Deep-learning-based clustering of OCT images for biomarker discovery in age-related macular degeneration (Pinnacle study report 4)

Read original: arXiv:2405.09549 - Published 5/17/2024 by Robbie Holland, Rebecca Kaye, Ahmed M. Hagag, Oliver Leingang, Thomas R. P. Taylor, Hrvoje Bogunovi'c, Ursula Schmidt-Erfurth, Hendrik P. N. Scholl, Daniel Rueckert, Andrew J. Lotery and 2 others
Total Score

0

🔗

Sign in to get full access

or

If you already have an account, we'll log you in

Overview

  • Current disease management relies on grading systems that lack prognostic value
  • Proposals for new biomarkers are limited to anecdotal observations
  • This work introduces a deep learning-based system to accelerate biomarker discovery for age-related macular degeneration (AMD)

Plain English Explanation

Diseases are currently managed using grading systems, which divide patients into broad categories based on their condition. However, these categories often don't provide accurate predictions about a patient's prognosis or future health. Additionally, new potential biomarkers (measurable indicators of a disease or condition) are typically proposed based on limited observations, rather than a more comprehensive approach.

This research paper presents a deep learning-based system that aims to accelerate the discovery of new biomarkers for age-related macular degeneration (AMD), a leading cause of vision loss. The system first trains a neural network using a technique called contrastive learning to identify features in 46,496 retinal images that are relevant to both known and unknown AMD biomarkers, without using any clinical annotations.

To interpret the discovered biomarkers, the researchers divide the images into 30 clusters, each containing similar features. They then conduct interviews with two independent teams of retinal specialists, who describe the distinct characteristics they observe in each cluster. This process allows the researchers to identify known biomarkers, as well as new potential biomarkers or combinations of biomarkers that are not yet used in current grading systems.

The discovered biomarkers demonstrate the ability to separate different stages of AMD, such as incomplete from complete retinal atrophy, and intraretinal from subretinal fluid. In simulations, the biomarkers also outperformed clinically-used grading systems in predicting a patient's prognosis.

Technical Explanation

The researchers first trained a neural network using self-supervised contrastive learning on 46,496 retinal optical coherence tomography (OCT) images. This allowed the network to discover features related to both known and unknown AMD biomarkers, without requiring any clinical annotations.

To interpret the discovered biomarkers, the researchers partitioned the images into 30 clusters, each containing similar features. They then conducted two parallel 1.5-hour semi-structured interviews with independent teams of retinal specialists, who described the distinct characteristics they observed in each cluster using clinical language.

The interviews revealed that both teams independently identified clearly distinct characteristics in 27 of the 30 clusters, with 23 of those related to AMD. Seven of the clusters corresponded to known biomarkers already used in established grading systems, while 16 depicted biomarker combinations or subtypes that are either not yet used, recently proposed, or previously unknown.

The discovered biomarkers were able to separate incomplete from complete retinal atrophy, intraretinal from subretinal fluid, and thick from thin choroids. In simulations, the biomarkers also outperformed clinically-used grading systems in terms of prognostic value.

Critical Analysis

The research demonstrates the potential of using deep learning-based approaches to accelerate the discovery of novel biomarkers, going beyond the limited set of biomarkers currently used in clinical grading systems. However, the authors acknowledge that the proposed system is still a proof-of-concept and requires further validation and testing on larger datasets before it can be widely adopted.

Additionally, the reliance on semi-structured interviews with retinal specialists to interpret the discovered biomarkers may introduce some subjectivity and bias. It would be valuable to explore more systematic and quantitative methods for biomarker interpretation, potentially leveraging multimodal normative modeling approaches to better understand the clinical significance of the discovered features.

Conclusion

This research presents a promising deep learning-based system for accelerating the discovery of novel biomarkers for age-related macular degeneration. By leveraging self-supervised contrastive learning and input from retinal specialists, the system was able to identify both known and new potential biomarkers that outperform current grading systems in prognostic value. While further validation is needed, this work highlights the potential of AI-driven biomarker discovery to improve disease management and patient outcomes.



This summary was produced with help from an AI and may contain inaccuracies - check out the links to read the original source documents!

Follow @aimodelsfyi on 𝕏 →

Related Papers

🔗

Total Score

0

Deep-learning-based clustering of OCT images for biomarker discovery in age-related macular degeneration (Pinnacle study report 4)

Robbie Holland, Rebecca Kaye, Ahmed M. Hagag, Oliver Leingang, Thomas R. P. Taylor, Hrvoje Bogunovi'c, Ursula Schmidt-Erfurth, Hendrik P. N. Scholl, Daniel Rueckert, Andrew J. Lotery, Sobha Sivaprasad, Martin J. Menten

Diseases are currently managed by grading systems, where patients are stratified by grading systems into stages that indicate patient risk and guide clinical management. However, these broad categories typically lack prognostic value, and proposals for new biomarkers are currently limited to anecdotal observations. In this work, we introduce a deep-learning-based biomarker proposal system for the purpose of accelerating biomarker discovery in age-related macular degeneration (AMD). It works by first training a neural network using self-supervised contrastive learning to discover, without any clinical annotations, features relating to both known and unknown AMD biomarkers present in 46,496 retinal optical coherence tomography (OCT) images. To interpret the discovered biomarkers, we partition the images into 30 subsets, termed clusters, that contain similar features. We then conduct two parallel 1.5-hour semi-structured interviews with two independent teams of retinal specialists that describe each cluster in clinical language. Overall, both teams independently identified clearly distinct characteristics in 27 of 30 clusters, of which 23 were related to AMD. Seven were recognised as known biomarkers already used in established grading systems and 16 depicted biomarker combinations or subtypes that are either not yet used in grading systems, were only recently proposed, or were unknown. Clusters separated incomplete from complete retinal atrophy, intraretinal from subretinal fluid and thick from thin choroids, and in simulation outperformed clinically-used grading systems in prognostic value. Overall, contrastive learning enabled the automatic proposal of AMD biomarkers that go beyond the set used by clinically established grading systems. Ultimately, we envision that equipping clinicians with discovery-oriented deep-learning tools can accelerate discovery of novel prognostic biomarkers.

Read more

5/17/2024

Enhancing Retinal Disease Classification from OCTA Images via Active Learning Techniques
Total Score

0

Enhancing Retinal Disease Classification from OCTA Images via Active Learning Techniques

Jacob Thrasher, Annahita Amireskandari, Prashnna Gyawali

Eye diseases are common in older Americans and can lead to decreased vision and blindness. Recent advancements in imaging technologies allow clinicians to capture high-quality images of the retinal blood vessels via Optical Coherence Tomography Angiography (OCTA), which contain vital information for diagnosing these diseases and expediting preventative measures. OCTA provides detailed vascular imaging as compared to the solely structural information obtained by common OCT imaging. Although there have been considerable studies on OCT imaging, there have been limited to no studies exploring the role of artificial intelligence (AI) and machine learning (ML) approaches for predictive modeling with OCTA images. In this paper, we explore the use of deep learning to identify eye disease in OCTA images. However, due to the lack of labeled data, the straightforward application of deep learning doesn't necessarily yield good generalization. To this end, we utilize active learning to select the most valuable subset of data to train our model. We demonstrate that active learning subset selection greatly outperforms other strategies, such as inverse frequency class weighting, random undersampling, and oversampling, by up to 49% in F1 evaluation.

Read more

7/23/2024

Beyond the Eye: A Relational Model for Early Dementia Detection Using Retinal OCTA Images
Total Score

0

Beyond the Eye: A Relational Model for Early Dementia Detection Using Retinal OCTA Images

Shouyue Liu, Jinkui Hao, Yonghuai Liu, Huazhu Fu, Xinyu Guo, Shuting Zhang, Yitian Zhao

Early detection of dementia, such as Alzheimer's disease (AD) or mild cognitive impairment (MCI), is essential to enable timely intervention and potential treatment. Accurate detection of AD/MCI is challenging due to the high complexity, cost, and often invasive nature of current diagnostic techniques, which limit their suitability for large-scale population screening. Given the shared embryological origins and physiological characteristics of the retina and brain, retinal imaging is emerging as a potentially rapid and cost-effective alternative for the identification of individuals with or at high risk of AD. In this paper, we present a novel PolarNet+ that uses retinal optical coherence tomography angiography (OCTA) to discriminate early-onset AD (EOAD) and MCI subjects from controls. Our method first maps OCTA images from Cartesian coordinates to polar coordinates, allowing approximate sub-region calculation to implement the clinician-friendly early treatment of diabetic retinopathy study (ETDRS) grid analysis. We then introduce a multi-view module to serialize and analyze the images along three dimensions for comprehensive, clinically useful information extraction. Finally, we abstract the sequence embedding into a graph, transforming the detection task into a general graph classification problem. A regional relationship module is applied after the multi-view module to excavate the relationship between the sub-regions. Such regional relationship analyses validate known eye-brain links and reveal new discriminative patterns.

Read more

8/12/2024

Machine Learning-Based Prediction of Key Genes Correlated to the Subretinal Lesion Severity in a Mouse Model of Age-Related Macular Degeneration
Total Score

0

Machine Learning-Based Prediction of Key Genes Correlated to the Subretinal Lesion Severity in a Mouse Model of Age-Related Macular Degeneration

Kuan Yan, Yue Zeng, Dai Shi, Ting Zhang, Dmytro Matsypura, Mark C. Gillies, Ling Zhu, Junbin Gao

Age-related macular degeneration (AMD) is a major cause of blindness in older adults, severely affecting vision and quality of life. Despite advances in understanding AMD, the molecular factors driving the severity of subretinal scarring (fibrosis) remain elusive, hampering the development of effective therapies. This study introduces a machine learning-based framework to predict key genes that are strongly correlated with lesion severity and to identify potential therapeutic targets to prevent subretinal fibrosis in AMD. Using an original RNA sequencing (RNA-seq) dataset from the diseased retinas of JR5558 mice, we developed a novel and specific feature engineering technique, including pathway-based dimensionality reduction and gene-based feature expansion, to enhance prediction accuracy. Two iterative experiments were conducted by leveraging Ridge and ElasticNet regression models to assess biological relevance and gene impact. The results highlight the biological significance of several key genes and demonstrate the framework's effectiveness in identifying novel therapeutic targets. The key findings provide valuable insights for advancing drug discovery efforts and improving treatment strategies for AMD, with the potential to enhance patient outcomes by targeting the underlying genetic mechanisms of subretinal lesion development.

Read more

9/10/2024