Ophthalmology (Eye & Ear Hospital) - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 10 of 22
  • Item
    No Preview Available
    Exploring the contribution of ARMS2 and HTRA1 genetic risk factors in age-related macular degeneration
    Pan, Y ; Fu, Y ; Baird, PN ; Guymer, RH ; Das, T ; Iwata, T (PERGAMON-ELSEVIER SCIENCE LTD, 2023-11)
    Age-related macular degeneration (AMD) is the leading cause of severe irreversible central vision loss in individuals over 65 years old. Genome-wide association studies (GWASs) have shown that the region at chromosome 10q26, where the age-related maculopathy susceptibility (ARMS2/LOC387715) and HtrA serine peptidase 1 (HTRA1) genes are located, represents one of the strongest associated loci for AMD. However, the underlying biological mechanism of this genetic association has remained elusive. In this article, we extensively review the literature by us and others regarding the ARMS2/HTRA1 risk alleles and their functional significance. We also review the literature regarding the presumed function of the ARMS2 protein and the molecular processes of the HTRA1 protein in AMD pathogenesis in vitro and in vivo, including those of transgenic mice overexpressing HtrA1/HTRA1 which developed Bruch's membrane (BM) damage, choroidal neovascularization (CNV), and polypoidal choroidal vasculopathy (PCV), similar to human AMD patients. The elucidation of the molecular mechanisms of the ARMS2 and HTRA1 susceptibility loci has begun to untangle the complex biological pathways underlying AMD pathophysiology, pointing to new testable paradigms for treatment.
  • Item
    Thumbnail Image
    Deep Learning Applied to Automated Segmentation of Geographic Atrophy in Fundus Autofluorescence Images.
    Arslan, J ; Samarasinghe, G ; Sowmya, A ; Benke, KK ; Hodgson, LAB ; Guymer, RH ; Baird, PN (Association for Research in Vision and Ophthalmology (ARVO), 2021-07-01)
    PURPOSE: This study describes the development of a deep learning algorithm based on the U-Net architecture for automated segmentation of geographic atrophy (GA) lesions in fundus autofluorescence (FAF) images. METHODS: Image preprocessing and normalization by modified adaptive histogram equalization were used for image standardization to improve effectiveness of deep learning. A U-Net-based deep learning algorithm was developed and trained and tested by fivefold cross-validation using FAF images from clinical datasets. The following metrics were used for evaluating the performance for lesion segmentation in GA: dice similarity coefficient (DSC), DSC loss, sensitivity, specificity, mean absolute error (MAE), accuracy, recall, and precision. RESULTS: In total, 702 FAF images from 51 patients were analyzed. After fivefold cross-validation for lesion segmentation, the average training and validation scores were found for the most important metric, DSC (0.9874 and 0.9779), for accuracy (0.9912 and 0.9815), for sensitivity (0.9955 and 0.9928), and for specificity (0.8686 and 0.7261). Scores for testing were all similar to the validation scores. The algorithm segmented GA lesions six times more quickly than human performance. CONCLUSIONS: The deep learning algorithm can be implemented using clinical data with a very high level of performance for lesion segmentation. Automation of diagnostics for GA assessment has the potential to provide savings with respect to patient visit duration, operational cost and measurement reliability in routine GA assessments. TRANSLATIONAL RELEVANCE: A deep learning algorithm based on the U-Net architecture and image preprocessing appears to be suitable for automated segmentation of GA lesions on clinical data, producing fast and accurate results.
  • Item
    Thumbnail Image
    Model Structure Uncertainty in the Characterization and Growth of Geographic Atrophy
    Arslan, J ; Benke, KK ; Samarasinghe, G ; Sowmya, A ; Guymer, RH ; Baird, PN (ASSOC RESEARCH VISION OPHTHALMOLOGY INC, 2021-05)
    PURPOSE: To identify the most suitable model for assessing the rate of growth of total geographic atrophy (GA) by analysis of model structure uncertainty. METHODS: Model structure uncertainty refers to unexplained variability arising from the choice of mathematical model and represents an example of epistemic uncertainty. In this study, we quantified this uncertainty to help identify a model most representative of GA progression. Fundus autofluorescence (FAF) images and GA progression data (i.e., total GA area estimation at each presentation) were acquired using Spectralis HRA+OCT instrumentation and RegionFinder software. Six regression models were evaluated. Models were compared using various statistical tests, [i.e., coefficient of determination (r2), uncertainty metric (U), and test of significance for the correlation coefficient, r], as well as adherence to expected physical and clinical assumptions of GA growth. RESULTS: Analysis was carried out for 81 GA-affected eyes, 531 FAF images (range: 3-17 images per eye), over median of 57 months (IQR: 42, 74), with a mean baseline lesion size of 2.62 ± 4.49 mm2 (range: 0.11-20.69 mm2). The linear model proved to be the most representative of total GA growth, with lowest average uncertainty (original scale: U = 0.025, square root scale: U = 0.014), high average r2 (original scale: 0.92, square root scale: 0.93), and applicability of the model was supported by a high correlation coefficient, r, with statistical significance (P = 0.01). CONCLUSIONS: Statistical analysis of uncertainty suggests that the linear model provides an effective and practical representation of the rate and progression of total GA growth based on data from patient presentations in clinical settings. TRANSLATIONAL RELEVANCE: Identification of correct model structure to characterize rate of growth of total GA in the retina using FAF images provides an objective metric for comparing interventions and charting GA progression in clinical presentations.
  • Item
    Thumbnail Image
    Binding of Gtf2i-β/δ transcription factors to the ARMS2 gene leads to increased circulating HTRA1 in AMD patients and in vitro
    Pan, Y ; Iejima, D ; Nakayama, M ; Suga, A ; Noda, T ; Kaur, I ; Das, T ; Chakrabarti, S ; Guymer, RH ; DeAngelis, MM ; Yamamoto, M ; Baird, PN ; Iwata, T (ELSEVIER, 2021)
    The disease-initiating molecular events for age-related macular degeneration (AMD), a multifactorial retinal disease affecting many millions of elderly individuals worldwide, are still unknown. Of the over 30 risk and protective loci so far associated with AMD through whole genome-wide association studies (GWAS), the Age-Related Maculopathy Susceptibility 2 (ARMS2) gene locus represents one of the most highly associated risk regions for AMD. A unique insertion/deletion (in/del) sequence located immediately upstream of the High Temperature Requirement A1 (HTRA1) gene in this region confers high risk for AMD. Using electrophoretic mobility shift assay (EMSA), we identified that two Gtf2i-β/δ transcription factor isoforms bind to the cis-element 5'- ATTAATAACC-3' contained in this in/del sequence. The binding of these transcription factors leads to enhanced upregulation of transcription of the secretory serine protease HTRA1 in transfected cells and AMD patient-derived induced pluripotent stem cells (iPSCs). Overexpression of Htra1 in mice using a CAG-promoter demonstrated increased blood concentration of Htra1 protein, caused upregulation of vascular endothelial growth factor (VEGF), and produced a choroidal neovascularization (CNV)-like phenotype. Finally, a comparison of 478 AMD patients to 481 healthy, age-matched controls from Japan, India, Australia, and the USA showed a statistically increased level of secreted HTRA1 blood concentration in AMD patients compared with age-matched controls. Taken together, these results suggest a common mechanism across ethnicities whereby increased systemic blood circulation of secreted serine protease HTRA1 leads to subsequent degradation of Bruch's membrane and eventual CNV in AMD.
  • Item
    No Preview Available
    A large genome-wide association study of age-related macular degeneration highlights contributions of rare and common variants
    Fritsche, LG ; Igl, W ; Bailey, JNC ; Grassmann, F ; Sengupta, S ; Bragg-Gresham, JL ; Burdon, KP ; Hebbring, SJ ; Wen, C ; Gorski, M ; Kim, IK ; Cho, D ; Zack, D ; Souied, E ; Scholl, HPN ; Bala, E ; Lee, KE ; Hunter, DJ ; Sardell, RJ ; Mitchell, P ; Merriam, JE ; Cipriani, V ; Hoffman, JD ; Schick, T ; Lechanteur, YTE ; Guymer, RH ; Johnson, MP ; Jiang, Y ; Stanton, CM ; Buitendijk, GHS ; Zhan, X ; Kwong, AM ; Boleda, A ; Brooks, M ; Gieser, L ; Ratnapriya, R ; Branham, KE ; Foerster, JR ; Heckenlively, JR ; Othman, MI ; Vote, BJ ; Liang, HH ; Souzeau, E ; McAllister, IL ; Isaacs, T ; Hall, J ; Lake, S ; Mackey, DA ; Constable, IJ ; Craig, JE ; Kitchner, TE ; Yang, Z ; Su, Z ; Luo, H ; Chen, D ; Hong, O ; Flagg, K ; Lin, D ; Mao, G ; Ferreyra, H ; Starke, K ; von Strachwitz, CN ; Wolf, A ; Brandl, C ; Rudolph, G ; Olden, M ; Morrison, MA ; Morgan, DJ ; Schu, M ; Ahn, J ; Silvestri, G ; Tsironi, EE ; Park, KH ; Farrer, LA ; Orlin, A ; Brucker, A ; Li, M ; Curcio, CA ; Mohand-Said, S ; Sahel, J-M ; Audo, I ; Benchaboune, M ; Cree, AJ ; Rennie, CA ; Goverdhan, SV ; Grunin, M ; Hagbi-Levi, S ; Campochiaro, P ; Katsanis, N ; Holz, FG ; Blond, F ; Blanche, H ; Deleuze, J-F ; Igo, RP ; Truitt, B ; Peachey, NS ; Meuer, SM ; Myers, CE ; Moore, EL ; Klein, R ; Hauser, MA ; Postel, EA ; Courtenay, MD ; Schwartz, SG ; Kovach, JL ; Scott, WK ; Liew, G ; Tan, AG ; Gopinath, B ; Merriam, JC ; Smith, RT ; Khan, JC ; Shahid, H ; Moore, AT ; McGrath, JA ; Laux, R ; Brantley, MA ; Agarwal, A ; Ersoy, L ; Caramoy, A ; Langmann, T ; Saksens, NTM ; de Jong, EK ; Hoyng, CB ; Cain, MS ; Richardson, AJ ; Martin, TM ; Blangero, J ; Weeks, DE ; Dhillon, B ; van Duijn, CM ; Doheny, KF ; Romm, J ; Klaver, CCW ; Hayward, C ; Gorin, MB ; Klein, ML ; Baird, PN ; den Hollander, AI ; Fauser, S ; Yates, JRW ; Allikmets, R ; Wang, JJ ; Schaumberg, DA ; Klein, BEK ; Hagstrom, SA ; Chowers, I ; Lotery, AJ ; Leveillard, T ; Zhang, K ; Brilliant, MH ; Hewitt, AW ; Swaroop, A ; Chew, EY ; Pericak-Vance, MA ; DeAngelis, M ; Stambolian, D ; Haines, JL ; Iyengar, SK ; Weber, BHF ; Abecasis, GR ; Heid, IM (NATURE PUBLISHING GROUP, 2016-02)
    Advanced age-related macular degeneration (AMD) is the leading cause of blindness in the elderly, with limited therapeutic options. Here we report on a study of >12 million variants, including 163,714 directly genotyped, mostly rare, protein-altering variants. Analyzing 16,144 patients and 17,832 controls, we identify 52 independently associated common and rare variants (P < 5 × 10(-8)) distributed across 34 loci. Although wet and dry AMD subtypes exhibit predominantly shared genetics, we identify the first genetic association signal specific to wet AMD, near MMP9 (difference P value = 4.1 × 10(-10)). Very rare coding variants (frequency <0.1%) in CFH, CFI and TIMP3 suggest causal roles for these genes, as does a splice variant in SLC16A8. Our results support the hypothesis that rare coding variants can pinpoint causal genes within known genetic loci and illustrate that applying the approach systematically to detect new loci requires extremely large sample sizes.
  • Item
    No Preview Available
    Identification of a rare coding variant in complement 3 associated with age-related macular degeneration
    Zhan, X ; Larson, DE ; Wang, C ; Koboldt, DC ; Sergeev, YV ; Fulton, RS ; Fulton, LL ; Fronick, CC ; Branham, KE ; Bragg-Gresham, J ; Jun, G ; Hu, Y ; Kang, HM ; Liu, D ; Othman, M ; Brooks, M ; Ratnapriya, R ; Boleda, A ; Grassmann, F ; von Strachwitz, C ; Olson, LM ; Buitendijk, GHS ; Hofman, A ; van Duijn, CM ; Cipriani, V ; Moore, AT ; Shahid, H ; Jiang, Y ; Conley, YP ; Morgan, DJ ; Kim, IK ; Johnson, MP ; Cantsilieris, S ; Richardson, AJ ; Guymer, RH ; Luo, H ; Ouyang, H ; Licht, C ; Pluthero, FG ; Zhang, MM ; Zhang, K ; Baird, PN ; Blangero, J ; Klein, ML ; Farrer, LA ; DeAngelis, MM ; Weeks, DE ; Gorin, MB ; Yates, JRW ; Klaver, CCW ; Pericak-Vance, MA ; Haines, JL ; Weber, BHF ; Wilson, RK ; Heckenlively, JR ; Chew, EY ; Stambolian, D ; Mardis, ER ; Swaroop, A ; Abecasis, GR (NATURE PORTFOLIO, 2013-11)
    Macular degeneration is a common cause of blindness in the elderly. To identify rare coding variants associated with a large increase in risk of age-related macular degeneration (AMD), we sequenced 2,335 cases and 789 controls in 10 candidate loci (57 genes). To increase power, we augmented our control set with ancestry-matched exome-sequenced controls. An analysis of coding variation in 2,268 AMD cases and 2,268 ancestry-matched controls identified 2 large-effect rare variants: previously described p.Arg1210Cys encoded in the CFH gene (case frequency (fcase) = 0.51%; control frequency (fcontrol) = 0.02%; odds ratio (OR) = 23.11) and newly identified p.Lys155Gln encoded in the C3 gene (fcase = 1.06%; fcontrol = 0.39%; OR = 2.68). The variants suggest decreased inhibition of C3 by complement factor H, resulting in increased activation of the alternative complement pathway, as a key component of disease biology.
  • Item
    No Preview Available
    Seven new loci associated with age-related macular degeneration
    Fritsche, LG ; Chen, W ; Schu, M ; Yaspan, BL ; Yu, Y ; Thorleifsson, G ; Zack, DJ ; Arakawa, S ; Cipriani, V ; Ripke, S ; Igo, RP ; Buitendijk, GHS ; Sim, X ; Weeks, DE ; Guymer, RH ; Merriam, JE ; Francis, PJ ; Hannum, G ; Agarwal, A ; Armbrecht, AM ; Audo, I ; Aung, T ; Barile, GR ; Benchaboune, M ; Bird, AC ; Bishop, PN ; Branham, KE ; Brooks, M ; Brucker, AJ ; Cade, WH ; Cain, MS ; Campochiaroll, PA ; Chan, C-C ; Cheng, C-Y ; Chew, EY ; Chin, KA ; Chowers, I ; Clayton, DG ; Cojocaru, R ; Conley, YP ; Cornes, BK ; Daly, MJ ; Dhillon, B ; Edwards, A ; Evangelou, E ; Fagemess, J ; Ferreyra, HA ; Friedman, JS ; Geirsdottir, A ; George, RJ ; Gieger, C ; Gupta, N ; Hagstrom, SA ; Harding, SP ; Haritoglou, C ; Heckenlively, JR ; Hoz, FG ; Hughes, G ; Ioannidis, JPA ; Ishibashi, T ; Joseph, P ; Jun, G ; Kamatani, Y ; Katsanis, N ; Keilhauer, CN ; Khan, JC ; Kim, IK ; Kiyohara, Y ; Klein, BEK ; Klein, R ; Kovach, JL ; Kozak, I ; Lee, CJ ; Lee, KE ; Lichtner, P ; Lotery, AJ ; Meitinger, T ; Mitchell, P ; Mohand-Saied, S ; Moore, AT ; Morgan, DJ ; Morrison, MA ; Myers, CE ; Naj, AC ; Nakamura, Y ; Okada, Y ; Orlin, A ; Ortube, MC ; Othman, MI ; Pappas, C ; Park, KH ; Pauer, GJT ; Peachey, NS ; Poch, O ; Priya, RR ; Reynolds, R ; Richardson, AJ ; Ripp, R ; Rudolph, G ; Ryu, E ; Sahel, J-A ; Schaumberg, DA ; Scholl, HPN ; Schwartz, SG ; Scott, WK ; Shahid, H ; Sigurdsson, H ; Silvestri, G ; Sivakumaran, TA ; Smith, RT ; Sobrin, L ; Souied, EH ; Stambolian, DE ; Stefansson, H ; Sturgill-Short, GM ; Takahashi, A ; Tosakulwong, N ; Truitt, BJ ; Tsironi, EE ; Uitterlinden, AG ; van Duijn, CM ; Vijaya, L ; Vingerling, JR ; Vithana, EN ; Webster, AR ; Wichmann, H-E ; Winkler, TW ; Wong, TY ; Wright, AF ; Zelenika, D ; Zhang, M ; Zhao, L ; Zhang, K ; Klein, ML ; Hageman, GS ; Lathrop, GM ; Stefansson, K ; Allikmets, R ; Baird, PN ; Gorin, MB ; Wang, JJ ; Klaver, CCW ; Seddon, JM ; Pericak-Vance, MA ; Iyengar, SK ; Yates, JRW ; Swaroop, A ; Weber, BHF ; Kubo, M ; DeAngelis, MM ; Leveillard, T ; Thorsteinsdottir, U ; Haines, JL ; Farrer, LA ; Heid, IM ; Abecasis, GR (NATURE PORTFOLIO, 2013-04)
    Age-related macular degeneration (AMD) is a common cause of blindness in older individuals. To accelerate the understanding of AMD biology and help design new therapies, we executed a collaborative genome-wide association study, including >17,100 advanced AMD cases and >60,000 controls of European and Asian ancestry. We identified 19 loci associated at P < 5 × 10(-8). These loci show enrichment for genes involved in the regulation of complement activity, lipid metabolism, extracellular matrix remodeling and angiogenesis. Our results include seven loci with associations reaching P < 5 × 10(-8) for the first time, near the genes COL8A1-FILIP1L, IER3-DDR1, SLC16A8, TGFBR1, RAD51B, ADAMTS9 and B3GALTL. A genetic risk score combining SNP genotypes from all loci showed similar ability to distinguish cases and controls in all samples examined. Our findings provide new directions for biological, genetic and therapeutic studies of AMD.
  • Item
    No Preview Available
    Clinical validation of a genetic model to estimate the risk of developing choroidal neovascular age-related macular degeneration.
    Hageman, GS ; Gehrs, K ; Lejnine, S ; Bansal, AT ; Deangelis, MM ; Guymer, RH ; Baird, PN ; Allikmets, R ; Deciu, C ; Oeth, P ; Perlee, LT (Springer Science and Business Media LLC, 2011-07)
    Predictive tests for estimating the risk of developing late-stage neovascular age-related macular degeneration (AMD) are subject to unique challenges. AMD prevalence increases with age, clinical phenotypes are heterogeneous and control collections are prone to high false-negative rates, as many control subjects are likely to develop disease with advancing age. Risk prediction tests have been presented previously, using up to ten genetic markers and a range of self-reported non-genetic variables such as body mass index (BMI) and smoking history. In order to maximise the accuracy of prediction for mainstream genetic testing, we sought to derive a test comparable in performance to earlier testing models but based purely on genetic markers, which are static through life and not subject to misreporting. We report a multicentre assessment of a larger panel of single nucleotide polymorphisms (SNPs) than previously analysed, to improve further the classification performance of a predictive test to estimate the risk of developing choroidal neovascular (CNV) disease. We developed a predictive model based solely on genetic markers and avoided inclusion of self-reported variables (eg smoking history) or non-static factors (BMI, education status) that might otherwise introduce inaccuracies in calculating individual risk estimates. We describe the performance of a test panel comprising 13 SNPs genotyped across a consolidated collection of four patient cohorts obtained from academic centres deemed appropriate for pooling. We report on predictive effect sizes and their classification performance. By incorporating multiple cohorts of homogeneous ethnic origin, we obtained >80 per cent power to detect differences in genetic variants observed between cases and controls. We focused our study on CNV, a subtype of advanced AMD associated with a severe and potentially treatable form of the disease. Lastly, we followed a two-stage strategy involving both test model development and test model validation to present estimates of classification performance anticipated in the larger clinical setting. The model contained nine SNPs tagging variants in the regulators of complement activation (RCA) locus spanning the complement factor H (CFH), complement factor H-related 4 (CFHR4), complement factor H-related 5 (CFHR5) and coagulation factor XIII B subunit (F13B) genes; the four remaining SNPs targeted polymorphisms in the complement component 2 (C2), complement factor B (CFB), complement component 3 (C3) and age-related maculopathy susceptibility protein 2 (ARMS2) genes. The pooled sample size (1,132 CNV cases, 822 controls) allowed for both model development and model validation to confirm the accuracy of risk prediction. At the validation stage, our test model yielded 82 per cent sensitivity and 63 per cent specificity, comparable with metrics reported with earlier testing models that included environmental risk factors. Our test had an area under the curve of 0.80, reflecting a modest improvement compared with tests reported with fewer SNPs.
  • Item
    No Preview Available
    Can HMG Co-A reductase inhibitors ("statins") slow the progression of age-related macular degeneration? The Age-Related Maculopathy Statin Study (ARMSS)
    Guymer, RH ; Dimitrov, PN ; Varsamidis, M ; Lim, LL ; Baird, PN ; Vingrys, AJ ; Robman, L (DOVE MEDICAL PRESS LTD, 2008)
    Age-related macular degeneration (AMD) is responsible for the majority of visual impairment in the Western world. The role of cholesterol-lowering medications, HMG Co-A reductase inhibitors or statins, in reducing the risk of AMD or of delaying its progression has not been fully investigated. A 3-year prospective randomized controlled trial of 40 mg simvastatin per day compared to placebo in subjects at high risk of AMD progression is described. This paper outlines the primary aims of the Age-Related Maculopathy Statin Study (ARMSS), and the methodology involved. Standardized clinical grading of macular photographs and comparison of serial macular digital photographs, using the International grading scheme, form the basis for assessment of primary study outcomes. In addition, macular function is assessed at each visit with detailed psychophysical measurements of rod and cone function. Information collected in this study will assist in the assessment of the potential value of HMG Co-A reductase inhibitors (statins) in reducing the risk of AMD progression.
  • Item
    Thumbnail Image
    Common variants near FRK/COL10A1 and VEGFA are associated with advanced age-related macular degeneration
    Yu, Y ; Bhangale, TR ; Fagerness, J ; Ripke, S ; Thorleifsson, G ; Tan, PL ; Souied, EH ; Richardson, AJ ; Merriam, JE ; Buitendijk, GHS ; Reynolds, R ; Raychaudhuri, S ; Chin, KA ; Sobrin, L ; Evangelou, E ; Lee, PH ; Lee, AY ; Leveziel, N ; Zack, DJ ; Campochiaro, B ; Campochiaro, P ; Smith, RT ; Barile, GR ; Guymer, RH ; Hogg, R ; Chakravarthy, U ; Robman, LD ; Gustafsson, O ; Sigurdsson, H ; Ortmann, W ; Behrens, TW ; Stefansson, K ; Uitterlinden, AG ; van Duijn, CM ; Vingerling, JR ; Klaver, CCW ; Allikmets, R ; Brantley, MA ; Baird, PN ; Katsanis, N ; Thorsteinsdottir, U ; Ioannidis, JPA ; Daly, MJ ; Graham, RR ; Seddon, JM (OXFORD UNIV PRESS, 2011-09-15)
    Despite significant progress in the identification of genetic loci for age-related macular degeneration (AMD), not all of the heritability has been explained. To identify variants which contribute to the remaining genetic susceptibility, we performed the largest meta-analysis of genome-wide association studies to date for advanced AMD. We imputed 6 036 699 single-nucleotide polymorphisms with the 1000 Genomes Project reference genotypes on 2594 cases and 4134 controls with follow-up replication of top signals in 5640 cases and 52 174 controls. We identified two new common susceptibility alleles, rs1999930 on 6q21-q22.3 near FRK/COL10A1 [odds ratio (OR) 0.87; P = 1.1 × 10(-8)] and rs4711751 on 6p12 near VEGFA (OR 1.15; P = 8.7 × 10(-9)). In addition to the two novel loci, 10 previously reported loci in ARMS2/HTRA1 (rs10490924), CFH (rs1061170, and rs1410996), CFB (rs641153), C3 (rs2230199), C2 (rs9332739), CFI (rs10033900), LIPC (rs10468017), TIMP3 (rs9621532) and CETP (rs3764261) were confirmed with genome-wide significant signals in this large study. Loci in the recently reported genes ABCA1 and COL8A1 were also detected with suggestive evidence of association with advanced AMD. The novel variants identified in this study suggest that angiogenesis (VEGFA) and extracellular collagen matrix (FRK/COL10A1) pathways contribute to the development of advanced AMD.