1
|
Naskar S, Sharma S, Kuotsu K, Halder S, Pal G, Saha S, Mondal S, Biswas UK, Jana M, Bhattacharjee S. The biomedical applications of artificial intelligence: an overview of decades of research. J Drug Target 2025; 33:717-748. [PMID: 39744873 DOI: 10.1080/1061186x.2024.2448711] [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: 10/31/2024] [Revised: 12/13/2024] [Accepted: 12/26/2024] [Indexed: 01/11/2025]
Abstract
A significant area of computer science called artificial intelligence (AI) is successfully applied to the analysis of intricate biological data and the extraction of substantial associations from datasets for a variety of biomedical uses. AI has attracted significant interest in biomedical research due to its features: (i) better patient care through early diagnosis and detection; (ii) enhanced workflow; (iii) lowering medical errors; (v) lowering medical costs; (vi) reducing morbidity and mortality; (vii) enhancing performance; (viii) enhancing precision; and (ix) time efficiency. Quantitative metrics are crucial for evaluating AI implementations, providing insights, enabling informed decisions, and measuring the impact of AI-driven initiatives, thereby enhancing transparency, accountability, and overall impact. The implementation of AI in biomedical fields faces challenges such as ethical and privacy concerns, lack of awareness, technology unreliability, and professional liability. A brief discussion is given of the AI techniques, which include Virtual screening (VS), DL, ML, Hidden Markov models (HMMs), Neural networks (NNs), Generative models (GMs), Molecular dynamics (MD), and Structure-activity relationship (SAR) models. The study explores the application of AI in biomedical fields, highlighting its enhanced predictive accuracy, treatment efficacy, diagnostic efficiency, faster decision-making, personalised treatment strategies, and precise medical interventions.
Collapse
Affiliation(s)
- Sweet Naskar
- Department of Pharmaceutics, Institute of Pharmacy, Kalyani, West Bengal, India
| | - Suraj Sharma
- Department of Pharmaceutics, Sikkim Professional College of Pharmaceutical Sciences, Sikkim, India
| | - Ketousetuo Kuotsu
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata, West Bengal, India
| | - Suman Halder
- Medical Department, Department of Indian Railway, Kharagpur Division, Kharagpur, West Bengal, India
| | - Goutam Pal
- Service Dispensary, ESI Hospital, Hoogly, West Bengal, India
| | - Subhankar Saha
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata, West Bengal, India
| | - Shubhadeep Mondal
- Department of Pharmacology, Momtaz Begum Pharmacy College, Rajarhat, West Bengal, India
| | - Ujjwal Kumar Biswas
- School of Pharmaceutical Science (SPS), Siksha O Anusandhan (SOA) University, Bhubaneswar, Odisha, India
| | - Mayukh Jana
- School of Pharmacy, Centurion University of Technology and Management, Centurion University, Bhubaneswar, Odisha, India
| | - Sunirmal Bhattacharjee
- Department of Pharmaceutics, Bharat Pharmaceutical Technology, Amtali, Agartala, Tripura, India
| |
Collapse
|
2
|
Nguyen N, Bernatsky S, Colmegna I, Berger C, Carrier N, Allard-Chamard H, Liang P, Roux S, Boire G, Hudson M. Predicting Response to Tumor Necrosis Factor Inhibitors in Rheumatoid Arthritis Using an Extended Set of Clinical Variables: An Unmet Need. Int J Rheum Dis 2025; 28:e70281. [PMID: 40387307 DOI: 10.1111/1756-185x.70281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 04/17/2025] [Accepted: 05/08/2025] [Indexed: 05/20/2025]
Affiliation(s)
- Nam Nguyen
- Department of Medicine, McGill University, Montreal, Canada
| | - Sasha Bernatsky
- Division of Clinical Epidemiology, Department of Medicine, McGill University Health Centre, Montreal, Canada
- Division of Rheumatology, Department of Medicine, McGill University Health Centre, Montreal, Canada
| | - Ines Colmegna
- Division of Rheumatology, Department of Medicine, McGill University Health Centre, Montreal, Canada
- Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Claudie Berger
- Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Nathalie Carrier
- Centre Intégré Universitaire de santé et de Services Sociaux de L'Estrie - Centre Hospitalier Universitaire de Sherbrooke (CIUSSSE-CHUS), Sherbrooke, Canada
| | - Hugues Allard-Chamard
- Centre Intégré Universitaire de santé et de Services Sociaux de L'Estrie - Centre Hospitalier Universitaire de Sherbrooke (CIUSSSE-CHUS), Sherbrooke, Canada
- Division of Rheumatology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Canada
| | - Patrick Liang
- Centre Intégré Universitaire de santé et de Services Sociaux de L'Estrie - Centre Hospitalier Universitaire de Sherbrooke (CIUSSSE-CHUS), Sherbrooke, Canada
- Division of Rheumatology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Canada
| | - Sophie Roux
- Centre Intégré Universitaire de santé et de Services Sociaux de L'Estrie - Centre Hospitalier Universitaire de Sherbrooke (CIUSSSE-CHUS), Sherbrooke, Canada
- Division of Rheumatology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Canada
| | - Gilles Boire
- Centre Intégré Universitaire de santé et de Services Sociaux de L'Estrie - Centre Hospitalier Universitaire de Sherbrooke (CIUSSSE-CHUS), Sherbrooke, Canada
- Division of Rheumatology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Canada
| | - Marie Hudson
- Department of Medicine, McGill University, Montreal, Canada
- Division of Rheumatology, Jewish General Hospital and the Lady Davis Institute for Medical Research, Montreal, Canada
| |
Collapse
|
3
|
Benavent D, Carmona L, García Llorente JF, Montoro M, Ramirez S, Otón T, Loza E, Gómez-Centeno A. Artificial intelligence to predict treatment response in rheumatoid arthritis and spondyloarthritis: a scoping review. Rheumatol Int 2025; 45:91. [PMID: 40192881 PMCID: PMC11976819 DOI: 10.1007/s00296-025-05825-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 02/28/2025] [Indexed: 04/10/2025]
Abstract
To analyse the types and applications of artificial intelligence (AI) technologies to predict treatment response in rheumatoid arthritis (RA) and spondyloarthritis (SpA). A comprehensive search in Medline, Embase, and Cochrane databases (up to August 2024) identified studies using AI to predict treatment response in RA and SpA. Data on study design, AI methodologies, data sources, and outcomes were extracted and synthesized. Findings were summarized descriptively. Of the 4257 articles identified, 89 studies met the inclusion criteria (74 on RA, 7 on SpA, 4 on Psoriatic Arthritis and 4 a mix of them). AI models primarily employed supervised machine learning techniques (e.g., random forests, support vector machines), unsupervised clustering, and deep learning. Data sources included electronic medical records, clinical biomarkers, genetic and proteomic data, and imaging. Predictive performance varied by methodology, with accuracy ranging from 60 to 70% and AUC values between 0.63 and 0.92. Multi-omics approaches and imaging-based models showed promising results in predicting responses to biologic DMARDs and JAK inhibitors but methodological heterogeneity limited generalizability. AI technologies exhibit substantial potential in predicting treatment responses in RA and SpA, enhancing personalized medicine. However, challenges such as methodological variability, data integration, and external validation remain. Future research should focus on refining AI models, ensuring their robustness across diverse patient populations, and facilitating their integration into clinical practice to optimize therapeutic decision-making in rheumatology.
Collapse
Affiliation(s)
- Diego Benavent
- Rheumatology Department, Hospital Universitari de Bellvitge, Carrer de la Feixa Llarga, s/n, L'Hospitalet de Llobregat, 08907, Barcelona, Spain.
| | - Loreto Carmona
- Instituto de Salud Musculoesquelética, 28039, Madrid, Spain
| | | | | | | | - Teresa Otón
- Instituto de Salud Musculoesquelética, 28039, Madrid, Spain
| | - Estíbaliz Loza
- Instituto de Salud Musculoesquelética, 28039, Madrid, Spain
| | - Antonio Gómez-Centeno
- Rheumatology Department, Parc Taulí Hospital UniversitariInstitut d'Investigació i Innovació Parc Taulí (I3PT), Sabadell, 28108, Barcelona, Spain
| |
Collapse
|
4
|
Garg S, Kitchen R, Gupta R, Pearson E. Applications of AI in Predicting Drug Responses for Type 2 Diabetes. JMIR Diabetes 2025; 10:e66831. [PMID: 40146874 PMCID: PMC11967697 DOI: 10.2196/66831] [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: 09/24/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 03/29/2025] Open
Abstract
Unlabelled Type 2 diabetes mellitus has seen a continuous rise in prevalence in recent years, and a similar trend has been observed in the increased availability of glucose-lowering drugs. There is a need to understand the variation in treatment response to these drugs to be able to predict people who will respond well or poorly to a drug. Electronic health records, clinical trials, and observational studies provide a huge amount of data to explore predictors of drug response. The use of artificial intelligence (AI), which includes machine learning and deep learning techniques, has the capacity to improve the prediction of treatment response in patients. AI can assist in the analysis of vast datasets to identify patterns and may provide valuable information on selecting an effective drug. Predicting an individual's response to a drug can aid in treatment selection, optimizing therapy, exploring new therapeutic options, and personalized medicine. This viewpoint highlights the growing evidence supporting the potential of AI-based methods to predict drug response with accuracy. Furthermore, the methods highlight a trend toward using ensemble methods as preferred models in drug response prediction studies.
Collapse
Affiliation(s)
- Shilpa Garg
- Diabetes Endocrinology and Reproductive Biology, School of Medicine, University of Dundee, Ninewells Avenue, Dundee, DD1 9SY, United Kingdom, 44 7443787733
| | | | | | - Ewan Pearson
- Diabetes Endocrinology and Reproductive Biology, School of Medicine, University of Dundee, Ninewells Avenue, Dundee, DD1 9SY, United Kingdom, 44 7443787733
| |
Collapse
|
5
|
Chen S, Nie R, Shen X, Wang Y, Luan H, Zeng X, Chen Y, Yuan H. Associations between age, red cell distribution width and 180-day and 1-year mortality in giant cell arteritis patients: mediation analyses and machine learning in a cohort study. Arthritis Res Ther 2025; 27:25. [PMID: 39923097 PMCID: PMC11806560 DOI: 10.1186/s13075-025-03477-z] [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: 09/09/2024] [Accepted: 01/09/2025] [Indexed: 02/10/2025] Open
Abstract
OBJECTIVE The aim of this study was to investigate the correlation between age, red cell distribution width (RDW) levels, and 180-day and 1-year mortality in giant cell arteritis (GCA) patients hospitalized or admitted to the ICU. METHODS Clinical data from GCA patients were extracted from the MIMIC-IV (3.0) database. Logistic and Cox regression analyses, Kaplan-Meier (KM) survival analysis, restricted cubic spline (RCS) analysis, and mediation effect analysis were employed to investigate the association between age, RDW levels, and 180-day and 1-year mortality in GCA patients hospitalized or admitted to the ICU. Predictive models were constructed using machine learning algorithms, and SHapley Additive exPlanations (SHAP) analysis was applied to evaluate the contributions of age and RDW levels to mortality in this patient population. RESULTS A total of 228 GCA patients were eligible for analysis. Our study identified both age and RDW levels (both with OR > 1, P < 0.05) as significant predictors of 180-day and 1-year mortality in GCA patients hospitalized or admitted to the ICU using multivariate logistic regression analysis. In multivariate Cox regression analysis, both age and RDW (both with HR > 1, P < 0.05) also emerged as prognostic risk factors for 180-day and 1-year mortality in this patient population. KM survival analysis further showed that GCA patients hospitalized or admitted to the ICU with higher age or elevated RDW levels had significantly lower survival rates compared to younger patients or those with lower RDW levels (P < 0.0001). Moreover, RCS analysis indicated a strong nonlinear relationship between RDW levels (threshold: 17.53%) and 1-year mortality in this population. Additionally, RDW levels were found to modestly mediate the relationship between age (per 10-year increase) and 180-day or 1-year mortality in GCA patients hospitalized or admitted to the ICU. The results of the machine learning analysis indicated that the model built using the random forest algorithm performed the best, with an area under the curve of 0.879. Furthermore, SHAP analysis revealed that both age and RDW levels made significant contributions to the prediction of mortality in GCA patients hospitalized or admitted to the ICU. CONCLUSIONS Older age and higher RDW levels were identified as independent risk factors for increased 180-day and 1-year mortality in GCA patients hospitalized or admitted to the ICU. Furthermore, elevated RDW levels modestly mediated the relationship between age and 180-day or 1-year mortality in this patient population.
Collapse
Affiliation(s)
- Si Chen
- Department of Clinical Laboratory, Beijing Anzhen Hospital, Capital Medical University, Anzhen Road No. 2, Chaoyang District, Beijing, 100029, China
| | - Rui Nie
- Department of Clinical Laboratory, Beijing Anzhen Hospital, Capital Medical University, Anzhen Road No. 2, Chaoyang District, Beijing, 100029, China
| | - Xiaoran Shen
- Department of Clinical Laboratory, Beijing Anzhen Hospital, Capital Medical University, Anzhen Road No. 2, Chaoyang District, Beijing, 100029, China
| | - Yan Wang
- Department of Clinical Laboratory, Beijing Anzhen Hospital, Capital Medical University, Anzhen Road No. 2, Chaoyang District, Beijing, 100029, China
| | - Haixia Luan
- Department of Clinical Laboratory, Beijing Anzhen Hospital, Capital Medical University, Anzhen Road No. 2, Chaoyang District, Beijing, 100029, China
| | - Xiaoli Zeng
- Department of Clinical Laboratory, Beijing Anzhen Hospital, Capital Medical University, Anzhen Road No. 2, Chaoyang District, Beijing, 100029, China
| | - Yanhua Chen
- School of Computer and Artificial Intelligence, Zhengzhou University, No. 100, Science Avenue, High-Tech Zone, Zhengzhou, Henan, 450001, China.
| | - Hui Yuan
- Department of Clinical Laboratory, Beijing Anzhen Hospital, Capital Medical University, Anzhen Road No. 2, Chaoyang District, Beijing, 100029, China.
| |
Collapse
|
6
|
Alten R, Behar C, Merckaert P, Afari E, Vannier-Moreau V, Ohayon A, Connolly SE, Najm A, Juge PA, Liu G, Rai A, Elbez Y, Lozenski K. Predicting abatacept retention using machine learning. Arthritis Res Ther 2025; 27:20. [PMID: 39893489 PMCID: PMC11786492 DOI: 10.1186/s13075-025-03484-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 01/16/2025] [Indexed: 02/04/2025] Open
Abstract
BACKGROUND The incorporation of machine learning is becoming more prevalent in the clinical setting. By predicting clinical outcomes, machine learning can provide clinicians with a valuable tool for refining precision medicine approaches and improving treatment outcomes. METHODS This was a post hoc analysis of pooled patient-level data from the global, real-world ACTION and ASCORE trials in patients with rheumatoid arthritis (RA) initiating abatacept. Patient demographic and disease characteristics were input across 10 machine learning models used to predict 12-month treatment retention. Retention was defined as treatment for > 365 days or ≤ 365 days in patients who achieved remission or major clinical response (based on European Alliance of Associations for Rheumatology response criteria). The pooled dataset was split into a training/validation cohort for model development and a test cohort for an unbiased evaluation of performance. SHapley Additive exPlanation (SHAP) values determined the level of importance and directionality for key patient features predicting abatacept retention. RESULTS The pooled ACTION and ASCORE dataset included 5320 patients with RA (mean [standard deviation] age 57.7 [12.7] years; 79% female). The 12-month abatacept retention rate was 61% (n = 3236) with a discontinuation rate of 39% (n = 2037). In the training set (n = 4218), the gradient-boosting classifier model demonstrated the best performance (testing accuracy: 62%). This model had an area under the receiver operating characteristic curve (95% confidence interval) of 0.620 (0.586, 0.653) and F1 score of 0.659 (0.625, 0.689) in the test set of patients (n = 1055). Using this model, the five most important variables predicting 12-month abatacept retention were low body mass index (BMI), low American College of Rheumatology functional status class, anti-citrullinated protein antibody (ACPA) positivity, low Patient Global Assessment, and younger age. CONCLUSIONS The gradient-boosting classifier model identified key patient features predictive of abatacept retention from this large, real-world study population. The SHAP values conveyed the directionality and importance of BMI, functional status, ACPA serostatus, Patient Global Assessment, and age for abatacept retention. Findings are consistent with previous observations and help validate the machine learning approach for predictive modelling in RA treatment, and may help inform clinical decision making. TRIAL REGISTRATION NCT02109666 (ACTION), NCT02090556 (ASCORE).
Collapse
Affiliation(s)
- Rieke Alten
- Schlosspark-Klinik University, Berlin, Germany.
| | | | | | | | | | | | | | - Aurélie Najm
- School of Infection and Immunity, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | | | | | - Angshu Rai
- Bristol Myers Squibb, Princeton, NJ, USA
| | | | | |
Collapse
|
7
|
Alsaber AR, Al-Herz A, Alawadhi B, Doush IA, Setiya P, AL-Sultan AT, Saleh K, Al-Awadhi A, Hasan E, Al-Kandari W, Mokaddem K, Ghanem AA, Attia Y, Hussain M, AlHadhood N, Ali Y, Tarakmeh H, Aldabie G, AlKadi A, Alhajeri H. Machine learning-based remission prediction in rheumatoid arthritis patients treated with biologic disease-modifying anti-rheumatic drugs: findings from the Kuwait rheumatic disease registry. Front Big Data 2024; 7:1406365. [PMID: 39421133 PMCID: PMC11484091 DOI: 10.3389/fdata.2024.1406365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 09/12/2024] [Indexed: 10/19/2024] Open
Abstract
Background Rheumatoid arthritis (RA) is a common condition treated with biological disease-modifying anti-rheumatic medicines (bDMARDs). However, many patients exhibit resistance, necessitating the use of machine learning models to predict remissions in patients treated with bDMARDs, thereby reducing healthcare costs and minimizing negative effects. Objective The study aims to develop machine learning models using data from the Kuwait Registry for Rheumatic Diseases (KRRD) to identify clinical characteristics predictive of remission in RA patients treated with biologics. Methods The study collected follow-up data from 1,968 patients treated with bDMARDs from four public hospitals in Kuwait from 2013 to 2022. Machine learning techniques like lasso, ridge, support vector machine, random forest, XGBoost, and Shapley additive explanation were used to predict remission at a 1-year follow-up. Results The study used the Shapley plot in explainable Artificial Intelligence (XAI) to analyze the effects of predictors on remission prognosis across different types of bDMARDs. Top clinical features were identified for patients treated with bDMARDs, each associated with specific mean SHAP values. The findings highlight the importance of clinical assessments and specific treatments in shaping treatment outcomes. Conclusion The proposed machine learning model system effectively identifies clinical features predicting remission in bDMARDs, potentially improving treatment efficacy in rheumatoid arthritis patients.
Collapse
Affiliation(s)
- Ahmad R. Alsaber
- College of Business and Economics, American University of Kuwait, Salmiya, Kuwait
| | - Adeeba Al-Herz
- Department of Rheumatology, Al-Amiri Hospital, Kuwait City, Kuwait
| | - Balqees Alawadhi
- Department of Food and Nutritional Sciences, The Public Authority for Applied Education & Training, Shuwaikh Industrial, Kuwait
| | - Iyad Abu Doush
- College of Engineering and Applied Sciences, American University of Kuwait, Salmiya, Kuwait
- Computer Science Department, Yarmouk University, Irbid, Jordan
| | - Parul Setiya
- College of Agriculture, Govind Ballabh Pant University of Agriculture and Technology, Pantnagar, India
| | - Ahmad T. AL-Sultan
- Department of Community Medicine and Behavioral Sciences, Kuwait University, Safat, Kuwait
| | - Khulood Saleh
- Department of Rheumatology, Farwaniya Hospital, Kuwait City, Kuwait
| | - Adel Al-Awadhi
- Department of Rheumatology, Al-Amiri Hospital, Kuwait City, Kuwait
| | - Eman Hasan
- Department of Rheumatology, Al-Amiri Hospital, Kuwait City, Kuwait
| | | | - Khalid Mokaddem
- Department of Rheumatology, Al-Amiri Hospital, Kuwait City, Kuwait
| | - Aqeel A. Ghanem
- Department of Rheumatology, Mubarak Al-Kabeer Hospital, Kuwait City, Kuwait
| | - Yousef Attia
- Department of Rheumatology, Al-Amiri Hospital, Kuwait City, Kuwait
| | - Mohammed Hussain
- Department of Rheumatology, Al-Amiri Hospital, Kuwait City, Kuwait
| | - Naser AlHadhood
- Department of Rheumatology, Farwaniya Hospital, Kuwait City, Kuwait
| | - Yaser Ali
- Department of Rheumatology, Mubarak Al-Kabeer Hospital, Kuwait City, Kuwait
| | - Hoda Tarakmeh
- Department of Rheumatology, Mubarak Al-Kabeer Hospital, Kuwait City, Kuwait
| | - Ghaydaa Aldabie
- Department of Rheumatology, Farwaniya Hospital, Kuwait City, Kuwait
| | - Amjad AlKadi
- Department of Rheumatology, Al-Sabah Hospital, Kuwait City, Kuwait
| | - Hebah Alhajeri
- Department of Rheumatology, Mubarak Al-Kabeer Hospital, Kuwait City, Kuwait
| |
Collapse
|
8
|
Mendoza-Pinto C, Sánchez-Tecuatl M, Berra-Romani R, Maya-Castro ID, Etchegaray-Morales I, Munguía-Realpozo P, Cárdenas-García M, Arellano-Avendaño FJ, García-Carrasco M. Machine learning in the prediction of treatment response in rheumatoid arthritis: A systematic review. Semin Arthritis Rheum 2024; 68:152501. [PMID: 39226650 DOI: 10.1016/j.semarthrit.2024.152501] [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: 01/14/2024] [Revised: 05/14/2024] [Accepted: 06/11/2024] [Indexed: 09/05/2024]
Abstract
OBJECTIVE This study aimed to investigate the current status and performance of machine learning (ML) approaches in providing reproducible treatment response predictions. METHODS This systematic review was conducted in accordance with the PRISMA statement and the CHARMS checklist. We searched PubMed, Cochrane Library, Web of Science, Scopus, and EBSCO databases for cohort studies that derived and/or validated ML models focused on predicting rheumatoid arthritis (RA) treatment response. We extracted data and critically appraised studies based on the Transparent Reporting of a Multivariable Prediction Model for Individual Prognosis or Diagnosis (TRIPOD) and Prediction Model Risk of Bias Assessment Tool (PROBAST) guidelines. RESULTS From 210 unduplicated records identified by the literature search, we retained 29 eligible studies. Of these studies, 10 developed a predictive model and reported a mean adherence to the TRIPOD guidelines of 45.6 % (95 % CI: 38.3-52.8 %). The remaining 19 studies not only developed a predictive model but also validated it externally, with a mean adherence of 42.9 % (95 % CI: 39.1-46.6 %). Most of the articles had an unclear risk of bias (41.4 %), followed by a high risk of bias, which was present in 37.9 %. CONCLUSIONS In recent years, ML methods have been increasingly used to predict treatment response in RA. Our critical appraisal revealed unclear and high risk of bias in most of the identified models, suggesting that researchers can do more to address the risk of bias and increase transparency, including the use of calibration measures and reporting methods for handling missing data. FUNDING None.
Collapse
Affiliation(s)
- Claudia Mendoza-Pinto
- Department of Rheumatology, Medicine School, Benemérita Universidad Autónoma de Puebla, Mexico; Rheumatology and Autoimmune Diseases Research Unit, Specialties Hospital UMAE-CIBIOR, Instituto Mexicano del Seguro Social, Puebla, Mexico
| | - Marcial Sánchez-Tecuatl
- Electronics Department, National Institute of Astrophysics, Optics and Electronics, Puebla, Mexico
| | - Roberto Berra-Romani
- Department of Biomedicine, Medicine School, Benemérita Universidad Autónoma de Puebla, Mexico
| | | | - Ivet Etchegaray-Morales
- Department of Rheumatology, Medicine School, Benemérita Universidad Autónoma de Puebla, Mexico
| | - Pamela Munguía-Realpozo
- Department of Rheumatology, Medicine School, Benemérita Universidad Autónoma de Puebla, Mexico; Rheumatology and Autoimmune Diseases Research Unit, Specialties Hospital UMAE-CIBIOR, Instituto Mexicano del Seguro Social, Puebla, Mexico.
| | - Maura Cárdenas-García
- Cell Physiology Laboratory, Medicine School, Benemérita Universidad Autónoma de Puebla, Mexico.
| | | | | |
Collapse
|
9
|
Nair A, Alagha MA, Cobb J, Jones G. Assessing the Value of Imaging Data in Machine Learning Models to Predict Patient-Reported Outcome Measures in Knee Osteoarthritis Patients. Bioengineering (Basel) 2024; 11:824. [PMID: 39199782 PMCID: PMC11351307 DOI: 10.3390/bioengineering11080824] [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: 07/04/2024] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 09/01/2024] Open
Abstract
Knee osteoarthritis (OA) affects over 650 million patients worldwide. Total knee replacement is aimed at end-stage OA to relieve symptoms of pain, stiffness and reduced mobility. However, the role of imaging modalities in monitoring symptomatic disease progression remains unclear. This study aimed to compare machine learning (ML) models, with and without imaging features, in predicting the two-year Western Ontario and McMaster Universities Arthritis Index (WOMAC) score for knee OA patients. We included 2408 patients from the Osteoarthritis Initiative (OAI) database, with 629 patients from the Multicenter Osteoarthritis Study (MOST) database. The clinical dataset included 18 clinical features, while the imaging dataset contained an additional 10 imaging features. Minimal Clinically Important Difference (MCID) was set to 24, reflecting meaningful physical impairment. Clinical and imaging dataset models produced similar area under curve (AUC) scores, highlighting low differences in performance AUC < 0.025). For both clinical and imaging datasets, Gradient Boosting Machine (GBM) models performed the best in the external validation, with a clinically acceptable AUC of 0.734 (95% CI 0.687-0.781) and 0.747 (95% CI 0.701-0.792), respectively. The five features identified included educational background, family history of osteoarthritis, co-morbidities, use of osteoporosis medications and previous knee procedures. This is the first study to demonstrate that ML models achieve comparable performance with and without imaging features.
Collapse
Affiliation(s)
- Abhinav Nair
- MSk Lab, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | - M. Abdulhadi Alagha
- MSk Lab, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
- Data Science Institute, London School of Economics and Political Science, London, UK
| | - Justin Cobb
- MSk Lab, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | - Gareth Jones
- MSk Lab, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
10
|
Salehi F, Lopera Gonzalez LI, Bayat S, Kleyer A, Zanca D, Brost A, Schett G, Eskofier BM. Machine Learning Prediction of Treatment Response to Biological Disease-Modifying Antirheumatic Drugs in Rheumatoid Arthritis. J Clin Med 2024; 13:3890. [PMID: 38999454 PMCID: PMC11242607 DOI: 10.3390/jcm13133890] [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: 05/31/2024] [Revised: 06/25/2024] [Accepted: 06/27/2024] [Indexed: 07/14/2024] Open
Abstract
Background: Disease-modifying antirheumatic drugs (bDMARDs) have shown efficacy in treating Rheumatoid Arthritis (RA). Predicting treatment outcomes for RA is crucial as approximately 30% of patients do not respond to bDMARDs and only half achieve a sustained response. This study aims to leverage machine learning to predict both initial response at 6 months and sustained response at 12 months using baseline clinical data. Methods: Baseline clinical data were collected from 154 RA patients treated at the University Hospital in Erlangen, Germany. Five machine learning models were compared: Extreme Gradient Boosting (XGBoost), Adaptive Boosting (AdaBoost), K-nearest neighbors (KNN), Support Vector Machines (SVM), and Random Forest. Nested cross-validation was employed to ensure robustness and avoid overfitting, integrating hyperparameter tuning within its process. Results: XGBoost achieved the highest accuracy for predicting initial response (AUC-ROC of 0.91), while AdaBoost was the most effective for sustained response (AUC-ROC of 0.84). Key predictors included the Disease Activity Score-28 using erythrocyte sedimentation rate (DAS28-ESR), with higher scores at baseline associated with lower response chances at 6 and 12 months. Shapley additive explanations (SHAP) identified the most important baseline features and visualized their directional effects on treatment response and sustained response. Conclusions: These findings can enhance RA treatment plans and support clinical decision-making, ultimately improving patient outcomes by predicting response before starting medication.
Collapse
Affiliation(s)
- Fatemeh Salehi
- Machine Learning and Data Analytics Laboratory, Department Artificial Intelligence in Biomedical Engineering, Friedrich Alexander University Erlangen-Nuremberg, 91052 Erlangen, Germany; (D.Z.); (B.M.E.)
| | - Luis I. Lopera Gonzalez
- Instutue of Digital Health, Friedrich Alexander University Erlangen-Nuremberg, 91052 Erlangen, Germany;
| | - Sara Bayat
- Department of Internal Medicine 3, Rheumatology and Immunology, University Hospital Erlangen, 91054 Erlangen, Germany; (S.B.); (G.S.)
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
| | - Arnd Kleyer
- Department of Rheumatology and Clinical Immunology, Charité—University Medicine Berlin, 10117 Berlin, Germany;
| | - Dario Zanca
- Machine Learning and Data Analytics Laboratory, Department Artificial Intelligence in Biomedical Engineering, Friedrich Alexander University Erlangen-Nuremberg, 91052 Erlangen, Germany; (D.Z.); (B.M.E.)
| | | | - Georg Schett
- Department of Internal Medicine 3, Rheumatology and Immunology, University Hospital Erlangen, 91054 Erlangen, Germany; (S.B.); (G.S.)
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
| | - Bjoern M. Eskofier
- Machine Learning and Data Analytics Laboratory, Department Artificial Intelligence in Biomedical Engineering, Friedrich Alexander University Erlangen-Nuremberg, 91052 Erlangen, Germany; (D.Z.); (B.M.E.)
- Translational Digital Health Group, Institute of AI for Health, Helmholtz Center Munich—German Research Center for Environmental Health, 85764 Neuherberg, Germany
| |
Collapse
|
11
|
Shi Y, Zhou M, Chang C, Jiang P, Wei K, Zhao J, Shan Y, Zheng Y, Zhao F, Lv X, Guo S, Wang F, He D. Advancing precision rheumatology: applications of machine learning for rheumatoid arthritis management. Front Immunol 2024; 15:1409555. [PMID: 38915408 PMCID: PMC11194317 DOI: 10.3389/fimmu.2024.1409555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 05/24/2024] [Indexed: 06/26/2024] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease causing progressive joint damage. Early diagnosis and treatment is critical, but remains challenging due to RA complexity and heterogeneity. Machine learning (ML) techniques may enhance RA management by identifying patterns within multidimensional biomedical data to improve classification, diagnosis, and treatment predictions. In this review, we summarize the applications of ML for RA management. Emerging studies or applications have developed diagnostic and predictive models for RA that utilize a variety of data modalities, including electronic health records, imaging, and multi-omics data. High-performance supervised learning models have demonstrated an Area Under the Curve (AUC) exceeding 0.85, which is used for identifying RA patients and predicting treatment responses. Unsupervised learning has revealed potential RA subtypes. Ongoing research is integrating multimodal data with deep learning to further improve performance. However, key challenges remain regarding model overfitting, generalizability, validation in clinical settings, and interpretability. Small sample sizes and lack of diverse population testing risks overestimating model performance. Prospective studies evaluating real-world clinical utility are lacking. Enhancing model interpretability is critical for clinician acceptance. In summary, while ML shows promise for transforming RA management through earlier diagnosis and optimized treatment, larger scale multisite data, prospective clinical validation of interpretable models, and testing across diverse populations is still needed. As these gaps are addressed, ML may pave the way towards precision medicine in RA.
Collapse
Affiliation(s)
- Yiming Shi
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Mi Zhou
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Cen Chang
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Ping Jiang
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Kai Wei
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Jianan Zhao
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Yu Shan
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Yixin Zheng
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Fuyu Zhao
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Xinliang Lv
- Traditional Chinese Medicine Hospital of Inner Mongolia Autonomous Region, Hohhot, Inner Mongolia Autonomous Region, China
| | - Shicheng Guo
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fubo Wang
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Department of Urology, Affiliated Tumor Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
| | - Dongyi He
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
12
|
Sharma SD, Bluett J. Towards Personalized Medicine in Rheumatoid Arthritis. Open Access Rheumatol 2024; 16:89-114. [PMID: 38779469 PMCID: PMC11110814 DOI: 10.2147/oarrr.s372610] [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: 01/04/2024] [Accepted: 05/03/2024] [Indexed: 05/25/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic, incurable, multisystem, inflammatory disease characterized by synovitis and extra-articular features. Although several advanced therapies targeting inflammatory mechanisms underlying the disease are available, no advanced therapy is universally effective. Therefore, a ceiling of treatment response is currently accepted where no advanced therapy is superior to another. The current challenge for medical research is the discovery and integration of predictive markers of drug response that can be used to personalize medicine so that the patient is started on "the right drug at the right time". This review article summarizes our current understanding of predicting response to anti-rheumatic drugs in RA, obstacles impeding the development of personalized medicine approaches and future research priorities to overcome these barriers.
Collapse
Affiliation(s)
- Seema D Sharma
- Centre for Musculoskeletal Research, Division of Musculoskeletal & Dermatological Sciences, School of Biological Sciences, University of Manchester, Manchester, UK
| | - James Bluett
- Centre for Musculoskeletal Research, Division of Musculoskeletal & Dermatological Sciences, School of Biological Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
13
|
Ukalovic D, Leeb BF, Rintelen B, Eichbauer-Sturm G, Spellitz P, Puchner R, Herold M, Stetter M, Ferincz V, Resch-Passini J, Zwerina J, Zimmermann-Rittereiser M, Fritsch-Stork R. Prediction of ineffectiveness of biological drugs using machine learning and explainable AI methods: data from the Austrian Biological Registry BioReg. Arthritis Res Ther 2024; 26:44. [PMID: 38331930 PMCID: PMC10851538 DOI: 10.1186/s13075-024-03277-x] [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: 10/20/2023] [Accepted: 01/25/2024] [Indexed: 02/10/2024] Open
Abstract
OBJECTIVES Machine learning models can support an individualized approach in the choice of bDMARDs. We developed prediction models for 5 different bDMARDs using machine learning methods based on patient data derived from the Austrian Biologics Registry (BioReg). METHODS Data from 1397 patients and 19 variables with at least 100 treat-to-target (t2t) courses per drug were derived from the BioReg biologics registry. Different machine learning algorithms were trained to predict the risk of ineffectiveness for each bDMARD within the first 26 weeks. Cross-validation and hyperparameter optimization were applied to generate the best models. Model quality was assessed by area under the receiver operating characteristic (AUROC). Using explainable AI (XAI), risk-reducing and risk-increasing factors were extracted. RESULTS The best models per drug achieved an AUROC score of the following: abatacept, 0.66 (95% CI, 0.54-0.78); adalimumab, 0.70 (95% CI, 0.68-0.74); certolizumab, 0.84 (95% CI, 0.79-0.89); etanercept, 0.68 (95% CI, 0.55-0.87); tocilizumab, 0.72 (95% CI, 0.69-0.77). The most risk-increasing variables were visual analytic scores (VAS) for abatacept and etanercept and co-therapy with glucocorticoids for adalimumab. Dosage was the most important variable for certolizumab and associated with a lower risk of non-response. Some variables, such as gender and rheumatoid factor (RF), showed opposite impacts depending on the bDMARD. CONCLUSION Ineffectiveness of biological drugs could be predicted with promising accuracy. Interestingly, individual parameters were found to be associated with drug responses in different directions, indicating highly complex interactions. Machine learning can be of help in the decision-process by disentangling these relations.
Collapse
Affiliation(s)
| | - Burkhard F Leeb
- Rheumatological Practice, Private Office, Hollabrunn, Austria
| | - Bernhard Rintelen
- Lower Austrian State Hospital Stockerau, 2nd Department of Medicine, Lower Austrian Competence Center for Rheumatology, Karl Landsteiner Institute for Clinical Rheumatology, Stockerau, Austria
| | | | - Peter Spellitz
- Rheuma-Center Wien-Oberlaa, Department of Rheumatology, Vienna, Austria
| | | | - Manfred Herold
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Miriam Stetter
- Rheumatological Practice, Private Office, Amstetten, Austria
| | - Vera Ferincz
- Department of Internal Medicine, University Hospital St. Pölten, St. Pölten, Austria
| | | | - Jochen Zwerina
- Hanusch Krankenhaus, Vienna, Austria
- Ludwig Boltzmann Institute of Osteology, Vienna, Austria
| | | | - Ruth Fritsch-Stork
- Health Care Center Mariahilf of ÖGK, Vienna, Austria
- Biologica Registry BioReg, Stockerau, Austria
- Medical Faculty, Sigmund Freud Private University Vienna, Vienna, Austria
| |
Collapse
|
14
|
Danieli MG, Brunetto S, Gammeri L, Palmeri D, Claudi I, Shoenfeld Y, Gangemi S. Machine learning application in autoimmune diseases: State of art and future prospectives. Autoimmun Rev 2024; 23:103496. [PMID: 38081493 DOI: 10.1016/j.autrev.2023.103496] [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: 11/12/2023] [Accepted: 11/29/2023] [Indexed: 04/30/2024]
Abstract
Autoimmune diseases are a group of disorders resulting from an alteration of immune tolerance, characterized by the formation of autoantibodies and the consequent development of heterogeneous clinical manifestations. Diagnosing autoimmune diseases is often complicated, and the available prognostic tools are limited. Machine learning allows us to analyze large amounts of data and carry out complex calculations quickly and with minimal effort. In this work, we examine the literature focusing on the use of machine learning in the field of the main systemic (systemic lupus erythematosus and rheumatoid arthritis) and organ-specific autoimmune diseases (type 1 diabetes mellitus, autoimmune thyroid, gastrointestinal, and skin diseases). From our analysis, interesting applications of machine learning emerged for developing algorithms useful in the early diagnosis of disease or prognostic models (risk of complications, therapeutic response). Subsequent studies and the creation of increasingly rich databases to be supplied to the algorithms will eventually guide the clinician in the diagnosis, allowing intervention when the pathology is still in an early stage and immediately directing towards a correct therapeutic approach.
Collapse
Affiliation(s)
- Maria Giovanna Danieli
- SOS Immunologia delle Malattie Rare e dei Trapianti. AOU delle Marche & Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, via Tronto 10/A, 60126 Torrette di Ancona, Italy; Postgraduate School of Allergy and Clinical Immunology, Università Politecnica delle Marche, via Tronto 10/A, 60126 Ancona, Italy.
| | - Silvia Brunetto
- Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy
| | - Luca Gammeri
- Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy
| | - Davide Palmeri
- Postgraduate School of Allergy and Clinical Immunology, Università Politecnica delle Marche, via Tronto 10/A, 60126 Ancona, Italy
| | - Ilaria Claudi
- Postgraduate School of Allergy and Clinical Immunology, Università Politecnica delle Marche, via Tronto 10/A, 60126 Ancona, Italy
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, and Reichman University Herzliya, Israel.
| | - Sebastiano Gangemi
- Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy.
| |
Collapse
|
15
|
Valdivieso Shephard JL, Alvarez Robles EJ, Cámara Hijón C, Hernandez Breijo B, Novella-Navarro M, Bogas Schay P, Cuesta de la Cámara R, Balsa Criado A, López Granados E, Plasencia Rodríguez C. Predicting anti-TNF treatment response in rheumatoid arthritis: An artificial intelligence-driven model using cytokine profile and routine clinical practice parameters. Heliyon 2024; 10:e22925. [PMID: 38163219 PMCID: PMC10754867 DOI: 10.1016/j.heliyon.2023.e22925] [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: 06/27/2023] [Revised: 11/18/2023] [Accepted: 11/22/2023] [Indexed: 01/03/2024] Open
Abstract
Introduction Rheumatoid arthritis (RA) is a heterogeneous disease in which therapeutic strategies used have evolved dramatically. Despite significant progress in treatment strategies such as the development of anti-TNF drugs, it is still not possible to differentiate those patients who will respond from who will not. This can lead to effective-treatment delays and unnecessary costs. The aim of this study was to utilize a profile of the patient's characteristics, clinical parameters, immune status (cytokine profile) and artificial intelligence to assess the feasibility of developing a tool that could allow us to predict which patients will respond to treatment with anti-TNF drugs. Methods This study included 38 patients with RA from the RA-Paz cohort. Clinical activity was measured at baseline and after 6 months of treatment. The cytokines measured before the start of anti-TNF treatment were IL-1, IL-12, IL-10, IL-2, IL-4, IFNg, TNFa, and IL-6. Statistical analyses were performed using the Wilcoxon-Rank-Sum Test and the Benjamini-Hochberg method. The predictive model viability was explored using the 5-fold cross-validation scheme in order to train the logistic regression models. Results Statistically significant differences were found in parameters such as IL-6, IL-2, CRP and DAS-ESR. The predictive model performed to an acceptable level in correctly classifying patients (ROC-AUC 0.804167 to 0.891667), suggesting that it would be possible to develop a clinical classification tool. Conclusions Using a combination of parameters such as IL-6, IL-2, CRP and DAS-ESR, it was possible to develop a predictive model that can acceptably discriminate between remitters and non-remitters. However, this model needs to be replicated in a larger cohort to confirm these findings.
Collapse
Affiliation(s)
| | | | | | - Borja Hernandez Breijo
- Immunology-Rheumatology Research Group, Hospital Universitario La Paz-Idipaz, Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
16
|
Gilvaz VJ, Reginato AM. Artificial intelligence in rheumatoid arthritis: potential applications and future implications. Front Med (Lausanne) 2023; 10:1280312. [PMID: 38034534 PMCID: PMC10687464 DOI: 10.3389/fmed.2023.1280312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 10/13/2023] [Indexed: 12/02/2023] Open
Abstract
The widespread adoption of digital health records, coupled with the rise of advanced diagnostic testing, has resulted in an explosion of patient data, comparable in scope to genomic datasets. This vast information repository offers significant potential for improving patient outcomes and decision-making, provided one can extract meaningful insights from it. This is where artificial intelligence (AI) tools like machine learning (ML) and deep learning come into play, helping us leverage these enormous datasets to predict outcomes and make informed decisions. AI models can be trained to analyze and interpret patient data, including physician notes, laboratory testing, and imaging, to aid in the management of patients with rheumatic diseases. As one of the most common autoimmune diseases, rheumatoid arthritis (RA) has attracted considerable attention, particularly concerning the evolution of diagnostic techniques and therapeutic interventions. Our aim is to underscore those areas where AI, according to recent research, demonstrates promising potential to enhance the management of patients with RA.
Collapse
Affiliation(s)
- Vinit J. Gilvaz
- Division of Rheumatology, Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Anthony M. Reginato
- Division of Rheumatology, Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI, United States
- Department of Dermatology, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI, United States
| |
Collapse
|
17
|
Hegarty C, Neto N, Cahill P, Floudas A. Computational approaches in rheumatic diseases - Deciphering complex spatio-temporal cell interactions. Comput Struct Biotechnol J 2023; 21:4009-4020. [PMID: 37649712 PMCID: PMC10462794 DOI: 10.1016/j.csbj.2023.08.005] [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: 04/04/2023] [Revised: 08/04/2023] [Accepted: 08/04/2023] [Indexed: 09/01/2023] Open
Abstract
Inflammatory arthritis, including rheumatoid (RA), and psoriatic (PsA) arthritis, are clinically and immunologically heterogeneous diseases with no identified cure. Chronic inflammation of the synovial tissue ushers loss of function of the joint that severely impacts the patient's quality of life, eventually leading to disability and life-threatening comorbidities. The pathogenesis of synovial inflammation is the consequence of compounded immune and stromal cell interactions influenced by genetic and environmental factors. Deciphering the complexity of the synovial cellular landscape has accelerated primarily due to the utilisation of bulk and single cell RNA sequencing. Particularly the capacity to generate cell-cell interaction networks could reveal evidence of previously unappreciated processes leading to disease. However, there is currently a lack of universal nomenclature as a result of varied experimental and technological approaches that discombobulates the study of synovial inflammation. While spatial transcriptomic analysis that combines anatomical information with transcriptomic data of synovial tissue biopsies promises to provide more insights into disease pathogenesis, in vitro functional assays with single-cell resolution will be required to validate current bioinformatic applications. In order to provide a comprehensive approach and translate experimental data to clinical practice, a combination of clinical and molecular data with machine learning has the potential to enhance patient stratification and identify individuals at risk of arthritis that would benefit from early therapeutic intervention. This review aims to provide a comprehensive understanding of the effect of computational approaches in deciphering synovial inflammation pathogenesis and discuss the impact that further experimental and novel computational tools may have on therapeutic target identification and drug development.
Collapse
Affiliation(s)
- Ciara Hegarty
- Translational Immunology lab, School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Nuno Neto
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Ireland
| | - Paul Cahill
- Vascular Biology lab, School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Achilleas Floudas
- Translational Immunology lab, School of Biotechnology, Dublin City University, Dublin, Ireland
| |
Collapse
|
18
|
Curtis JR, Yun H, Chen L, Ford SS, van Hoogstraten H, Fiore S, Ford K, Praestgaard A, Rehberg M, Choy E. Real-World Sarilumab Use and Rule Testing to Predict Treatment Response in Patients with Rheumatoid Arthritis: Findings from the RISE Registry. Rheumatol Ther 2023; 10:1055-1072. [PMID: 37349636 PMCID: PMC10326227 DOI: 10.1007/s40744-023-00568-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 05/30/2023] [Indexed: 06/24/2023] Open
Abstract
INTRODUCTION Clinical trial findings may not be generalizable to routine practice. This study evaluated sarilumab effectiveness in patients with rheumatoid arthritis (RA) and tested the real-world applicability of a response prediction rule, derived from trial data using machine learning (based on C-reactive protein [CRP] > 12.3 mg/l and seropositivity [anticyclic citrullinated peptide antibodies, ACPA +]). METHODS Sarilumab initiators from the ACR-RISE Registry, with ≥ 1 prescription on/after its FDA approval (2017-2020), were divided into three cohorts based on progressively restrictive criteria: Cohort A (had active disease), Cohort B (met eligibility criteria of a phase 3 trial in RA patients with inadequate response/intolerance to tumor necrosis factor inhibitors [TNFi]), and Cohort C (characteristics matched to the phase 3 trial baseline). Mean changes in Clinical Disease Activity Index (CDAI) and Routine Assessment of Patient Index Data 3 (RAPID3) were evaluated at 6 and 12 months. In a separate cohort, predictive rule was tested based on CRP levels and seropositive status (ACPA and/or rheumatoid factor); patients were categorized into rule-positive (seropositive with CRP > 12.3 mg/l) and rule-negative groups to compare the odds of achieving CDAI low disease activity (LDA)/remission and minimal clinically important difference (MCID) over 24 weeks. RESULTS Among sarilumab initiators (N = 2949), treatment effectiveness was noted across cohorts, with greater improvement noted for Cohort C at 6 and 12 months. Among the predictive rule cohort (N = 205), rule-positive (vs. rule-negative) patients were more likely to reach LDA (odds ratio: 1.5 [0.7, 3.2]) and MCID (1.1 [0.5, 2.4]). Sensitivity analyses (CRP > 5 mg/l) showed better response to sarilumab in rule-positive patients. CONCLUSIONS In real-world setting, sarilumab demonstrated treatment effectiveness, with greater improvements in the most selective population, mirroring phase 3 TNFi-refractory and rule-positive RA patients. Seropositivity appeared a stronger driver for treatment response than CRP, although optimization of the rule in routine practice requires further data.
Collapse
Affiliation(s)
- Jeffrey R Curtis
- University of Alabama at Birmingham, Birmingham, AL, 35233, USA.
| | - Huifeng Yun
- University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Lang Chen
- University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | | | | | | | | | | | | | - Ernest Choy
- CREATE Centre, Cardiff University, Cardiff, UK
| |
Collapse
|
19
|
Wang J, Tian Y, Zhou T, Tong D, Ma J, Li J. A survey of artificial intelligence in rheumatoid arthritis. RHEUMATOLOGY AND IMMUNOLOGY RESEARCH 2023; 4:69-77. [PMID: 37485476 PMCID: PMC10362600 DOI: 10.2478/rir-2023-0011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/14/2023] [Indexed: 07/25/2023]
Abstract
The article offers a survey of currently notable artificial intelligence methods (released between 2019-2023), with a particular emphasis on the latest advancements in detecting rheumatoid arthritis (RA) at an early stage, providing early treatment, and managing the disease. We discussed challenges in these areas followed by specific artificial intelligence (AI) techniques and summarized advances, relevant strengths, and obstacles. Overall, the application of AI in the fields of RA has the potential to enable healthcare professionals to detect RA at an earlier stage, thereby facilitating timely intervention and better disease management. However, more research is required to confirm the precision and dependability of AI in RA, and several problems such as technological and ethical concerns related to these approaches must be resolved before their widespread adoption.
Collapse
Affiliation(s)
- Jiaqi Wang
- Research Center for Healthcare Data Science, Zhejiang Laboratory, Hangzhou311121, Zhejiang Province, China
| | - Yu Tian
- Engineering Research Center of EMR and Intelligent Expert System, Ministry of Education, Key Laboratory for Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou310027, Zhejiang Province, China
| | - Tianshu Zhou
- Research Center for Healthcare Data Science, Zhejiang Laboratory, Hangzhou311121, Zhejiang Province, China
| | - Danyang Tong
- Research Center for Healthcare Data Science, Zhejiang Laboratory, Hangzhou311121, Zhejiang Province, China
| | - Jing Ma
- Research Center for Healthcare Data Science, Zhejiang Laboratory, Hangzhou311121, Zhejiang Province, China
| | - Jingsong Li
- Research Center for Healthcare Data Science, Zhejiang Laboratory, Hangzhou311121, Zhejiang Province, China
- Engineering Research Center of EMR and Intelligent Expert System, Ministry of Education, Key Laboratory for Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou310027, Zhejiang Province, China
| |
Collapse
|
20
|
Efficacy of a machine learning-based approach in predicting neurological prognosis of cervical spinal cord injury patients following urgent surgery within 24 h after injury. J Clin Neurosci 2023; 107:150-156. [PMID: 36376152 DOI: 10.1016/j.jocn.2022.11.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 10/12/2022] [Accepted: 11/05/2022] [Indexed: 11/13/2022]
Abstract
We aimed to develop a machine learning (ML) model for predicting the neurological outcomes of cervical spinal cord injury (CSCI). We retrospectively analyzed 135 patients with CSCI who underwent surgery within 24 h after injury. Patients were assessed with the American Spinal Injury Association Impairment Scale (AIS; grades A to E) 6 months after injury. A total of 34 features extracted from demographic variables, surgical factors, laboratory variables, neurological status, and radiological findings were analyzed. The ML model was created using Light GBM, XGBoost, and CatBoost. We evaluated Shapley Additive Explanations (SHAP) values to determine the variables that contributed most to the prediction models. We constructed multiclass prediction models for the five AIS grades and binary classification models to predict more than one-grade improvement in AIS 6 months after injury. Of the ML models used, CatBoost showed the highest accuracy (0.800) for the prediction of AIS grade and the highest AUC (0.90) for predicting improvement in AIS. AIS grade at admission, intramedullary hemorrhage, longitudinal extent of intramedullary T2 hyperintensity, and HbA1c were identified as important features for these prediction models. The ML models successfully predicted neurological outcomes 6 months after injury following urgent surgery in patients with CSCI.
Collapse
|
21
|
Loh HW, Ooi CP, Seoni S, Barua PD, Molinari F, Acharya UR. Application of explainable artificial intelligence for healthcare: A systematic review of the last decade (2011-2022). COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2022; 226:107161. [PMID: 36228495 DOI: 10.1016/j.cmpb.2022.107161] [Citation(s) in RCA: 155] [Impact Index Per Article: 51.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/16/2022] [Accepted: 09/25/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND AND OBJECTIVES Artificial intelligence (AI) has branched out to various applications in healthcare, such as health services management, predictive medicine, clinical decision-making, and patient data and diagnostics. Although AI models have achieved human-like performance, their use is still limited because they are seen as a black box. This lack of trust remains the main reason for their low use in practice, especially in healthcare. Hence, explainable artificial intelligence (XAI) has been introduced as a technique that can provide confidence in the model's prediction by explaining how the prediction is derived, thereby encouraging the use of AI systems in healthcare. The primary goal of this review is to provide areas of healthcare that require more attention from the XAI research community. METHODS Multiple journal databases were thoroughly searched using PRISMA guidelines 2020. Studies that do not appear in Q1 journals, which are highly credible, were excluded. RESULTS In this review, we surveyed 99 Q1 articles covering the following XAI techniques: SHAP, LIME, GradCAM, LRP, Fuzzy classifier, EBM, CBR, rule-based systems, and others. CONCLUSION We discovered that detecting abnormalities in 1D biosignals and identifying key text in clinical notes are areas that require more attention from the XAI research community. We hope this is review will encourage the development of a holistic cloud system for a smart city.
Collapse
Affiliation(s)
- Hui Wen Loh
- School of Science and Technology, Singapore University of Social Sciences, Singapore
| | - Chui Ping Ooi
- School of Science and Technology, Singapore University of Social Sciences, Singapore
| | - Silvia Seoni
- Department of Electronics and Telecommunications, Biolab, Politecnico di Torino, Torino 10129, Italy
| | - Prabal Datta Barua
- Faculty of Engineering and Information Technology, University of Technology Sydney, Australia; School of Business (Information Systems), Faculty of Business, Education, Law & Arts, University of Southern Queensland, Australia
| | - Filippo Molinari
- Department of Electronics and Telecommunications, Biolab, Politecnico di Torino, Torino 10129, Italy
| | - U Rajendra Acharya
- School of Science and Technology, Singapore University of Social Sciences, Singapore; School of Business (Information Systems), Faculty of Business, Education, Law & Arts, University of Southern Queensland, Australia; School of Engineering, Ngee Ann Polytechnic, Singapore; Department of Bioinformatics and Medical Engineering, Asia University, Taiwan; Research Organization for Advanced Science and Technology (IROAST), Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
22
|
Koo BS, Eun S, Shin K, Hong S, Kim YG, Lee CK, Yoo B, Oh JS. Differences in trajectory of disease activity according to biologic and targeted synthetic disease-modifying anti-rheumatic drug treatment in patients with rheumatoid arthritis. Arthritis Res Ther 2022; 24:233. [PMID: 36242075 PMCID: PMC9563490 DOI: 10.1186/s13075-022-02918-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 10/05/2022] [Indexed: 11/18/2022] Open
Abstract
Background
The purpose of this study was to stratify patients with rheumatoid arthritis (RA) according to the trend of disease activity by trajectory-based clustering and to identify contributing factors for treatment response to biologic and targeted synthetic disease-modifying anti-rheumatic drugs (DMARDs) according to trajectory groups. Methods We analyzed the data from a nationwide RA cohort from the Korean College of Rheumatology Biologics and Targeted Therapy registry. Patients treated with second-line biologic and targeted synthetic DMARDs were included. Trajectory modeling for clustering was used to group the disease activity trend. The contributing factors using the machine learning model of SHAP (SHapley Additive exPlanations) values for each trajectory were investigated. Results The trends in the disease activity of 688 RA patients were clustered into 4 groups: rapid decrease and stable disease activity (group 1, n = 319), rapid decrease followed by an increase (group 2, n = 36), slow and continued decrease (group 3, n = 290), and no decrease in disease activity (group 4, n = 43). SHAP plots indicated that the most important features of group 2 compared to group 1 were the baseline erythrocyte sedimentation rate (ESR), prednisolone dose, and disease activity score with 28-joint assessment (DAS28) (SHAP value 0.308, 0.157, and 0.103, respectively). The most important features of group 3 compared to group 1 were the baseline ESR, DAS28, and estimated glomerular filtration rate (eGFR) (SHAP value 0.175, 0.164, 0.042, respectively). The most important features of group 4 compared to group 1 were the baseline DAS28, ESR, and blood urea nitrogen (BUN) (SHAP value 0.387, 0.153, 0.144, respectively). Conclusions The trajectory-based approach was useful for clustering the treatment response of biologic and targeted synthetic DMARDs in patients with RA. In addition, baseline DAS28, ESR, prednisolone dose, eGFR, and BUN were important contributing factors for 4-year trajectories.
Collapse
Affiliation(s)
- Bon San Koo
- Department of Internal Medicine, Inje University Seoul Paik Hospital, Inje University College of Medicine, Seoul, South Korea
| | - Seongho Eun
- Department of Management Engineering, College of Business, KAIST, Seoul, South Korea
| | - Kichul Shin
- Division of Rheumatology, Seoul Metropolitan Government-Seoul National University Hospital Boramae Medical Center, Seoul, South Korea
| | - Seokchan Hong
- Division of Rheumatology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Yong-Gil Kim
- Division of Rheumatology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Chang-Keun Lee
- Division of Rheumatology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Bin Yoo
- Division of Rheumatology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Ji Seon Oh
- Department of Information Medicine, Big Data Research Center, Asan Medical Center, Seoul, South Korea.
| |
Collapse
|
23
|
Momtazmanesh S, Nowroozi A, Rezaei N. Artificial Intelligence in Rheumatoid Arthritis: Current Status and Future Perspectives: A State-of-the-Art Review. Rheumatol Ther 2022; 9:1249-1304. [PMID: 35849321 PMCID: PMC9510088 DOI: 10.1007/s40744-022-00475-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 06/24/2022] [Indexed: 11/23/2022] Open
Abstract
Investigation of the potential applications of artificial intelligence (AI), including machine learning (ML) and deep learning (DL) techniques, is an exponentially growing field in medicine and healthcare. These methods can be critical in providing high-quality care to patients with chronic rheumatological diseases lacking an optimal treatment, like rheumatoid arthritis (RA), which is the second most prevalent autoimmune disease. Herein, following reviewing the basic concepts of AI, we summarize the advances in its applications in RA clinical practice and research. We provide directions for future investigations in this field after reviewing the current knowledge gaps and technical and ethical challenges in applying AI. Automated models have been largely used to improve RA diagnosis since the early 2000s, and they have used a wide variety of techniques, e.g., support vector machine, random forest, and artificial neural networks. AI algorithms can facilitate screening and identification of susceptible groups, diagnosis using omics, imaging, clinical, and sensor data, patient detection within electronic health record (EHR), i.e., phenotyping, treatment response assessment, monitoring disease course, determining prognosis, novel drug discovery, and enhancing basic science research. They can also aid in risk assessment for incidence of comorbidities, e.g., cardiovascular diseases, in patients with RA. However, the proposed models may vary significantly in their performance and reliability. Despite the promising results achieved by AI models in enhancing early diagnosis and management of patients with RA, they are not fully ready to be incorporated into clinical practice. Future investigations are required to ensure development of reliable and generalizable algorithms while they carefully look for any potential source of bias or misconduct. We showed that a growing body of evidence supports the potential role of AI in revolutionizing screening, diagnosis, and management of patients with RA. However, multiple obstacles hinder clinical applications of AI models. Incorporating the machine and/or deep learning algorithms into real-world settings would be a key step in the progress of AI in medicine.
Collapse
Affiliation(s)
- Sara Momtazmanesh
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Dr. Gharib St, Keshavarz Blvd, Tehran, Iran
| | - Ali Nowroozi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Dr. Gharib St, Keshavarz Blvd, Tehran, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
24
|
Taylor PC, Matucci Cerinic M, Alten R, Avouac J, Westhovens R. Managing inadequate response to initial anti-TNF therapy in rheumatoid arthritis: optimising treatment outcomes. Ther Adv Musculoskelet Dis 2022; 14:1759720X221114101. [PMID: 35991524 PMCID: PMC9386864 DOI: 10.1177/1759720x221114101] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 06/29/2022] [Indexed: 11/19/2022] Open
Abstract
Anti-tumour necrosis factors (anti-TNFs) are established as first-line biological therapy for rheumatoid arthritis (RA) with over two decades of accumulated clinical experience. Anti-TNFs have well established efficacy/safety profiles along with additional benefits on various comorbidities. However, up to 40% of patients may respond inadequately to an initial anti-TNF treatment because of primary non-response, loss of response, or intolerance. Following inadequate response (IR) to anti-TNF treatment, clinicians can consider switching to an alternative anti-TNF (cycling) or to another class of targeted drug with a different mechanism of action, such as Janus kinase inhibitors, interleukin-6 receptor blockers, B-cell depletion agents, and co-stimulation inhibitors (swapping). While European League Against Rheumatism recommendations for pharmacotherapeutic management of RA, published in 2020, are widely regarded as helpful guides to clinical practice, they do not provide any clear recommendations on therapeutic choices following an IR to first-line anti-TNF. This suggests that both cycling and swapping treatment strategies are of equal value, but that the treating physician must take the patient’s individual characteristics into account. This article considers which patient characteristics influence clinical decision-making processes, including the reason for treatment failure, previous therapies, comorbidities, extra-articular manifestations, pregnancy, patient preference and cost-effectiveness, and what evidence is available to support decisions made by the physician.
Collapse
Affiliation(s)
- Peter C Taylor
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Old Rd, Headington, Oxford OX3 7LD, UK
| | - Marco Matucci Cerinic
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Rieke Alten
- Department of Internal Medicine, Rheumatology, Clinical Immunology and Osteology, Schlosspark-Klinik University Medicine Berlin, Berlin, Germany
| | - Jérôme Avouac
- AP-HP Centre, Université de Paris, Hôpital Cochin, Service de Rhumatologie, Paris, France
| | - Rene Westhovens
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration and Division of Rheumatology, KU Leuven, Leuven, Belgium
| |
Collapse
|
25
|
Mahroum N, Elsalti A, Alwani A, Seida I, Alrais M, Seida R, Esirgun SN, Abali T, Kiyak Z, Zoubi M, Shoenfeld Y. The mosaic of autoimmunity - Finally discussing in person. The 13 th international congress on autoimmunity 2022 (AUTO13) Athens. Autoimmun Rev 2022; 21:103166. [PMID: 35932955 PMCID: PMC9349027 DOI: 10.1016/j.autrev.2022.103166] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 07/31/2022] [Indexed: 11/29/2022]
Abstract
While autoimmunity is a branch of medicine linked to every single organ system via direct and indirect pathways, meeting in person to discuss autoimmunity during the 13th international congress on autoimmunity (AUTO13) with participants from all over the world had a very good reason. The mechanisms involved in autoimmune diseases are of extreme importance and in fact critical in understanding the course of diseases as well as selecting proper therapies. COVID-19 has served as a great example of how autoimmunity is deeply involved in the disease and directly correlated to severity, morbidity, and mortality. For instance, initially the term cytokine storm dominated, then COVID-19 was addressed as the new member of the hyperferritinemic syndrome, and also the use of immunosuppressants in patients with COVID-19 throughout the pandemic, all shed light on the fundamental role of autoimmunity. Unsurprisingly, SARS-CoV-2 was called the “autoimmune virus” during AUTO13. Subsequently, the correlation between autoimmunity and COVID-19 vaccines and post-COVID, all were discussed from different autoimmune aspects during the congress. In addition, updates on the mechanisms of diseases, autoantibodies, novel diagnostics and therapies in regard to autoimmune diseases such as antiphospholipid syndrome, systemic lupus erythematosus, systemic sclerosis and others, were discussed in dedicated sessions. Due to the magnificence of the topics discussed, we aimed to bring in our article hereby, the pearls of AUTO13 in terms of updates, new aspects of autoimmunity, and interesting findings. While more than 500 abstract were presented, concluding all the topics was not in reach, hence major findings were summarized.
Collapse
Affiliation(s)
- Naim Mahroum
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey.
| | - Abdulrahman Elsalti
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Abdulkarim Alwani
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Isa Seida
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Mahmoud Alrais
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Ravend Seida
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Sevval Nil Esirgun
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Tunahan Abali
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Zeynep Kiyak
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Majdi Zoubi
- Department of Internal Medicine B, HaEmek Medical Center, Afula, Israel, Affiliated to Technion, Faculty of Medicine, Haifa, Israel
| | | |
Collapse
|
26
|
Machine learning-based prediction of relapse in rheumatoid arthritis patients using data on ultrasound examination and blood test. Sci Rep 2022; 12:7224. [PMID: 35508670 PMCID: PMC9068780 DOI: 10.1038/s41598-022-11361-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 04/22/2022] [Indexed: 11/08/2022] Open
Abstract
Recent effective therapies enable most rheumatoid arthritis (RA) patients to achieve remission; however, some patients experience relapse. We aimed to predict relapse in RA patients through machine learning (ML) using data on ultrasound (US) examination and blood test. Overall, 210 patients with RA in remission at baseline were dichotomized into remission (n = 150) and relapse (n = 60) based on the disease activity at 2-year follow-up. Three ML classifiers [Logistic Regression, Random Forest, and extreme gradient boosting (XGBoost)] and data on 73 features (14 US examination data, 54 blood test data, and five data on patient information) at baseline were used for predicting relapse. The best performance was obtained using the XGBoost classifier (area under the receiver operator characteristic curve (AUC) = 0.747), compared with Random Forest and Logistic Regression (AUC = 0.719 and 0.701, respectively). In the XGBoost classifier prediction, ten important features, including wrist/metatarsophalangeal superb microvascular imaging scores, were selected using the recursive feature elimination method. The performance was superior to that predicted by researcher-selected features, which are conventional prognostic markers. These results suggest that ML can provide an accurate prediction of relapse in RA patients, and the use of predictive algorithms may facilitate personalized treatment options.
Collapse
|
27
|
White IR, Kleinstein SE, Praet C, Chamberlain C, McHale D, Maia JM, Xie P, Goldstein DB, Urban TJ, Shea PR. A genome-wide screen for variants influencing certolizumab pegol response in a moderate to severe rheumatoid arthritis population. PLoS One 2022; 17:e0261165. [PMID: 35413058 PMCID: PMC9004786 DOI: 10.1371/journal.pone.0261165] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 11/24/2021] [Indexed: 12/14/2022] Open
Abstract
Certolizumab pegol (CZP) is a PEGylated Fc-free tumor necrosis factor (TNF) inhibitor antibody approved for use in the treatment of rheumatoid arthritis (RA), Crohn’s disease, psoriatic arthritis, axial spondyloarthritis and psoriasis. In a clinical trial of patients with severe RA, CZP improved disease symptoms in approximately half of patients. However, variability in CZP efficacy remains a problem for clinicians, thus, the aim of this study was to identify genetic variants predictive of CZP response. We performed a genome-wide association study (GWAS) of 302 RA patients treated with CZP in the REALISTIC trial to identify common single nucleotide polymorphisms (SNPs) associated with treatment response. Whole-exome sequencing was also performed for 74 CZP extreme responders and non-responders within the same population, as well as 1546 population controls. No common SNPs or rare functional variants were significantly associated with CZP response, though a non-significant enrichment in the RA-implicated KCNK5 gene was observed. Two SNPs near spondin-1 and semaphorin-4G approached genome-wide significance. The results of the current study did not provide an unambiguous predictor of CZP response.
Collapse
Affiliation(s)
- Ian R. White
- Experimental Medicine and Diagnostics, UCB Celltech, Slough, United Kingdom
| | - Sarah E. Kleinstein
- Institute for Genomic Medicine, Columbia University, New York, New York, United States of America
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | | | - Chris Chamberlain
- Experimental Medicine and Diagnostics, UCB Celltech, Slough, United Kingdom
| | - Duncan McHale
- Experimental Medicine and Diagnostics, UCB Celltech, Slough, United Kingdom
| | - Jessica M. Maia
- Institute for Genomic Medicine, Columbia University, New York, New York, United States of America
| | - Pingxing Xie
- Institute for Genomic Medicine, Columbia University, New York, New York, United States of America
- Faculty of Medicine, McGill University, Montréal, Québec, Canada
| | - David B. Goldstein
- Institute for Genomic Medicine, Columbia University, New York, New York, United States of America
| | - Thomas J. Urban
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Patrick R. Shea
- Institute for Genomic Medicine, Columbia University, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
28
|
Kedra J, Davergne T, Braithwaite B, Servy H, Gossec L. Machine learning approaches to improve disease management of patients with rheumatoid arthritis: review and future directions. Expert Rev Clin Immunol 2021; 17:1311-1321. [PMID: 34890271 DOI: 10.1080/1744666x.2022.2017773] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Although the management of rheumatoid arthritis (RA) has improved in major way over the last decades, this disease still leads to an important burden for patients and society, and there is a need to develop more personalized approaches. Machine learning (ML) methods are more and more used in health-related studies and can be applied to different sorts of data (clinical, radiological, or 'omics' data). Such approaches may improve the management of patients with RA. AREAS COVERED In this paper, we propose a review regarding ML approaches applied to RA. A scoping literature search was performed in PubMed, in September 2021 using the following MeSH terms: 'arthritis, rheumatoid' and 'machine learning'. Based on this search, the usefulness of ML methods for RA diagnosis, monitoring, and prediction of response to treatment and RA outcomes, is discussed. EXPERT OPINION ML methods have the potential to revolutionize RA-related research and improve disease management and patient care. Nevertheless, these models are not yet ready to contribute fully to rheumatologists' daily practice. Indeed, these methods raise technical, methodological, and ethical issues, which should be addressed properly to allow their implementation. Collaboration between data scientists, clinical researchers, and physicians is therefore required to move this field forward.
Collapse
Affiliation(s)
- Joanna Kedra
- Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France.,Rheumatology Department, Pitié-Salpêtrière Hospital, AP-HP, Paris, France
| | - Thomas Davergne
- Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France
| | | | | | - Laure Gossec
- Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France.,Rheumatology Department, Pitié-Salpêtrière Hospital, AP-HP, Paris, France
| |
Collapse
|