Skip to main content
Erschienen in: Graefe's Archive for Clinical and Experimental Ophthalmology 11/2023

Open Access 09.05.2023 | Mini Review

Artificial intelligence in retinal disease: clinical application, challenges, and future directions

verfasst von: Malena Daich Varela, Sagnik Sen, Thales Antonio Cabral De Guimaraes, Nathaniel Kabiri, Nikolas Pontikos, Konstantinos Balaskas, Michel Michaelides

Erschienen in: Graefe's Archive for Clinical and Experimental Ophthalmology | Ausgabe 11/2023

Abstract

Retinal diseases are a leading cause of blindness in developed countries, accounting for the largest share of visually impaired children, working-age adults (inherited retinal disease), and elderly individuals (age-related macular degeneration). These conditions need specialised clinicians to interpret multimodal retinal imaging, with diagnosis and intervention potentially delayed. With an increasing and ageing population, this is becoming a global health priority. One solution is the development of artificial intelligence (AI) software to facilitate rapid data processing. Herein, we review research offering decision support for the diagnosis, classification, monitoring, and treatment of retinal disease using AI. We have prioritised diabetic retinopathy, age-related macular degeneration, inherited retinal disease, and retinopathy of prematurity. There is cautious optimism that these algorithms will be integrated into routine clinical practice to facilitate access to vision-saving treatments, improve efficiency of healthcare systems, and assist clinicians in processing the ever-increasing volume of multimodal data, thereby also liberating time for doctor-patient interaction and co-development of personalised management plans.
Hinweise

Publisher's note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Retinal diseases are a significant cause of visual impairment and blindness, both in adults (secondary to age-related macular degeneration (AMD) and diabetic retinopathy (DR)) [1] and in children (due to inherited retinal disorders (IRD) and retinopathy of prematurity (ROP)) [2]. Diagnosing these conditions usually involves multimodal testing and multiple consultations with retina specialists, often not available in a timely manner, which can result in delays in sight-saving treatments. For rare diseases, it can take several years for a final diagnosis (‘diagnostic odyssey’), resulting in uncertainty about the prognosis and delay in appropriate care.
Healthcare data increases by approximately 50% every year, making it one of the fastest-growing digital areas [3]. Genomic data alone is as demanding in terms of data acquisition, storage, distribution, and analysis as astronomy or social media content [4]. Ophthalmology is one of the leading data generators, with 30 million optical coherence tomography (OCT) scans performed yearly in the USA [5]. This ever-increasing vast amount of data, alongside the development of cutting-edge digital technology, has made ophthalmology a pioneer in digital innovation and healthcare artificial intelligence (AI).
AI has been rapidly developing in multiple areas of medicine, including, dermatologist-level performance at detecting skin cancer [6], highly accurate classification of pulmonary tuberculosis [7], and genetic variant calling and classification [8]. AI-based ophthalmology telemedicine has been beneficial during the COVID-19 pandemic [9], and remote evaluation and analysis of retinal imaging may be useful in decreasing diagnostic time and facilitating triaging and classification [10, 11].
Development of highly sensitive and sensible AI-based tools requires transdisciplinary collaboration between clinicians and software engineers. Herein, we will provide an overview of current methodologies used in AI system development and validation and focus on clinical application in prioritising retinal diseases.

AI methodology overview

The most common techniques to develop AI-based healthcare tools will be summarised below and in Figs. 1 and 2:
  • AI is a phenomenon in which non-living entities mimic human intelligence [12]. It is an umbrella term encompassing a spectrum of computing programs. ‘Rule-based’, ‘hard-coded’ or ‘symbolic AI’ has existed for many decades and is the basis of any software system, from a traffic light management system to the autopilot flying every plane. In healthcare, symbolic AI has multiple applications, e.g. calculating cardiovascular risk index or eGFR.
  • Machine learning (ML) is an AI subfield in which a program achieves a task by being exposed to vast volumes of data and gradually learning to recognise patterns within the data, allocating data to distinct classes [13]. It involves ‘soft coding’, which means that the model learns from examples instead of being programmed with rules [12]. ML models can be supervised (based on data labelled by humans), unsupervised (i.e., grouping features within categories), or reinforcement learning (the system accumulates its own feedback to improve through a reward function) [14]. In medicine, supervision is the most common.
  • Nonneural network-supervised ML algorithms are useful in healthcare for prediction modelling and evaluating associations and best-fitted lines between two (linear regression, parametric) or multiple variables (random forest, non-parametric). The latter combines different inputs using a network of flowcharts (known as decision trees); each tree creates an outcome, and a collective one will be made by combining all the singular outputs [15]. Non-neural networks are often combined with deep neural network (DNN) architectures and achieve improved performance (Fig. 1) [16].
  • Deep learning (DL) is a subdivision of ML, defined by the presence of multiple layers of artificial neural networks (ANN) [17]. An ANN is composed of an input layer of multiple nodes—‘artificial neurons’—that represent characteristics to be analysed, e.g. pixels on an image, diagnoses (International Classification of Disease (ICD) coded), age, nucleotide changes, etc.; connected to one or more hidden layers that sum and analyse all inputs, and transmit a final value to an output layer (Fig. 2A).
  • DNN corresponds to multi-layered DL algorithms (with often over 100 hidden layers), which are currently the gold standard for image classification [15]. As more layers are added, an iterative training phenomenon starts occurring, by which deep layers combine stimuli sent from other layers and design new stimuli, improving the output layer and ultimately leading to better diagnoses [8].
  • Convolutional neural network (CNN) is a type of DNN particularly useful for image and video analysis [15]. These algorithms divide the files into pixels, convert them into numbers or symbols, analyse them by multiple convolutional layers that filter, merge, mask, and/or multiply features, and feed the results to a dense neural network that will create an output layer [18]. Fully convolutional networks (FCN) feed the output layers themselves, without the final step of dense layers (Fig. 2A) [17].

Useful concepts to better understand AI literature

There are different types of models, depending on the outcome prediction. (i) Classification models apply to categorical outputs, such as classifying retinal images into with or without DR; (ii) segmentation models are specialised for image processing and analysis, detecting presence or absence of features (e.g., intraretinal fluid), segmenting images into known anatomical correlates, or classifying them into diagnostic categories; (iii) regression models for when a quantitative output is needed, such as predict central macular thickness from an OCT file [19, 20].
Different performance metrics are used to present results of each model type. Dice similarity coefficient (dice score) and intraclass correlation coefficient (ICC) are metrics of segmentation accuracy suitable for evaluating performance of image segmentation DL algorithms, ranging from 0 to 1 [21]. There are multiple performance metrics for classification and regression model algorithms, such as (i) receiver operating characteristic (ROC) curves, that plot true positives (sensitivity) against false positives (1 = specificity) [22]; (ii) the area under the curve (AUC, also known as AUROC) ranging from 0 to 1, with 1 indicating a perfect algorithm [23]; (iii) precision-recall curves (PRC), which associate positive predictive value with sensitivity (also known as recall or true positive rate) [24]; (iv) the accuracy statistical score; (v) absolute difference; (vi) Pearson’s correlation between said parameters (the latter also goes from 0 and 1) [23].
The process of developing a supervised AI model generally involves three stages: (1) training, when the network is provided labelled images; (2) fine-tuning, where the model starts aiding the manual annotation and human graders to correct it and improve it; (3) validation or testing, where the algorithm is tested on a hold-out dataset annotated by human graders and kept separate from the training dataset (internal validation). External validation on datasets of completely independent origin than the training dataset is the gold standard for validation/performance evaluation, indicating generalizability (Fig. 2B). [23]

Selected retinal diseases for which AI-based tools have been developed

Diabetic retinopathy (DR)

Recent studies have shown that AI-based DR screening systems can achieve adequate levels of safety [2529]. These algorithms include classical expert-designed image analysis, mathematical morphology, and transformations [3033]. One of the approaches tested was to classify colour fundus images from training datasets into referable DR (moderate or advanced stage) or non-referable DR (no or mild DR, Table 1). These studies either built their own CNNs or used pretrained ones like AlexNet [34], Inception V3 [35], Inception-Resnet-V2 [36], and Resnet152 [37]. Other studies tried to detect DR based on fixed features such as red lesions [3839], microaneurysms [40], exudates, and blood-vessel segmentation [41, 42]. Lastly, other groups introduced a method to detect DR and diabetic macular oedema (DMO) using a CNN model, being able to detect the exact stage of DR; these studies are summarised in Table 1 [4960, 95].
Table 1
Artificial intelligence in retinal disease—methods, cohorts, and overall results
Condition
Imaging analysed
Database (n)
AI tool
Task
Performance (metrics provided by each paper)
Publication
DR
Colour
DiaretDB0 (130), DiaretDB1 (89), and DrimDB (125)
CNN
Referable/non-referable DR
Accuracy 99.17% (DiaretDB0), 98.53% (DiaretDB1), 99.18% (DrimDB)
Sensitivity 100% (DiaretDB0), 99.2% (DiaretDB1), 100% (DrimDB)
Specificity 98.4% (DiaretDB0), 97.97% (DiaretDB1), 98.44% (DrimDB)
Adem et al. [41]
Colour
Kaggle (88,702), DiaretDB1 (89), and E-ophtha (107,799)
CNN
Referable/non-referable DR
AUC 0.954 (Kaggle), 0.949 (E-ophtha)
Quellec et al. [43]
Colour
Kaggle (35,000)
CNN-ResNet34
Referable/non-referable DR
Sensitivity 85%
Specificity 86%
Esfahani et al. [44]
Colour
Messidor-2 (1748), Kaggle (88,702), and DR2 (520)
CNN
Referable/non-referable DR
Accuracy 98.2% (Messidor-2), 98% (DR2)
Pires et al. [45]
Colour
Own dataset (30,244)
CNN (Inception V3, Inception-Resnet-V2, and Resnet152)
Referable/non-referable DR
AUC 0.946
Accuracy 88.21%
Sensitivity 85.57%
Specificity 90.85%
Jiang et al. [46]
Colour
Own dataset (60,000) and STARE (131)
CNN (WP-CNN, ResNet, SeNet, and DenseNet)
Referable/non-referable DR
AUC 0.9823 (Own dataset), 0.951 (STARE)
Accuracy 94.23% (Own dataset), 90.84% (STARE)
Sensitivity 90.94% (Own dataset)
Specificity 90.85% (Own dataset)
Liu YP et al. [47]
Colour
DIARETDB1 (89), DIARETDB0 (130), Kaggle (15,919), Messidor (1200), Messidor-2 (874), IDRiD (103), and DDR (4105)
CNN (VGG16, custom CNN)
Referable/non-referable DR
AUC 0.786 (Kaggle, Messidor), 0.764 (Messidor-2), 0.912 (IDRiD, DDR)
Accuracy 82.1% (Kaggle, Messidor), 91.1% (Messidor-2), 94% (IDRiD, DDR)
Zago et al. [48]
Colour
Messidor-2 (1748)
CNN
Different clinical stages of DR
AUC 0.98
Sensitivity 96.8%
Specificity 87%
Abramoff et al. [49]
Colour
Kaggle (80,000)
CNN
Different clinical stages of DR
Accuracy 75%
Sensitivity 30%
Specificity 95%
Pratt et al. [50]
Colour
Kaggle (2000)
DNN, CNN (VGGNET architecture), BNN
Different clinical stages of DR
Accuracy: BNN = 42%, DNN = 86.3%, CNN = 78.3%
Dutta S et al. [51]
Colour
Kaggle (166)
CNN (InceptionNet V3, AlexNet, and VGG16)
Different clinical stages of DR
Accuracy: AlexNet = 37.43%, VGG16 = 50.03%, InceptionNet V3 = 63.23%
Wang X et al. [52]
Colour
Kaggle (35,126)
CNN (AlexNet, VggNet, GoogleNet, and ResNet)
Different clinical stages of DR
AUC 0.9786 (VggNet)
Accuracy 95.68% (VggNet)
Sensitivity 90.78% (VggNet)
Specificity 97.43% (VggNet)
Wan S et al. [53]
Colour
MESSIDOR (1200)
CNN (AlexNet, VggNet16, custom CNN)
Different clinical stages of DR
Accuracy 98.15%
Sensitivity 98.94%
Specificity 97.87%
Mobben-ur-Rehman et al. [54]
Colour
Own dataset (13,767)
CNN (ResNet50, InceptionV3, InceptionResNetV2, Xception, and DenseNets)
Different clinical stages of DR
96.5%, 98.1%, 98.9%
Zhang et al. [55]
Colour
Kaggle (22,700) and IDRiD (516)
CNN (AlexNet)
Different clinical stages of DR
Accuracy 90.07%
Harangi et al. [56]
Colour
DDR (13,673)
CNN (GoogLeNet, ResNet-18, DenseNet-121, VGG-16, and SE-BN-Inception)
Different clinical stages of DR
Accuracy 82.84%
Li T et al. [57]
Colour
Messidor (1190)
CNN (modified Alexnet)
Different clinical stages of DR
Accuracy 96.35%
Sensitivity 92.35%
Specificity 97.45%
Shanthi T et al. [58]
Colour
Own dataset (9194) and Messidor (1200)
CNN
Different clinical stages of DR
Accuracy 92.95% (own dataset)
Sensitivity 99.39% (own dataset), 99.93% (Messidor)
Specificity 92.59% (own dataset), 96.2% (Messidor)
Wang J et al. [59]
Colour
Messidor (1200) and IDRiD (516)
CNN (ResNet50)
Different clinical stages of DR
AUC 0.963 (Messidor)
Accuracy 92.6% (Messidor), 65.1% (IDRiD)
Sensitivity 92% (Messidor)
Li X et al. [60]
AMD
Colour
407 eyes with nonadvanced AMD
DL
Distinguishes between low and high-risk AMD by quantifying drusen location, area, and size
For drusen area: ICC > 0.85; for diameter: ICC = 0.69; for AMD risk assessment: ROC = 0.948 and 0.954
Van Grinsven et al. [61]
Colour, OCT, and IR
278 eyes with/without reticular pseudodrusen (RPD)
DL
Automatic quantification of RPD
ROC = 0.94 and 0.958; κ agreement = 0.911; ICC = 0.704
Van Grinsven et al. [62]
Colour
2951 subjects from AREDS (834 progressors)
DL
Association between genetic variants and transition to advanced AMD
AUC: 5 years = 0.885; 10 years = 0.915
Seddon et al. [63]
Colour and OCT
280 eyes from 140 participants
DL
Prediction of progression to late AMD
AUC = 0.85
Wu et al. [64]
Colour and microperimetry
280 eyes from 140 participants
DL
Predictive value of pointwise sensitivity and low luminance deficits for AMD progression
AUC = 0.8
Wu et al. [65]
Colour
 > 4600 participants from AREDS
DL
Predict progression to advanced dry or wet AMD
Accuracy = 0.86 (1 year) and 0.86 (2 years); specificity = 0.85 (1 year) and 0.84 (2 years); sensitivity = 0.91 (1 year) and 0.92 (2 years)
Bhuiyan et al. [66]
Colour
1351 subjects from AREDS (> 31,000 images)
DL
Predict progression to advanced dry or wet AMD
AUC = 0.85
Yan et al. [67]
Colour
67,401 colour fundus images from 4613 study participants
DL
Estimate 5-year risk of progression to wet AMD and geographic atrophy based on 9-step AREDS severity scale
Weighted κ scores = 0.77 for the 4-step and 0.74 for the 9-step AMD severity scales
Burlina et al. [68]
Colour
4507 AREDS participants and 2169 BMES participants
DL
Validation of a risk scoring system for prediction of progression
Sensitivity = 0.87; specificity = 0.73
Chiu et al. [69]
OCT
2795 patients
DL
Prediction to nAMD within a 6-month window
AUC = 0.74 (conversion scan ground truth) and 0.886 (1st injection ground truth)
Yim et al. [70]
OCT
671 AMD fellow eyes with 13,954 observations
DL
Predict progression to wet AMD
AUC = 0.96 ± 0.02 (3 months); 0.97 ± 0.02 (21 months)
Banerjee et al. [71]
OCT
686 fellow eyes with non-neovascular AMD at baseline
DL
Predict conversion from non-neovascular to neovascular AMD
Drusen are within 3 mm of fovea (HR = 1.45); mean drusen reflectivity (HR = 3.97)
Hallak et al. [72]
OCT
2146 OCT scans of 330 AMD eyes (244 patients)
DL
Predict neovascular AMD progression within 5 years
AUC = 0.74 (5 years), 0.92 (11 months), 0.86 (16 months), 0.7 (18 months), and 0.79 (48 months)
de Sisternes et al. [73]
OCT
71 eyes of patients with early AMD and contralateral neovascular AMD (9088 OCT B-scans)
CNN
Prediction of conversion from early/intermediate to advanced neovascular AMD
AUC = 0.87 (VGG16) and 0.91 (AMDnet)
Russakoff et al. [74]
OCT
495 eyes
DL
Predictive model to assess risk of conversion to advanced AMD
AUC = 0.68 for CNV and 0.8 for geographic atrophy
Schmidt-Erfurth et al. [75]
OCT
2712 OCT B-scans
DL
Segmentation of features associated with AMD
Dice = 0.63 ± 0.15; ICC = 0.66 ± 0.22
Liefers et al. [76]
OCT
930 OCT B-scans from 93 eyes of patients with neovascular AMD
CNN
Segmentation of features associated with neovascular AMD
Dice = 0.78 (IRF), 0.82 (SRF), 0.75 (SHRM), and 0.8 (PED); ICCs = 0.98 (IRF), 0.98 (SRF), 0.97 (SHRM), and 0.98 (PED)
Lee et al. [77]
RP
Colour
1128 RP and 517 healthy
CNN
Diagnose RP
AUROC 96.74%
Chen et al. [78]
Colour
99 RP and 21 healthy
FCN
Diagnose RP
Accuracy 99.52%
Arsalan et al. [79]
RP, best disease (BD), and Stargardt
FAF
73 healthy, 125 Stargardt, 160 RP, 125 BD
CNN
Classify images into each group
Accuracy 0.95
Miere et al. [80]
Stargardt
OCT
102 healthy (33 participants) and 647 Stargardt (60 patients)
CNN
Differentiate between Stargardt and healthy
Accuracy 99.6%
Shah et al. [81]
BVMD and AVMD
FAF and OCT
118 BVMD eyes and 96 AVMD eyes
CNN
Differentiate between BVMD and AVMD
AUROC 0.880
Crincoli et al. [23]
Stargardt and PRPH2-related pattern dystrophy
FAF
304 Stargardt (40 patients) and 66
PRPH2 (9 patients)
CNN
Differentiate between Stargardt and PRPH2-related pattern dystrophy
AUROC 0.890
Miere et al. [82]
ABCA4-, RP1L1-, and EYS-related retinopathy
OCT
58 IRD and 17 healthy
DL
Predict causative gene
ABCA4 100% accuracy; RP1L1 66.7 to 87.5%; EYS 82.4 to 100%; healthy 73.7 to 100%
Fujinami-Yokokawa et al. [83]
Stargardt disease
FAF
47 images (24 patients)
CNN
Segment flecks
Dice score: 0.54 ± 0.14 for diffuse speckled patterns; 0.71 ± 0.08 for discrete flecks
Charng et al. [84]
Stargardt disease and AMD
FAF
320 healthy
320 AMD and 100 Stargardt
CNN & FCN
Detect and segment atrophy
Atrophy screening: AMD 0.98 accuracy; Stargardt 0.95
Segmentation: AMD overlapping ratio of 0.89 ± 0.06; Stargardt: 0.78 ± 0.17
Wang et al. [85]
Stargardt and pattern dystrophy
FAF
110 AMD, 204 Stargardt, and pattern dystrophy
CNN
Differentiate between AMD and IRD-associated macular atrophy
AUROC 0.981
Miere et al. [86]
Stargardt disease
OCT
87 scan sets (22 patients)
FCN
Detect outer and inner limits of the retina
Mean difference: 2.10 µm and 0.059 mm3 in central macular thickness and volume between model and annotators
Kugelman et al. [87]
AOSLO
142 controls and 148 Stargardt
FCN
Identify cone photoreceptors
Dice score: 0.9431 ± 0.0482
Davidson et al. [88]
RP and CHM
OCT
300 B-scans with RP and 300 with CHM
FCN
EZ segmentation
Similarity of 0.894 ± 0.102 automatic vs manual grading for RP; 0.912 ± 0.055 for CHM
Camino et al. [89]
CHM
OCT
16 eyes CHM and 5 healthy
Nonneural (RF)
EZ segmentation
0.876 ± 0.066 Jaccard similarity index
Wang et al. [90]
USH2A-related RP
OCT
126 volume scans (126 patients)
CNN
EZ segmentation
Dice score 0.79 ± 0.27
Loo et al. [91]
OCT
86 volume scans (86 patients)
CNN
EZ segmentation
Dice score 0.867 ± 0.105
Wang et al. [92]
RP
OCT and IR
2918 (314 patients)
CNN and FCN
Predict VA below or above 20/40
AUROC 0.85
Liu et al. [93]
Blue cone monochromacy (BCM)
OCT
26 IRD, 16 BCM, 3 normal (patients)
Nonneural
Predict foveal sensitivity and VA
0.174 RMSE for VA and 2.91 for sensitivity
Sumaroka et al. [94]
RP, retinitis pigmentosa; CNN, convolutional neural network; AUROC, area under the receiver operating characteristic; FROC, free-response receiver operating characteristics; ICC, intraclass correlation coefficients; FCN, fully convolutional network; FAF, fundus autofluorescence; OCT, optical coherence tomography; BVMD, best vitelliform macular dystrophy; HR, hazard ratio; AVMD, adult-onset vitelliform macular dystrophy; DL, deep learning; AMD, age-related macular degeneration; IRD, inherited retinal dystrophy; AREDS, age-related eye disease study; BMES, Blue Mountains Eye Study; AOSLO, adaptive optics scanning laser ophthalmoscopy; CHM, choroideremia; EZ, ellipsoid zone; VA, visual acuity; RMSE, root-mean-square error; IRF, intraretinal fluid; SHRM, subretinal hyperreflective material; PED, pigment epithelial detachment; SRF, subretinal fluid
Wong et al. [96] developed a model to classify DR stages based on microaneurysms and haemorrhages, while others used exudates, blood vessel mapping, and the optic disc. [97, 98] The sensitivity of automatic DR screening has been reported as ranging from 75 to 94.7%, with comparable specificity and accuracy [99]. Several publicly available retinal datasets have been used to train, validate, and test these AI systems, and also to compare performance against other systems; namely, DIARETDB1, Kaggle, E-ophtha, DDR, DRIVE, HRF, Messidor, Messidor-2, STARE, CHASE DB1, Indian Diabetic Retinopathy Image Dataset (IDRiD), ROC, and DR2 [57, 100108]. Several studies have used these datasets to detect red lesions, microaneurysms, DR lesions, exudates, individual DR stages, and blood vessel segmentation [38, 40, 41, 43, 52, 109, 110].
Another area of focus is the detection of DMO, currently assessed by OCT as the gold standard. AI-based groups have tried detecting DMO from colour fundus photography based on exudates and accurate identification of the macula. Automated detection via OCT imaging is ongoing, focusing on retinal layer segmentation [111, 112] and specific lesion (e.g. cysts) identification [113118]. Recently, DL has also been used to detect macular thickening based on colour photographs, and it has been found to be comparable to OCT-measured thickness [119].
Multiple programs have tried to use AI-based methods in population-based screening for DR. The United States Food and Drug Administration (US FDA) has recently approved IDx-DR, a CNN for screening DR stages in adults aged 22 years or older [49, 120]. Initial versions of IDx-DR have been evaluated as part of the Iowa Detection Programme and have shown good results in White, North African, and Sub-Saharan populations [25]. Similar software, like the RetmarkerDR in Portugal and EyeArt in Canada, have been tested in local screening programs [121, 122]. Multiple South-Asian eye institutes are also involved in development and validation of AI-based algorithms in DR [95, 123, 124]. Recently, a Singapore-based DL tool has shown comparable diagnostic accuracy to manual grading, and a semi-automated DL model involving a secondary human assessment may prove to be the most cost-effective model [125, 126]. Their real-world performance remains to be tested [127].
The use of AI with DL tools has great potential in AMD, both for diagnostic purposes—while allowing for a more efficient and accurate approach—to prognostication of affected individuals and perhaps to directly determine (predict) efficacy of treatments. The most common imaging modalities being explored in the field of AI for AMD are OCT, colour fundus image, and fundus autofluorescence (FAF). OCT-angiography (OCTA) has also been used in DL approaches to diagnose and classify AMD, achieving high accuracy and sensitivity [128, 129]. Due to the huge number of studies, selected key ones will be discussed, with a summary of a broad range of studies in Table 1.
One of the first attempts to evaluate ML algorithms in risk assessment of AMD was a European study by van Grinsven et al. that aimed to detect and quantify drusen on colour fundus photographs in eyes without and with early to moderate AMD [61]. This study demonstrated that the proposed system was in keeping with experienced human observers in detecting the presence of drusen as well as estimating the area, with an ICC greater than 0.85. For AMD risk assessment, it achieved a ROC of 0.948 and 0.954—similar performance to human graders. Subsequently, the same group explored another algorithm for automatic detection of reticular pseudodrusen (RPD) [62]. This followed a multimodal imaging approach using colour fundus, FAF, and near-infrared images, with automated quantification having similar performance to the observers.
In 2018, Schmidt-Erfuth et al. evaluated the predictive potential of ML in terms of best-corrected visual acuity (BCVA) by analysing OCT volume scan features—intraretinal fluid (IRF), subretinal fluid (SRF), and pigment epithelial detachment (PED) [130]. A modest correlation was found between BCVA and OCT at baseline (R2 = 0.21), while functional outcome prediction accuracy increased in linear fashion. The same group then explored automated quantification of fluid volumes using a DL method and a CNN, using OCT data from the HARBOR study (NCT00891735) for neovascular AMD (nAMD) [131]. Retinal fluid volumes (IRF, SRF, and PED) were then validated by the authors as important biomarkers in nAMD [132].
A more recent study attempted to introduce an AI system that combines 3D OCT images and automatic tissue maps in individuals with unilateral nAMD to predict progression in the contralateral eye [70]. It achieved a sensitivity of 80% at 55% specificity and 34% specificity at 90% sensitivity while being able to identify high-risk groups and changes in anatomy before conversion to nAMD, outperforming 5 out of 6 experts. Also, the age-related eye disease studies (AREDS and AREDS2) used DL algorithms and survival analysis to predict risk of late AMD, which achieved high prognostic accuracy [133].
Several segmentation models have been described in AMD. In 2018, De Fauw et al. created a landmark OCT image segmentation model that utilised a DL framework to perform segmentation and automated diagnosis of retinal diseases [134]. Subsequently, Liefers et al. validated a DL model for segmentation of retinal features specifically in individuals with atrophic AMD and nAMD, with results comparable to independent observers [135]. A further automated segmentation algorithm with a CNN has been explored to quantify IRF, SRF, PED, and subretinal hyperreflective material (SHRM) in nAMD [136]. There was good agreement for both the segmentation and detection of lesions between clinicians and the network (dice scores ≥ 0.75 for all features). Two applications with validated automated DL segmentation algorithms are currently commercially available: RetinAI (Medical AG, Switzerland) and RetInSight (Vienna, Austria) [137].
Dry AMD with geographic atrophy (GA) has also been actively investigated. Zhang et al. developed a DL model that segments and classifies GA on OCT images, achieving similar performance to manual specialist assessment [138]. Another group segmented GA in both OCT and FAF images and had reasonable agreement, with better performance (highest dice) in FAF [139]. GA algorithms have also been used to predict VA, with certain features such as photoreceptor degeneration having high predictive significance [140].

Inherited retinal disorders (IRD)

AI algorithms using multimodal imaging techniques have been developed to facilitate the diagnosis [78], classification [80], decipher the genetic aetiology [83], and measure the progression rate of IRD [89, 84].
Chen et al. have developed a CNN that detects if a patient has retinitis pigmentosa (RP) by analysing colour fundus images, with an overall accuracy of 96% (versus 81.5% from four ophthalmology experts) [78]. Another group proposed an FCN that detects pigment in colour images and diagnoses RP with an accuracy of 99.5% [79].
To predict aetiologies, Miere et al. have created a CNN model that can distinguish between FAF images from patients with Stargardt disease (STGD), RP, and best disease (BD), with an overall accuracy of 0.95 [80]. Furthermore, Fujinami-Yokokawa et al. used OCT images to predict causative genes (ABCA4, RP1L1, and EYS) through a DL platform [83]. They achieved an accuracy of 100% for ABCA4, 66.7 to 87.5% for RP1L1, 82.4 to 100% for EYS, and 73.7 to 100% for healthy control images. Miere et al. also created a CNN that is able to outperform specialists in distinguishing between FAF images of STGD and PRPH2-related macular dystrophy (AUROC 0.890 versus experts 0.816) [82]. Shah et al. also achieved an accuracy of 99.6% with a model distinguishing between OCT images from patients with STGD and controls [81]. Crincoli et al. combined image processing with a CNN to differentiate between BD and adult-onset vitelliform macular dystrophy using FAF and OCT images, with an AUROC of 0.880 [23]. Moreover, this endeavour has been recently markedly upscaled by Pontikos et al. to differentiate between 36 gene classes by exploiting multimodal imaging [141]. However, further development is needed, given more than 300 genes are known to cause IRD to date.
STGD is the most prevalent inherited macular dystrophy, and it can affect both children and adults, with multiple ongoing clinical trials [142]. Charng et al. developed a CNN algorithm that segments flecks and is able to monitor their progression over time [84]. They obtained an overall agreement between manual and automatic segmentation of 0.54 ± 0.14 dice score for diffuse speckled patterns and 0.71 ± 0.08 for discrete flecks. Wang et al. also used FAF images, detecting and quantifying areas of atrophy in STGD and AMD [85]. They obtained an accuracy of 0.98 for differentiating normal eyes from those with AMD-related atrophy and 0.95 for eyes with STGD. Atrophic areas were also segmented manually and automatically, with an overlap ratio of 0.89 ± 0.06 in AMD and 0.78 ± 0.17 in STGD [85]. Miere et al. also assessed atrophy and developed a CNN that differentiates between FAF images with GA secondary to AMD and IRD-associated, with an AUROC of 0.981 [86].
Automatic macular OCT segmentation by the device manufacturers is often inaccurate in IRD, requiring manual correction in over one-third of scans [143]. OCT images of STGD were used to create an improved DL-based algorithm that is able to segment the inner and outer retinal limits, providing faster and better macular thickness and volume quantification [87]. Lastly, adaptive optics scanning light ophthalmoscopy images of STGD have also been used to develop an FCN that is able to accurately count macular cones (dice score: 0.9431 ± 0.0482) [88].
Other tools are being designed to assess disease severity and potentially have applications in determining eligibility for interventional trials. A CNN has been developed by Camino et al. that segments preserved EZ area on OCT images from patients with RP and choroideremia (CHM) [89]. This tool reached 0.894 ± 0.102 similarity between automatic and manual grading for RP and 0.912 ± 0.055 for CHM. Loo et al. also targeted EZ segmentation and validated their algorithm for macular telangiectasia in patients with USH2A-related RP, with excellent applicability (dice score 0.79 ± 0.27) [91]. Similarly, Wang et al. also tested an EZ segmentation CNN in USH2A-RP and obtained a Dice score of 0.867 ± 0.105 [92]. CHM EZ segmentation was then attempted by Wang et al. through a nonneural random forest approach and reached a Jaccard similarity index between manual and automated segmentation of 0.876 ± 0.066 [90].
Predicting VA based on OCT and infrared images in RP has been assessed by Liu et al. They were able to determine if a patient with RP had VA below or above 20/40, with an AUC of 0.85 [93]. Sumaroka et al. also developed a nonneural network to predict foveal sensitivity (Humphrey visual field testing), VA, and possible outcome of therapy in patients with blue cone monochromacy based on OCT scans, with good results [94].

Retinopathy of prematurity (ROP)

ROP is an important cause of preventable childhood blindness worldwide [144]. ROP causes abnormal blood vessel growth and can be detected by trained ophthalmologists using indirect ophthalmoscopy, with access to adequate, timely screening potentially limited due to the requirement of highly trained personnel and equipment. DL-based detection and staging of ROP[145] by evaluation of posterior pole fundus images has been attempted with high sensitivity and specificity [146]. Authors have developed a ROP vascular severity score with good correlation with the labels set by the International Classification of Retinopathy of Prematurity committee [147]. The DeepROP score [148] and i-ROP DL system are DL algorithms developed to evaluate clinically significant severe ROP at the posterior pole [149]. ROP plus disease, a more aggressive form of ROP, is often difficult to diagnose given the lack of consensus among ophthalmologists; several authors have evaluated automated algorithms that may be able to objectively diagnose plus disease [150153].
These study limitations are the review of the literature in a non-systematic approach, possibly leading to some papers being omitted or not adequately prioritised; and editorial restrictions, which prevented us from doing a comprehensive review of AI applications in all retinal disorders. Substantial research has been undertaken in other fields of medical retina (e.g., uveitis and oncology), which will be reviewed in a subsequent project. [154, 155]

Concluding remarks and future directions

Retinal disease has been at the forefront of AI in ophthalmology, with the first AI-related publication being on DR. Since then, research groups focusing their efforts on AI have multiplied around the world, targeting all aspects of the patient journey, including diagnosis, triage, and prognostication, by leveraging multiple imaging (and functional) modalities, as well as a range of AI tools. A shortage of medical professionals is anticipated in the short term, likely further increasing healthcare inequalities and challenging our ability to improve care for preventable diseases [156]. AI represents one important approach to help meet these challenges and moreover facilitate improvements in patient care—both at the individual level with more timely, accurate, and bespoke management, as well as population-level, large-scale healthcare. Ever-improving DNN and CNN algorithms can become a helping hand for healthcare to lean on towards meeting current capability endpoints.
Despite the huge promise, many challenges remain for AI in ophthalmology, including, (i) the need for larger, more diverse, and representative datasets that fully represent real life, (ii) the closer collaboration by experts (both national and international) to develop disease-specific consensus and subsequently provide a comprehensive large volume of image grading, and (iii) greater synergy between healthcare professionals, patients, and data scientists, communicating and improving the software interface as it is being iteratively created, and ensuring it complements the human interaction that underpins the practice of medicine, rather than seeking to replace it [157]. Further uses of AI are yet to be explored, such as multimodal inputs to determine the best candidates for interventional clinical trials, the selection of the ideal anti-VEGF and therapeutic scheme in nAMD, and the estimation of functional impairment based on structural parameters for IRD, among others.
The future of healthcare will increasingly incorporate the advantages that AI can provide to improve the lives of our patients and no doubt perform assessments quicker and more accurately than retina specialists can currently sustainably provide, allowing us to spend more time being better clinicians and scientists. Nevertheless, as always with new technology, there will be new learnings and surprises along the way.

Declarations

Ethical approval

This article does not contain any studies with animals or human participants performed by any of the authors.

Conflict of interest

The authors alone are responsible for the content and writing of this article. MM consults for MeiraGTx Ltd. All other authors certify that they have no affiliations with or involvement in any organisation or entity with any financial interest (such as honoraria; educational grants; participation in speakers’ bureaus; membership, employment, consultancies, stock ownership, or other equity interest; expert testimony or patent-licensing arrangements) or non-financial interest (such as personal or professional relationships, affiliations, knowledge, or beliefs) in the subject matter or materials discussed in this manuscript.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Jetzt e.Med zum Sonderpreis bestellen!

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Jetzt bestellen und 100 € sparen!

Gräfes Archiv

Online-Abonnement

Literatur
5.
Zurück zum Zitat Ting DSW, Lin H, Ruamviboonsuk P et al (2020) Artificial intelligence, the internet of things, and virtual clinics: ophthalmology at the digital translation forefront. Lancet Digit Health 2:e8–e9CrossRefPubMed Ting DSW, Lin H, Ruamviboonsuk P et al (2020) Artificial intelligence, the internet of things, and virtual clinics: ophthalmology at the digital translation forefront. Lancet Digit Health 2:e8–e9CrossRefPubMed
8.
Zurück zum Zitat Dias R, Torkamani A (2019) Artificial intelligence in clinical and genomic diagnostics. Genome Med 11:1–12CrossRef Dias R, Torkamani A (2019) Artificial intelligence in clinical and genomic diagnostics. Genome Med 11:1–12CrossRef
13.
Zurück zum Zitat El Naqa I, Murphy MJ (2015) What is machine learning?. Springer International Publishing, Berlin, pp 3–11 El Naqa I, Murphy MJ (2015) What is machine learning?. Springer International Publishing, Berlin, pp 3–11
14.
Zurück zum Zitat Tizhoosh HR (2005, December) Reinforcement learning based on actions and opposite actions. In International conference on artificial intelligence and machine learning, vol 414 Tizhoosh HR (2005, December) Reinforcement learning based on actions and opposite actions. In International conference on artificial intelligence and machine learning, vol 414
16.
Zurück zum Zitat Ghods A, Cook DJ (2021) A survey of deep network techniques all classifiers can adopt. Data Min Knowl Discov 35:46–87CrossRefPubMed Ghods A, Cook DJ (2021) A survey of deep network techniques all classifiers can adopt. Data Min Knowl Discov 35:46–87CrossRefPubMed
18.
Zurück zum Zitat Dhillon A, Verma GK (2020) Convolutional neural network: a review of models, methodologies and applications to object detection. Progress in Artificial Intelligence 9:85–112CrossRef Dhillon A, Verma GK (2020) Convolutional neural network: a review of models, methodologies and applications to object detection. Progress in Artificial Intelligence 9:85–112CrossRef
24.
Zurück zum Zitat Chicco D (2017) Ten quick tips for machine learning in computational biology. BioData Min 10:1–17CrossRef Chicco D (2017) Ten quick tips for machine learning in computational biology. BioData Min 10:1–17CrossRef
35.
Zurück zum Zitat Szegedy C, Vanhoucke V, Ioffe S, Shlens J, Wojna Z (2016) Rethinking the inception architecture for computer vision. In Proceedings of the IEEE conference on computer vision and pattern recognition, Las Vegas, pp 2818–2826 Szegedy C, Vanhoucke V, Ioffe S, Shlens J, Wojna Z (2016) Rethinking the inception architecture for computer vision. In Proceedings of the IEEE conference on computer vision and pattern recognition, Las Vegas, pp 2818–2826
37.
Zurück zum Zitat He K, Zhang X, Ren S, Sun J (2016) Deep residual learning for image recognition. In Proceedings of the IEEE conference on computer vision and pattern recognition, Las Vegas, pp 770–778 He K, Zhang X, Ren S, Sun J (2016) Deep residual learning for image recognition. In Proceedings of the IEEE conference on computer vision and pattern recognition, Las Vegas, pp 770–778
42.
44.
Zurück zum Zitat Esfahani M T, Ghaderi M, Kafiyeh R (2018) Classification of diabetic and normal fundus images using new deep learning method. Leonardo Electron J Pract Technol 17(32):233–248 Esfahani M T, Ghaderi M, Kafiyeh R (2018) Classification of diabetic and normal fundus images using new deep learning method. Leonardo Electron J Pract Technol 17(32):233–248
46.
Zurück zum Zitat Jiang H, Yang K, Gao M, Zhang D, Ma H, Qian W (2019) An interpretable ensemble deep learning model for diabetic retinopathy disease classification. In: 2019 41st Annual International Conference of the IEEE Engineering in Medicine and Biology Society (EMBC), Berlin, Germany, pp 2045–2048. https://doi.org/10.1109/EMBC.2019.8857160 Jiang H, Yang K, Gao M, Zhang D, Ma H, Qian W (2019) An interpretable ensemble deep learning model for diabetic retinopathy disease classification. In: 2019 41st Annual International Conference of the IEEE Engineering in Medicine and Biology Society (EMBC), Berlin, Germany, pp 2045–2048. https://​doi.​org/​10.​1109/​EMBC.​2019.​8857160
52.
56.
Zurück zum Zitat Harangi B, Toth J, Baran A, Hajdu A (2019) Automatic screening of fundus images using a combination of convolutional neural network and hand-crafted features. In: 2019 41st Annual International Conference of the IEEE Engineering in Medicine and Biology Society (EMBC), Berlin, pp 2699–2702. https://doi.org/10.1109/EMBC.2019.8857073 Harangi B, Toth J, Baran A, Hajdu A (2019) Automatic screening of fundus images using a combination of convolutional neural network and hand-crafted features. In: 2019 41st Annual International Conference of the IEEE Engineering in Medicine and Biology Society (EMBC), Berlin, pp 2699–2702. https://​doi.​org/​10.​1109/​EMBC.​2019.​8857073
61.
Zurück zum Zitat van Grinsven MJJP, Lechanteur YTE, van de Ven JPH et al (2013) Automatic drusen quantification and risk assessment of age-related macular degeneration on color fundus images. Invest Ophthalmol Vis Sci 54:3019–3027CrossRefPubMed van Grinsven MJJP, Lechanteur YTE, van de Ven JPH et al (2013) Automatic drusen quantification and risk assessment of age-related macular degeneration on color fundus images. Invest Ophthalmol Vis Sci 54:3019–3027CrossRefPubMed
62.
Zurück zum Zitat Van Grinsven MJJP, Buitendijk GHS, Brussee C et al (2015) Automatic identification of reticular pseudodrusen using multimodal retinal image analysis. Invest Ophthalmol Vis Sci 56:633–639CrossRefPubMed Van Grinsven MJJP, Buitendijk GHS, Brussee C et al (2015) Automatic identification of reticular pseudodrusen using multimodal retinal image analysis. Invest Ophthalmol Vis Sci 56:633–639CrossRefPubMed
70.
Zurück zum Zitat Yim J, Chopra R, Spitz T et al (2020) Predicting conversion to wet age-related macular degeneration using deep learning. Nat Med 26:892–899CrossRefPubMed Yim J, Chopra R, Spitz T et al (2020) Predicting conversion to wet age-related macular degeneration using deep learning. Nat Med 26:892–899CrossRefPubMed
80.
Zurück zum Zitat Miere A, Le Meur T, Bitton K, Pallone C, Semoun O, Capuano V, Colantuono D, Taibouni K, Chenoune Y, Astroz P, Berlemont S, Petit E, Souied E (2020) Deep learning-based classification of inherited retinal diseases using fundus autofluorescence. J Clin Med 9(10):3303. https://doi.org/10.3390/jcm9103303 Miere A, Le Meur T, Bitton K, Pallone C, Semoun O, Capuano V, Colantuono D, Taibouni K, Chenoune Y, Astroz P, Berlemont S, Petit E, Souied E (2020) Deep learning-based classification of inherited retinal diseases using fundus autofluorescence. J Clin Med 9(10):3303. https://​doi.​org/​10.​3390/​jcm9103303
82.
Zurück zum Zitat Miere A, Zambrowski O, Kessler A, Mehanna C-J, Pallone C, Seknazi D, Denys P, Amoroso F, Petit E, Souied EH (2021) Deep learning to distinguish abca4-related stargardt disease from prph2-related pseudo-stargardt pattern dystrophy. J Clin Med 10(24):5742. https://doi.org/10.3390/s20123454 Miere A, Zambrowski O, Kessler A, Mehanna C-J, Pallone C, Seknazi D, Denys P, Amoroso F, Petit E, Souied EH (2021) Deep learning to distinguish abca4-related stargardt disease from prph2-related pseudo-stargardt pattern dystrophy. J Clin Med 10(24):5742. https://​doi.​org/​10.​3390/​s20123454
85.
Zurück zum Zitat Wang Z, Sadda SR, Hu Z (2019) Deep learning for automated screening and semantic segmentation of age-related and juvenile atrophic macular degeneration. In: Proc. SPIE 10950, Medical Imaging 2019: Computer-Aided Diagnosis, 109501Q. https://doi.org/10.1117/12.2511538 Wang Z, Sadda SR, Hu Z (2019) Deep learning for automated screening and semantic segmentation of age-related and juvenile atrophic macular degeneration. In: Proc. SPIE 10950, Medical Imaging 2019: Computer-Aided Diagnosis, 109501Q. https://​doi.​org/​10.​1117/​12.​2511538
92.
Zurück zum Zitat Wang YZ, Birch DG (2022) Performance of deep learning models in automatic measurement of ellipsoid zone area on baseline optical coherence tomography (OCT) images from the rate of progression of USH2A-Related retinal degeneration (RUSH2A) study. Front Med (Lausanne) 9:932498. https://doi.org/10.3389/fmed.2022.932498 Wang YZ, Birch DG (2022) Performance of deep learning models in automatic measurement of ellipsoid zone area on baseline optical coherence tomography (OCT) images from the rate of progression of USH2A-Related retinal degeneration (RUSH2A) study. Front Med (Lausanne) 9:932498. https://​doi.​org/​10.​3389/​fmed.​2022.​932498
96.
Zurück zum Zitat Wong LY, Acharya R, Venkatesh YV, Chee C, Min LC (2008) Identification of different stages of diabetic retinopathy using retinal optical images. Inf Sci 178(106):21 Wong LY, Acharya R, Venkatesh YV, Chee C, Min LC (2008) Identification of different stages of diabetic retinopathy using retinal optical images. Inf Sci 178(106):21
100.
Zurück zum Zitat Kauppi T, Pietilä J, Kalesnykiene V, Kamarainen JK, Lensu L, Sorri I, Raninen A, Voutilainen R, Uusitalo H, Kälviäinen H (2007, September) The diaretdb1 diabetic retinopathy database and evaluation protocol. In BMVC 1(1):10 Kauppi T, Pietilä J, Kalesnykiene V, Kamarainen JK, Lensu L, Sorri I, Raninen A, Voutilainen R, Uusitalo H, Kälviäinen H (2007, September) The diaretdb1 diabetic retinopathy database and evaluation protocol. In BMVC 1(1):10
114.
Zurück zum Zitat Schlegl T, Glodan A-M, Podkowinski D, Waldstein SM, Gerendas BS, Schmidt-Erfurth U, Langs G (2015) Automatic segmentation and classifcation of intraretinal cystoid fuid and subretinal fuid in 3d-oct using convolutional neural networks. Investig Ophthalmol Vis Sci 56(7):5920 Schlegl T, Glodan A-M, Podkowinski D, Waldstein SM, Gerendas BS, Schmidt-Erfurth U, Langs G (2015) Automatic segmentation and classifcation of intraretinal cystoid fuid and subretinal fuid in 3d-oct using convolutional neural networks. Investig Ophthalmol Vis Sci 56(7):5920
117.
Zurück zum Zitat Venhuizen FG, van Ginneken B, Bloemen B, van Grinsven MJJP, Philipsen R, Hoyng C, Theelen T, Sánchez CI (2015) Automated age-related macular degeneration classification in OCT using unsupervised feature learning. In: Proc. SPIE 9414, Medical Imaging 2015: Computer-Aided Diagnosis, 94141I. https://doi.org/10.1117/12.2081521 Venhuizen FG, van Ginneken B, Bloemen B, van Grinsven MJJP, Philipsen R, Hoyng C, Theelen T, Sánchez CI (2015) Automated age-related macular degeneration classification in OCT using unsupervised feature learning. In: Proc. SPIE 9414, Medical Imaging 2015: Computer-Aided Diagnosis, 94141I. https://​doi.​org/​10.​1117/​12.​2081521
121.
Zurück zum Zitat Ribeiro L, Oliveira C, M, Neves C, Ramos J, D, Ferreira H, Cunha-Vaz J (2015) Screening for diabetic retinopathy in the central region of portugal. Added Value of Automated ‘Disease/No Disease' Grading. Ophthalmologica 233:96–103. https://doi.org/10.1159/000368426 Ribeiro L, Oliveira C, M, Neves C, Ramos J, D, Ferreira H, Cunha-Vaz J (2015) Screening for diabetic retinopathy in the central region of portugal. Added Value of Automated ‘Disease/No Disease' Grading. Ophthalmologica 233:96–103. https://​doi.​org/​10.​1159/​000368426
130.
Zurück zum Zitat Schmidt-Erfurth U, Bogunovic H, Sadeghipour A et al (2018) Machine learning to analyze the prognostic value of current imaging biomarkers in neovascular age-related macular degeneration. Ophthalmol Retina 2:24–30CrossRefPubMed Schmidt-Erfurth U, Bogunovic H, Sadeghipour A et al (2018) Machine learning to analyze the prognostic value of current imaging biomarkers in neovascular age-related macular degeneration. Ophthalmol Retina 2:24–30CrossRefPubMed
131.
Zurück zum Zitat Schmidt-Erfurth U, Vogl W-D, Jampol LM, Bogunović H (2020) Application of automated quantification of fluid volumes to anti–VEGF therapy of neovascular age-related macular degeneration. Ophthalmology 127:1211–1219CrossRefPubMed Schmidt-Erfurth U, Vogl W-D, Jampol LM, Bogunović H (2020) Application of automated quantification of fluid volumes to anti–VEGF therapy of neovascular age-related macular degeneration. Ophthalmology 127:1211–1219CrossRefPubMed
132.
Zurück zum Zitat Keenan TDL, Chakravarthy U, Loewenstein A et al (2021) Automated quantitative assessment of retinal fluid volumes as important biomarkers in neovascular age-related macular degeneration. Am J Ophthalmol 224:267–281CrossRefPubMedPubMedCentral Keenan TDL, Chakravarthy U, Loewenstein A et al (2021) Automated quantitative assessment of retinal fluid volumes as important biomarkers in neovascular age-related macular degeneration. Am J Ophthalmol 224:267–281CrossRefPubMedPubMedCentral
133.
Zurück zum Zitat Peng Y, Keenan TD, Chen Q et al (2020) Predicting risk of late age-related macular degeneration using deep learning. NPJ Digit Med 3:1–10CrossRef Peng Y, Keenan TD, Chen Q et al (2020) Predicting risk of late age-related macular degeneration using deep learning. NPJ Digit Med 3:1–10CrossRef
135.
Zurück zum Zitat Liefers B, Taylor P, Alsaedi A et al (2021) Quantification of key retinal features in early and late age-related macular degeneration using deep learning. Am J Ophthalmol 226:1–12CrossRefPubMed Liefers B, Taylor P, Alsaedi A et al (2021) Quantification of key retinal features in early and late age-related macular degeneration using deep learning. Am J Ophthalmol 226:1–12CrossRefPubMed
136.
Zurück zum Zitat Lee H, Kang KE, Chung H, Kim HC (2018) Automated segmentation of lesions including subretinal hyperreflective material in neovascular age-related macular degeneration. Am J Ophthalmol 191:64–75CrossRefPubMed Lee H, Kang KE, Chung H, Kim HC (2018) Automated segmentation of lesions including subretinal hyperreflective material in neovascular age-related macular degeneration. Am J Ophthalmol 191:64–75CrossRefPubMed
142.
Zurück zum Zitat Tanna P, Strauss RW, Fujinami K, Michaelides M (2017) Stargardt disease: clinical features, molecular genetics, animal models and therapeutic options. Br J Ophthalmol 101:25–30CrossRefPubMed Tanna P, Strauss RW, Fujinami K, Michaelides M (2017) Stargardt disease: clinical features, molecular genetics, animal models and therapeutic options. Br J Ophthalmol 101:25–30CrossRefPubMed
Metadaten
Titel
Artificial intelligence in retinal disease: clinical application, challenges, and future directions
verfasst von
Malena Daich Varela
Sagnik Sen
Thales Antonio Cabral De Guimaraes
Nathaniel Kabiri
Nikolas Pontikos
Konstantinos Balaskas
Michel Michaelides
Publikationsdatum
09.05.2023
Verlag
Springer Berlin Heidelberg
Erschienen in
Graefe's Archive for Clinical and Experimental Ophthalmology / Ausgabe 11/2023
Print ISSN: 0721-832X
Elektronische ISSN: 1435-702X
DOI
https://doi.org/10.1007/s00417-023-06052-x

Weitere Artikel der Ausgabe 11/2023

Graefe's Archive for Clinical and Experimental Ophthalmology 11/2023 Zur Ausgabe

Neu im Fachgebiet Augenheilkunde

Metastase in der periokulären Region

Metastasen Leitthema

Orbitale und periokuläre metastatische Tumoren galten früher als sehr selten. Aber mit der ständigen Aktualisierung von Medikamenten und Nachweismethoden für die Krebsbehandlung werden neue Chemotherapien und Strahlenbehandlungen eingesetzt. Die …

Staging und Systemtherapie bei okulären und periokulären Metastasen

Metastasen Leitthema

Metastasen bösartiger Erkrankungen sind die häufigsten Tumoren, die im Auge diagnostiziert werden. Sie treten bei ungefähr 5–10 % der Patienten mit soliden Tumoren im Verlauf der Erkrankung auf. Besonders häufig sind diese beim Mammakarzinom und …

CME: Wundheilung nach Trabekulektomie

Trabekulektomie CME-Artikel

Wird ein Glaukom chirurgisch behandelt, ist die anschließende Wundheilung von entscheidender Bedeutung. In diesem CME-Kurs lernen Sie, welche Pathomechanismen der Vernarbung zugrunde liegen, wie perioperativ therapiert und Operationsversagen frühzeitig erkannt werden kann.

„standard operating procedures“ (SOP) – Vorschlag zum therapeutischen Management bei periokulären sowie intraokulären Metastasen

Metastasen Leitthema

Peri- sowie intraokuläre Metastasen sind insgesamt gesehen selten und meist Zeichen einer fortgeschrittenen primären Tumorerkrankung. Die Therapie ist daher zumeist palliativ und selten kurativ. Zudem ist die Therapiefindung sehr individuell. Die …

Update Augenheilkunde

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.