1
|
Aleman TS, Roman AJ, Uyhazi KE, Jiang YY, Bedoukian EC, Sumaroka A, Wu V, Swider M, Viarbitskaya I, Russell RC, Shagena EO, Santos AJ, Serrano LW, Parchinski KM, Kim RJ, Weber ML, Garafalo AV, Thompson DA, Maguire AM, Bennett J, Scoles DH, O'Neil EC, Morgan JIW, Cideciyan AV. Retinal Degeneration Associated With Biallelic RDH12 Variants: Longitudinal Evaluation of Retinal Structure and Visual Function in Pediatric Patients. Invest Ophthalmol Vis Sci 2024; 65:30. [PMID: 39693083 DOI: 10.1167/iovs.65.14.30] [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: 12/19/2024] Open
Abstract
Purpose The purpose of this study was to determine the natural history of the photoreceptor disease in a large group of pediatric patients with RHD12-associated Leber congenital amaurosis (RDH12-LCA), to estimate the changes expected over the duration of a clinical trial, and to define the relationship between the photoreceptor loss and visual dysfunction. Methods Forty-six patients representing 36 families were included. The great majority of patients were under the age of 18 years. Patients underwent complete ophthalmic examinations and imaging with various modalities including adaptive optics scanning laser ophthalmoscopy. Visual function was assessed with static and kinetic perimetry, and full-field stimulus test (FST) under dark- and light-adapted conditions. Results Patients had a severe and early onset retinal degeneration (EORD). Visual acuity losses showed a progression rate of 0.04 logMAR per year. A small foveal island could be retained but showed degeneration over time. Foveal cone sensitivity losses were predictable by the loss of photoreceptors. Peripapillary retina could be retained with no significant progression detectable. Peripapillary rod sensitivity was substantially less than expected from photoreceptor structure pointing to a large improvement potential. FST sensitivities were reliably recordable in pediatric patients and showed a small but significant improvement with age. Locally and globally, loss of rod sensitivity tended to be larger than loss of cone sensitivity. Conclusions Foveal cones of RDH12-LCA should be targeted with treatments aimed to slow progression, whereas peripapillary rod photoreceptors should be targeted with treatments aimed to improve night vision. Pediatric FST testing may be complicated by age-related maturation of decision making regarding threshold criteria.
Collapse
Affiliation(s)
- Tomas S Aleman
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
- Division of Ophthalmology and The Individualized Medical Genetics Center, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States
| | - Alejandro J Roman
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Katherine E Uyhazi
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Yu You Jiang
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Emma C Bedoukian
- Division of Ophthalmology and The Individualized Medical Genetics Center, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States
| | - Alexander Sumaroka
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Vivian Wu
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Malgorzata Swider
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Iryna Viarbitskaya
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Robert C Russell
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Elizabeth O Shagena
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Arlene J Santos
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Leona W Serrano
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Kelsey M Parchinski
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Rebecca J Kim
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Mariejel L Weber
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Alexandra V Garafalo
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Dorothy A Thompson
- Great Ormond Street Hospital for Children, NHS Foundation Trust, London, United Kingdom
| | - Albert M Maguire
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
- Division of Ophthalmology and The Individualized Medical Genetics Center, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States
| | - Jean Bennett
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Drew H Scoles
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
- Division of Ophthalmology and The Individualized Medical Genetics Center, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States
| | - Erin C O'Neil
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
- Division of Ophthalmology and The Individualized Medical Genetics Center, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States
| | - Jessica I W Morgan
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Artur V Cideciyan
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| |
Collapse
|
2
|
Wynne N, Jiang YY, Aleman TS, Morgan JIW. Retinal Sensitivity in Comparison to Cone Density in Choroideremia. Invest Ophthalmol Vis Sci 2024; 65:6. [PMID: 39625441 PMCID: PMC11620006 DOI: 10.1167/iovs.65.14.6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 11/06/2024] [Indexed: 12/08/2024] Open
Abstract
Purpose Choroideremia (CHM) is an X-linked inherited retinal degeneration causing loss of photoreceptors, retinal pigment epithelium, and choriocapillaris. Structural abnormalities of the cone photoreceptor mosaic have been reported even within the retained island of functioning retina. Here, we describe the relationship between cone density and visual sensitivity within the retained central retina in CHM. Methods The cone mosaics of 31 patients aged 10 to 57 years with CHM were imaged using a multi-modal, custom-built adaptive optics scanning light ophthalmoscope (AOSLO). Retinal sensitivity was measured using fundus tracking perimetry, cross-sectional retinal structure with optical coherence tomography. The relationship between sensitivity at each retinal location tested and structural parameters of local disease severity (cone density and distance to border) was explored using unpaired t-tests and linear regressions. Ellipsoid zone area was also investigated. Results Three hundred fourteen individual regions of interest were analyzed. Cone density and retinal sensitivities were significantly decreased compared with healthy controls (P < 0.0002). There was a statistically significant correlation between cone density and visual sensitivity within the fovea and parafovea (P < 0.0005), but not in the perifovea (P > 0.1). There was a significant relationship between the distance to the atrophic border and sensitivity from 400 µm to 1600 µm eccentricity (P < 0.001). Mean sensitivity was significantly related to the ellipsoid extent (P < 0.0001). Conclusions The correlation between cone density and retinal sensitivity within 1 mm of the foveal center in CHM suggests central sensitivity loss is driven by cone loss. Outside of the central fixation point, proximity to the atrophic border is correlated with sensitivity, suggesting the presence of a functional transition zone.
Collapse
Affiliation(s)
- Niamh Wynne
- Scheie Eye Institute, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, United States
| | - Yu You Jiang
- Scheie Eye Institute, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, United States
- Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Tomas S. Aleman
- Scheie Eye Institute, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, United States
- Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Jessica I. W. Morgan
- Scheie Eye Institute, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, United States
- Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| |
Collapse
|
3
|
Poli FE, MacLaren RE, Cehajic-Kapetanovic J. Retinal Patterns and the Role of Autofluorescence in Choroideremia. Genes (Basel) 2024; 15:1471. [PMID: 39596671 PMCID: PMC11593989 DOI: 10.3390/genes15111471] [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: 10/05/2024] [Revised: 11/05/2024] [Accepted: 11/09/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Choroideremia is a monogenic inherited retinal dystrophy that manifests in males with night blindness, progressive loss of peripheral vision, and ultimately profound sight loss, commonly by middle age. It is caused by genetic defects of the CHM gene, which result in a deficiency in Rab-escort protein-1, a key element for intracellular trafficking of vesicles, including those carrying melanin. As choroideremia primarily affects the retinal pigment epithelium, fundus autofluorescence, which focuses on the fluorescent properties of pigments within the retina, is an established imaging modality used for the assessment and monitoring of affected patients. METHODS AND RESULTS In this manuscript, we demonstrate the use of both short-wavelength blue and near-infrared autofluorescence and how these imaging modalities reveal distinct disease patterns in choroideremia. In addition, we show how these structural measurements relate to retinal functional measures, namely microperimetry, and discuss the potential role of these retinal imaging modalities in clinical practice and research studies. Moreover, we discuss the mechanisms underlying retinal autofluorescence patterns by imaging with a particular focus on melanin pigment. CONCLUSIONS This could be of particular significance given the current progress in therapeutic options, including gene replacement therapy.
Collapse
Affiliation(s)
- Federica E. Poli
- Nuffield Laboratory of Ophthalmology, University of Oxford, Oxford OX3 9DU, UK
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
- Royal Berkshire NHS Foundation Trust, Reading RG1 5AN, UK
| | - Robert E. MacLaren
- Nuffield Laboratory of Ophthalmology, University of Oxford, Oxford OX3 9DU, UK
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Jasmina Cehajic-Kapetanovic
- Nuffield Laboratory of Ophthalmology, University of Oxford, Oxford OX3 9DU, UK
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
| |
Collapse
|
4
|
Prétot D, Della Volpe Waizel M, Kaminska K, Valmaggia P, Placidi G, Falsini B, Fries FN, Szentmáry N, Rivolta C, Scholl HPN, Calzetti G. Retinal oxygen metabolic function in choroideremia and retinitis pigmentosa. Graefes Arch Clin Exp Ophthalmol 2024:10.1007/s00417-024-06659-8. [PMID: 39394491 DOI: 10.1007/s00417-024-06659-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 09/13/2024] [Accepted: 10/03/2024] [Indexed: 10/13/2024] Open
Abstract
PURPOSE To measure the retinal oxygen metabolic function with retinal oximetry (RO) in patients with choroideremia (CHM) and compare these findings with retinitis pigmentosa (RP) patients and controls. METHODS Prospective observational study including 18 eyes of 9 molecularly confirmed CHM patients (9♂; 40.2 ± 21.2 years (mean ± SD), 77 eyes from 39 patients with RP (15♀ 24♂; 45.6 ± 14.7 years) and 100 eyes from 53 controls (31♀ 22♂; 40.2 ± 13.4 years). Main outcome parameters were the mean arterial (A-SO2; %), venular (V-SO2; %) oxygen saturation, and their difference (A-V SO2; %) recorded with the oxygen saturation tool of the Retinal Vessel Analyzer (IMEDOS Systems UG, Germany). Statistical analyses were performed with linear mixed-effects models. RESULTS Eyes suffering from CHM differed significantly from both RP and control eyes, when the retinal oxygen metabolic parameters were taken into account. While RP showed significantly higher A-SO2 and V-SO2 values when compared to controls, CHM showed opposite findings with significantly lower values when compared to both RP and controls (P < 0.001). The A-V SO2, which represents the retinal oxygen metabolic consumption, showed significantly lower values in CHM compared to controls. CONCLUSION The retina in CHM is a relatively hypoxic environment. The decrease in oxygen levels may be due to the profound choroidal degeneration, leading to decreased oxygen flux to the retina. RO measurements may help understand the pathogenesis of CHM and RP. These findings may provide useful details to inform the planning of clinical trials of emerging therapies for CHM. KEY MESSAGES What was known before? Retinal oxygen metabolic function measured with retinal oximetry (RO) shows significant alterations in patients with retinitis pigmentosa. WHAT THIS STUDY ADDS RO function in choroideremia is significantly altered when compared to controls. Furthermore, RO in choroideremia shows opposing findings within different oxygen metabolic parameters to those that were so far known for retinitis pigmentosa. By providing insights into the retinal oxygen metabolic mechanisms, RO can help understand the underlying pathophysiology in choroideremia.
Collapse
Affiliation(s)
- Dominique Prétot
- Department of Ophthalmology, University Hospital Basel, University of Basel, Basel, Switzerland
- Heuberger Eye Clinic, Olten, Switzerland
| | - Maria Della Volpe Waizel
- Department of Ophthalmology, University Hospital Basel, University of Basel, Basel, Switzerland
- Dr. Rolf M. Schwiete Center for Limbal Stem Cell and Congenital Aniridia Research, Saarland University, Homburg, Saar, Germany
| | - Karolina Kaminska
- Department of Ophthalmology, University Hospital Basel, University of Basel, Basel, Switzerland
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
| | - Philippe Valmaggia
- Department of Ophthalmology, University Hospital Basel, University of Basel, Basel, Switzerland
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - Giorgio Placidi
- Ophthalmology Unit, Fondazione Policlinico Universitario ''A. Gemelli'' IRCCS/Università Cattolica del S. Cuore, Rome, Italy
| | - Benedetto Falsini
- Ophthalmology Unit, Fondazione Policlinico Universitario ''A. Gemelli'' IRCCS/Università Cattolica del S. Cuore, Rome, Italy
| | - Fabian N Fries
- Dr. Rolf M. Schwiete Center for Limbal Stem Cell and Congenital Aniridia Research, Saarland University, Homburg, Saar, Germany
- Department of Ophthalmology, Saarland University Medical Center, Homburg, Saar, Germany
| | - Nóra Szentmáry
- Dr. Rolf M. Schwiete Center for Limbal Stem Cell and Congenital Aniridia Research, Saarland University, Homburg, Saar, Germany
- Department of Ophthalmology, Saarland University Medical Center, Homburg, Saar, Germany
| | - Carlo Rivolta
- Department of Ophthalmology, University Hospital Basel, University of Basel, Basel, Switzerland
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Genetics and Genome Biology, University of Leicester, Leicester, LE1 7RH, UK
| | - Hendrik P N Scholl
- Department of Ophthalmology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Giacomo Calzetti
- Department of Ophthalmology, University Hospital Basel, University of Basel, Basel, Switzerland.
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland.
- Vista Vision Eye Clinic, Brescia, Italy.
| |
Collapse
|
5
|
Bonneau S, Kulbay M, Kahn-Ali S, Qian CX. Exploring the impact of Choroideremia on women with phenotypic and/or genotypic evidence of disease: insights from a global survey. Ophthalmic Genet 2024; 45:452-461. [PMID: 38847528 DOI: 10.1080/13816810.2024.2357705] [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: 01/20/2024] [Revised: 05/02/2024] [Accepted: 05/08/2024] [Indexed: 06/22/2024]
Abstract
INTRODUCTION Choroideremia (CHM) is an X-linked inherited retinal disease mostly affecting males. However, women with phenotypic and/or genotypic evidence of CHM may develop degenerative visual disability with advancing age. Our objective was to determine the visual impacts of phenotypic and/or genotypic evidence of CHM in women and its associated psychosocial burden and influence on activities of daily living (ADLs). METHODS We conducted an international cross-sectional survey from April to December 2022 using an e-questionnaire distributed through not-for-profit stakeholder organizations and social media plat-forms. RESULTS With a total of 55 respondents (n = 55), most women with phenotypic and/or genotypic evidence of CHM (76%) reported a change in their visual acuity. When assessing its impact on ADLs, Pearson's correlation coefficient showed a negative correlation between driving (p = 0.046) and mobility capabil-ities (0.046) with the respondent's age. More than half of women reported being afraid, anxious, and stressed, with women below the age of 50 years old reporting a significantly higher level of distress and hopelessness (p = 0.003), anxiety (p = 0.00007), issues with relaxing (p = 0.025), and negative personal thoughts (p = 0.042). CONCLUSION Overall, this survey outlines both physical and psychological burden of being a woman with phenotypic and/or genotypic evidence of CHM. Given the limited clinical research in females affected by CHM, this patient-centered survey is a crucial advocacy tool for these individuals.
Collapse
Affiliation(s)
- Steven Bonneau
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Centre Universitaire affilié à l'Université de Montréal, Montréal, Québec, Canada
- Department of Surgery, Division of Ophthalmology, University of Sherbrooke, Sherbrooke, Québec, Canada
| | - Merve Kulbay
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Centre Universitaire affilié à l'Université de Montréal, Montréal, Québec, Canada
- Department of Ophthalmology & Visual Sciences, McGill University, Montreal, Quebec, Canada
| | - Shigufa Kahn-Ali
- Department of Ophthalmology, Centre Universitaire d'Ophtalmologie (CUO), Hôpital Maisonneuve-Rosemont, University of Montreal, Montréal, Québec, Canada
| | - Cynthia X Qian
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Centre Universitaire affilié à l'Université de Montréal, Montréal, Québec, Canada
- Department of Ophthalmology & Visual Sciences, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
6
|
Baffour-Awuah KA, Taylor LJ, Josan AS, Jolly JK, MacLaren RE. Investigating the impact of asymmetric macular sensitivity on visual acuity chart reading in choroideremia. Ophthalmic Physiol Opt 2024; 44:1188-1201. [PMID: 38989810 DOI: 10.1111/opo.13356] [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: 11/22/2023] [Revised: 06/08/2024] [Accepted: 06/10/2024] [Indexed: 07/12/2024]
Abstract
INTRODUCTION Degeneration in choroideremia, unlike typical centripetal photoreceptor degenerations, is centred temporal to the fovea. Once the fovea is affected, the nasal visual field (temporal retina) is relatively spared, and the preferred retinal locus shifts temporally. Therefore, when reading left to right, only the right eye reads into a scotoma. We investigate how this unique property affects the ability to read an eye chart. METHODS Standard- and low-luminance visual acuity (VA) for right and left eyes were measured with the Early Treatment of Diabetic Retinopathy Study (ETDRS) chart. Letters in each line were labelled by column position. The numbers of letter errors for each position across the whole chart were summed to produce total column error scores for each participant. Macular sensitivity was assessed using microperimetry. Central sensitivity asymmetry was determined by the temporal-versus-nasal central macular difference and subsequently correlated to a weighted ETDRS column error score. Healthy volunteers and participants with X-linked retinitis pigmentosa GTPase regulator associated retinitis pigmentosa (RPGR-RP) were used as controls. RESULTS Thirty-nine choroideremia participants (median age 44.9 years [IQR 35.7-53.5]), 23 RPGR-RP participants (median age 30.8 years [IQR 26.5-40.5]) and 35 healthy controls (median age 23.8 years [IQR 20.3-29.0]) were examined. In choroideremia, standard VA in the right eye showed significantly greater ETDRS column errors on the temporal side compared with the nasal side (p = 0.002). This significantly correlated with greater asymmetry in temporal-versus-nasal central macular sensitivity (p = 0.04). No significant patterns in ETDRS column errors or central macular sensitivity were seen in the choroideremia left eyes, nor in RPGR-RP and control eyes. CONCLUSION Difficulty in tracking across lines during ETDRS VA testing may cause excess errors independent of true VA. VA assessment with single-letter optotype systems may be more suitable, particularly for patients with choroideremia, and potentially other retinal diseases with asymmetric central macular sensitivity or large central scotomas including geographic atrophy.
Collapse
Affiliation(s)
- Kwame A Baffour-Awuah
- Oxford Eye Hospital, Oxford University Hospitals NHS Trust, Oxford, UK
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Laura J Taylor
- Oxford Eye Hospital, Oxford University Hospitals NHS Trust, Oxford, UK
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Amandeep S Josan
- Oxford Eye Hospital, Oxford University Hospitals NHS Trust, Oxford, UK
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Jasleen K Jolly
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- Vision and Eye Research Institute, Anglia Ruskin University, Cambridge, UK
| | - Robert E MacLaren
- Oxford Eye Hospital, Oxford University Hospitals NHS Trust, Oxford, UK
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
7
|
Gocuk SA, Ayton LN, Edwards TL, McGuinness MB, Maclaren RE, Taylor LJ, Jolly JK. Longitudinal assessment of female carriers of choroideremia using multimodal retinal imaging. Br J Ophthalmol 2024:bjo-2024-325578. [PMID: 39122355 DOI: 10.1136/bjo-2024-325578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 07/20/2024] [Indexed: 08/12/2024]
Abstract
BACKGROUND/AIMS Female choroideremia carriers present with a spectrum of disease severity. Unlike in men, the rate of disease progression has not been well characterised in carriers. This longitudinal study aimed to determine the rate of retinal degeneration in choroideremia carriers, using multimodal imaging and microperimetry. METHODS Choroideremia carriers previously seen at Oxford Eye Hospital (United Kingdom) between 2012 and 2017 returned for testing between 2015 and 2023, providing up to 11 years' follow-up data. Participants had optical coherence tomography, fundus-tracked microperimetry and fundus autofluorescence (FAF) imaging performed. RESULTS Thirty-four eyes of 17 choroideremia carriers were examined using multimodal imaging. Median age was 44 (range: 15-73) years at baseline and median follow-up duration was 7 (range: 1-11) years. At baseline, phenotype was classified as fine (n=5 eyes), coarse (n=13 eyes), geographic (n=12 eyes) or male pattern (n=4 eyes). Thirteen patients showed no change in phenotype classification, four showed slight changes associated with choroideremia-related retinal degeneration. Despite this, carriers with severe retinal phenotypes had a statistically significant decline in average retinal sensitivity (-0.7 dB and -0.8 dB per year, respectively, p<0.001), area of geographic loss defined by FAF (+2.5 mm2 and +3.7 mm2 per year, respectively, p<0.001) and thinning of the photoreceptor complex (up to -2.8 microns and -10.3 microns per year, p<0.001). CONCLUSION Choroideremia carriers, particularly those with severe retinal phenotypes, exhibit progressive retinal degeneration, as evident by multimodal imaging biomarkers and functional testing. Clinicians should not rely on retinal severity classification alone to assess disease progression.
Collapse
Affiliation(s)
- Sena A Gocuk
- Department of Optometry and Vision Sciences, The University of Melbourne, Melbourne, Victoria, Australia
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
- Ophthalmology, Department of Surgery, The University of Melbourne, Melbourne, Victoria, Australia
| | - Lauren N Ayton
- Department of Optometry and Vision Sciences, The University of Melbourne, Melbourne, Victoria, Australia
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
- Ophthalmology, Department of Surgery, The University of Melbourne, Melbourne, Victoria, Australia
| | - Thomas L Edwards
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
- Ophthalmology, Department of Surgery, The University of Melbourne, Melbourne, Victoria, Australia
| | - Myra B McGuinness
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
- Ophthalmology, Department of Surgery, The University of Melbourne, Melbourne, Victoria, Australia
- Centre for Epidemiology and Biostatistics, The University of Melbourne, Melbourne, Victoria, Australia
| | - Robert E Maclaren
- Oxford Eye Hospital, Oxford University Hospital NHS Foundation Trust, Oxford, UK
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, The University of Oxford, Oxford, UK
| | - Laura J Taylor
- Oxford Eye Hospital, Oxford University Hospital NHS Foundation Trust, Oxford, UK
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, The University of Oxford, Oxford, UK
| | - Jasleen K Jolly
- Department of Optometry and Vision Sciences, The University of Melbourne, Melbourne, Victoria, Australia
- Oxford Eye Hospital, Oxford University Hospital NHS Foundation Trust, Oxford, UK
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, The University of Oxford, Oxford, UK
- Vision and Eye Research Institute, Anglia Ruskin University, Cambridge, UK
| |
Collapse
|
8
|
Romano F, Barbieri L, Staurenghi G, Salvetti AP. Hyperreflective Ganglion Cell Layer Band in Choroideremia. Retina 2024; 44:e42-e43. [PMID: 37972966 DOI: 10.1097/iae.0000000000004003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Affiliation(s)
- Francesco Romano
- Eye Clinic, Department of Biomedical and Clinical Sciences, Ospedale Luigi Sacco, University of Milan, Milan, Italy
- Harvard Retinal Imaging Lab, Harvard Medical School, Massachusetts Eye and Ear, Boston, Massachusetts; and
- Retina Service, Department of Ophthalmology, Massachusetts Eye and Ear, Boston, Massachusetts
| | - Lucrezia Barbieri
- Eye Clinic, Department of Biomedical and Clinical Sciences, Ospedale Luigi Sacco, University of Milan, Milan, Italy
| | - Giovanni Staurenghi
- Eye Clinic, Department of Biomedical and Clinical Sciences, Ospedale Luigi Sacco, University of Milan, Milan, Italy
| | - Anna Paola Salvetti
- Eye Clinic, Department of Biomedical and Clinical Sciences, Ospedale Luigi Sacco, University of Milan, Milan, Italy
| |
Collapse
|
9
|
Pierce EA, Aleman TS, Jayasundera KT, Ashimatey BS, Kim K, Rashid A, Jaskolka MC, Myers RL, Lam BL, Bailey ST, Comander JI, Lauer AK, Maguire AM, Pennesi ME. Gene Editing for CEP290-Associated Retinal Degeneration. N Engl J Med 2024; 390:1972-1984. [PMID: 38709228 PMCID: PMC11389875 DOI: 10.1056/nejmoa2309915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/07/2024]
Abstract
BACKGROUND CEP290-associated inherited retinal degeneration causes severe early-onset vision loss due to pathogenic variants in CEP290. EDIT-101 is a clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated protein 9 (Cas9) gene-editing complex designed to treat inherited retinal degeneration caused by a specific damaging variant in intron 26 of CEP290 (IVS26 variant). METHODS We performed a phase 1-2, open-label, single-ascending-dose study in which persons 3 years of age or older with CEP290-associated inherited retinal degeneration caused by a homozygous or compound heterozygous IVS26 variant received a subretinal injection of EDIT-101 in the worse (study) eye. The primary outcome was safety, which included adverse events and dose-limiting toxic effects. Key secondary efficacy outcomes were the change from baseline in the best corrected visual acuity, the retinal sensitivity detected with the use of full-field stimulus testing (FST), the score on the Ora-Visual Navigation Challenge mobility test, and the vision-related quality-of-life score on the National Eye Institute Visual Function Questionnaire-25 (in adults) or the Children's Visual Function Questionnaire (in children). RESULTS EDIT-101 was injected in 12 adults 17 to 63 years of age (median, 37 years) at a low dose (in 2 participants), an intermediate dose (in 5), or a high dose (in 5) and in 2 children 9 and 14 years of age at the intermediate dose. At baseline, the median best corrected visual acuity in the study eye was 2.4 log10 of the minimum angle of resolution (range, 3.9 to 0.6). No serious adverse events related to the treatment or procedure and no dose-limiting toxic effects were recorded. Six participants had a meaningful improvement from baseline in cone-mediated vision as assessed with the use of FST, of whom 5 had improvement in at least one other key secondary outcome. Nine participants (64%) had a meaningful improvement from baseline in the best corrected visual acuity, the sensitivity to red light as measured with FST, or the score on the mobility test. Six participants had a meaningful improvement from baseline in the vision-related quality-of-life score. CONCLUSIONS The safety profile and improvements in photoreceptor function after EDIT-101 treatment in this small phase 1-2 study support further research of in vivo CRISPR-Cas9 gene editing to treat inherited retinal degenerations due to the IVS26 variant of CEP290 and other genetic causes. (Funded by Editas Medicine and others; BRILLIANCE ClinicalTrials.gov number, NCT03872479.).
Collapse
Affiliation(s)
- Eric A Pierce
- From the Ocular Genomics Institute, Department of Ophthalmology, Mass Eye and Ear and Harvard Medical School, Boston (E.A.P., J.I.C.), and Editas Medicine, Cambridge (B.S.A., K.K., A.R., M.C.J., R.L.M.) - both in Massachusetts; the Scheie Eye Institute and the Division of Ophthalmology of the Children's Hospital of Philadelphia, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia (T.S.A., A.M.M.); the University of Michigan Kellogg Eye Center, Ann Arbor (K.T.J.); the Bascom Palmer Eye Institute, University of Miami, Miami (B.L.L.); and the Casey Eye Institute, Oregon Health and Science University, Portland (S.T.B., A.K.L., M.E.P.)
| | - Tomas S Aleman
- From the Ocular Genomics Institute, Department of Ophthalmology, Mass Eye and Ear and Harvard Medical School, Boston (E.A.P., J.I.C.), and Editas Medicine, Cambridge (B.S.A., K.K., A.R., M.C.J., R.L.M.) - both in Massachusetts; the Scheie Eye Institute and the Division of Ophthalmology of the Children's Hospital of Philadelphia, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia (T.S.A., A.M.M.); the University of Michigan Kellogg Eye Center, Ann Arbor (K.T.J.); the Bascom Palmer Eye Institute, University of Miami, Miami (B.L.L.); and the Casey Eye Institute, Oregon Health and Science University, Portland (S.T.B., A.K.L., M.E.P.)
| | - Kanishka T Jayasundera
- From the Ocular Genomics Institute, Department of Ophthalmology, Mass Eye and Ear and Harvard Medical School, Boston (E.A.P., J.I.C.), and Editas Medicine, Cambridge (B.S.A., K.K., A.R., M.C.J., R.L.M.) - both in Massachusetts; the Scheie Eye Institute and the Division of Ophthalmology of the Children's Hospital of Philadelphia, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia (T.S.A., A.M.M.); the University of Michigan Kellogg Eye Center, Ann Arbor (K.T.J.); the Bascom Palmer Eye Institute, University of Miami, Miami (B.L.L.); and the Casey Eye Institute, Oregon Health and Science University, Portland (S.T.B., A.K.L., M.E.P.)
| | - Bright S Ashimatey
- From the Ocular Genomics Institute, Department of Ophthalmology, Mass Eye and Ear and Harvard Medical School, Boston (E.A.P., J.I.C.), and Editas Medicine, Cambridge (B.S.A., K.K., A.R., M.C.J., R.L.M.) - both in Massachusetts; the Scheie Eye Institute and the Division of Ophthalmology of the Children's Hospital of Philadelphia, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia (T.S.A., A.M.M.); the University of Michigan Kellogg Eye Center, Ann Arbor (K.T.J.); the Bascom Palmer Eye Institute, University of Miami, Miami (B.L.L.); and the Casey Eye Institute, Oregon Health and Science University, Portland (S.T.B., A.K.L., M.E.P.)
| | - Keunpyo Kim
- From the Ocular Genomics Institute, Department of Ophthalmology, Mass Eye and Ear and Harvard Medical School, Boston (E.A.P., J.I.C.), and Editas Medicine, Cambridge (B.S.A., K.K., A.R., M.C.J., R.L.M.) - both in Massachusetts; the Scheie Eye Institute and the Division of Ophthalmology of the Children's Hospital of Philadelphia, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia (T.S.A., A.M.M.); the University of Michigan Kellogg Eye Center, Ann Arbor (K.T.J.); the Bascom Palmer Eye Institute, University of Miami, Miami (B.L.L.); and the Casey Eye Institute, Oregon Health and Science University, Portland (S.T.B., A.K.L., M.E.P.)
| | - Alia Rashid
- From the Ocular Genomics Institute, Department of Ophthalmology, Mass Eye and Ear and Harvard Medical School, Boston (E.A.P., J.I.C.), and Editas Medicine, Cambridge (B.S.A., K.K., A.R., M.C.J., R.L.M.) - both in Massachusetts; the Scheie Eye Institute and the Division of Ophthalmology of the Children's Hospital of Philadelphia, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia (T.S.A., A.M.M.); the University of Michigan Kellogg Eye Center, Ann Arbor (K.T.J.); the Bascom Palmer Eye Institute, University of Miami, Miami (B.L.L.); and the Casey Eye Institute, Oregon Health and Science University, Portland (S.T.B., A.K.L., M.E.P.)
| | - Michael C Jaskolka
- From the Ocular Genomics Institute, Department of Ophthalmology, Mass Eye and Ear and Harvard Medical School, Boston (E.A.P., J.I.C.), and Editas Medicine, Cambridge (B.S.A., K.K., A.R., M.C.J., R.L.M.) - both in Massachusetts; the Scheie Eye Institute and the Division of Ophthalmology of the Children's Hospital of Philadelphia, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia (T.S.A., A.M.M.); the University of Michigan Kellogg Eye Center, Ann Arbor (K.T.J.); the Bascom Palmer Eye Institute, University of Miami, Miami (B.L.L.); and the Casey Eye Institute, Oregon Health and Science University, Portland (S.T.B., A.K.L., M.E.P.)
| | - Rene L Myers
- From the Ocular Genomics Institute, Department of Ophthalmology, Mass Eye and Ear and Harvard Medical School, Boston (E.A.P., J.I.C.), and Editas Medicine, Cambridge (B.S.A., K.K., A.R., M.C.J., R.L.M.) - both in Massachusetts; the Scheie Eye Institute and the Division of Ophthalmology of the Children's Hospital of Philadelphia, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia (T.S.A., A.M.M.); the University of Michigan Kellogg Eye Center, Ann Arbor (K.T.J.); the Bascom Palmer Eye Institute, University of Miami, Miami (B.L.L.); and the Casey Eye Institute, Oregon Health and Science University, Portland (S.T.B., A.K.L., M.E.P.)
| | - Byron L Lam
- From the Ocular Genomics Institute, Department of Ophthalmology, Mass Eye and Ear and Harvard Medical School, Boston (E.A.P., J.I.C.), and Editas Medicine, Cambridge (B.S.A., K.K., A.R., M.C.J., R.L.M.) - both in Massachusetts; the Scheie Eye Institute and the Division of Ophthalmology of the Children's Hospital of Philadelphia, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia (T.S.A., A.M.M.); the University of Michigan Kellogg Eye Center, Ann Arbor (K.T.J.); the Bascom Palmer Eye Institute, University of Miami, Miami (B.L.L.); and the Casey Eye Institute, Oregon Health and Science University, Portland (S.T.B., A.K.L., M.E.P.)
| | - Steven T Bailey
- From the Ocular Genomics Institute, Department of Ophthalmology, Mass Eye and Ear and Harvard Medical School, Boston (E.A.P., J.I.C.), and Editas Medicine, Cambridge (B.S.A., K.K., A.R., M.C.J., R.L.M.) - both in Massachusetts; the Scheie Eye Institute and the Division of Ophthalmology of the Children's Hospital of Philadelphia, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia (T.S.A., A.M.M.); the University of Michigan Kellogg Eye Center, Ann Arbor (K.T.J.); the Bascom Palmer Eye Institute, University of Miami, Miami (B.L.L.); and the Casey Eye Institute, Oregon Health and Science University, Portland (S.T.B., A.K.L., M.E.P.)
| | - Jason I Comander
- From the Ocular Genomics Institute, Department of Ophthalmology, Mass Eye and Ear and Harvard Medical School, Boston (E.A.P., J.I.C.), and Editas Medicine, Cambridge (B.S.A., K.K., A.R., M.C.J., R.L.M.) - both in Massachusetts; the Scheie Eye Institute and the Division of Ophthalmology of the Children's Hospital of Philadelphia, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia (T.S.A., A.M.M.); the University of Michigan Kellogg Eye Center, Ann Arbor (K.T.J.); the Bascom Palmer Eye Institute, University of Miami, Miami (B.L.L.); and the Casey Eye Institute, Oregon Health and Science University, Portland (S.T.B., A.K.L., M.E.P.)
| | - Andreas K Lauer
- From the Ocular Genomics Institute, Department of Ophthalmology, Mass Eye and Ear and Harvard Medical School, Boston (E.A.P., J.I.C.), and Editas Medicine, Cambridge (B.S.A., K.K., A.R., M.C.J., R.L.M.) - both in Massachusetts; the Scheie Eye Institute and the Division of Ophthalmology of the Children's Hospital of Philadelphia, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia (T.S.A., A.M.M.); the University of Michigan Kellogg Eye Center, Ann Arbor (K.T.J.); the Bascom Palmer Eye Institute, University of Miami, Miami (B.L.L.); and the Casey Eye Institute, Oregon Health and Science University, Portland (S.T.B., A.K.L., M.E.P.)
| | - Albert M Maguire
- From the Ocular Genomics Institute, Department of Ophthalmology, Mass Eye and Ear and Harvard Medical School, Boston (E.A.P., J.I.C.), and Editas Medicine, Cambridge (B.S.A., K.K., A.R., M.C.J., R.L.M.) - both in Massachusetts; the Scheie Eye Institute and the Division of Ophthalmology of the Children's Hospital of Philadelphia, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia (T.S.A., A.M.M.); the University of Michigan Kellogg Eye Center, Ann Arbor (K.T.J.); the Bascom Palmer Eye Institute, University of Miami, Miami (B.L.L.); and the Casey Eye Institute, Oregon Health and Science University, Portland (S.T.B., A.K.L., M.E.P.)
| | - Mark E Pennesi
- From the Ocular Genomics Institute, Department of Ophthalmology, Mass Eye and Ear and Harvard Medical School, Boston (E.A.P., J.I.C.), and Editas Medicine, Cambridge (B.S.A., K.K., A.R., M.C.J., R.L.M.) - both in Massachusetts; the Scheie Eye Institute and the Division of Ophthalmology of the Children's Hospital of Philadelphia, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia (T.S.A., A.M.M.); the University of Michigan Kellogg Eye Center, Ann Arbor (K.T.J.); the Bascom Palmer Eye Institute, University of Miami, Miami (B.L.L.); and the Casey Eye Institute, Oregon Health and Science University, Portland (S.T.B., A.K.L., M.E.P.)
| |
Collapse
|
10
|
Xu P, Cooper RF, Jiang YY, Morgan JIW. Parafoveal cone function in choroideremia assessed with adaptive optics optoretinography. Sci Rep 2024; 14:8339. [PMID: 38594294 PMCID: PMC11004114 DOI: 10.1038/s41598-024-58059-x] [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: 12/13/2023] [Accepted: 03/25/2024] [Indexed: 04/11/2024] Open
Abstract
Choroideremia (CHM) is an X-linked retinal degeneration leading to loss of the photoreceptors, retinal pigment epithelium (RPE), and choroid. Adaptive optics optoretinography is an emerging technique for noninvasive, objective assessment of photoreceptor function. Here, we investigate parafoveal cone function in CHM using adaptive optics optoretinography and compare with cone structure and clinical assessments of vision. Parafoveal cone mosaics of 10 CHM and four normal-sighted participants were imaged with an adaptive optics scanning light ophthalmoscope. While acquiring video sequences, a 2 s 550Δ10 nm, 450 nW/deg2 stimulus was presented. Videos were registered and the intensity of each cone in each frame was extracted, normalized, standardized, and aggregated to generate the population optoretinogram (ORG) over time. A gamma-pdf was fit to the ORG and the peak was extracted as ORG amplitude. CHM ORG amplitudes were compared to normal and were correlated with bound cone density, ellipsoid zone to RPE/Bruch's membrane (EZ-to-RPE/BrM) distance, and foveal sensitivity using Pearson correlation analysis. ORG amplitude was significantly reduced in CHM compared to normal (0.22 ± 0.15 vs. 1.34 ± 0.31). In addition, CHM ORG amplitude was positively correlated with cone density, EZ-to-RPE/BrM distance, and foveal sensitivity. Our results demonstrate promise for using ORG as a biomarker of photoreceptor function.
Collapse
Affiliation(s)
- Peiluo Xu
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Robert F Cooper
- Department of Ophthalmology, Joint Department of Biomedical Engineering, Medical College of Wisconsin, Marquette University and Medical College of Wisconsin, Milwaukee, WI, 53233, USA
| | - Yu You Jiang
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jessica I W Morgan
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
11
|
Rohowetz LJ, Kunkler AL, Sengillo JD, Lazzarini TA, Lam BL, Berrocal AM. Choroideremia presenting as vision loss secondary to choroidal neovascularization. Ophthalmic Genet 2024; 45:175-179. [PMID: 37575054 DOI: 10.1080/13816810.2023.2245117] [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: 04/16/2023] [Revised: 06/23/2023] [Accepted: 08/01/2023] [Indexed: 08/15/2023]
Abstract
BACKGROUND Choroidal neovascularization (CNV) is a rare complication of choroideremia that occurs secondary to relative atrophy of the retinal pigment epithelium and eventual rupture of Bruch's membrane. The ideal management of CNV in choroideremia is unclear. MATERIALS AND METHODS Case report. OBSERVATIONS A 14-year-old male with no known ocular history presented to the eye emergency department complaining of a central scotoma in the right eye for 4 days. He had no past medical history and family history was unremarkable for known ocular disease. Visual acuity was 20/70 in the right eye and 20/30 in the left eye. Posterior segment exam revealed chorioretinal atrophy extending from the outer macula to the midperiphery in both eyes. There was CNV with associated subretinal hemorrhage in the right eye. Optical coherence tomography demonstrated the presence of CNV with subretinal fluid in the right eye and parafoveal outer retinal atrophy in both eyes. Genetic testing revealed a hemizygous exon 2 deletion on the CHM gene, pathogenic for choroideremia. The patient received a total of 3 injections 4 weeks apart followed by 1 injection 6 weeks later with resolution of the subretinal hemorrhage and reduction in CNV size with improvement in visual acuity to 20/20 at last follow-up exam. CONCLUSIONS AND IMPORTANCE Choroidal neovascularization is a rare cause of central vision loss in patients with choroideremia. In this report, we demonstrate a good functional and anatomic response to intravitreal bevacizumab in a 14-year-old patient with undiagnosed choroideremia who presented with CNV-induced central vision loss.
Collapse
Affiliation(s)
- Landon J Rohowetz
- Department of Ophthalmology, Bascom Palmer Eye Institute, Miami, Florida, USA
| | - Anne L Kunkler
- Department of Ophthalmology, Bascom Palmer Eye Institute, Miami, Florida, USA
| | - Jesse D Sengillo
- Department of Ophthalmology, Bascom Palmer Eye Institute, Miami, Florida, USA
| | - Thomas A Lazzarini
- Department of Ophthalmology, Bascom Palmer Eye Institute, Miami, Florida, USA
| | - Byron L Lam
- Department of Ophthalmology, Bascom Palmer Eye Institute, Miami, Florida, USA
| | - Audina M Berrocal
- Department of Ophthalmology, Bascom Palmer Eye Institute, Miami, Florida, USA
| |
Collapse
|
12
|
Wynne N, Jiang YY, Aleman TS, Morgan JIW. FOVEAL PHENOTYPES IN CHOROIDEREMIA ON ADAPTIVE OPTICS SCANNING LIGHT OPHTHALMOSCOPY. Retina 2024; 44:659-668. [PMID: 38531059 PMCID: PMC10972540 DOI: 10.1097/iae.0000000000003995] [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] [Indexed: 03/28/2024]
Abstract
PURPOSE Choroideremia is an X-linked inherited retinal degeneration involving the choriocapillaris, retinal pigment epithelium, and photoreceptors. Adaptive optics scanning light ophthalmoscopy allows visualization of retinal structure at the level of individual cells and is well poised to provide insight into the pathophysiologic mechanisms underpinning the retinal degeneration in choroideremia. METHODS Foveal adaptive optics scanning light ophthalmoscopy images of 102 eyes of 54 individuals with choroideremia were analyzed. Measures were compared with those from standard clinical imaging. Visual acuity was also measured and compared with quantitative foveal metrics. RESULTS The 3 distinct phenotypes observed were: relatively normal (5 eyes, 4 individuals), spiderweb (9 eyes, 7 individuals), and salt and pepper (87 eyes, 47 individuals). Peak cone density (86 eyes of 51 individuals) was significantly lower in choroideremia than in healthy retinas (P < 0.0001, range: 29,382-157,717 cones/mm2). Peak cone density was significantly related to extent of retained ellipsoid zone on en face optical coherence tomography (r2 = 0.47, P = 0.0009) and inversely related to visual acuity (r2 = 0.20, P = 0.001). CONCLUSION Distinct phenotypes can be observed on adaptive optics scanning light ophthalmoscopy imaging in choroideremia that cannot always be discerned on standard clinical imaging. Quantitative measures on adaptive optics imaging are related to the structural and functional severity of disease.
Collapse
Affiliation(s)
- Niamh Wynne
- Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Yu You Jiang
- Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania, Philadelphia, PA, United States
| | - Tomas S Aleman
- Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania, Philadelphia, PA, United States
| | - Jessica IW Morgan
- Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
13
|
Jolly JK, Rodda BM, Edwards TL, Ayton LN, Ruddle JB. Optical coherence tomography in children with inherited retinal disease. Clin Exp Optom 2024; 107:255-266. [PMID: 38252959 DOI: 10.1080/08164622.2023.2294807] [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: 02/13/2023] [Accepted: 12/06/2023] [Indexed: 01/24/2024] Open
Abstract
Recent advances have led to therapeutic options becoming available for people with inherited retinal disease. In particular, gene therapy has been shown to hold great promise for slowing vision loss from inherited retinal disease. Recent studies suggest that gene therapy is likely to be most effective when implemented early in the disease process, making consideration of paediatric populations important. It is therefore necessary to have a comprehensive understanding of retinal imaging in children with inherited retinal diseases, in order to monitor disease progression and to determine which early retinal biomarkers may be used as outcome measures in future clinical trials. In addition, as many optometrists will review children with an inherited retinal disease, an understanding of the expected imaging outcomes can improve clinical care. This review focuses on the most common imaging modality used in research assessment of paediatric inherited retinal diseases: optical coherence tomography. Optical coherence tomography findings can be used in both the clinical and research setting. In particular, the review discusses current knowledge of optical coherence tomography findings in eight paediatric inherited retinal diseases - Stargardt disease, Bests disease, Leber's congenital amaurosis, choroideremia, RPGR related retinitis pigmentosa, Usher syndrome, X-linked retinoschisis and, Batten disease.
Collapse
Affiliation(s)
- Jasleen K Jolly
- Vision and Eye Research Institute, Anglia Ruskin University, Cambridge, UK
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Brent M Rodda
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, Victoria, Australia
| | - Thomas L Edwards
- Ophthalmology, Department of Surgery, The University of Melbourne, East Melbourne, Victoria, Australia
- Centre for Eye Research Australia, The Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Lauren N Ayton
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, Victoria, Australia
- Ophthalmology, Department of Surgery, The University of Melbourne, East Melbourne, Victoria, Australia
- Centre for Eye Research Australia, The Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Jonathan B Ruddle
- Centre for Eye Research Australia, The Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
- Department of Ophthalmology, Royal Children's Hospital, Parkville, Victoria, Australia
| |
Collapse
|
14
|
Waldock WJ, Taylor LJ, Sperring S, Staurenghi F, Martinez-Fernandez de la Camara C, Whitfield J, Clouston P, Yusuf IH, MacLaren RE. A hypomorphic variant of choroideremia is associated with a novel intronic mutation that leads to exon skipping. Ophthalmic Genet 2024; 45:210-217. [PMID: 38273808 DOI: 10.1080/13816810.2023.2270554] [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: 07/31/2023] [Accepted: 10/09/2023] [Indexed: 01/27/2024]
Abstract
INTRODUCTION Molecular confirmation of pathogenic sequence variants in the CHM gene is required prior to enrolment in retinal gene therapy clinical trials for choroideremia. Individuals with mild choroideremia have been reported. The molecular basis of genotype-phenotype associations is of clinical relevance since it may impact on selection for retinal gene therapy. METHODS AND MATERIALS Genetic testing and RNA analysis were undertaken in a patient with mild choroideremia to confirm the pathogenicity of a novel intronic variant in CHM and to explore the mechanism underlying the mild clinical phenotype. RESULTS A 42-year-old male presented with visual field loss. Fundoscopy and autofluorescence imaging demonstrated mild choroideremia for his age. Genetic analysis revealed a variant at a splice acceptor site in the CHM gene (c.1350-3C > G). RNA analysis demonstrated two out-of-frame transcripts, suggesting pathogenicity, without any detectable wildtype transcripts. One of the two out-of-frame transcripts is present in very low levels in healthy controls. DISCUSSION Mild choroideremia may result from +3 or -3 splice site variants in CHM. It is presumed that the resulting mRNA transcripts may be partly functional, thereby preventing the development of the null phenotype. Choroideremia patients with such variants may present challenges for gene therapy since there may be residual transcript activity which could result in long-lasting visual function which is atypical for this disease.
Collapse
Affiliation(s)
| | - Laura J Taylor
- Oxford Eye Hospital, Oxford University Hospitals NHS Trust, Oxford, UK
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Sian Sperring
- Oxford Eye Hospital, Oxford University Hospitals NHS Trust, Oxford, UK
| | - Federica Staurenghi
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Cristina Martinez-Fernandez de la Camara
- Oxford Eye Hospital, Oxford University Hospitals NHS Trust, Oxford, UK
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | | | - Penny Clouston
- Oxford Regional Genetics Laboratories, Churchill Hospital, Oxford, UK
| | - Imran H Yusuf
- Oxford Eye Hospital, Oxford University Hospitals NHS Trust, Oxford, UK
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Robert E MacLaren
- Oxford Eye Hospital, Oxford University Hospitals NHS Trust, Oxford, UK
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
15
|
Tsui JC, Aleman TS, Tapino PJ, Kim BJ. Detailed Phenotype Supports Pathogenicity of Hypomorphic Variant in ABCC6-Associated Pattern Dystrophy. Case Rep Ophthalmol 2024; 15:497-506. [PMID: 39015234 PMCID: PMC11250126 DOI: 10.1159/000538045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 02/25/2024] [Indexed: 07/18/2024] Open
Abstract
Introduction We report a case of pseudoxanthoma elasticum (PXE) with an atypical phenotype likely related to a hypomorphic variant in ABCC6. Case Presentation A 66-year-old Caucasian female with a history of a maculopathy interpreted as either age-related macular degeneration or a pattern dystrophy underwent a detailed ophthalmic evaluation. Visual acuities were 20/25, OD, and 20/20, OS. Spectral domain optical coherence and fluorescein angiography demonstrated outer retinal disruptions and breaks in retinal pigment epithelium (RPE)/Bruch's membrane bilaterally, consistent with angioid streaks. A large area of hypo- and hyperautofluorescence extending from the central retina into the peripapillary retina was documented with short-wavelength excitation autofluorescence. The area of hypoautofluorescence, which was much larger on near-infrared excitation, spared the temporal retina. Two-color dark-adapted perimetries documented severe rod sensitivity losses and less severe cone sensitivity abnormalities co-localizing with the RPE abnormalities. No obvious skin findings were observed, and initial dermatologic biopsy was negative. Gene screening identified a pathogenic ABCC6 gene variant c.1552C>T and a previously reported variant of uncertain significance c.1171A>G. A second dermatologic biopsy demonstrated positive findings consistent with PXE. Conclusion Although this patient had minimal skin findings, this patient had characteristic structural and functional abnormalities of a pattern dystrophy with angioid streaks and histologic evidence of PXE, suggesting compound heterozygous variants involving the hypomorphic ABCC6 c.1171A>G variant. These findings support the pathogenic role of both variants.
Collapse
Affiliation(s)
- Jonathan C. Tsui
- Scheie Eye Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Ophthalmology, Veterans Affairs New Jersey Healthcare System, East Orange, NJ, USA
| | - Tomas S. Aleman
- Scheie Eye Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Advanced Retinal and Ocular Therapeutics (CAROT), University of Pennsylvania, Philadelphia, PA, USA
- Division of Ophthalmology, Children’s Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
| | - Paul J. Tapino
- Scheie Eye Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Benjamin J. Kim
- Scheie Eye Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
16
|
DeLuca AP, Whitmore SS, Tatro NJ, Andorf JL, Faga BP, Faga LA, Colins MM, Luse MA, Fenner BJ, Stone EM, Scheetz TE. Using Goldmann Visual Field Volume to Track Disease Progression in Choroideremia. OPHTHALMOLOGY SCIENCE 2023; 3:100397. [PMID: 38025158 PMCID: PMC10630671 DOI: 10.1016/j.xops.2023.100397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 08/31/2023] [Accepted: 09/06/2023] [Indexed: 12/01/2023]
Abstract
Purpose Choroideremia is an X-linked choroidopathy caused by pathogenic variants in the CHM gene. It is characterized by the early appearance of multiple scotomas in the peripheral visual field that spread and coalesce, usually sparing central vision until late in the disease. These features make quantitative monitoring of visual decline particularly challenging. Here, we describe a novel computational approach to convert Goldmann visual field (GVF) data into quantitative volumetric measurements. With this approach, we analyzed visual field loss in a longitudinal, retrospective cohort of patients with choroideremia. Design Single-center, retrospective, cohort study. Participants We analyzed data from 238 clinic visits of 56 molecularly-confirmed male patients with choroideremia from 41 families (range, 1-27 visits per patient). Patients had a median follow up of 4 years (range, 0-56 years) with an age range of 5 to 76 years at the time of their visits. Methods Clinical data from molecularly-confirmed patients with choroideremia, including GVF data, were included for analysis. Goldmann visual field records were traced using a tablet-based application, and the 3-dimensional hill of vision was interpolated for each trace. This procedure allowed quantification of visual field loss from data collected over decades with differing protocols, including different or incomplete isopters. Visual acuity (VA) data were collected and converted to logarithm of the minimum angle of resolution values. A delayed exponential mixed-effects model was used to evaluate the loss of visual field volume over time. Main Outcome Measures Visual acuity and GVF volume. Results The estimated mean age at disease onset was 12.6 years (standard deviation, 9.1 years; 95% quantile interval, 6.5-36.4 years). The mean field volume loss was 6.8% per year (standard deviation, 4.5%; 95% quantile interval, 1.9%-18.8%) based on exponential modeling. Field volume was more strongly correlated between eyes (r2 = 0.935) than best-corrected VA (r2 = 0.285). Conclusions Volumetric analysis of GVF data enabled quantification of peripheral visual function in patients with choroideremia and evaluation of disease progression. The methods presented here may facilitate the analysis of historical GVF data from patients with inherited retinal disease and other diseases associated with visual field loss. This work informs the creation of appropriate outcome measures in choroideremia therapeutic trials, particularly in trial designs. Financial Disclosures Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.
Collapse
Affiliation(s)
- Adam P. DeLuca
- The University of Iowa Institute for Vision Research & Department of Ophthalmology and Visual Sciences, Carver College of Medicine, The University of Iowa, Iowa City, Iowa
| | - S. Scott Whitmore
- The University of Iowa Institute for Vision Research & Department of Ophthalmology and Visual Sciences, Carver College of Medicine, The University of Iowa, Iowa City, Iowa
| | - Nicole J. Tatro
- The University of Iowa Institute for Vision Research & Department of Ophthalmology and Visual Sciences, Carver College of Medicine, The University of Iowa, Iowa City, Iowa
| | - Jeaneen L. Andorf
- The University of Iowa Institute for Vision Research & Department of Ophthalmology and Visual Sciences, Carver College of Medicine, The University of Iowa, Iowa City, Iowa
| | - Ben P. Faga
- The University of Iowa Institute for Vision Research & Department of Ophthalmology and Visual Sciences, Carver College of Medicine, The University of Iowa, Iowa City, Iowa
| | - Laurel A. Faga
- The University of Iowa Institute for Vision Research & Department of Ophthalmology and Visual Sciences, Carver College of Medicine, The University of Iowa, Iowa City, Iowa
| | - Malia M. Colins
- The University of Iowa Institute for Vision Research & Department of Ophthalmology and Visual Sciences, Carver College of Medicine, The University of Iowa, Iowa City, Iowa
| | - Meagan A. Luse
- The University of Iowa Institute for Vision Research & Department of Ophthalmology and Visual Sciences, Carver College of Medicine, The University of Iowa, Iowa City, Iowa
| | - Beau J. Fenner
- The University of Iowa Institute for Vision Research & Department of Ophthalmology and Visual Sciences, Carver College of Medicine, The University of Iowa, Iowa City, Iowa
- Department of Medical Retina, Singapore National Eye Centre, Singapore
- Singapore Eye Research Institute, Singapore
- Ophthalmology and Visual Sciences Academic Clinical Programme, SingHealth Duke-NUS Academic Medical Centre, Duke-NUS Graduate Medical School, Singapore
| | - Edwin M. Stone
- The University of Iowa Institute for Vision Research & Department of Ophthalmology and Visual Sciences, Carver College of Medicine, The University of Iowa, Iowa City, Iowa
| | - Todd E. Scheetz
- The University of Iowa Institute for Vision Research & Department of Ophthalmology and Visual Sciences, Carver College of Medicine, The University of Iowa, Iowa City, Iowa
| |
Collapse
|
17
|
Rodriguez-Martinez AC, Higgins BE, Tailor-Hamblin V, Malka S, Cheloni R, Collins AM, Bladen J, Henderson R, Moosajee M. Foveal Hypoplasia in CRB1-Related Retinopathies. Int J Mol Sci 2023; 24:13932. [PMID: 37762234 PMCID: PMC10531165 DOI: 10.3390/ijms241813932] [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: 07/25/2023] [Revised: 09/05/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
The CRB1 gene plays a role in retinal development and its maintenance. When disrupted, it gives a range of phenotypes such as early-onset severe retinal dystrophy/Leber congenital amaurosis (EOSRD/LCA), retinitis pigmentosa (RP), cone-rod dystrophy (CORD) and macular dystrophy (MD). Studies in CRB1 retinopathies have shown thickening and coarse lamination of retinal layers resembling an immature retina. Its role in foveal development has not yet been described; however, this retrospective study is the first to report foveal hypoplasia (FH) presence in a CRB1-related retinopathy cohort. Patients with pathogenic biallelic CRB1 variants from Moorfields Eye Hospital, London, UK, were collected. Demographic, clinical data and SD-OCT analyses with FH structural grading were performed. A total of 15 (48%) patients had EOSRD/LCA, 11 (35%) MD, 3 (9%) CORD and 2 (6%) RP. FH was observed in 20 (65%; CI: 0.47-0.79) patients, all of whom were grade 1. A significant difference in BCVA between patients with FH and without was found (p = 0.014). BCVA continued to worsen over time in both groups (p < 0.001), irrespective of FH. This study reports FH in a CRB1 cohort, supporting the role of CRB1 in foveal development. FH was associated with poorer BCVA and abnormal retinal morphology. Nonetheless, its presence did not alter the disease progression.
Collapse
Affiliation(s)
- Ana Catalina Rodriguez-Martinez
- UCL Institute of Ophthalmology, London EC1V 9EL, UK; (A.C.R.-M.); (B.E.H.); (V.T.-H.); (S.M.); (R.C.); (A.M.C.); (R.H.)
- Moorfields Eye Hospital NHS Foundation Trust, London EC1V 2PD, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 1LE, UK
| | - Bethany Elora Higgins
- UCL Institute of Ophthalmology, London EC1V 9EL, UK; (A.C.R.-M.); (B.E.H.); (V.T.-H.); (S.M.); (R.C.); (A.M.C.); (R.H.)
- Moorfields Eye Hospital NHS Foundation Trust, London EC1V 2PD, UK
| | - Vijay Tailor-Hamblin
- UCL Institute of Ophthalmology, London EC1V 9EL, UK; (A.C.R.-M.); (B.E.H.); (V.T.-H.); (S.M.); (R.C.); (A.M.C.); (R.H.)
- Moorfields Eye Hospital NHS Foundation Trust, London EC1V 2PD, UK
- UCL Experimental Psychology, London WC1H 0AP, UK
| | - Samantha Malka
- UCL Institute of Ophthalmology, London EC1V 9EL, UK; (A.C.R.-M.); (B.E.H.); (V.T.-H.); (S.M.); (R.C.); (A.M.C.); (R.H.)
- Moorfields Eye Hospital NHS Foundation Trust, London EC1V 2PD, UK
| | - Riccardo Cheloni
- UCL Institute of Ophthalmology, London EC1V 9EL, UK; (A.C.R.-M.); (B.E.H.); (V.T.-H.); (S.M.); (R.C.); (A.M.C.); (R.H.)
- Moorfields Eye Hospital NHS Foundation Trust, London EC1V 2PD, UK
| | - Alexander Mark Collins
- UCL Institute of Ophthalmology, London EC1V 9EL, UK; (A.C.R.-M.); (B.E.H.); (V.T.-H.); (S.M.); (R.C.); (A.M.C.); (R.H.)
| | - John Bladen
- King’s College Hospital NHS Foundation Trust, Strand, London WC2R 2LS, UK;
| | - Robert Henderson
- UCL Institute of Ophthalmology, London EC1V 9EL, UK; (A.C.R.-M.); (B.E.H.); (V.T.-H.); (S.M.); (R.C.); (A.M.C.); (R.H.)
- Moorfields Eye Hospital NHS Foundation Trust, London EC1V 2PD, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 1LE, UK
| | - Mariya Moosajee
- UCL Institute of Ophthalmology, London EC1V 9EL, UK; (A.C.R.-M.); (B.E.H.); (V.T.-H.); (S.M.); (R.C.); (A.M.C.); (R.H.)
- Moorfields Eye Hospital NHS Foundation Trust, London EC1V 2PD, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 1LE, UK
- The Francis Crick Institute, London NW1 1AT, UK
| |
Collapse
|
18
|
Poli FE, Yusuf IH, Jolly JK, Taylor LJ, Adeyoju D, Josan AS, Birtel J, Charbel Issa P, Cehajic-Kapetanovic J, Da Cruz L, MacLaren RE. Correlation Between Fundus Autofluorescence Pattern and Retinal Function on Microperimetry in Choroideremia. Transl Vis Sci Technol 2023; 12:24. [PMID: 37773503 PMCID: PMC10547012 DOI: 10.1167/tvst.12.9.24] [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: 03/16/2023] [Accepted: 08/29/2023] [Indexed: 10/01/2023] Open
Abstract
Purpose In patients with choroideremia, it is not known how smooth and mottled patterns on short-wavelength fundus autofluorescence (AF) imaging relate to retinal function. Methods A retrospective case-note review was undertaken on 190 patients with choroideremia at two specialist centers for retinal genetics. Twenty patients with both smooth and mottled zones on short-wavelength AF imaging and concurrent mesopic microperimetry assessments were included. Mean retinal sensitivities within the smooth and mottled zones were compared between choroideremia patients, and identical points on mesopic microperimetry collected from 12 age-matched controls. Longitudinal analyses were undertaken at 2 and 5 years in a subset of patients. Results In patients with choroideremia, mean retinal sensitivities at baseline were significantly greater in the smooth zone (26.1 ± 2.0 dB) versus the mottled zone (20.5 ± 4.2 dB) (P < 0.0001). Mean retinal sensitivities at baseline were similar in the smooth zone between choroideremia patients and controls (P = 0.054) but significantly impaired in the mottled zone in choroideremia compared to controls (P < 0.0001). The rate of decline in total sensitivity over 5 years was not significant in either the smooth or mottled zone in a small subset of choroideremia patients (n = 7; P = 0.344). Conclusions In choroideremia, retinal sensitivity as determined by microperimetry correlates with patterns on AF imaging: retinal function in the smooth zone, where the retinal pigment epithelium is anatomically preserved, is similar to controls, but retinal sensitivity in the mottled zone is impaired. Translational Relevance Patterns on AF imaging may represent a novel, objective outcome measure for clinical trials in choroideremia as a surrogate for retinal function.
Collapse
Affiliation(s)
- Federica E. Poli
- Nuffield Laboratory of Ophthalmology, University of Oxford, UK
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Imran H. Yusuf
- Nuffield Laboratory of Ophthalmology, University of Oxford, UK
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Jasleen K. Jolly
- Nuffield Laboratory of Ophthalmology, University of Oxford, UK
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Vision and Eye Research Institute, School of Medicine, Anglia Ruskin University, Cambridge, UK
| | - Laura J. Taylor
- Nuffield Laboratory of Ophthalmology, University of Oxford, UK
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Daniel Adeyoju
- Nuffield Laboratory of Ophthalmology, University of Oxford, UK
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Amandeep S. Josan
- Nuffield Laboratory of Ophthalmology, University of Oxford, UK
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Johannes Birtel
- Nuffield Laboratory of Ophthalmology, University of Oxford, UK
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Department of Ophthalmology, University Hospital of Bonn, Bonn, Germany
- Department of Ophthalmology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Peter Charbel Issa
- Nuffield Laboratory of Ophthalmology, University of Oxford, UK
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Jasmina Cehajic-Kapetanovic
- Nuffield Laboratory of Ophthalmology, University of Oxford, UK
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Lyndon Da Cruz
- Institute of Ophthalmology, University College London, London, UK
- Moorfields Eye Hospital, London, UK
| | - Robert E. MacLaren
- Nuffield Laboratory of Ophthalmology, University of Oxford, UK
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| |
Collapse
|
19
|
Gocuk SA, Jolly JK, Edwards TL, Ayton LN. Female carriers of X-linked inherited retinal diseases - Genetics, diagnosis, and potential therapies. Prog Retin Eye Res 2023; 96:101190. [PMID: 37406879 DOI: 10.1016/j.preteyeres.2023.101190] [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: 03/09/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 07/07/2023]
Abstract
Inherited retinal diseases (IRDs) are a group of heterogeneous conditions that cause progressive vision loss, typically due to monogenic mutations. Female carriers of X-linked IRDs have a single copy of the disease-causing gene, and therefore, may exhibit variable clinical signs that vary from near normal retina to severe disease and vision loss. The relationships between individual genetic mutations and disease severity in X-linked carriers requires further study. This review summarises the current literature surrounding the spectrum of disease seen in female carriers of choroideremia and X-linked retinitis pigmentosa. Various classification systems are contrasted to accurately grade retinal disease. Furthermore, genetic mechanisms at the early embryonic stage are explored to potentially explain the variability of disease seen in female carriers. Future research in this area will provide insight into the association between genotype and retinal phenotypes of female carriers, which will guide in the management of these patients. This review acknowledges the importance of identifying which patients may be at high risk of developing severe symptoms, and therefore should be considered for emerging treatments, such as retinal gene therapy.
Collapse
Affiliation(s)
- Sena A Gocuk
- Department of Optometry and Vision Sciences, The University of Melbourne, Melbourne, Victoria, Australia; Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, The University of Melbourne, Melbourne, Victoria, Australia
| | - Jasleen K Jolly
- Vision and Eye Research Institute, Anglia Ruskin University, Cambridge, UK
| | - Thomas L Edwards
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, The University of Melbourne, Melbourne, Victoria, Australia
| | - Lauren N Ayton
- Department of Optometry and Vision Sciences, The University of Melbourne, Melbourne, Victoria, Australia; Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, The University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
20
|
Xu P, Jiang YY, Morgan JIW. Cone Photoreceptor Morphology in Choroideremia Assessed Using Non-Confocal Split-Detection Adaptive Optics Scanning Light Ophthalmoscopy. Invest Ophthalmol Vis Sci 2023; 64:36. [PMID: 37504961 PMCID: PMC10383007 DOI: 10.1167/iovs.64.10.36] [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: 04/06/2023] [Accepted: 07/10/2023] [Indexed: 07/29/2023] Open
Abstract
Purpose Choroideremia (CHM) is an X-linked inherited retinal degeneration causing loss of the photoreceptors, retinal pigment epithelium, and choriocapillaris, although patients typically retain a central island of relatively preserved, functioning retina until late-stage disease. Here, we investigate cone photoreceptor morphology within the retained retinal island by examining cone inner segment area, density, circularity, and intercone space. Methods Using a custom-built, multimodal adaptive optics scanning light ophthalmoscope, nonconfocal split-detection images of the photoreceptor mosaic were collected at 1°, 2°, and 4° temporal to the fovea from 13 CHM and 12 control subjects. Cone centers were manually identified, and cone borders were segmented. A custom MATLAB script was used to extract area and circularity for each cone and calculate the percentage of intercone space in each region of interest. Bound cone density was also calculated. An unbalanced two-way ANOVA and Bonferroni post hoc tests were used to assess statistical differences between the CHM and control groups and along retinal eccentricity. Results Cone density was lower in the CHM group than in the control group (P < 0.001) and decreased with eccentricity from the fovea (P < 0.001). CHM cone inner segments were larger in area (P < 0.001) and more circular (P = 0.042) than those of the controls. Intercone space in CHM was also higher than in the controls (P < 0.001). Conclusions Cone morphology is altered in CHM compared to control, even within the centrally retained, functioning retinal area. Further studies are required to determine whether such morphology is a precursor to cone degeneration.
Collapse
Affiliation(s)
- Peiluo Xu
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Yu You Jiang
- Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania, United States
- Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Jessica I. W. Morgan
- Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania, United States
- Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| |
Collapse
|
21
|
Yusuf IH, MacLaren RE. Choroideremia: Toward Regulatory Approval of Retinal Gene Therapy. Cold Spring Harb Perspect Med 2023; 13:a041279. [PMID: 37277205 PMCID: PMC10691480 DOI: 10.1101/cshperspect.a041279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Choroideremia is an X-linked inherited retinal degeneration characterized by primary centripetal degeneration of the retinal pigment epithelium (RPE), with secondary degeneration of the choroid and retina. Affected individuals experience reduced night vision in early adulthood with blindness in late middle age. The underlying CHM gene encodes REP1, a protein involved in the prenylation of Rab GTPases essential for intracellular vesicle trafficking. Adeno-associated viral gene therapy has demonstrated some benefit in clinical trials for choroideremia. However, challenges remain in gaining regulatory approval. Choroideremia is slowly progressive, which presents difficulties in demonstrating benefit over short pivotal clinical trials that usually run for 1-2 years. Improvements in visual acuity are particularly challenging due to the initial negative effects of surgical detachment of the fovea. Despite these challenges, great progress toward a treatment has been made since choroideremia was first described in 1872.
Collapse
Affiliation(s)
- Imran H Yusuf
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, United Kingdom
- Oxford Eye Hospital, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, United Kingdom
| | - Robert E MacLaren
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, United Kingdom
- Oxford Eye Hospital, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, United Kingdom
| |
Collapse
|
22
|
Zhai Y, Xu M, Radziwon A, Dimopoulos IS, Crichton P, Mah R, MacLaren RE, Somani R, Tennant MT, MacDonald IM. AAV2-Mediated Gene Therapy for Choroideremia: 5-Year Results and Alternate Anti-sense Oligonucleotide Therapy. Am J Ophthalmol 2023; 248:145-156. [PMID: 36581191 DOI: 10.1016/j.ajo.2022.12.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 11/03/2022] [Accepted: 12/14/2022] [Indexed: 12/27/2022]
Abstract
PURPOSE To assess the long-term safety and efficacy of AAV2-REP1 in choroideremia (CHM) patients, and to test a potential antisense oligonucleotide therapy for CHM. DESIGN Extended, prospective phase 1/2 clinical trial and laboratory investigation. METHODS Five patients who received a single subfoveal injection of AAV2-REP1 were studied. The long-term safety was evaluated by ophthalmic examination, spectral domain optical coherence tomography, and fundus autofluorescence (FAF) for up to 5 years. Functional and structural changes were determined by different test modalities. Four antisense oligonucleotides (ASOs) were designed to treat the CHM c.1245-521A>G mutation, which was present in 2 patients within this trial. RESULTS Subject P3 experienced a localized intraretinal immune response that resulted in a significant loss of preserved retinal pigment epithelium (RPE). P4 experienced an exacerbation of peripheral retinoschisis. P2 had a constant ≥15-letter best-corrected visual acuity (BCVA) gain in the treated eye, whereas P5 had ≥15-letter BCVA improvement once in the untreated eye. The preserved FAF areas declined more rapidly in the treated eyes compared to the untreated eyes (P = .043). A customized 25-mer ASO recovered 83.2% to 95.0% of the normal RNA and 57.5% of the normal protein in fibroblasts from 2 trial patients. CONCLUSIONS Intraretinal inflammation triggered by AAV2-REP1 subretinal injection stabilized after 2 years but resulted in permanent damage to the retinal structure. Long-term progression of the disease was seen in both treated and untreated eyes, casting doubt as to the effectiveness of this approach in late-stage CHM. Alternative approaches such as ASO may have a therapeutic effect in a subgroup of CHM patients.
Collapse
Affiliation(s)
- Yi Zhai
- From the Department of Ophthalmology and Visual Sciences (Y.Z., M.X., A.R., R.S., M.T.T., I.M.M.), University of Alberta, Edmonton, Alberta, Canada
| | - Manlong Xu
- From the Department of Ophthalmology and Visual Sciences (Y.Z., M.X., A.R., R.S., M.T.T., I.M.M.), University of Alberta, Edmonton, Alberta, Canada
| | - Alina Radziwon
- From the Department of Ophthalmology and Visual Sciences (Y.Z., M.X., A.R., R.S., M.T.T., I.M.M.), University of Alberta, Edmonton, Alberta, Canada; Department of Medical Genetics (A.R., I.M.M.), University of Alberta, Edmonton, Alberta, Canada
| | - Ioannis S Dimopoulos
- Department of Ophthalmology (I.S.D., P.C., R.M.), University of Ottawa, Ottawa, Ontario, Canada
| | - Paul Crichton
- Department of Ophthalmology (I.S.D., P.C., R.M.), University of Ottawa, Ottawa, Ontario, Canada
| | - Rachel Mah
- Department of Ophthalmology (I.S.D., P.C., R.M.), University of Ottawa, Ottawa, Ontario, Canada
| | - Robert E MacLaren
- Nuffield Laboratory of Ophthalmology (R.E.M.), Department of Clinical Neurosciences, University of Oxford and NIHR Oxford Biomedical Research Centre, Oxfordshire, UK
| | - Rizwan Somani
- From the Department of Ophthalmology and Visual Sciences (Y.Z., M.X., A.R., R.S., M.T.T., I.M.M.), University of Alberta, Edmonton, Alberta, Canada
| | - Matthew T Tennant
- From the Department of Ophthalmology and Visual Sciences (Y.Z., M.X., A.R., R.S., M.T.T., I.M.M.), University of Alberta, Edmonton, Alberta, Canada
| | - Ian M MacDonald
- From the Department of Ophthalmology and Visual Sciences (Y.Z., M.X., A.R., R.S., M.T.T., I.M.M.), University of Alberta, Edmonton, Alberta, Canada; Department of Medical Genetics (A.R., I.M.M.), University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
23
|
O'Neil EC, Uyhazi KE, O'Connor K, Aleman IA, Pulido JS, Rossano JW, Aleman TS. DANON DISEASE: A MODEL OF PHOTORECEPTOR DEGENERATION SECONDARY TO PRIMARY RETINAL PIGMENT EPITHELIUM DISEASE. Retin Cases Brief Rep 2022; 16:707-713. [PMID: 36288619 DOI: 10.1097/icb.0000000000001125] [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: 06/16/2023]
Abstract
PURPOSE To describe in detail the retinal phenotype of LAMP2-associated Danon disease. METHODS Three LAMP2-positive patients from two unrelated families were studied with spectral-domain optical coherence tomography and with short-wavelength and near-infrared fundus autofluorescence (FAF) imaging. Visual function was measured with full-field electroretinography and chromatic perimetry. A patient with choroideremia was also studied for comparison. RESULTS A 45-year-old LAMP2-heterozygous woman, her 21-year-old hemizygous son, and an unrelated heterozygous 60-year-old woman had normal visual acuities. Central spectral-domain optical coherence tomographies were grossly normal in the younger two patients (mother and son). The oldest patient showed a tenuous interdigitation signal, interruptions of the inner segment ellipsoid zone band, and parafoveal outer nuclear layer thinning. Quantitatively, all patients had shorter than normal ellipsoid zone to retinal pigment epithelium distance in pericentral retina, normal at the foveola. A speckled hypoautofluorescence pattern on short-wavelength FAF contrasted with grossly abnormal near-infrared FAF in the heterozygous carriers. The oldest patient had reduced full-field electroretinography amplitudes (to ∼50% of normal) for rod- and cone-mediated responses and her perimetry showed severe rod dysfunction but substantial cone function. A disproportionate loss of the near-infrared FAF compared with the short-wavelength FAF, predominantly outer segment changes, and severe rod dysfunction with preserved cone function was similarly documented in a 9-year-old choroideremia hemizygous patient. CONCLUSION A disproportionate loss of the near-infrared FAF signal compared with the short-wavelength FAF signal, outer segment abnormalities, and severe rod dysfunction but relatively preserved cone vision suggests a stereotypical pattern of primary retinal pigment epithelial or parallel retinal pigment epithelial + photoreceptor disease in Danon disease.
Collapse
Affiliation(s)
- Erin C O'Neil
- University of Pennsylvania, Philadelphia, Pennsylvania
- Division of Ophthalmology of the Children's Hospital of Philadelphia, Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Keli O'Connor
- University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Jose S Pulido
- Department of Ophthalmology, Jefferson Kimmel Medical School, Wills Eye Hospital, Philadelphia, Pennsylvania; and
- Divisions of Pediatric Cardiology and Critical Care Medicine of The Children's Hospital of Philadelphia and the Department of Pediatrics of the University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Joseph W Rossano
- Divisions of Pediatric Cardiology and Critical Care Medicine of The Children's Hospital of Philadelphia and the Department of Pediatrics of the University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Tomas S Aleman
- University of Pennsylvania, Philadelphia, Pennsylvania
- Division of Ophthalmology of the Children's Hospital of Philadelphia, Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
24
|
Edwards MM, McLeod DS, Grebe R, Bhutto IA, Dahake R, Crumley K, Lutty GA. Glial remodeling and choroidal vascular pathology in eyes from two donors with Choroideremia. FRONTIERS IN OPHTHALMOLOGY 2022; 2:994566. [PMID: 38983545 PMCID: PMC11182301 DOI: 10.3389/fopht.2022.994566] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 09/26/2022] [Indexed: 07/11/2024]
Abstract
Choroideremia (CHM) is a recessive, X-linked disease that affects 1 in 50,000 people worldwide. CHM causes night blindness in teenage years with vision loss progressing over the next two to three decades. While CHM is known to cause progressive loss of retinal pigment epithelial (RPE) cells, photoreceptors and choroidal vessels, little attention has been given to retinal glial changes in eyes with CHM. In addition, while choroidal loss has been observed clinically, no histopathologic assessment of choroidal loss has been done. We investigated glial remodeling and activation as well as choriocapillaris changes and their association with RPE loss in postmortem eyes from two donors with CHM. Eyes were fixed and cryopreserved or the retina and choroid/RPE were processed as flatmounts with a small piece cut for transmission electron microscopy. A dense glial membrane, made up of vimentin and GFAP double-positive cells, occupied the subretinal space in the area of RPE and photoreceptor loss of both eyes. The membranes did not extend into the far periphery, where RPE and photoreceptors were viable. A glial membrane was also found on the vitreoretinal surface. Transmission electron microscopy analysis demonstrated prominence and disorganization of glial cells, which contained exosome-like vesicles. UEA lectin demonstrated complete absence of choriocapillaris in areas with RPE loss while some large choroidal vessels remained viable. In the far periphery, where the RPE monolayer was intact, choriocapillaris appeared normal. The extensive glial remodeling present in eyes with CHM should be taken into account when therapies such as stem cell replacement are considered as it could impede cells entering the retina. This gliosis would also need to be reversed to some extent for Müller cells to perform their normal homeostatic functions in the retina. Future studies investigating donor eyes as well as clinical imaging from carriers or those with earlier stages of CHM will prove valuable in understanding the glial changes, which could affect disease progression if they occur early. This would also provide insights into the progression of disease in the photoreceptor/RPE/choriocapillaris complex, which is crucial for identifying new treatments and finding the windows for treatment.
Collapse
Affiliation(s)
- Malia M Edwards
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - D Scott McLeod
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Rhonda Grebe
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Imran A Bhutto
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Richa Dahake
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Kelly Crumley
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Gerard A Lutty
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
25
|
Clinical and imaging findings of choroideremia in a pediatric patient due to a novel frameshift mutation. Am J Ophthalmol Case Rep 2022; 28:101718. [PMID: 36247208 PMCID: PMC9562444 DOI: 10.1016/j.ajoc.2022.101718] [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: 05/01/2022] [Revised: 09/15/2022] [Accepted: 10/02/2022] [Indexed: 11/21/2022] Open
Abstract
Purpose To describe the clinical characteristics, imaging findings and genetic testing results of a young simplex male with choroideremia. Observations A 6-year-old Hispanic-Chinese male was referred to the retina clinic for peripheral retinal pigmentary changes observed in both eyes on routine exam. The patient has an unremarkable family history and developmental history. Best corrected visual acuity was 20/25 in both eyes. Optical coherence tomography demonstrated attenuation of the ellipsoid and interdigitation zones. Widefield fundus autofluorescence demonstrated nummular hypo-autofluorescence peripherally in both eyes. Genetic testing revealed a variant originally described as a variant of uncertain significance (VUS) a c. 1775_1814del (p.Glu592Valfs*44) identified in the CHM gene, which was reclassified as pathogenic following segregation analysis. The patient was diagnosed with choroideremia due to a CHM pathogenic variant. Conclusions The multimodal imaging findings demonstrated here illustrate important clues to the diagnosis of Choroideremia in a simplex male.
Collapse
|
26
|
Widespread subclinical cellular changes revealed across a neural-epithelial-vascular complex in choroideremia using adaptive optics. Commun Biol 2022; 5:893. [PMID: 36100689 PMCID: PMC9470576 DOI: 10.1038/s42003-022-03842-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 08/12/2022] [Indexed: 11/08/2022] Open
Abstract
AbstractChoroideremia is an X-linked, blinding retinal degeneration with progressive loss of photoreceptors, retinal pigment epithelial (RPE) cells, and choriocapillaris. To study the extent to which these layers are disrupted in affected males and female carriers, we performed multimodal adaptive optics imaging to better visualize the in vivo pathogenesis of choroideremia in the living human eye. We demonstrate the presence of subclinical, widespread enlarged RPE cells present in all subjects imaged. In the fovea, the last area to be affected in choroideremia, we found greater disruption to the RPE than to either the photoreceptor or choriocapillaris layers. The unexpected finding of patches of photoreceptors that were fluorescently-labeled, but structurally and functionally normal, suggests that the RPE blood barrier function may be altered in choroideremia. Finally, we introduce a strategy for detecting enlarged cells using conventional ophthalmic imaging instrumentation. These findings establish that there is subclinical polymegathism of RPE cells in choroideremia.
Collapse
|
27
|
Malechka VV, Duong D, Bordonada KD, Turriff A, Blain D, Murphy E, Introne WJ, Gochuico BR, Adams DR, Zein WM, Brooks BP, Huryn LA, Solomon BD, Hufnagel RB. Investigating Determinants and Evaluating Deep Learning Training Approaches for Visual Acuity in Foveal Hypoplasia. OPHTHALMOLOGY SCIENCE 2022; 3:100225. [PMID: 36339947 PMCID: PMC9634033 DOI: 10.1016/j.xops.2022.100225] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 08/30/2022] [Accepted: 09/19/2022] [Indexed: 11/13/2022]
Abstract
Purpose To describe the relationships between foveal structure and visual function in a cohort of individuals with foveal hypoplasia (FH) and to estimate FH grade and visual acuity using a deep learning classifier. Design Retrospective cohort study and experimental study. Participants A total of 201 patients with FH were evaluated at the National Eye Institute from 2004 to 2018. Methods Structural components of foveal OCT scans and corresponding clinical data were analyzed to assess their contributions to visual acuity. To automate FH scoring and visual acuity correlations, we evaluated the following 3 inputs for training a neural network predictor: (1) OCT scans, (2) OCT scans and metadata, and (3) real OCT scans and fake OCT scans created from a generative adversarial network. Main Outcome Measures The relationships between visual acuity outcomes and determinants, such as foveal morphology, nystagmus, and refractive error. Results The mean subject age was 24.4 years (range, 1–73 years; standard deviation = 18.25 years) at the time of OCT imaging. The mean best-corrected visual acuity (n = 398 eyes) was equivalent to a logarithm of the minimal angle of resolution (LogMAR) value of 0.75 (Snellen 20/115). Spherical equivalent refractive error (SER) ranged from −20.25 diopters (D) to +13.63 D with a median of +0.50 D. The presence of nystagmus and a high-LogMAR value showed a statistically significant relationship (P < 0.0001). The participants whose SER values were farther from plano demonstrated higher LogMAR values (n = 382 eyes). The proportion of patients with nystagmus increased with a higher FH grade. Variability in SER with grade 4 (range, −20.25 D to +13.00 D) compared with grade 1 (range, −8.88 D to +8.50 D) was statistically significant (P < 0.0001). Our neural network predictors reliably estimated the FH grading and visual acuity (correlation to true value > 0.85 and > 0.70, respectively) for a test cohort of 37 individuals (98 OCT scans). Training the predictor on real OCT scans with metadata and fake OCT scans improved the accuracy over the model trained on real OCT scans alone. Conclusions Nystagmus and foveal anatomy impact visual outcomes in patients with FH, and computational algorithms reliably estimate FH grading and visual acuity.
Collapse
|
28
|
Parmann R, Greenstein VC, Tsang SH, Sparrow JR. Choroideremia Carriers: Dark-Adapted Perimetry and Retinal Structures. Invest Ophthalmol Vis Sci 2022; 63:4. [PMID: 35816046 PMCID: PMC9284471 DOI: 10.1167/iovs.63.8.4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose In choroideremia (CHM) carriers, scotopic sensitivity was assessed by dark adapted chromatic perimetry (DACP) and outer retinal structure was evaluated by multimodal imaging. Methods Nine carriers (18 eyes) and 13 healthy controls (13 eyes) underwent DACP testing with cyan and red stimuli. Analysis addressed peripapillary (4 test locations closest to the optic disc), macular (52 locations), and peripheral (60 locations outside the macula) regions. Responses were considered to be rod-mediated when cyan relative to red sensitivity was >5 dB. Fundus imaging included spectral domain optical coherence tomography (SD-OCT), short-wavelength (SW-AF), near-infrared (NIR-AF), ultrawide-field (200 degrees) pseudocolor fundus imaging, and quantitative (qAF) fundus autofluorescence. Results Detection of the cyan stimulus was rod mediated in essentially all test locations (99.7%). In the macular and peripheral areas, DACP sensitivity values were not significantly different from healthy eyes. In the peripapillary area, sensitivities were significantly decreased (P < 0.05). SD-OCT imaging ranged from hyper-reflective lesions and discontinuities of the outer retinal bands to hypertransmission of signal. SW-AF and NIR-AF images presented with peripapillary atrophy in seven patients (14 eyes). Mosaicism was detectable in SW-AF images in seven patients and in NIR-AF images in five patients. Frank hypo-autofluorescence was visible in eight patients with distinct chorioretinopathy in seven patients. The qAF values were below the 95% confidence interval (CI) of healthy age-matched individuals in 12 eyes. Conclusions Rod mediated scotopic sensitivity was comparable to that in control eyes in macular and peripheral areas but was decreased in the peripapillary area where changes in retinal structure were also most severe.
Collapse
Affiliation(s)
- Rait Parmann
- Departments of Ophthalmology, Columbia University, New York, NY, United States
| | | | - Stephen H Tsang
- Departments of Ophthalmology, Columbia University, New York, NY, United States.,Departments of Pathology and Cell Biology, Columbia University, New York, NY, United States
| | - Janet R Sparrow
- Departments of Ophthalmology, Columbia University, New York, NY, United States.,Departments of Pathology and Cell Biology, Columbia University, New York, NY, United States
| |
Collapse
|
29
|
Ratra D, Chattree S, Raviselvan M, Pradhan A, Giridhar S. Structural and functional phenotypic features and molecular analysis of Indian patients with Bietti crystalline dystrophy. Indian J Ophthalmol 2022; 70:2526-2532. [PMID: 35791149 PMCID: PMC9426155 DOI: 10.4103/ijo.ijo_2146_21] [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] [Indexed: 12/02/2022] Open
Abstract
Purpose: Bietti crystalline dystrophy (BCD) is a rare retinal dystrophy, uncommon in Indians. This study describes the various phenotypic features seen in the Indian population. Methods: In this retrospective, descriptive case series, records of patients with either clinical or molecular diagnosis of BCD from 2009 to 2020 were perused. Phenotypic and genotype information was collected and analyzed. Results: This study included 58 patients of BCD (31 males) aged 21–79 years (mean: 47 ± 14 years). The age at onset ranged from 7 to 41 years (mean: 28.8 ± 5.1 years). Vision ranged from 20/20 to counting fingers. There were 18 (31%) patients with stage 1 with crystals and mild retinochoroidal atrophy, 22 (38%) with stage 2 with atrophy extending beyond arcades, and 18 (31%) with absent crystals and extensive atrophy of stage 3. Choroidal neovascular membrane was seen in four patients. The optical coherence tomography showed retinochoroidal thinning (84.6%), outer retinal tubulations (71.8%), and paradoxical foveal thickening with interlaminar bridges (7.7%). Electrophysiology and visual fields showed reduced responses in advanced retinochoroidal changes. Molecular confirmation was available in five patients; five mutations were seen in the CYP4V2. Conclusion: A wide variation is seen in the phenotypic picture of BCD. A molecular diagnosis is helpful in differentiating from other retinal dystrophies. The OCT shows the peculiar feature of the interlaminar bridge in early cases with photoreceptor loss. Further investigations into this OCT feature may provide insights into the pathogenesis of BCD. A genotype–phenotype correlation could not be done.
Collapse
Affiliation(s)
- Dhanashree Ratra
- Department of Vitreoretinal Diseases, Medical Research Foundation, Sankara Nethralaya, Chennai, Tamil Nadu, India
| | - Surabhi Chattree
- Department of Vitreoretinal Diseases, Medical Research Foundation, Sankara Nethralaya, Chennai, Tamil Nadu, India
| | - Munispriyan Raviselvan
- Department of Vitreoretinal Diseases, Medical Research Foundation, Sankara Nethralaya, Chennai, Tamil Nadu, India
| | - Arkaprava Pradhan
- Department of Vitreoretinal Diseases, Medical Research Foundation, Sankara Nethralaya, Chennai, Tamil Nadu, India
| | - Sneha Giridhar
- Department of Vitreoretinal Diseases, Medical Research Foundation, Sankara Nethralaya, Chennai, Tamil Nadu, India
| |
Collapse
|
30
|
AAV2-hCHM Subretinal Delivery to the Macula in Choroideremia: Two Year Interim Results of an Ongoing Phase I/II Gene Therapy Trial. Ophthalmology 2022; 129:1177-1191. [PMID: 35714735 DOI: 10.1016/j.ophtha.2022.06.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 06/03/2022] [Accepted: 06/07/2022] [Indexed: 11/22/2022] Open
Abstract
PURPOSE To assess the safety of the subretinal delivery of a recombinant adeno-associated virus serotype 2 (AAV2) vector carrying a human CHM-encoding cDNA in choroideremia (CHM). DESIGN Prospective, open-label, non-randomized, dose-escalation, phase 1/2 clinical trial. SUBJECTS, PARTICIPANTS, AND/OR CONTROLS Fifteen CHM patients (ages 20-57 years at dosing). METHODS, INTERVENTION, OR TESTING Patients received uniocular subfoveal injections of low dose (up to 5x1010 vector genome (vg) per eye, n=5) or high dose (up to 1x1011 vg per eye, n=10) AAV2-hCHM. Patients were evaluated pre- and post-operatively for two years with ophthalmic examinations, multimodal retinal imaging and psychophysical testing. MAIN OUTCOME Measures: visual acuity (VA), perimetry (10-2 protocol), spectral-domain optical coherence tomography (SD-OCT) and short-wavelength fundus autofluorescence (SW-FAF). RESULTS We detected no vector-related or systemic toxicities. VA returned to within 15 letters of baseline in all but two patients (one developed acute foveal thinning, another patient, a macular hole); the rest showed no gross changes in foveal structure at two years. There were no significant differences between intervention and control eyes in mean light-adapted sensitivity by perimetry, or in the lateral extent of retinal pigment epithelium (RPE) relative preservation by SD-OCT and SW-FAF. Microperimetry showed non-significant (<3SD of the intervisit variability) gains in sensitivity in some locations and participants in the intervention eye. There were no obvious dose-dependent relationships. CONCLUSIONS VA was within 15 letters of baseline after the subfoveal AAV2-hCHM injections in 13/15 (87%) of the patients. Acute foveal thinning with unchanged perifoveal function in one patient and macular hole in a second suggests foveal vulnerability to the subretinal injections. Longer observation intervals will help establish the significance of the minor differences in sensitivities and rate of disease progression observed between intervention and control eyes.
Collapse
|
31
|
Morgan JIW, Jiang YY, Vergilio GK, Serrano LW, Pearson DJ, Bennett J, Maguire AM, Aleman TS. Short-term Assessment of Subfoveal Injection of Adeno-Associated Virus-Mediated hCHM Gene Augmentation in Choroideremia Using Adaptive Optics Ophthalmoscopy. JAMA Ophthalmol 2022; 140:411-420. [PMID: 35266957 PMCID: PMC8914909 DOI: 10.1001/jamaophthalmol.2022.0158] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 01/17/2022] [Indexed: 11/14/2022]
Abstract
Importance Subretinal injection for gene augmentation in retinal degenerations forcefully detaches the neural retina from the retinal pigment epithelium, potentially damaging photoreceptors and/or retinal pigment epithelium cells. Objective To use adaptive optics scanning light ophthalmoscopy (AOSLO) to assess the short-term integrity of the cone mosaic following subretinal injections of adeno-associated virus vector designed to deliver a functional version of the CHM gene (AAV2-hCHM) in patients with choroideremia. Design, Setting, and Participants This longitudinal case series study enrolled adult patients with choroideremia from February 2015 to January 2016 in the US. To be included in the study, study participants must have received uniocular subfoveal injections of low-dose (5 × 1010 vector genome per eye) or high-dose (1 × 1011 vector genome per eye) AAV2-hCHM. Analysis began February 2015. Main Outcomes and Measures The macular regions of both eyes were imaged before and 1 month after injection using a custom-built multimodal AOSLO. Postinjection cone inner segment mosaics were compared with preinjection mosaics at multiple regions of interest. Colocalized spectral-domain optical coherence tomography and dark-adapted cone sensitivity was also acquired at each time point. Results Nine study participants ranged in age from 26 to 50 years at the time of enrollment, and all were White men. Postinjection AOSLO images showed preservation of the cone mosaic in all 9 AAV2-hCHM-injected eyes. Mosaics appeared intact and contiguous 1 month postinjection, with the exception of foveal disruption in 1 patient. Optical coherence tomography showed foveal cone outer segment shortening postinjection. Cone-mediated sensitivities were unchanged in 8 of 9 injected and 9 of 9 uninjected eyes. One participant showed acute loss of foveal optical coherence tomography cone outer segment-related signals along with cone sensitivity loss that colocalized with disruption of the mosaic on AOSLO. Conclusions and Relevance Integrity of the cone mosaic is maintained following subretinal delivery of AAV2-hCHM, providing strong evidence in support of the safety of the injections. Minor foveal thinning observed following surgery corresponds with short-term cone outer segment shortening rather than cone cell loss. Foveal cone loss in 1 participant raises the possibility of individual vulnerability to the subretinal injection.
Collapse
Affiliation(s)
- Jessica I. W. Morgan
- Scheie Eye Institute, University of Pennsylvania, Philadelphia
- Center for Advanced Retinal & Ocular Therapeutics, University of Pennsylvania, Philadelphia
| | - Yu You Jiang
- Scheie Eye Institute, University of Pennsylvania, Philadelphia
- Center for Advanced Retinal & Ocular Therapeutics, University of Pennsylvania, Philadelphia
| | - Grace K. Vergilio
- Scheie Eye Institute, University of Pennsylvania, Philadelphia
- Center for Advanced Retinal & Ocular Therapeutics, University of Pennsylvania, Philadelphia
| | - Leona W. Serrano
- Scheie Eye Institute, University of Pennsylvania, Philadelphia
- Center for Advanced Retinal & Ocular Therapeutics, University of Pennsylvania, Philadelphia
| | - Denise J. Pearson
- Scheie Eye Institute, University of Pennsylvania, Philadelphia
- Center for Advanced Retinal & Ocular Therapeutics, University of Pennsylvania, Philadelphia
| | - Jean Bennett
- Scheie Eye Institute, University of Pennsylvania, Philadelphia
- Center for Advanced Retinal & Ocular Therapeutics, University of Pennsylvania, Philadelphia
| | - Albert M. Maguire
- Scheie Eye Institute, University of Pennsylvania, Philadelphia
- Center for Advanced Retinal & Ocular Therapeutics, University of Pennsylvania, Philadelphia
| | - Tomas S. Aleman
- Scheie Eye Institute, University of Pennsylvania, Philadelphia
- Center for Advanced Retinal & Ocular Therapeutics, University of Pennsylvania, Philadelphia
| |
Collapse
|
32
|
Bilateral visual acuity decline in males with choroideremia: a pooled, cross-sectional meta-analysis. BMC Ophthalmol 2022; 22:29. [PMID: 35034620 PMCID: PMC8762852 DOI: 10.1186/s12886-022-02250-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 12/30/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Choroideremia is a rare inherited retinal disease that leads to blindness. Visual acuity (VA) is a key outcome measure in choroideremia treatment studies, but VA decline rates change with age. An accurate understanding of the natural deterioration of VA in choroideremia is important to assess the treatment effect of new therapies in which VA is the primary outcome measure. We conducted a meta-analysis of data on individuals with choroideremia to determine the rate of VA deterioration between the better- and worse-seeing eye (BSE and WSE, respectively). METHODS Data were collected from the prospective Natural History of the Progression of Choroideremia (NIGHT) study (613 eyes, baseline data only), studies included in a recent meta-analysis, and studies identified in a targeted literature search performed on March 25, 2020, including individual best-corrected VA (BCVA) and age data in male individuals with choroideremia. Best-corrected VA decline rates (measured by logMAR units) by age and trends in BCVA decline rates in the BSE and WSE were evaluated. RESULTS: Data from 1037 males (1602 eyes; mean age, 41.8 years) were included. Before and after an age cutoff of 33.8 years, BCVA decline rates for the WSE were 0.0086 and 0.0219 logMAR per year, respectively. Before and after an age cutoff of 39.1 years, BCVA decline rates for the BSE were 0.00001 and 0.0203 logMAR per year, respectively. Differences in absolute BCVA and decline rates increased between the 2 eyes until age ~ 40; thereafter, differences in absolute BCVA and decline rates were similar between eyes. CONCLUSIONS Using the largest choroideremia data set to date, this analysis demonstrates accelerated BCVA decline beginning between 30 and 40 years of age. Disparate interocular progression rates were observed before the transition age, with similar interocular progression rates after the transition age.
Collapse
|
33
|
Young BK, Shen LL, Del Priore LV. An In Silica Model for RPE Loss Patterns in Choroideremia. Invest Ophthalmol Vis Sci 2021; 62:10. [PMID: 34779822 PMCID: PMC8606796 DOI: 10.1167/iovs.62.14.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose To use empirical data to develop a model of cell loss in choroideremia that predicts the known exponential rate of RPE loss and central, scalloped preservation pattern seen in this disease. Methods A computational model of RPE loss was created in Python 3.7, which constructed an array of RPE cells clusters, binarized as either live or atrophic. Two rules were applied to this model: the background effect gave each cell a chance of dying defined by a background function, and the neighbor effect increased the chance of RPE cell death if a neighbor were dead. The known anatomic distribution of rods, RPE, choriocapillaris density, amacrine, ganglion, and cone cells were derived from the literature and applied to this model. Atrophy growth rates were measured over arbitrary time units and fit to the known exponential decay model. The main outcome measures: included topography of atrophy over time and fit of simulated residual RPE area to exponential decay. Results A background effect alone can simulate exponential decay, but does not simulate the central island preservation seen in choroideremia. An additive neighbor effect alone does not simulate exponential decay. When the neighbor effect multiplies the background effect using the rod density function, our model follows an exponential decay, similar to previous observations. Also, our model predicts a residual island of RPE that resembles the topographic distribution of residual RPE seen in choroideremia. Conclusions The pattern of RPE loss in choroideremia can be predicted by applying simple rules. The RPE preservation pattern typically seen in choroideremia may be related to the underlying pattern of rod density. Further studies are needed to validate these findings.
Collapse
Affiliation(s)
- Benjamin K Young
- W.K. Kellogg Eye Center, University of Michigan School of Medicine, Ann Arbor, MI, United States
| | - Liangbo L Shen
- Department of Ophthalmology, University of California San Francisco, San Francisco, San Francisco, CA, United States
| | - Lucian V Del Priore
- Department of Ophthalmology and Visual Sciences, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
34
|
Nuzbrokh Y, Ragi SD, Tsang SH. Gene therapy for inherited retinal diseases. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1278. [PMID: 34532415 PMCID: PMC8421966 DOI: 10.21037/atm-20-4726] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 11/04/2020] [Indexed: 01/04/2023]
Abstract
Inherited retinal diseases (IRDs) are a genetically variable collection of devastating disorders that lead to significant visual impairment. Advances in genetic characterization over the past two decades have allowed identification of over 260 causative mutations associated with inherited retinal disorders. Thought to be incurable, gene supplementation therapy offers great promise in treating various forms of these blinding conditions. In gene replacement therapy, a disease-causing gene is replaced with a functional copy of the gene. These therapies are designed to slow disease progression and hopefully restore visual function. Gene therapies are typically delivered to target retinal cells by subretinal (SR) or intravitreal (IVT) injection. The historic Food and Drug Administration (FDA) approval of voretigene neparvovec for RPE65-associated Leber's congenital amaurosis (LCA) spurred tremendous optimism surrounding retinal gene therapy for various other monogenic IRDs. Novel disease-causing mutations continue to be discovered annually, and targeted genetic therapy is now under development in clinical and preclinical models for many IRDs. Numerous clinical trials for other IRDs are ongoing or have recently completed. Disorders being targeted for genetic therapy include retinitis pigmentosa (RP), choroideremia (CHM), achromatopsia (ACHM), Leber's hereditary optic neuropathy, usher syndrome (USH), X-linked retinoschisis, and Stargardt disease. Here, we provide an update of completed, ongoing, and planned clinical trials using gene supplementation strategies for retinal degenerative disorders.
Collapse
Affiliation(s)
- Yan Nuzbrokh
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, New York, NY, USA.,Jonas Children's Vision Care, New York, NY, USA.,Renaissance School of Medicine at Stony Brook University, Stony Brook, New York, NY, USA
| | - Sara D Ragi
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, New York, NY, USA.,Jonas Children's Vision Care, New York, NY, USA
| | - Stephen H Tsang
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, New York, NY, USA.,Jonas Children's Vision Care, New York, NY, USA.,Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
35
|
Fry LE, Patrício MI, Jolly JK, Xue K, MacLaren RE. Expression of Rab Prenylation Pathway Genes and Relation to Disease Progression in Choroideremia. Transl Vis Sci Technol 2021; 10:12. [PMID: 34254989 PMCID: PMC8287038 DOI: 10.1167/tvst.10.8.12] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Choroideremia results from the deficiency of Rab Escort Protein 1 (REP1), encoded by CHM, involved in the prenylation of Rab GTPases. Here, we investigate whether the transcription and expression of other genes involved in the prenylation of Rab proteins correlates with disease progression in a cohort of patients with choroideremia. Methods Rates of retinal pigment epithelial area loss in 41 patients with choroideremia were measured using fundus autofluorescence imaging for up to 4 years. From lysates of cultured skin fibroblasts donated by patients (n = 15) and controls (n = 14), CHM, CHML, RABGGTB and RAB27A mRNA expression, and REP1 and REP2 protein expression were compared. Results The central autofluorescent island area loss in patients with choroideremia occurred with a mean half-life of 5.89 years (95% confidence interval [CI] = 5.09-6.70), with some patients demonstrating relatively fast or slow rates of progression (range = 3.3-14.1 years). Expression of CHM mRNA and REP1 protein were significantly decreased in all patients. No difference in expression of CHML, RABGGTB, RAB27A, or REP2 was seen between patients and controls. No correlation was seen between expression of the genes analyzed and rates of retinal degeneration. Non-sense induced transcriptional compensation of CHML, a CHM-like retrogene, was not observed in patients with CHM variants predicted to undergo non-sense mediated decay. Conclusions Patients with choroideremia, who are deficient for REP1, show normal levels of expression of other genes involved in Rab prenylation, which do not appear to play any modifying role in the rate of disease progression. Translational Relevance There remains little evidence for selection of patients for choroideremia gene therapy based on genotype.
Collapse
Affiliation(s)
- Lewis E Fry
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK.,Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Maria I Patrício
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK.,Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Jasleen K Jolly
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK.,Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Kanmin Xue
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK.,Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Robert E MacLaren
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK.,Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| |
Collapse
|
36
|
Compound Heterozygous Mutations in ZNF408 in a Patient with a Late Onset Pigmentary Retinopathy and Relatively Preserved Central Retina. Doc Ophthalmol 2021; 143:305-312. [PMID: 34259982 DOI: 10.1007/s10633-021-09847-7] [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: 03/08/2021] [Accepted: 06/24/2021] [Indexed: 12/31/2022]
Abstract
PURPOSE To describe in detail the phenotype of a patient with compound heterozygous mutations in ZNF408 and an adult-onset pigmentary retinopathy rather than familial exudative vitreoretinopathy as expected with heterozygous mutations in this gene. METHODS A 70-year-old male presented with a pigmentary retinopathy, which prompted a genetic evaluation that revealed two variants in trans in the ZNF408 gene. He underwent an ophthalmic examination, kinetic fields, electroretinography (ERG), spectral-domain optical coherence tomography (SD-OCT), fundus autofluorescence, wide-angle fluorescein angiography and near-infrared imaging. RESULTS Visual acuity was 20/20 for both eyes. Fundus examination showed epiretinal membrane, vascular attenuation and peripheral bone spicule pigmentation in both eyes. Fluorescein angiography showed no vascular anomalies in both eyes. Fundus autofluorescence showed a preserved island of fundus autofluorescence centrally. Visual field by kinetic perimetry (V-4e stimulus) showed generalized constriction to 40 degrees of eccentricity and by an I-4e target showed generalized constriction to 10 degrees of eccentricity. ERG showed detectable but reduced cone-mediated responses. SD-OCT demonstrated preserved outer nuclear layer thickness centrally, which decreased with eccentricity. Static perimetry showed substantial rod and cone sensitivities centrally that declined with eccentricity. A next-generation sequencing panel revealed bi-allelic variants (p.Arg567Ter; c.1699C > T and p.Leu566His; c.1697 T > A) in the ZNF408 gene. CONCLUSIONS ZNF408-associated retinal dystrophies can present with predominantly retinal findings and should be considered in the differential diagnosis of retinitis pigmentosa. Our study revealed a novel variant p.L566H, which to our knowledge has not previously been reported.
Collapse
|
37
|
Wynne N, Carroll J, Duncan JL. Promises and pitfalls of evaluating photoreceptor-based retinal disease with adaptive optics scanning light ophthalmoscopy (AOSLO). Prog Retin Eye Res 2021; 83:100920. [PMID: 33161127 PMCID: PMC8639282 DOI: 10.1016/j.preteyeres.2020.100920] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/28/2020] [Accepted: 10/31/2020] [Indexed: 12/15/2022]
Abstract
Adaptive optics scanning light ophthalmoscopy (AOSLO) allows visualization of the living human retina with exquisite single-cell resolution. This technology has improved our understanding of normal retinal structure and revealed pathophysiological details of a number of retinal diseases. Despite the remarkable capabilities of AOSLO, it has not seen the widespread commercial adoption and mainstream clinical success of other modalities developed in a similar time frame. Nevertheless, continued advancements in AOSLO hardware and software have expanded use to a broader range of patients. Current devices enable imaging of a number of different retinal cell types, with recent improvements in stimulus and detection schemes enabling monitoring of retinal function, microscopic structural changes, and even subcellular activity. This has positioned AOSLO for use in clinical trials, primarily as exploratory outcome measures or biomarkers that can be used to monitor disease progression or therapeutic response. AOSLO metrics could facilitate patient selection for such trials, to refine inclusion criteria or to guide the choice of therapy, depending on the presence, absence, or functional viability of specific cell types. Here we explore the potential of AOSLO retinal imaging by reviewing clinical applications as well as some of the pitfalls and barriers to more widespread clinical adoption.
Collapse
Affiliation(s)
- Niamh Wynne
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Joseph Carroll
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jacque L Duncan
- Department of Ophthalmology, University of California, San Francisco, CA, USA.
| |
Collapse
|
38
|
Abstract
PURPOSE To evaluate choroidal features in young patients affected by choroideremia (CHM). METHODS Young CHM patients and control subjects were recruited at the Eye Clinic in Florence. High-resolution choroidal imaging was obtained using swept-source optical coherence tomography with long optical coherence tomography scans (12 × 9 mm optical coherence tomography scans). We considered the subfoveal choroidal area within 9 mm of the optic disk in the horizontal plane and the subfoveal choroidal area within a 3-mm diameter centered over the fovea. The subfoveal choroidal thickness, total choroidal area, luminal area, stromal area, and choroidal vascularity index were assessed using the "ImageJ" software in both groups. RESULTS Eight patients (16 eyes; mean age, 19.3 ± 5.2 years) and seven control subjects (14 eyes; mean age, 19.0 ± 5.0 years) were included in this study. Best-corrected visual acuity was 20/20 in both eyes of seven CHM patients and in all control subjects and 20/25 in both eyes in one CHM patient. Mean subfoveal choroidal thickness did not differ between CHM patients and control subjects. Luminal area9mm, stromal area9mm, and total choroidal area9mm were reduced in patients compared with the control group. Luminal area3mm, stromal area3mm, and total choroidal area3mm did not differ between patients and control subjects. Choroidal vascularity index9mm and choroidal vascularity index3mm were not different between patients and control subjects. CONCLUSION There are no differences in the choroidal vascularity index between young CHM patients and control subjects; this result suggests a simultaneous, proportional impairment of both the stromal and vascular components of the choroid in the early stages of the disease.
Collapse
|
39
|
Aleman TS, O'Neil EC, O'Connor K, Jiang YY, Aleman IA, Bennett J, Morgan JIW, Toussaint BW. Bardet-Biedl syndrome-7 ( BBS7) shows treatment potential and a cone-rod dystrophy phenotype that recapitulates the non-human primate model. Ophthalmic Genet 2021; 42:252-265. [PMID: 33729075 DOI: 10.1080/13816810.2021.1888132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Purpose: To provide a detailed ophthalmic phenotype of two male patients with Bardet-Biedl Syndrome (BBS) due to mutations in the BBS7 geneMethods: Two brothers ages 26 (Patient 1, P1) and 23 (P2) underwent comprehensive ophthalmic evaluations over three years. Visual function was assessed with full-field electroretinograms (ffERGs), kinetic and chromatic perimetry, multimodal imaging with spectral domain optical coherence tomography (SD-OCT), fundus autofluorescence (FAF) with short- (SW) and near-infrared (NIR) excitation lights and adaptive optics scanning light ophthalmoscopy (AOSLO).Results: Both siblings had a history of obesity and postaxial polydactyly; P2 had diagnoses of type 1 Diabetes Mellitus, Addison's disease, high-functioning autism-spectrum disorder and -12D myopia. Visual acuities were better than 20/30. Kinetic fields were moderately constricted. Cone-mediated ffERGs were undetectable, rod ERGs were ~80% of normal mean. Static perimetry showed severe central cone and rod dysfunction. Foveal to parafoveal hypoautofluorescence, most obvious on NIR-FAF, co-localized with outer segment shortening/loss and outer nuclear layer thinning by SD-OCT, and with reduced photoreceptors densities by AOSLO. A structural-functional dissociation was confirmed for cone- and rod-mediated parameters. Worsening of the above abnormalities was documented by SD-OCT and FAF in P2 at 3 years. Gene screening identified compound heterozygous mutations in BBS7 (p.Val266Glu: c.797 T > A of maternal origin; c.1781_1783delCAT, paternal) in both patients.Conclusions: BBS7-associated retinal degeneration may present as a progressive cone-rod dystrophy pattern, reminiscent of both the murine and non-human primate models of the disease. Predominantly central retinal abnormalities in both cone and rod photoreceptors showed a structural-functional dissociation, an ideal scenario for gene augmentation treatments.
Collapse
Affiliation(s)
- Tomas S Aleman
- Scheie Eye Institute at the Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Division of Ophthalmology of the Children's Hospital of Philadelphia, Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Erin C O'Neil
- Scheie Eye Institute at the Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Division of Ophthalmology of the Children's Hospital of Philadelphia, Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Keli O'Connor
- Scheie Eye Institute at the Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yu You Jiang
- Scheie Eye Institute at the Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Isabella A Aleman
- Scheie Eye Institute at the Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jean Bennett
- Scheie Eye Institute at the Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jessica I W Morgan
- Scheie Eye Institute at the Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Brian W Toussaint
- Christiana Care Health System, Wilmington, Delaware, USA.,Department of Ophthalmology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
40
|
Aleman TS, Miller AJ, Maguire KH, Aleman EM, Serrano LW, O'Connor KB, Bedoukian EC, Leroy BP, Maguire AM, Bennett J. A Virtual Reality Orientation and Mobility Test for Inherited Retinal Degenerations: Testing a Proof-of-Concept After Gene Therapy. Clin Ophthalmol 2021; 15:939-952. [PMID: 33688162 PMCID: PMC7936670 DOI: 10.2147/opth.s292527] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 01/22/2021] [Indexed: 12/19/2022] Open
Abstract
Purpose To test the ability of a virtual reality (VR) orientation and mobility (O&M) protocol to serve a measure of functional vision for patients with inherited retinal degenerations (IRDs). Methods A VR-O&M protocol designed using a commercially available VR hardware was tested in normally sighted control subjects (n=7; ages 10–35yo; Average 22.5yo) and patients with RPE65-associated Leber Congenital Amaurosis (n=3; ages 7–18yo; Average 12.7yo), in two of them before and after gene therapy. Patients underwent perimetry and full-field sensitivity testing. VR-O&M parameters correlated with the visual dysfunction. Results Visual acuities in RPE65 patients were on average worse than 20/200, dark-adapted sensitivity losses >5 log units, and fields constricted between 20° and 40°. Before treatment, patients required ~1000-fold brighter environment to navigate, had at least x4 more collisions, and were slower both to orient and navigate compared to control subjects. Improvements in cone- (by 1–2 L.u.) and rod-mediated (by >4 L.u.) sensitivities post-treatment led to fewer collisions (at least by half) at ~100-fold dimmer luminances, and to x4 times faster navigation times. Conclusion This study provides proof-of-concept data in support for the use of VR-O&M systems to quantify the impact that the visual dysfunction and improvement of vision following treatments has on functional vision in IRDs. The VR-O&M was useful in potentially challenging scenarios such as in pediatric patients with severe IRDs. Translational Relevance A VR-O&M test will provide much needed flexibility, both in its deployment as well as in the possibility to test various attributes of vision that may be impacted by gene therapy in the setting of translational studies. Precis This study provides proof-of-concept data in support for the use of a virtual reality orientation and mobility test to quantify the impact of the disease and of treatments thereof on functional vision in inherited retinal degenerations.
Collapse
Affiliation(s)
- Tomas S Aleman
- Scheie Eye Institute at the Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania, Philadelphia, PA, USA.,Division of Ophthalmology at the Children's Hospital of Philadelphia of the Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA, USA
| | - Alexander J Miller
- Neurology Virtual Reality Laboratory of the Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Katherine H Maguire
- Scheie Eye Institute at the Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| | - Elena M Aleman
- Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| | - Leona W Serrano
- Scheie Eye Institute at the Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| | - Keli B O'Connor
- Scheie Eye Institute at the Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| | - Emma C Bedoukian
- Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Bart P Leroy
- Division of Ophthalmology at the Children's Hospital of Philadelphia of the Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA, USA.,Department of Pediatrics, Ghent University, Ghent, Belgium.,Department of Ophthalmology, Ghent University, Ghent, Belgium.,Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Albert M Maguire
- Scheie Eye Institute at the Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania, Philadelphia, PA, USA.,Division of Ophthalmology at the Children's Hospital of Philadelphia of the Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA, USA
| | - Jean Bennett
- Scheie Eye Institute at the Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
41
|
O'Hare F, Edwards TL, Hu ML, Hickey DG, Zhang AC, Wang JH, Liu Z, Ayton LN. An optometrist's guide to the top candidate inherited retinal diseases for gene therapy. Clin Exp Optom 2021; 104:431-443. [PMID: 33689629 DOI: 10.1080/08164622.2021.1878851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
This review presents the phenotypic and genotypic profiles of a select group of inherited retinal diseases (IRDs) that are currently the focus of retinal gene therapy trials globally. Research progress in IRD treatment trials may soon lead to their availability in Australia and New Zealand, as either approved treatment or a clinical trial. The salient clinical characteristics of retinitis pigmentosa-the largest IRD category-are highlighted, with specific reference to RPE65-associated Leber congenital amaurosis, followed by other specific IRDs, namely choroideremia and ABCA4-associated Stargardt disease. These IRDs are selected based on their candidacy for gene therapy. Guidance on the clinical diagnostic tests that support each of these diagnoses will be presented. More broadly, the most useful structure and function measures to monitor IRD progression is discussed, along with the key assessments that offer differential diagnostic insight. This review is intended to be a clinical guide for optometrists, to assist in assessment and management of individuals who may be eligible for current and future gene therapies. A companion article in this issue will provide an overview of the basic principles of gene therapy and its development as a new treatment for inherited retinal diseases.
Collapse
Affiliation(s)
- Fleur O'Hare
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia.,Department of Surgery (Ophthalmology), The University of Melbourne, Melbourne, Australia.,Department of Optometry and Vision Sciences, The University of Melbourne, Melbourne, Australia
| | - Thomas L Edwards
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia.,Department of Surgery (Ophthalmology), The University of Melbourne, Melbourne, Australia
| | - Monica L Hu
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
| | - Doron G Hickey
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia.,Department of Surgery (Ophthalmology), The University of Melbourne, Melbourne, Australia
| | - Alexis C Zhang
- Department of Surgery (Ophthalmology), The University of Melbourne, Melbourne, Australia.,Department of Optometry and Vision Sciences, The University of Melbourne, Melbourne, Australia
| | - Jiang-Hui Wang
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
| | - Zhengyang Liu
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
| | - Lauren N Ayton
- Department of Surgery (Ophthalmology), The University of Melbourne, Melbourne, Australia.,Department of Optometry and Vision Sciences, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
42
|
Iovino C, Di Iorio V, Testa F, Bombace V, Melillo P, Vupparaboina KK, Chhablani J, Simonelli F. Choroidal Vascularity Features in Patients with Choroideremia and Cystoid Spaces. Diagnostics (Basel) 2021; 11:diagnostics11030382. [PMID: 33668232 PMCID: PMC7996232 DOI: 10.3390/diagnostics11030382] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 02/20/2021] [Accepted: 02/21/2021] [Indexed: 11/16/2022] Open
Abstract
Cystoid spaces (CSs) are a common retinal finding in choroideremia (CHM) patients. The aim of this study was to analyze the vascular features of the choroid associated with the presence of CSs in patients with confirmed genetic diagnosis of CHM. A total of 33 patients (33 eyes) were enrolled in this retrospective cross-sectional study and divided into two groups based on the presence (17 eyes) or absence (16 eyes) of CSs. Choroidal features were evaluated on spectral-domain optical coherence tomography including subfoveal choroidal thickness (CT), total choroidal area (TCA), luminal choroidal area (LCA), and stromal choroidal area (SCA). The choroidal vascularity index (CVI) was then calculated in all study eyes. All structural choroidal parameters were calculated both on the entire length of the B-scan and in the central subfoveal 1500 μm. The average age was 37.3 ± 11.6 and 31.4 ± 16.7 years (p = 0.25) and mean logMAR best-corrected visual acuity was 0.11 ± 0.20 and 0.20 ± 0.57 (p = 0.54) in the CHM groups with and without CSs, respectively. There were no significant differences in subfoveal CT, and TCA, LCA, SCA, and CVI evaluated on either the entire scan or in the central 1500 μm (all p > 0.05). All choroidal vasculature parameters exhibited no significant differences between CHM eyes with and without CSs. Our results suggest that the choroid may not be involved in the development of CSs in patients with CHM.
Collapse
Affiliation(s)
- Claudio Iovino
- Eye Clinic, Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania ‘Luigi Vanvitelli’, Via Pansini 5, 80131 Naples, Italy; (C.I.); (V.D.I.); (F.T.); (V.B.); (P.M.)
| | - Valentina Di Iorio
- Eye Clinic, Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania ‘Luigi Vanvitelli’, Via Pansini 5, 80131 Naples, Italy; (C.I.); (V.D.I.); (F.T.); (V.B.); (P.M.)
| | - Francesco Testa
- Eye Clinic, Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania ‘Luigi Vanvitelli’, Via Pansini 5, 80131 Naples, Italy; (C.I.); (V.D.I.); (F.T.); (V.B.); (P.M.)
| | - Viviana Bombace
- Eye Clinic, Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania ‘Luigi Vanvitelli’, Via Pansini 5, 80131 Naples, Italy; (C.I.); (V.D.I.); (F.T.); (V.B.); (P.M.)
| | - Paolo Melillo
- Eye Clinic, Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania ‘Luigi Vanvitelli’, Via Pansini 5, 80131 Naples, Italy; (C.I.); (V.D.I.); (F.T.); (V.B.); (P.M.)
| | - Kiran Kumar Vupparaboina
- Smt. Kanuri Santhamma Centre for Vitreo-Retinal Diseases, L V Prasad Eye Institute, Hyderabad 500034, India;
| | - Jay Chhablani
- Department of Ophthalmology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA;
| | - Francesca Simonelli
- Eye Clinic, Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania ‘Luigi Vanvitelli’, Via Pansini 5, 80131 Naples, Italy; (C.I.); (V.D.I.); (F.T.); (V.B.); (P.M.)
- Correspondence: ; Tel.: +39-081-7704501
| |
Collapse
|
43
|
Zeitz C, Nassisi M, Laurent-Coriat C, Andrieu C, Boyard F, Condroyer C, Démontant V, Antonio A, Lancelot ME, Frederiksen H, Kloeckener-Gruissem B, El-Shamieh S, Zanlonghi X, Meunier I, Roux AF, Mohand-Saïd S, Sahel JA, Audo I. CHM mutation spectrum and disease: An update at the time of human therapeutic trials. Hum Mutat 2021; 42:323-341. [PMID: 33538369 DOI: 10.1002/humu.24174] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 12/21/2020] [Accepted: 01/20/2021] [Indexed: 12/15/2022]
Abstract
Choroideremia is an X-linked inherited retinal disorder (IRD) characterized by the degeneration of retinal pigment epithelium, photoreceptors, choriocapillaris and choroid affecting males with variable phenotypes in female carriers. Unlike other IRD, characterized by a large clinical and genetic heterogeneity, choroideremia shows a specific phenotype with causative mutations in only one gene, CHM. Ongoing gene replacement trials raise further interests in this disorder. We describe here the clinical and genetic data from a French cohort of 45 families, 25 of which carry novel variants, in the context of 822 previously reported choroideremia families. Most of the variants represent loss-of-function mutations with eleven families having large (i.e. ≥6 kb) genomic deletions, 18 small insertions, deletions or insertion deletions, six showing nonsense variants, eight splice site variants and two missense variants likely to affect splicing. Similarly, 822 previously published families carry mostly loss-of-function variants. Recurrent variants are observed worldwide, some of which linked to a common ancestor, others arisen independently in specific CHM regions prone to mutations. Since all exons of CHM may harbor variants, Sanger sequencing combined with quantitative polymerase chain reaction or multiplex ligation-dependent probe amplification experiments are efficient to achieve the molecular diagnosis in patients with typical choroideremia features.
Collapse
Affiliation(s)
- Christina Zeitz
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Marco Nassisi
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | | | - Camille Andrieu
- CHNO des Quinze-Vingts, DHU Sight Restore, INSERM-DHOS CIC1423, Paris, France
| | - Fiona Boyard
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | | | - Vanessa Démontant
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Aline Antonio
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | | | - Helen Frederiksen
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Barbara Kloeckener-Gruissem
- Institute of Medical Molecular Genetics, University of Zurich, Schlieren, Switzerland.,Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Said El-Shamieh
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France.,Department of Medical Laboratory Technology, Faculty of Health Sciences, Beirut Arab University, Beirut, Lebanon
| | - Xavier Zanlonghi
- Clinique Pluridisciplinaire Jules Verne, Institut Ophtalmologique de l'Ouest, Nantes, France
| | - Isabelle Meunier
- National Reference Centre for Inherited Sensory Diseases, University of Montpellier, Montpellier University Hospital, Montpellier, France.,Institute for Neurosciences of Montpellier (INM), University of Montpellier, INSERM, Montpellier, France
| | - Anne-Françoise Roux
- Laboratoire de Génétique Moléculaire, CHU de Montpellier, Université de Montpellier, Montpellier, France
| | - Saddek Mohand-Saïd
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France.,CHNO des Quinze-Vingts, DHU Sight Restore, INSERM-DHOS CIC1423, Paris, France
| | - José-Alain Sahel
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France.,CHNO des Quinze-Vingts, DHU Sight Restore, INSERM-DHOS CIC1423, Paris, France.,Fondation Ophtalmologique Adolphe de Rothschild, Paris, France.,Académie des Sciences-Institut de France, Paris, France.,Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Isabelle Audo
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France.,CHNO des Quinze-Vingts, DHU Sight Restore, INSERM-DHOS CIC1423, Paris, France.,Department of Genetics, UCL-Institute of Ophthalmology, London, UK
| |
Collapse
|
44
|
Abstract
PURPOSE Choroideremia (CHM) is a rare inherited retinal degeneration resulting from mutation of the CHM gene, which results in absence of functional Rab escort protein 1 (REP1). We evaluated retinal gene therapy with an adeno-associated virus vector that used to deliver a functional version of the CHM gene (AAV2-REP1). METHODS THOR (NCT02671539) is a Phase 2, open-label, single-center, randomized study. Six male patients (51-60 years) with CHM received AAV2-REP1, by a single 0.1-mL subretinal injection of 10 genome particles during vitrectomy. Twelve-month data are reported. RESULTS In study eyes, 4 patients experienced minor changes in best-corrected visual acuity (-4 to +1 Early Treatment Diabetic Retinopathy Study [ETDRS] letters); one gained 17 letters and another lost 14 letters. Control eyes had changes of -2 to +4 letters. In 5/6 patients, improvements in mean (95% confidence intervals) retinal sensitivity (2.3 [4.0] dB), peak retinal sensitivity (2.8 [3.5] dB), and gaze fixation area (-36.1 [66.9] deg) were recorded. Changes in anatomical endpoints were similar between study and control eyes. Adverse events were consistent with the surgical procedure. CONCLUSION Gene therapy with AAV2-REP1 can maintain, and in some cases, improve, visual acuity in CHM. Longer term follow-up is required to establish whether these benefits are maintained.
Collapse
|
45
|
Maguire AM, Bennett J, Aleman EM, Leroy BP, Aleman TS. Clinical Perspective: Treating RPE65-Associated Retinal Dystrophy. Mol Ther 2021; 29:442-463. [PMID: 33278565 PMCID: PMC7854308 DOI: 10.1016/j.ymthe.2020.11.029] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/16/2020] [Accepted: 11/25/2020] [Indexed: 12/25/2022] Open
Abstract
Until recently, there was no approved treatment for a retinal degenerative disease. Subretinal injection of a recombinant adeno-associated virus (AAV) delivering the normal copy of the human RPE65 cDNA led to reversal of blindness first in animal models and then in humans. This led to the first US Food and Drug Administration (FDA)-approved gene therapy product for a genetic disease, voretigene neparvovec-rzyl (Luxturna). Luxturna was then approved by the European Medicines Association and is now available in the US through Spark Therapeutics and worldwide through Novartis. Not only has treatment with Luxturna changed the lives of people previously destined to live a life of blindness, but it has fueled interest in developing additional gene therapy reagents targeting numerous other genetic forms of inherited retinal disease. This review describes many of the considerations for administration of Luxturna and describes how lessons from experience with Luxturna could lead to additional gene-based treatments of blindness.
Collapse
Affiliation(s)
- Albert M Maguire
- Center for Advanced Retinal and Ocular Therapeutics (CAROT), Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; The Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
| | - Jean Bennett
- Center for Advanced Retinal and Ocular Therapeutics (CAROT), Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; The Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
| | - Elena M Aleman
- Center for Advanced Retinal and Ocular Therapeutics (CAROT), Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Bart P Leroy
- Center for Advanced Retinal and Ocular Therapeutics (CAROT), Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Ophthalmology and Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
| | - Tomas S Aleman
- Center for Advanced Retinal and Ocular Therapeutics (CAROT), Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; The Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA.
| |
Collapse
|
46
|
Abstract
PURPOSE Choroideremia is an incurable, X-linked, recessive retinal dystrophy caused by loss of function mutations in the CHM gene. It is estimated to affect approximately 1 in 50,000 male patients. It is characterized by progressive degeneration of the retinal pigment epithelium, choroid, and photoreceptors, resulting in visual impairment and blindness. There is an unmet need in choroideremia, because currently, there are no approved treatments available for patients with the disease. METHODS We review the patient journey, societal impact, and emerging treatments for patients with choroideremia. RESULTS Its relative rarity and similarities with other retinal diseases in early years mean that diagnosis of choroideremia can often be delayed. Furthermore, its impact on affected individuals, and wider society, is also likely underestimated. AAV2-mediated gene therapy is an investigational treatment that aims to replace the faulty CHM gene. Early-phase studies reported potentially important visual acuity gains and maintenance of vision in some patients, and a large Phase 3 program is now underway. CONCLUSION Choroideremia is a disease with a significant unmet need. Interventions that can treat progression of the disease and improve visual and functional outcomes have the potential to reduce health care costs and enhance patient quality of life.
Collapse
|
47
|
Meschede IP, Burgoyne T, Tolmachova T, Seabra MC, Futter CE. Chronically shortened rod outer segments accompany photoreceptor cell death in Choroideremia. PLoS One 2020; 15:e0242284. [PMID: 33201897 PMCID: PMC7671558 DOI: 10.1371/journal.pone.0242284] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 10/29/2020] [Indexed: 01/28/2023] Open
Abstract
X-linked choroideremia (CHM) is a disease characterized by gradual retinal degeneration caused by loss of the Rab Escort Protein, REP1. Despite partial compensation by REP2 the disease is characterized by prenylation defects in multiple members of the Rab protein family that are master regulators of membrane traffic. Remarkably, the eye is the only organ affected in CHM patients, possibly because of the huge membrane traffic burden of the post mitotic photoreceptors, which synthesise outer segments, and the adjacent retinal pigment epithelium that degrades the spent portions each day. In this study, we aimed to identify defects in membrane traffic that might lead to photoreceptor cell death in CHM. In a heterozygous null female mouse model of CHM (Chmnull/WT), degeneration of the photoreceptor layer was clearly evident from increased numbers of TUNEL positive cells compared to age matched controls, small numbers of cells exhibiting signs of mitochondrial stress and greatly increased microglial infiltration. However, most rod photoreceptors exhibited remarkably normal morphology with well-formed outer segments and no discernible accumulation of transport vesicles in the inner segment. The major evidence of membrane trafficking defects was a shortening of rod outer segments that was evident at 2 months of age but remained constant over the period during which the cells die. A decrease in rhodopsin density found in the outer segment may underlie the outer segment shortening but does not lead to rhodopsin accumulation in the inner segment. Our data argue against defects in rhodopsin transport or outer segment renewal as triggers of cell death in CHM.
Collapse
Affiliation(s)
| | | | | | - Miguel C. Seabra
- UCL Institute of Ophthalmology, London, United Kingdom
- Imperial College London, London, United Kingdom
- CEDOC, NOVA Universidade Nova de Lisboa, Lisbon, Portugal
| | | |
Collapse
|
48
|
Hayashi T, Kameya S, Mizobuchi K, Kubota D, Kikuchi S, Yoshitake K, Mizota A, Murakami A, Iwata T, Nakano T. Genetic defects of CHM and visual acuity outcome in 24 choroideremia patients from 16 Japanese families. Sci Rep 2020; 10:15883. [PMID: 32985515 PMCID: PMC7522719 DOI: 10.1038/s41598-020-72623-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 09/02/2020] [Indexed: 11/15/2022] Open
Abstract
Choroideremia (CHM) is an incurable progressive chorioretinal dystrophy. Little is known about the natural disease course of visual acuity in the Japanese population. We aimed to investigate the genetic spectrum of the CHM gene and visual acuity outcomes in 24 CHM patients from 16 Japanese families. We measured decimal best-corrected visual acuity (BCVA) at presentation and follow-up, converted to logMAR units for statistical analysis. Sanger and/or whole-exome sequencing were performed to identify pathogenic CHM variants/deletions. The median age at presentation was 37.0 years (range, 5–76 years). The mean follow-up interval was 8.2 years. BCVA of the better-seeing eye at presentation was significantly worsened with increasing age (r = 0.515, p < 0.01), with a high rate of BCVA decline in patients > 40 years old. A Kaplan–Meier survival curve suggested that a BCVA of Snellen equivalent 20/40 at follow-up remains until the fifties. Fourteen pathogenic variants, 6 of which were novel [c.49 + 5G > A, c.116 + 5G > A, p.(Gly176Glu, Glu177Ter), p.Tyr531Ter, an exon 2 deletion, and a 5.0-Mb deletion], were identified in 15 families. No variant was found in one family only. Our BCVA outcome data are useful for predicting visual prognosis and determining the timing of intervention in Japanese patients with CHM variants.
Collapse
Affiliation(s)
- Takaaki Hayashi
- Department of Ophthalmology, Katsushika Medical Center, The Jikei University School of Medicine, 6-41-2 Aoto, Katsushika-ku, Tokyo, 125-8506, Japan. .,Department of Ophthalmology, The Jikei University School of Medicine, Tokyo, Japan.
| | - Shuhei Kameya
- Department of Ophthalmology, Nippon Medical School, Chiba Hokusoh Hospital, Chiba, Japan
| | - Kei Mizobuchi
- Department of Ophthalmology, The Jikei University School of Medicine, Tokyo, Japan
| | - Daiki Kubota
- Department of Ophthalmology, Nippon Medical School, Chiba Hokusoh Hospital, Chiba, Japan
| | - Sachiko Kikuchi
- Department of Ophthalmology, Nippon Medical School, Chiba Hokusoh Hospital, Chiba, Japan
| | - Kazutoshi Yoshitake
- Division of Molecular and Cellular Biology, National Institute of Sensory Organs, National Hospital Organization, Tokyo Medical Center, Tokyo, Japan
| | - Atsushi Mizota
- Department of Ophthalmology, Teikyo University School of Medicine, Tokyo, Japan
| | - Akira Murakami
- Department of Ophthalmology, Juntendo University, Faculty of Medicine, Tokyo, Japan
| | - Takeshi Iwata
- Division of Molecular and Cellular Biology, National Institute of Sensory Organs, National Hospital Organization, Tokyo Medical Center, Tokyo, Japan
| | - Tadashi Nakano
- Department of Ophthalmology, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
49
|
De Silva SR, Arno G, Robson AG, Fakin A, Pontikos N, Mohamed MD, Bird AC, Moore AT, Michaelides M, Webster AR, Mahroo OA. The X-linked retinopathies: Physiological insights, pathogenic mechanisms, phenotypic features and novel therapies. Prog Retin Eye Res 2020; 82:100898. [PMID: 32860923 DOI: 10.1016/j.preteyeres.2020.100898] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 08/07/2020] [Accepted: 08/21/2020] [Indexed: 02/08/2023]
Abstract
X-linked retinopathies represent a significant proportion of monogenic retinal disease. They include progressive and stationary conditions, with and without syndromic features. Many are X-linked recessive, but several exhibit a phenotype in female carriers, which can help establish diagnosis and yield insights into disease mechanisms. The presence of affected carriers can misleadingly suggest autosomal dominant inheritance. Some disorders (such as RPGR-associated retinopathy) show diverse phenotypes from variants in the same gene and also highlight limitations of current genetic sequencing methods. X-linked disease frequently arises from loss of function, implying potential for benefit from gene replacement strategies. We review X-inactivation and X-linked inheritance, and explore burden of disease attributable to X-linked genes in our clinically and genetically characterised retinal disease cohort, finding correlation between gene transcript length and numbers of families. We list relevant genes and discuss key clinical features, disease mechanisms, carrier phenotypes and novel experimental therapies. We consider in detail the following: RPGR (associated with retinitis pigmentosa, cone and cone-rod dystrophy), RP2 (retinitis pigmentosa), CHM (choroideremia), RS1 (X-linked retinoschisis), NYX (complete congenital stationary night blindness (CSNB)), CACNA1F (incomplete CSNB), OPN1LW/OPN1MW (blue cone monochromacy, Bornholm eye disease, cone dystrophy), GPR143 (ocular albinism), COL4A5 (Alport syndrome), and NDP (Norrie disease and X-linked familial exudative vitreoretinopathy (FEVR)). We use a recently published transcriptome analysis to explore expression by cell-type and discuss insights from electrophysiology. In the final section, we present an algorithm for genes to consider in diagnosing males with non-syndromic X-linked retinopathy, summarise current experimental therapeutic approaches, and consider questions for future research.
Collapse
Affiliation(s)
- Samantha R De Silva
- UCL Institute of Ophthalmology, University College London, UK; Moorfields Eye Hospital NHS Foundation Trust, London, UK
| | - Gavin Arno
- UCL Institute of Ophthalmology, University College London, UK; Moorfields Eye Hospital NHS Foundation Trust, London, UK
| | - Anthony G Robson
- UCL Institute of Ophthalmology, University College London, UK; Moorfields Eye Hospital NHS Foundation Trust, London, UK
| | - Ana Fakin
- UCL Institute of Ophthalmology, University College London, UK; Moorfields Eye Hospital NHS Foundation Trust, London, UK; Ljubljana University Medical Centre, Ljubljana, Slovenia
| | - Nikolas Pontikos
- UCL Institute of Ophthalmology, University College London, UK; Moorfields Eye Hospital NHS Foundation Trust, London, UK
| | - Moin D Mohamed
- Department of Ophthalmology, Guy's & St Thomas' NHS Foundation Trust, London, UK
| | - Alan C Bird
- UCL Institute of Ophthalmology, University College London, UK; Moorfields Eye Hospital NHS Foundation Trust, London, UK
| | - Anthony T Moore
- UCL Institute of Ophthalmology, University College London, UK; Moorfields Eye Hospital NHS Foundation Trust, London, UK; Department of Ophthalmology, UCSF School of Medicine, San Francisco, CA, USA
| | - Michel Michaelides
- UCL Institute of Ophthalmology, University College London, UK; Moorfields Eye Hospital NHS Foundation Trust, London, UK
| | - Andrew R Webster
- UCL Institute of Ophthalmology, University College London, UK; Moorfields Eye Hospital NHS Foundation Trust, London, UK
| | - Omar A Mahroo
- UCL Institute of Ophthalmology, University College London, UK; Moorfields Eye Hospital NHS Foundation Trust, London, UK; Department of Ophthalmology, Guy's & St Thomas' NHS Foundation Trust, London, UK; Section of Ophthalmology, King's College London, UK; Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| |
Collapse
|
50
|
Morgan JIW, Chen M, Huang AM, Jiang YY, Cooper RF. Cone Identification in Choroideremia: Repeatability, Reliability, and Automation Through Use of a Convolutional Neural Network. Transl Vis Sci Technol 2020; 9:40. [PMID: 32855844 PMCID: PMC7424931 DOI: 10.1167/tvst.9.2.40] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 04/10/2020] [Indexed: 11/24/2022] Open
Abstract
Purpose Adaptive optics imaging has enabled the visualization of photoreceptors both in health and disease. However, there remains a need for automated accurate cone photoreceptor identification in images of disease. Here, we apply an open-source convolutional neural network (CNN) to automatically identify cones in images of choroideremia (CHM). We further compare the results to the repeatability and reliability of manual cone identifications in CHM. Methods We used split-detection adaptive optics scanning laser ophthalmoscopy to image the inner segment cone mosaic of 17 patients with CHM. Cones were manually identified twice by one experienced grader and once by two additional experienced graders in 204 regions of interest (ROIs). An open-source CNN either pre-trained on normal images or trained on CHM images automatically identified cones in the ROIs. True and false positive rates and Dice's coefficient were used to determine the agreement in cone locations between data sets. Interclass correlation coefficient was used to assess agreement in bound cone density. Results Intra- and intergrader agreement for cone density is high in CHM. CNN performance increased when it was trained on CHM images in comparison to normal, but had lower agreement than manual grading. Conclusions Manual cone identifications and cone density measurements are repeatable and reliable for images of CHM. CNNs show promise for automated cone selections, although additional improvements are needed to equal the accuracy of manual measurements. Translational Relevance These results are important for designing and interpreting longitudinal studies of cone mosaic metrics in disease progression or treatment intervention in CHM.
Collapse
Affiliation(s)
- Jessica I W Morgan
- Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA, USA.,Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| | - Min Chen
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew M Huang
- Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA, USA
| | - Yu You Jiang
- Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA, USA.,Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| | - Robert F Cooper
- Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA, USA.,Department of Psychology, University of Pennsylvania, Philadelphia, PA, USA.,Currently at the Joint Department of Biomedical Engineering, Marquette University and Medical College of Wisconsin and the Department of Ophthalmology, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|