision Therapeutics, San Diego, CA, USA), a visual cycle inhibitor, and EME research into the value of lutein and zeaxanthin supplements, using short-term measures of retinal function. In AMD, we suggest trials of fenretinide and of a potent statin. There is epidemiological evidence from the USA that the drug, levodopa, used for treating Parkinson's disease, may reduce the incidence of AMD. We suggest that similar research should be carried out using the large general practice databases in the UK. Ideally, future research should be at earlier stages in both diseases, before vision is impaired, using sensitive measures of macular function. This may require early detection of AMD by screening. STUDY REGISTRATION This study is registered as PROSPERO CRD42016038708. FUNDING The National Institute for Health Research HTA programme.
Collapse
Affiliation(s)
- Norman Waugh
- Division of Health Sciences, University of Warwick, Coventry, UK
| | | | | | - Pamela Royle
- Division of Health Sciences, University of Warwick, Coventry, UK
| | | | | | - Noemi Lois
- Ophthalmology, Royal Victoria Hospital, Belfast, UK.,Wellcome-Wolfson Centre for Experimental Medicine, Queens University, Belfast, UK
| |
Collapse
|
43
|
MORPHOLOGIC FEATURES OF THE RETINAL PIGMENT EPITHELIUM AND ASSOCIATED CHORIORETINAL CHARACTERISTICS IN EYES WITH EARLY AGE-RELATED MACULAR DEGENERATION AND SUBRETINAL DRUSENOID DEPOSITS. Retina 2019; 40:686-694. [PMID: 30950969 DOI: 10.1097/iae.0000000000002528] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE To investigate the features of the retinal pigment epithelium (RPE) on optical coherence tomography in eyes with early age-related macular degeneration with subretinal drusenoid deposit. METHODS We classified the eyes into three types: nonundulating RPE, undulating RPE, and wedge-shaped RPE. We compared the retinal vessel densities, retinal thickness, and choroidal thickness of a 3-mm-diameter zone. RESULTS We classified 33, 27, and 20 as nonundulating RPE, undulating RPE, and wedge-shaped RPE eyes, respectively. The vascular densities of the superficial and deep capillary plexus showed differences; nonundulating RPE group (23.93 ± 2.26% and 23.54 ± 1.78%), undulating RPE group (22.29 ± 2.80% and 21.94 ± 2.42%), and wedge-shaped RPE group (21.93 ± 2.70% and 20.63 ± 2.42%, P = 0.010 and P < 0.001). The mean retinal thickness and choroidal thickness were also different, nonundulating RPE group (298.26 ± 13.81 µm and 180.08 ± 55.49 µm), undulating RPE group (285.29 ± 21.88 µm and 148.45 ± 55.08 µm), and wedge-shaped RPE group (274.86 ± 20.62 µm and 135.75 ± 39.77 µm) (P = 0.001 and P = 0.007). CONCLUSION Altered features of the RPE on optical coherence tomography may indicate advancement in disease and be part of an overall degeneration process in these eyes.
Collapse
|
44
|
Optical coherence tomography evidence of macular ganglion cell-inner plexiform layer thinning in eyes with subretinal drusenoid deposits. Eye (Lond) 2019; 33:1290-1296. [PMID: 30926911 DOI: 10.1038/s41433-019-0405-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 01/30/2019] [Accepted: 03/01/2019] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND/OBJECTIVES The purpose of this study was to evaluate macular ganglion cell layer-inner plexiform layer (GCL-IPL) and choroidal thickness in early age-related macular degeneration (AMD) in eyes with subretinal drusenoid deposits (SDD). SUBJECTS/METHODS Comprehensive ophthalmological examination was performed. Near infrared reflectance and raster images using enhanced depth imaging were acquired with spectral domain optical coherence tomography. Drusen and SDD were diagnosed based on raster scans and near infrared reflectance. GCL-IPL maps were generated with automated segmentation and choroidal thickness maps were obtained by manually delineating the choroid-scleral boundary. RESULTS Forty-eight eyes from 48 patients (mean age 77.5 ± 5.7, range 68-90 years) with a diagnosis of early AMD and 42 eyes of 42 age-matched control subjects (mean age 76.9 ± 5.7, range 67-88 years) were included. Of these, 28 eyes (58.3%) had drusen alone, 4 eyes (8.3%) had SDD alone, and 16 eyes (33.3%) had drusen associated with SDD. Compared with controls, average choroidal thickness was significantly decreased in AMD eyes (P < 0.05). There was no significant difference in choroidal thickness in eyes with SDD with respect to those with drusen alone. GCL-IPL thickness was reduced in an annular pattern at the 3 and 6 mm macular areas in AMD patients with respect to controls (P < 0.05). GCL-IPL thickness at 3 mm was significantly reduced in eyes with SDD with respect to those with drusen alone (P = 0.03). CONCLUSIONS The GCL-IPL is reduced in thickness with an annular pattern in early AMD and is significantly thinner in eyes with SDD.
Collapse
|
45
|
Owen LA, Shakoor A, Morgan DJ, Hejazi AA, McEntire MW, Brown JJ, Farrer LA, Kim I, Vitale A, DeAngelis MM. The Utah Protocol for Postmortem Eye Phenotyping and Molecular Biochemical Analysis. Invest Ophthalmol Vis Sci 2019; 60:1204-1212. [PMID: 30924847 PMCID: PMC6440527 DOI: 10.1167/iovs.18-24254] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 01/31/2019] [Indexed: 12/14/2022] Open
Abstract
Purpose Current understanding of local disease pathophysiology in AMD is limited. Analysis of the human disease-affected tissue is most informative, as gene expression, expressed quantitative trait loci, microenvironmental, and epigenetic changes can be tissue, cell type, and location specific. Development of a novel translational treatment and prevention strategies particularly for earlier forms of AMD are needed, although access to human ocular tissue analysis is challenging. We present a standardized protocol to study rapidly processed postmortem donor eyes for molecular biochemical and genomic studies. Methods We partnered with the Utah Lions Eye Bank to obtain donor human eyes, blood, and vitreous, within 6 hours postmortem. Phenotypic analysis was performed using spectral-domain optical coherence tomography (SD-OCT) and color fundus photography. Macular and extramacular tissues were immediately isolated, and the neural retina and retinal pigment epithelium/choroid from each specimen were separated and preserved. Ocular disease phenotype was analyzed using clinically relevant grading criteria by a group of four ophthalmologists incorporating data from SD-OCT retinal images, fundus photographs, and medical records. Results The use of multimodal imaging leads to greater resolution of retinal pathology, allowing greater phenotypic rigor for both interobserver phenotype and known clinical diagnoses. Further, our analysis resulted in excellent quality RNA, which demonstrated appropriate tissue segregation. Conclusions The Utah protocol is a standardized methodology for analysis of disease mechanisms in AMD. It uniquely allows for simultaneous rigorous phenotypic, molecular biochemical, and genomic analysis of both systemic and local tissues. This better enables the development of disease biomarkers and therapeutic interventions.
Collapse
Affiliation(s)
- Leah A. Owen
- Department of Ophthalmology and Visual Sciences, University of Utah, School of Medicine, Salt Lake City, Utah, United States
| | - Akbar Shakoor
- Department of Ophthalmology and Visual Sciences, University of Utah, School of Medicine, Salt Lake City, Utah, United States
| | - Denise J. Morgan
- Department of Ophthalmology and Visual Sciences, University of Utah, School of Medicine, Salt Lake City, Utah, United States
| | - Andre A. Hejazi
- Department of Ophthalmology and Visual Sciences, University of Utah, School of Medicine, Salt Lake City, Utah, United States
| | | | - Jared J. Brown
- Utah Lions Eye Bank, Salt Lake City, Utah, United States
| | - Lindsay A. Farrer
- Departments of Medicine (Biomedical Genetics), Neurology, Ophthalmology, Epidemiology, and Biostatistics, Boston University Schools of Medicine and Public Health, Boston, Massachusetts, United States
| | - Ivana Kim
- Retina Service, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts, United States
| | - Albert Vitale
- Department of Ophthalmology and Visual Sciences, University of Utah, School of Medicine, Salt Lake City, Utah, United States
| | - Margaret M. DeAngelis
- Department of Ophthalmology and Visual Sciences, University of Utah, School of Medicine, Salt Lake City, Utah, United States
- Department of Pharmacotherapy, College of Pharmacy, University of Utah, Salt Lake City, Utah, United States
- Department of Population Health Sciences, University of Utah, School of Medicine, Salt Lake City, Utah, United States
| |
Collapse
|
46
|
Extensive Macular Atrophy with Pseudodrusen Imaged with OCT Angiography. Case Rep Ophthalmol Med 2018; 2018:8213097. [PMID: 30425871 PMCID: PMC6218749 DOI: 10.1155/2018/8213097] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 10/01/2018] [Accepted: 10/17/2018] [Indexed: 11/23/2022] Open
Abstract
This report describes the first case of extensive macular atrophy with pseudodrusen (EMAP) imaged with optical coherence tomography angiography (OCTA). A 58-year-old Caucasian man presented with decreased central vision in both eyes. Fundus examination showed large areas of macular atrophy centered on the fovea surrounded by diffuse reticular pseudodrusen. Spectral domain OCT (SDOCT) revealed outer retinal and choriocapillaris atrophy. OCTA demonstrated marked absence of choriocapillaris flow. Extensive macular atrophy with pseudodrusen is a rare clinical entity and a new extreme phenotype of macular degenerations that could shed more light on the role of pseudodrusen and choriocapillaris compromise in the pathogenesis of AMD.
Collapse
|
47
|
Baek JH, Lim D, Park KH, Chae JB, Jang H, Lee J, Chung H. Quantitative proteomic analysis of aqueous humor from patients with drusen and reticular pseudodrusen in age-related macular degeneration. BMC Ophthalmol 2018; 18:289. [PMID: 30404605 PMCID: PMC6222993 DOI: 10.1186/s12886-018-0941-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 10/12/2018] [Indexed: 01/12/2023] Open
Abstract
Background To identify novel biomarkers related to the pathogenesis of dry age-related macular degeneration (AMD), we adopted a human retinal pigment epithelial (RPE) cell culture model that mimics some features of dry AMD including the accumulation of intra- and sub-RPE deposits. Then, we investigated the aqueous humor (AH) proteome using a data-independent acquisition method (sequential window acquisition of all theoretical fragment ion mass spectrometry) for dry AMD patients and controls. Methods After uniformly pigmented polarized monolayers of human fetal primary RPE (hfRPE) cells were established, the cells were exposed to 4-hydroxy-2-nonenal (4-HNE), followed by Western blotting, immunofluorescence analysis and ELISA of cells or conditioned media for several proteins of interest. Data-dependent acquisition for identification of the AH proteome and SWATH-based mass spectrometry were performed for 11 dry AMD patients according to their phenotypes (including soft drusen and reticular pseudodrusen [RPD]) and 2 controls (3 groups). Results Increased intra- and sub-RPE deposits were observed in 4-HNE-treated hfRPE cells compared with control cultures based on APOA1, cathepsin D, and clusterin immunoreactivity. Additionally, the differential abundance of proteins in apical and basal chambers with or without 4-HNE treatment confirmed the polarized secretion of proteins from hfRPE cells. A total of 119 proteins were quantified in dry AMD patients and controls by SWATH-MS. Sixty-five proteins exhibited significantly altered abundance among the three groups. A two-dimensional principal component analysis plot was generated to identify typical proteins related to the pathogenesis of dry AMD. Among the identified proteins, eight proteins, including APOA1, CFHR2, and CLUS, were previously considered major components or regulators of drusen. Three proteins (SERPINA4, LUM, and KERA proteins) have not been previously described as components of drusen or as being related to dry AMD. Interestingly, the LUM and KERA proteins, which are related to extracellular matrix organization, were upregulated in both RPD and soft drusen. Conclusions Differential protein expression in the AH between patients with drusen and RPD was quantified using SWATH-MS in the present study. Detailed proteomic analyses of dry AMD patients might provide insights into the in vivo biology of drusen and RPD. Electronic supplementary material The online version of this article (10.1186/s12886-018-0941-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Je-Hyun Baek
- R&D Center for Clinical Mass Spectrometry, Seegene Medical Foundation, Seoul, 04805, South Korea
| | - Daehan Lim
- Department of Ophthalmology, Konkuk University School of Medicine, Konkuk University Medical Center, 120-1 Neungdong-ro, Gwangjin-gu, Seoul, Republic of Korea
| | - Kyu Hyung Park
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, 13620, South Korea
| | - Jae-Byoung Chae
- Department of Ophthalmology, Konkuk University School of Medicine, Konkuk University Medical Center, 120-1 Neungdong-ro, Gwangjin-gu, Seoul, Republic of Korea
| | - Hyoik Jang
- Department of Ophthalmology, Konkuk University School of Medicine, Konkuk University Medical Center, 120-1 Neungdong-ro, Gwangjin-gu, Seoul, Republic of Korea
| | - Jonghyun Lee
- Department of Ophthalmology, Ilsan Paik Hospital, Inje University College of Medicine, Goyang, 10380, South Korea
| | - Hyewon Chung
- Department of Ophthalmology, Konkuk University School of Medicine, Konkuk University Medical Center, 120-1 Neungdong-ro, Gwangjin-gu, Seoul, Republic of Korea.
| |
Collapse
|
48
|
Lyssenko NN, Haider N, Picataggi A, Cipollari E, Jiao W, Phillips MC, Rader DJ, Chavali VRM. Directional ABCA1-mediated cholesterol efflux and apoB-lipoprotein secretion in the retinal pigment epithelium. J Lipid Res 2018; 59:1927-1939. [PMID: 30076206 DOI: 10.1194/jlr.m087361] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 06/21/2018] [Indexed: 12/15/2022] Open
Abstract
Cholesterol-containing soft drusen and subretinal drusenoid deposits (SDDs) occur at the basolateral and apical side of the retinal pigment epithelium (RPE), respectively, in the chorioretina and are independent risk factors for late age-related macular degeneration (AMD). Cholesterol in these deposits could originate from the RPE as nascent HDL or apoB-lipoprotein. We characterized cholesterol efflux and apoB-lipoprotein secretion in RPE cells. Human RPE cells, ARPE-19, formed nascent HDL that was similar in physicochemical properties to nascent HDL formed by other cell types. In highly polarized primary human fetal RPE (phfRPE) monolayers grown in low-lipid conditions, cholesterol efflux to HDL was moderately directional to the apical side and much stronger than ABCA1-mediated efflux to apoA-I at both sides; ABCA1-mediated efflux was weak and equivalent between the two sides. Feeding phfRPE monolayers with oxidized or acetylated LDL increased intracellular levels of free and esterified cholesterol and substantially raised ABCA1-mediated cholesterol efflux at the apical side. phfRPE monolayers secreted apoB-lipoprotein preferentially to the apical side in low-lipid and oxidized LDL-feeding conditions. These findings together with evidence from human genetics and AMD pathology suggest that RPE-generated HDL may contribute lipid to SDDs.
Collapse
Affiliation(s)
- Nicholas N Lyssenko
- Division of Translational Medicine and Human Genetics, Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Naqi Haider
- Department of Ophthalmology, Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA
| | - Antonino Picataggi
- Division of Translational Medicine and Human Genetics, Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Eleonora Cipollari
- Division of Translational Medicine and Human Genetics, Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Wanzhen Jiao
- Department of Ophthalmology, Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA
| | - Michael C Phillips
- Division of Translational Medicine and Human Genetics, Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Daniel J Rader
- Division of Translational Medicine and Human Genetics, Department of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | | |
Collapse
|
49
|
LONG-TERM VISUAL CHANGES IN INITIALLY STRONGER FELLOW EYES IN PATIENTS WITH UNILATERAL TYPE 3 NEOVASCULARIZATION. Retina 2018; 39:1672-1681. [PMID: 29979454 DOI: 10.1097/iae.0000000000002239] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE To evaluate long-term visual changes in initially stronger fellow eyes in patients with unilateral Type 3 neovascularization. METHODS This retrospective study included 102 patients who were newly diagnosed with unilateral Type 3 neovascularization and in whom the best-corrected visual acuity (BCVA) of the fellow eye was initially better than that of the involved eye. All patients were treated with intravitreal anti-vascular endothelial growth factor injections. The BCVAs were compared at diagnosis, 12 months, 24 months, and the final visit. In patients who experienced ≥3 lines of visual deterioration in the BCVA of the fellow eye, the reason for visual deterioration was also verified. RESULTS The patients were followed for 45.9 ± 18.5 months after diagnosis. At diagnosis, the fellow-eye BCVA was better than that of the initially involved eye in all 102 patients. However, the fellow-eye visual acuity was the same or worse than that of the initially involved eye in 13 patients (12.7%) at 12 months, in 20 patients (19.6%) at 24 months, and in 24 patients (23.5%) at the final visit. At the final visit, 53 patients (51.9%) had experienced ≥3 lines of deterioration in the BCVA of the fellow eye. Fellow-eye neovascularization occurred in 42 patients, and geographic atrophy involving the fovea was noted in the remaining 11 patients. CONCLUSION Deterioration of the visual acuity of the fellow eye is frequently noted in unilateral Type 3 neovascularization. As a result of this deterioration, the initially stronger fellow eye did not remain stronger in 23.5% of the patients, suggesting the need for long-term strict treatment of the initially involved eye even when the visual acuity of the fellow eye is good.
Collapse
|
50
|
Spaide RF, Ooto S, Curcio CA. Subretinal drusenoid deposits AKA pseudodrusen. Surv Ophthalmol 2018; 63:782-815. [PMID: 29859199 DOI: 10.1016/j.survophthal.2018.05.005] [Citation(s) in RCA: 175] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 05/17/2018] [Accepted: 05/21/2018] [Indexed: 01/30/2023]
Abstract
A distinction between conventional drusen and pseudodrusen was first made in 1990, and more recently knowledge of pseudodrusen, more accurately called subretinal drusenoid deposits (SDDs), has expanded. Pseudodrusen have a bluish-white appearance by biomicroscopy and color fundus photography. Using optical coherence tomography, pseudodrusen were found to be accumulations of material internal to the retinal pigment epithelium that could extend internally through the ellipsoid zone. These deposits are more commonly seen in older eyes with thinner choroids. Histologic evaluation of these deposits revealed aggregations of material in the subretinal space between photoreceptors and retinal pigment epithelium. SDDs contain some proteins in common with soft drusen but differ in lipid composition. Many studies reported that SDDs are strong independent risk factors for late age-related macular degeneration. Geographic atrophy and type 3 neovascularization are particularly associated with SDD. Unlike conventional drusen, eyes with SDD show slow dark adaptation and poor contrast sensitivity. Outer retinal atrophy develops in eyes with regression of SDD, a newly recognized form of late age-related macular degeneration. Advances in imaging technology have enabled many insights into this condition, including associated photoreceptor, retinal pigment epithelium, and underlying choroidal changes.
Collapse
Affiliation(s)
- Richard F Spaide
- Vitreous Retina Macula Consultants of New York and LuEsther T. Mertz Retinal Research Center, Manhattan Eye, Ear and Throat Hospital, New York, New York, USA.
| | - Sotaro Ooto
- Vitreous Retina Macula Consultants of New York and LuEsther T. Mertz Retinal Research Center, Manhattan Eye, Ear and Throat Hospital, New York, New York, USA; Department of Ophthalmology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Christine A Curcio
- Department of Ophthalmology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabamas, USA
| |
Collapse
|