1
|
Pasculli G, Virgolin M, Myles P, Vidovszky A, Fisher C, Biasin E, Mourby M, Pappalardo F, D'Amico S, Torchia M, Chebykin A, Carbone V, Emili L, Roeshammar D. Synthetic Data in Healthcare and Drug Development: Definitions, Regulatory Frameworks, Issues. CPT Pharmacometrics Syst Pharmacol 2025; 14:840-852. [PMID: 40193292 PMCID: PMC12072219 DOI: 10.1002/psp4.70021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/01/2025] [Accepted: 03/10/2025] [Indexed: 04/09/2025] Open
Abstract
With the recent and evolving regulatory frameworks regarding the usage of Artificial Intelligence (AI) in both drug and medical device development, the differentiation between data derived from observed ('true' or 'real') sources and artificial data obtained using process-driven and/or (data-driven) algorithmic processes is emerging as a critical consideration in clinical research and regulatory discourse. We conducted a critical literature review that revealed evidence of the current ambivalent usage of the term "synthetic" (along with derivative terms) to refer to "true/observed" data in the context of clinical trials and AI-generated data (or "artificial" data). This paper, stemming from a critical evaluation of different perspectives captured from the scientific literature and recent regulatory endeavors, seeks to elucidate this distinction, exploring their respective utilities, regulatory stances, and upcoming needs, as well as the potential for both data types in advancing medical science and therapeutic development.
Collapse
Affiliation(s)
| | - Marco Virgolin
- InSilicoTrials Technologies B.V.s‐Hertogenboschthe Netherlands
| | - Puja Myles
- Medicines and Healthcare products Regulatory AgencyLondonUK
| | | | | | | | - Miranda Mourby
- Centre for Health, Law, and Emerging Technologies (HeLEX), Faculty of LawUniversity of OxfordOxfordUK
| | | | - Saverio D'Amico
- Humanitas Clinical and Research Center‐IRCCSMilanItaly
- Train s.r.l.MilanItaly
| | | | | | | | - Luca Emili
- InSilicoTrials Technologies S.p.A.TriesteItaly
| | | |
Collapse
|
2
|
Fitzke H, Fayzan T, Watkins J, Galimov E, Pierce BF. Real-world evidence: state-of-the-art and future perspectives. J Comp Eff Res 2025; 14:e240130. [PMID: 40051332 PMCID: PMC11963347 DOI: 10.57264/cer-2024-0130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 01/31/2025] [Indexed: 03/22/2025] Open
Abstract
Recent developments in digital infrastructure, advanced analytical approaches, and regulatory settings have facilitated the broadened use of real-world evidence (RWE) in population health management and evaluation of novel health technologies. RWE has uniquely contributed to improving human health by addressing unmet clinical needs, from assessing the external validity of clinical trial data to discovery of new disease phenotypes. In this perspective, we present exemplars across various health areas that have been impacted by real-world data and RWE, and we provide insights into further opportunities afforded by RWE. By deploying robust methodologies and transparently reporting caveats and limitations, real-world data accessed via secure data environments can support proactive healthcare management and accelerate access to novel interventions in England.
Collapse
Affiliation(s)
- Heather Fitzke
- Discover-NOW, Imperial College Health Partners, London, UK
| | - Tamanah Fayzan
- Discover-NOW, Imperial College Health Partners, London, UK
| | | | - Evgeny Galimov
- Discover-NOW, Imperial College Health Partners, London, UK
| | | |
Collapse
|
3
|
Cummings JL, Zhou Y, Van Stone A, Cammann D, Tonegawa-Kuji R, Fonseca J, Cheng F. Drug repurposing for Alzheimer's disease and other neurodegenerative disorders. Nat Commun 2025; 16:1755. [PMID: 39971900 PMCID: PMC11840136 DOI: 10.1038/s41467-025-56690-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 01/24/2025] [Indexed: 02/21/2025] Open
Abstract
Repurposed drugs provide a rich source of potential therapies for Alzheimer's disease (AD) and other neurodegenerative disorders (NDD). Repurposed drugs have information from non-clinical studies, phase 1 dosing, and safety and tolerability data collected with the original indication. Computational approaches, "omic" studies, drug databases, and electronic medical records help identify candidate therapies. Generic repurposed agents lack intellectual property protection and are rarely advanced to late-stage trials for AD/NDD. In this review we define repurposing, describe the advantages and challenges of repurposing, offer strategies for overcoming the obstacles, and describe the key contributions of repurposing to the drug development ecosystem.
Collapse
Affiliation(s)
- Jeffrey L Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, Kirk Kerkorian School of Medicine, University of Nevada, Las Vegas (UNLV), Las Vegas, NV, 89106, USA.
| | - Yadi Zhou
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, 44195, USA
| | | | - Davis Cammann
- Nevada Institute of Personalized Medicine, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA
| | - Reina Tonegawa-Kuji
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, 44195, USA
| | - Jorge Fonseca
- Howard R Hughes College of Engineering, Department of Computer Science, University of Nevada, Las Vegas (UNLV), Las Vegas, NV, 89154, USA
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio, 44106, USA
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, 44195, USA
| |
Collapse
|
4
|
Choi S, Byun JM, Park SS, Han J, Oh S, Jung S, Park H, Han S, Lee JY, Koh Y, Jeon YW, Yahng SA, Shin SH, Yoon SS, Min CK. Efficacy and Safety of Bispecific T-Cell Engagers in Relapsed/Refractory Multiple Myeloma: A Real-World Data-Based Case-Controlled Study. Transplant Cell Ther 2025; 31:74.e1-74.e11. [PMID: 39608453 DOI: 10.1016/j.jtct.2024.11.010] [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: 08/29/2024] [Revised: 10/16/2024] [Accepted: 11/19/2024] [Indexed: 11/30/2024]
Abstract
Although bispecific T-cell engager (BiTE) is a promising treatment for relapsed/refractory multiple myeloma (RRMM), it needs to be evaluated in a real-world setting. This study aimed to evaluate the efficacy and safety of BiTEs compared with a synthetic standard of care (SOC). From a multicenter registry database of 474 patients with RRMM who received third- or more advanced-line treatments between January 2021 and October 2023, 1:1 propensity score-matched BiTE cohort (n = 71) and SOC cohort (n = 71) were established. Matching was based on age, sex, number of prior therapies, international staging system at diagnosis, and baseline biochemical characteristics. Compared with the matched SOC cohort, the matched BiTE cohort demonstrated a significant improvement in median progression-free survival (PFS, 19.2 vs 5.4 months, hazard ratio (HR) = .50 [95% CI, .33 to .78], p < .01). However, the overall survival (OS) was not significantly different between the two cohorts. Safety profiles showed that 37 (52%) patients in the matched BiTE cohort experienced cytokine release syndrome, mostly grade 1 (n = 29, 41%), with rare occurrences of neurotoxicity (n = 4, 5.6%). Infections were significantly more common in the matched BiTE cohort compared with the matched SOC cohort (81% vs. 49%, p < .01). Non-B-cell mutation antigen (BCMA)-targeted BiTEs improved 6-month OS rates compared with BCMA-targeted BiTEs in monotherapy (94% [95% CI, 84 to 100] vs. 65% [95% CI, 45 to 95], p = .04) and combination with daratumumab (100% [95% CI, 100 to 100] vs. 77% [95% CI, 57 to 100], p = .20). Non-BCMA-targeted BiTEs also provided benefit for 6-month PFS rate compared with the BCMA-targeted BiTE cohort in monotherapy (76% [95% CI, 59 to 100] vs. 50% [95% CI, 31 to 82], p = .11) and combination with daratumumab (100% [95% CI, 100 to 100] vs. 69% [95% CI, 48 to 99], p = .10). Quantitative bias and sensitivity analyses confirmed the robustness of these results. This real-world data-based study underscores the potential of BiTEs to significantly enhance survival outcomes in patients with heavily treated RRMM and manageable safety profiles. The difference in clinical outcomes by BiTE targets warrants further investigation in larger clinical trials (ClinicalTrials.gov identifier: NCT06205823).
Collapse
Affiliation(s)
- Suein Choi
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Pharmacometrics Institute for Practical Education and Training (PIPET), College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ja Min Byun
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea; Cancer Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sung-Soo Park
- Catholic Research Network for Multiple Myeloma, Republic of Korea; Catholic Hematology Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| | - Jinsun Han
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Pharmacometrics Institute for Practical Education and Training (PIPET), College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sieun Oh
- Department of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seungpil Jung
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Pharmacometrics Institute for Practical Education and Training (PIPET), College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyejoon Park
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Pharmacometrics Institute for Practical Education and Training (PIPET), College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seunghoon Han
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Pharmacometrics Institute for Practical Education and Training (PIPET), College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jung Yeon Lee
- Catholic Research Network for Multiple Myeloma, Republic of Korea; Catholic Hematology Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Youngil Koh
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea; Cancer Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Young-Woo Jeon
- Catholic Research Network for Multiple Myeloma, Republic of Korea; Catholic Hematology Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Hematology, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung-Ah Yahng
- Catholic Research Network for Multiple Myeloma, Republic of Korea; Catholic Hematology Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Hematology, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Incheon, Republic of Korea
| | - Seung-Hwan Shin
- Catholic Research Network for Multiple Myeloma, Republic of Korea; Catholic Hematology Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Hematology, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sung-Soo Yoon
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea; Cancer Research Institute, Seoul National University Hospital, Seoul, Republic of Korea; Catholic Research Network for Multiple Myeloma, Republic of Korea
| | - Chang-Ki Min
- Catholic Research Network for Multiple Myeloma, Republic of Korea; Catholic Hematology Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
5
|
Imamura K, Izumi Y, Egawa N, Ayaki T, Nagai M, Nishiyama K, Watanabe Y, Murakami T, Hanajima R, Kataoka H, Kiriyama T, Nanaura H, Sugie K, Hirayama T, Kano O, Nakamori M, Maruyama H, Haji S, Fujita K, Atsuta N, Tatebe H, Tokuda T, Takahashi N, Morinaga A, Tabuchi R, Oe M, Kobayashi M, Lobello K, Morita S, Sobue G, Takahashi R, Inoue H. Protocol for a phase 2 study of bosutinib for amyotrophic lateral sclerosis using real-world data: induced pluripotent stem cell-based drug repurposing for amyotrophic lateral sclerosis medicine (iDReAM) study. BMJ Open 2024; 14:e082142. [PMID: 39461864 PMCID: PMC11529471 DOI: 10.1136/bmjopen-2023-082142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 09/24/2024] [Indexed: 10/29/2024] Open
Abstract
INTRODUCTION Amyotrophic lateral sclerosis (ALS) is a progressive, severe neurodegenerative disease caused by motor neuron death. Development of a medicine for ALS is urgently needed, and induced pluripotent cell-based drug repurposing identified a Src/c-Abl inhibitor, bosutinib, as a candidate for molecular targeted therapy of ALS. A phase 1 study confirmed the safety and tolerability of bosutinib in a 12-week treatment of ALS patients. The objectives of this study are to evaluate the efficacy and longer-term safety of bosutinib in ALS patients. METHODS AND ANALYSIS An open-label, multicentre phase 2 study was designed. The study consisted of a 12-week observation period, a 1-week transitional period, a 24-week study treatment period and a 4-week follow-up period. Following the transitional period, patients whose total Revised ALS Functional Rating Scale (ALSFRS-R) score declined by 1 to 4 points during the 12-week observation period were to receive bosutinib for 24 weeks. In this study, 25 ALS patients will be enrolled; patients will be randomly assigned to the following groups: 12 patients in the 200 mg quaque die (QD) group and 13 patients in the 300 mg QD group of bosutinib. The safety and exploratory efficacy of bosutinib in ALS patients for 24 weeks will be assessed. Efficacy using the ALSFRS-R score will be compared with the external published data from an edaravone study (MCI186-19) and registry data from a multicentre ALS cohort study, the Japanese Consortium for Amyotrophic Lateral Sclerosis Research. ETHICS AND DISSEMINATION This study was approved by the ethics committees of Kyoto University, Tokushima University, Kitasato University, Tottori University, Nara Medical University School of Medicine, Toho University and Hiroshima University. The findings will be disseminated in peer-reviewed journals and at scientific conferences. TRIAL REGISTRATION NUMBER jRCT2051220002; Pre-results, NCT04744532; Pre-results.
Collapse
Affiliation(s)
- Keiko Imamura
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Yuishin Izumi
- Department of Neurology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Naohiro Egawa
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takashi Ayaki
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Makiko Nagai
- Department of Neurology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Kazutoshi Nishiyama
- Department of Neurology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Yasuhiro Watanabe
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Takenobu Murakami
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Ritsuko Hanajima
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Hiroshi Kataoka
- Department of Neurology, Nara Medical University School of Medicine, Kashihara, Japan
| | - Takao Kiriyama
- Department of Neurology, Nara Medical University School of Medicine, Kashihara, Japan
| | - Hitoki Nanaura
- Department of Neurology, Nara Medical University School of Medicine, Kashihara, Japan
| | - Kazuma Sugie
- Department of Neurology, Nara Medical University School of Medicine, Kashihara, Japan
| | - Takehisa Hirayama
- Department of Neurology, Toho University Faculty of Medicine, Tokyo, Japan
| | - Osamu Kano
- Department of Neurology, Toho University Faculty of Medicine, Tokyo, Japan
| | - Masahiro Nakamori
- Department of Clinical Neuroscience and Therapeutics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hirofumi Maruyama
- Department of Clinical Neuroscience and Therapeutics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shotaro Haji
- Department of Neurology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Koji Fujita
- Department of Neurology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Naoki Atsuta
- Department of Neurology, Aichi Medical University, Nagakute, Japan
| | - Harutsugu Tatebe
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Takahiko Tokuda
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Naoto Takahashi
- Department of Hematology, Nephrology, and Rheumatology, Akita University Graduate School of Medicine, Akita, Japan
| | | | | | | | | | - Kasia Lobello
- Pfizer Worldwide Research and Development, Collegeville, Pennsylvania, USA
| | - Satoshi Morita
- Department of Biomedical Statistics and Bioinformatics, Kyoto University, Kyoto, Japan
- Institute for Advancement of Clinical and Translational Science (iACT), Kyoto University Hospital, Kyoto, Japan
| | - Gen Sobue
- Aichi Medical University, Nagakute, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Haruhisa Inoue
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Institute for Advancement of Clinical and Translational Science (iACT), Kyoto University Hospital, Kyoto, Japan
| |
Collapse
|
6
|
Kryukov M, Moriarty KP, Villamea M, O'Dwyer I, Chow O, Dormont F, Hernandez R, Bar-Joseph Z, Rufino B. Proxy endpoints - bridging clinical trials and real world data. J Biomed Inform 2024; 158:104723. [PMID: 39299565 DOI: 10.1016/j.jbi.2024.104723] [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: 06/20/2024] [Revised: 08/15/2024] [Accepted: 09/03/2024] [Indexed: 09/22/2024]
Abstract
OBJECTIVE Disease severity scores, or endpoints, are routinely measured during Randomized Controlled Trials (RCTs) to closely monitor the effect of treatment. In real-world clinical practice, although a larger set of patients is observed, the specific RCT endpoints are often not captured, which makes it hard to utilize real-world data (RWD) to evaluate drug efficacy in larger populations. METHODS To overcome this challenge, we developed an ensemble technique which learns proxy models of disease endpoints in RWD. Using a multi-stage learning framework applied to RCT data, we first identify features considered significant drivers of disease available within RWD. To create endpoint proxy models, we use Explainable Boosting Machines (EBMs) which allow for both end-user interpretability and modeling of non-linear relationships. RESULTS We demonstrate our approach on two diseases, rheumatoid arthritis (RA) and atopic dermatitis (AD). As we show, our combined feature selection and prediction method achieves good results for both disease areas, improving upon prior methods proposed for predictive disease severity scoring. CONCLUSION Having disease severity over time for a patient is important to further disease understanding and management. Our results open the door to more use cases in the space of RA and AD such as treatment effect estimates or prognostic scoring on RWD. Our framework may be extended beyond RA and AD to other diseases where the severity score is not well measured in electronic health records.
Collapse
Affiliation(s)
- Maxim Kryukov
- Data & Computational Science, R&D, Sanofi, Barcelona, Spain.
| | - Kathleen P Moriarty
- Data & Computational Science, R&D, Sanofi, 240 Richmond Street West, 3rd Floor, Toronto, M5V 1V6, Ontario, Canada.
| | | | - Ingrid O'Dwyer
- Data & Computational Science, R&D, Sanofi, 240 Richmond Street West, 3rd Floor, Toronto, M5V 1V6, Ontario, Canada.
| | - Ohn Chow
- Clinical Immunology and Inflammation, R&D, Sanofi, 450 Water St, MA, Cambridge, 02141, MA, United States.
| | - Flavio Dormont
- Clinical Real World Evidence, R&D, Sanofi, 46 Av. de la Grande Armée, Paris, 75017, Île-de-France, France.
| | - Ramon Hernandez
- Clinical Real World Evidence, R&D, Sanofi, 46 Av. de la Grande Armée, Paris, 75017, Île-de-France, France.
| | - Ziv Bar-Joseph
- Data & Computational Science, R&D, Sanofi, 450 Water St, MA, Cambridge, 02141, MA, United States.
| | - Brandon Rufino
- Data & Computational Science, R&D, Sanofi, 240 Richmond Street West, 3rd Floor, Toronto, M5V 1V6, Ontario, Canada.
| |
Collapse
|
7
|
Vallée A. Envisioning the Future of Personalized Medicine: Role and Realities of Digital Twins. J Med Internet Res 2024; 26:e50204. [PMID: 38739913 PMCID: PMC11130780 DOI: 10.2196/50204] [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: 06/22/2023] [Revised: 10/01/2023] [Accepted: 12/29/2023] [Indexed: 05/16/2024] Open
Abstract
Digital twins have emerged as a groundbreaking concept in personalized medicine, offering immense potential to transform health care delivery and improve patient outcomes. It is important to highlight the impact of digital twins on personalized medicine across the understanding of patient health, risk assessment, clinical trials and drug development, and patient monitoring. By mirroring individual health profiles, digital twins offer unparalleled insights into patient-specific conditions, enabling more accurate risk assessments and tailored interventions. However, their application extends beyond clinical benefits, prompting significant ethical debates over data privacy, consent, and potential biases in health care. The rapid evolution of this technology necessitates a careful balancing act between innovation and ethical responsibility. As the field of personalized medicine continues to evolve, digital twins hold tremendous promise in transforming health care delivery and revolutionizing patient care. While challenges exist, the continued development and integration of digital twins hold the potential to revolutionize personalized medicine, ushering in an era of tailored treatments and improved patient well-being. Digital twins can assist in recognizing trends and indicators that might signal the presence of diseases or forecast the likelihood of developing specific medical conditions, along with the progression of such diseases. Nevertheless, the use of human digital twins gives rise to ethical dilemmas related to informed consent, data ownership, and the potential for discrimination based on health profiles. There is a critical need for robust guidelines and regulations to navigate these challenges, ensuring that the pursuit of advanced health care solutions does not compromise patient rights and well-being. This viewpoint aims to ignite a comprehensive dialogue on the responsible integration of digital twins in medicine, advocating for a future where technology serves as a cornerstone for personalized, ethical, and effective patient care.
Collapse
Affiliation(s)
- Alexandre Vallée
- Department of Epidemiology and Public Health, Foch Hospital, Suresnes, France
| |
Collapse
|
8
|
Kim M, Patrick K, Nebeker C, Godino J, Stein S, Klasnja P, Perski O, Viglione C, Coleman A, Hekler E. The Digital Therapeutics Real-World Evidence Framework: An Approach for Guiding Evidence-Based Digital Therapeutics Design, Development, Testing, and Monitoring. J Med Internet Res 2024; 26:e49208. [PMID: 38441954 PMCID: PMC10951831 DOI: 10.2196/49208] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 01/13/2024] [Accepted: 01/29/2024] [Indexed: 03/07/2024] Open
Abstract
Digital therapeutics (DTx) are a promising way to provide safe, effective, accessible, sustainable, scalable, and equitable approaches to advance individual and population health. However, developing and deploying DTx is inherently complex in that DTx includes multiple interacting components, such as tools to support activities like medication adherence, health behavior goal-setting or self-monitoring, and algorithms that adapt the provision of these according to individual needs that may change over time. While myriad frameworks exist for different phases of DTx development, no single framework exists to guide evidence production for DTx across its full life cycle, from initial DTx development to long-term use. To fill this gap, we propose the DTx real-world evidence (RWE) framework as a pragmatic, iterative, milestone-driven approach for developing DTx. The DTx RWE framework is derived from the 4-phase development model used for behavioral interventions, but it includes key adaptations that are specific to the unique characteristics of DTx. To ensure the highest level of fidelity to the needs of users, the framework also incorporates real-world data (RWD) across the entire life cycle of DTx development and use. The DTx RWE framework is intended for any group interested in developing and deploying DTx in real-world contexts, including those in industry, health care, public health, and academia. Moreover, entities that fund research that supports the development of DTx and agencies that regulate DTx might find the DTx RWE framework useful as they endeavor to improve how DTxcan advance individual and population health.
Collapse
Affiliation(s)
- Meelim Kim
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla, CA, United States
- Department of Preventive Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- The Qualcomm Institute, University of California San Diego, La Jolla, CA, United States
- The Design Lab, University of California San Diego, La Jolla, CA, United States
| | - Kevin Patrick
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla, CA, United States
- The Qualcomm Institute, University of California San Diego, La Jolla, CA, United States
| | - Camille Nebeker
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla, CA, United States
- The Qualcomm Institute, University of California San Diego, La Jolla, CA, United States
- The Design Lab, University of California San Diego, La Jolla, CA, United States
| | - Job Godino
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla, CA, United States
- The Qualcomm Institute, University of California San Diego, La Jolla, CA, United States
- Laura Rodriguez Research Institute, Family Health Centers of San Diego, San Diego, CA, United States
| | | | - Predrag Klasnja
- School of Information, University of Michigan, Ann Arbor, MI, United States
| | - Olga Perski
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla, CA, United States
- Faculty of Social Sciences, Tampere University, Tampere, Finland
| | - Clare Viglione
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla, CA, United States
| | - Aaron Coleman
- Small Steps Labs LLC dba Fitabase Inc, San Diego, CA, United States
| | - Eric Hekler
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla, CA, United States
- The Qualcomm Institute, University of California San Diego, La Jolla, CA, United States
- The Design Lab, University of California San Diego, La Jolla, CA, United States
| |
Collapse
|
9
|
Cheng F, Wang F, Tang J, Zhou Y, Fu Z, Zhang P, Haines JL, Leverenz JB, Gan L, Hu J, Rosen-Zvi M, Pieper AA, Cummings J. Artificial intelligence and open science in discovery of disease-modifying medicines for Alzheimer's disease. Cell Rep Med 2024; 5:101379. [PMID: 38382465 PMCID: PMC10897520 DOI: 10.1016/j.xcrm.2023.101379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 08/15/2023] [Accepted: 12/19/2023] [Indexed: 02/23/2024]
Abstract
The high failure rate of clinical trials in Alzheimer's disease (AD) and AD-related dementia (ADRD) is due to a lack of understanding of the pathophysiology of disease, and this deficit may be addressed by applying artificial intelligence (AI) to "big data" to rapidly and effectively expand therapeutic development efforts. Recent accelerations in computing power and availability of big data, including electronic health records and multi-omics profiles, have converged to provide opportunities for scientific discovery and treatment development. Here, we review the potential utility of applying AI approaches to big data for discovery of disease-modifying medicines for AD/ADRD. We illustrate how AI tools can be applied to the AD/ADRD drug development pipeline through collaborative efforts among neurologists, gerontologists, geneticists, pharmacologists, medicinal chemists, and computational scientists. AI and open data science expedite drug discovery and development of disease-modifying therapeutics for AD/ADRD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA.
| | - Fei Wang
- Department of Population Health Sciences, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Jian Tang
- Mila-Quebec Institute for Learning Algorithms and CIFAR AI Research Chair, HEC Montreal, Montréal, QC H3T 2A7, Canada
| | - Yadi Zhou
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Zhimin Fu
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; College of Pharmacy, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Pengyue Zhang
- Department of Biostatistics and Health Data Science, Indiana University, Indianapolis, IN 46037, USA
| | - Jonathan L Haines
- Cleveland Institute for Computational Biology, and Department of Population & Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - James B Leverenz
- Lou Ruvo Center for Brain Health, Neurological Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Li Gan
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Jianying Hu
- IBM Research, Yorktown Heights, New York, NY 10598, USA
| | - Michal Rosen-Zvi
- AI for Accelerated Healthcare and Life Sciences Discovery, IBM Research Labs, Haifa 3498825, Israel; Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9190500, Israel
| | - Andrew A Pieper
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH 44106, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland OH 44106, USA; Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH, 44106, USA; Department of Neurosciences, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| | - Jeffrey Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, UNLV, Las Vegas, NV 89154, USA
| |
Collapse
|
10
|
Kurki S, Halla-Aho V, Haussmann M, Lähdesmäki H, Leinonen JV, Koskinen M. A comparative study of clinical trial and real-world data in patients with diabetic kidney disease. Sci Rep 2024; 14:1731. [PMID: 38243002 PMCID: PMC10798981 DOI: 10.1038/s41598-024-51938-3] [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: 10/18/2023] [Accepted: 01/11/2024] [Indexed: 01/21/2024] Open
Abstract
A growing body of research is focusing on real-world data (RWD) to supplement or replace randomized controlled trials (RCTs). However, due to the disparities in data generation mechanisms, differences are likely and necessitate scrutiny to validate the merging of these datasets. We compared the characteristics of RCT data from 5734 diabetic kidney disease patients with corresponding RWD from electronic health records (EHRs) of 23,523 patients. Demographics, diagnoses, medications, laboratory measurements, and vital signs were analyzed using visualization, statistical comparison, and cluster analysis. RCT and RWD sets exhibited significant differences in prevalence, longitudinality, completeness, and sampling density. The cluster analysis revealed distinct patient subgroups within both RCT and RWD sets, as well as clusters containing patients from both sets. We stress the importance of validation to verify the feasibility of combining RCT and RWD, for instance, in building an external control arm. Our results highlight general differences between RCT and RWD sets, which should be considered during the planning stages of an RCT-RWD study. If they are, RWD has the potential to enrich RCT data by providing first-hand baseline data, filling in missing data or by subgrouping or matching individuals, which calls for advanced methods to mitigate the differences between datasets.
Collapse
Affiliation(s)
- Samu Kurki
- Bayer Oy, Tuulikuja 2, 02100, Espoo, Finland.
| | | | - Manuel Haussmann
- Department of Computer Science, Aalto University, Espoo, Finland
| | - Harri Lähdesmäki
- Department of Computer Science, Aalto University, Espoo, Finland
| | | | | |
Collapse
|
11
|
Zang C, Zhang H, Xu J, Zhang H, Fouladvand S, Havaldar S, Cheng F, Chen K, Chen Y, Glicksberg BS, Chen J, Bian J, Wang F. High-throughput target trial emulation for Alzheimer's disease drug repurposing with real-world data. Nat Commun 2023; 14:8180. [PMID: 38081829 PMCID: PMC10713627 DOI: 10.1038/s41467-023-43929-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
Target trial emulation is the process of mimicking target randomized trials using real-world data, where effective confounding control for unbiased treatment effect estimation remains a main challenge. Although various approaches have been proposed for this challenge, a systematic evaluation is still lacking. Here we emulated trials for thousands of medications from two large-scale real-world data warehouses, covering over 10 years of clinical records for over 170 million patients, aiming to identify new indications of approved drugs for Alzheimer's disease. We assessed different propensity score models under the inverse probability of treatment weighting framework and suggested a model selection strategy for improved baseline covariate balancing. We also found that the deep learning-based propensity score model did not necessarily outperform logistic regression-based methods in covariate balancing. Finally, we highlighted five top-ranked drugs (pantoprazole, gabapentin, atorvastatin, fluticasone, and omeprazole) originally intended for other indications with potential benefits for Alzheimer's patients.
Collapse
Affiliation(s)
- Chengxi Zang
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
- Institute of Artificial Intelligence for Digital Health, Weill Cornell Medicine, New York, NY, USA
| | - Hao Zhang
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Jie Xu
- Department of Health Outcomes & Biomedical Informatics, University of Florida, Gainesville, FL, USA
| | - Hansi Zhang
- Department of Health Outcomes & Biomedical Informatics, University of Florida, Gainesville, FL, USA
| | - Sajjad Fouladvand
- Institude for Biomedical Informatics (IBI) and Department of Computer Science, University of Kentucky, Lexington, KY, USA
| | - Shreyas Havaldar
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Kun Chen
- Department of Statistics, University of Connecticut, Storrs, CT, USA
| | - Yong Chen
- Department of Biostatistics, Epidemiology and Informatics (DBEI), the Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Benjamin S Glicksberg
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jin Chen
- Institude for Biomedical Informatics (IBI) and Department of Computer Science, University of Kentucky, Lexington, KY, USA
| | - Jiang Bian
- Department of Health Outcomes & Biomedical Informatics, University of Florida, Gainesville, FL, USA
| | - Fei Wang
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA.
- Institute of Artificial Intelligence for Digital Health, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
12
|
Guo LL, Guo LY, Li J, Gu YW, Wang JY, Cui Y, Qian Q, Chen T, Jiang R, Zheng S. Characteristics and Admission Preferences of Pediatric Emergency Patients and Their Waiting Time Prediction Using Electronic Medical Record Data: Retrospective Comparative Analysis. J Med Internet Res 2023; 25:e49605. [PMID: 37910168 PMCID: PMC10652198 DOI: 10.2196/49605] [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: 06/03/2023] [Revised: 07/04/2023] [Accepted: 09/20/2023] [Indexed: 11/03/2023] Open
Abstract
BACKGROUND The growing number of patients visiting pediatric emergency departments could have a detrimental impact on the care provided to children who are triaged as needing urgent attention. Therefore, it has become essential to continuously monitor and analyze the admissions and waiting times of pediatric emergency patients. Despite the significant challenge posed by the shortage of pediatric medical resources in China's health care system, there have been few large-scale studies conducted to analyze visits to the pediatric emergency room. OBJECTIVE This study seeks to examine the characteristics and admission patterns of patients in the pediatric emergency department using electronic medical record (EMR) data. Additionally, it aims to develop and assess machine learning models for predicting waiting times for pediatric emergency department visits. METHODS This retrospective analysis involved patients who were admitted to the emergency department of Children's Hospital Capital Institute of Pediatrics from January 1, 2021, to December 31, 2021. Clinical data from these admissions were extracted from the electronic medical records, encompassing various variables of interest such as patient demographics, clinical diagnoses, and time stamps of clinical visits. These indicators were collected and compared. Furthermore, we developed and evaluated several computational models for predicting waiting times. RESULTS In total, 183,024 eligible admissions from 127,368 pediatric patients were included. During the 12-month study period, pediatric emergency department visits were most frequent among children aged less than 5 years, accounting for 71.26% (130,423/183,024) of the total visits. Additionally, there was a higher proportion of male patients (104,147/183,024, 56.90%) compared with female patients (78,877/183,024, 43.10%). Fever (50,715/183,024, 27.71%), respiratory infection (43,269/183,024, 23.64%), celialgia (9560/183,024, 5.22%), and emesis (6898/183,024, 3.77%) were the leading causes of pediatric emergency room visits. The average daily number of admissions was 501.44, and 18.76% (34,339/183,204) of pediatric emergency department visits resulted in discharge without a prescription or further tests. The median waiting time from registration to seeing a doctor was 27.53 minutes. Prolonged waiting times were observed from April to July, coinciding with an increased number of arrivals, primarily for respiratory diseases. In terms of waiting time prediction, machine learning models, specifically random forest, LightGBM, and XGBoost, outperformed regression methods. On average, these models reduced the root-mean-square error by approximately 17.73% (8.951/50.481) and increased the R2 by approximately 29.33% (0.154/0.525). The SHAP method analysis highlighted that the features "wait.green" and "department" had the most significant influence on waiting times. CONCLUSIONS This study offers a contemporary exploration of pediatric emergency room visits, revealing significant variations in admission rates across different periods and uncovering certain admission patterns. The machine learning models, particularly ensemble methods, delivered more dependable waiting time predictions. Patient volume awaiting consultation or treatment and the triage status emerged as crucial factors contributing to prolonged waiting times. Therefore, strategies such as patient diversion to alleviate congestion in emergency departments and optimizing triage systems to reduce average waiting times remain effective approaches to enhance the quality of pediatric health care services in China.
Collapse
Affiliation(s)
- Lin Lin Guo
- Children's Hospital Capital Institute of Pediatrics, Beijing, China
| | - Lin Ying Guo
- Children's Hospital Capital Institute of Pediatrics, Beijing, China
| | - Jiao Li
- Institute of Medical Information, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yao Wen Gu
- Institute of Medical Information, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jia Yang Wang
- Institute of Medical Information, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ying Cui
- Children's Hospital Capital Institute of Pediatrics, Beijing, China
| | - Qing Qian
- Institute of Medical Information, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ting Chen
- Department of Computer Science and Technology, Tsinghua University, Beijing, China
| | - Rui Jiang
- Department of Automation, Tsinghua University, Beijing, China
| | - Si Zheng
- Institute of Medical Information, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Computer Science and Technology, Tsinghua University, Beijing, China
| |
Collapse
|
13
|
Su Q, Cheng G, Huang J. A review of research on eligibility criteria for clinical trials. Clin Exp Med 2023; 23:1867-1879. [PMID: 36602707 PMCID: PMC9815064 DOI: 10.1007/s10238-022-00975-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 12/06/2022] [Indexed: 01/06/2023]
Abstract
The purpose of this paper is to systematically sort out and analyze the cutting-edge research on the eligibility criteria of clinical trials. Eligibility criteria are important prerequisites for the success of clinical trials. It directly affects the final results of the clinical trials. Inappropriate eligibility criteria will lead to insufficient recruitment, which is an important reason for the eventual failure of many clinical trials. We have investigated the research status of eligibility criteria for clinical trials on academic platforms such as arXiv and NIH. We have classified and sorted out all the papers we found, so that readers can understand the frontier research in this field. Eligibility criteria are the most important part of a clinical trial study. The ultimate goal of research in this field is to formulate more scientific and reasonable eligibility criteria and speed up the clinical trial process. The global research on the eligibility criteria of clinical trials is mainly divided into four main aspects: natural language processing, patient pre-screening, standard evaluation, and clinical trial query. Compared with the past, people are now using new technologies to study eligibility criteria from a new perspective (big data). In the research process, complex disease concepts, how to choose a suitable dataset, how to prove the validity and scientific of the research results, are challenges faced by researchers (especially for computer-related researchers). Future research will focus on the selection and improvement of artificial intelligence algorithms related to clinical trials and related practical applications such as databases, knowledge graphs, and dictionaries.
Collapse
Affiliation(s)
- Qianmin Su
- Department of Computer Science, School of Electronic and Electrical Engineering, Shanghai University of Engineering Science, No. 333 Longteng Road, Shanghai, 201620, China.
| | - Gaoyi Cheng
- Department of Computer Science, School of Electronic and Electrical Engineering, Shanghai University of Engineering Science, No. 333 Longteng Road, Shanghai, 201620, China
| | - Jihan Huang
- Center for Drug Clinical Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| |
Collapse
|
14
|
Hansford HJ, Cashin AG, Jones MD, Swanson SA, Islam N, Douglas SRG, Rizzo RRN, Devonshire JJ, Williams SA, Dahabreh IJ, Dickerman BA, Egger M, Garcia-Albeniz X, Golub RM, Lodi S, Moreno-Betancur M, Pearson SA, Schneeweiss S, Sterne JAC, Sharp MK, Stuart EA, Hernán MA, Lee H, McAuley JH. Reporting of Observational Studies Explicitly Aiming to Emulate Randomized Trials: A Systematic Review. JAMA Netw Open 2023; 6:e2336023. [PMID: 37755828 PMCID: PMC10534275 DOI: 10.1001/jamanetworkopen.2023.36023] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 08/22/2023] [Indexed: 09/28/2023] Open
Abstract
Importance Observational (nonexperimental) studies that aim to emulate a randomized trial (ie, the target trial) are increasingly informing medical and policy decision-making, but it is unclear how these studies are reported in the literature. Consistent reporting is essential for quality appraisal, evidence synthesis, and translation of evidence to policy and practice. Objective To assess the reporting of observational studies that explicitly aimed to emulate a target trial. Evidence Review We searched Medline, Embase, PsycINFO, and Web of Science for observational studies published between March 2012 and October 2022 that explicitly aimed to emulate a target trial of a health or medical intervention. Two reviewers double-screened and -extracted data on study characteristics, key predefined components of the target trial protocol and its emulation (eligibility criteria, treatment strategies, treatment assignment, outcome[s], follow-up, causal contrast[s], and analysis plan), and other items related to the target trial emulation. Findings A total of 200 studies that explicitly aimed to emulate a target trial were included. These studies included 26 subfields of medicine, and 168 (84%) were published from January 2020 to October 2022. The aim to emulate a target trial was explicit in 70 study titles (35%). Forty-three studies (22%) reported use of a published reporting guideline (eg, Strengthening the Reporting of Observational Studies in Epidemiology). Eighty-five studies (43%) did not describe all key items of how the target trial was emulated and 113 (57%) did not describe the protocol of the target trial and its emulation. Conclusion and Relevance In this systematic review of 200 studies that explicitly aimed to emulate a target trial, reporting of how the target trial was emulated was inconsistent. A reporting guideline for studies explicitly aiming to emulate a target trial may improve the reporting of the target trial protocols and other aspects of these emulation attempts.
Collapse
Affiliation(s)
- Harrison J. Hansford
- School of Health Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, Australia
- Centre for Pain IMPACT, Neuroscience Research Australia, Sydney, Australia
| | - Aidan G. Cashin
- School of Health Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, Australia
- Centre for Pain IMPACT, Neuroscience Research Australia, Sydney, Australia
| | - Matthew D. Jones
- School of Health Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, Australia
- Centre for Pain IMPACT, Neuroscience Research Australia, Sydney, Australia
| | - Sonja A. Swanson
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, Pennsylvania
- CAUSALab, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Nazrul Islam
- Oxford Population Health, Big Data Institute, University of Oxford, Oxford, United Kingdom
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Susan R. G. Douglas
- School of Health Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, Australia
| | - Rodrigo R. N. Rizzo
- School of Health Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, Australia
- Centre for Pain IMPACT, Neuroscience Research Australia, Sydney, Australia
| | - Jack J. Devonshire
- Centre for Pain IMPACT, Neuroscience Research Australia, Sydney, Australia
| | - Sam A. Williams
- Centre for Pain IMPACT, Neuroscience Research Australia, Sydney, Australia
| | - Issa J. Dahabreh
- CAUSALab, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Barbra A. Dickerman
- CAUSALab, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Matthias Egger
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
- Centre for Infectious Disease Epidemiology and Research, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Xabier Garcia-Albeniz
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- RTI Health Solutions, Barcelona, Spain
| | - Robert M. Golub
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Sara Lodi
- CAUSALab, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Margarita Moreno-Betancur
- Clinical Epidemiology & Biostatistics Unit, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - Sallie-Anne Pearson
- School of Population Health, Faculty of Medicine and Health, UNSW Sydney, New South Wales, Australia
| | - Sebastian Schneeweiss
- Division of Pharmacoepidemiology, Department of Medicine, Brigham & Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jonathan A. C. Sterne
- Department of Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- NIHR Bristol Biomedical Research Centre, Bristol, United Kingdom
- Health Data Research UK South-West, Bristol, United Kingdom
| | - Melissa K. Sharp
- Department of Public Health and Epidemiology, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Elizabeth A. Stuart
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Miguel A. Hernán
- CAUSALab, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Hopin Lee
- University of Exeter Medical School, Exeter, United Kingdom
| | - James H. McAuley
- School of Health Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, Australia
- Centre for Pain IMPACT, Neuroscience Research Australia, Sydney, Australia
| |
Collapse
|
15
|
Miller MI, Shih LC, Kolachalama VB. Machine Learning in Clinical Trials: A Primer with Applications to Neurology. Neurotherapeutics 2023; 20:1066-1080. [PMID: 37249836 PMCID: PMC10228463 DOI: 10.1007/s13311-023-01384-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2023] [Indexed: 05/31/2023] Open
Abstract
We reviewed foundational concepts in artificial intelligence (AI) and machine learning (ML) and discussed ways in which these methodologies may be employed to enhance progress in clinical trials and research, with particular attention to applications in the design, conduct, and interpretation of clinical trials for neurologic diseases. We discussed ways in which ML may help to accelerate the pace of subject recruitment, provide realistic simulation of medical interventions, and enhance remote trial administration via novel digital biomarkers and therapeutics. Lastly, we provide a brief overview of the technical, administrative, and regulatory challenges that must be addressed as ML achieves greater integration into clinical trial workflows.
Collapse
Affiliation(s)
- Matthew I Miller
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, 72 E. Concord Street, Evans 636, Boston, MA, 02118, USA
| | - Ludy C Shih
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, 02118, USA
| | - Vijaya B Kolachalama
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, 72 E. Concord Street, Evans 636, Boston, MA, 02118, USA.
- Department of Computer Science and Faculty of Computing & Data Sciences, Boston University, Boston, MA, 02115, USA.
| |
Collapse
|
16
|
Chen Z, Zhang H, Yang X, Wu S, He X, Xu J, Guo J, Prosperi M, Wang F, Xu H, Chen Y, Hu H, DeKosky ST, Farrer M, Guo Y, Wu Y, Bian J. Assess the documentation of cognitive tests and biomarkers in electronic health records via natural language processing for Alzheimer's disease and related dementias. Int J Med Inform 2023; 170:104973. [PMID: 36577203 PMCID: PMC11325083 DOI: 10.1016/j.ijmedinf.2022.104973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 12/11/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Cognitive tests and biomarkers are the key information to assess the severity and track the progression of Alzheimer's' disease (AD) and AD-related dementias (AD/ADRD), yet, both are often only documented in clinical narratives of patients' electronic health records (EHRs). In this work, we aim to (1) assess the documentation of cognitive tests and biomarkers in EHRs that can be used as real-world endpoints, and (2) identify, extract, and harmonize the different commonly used cognitive tests from clinical narratives using natural language processing (NLP) methods into categorical AD/ADRD severity. METHODS We developed a rule-based NLP pipeline to extract the cognitive tests and biomarkers from clinical narratives in AD/ADRD patients' EHRs. We aggregated the extracted results to the patient level and harmonized the cognitive test scores into severity categories using cutoffs determined based on both relevant literature and domain knowledge of AD/ADRD clinicians. RESULTS We identified an AD/ADRD cohort of 48,912 patients from the University of Florida (UF) Health system and identified 7 measurements (6 cognitive tests and 1 biomarker) that are frequently documented in our data. Our NLP pipeline achieved an overall F1-score of 0.9059 across the 7 measurements. Among the 6 cognitive tests, we were able to harmonize 4 cognitive test scores into severity categories, and the population characteristics of patients with different severity were described. We also identified several factors related to the availability of their documentation in EHRs. CONCLUSION This study demonstrates that our NLP pipelines can extract cognitive tests and biomarkers of AD/ADRD accurately for downstream studies. Although, the documentation of cognitive tests and biomarkers in EHRs appears to be low, RWD is still an important resource for AD/ADRD research. Nevertheless, providing standardized approach to document cognitive tests and biomarkers in EHRS are also warranted.
Collapse
Affiliation(s)
- Zhaoyi Chen
- Department of Health Outcomes and Biomedical Informatics, University of Florida, Gainesville, FL, USA
| | - Hansi Zhang
- Department of Health Outcomes and Biomedical Informatics, University of Florida, Gainesville, FL, USA
| | - Xi Yang
- Department of Health Outcomes and Biomedical Informatics, University of Florida, Gainesville, FL, USA
| | - Songzi Wu
- Department of Health Outcomes and Biomedical Informatics, University of Florida, Gainesville, FL, USA
| | - Xing He
- Department of Health Outcomes and Biomedical Informatics, University of Florida, Gainesville, FL, USA
| | - Jie Xu
- Department of Health Outcomes and Biomedical Informatics, University of Florida, Gainesville, FL, USA
| | - Jingchuan Guo
- Department of Pharmaceutical Outcomes and Policy, University of Florida, Gainesville, FL, USA
| | - Mattia Prosperi
- Department of Epidemiology, University of Florida, Gainesville, FL, USA
| | - Fei Wang
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Hua Xu
- Center for Translational AI in Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yong Chen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Hui Hu
- Channing Division of Network Medicine at Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Steven T DeKosky
- Department of Neurology, University of Florida, Gainesville, FL, USA
| | - Matthew Farrer
- Department of Neurology, University of Florida, Gainesville, FL, USA
| | - Yi Guo
- Department of Health Outcomes and Biomedical Informatics, University of Florida, Gainesville, FL, USA
| | - Yonghui Wu
- Department of Health Outcomes and Biomedical Informatics, University of Florida, Gainesville, FL, USA.
| | - Jiang Bian
- Department of Health Outcomes and Biomedical Informatics, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
17
|
Ling Y, Upadhyaya P, Chen L, Jiang X, Kim Y. Emulate randomized clinical trials using heterogeneous treatment effect estimation for personalized treatments: Methodology review and benchmark. J Biomed Inform 2023; 137:104256. [PMID: 36455806 PMCID: PMC9845190 DOI: 10.1016/j.jbi.2022.104256] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/28/2022] [Accepted: 11/24/2022] [Indexed: 11/30/2022]
Abstract
Big data and (deep) machine learning have been ambitious tools in digital medicine, but these tools focus mainly on association. Intervention in medicine is about the causal effects. The average treatment effect has long been studied as a measure of causal effect, assuming that all populations have the same effect size. However, no "one-size-fits-all" treatment seems to work in some complex diseases. Treatment effects may vary by patient. Estimating heterogeneous treatment effects (HTE) may have a high impact on developing personalized treatment. Lots of advanced machine learning models for estimating HTE have emerged in recent years, but there has been limited translational research into the real-world healthcare domain. To fill the gap, we reviewed and compared eleven recent HTE estimation methodologies, including meta-learner, representation learning models, and tree-based models. We performed a comprehensive benchmark experiment based on nationwide healthcare claim data with application to Alzheimer's disease drug repurposing. We provided some challenges and opportunities in HTE estimation analysis in the healthcare domain to close the gap between innovative HTE models and deployment to real-world healthcare problems.
Collapse
Affiliation(s)
- Yaobin Ling
- Center for Secure Artificial Intelligence for Healthcare, School of Biomedical Informatics, University of Texas Health Science Center at Houston, Fannin 7000, Houston, TX, United States.
| | - Pulakesh Upadhyaya
- Center for Secure Artificial Intelligence for Healthcare, School of Biomedical Informatics, University of Texas Health Science Center at Houston, Fannin 7000, Houston, TX, United States.
| | - Luyao Chen
- Center for Secure Artificial Intelligence for Healthcare, School of Biomedical Informatics, University of Texas Health Science Center at Houston, Fannin 7000, Houston, TX, United States.
| | - Xiaoqian Jiang
- Center for Secure Artificial Intelligence for Healthcare, School of Biomedical Informatics, University of Texas Health Science Center at Houston, Fannin 7000, Houston, TX, United States.
| | - Yejin Kim
- Center for Secure Artificial Intelligence for Healthcare, School of Biomedical Informatics, University of Texas Health Science Center at Houston, Fannin 7000, Houston, TX, United States.
| |
Collapse
|
18
|
Patry C, Sauer LD, Sander A, Krupka K, Fichtner A, Brezinski J, Geissbühler Y, Aubrun E, Grinienko A, Strologo LD, Haffner D, Oh J, Grenda R, Pape L, Topaloğlu R, Weber LT, Bouts A, Kim JJ, Prytula A, König J, Shenoy M, Höcker B, Tönshoff B. Emulation of the control cohort of a randomized controlled trial in pediatric kidney transplantation with Real-World Data from the CERTAIN Registry. Pediatr Nephrol 2022; 38:1621-1632. [PMID: 36264431 PMCID: PMC9584233 DOI: 10.1007/s00467-022-05777-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 09/02/2022] [Accepted: 09/29/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Randomized controlled trials in pediatric kidney transplantation are hampered by low incidence and prevalence of kidney failure in children. Real-World Data from patient registries could facilitate the conduct of clinical trials by substituting a control cohort. However, the emulation of a control cohort by registry data in pediatric kidney transplantation has not been investigated so far. METHODS In this multicenter comparative analysis, we emulated the control cohort (n = 54) of an RCT in pediatric kidney transplant patients (CRADLE trial; ClinicalTrials.gov NCT01544491) with data derived from the Cooperative European Paediatric Renal Transplant Initiative (CERTAIN) registry, using the same inclusion and exclusion criteria (CERTAIN cohort, n = 554). RESULTS Most baseline patient and transplant characteristics were well comparable between both cohorts. At year 1 posttransplant, a composite efficacy failure end point comprising biopsy-proven acute rejection, graft loss or death (5.8% ± 3.3% vs. 7.5% ± 1.1%, P = 0.33), and kidney function (72.5 ± 24.9 vs. 77.3 ± 24.2 mL/min/1.73 m2 P = 0.19) did not differ significantly between CRADLE and CERTAIN. Furthermore, the incidence and severity of BPAR (5.6% vs. 7.8%), the degree of proteinuria (20.2 ± 13.9 vs. 30.6 ± 58.4 g/mol, P = 0.15), and the key safety parameters such as occurrence of urinary tract infections (24.1% vs. 15.5%, P = 0.10) were well comparable. CONCLUSIONS In conclusion, usage of Real-World Data from patient registries such as CERTAIN to emulate the control cohort of an RCT is feasible and could facilitate the conduct of clinical trials in pediatric kidney transplantation. A higher resolution version of the Graphical abstract is available as Supplementary information.
Collapse
Affiliation(s)
- Christian Patry
- Department of Pediatrics I, University Children's Hospital Heidelberg, Heidelberg, Germany.
| | - Lukas D. Sauer
- Institute of Medical Biometry, University of Heidelberg, Heidelberg, Germany
| | - Anja Sander
- Institute of Medical Biometry, University of Heidelberg, Heidelberg, Germany
| | - Kai Krupka
- Department of Pediatrics I, University Children’s Hospital Heidelberg, Heidelberg, Germany
| | - Alexander Fichtner
- Department of Pediatrics I, University Children’s Hospital Heidelberg, Heidelberg, Germany
| | - Jolanda Brezinski
- Institute of Medical Biometry, University of Heidelberg, Heidelberg, Germany
| | | | | | | | - Luca Dello Strologo
- Renal Transplant Unit, Bambino Gesù Children’s Hospital, Pediatric subspecialities, Rome, Italy
| | - Dieter Haffner
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Hannover, Germany
| | - Jun Oh
- Pediatric Nephrology, University Hospital Hamburg, Hamburg, Germany
| | - Ryszard Grenda
- Department of Nephrology, Kidney Transplantation and Hypertension, Children’s Memorial Health Institute, Warsaw, Poland
| | - Lars Pape
- Clinic for Paediatrics III, Essen University Hospital, Essen, Germany
| | - Rezan Topaloğlu
- Department of Pediatric Nephrology, School of Medicine, Hacettepe University, Ankara, Turkey
| | - Lutz T. Weber
- Pediatric Nephrology, Children’s and Adolescents’ Hospital, University Hospital Cologne, Medical Faculty University of Cologne, Cologne, Germany
| | - Antonia Bouts
- Department of Pediatric Nephrology, Amsterdam University Medical Center, Emma Children’s Hospital, Amsterdam, The Netherlands
| | - Jon Jin Kim
- Department of Paediatric Nephrology, Nottingham University Hospital, Nottingham, UK
| | - Agnieszka Prytula
- Pediatric Nephrology and Rheumatology Department, Ghent University Hospital, Ghent, Belgium
| | - Jens König
- Department of General Pediatrics, University Children’s Hospital, Munster, Germany
| | - Mohan Shenoy
- Paediatric Nephrology, Royal Manchester Children’s Hospital, Manchester, UK
| | - Britta Höcker
- Department of Pediatrics I, University Children’s Hospital Heidelberg, Heidelberg, Germany
| | - Burkhard Tönshoff
- Department of Pediatrics I, University Children’s Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
19
|
Chen Z, Zhang H, George TJ, Guo Y, Prosperi M, Guo J, Braithwaite D, Wang F, Kibbe W, Wagner L, Bian J. Simulating Colorectal Cancer Trials Using Real-World Data. JCO Clin Cancer Inform 2022; 6:e2100195. [PMID: 35839432 PMCID: PMC9848597 DOI: 10.1200/cci.21.00195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 03/04/2022] [Accepted: 06/02/2022] [Indexed: 02/02/2023] Open
Abstract
PURPOSE Using real-world data (RWD)-based trial simulation approach, we aim to simulate colorectal cancer (CRC) trials and examine both effectiveness and safety end points in different simulation scenarios. METHODS We identified five phase III trials comparing new treatment regimens with an US Food and Drug Administration-approved first-line treatment in patients with metastatic CRC (ie, fluorouracil, leucovorin, and irinotecan) as the standard-of-care (SOC) control arm. Using Electronic Health Record-derived data from the OneFlorida network, we defined the study populations and outcome measures using the protocols from the original trials. Our design scenarios were (1) simulation of the SOC fluorouracil, leucovorin, and irinotecan arm and (2) comparative effectiveness research (CER) simulation of the control and experimental arms. For each scenario, we adjusted for random assignment, sampling, and dropout. We used overall survival (OS) and severe adverse events (SAEs) to measure effectiveness and safety. RESULTS We conducted CER simulations for two trials, and SOC simulations for three trials. The effect sizes of our simulated trials were stable across all simulation runs. Compared with the original trials, we observed longer OS and higher mean number of SAEs in both CER and SOC simulation. In the two CER simulations, hazard ratios associated with death from simulations were similar to that reported in the original trials. Consistent with the original trials, we found higher risk ratios of SAEs in the experiment arm, suggesting potentially higher toxicities from the new treatment regimen. We also observed similar SAE rates across all simulations compared with the original trials. CONCLUSION In this study, we simulated five CRC trials, and tested two simulation scenarios with several different configurations demonstrated that our simulations can robustly generate effectiveness and safety outcomes comparable with the original trials using real-world data.
Collapse
Affiliation(s)
- Zhaoyi Chen
- Department of Health Outcomes & Biomedical Informatics, College of Medicine, University of Florida, Gainesville, FL
- Cancer Informatics Share Resource, University of Florida Health Cancer Center, Gainesville, FL
| | - Hansi Zhang
- Department of Health Outcomes & Biomedical Informatics, College of Medicine, University of Florida, Gainesville, FL
| | - Thomas J. George
- Division of Hematology & Oncology, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL
| | - Yi Guo
- Department of Health Outcomes & Biomedical Informatics, College of Medicine, University of Florida, Gainesville, FL
- Cancer Informatics Share Resource, University of Florida Health Cancer Center, Gainesville, FL
| | - Mattia Prosperi
- Department of Epidemiology, College of Medicine & College of Public Health and Health Professions, University of Florida, Gainesville, FL
| | - Jingchuan Guo
- Department of Pharmaceutical Outcomes and Policy, College of Pharmacy, University of Florida, Gainesville, FL
| | - Dejana Braithwaite
- Department of Epidemiology, College of Medicine & College of Public Health and Health Professions, University of Florida, Gainesville, FL
- Department of Surgery, College of Medicine, University of Florida, Gainesville, FL
| | - Fei Wang
- Department of Population Health Sciences, Cornell University, New York, NY
| | - Warren Kibbe
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina
| | - Lynne Wagner
- Department of Social Sciences and Health Policy, Wake Forest School of Medicine, Winston-Salem, NC
| | - Jiang Bian
- Department of Health Outcomes & Biomedical Informatics, College of Medicine, University of Florida, Gainesville, FL
- Cancer Informatics Share Resource, University of Florida Health Cancer Center, Gainesville, FL
| |
Collapse
|
20
|
Kim Y, Zhang K, Savitz SI, Chen L, Schulz PE, Jiang X. Counterfactual analysis of differential comorbidity risk factors in Alzheimer's disease and related dementias. PLOS DIGITAL HEALTH 2022; 1:e0000018. [PMID: 36812506 PMCID: PMC9931358 DOI: 10.1371/journal.pdig.0000018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 01/21/2022] [Indexed: 11/19/2022]
Abstract
Alzheimer's disease and related dementias (ADRD) is a multifactorial disease that involves several different etiologic mechanisms with various comorbidities. There is also significant heterogeneity in the prevalence of ADRD across diverse demographics groups. Association studies on such heterogeneous comorbidity risk factors are limited in their ability to determine causation. We aim to compare counterfactual treatment effects of various comorbidity in ADRD in different racial groups (African Americans and Caucasians). We used 138,026 ADRD and 1:1 matched older adults without ADRD from nationwide electronic health records, which extensively cover a large population's long medical history in breadth. We matched African Americans and Caucasians based on age, sex, and high-risk comorbidities (hypertension, diabetes, obesity, vascular disease, heart disease, and head injury) to build two comparable cohorts. We derived a Bayesian network of 100 comorbidities and selected comorbidities with potential causal effect to ADRD. We estimated the average treatment effect (ATE) of the selected comorbidities on ADRD using inverse probability of treatment weighting. Late effects of cerebrovascular disease significantly predisposed older African Americans (ATE = 0.2715) to ADRD, but not in the Caucasian counterparts; depression significantly predisposed older Caucasian counterparts (ATE = 0.1560) to ADRD, but not in the African Americans. Our extensive counterfactual analysis using a nationwide EHR discovered different comorbidities that predispose older African Americans to ADRD compared to Caucasian counterparts. Despite the noisy and incomplete nature of the real-world data, the counterfactual analysis on the comorbidity risk factors can be a valuable tool to support the risk factor exposure studies.
Collapse
Affiliation(s)
- Yejin Kim
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
- Institute for Stroke and Cerebrovascular Disease, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Kai Zhang
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Sean I. Savitz
- Institute for Stroke and Cerebrovascular Disease, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Luyao Chen
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Paul E. Schulz
- Department of Neurology, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Xiaoqian Jiang
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
- Institute for Stroke and Cerebrovascular Disease, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| |
Collapse
|
21
|
Hogan WR, Shenkman EA, Robinson T, Carasquillo O, Robinson PS, Essner RZ, Bian J, Lipori G, Harle C, Magoc T, Manini L, Mendoza T, White S, Loiacono A, Hall J, Nelson D. The OneFlorida Data Trust: a centralized, translational research data infrastructure of statewide scope. J Am Med Inform Assoc 2021; 29:686-693. [PMID: 34664656 PMCID: PMC8922180 DOI: 10.1093/jamia/ocab221] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/03/2021] [Accepted: 09/29/2021] [Indexed: 01/22/2023] Open
Abstract
The OneFlorida Data Trust is a centralized research patient data repository created and managed by the OneFlorida Clinical Research Consortium ("OneFlorida"). It comprises structured electronic health record (EHR), administrative claims, tumor registry, death, and other data on 17.2 million individuals who received healthcare in Florida between January 2012 and the present. Ten healthcare systems in Miami, Orlando, Tampa, Jacksonville, Tallahassee, Gainesville, and rural areas of Florida contribute EHR data, covering the major metropolitan regions in Florida. Deduplication of patients is accomplished via privacy-preserving entity resolution (precision 0.97-0.99, recall 0.75), thereby linking patients' EHR, claims, and death data. Another unique feature is the establishment of mother-baby relationships via Florida vital statistics data. Research usage has been significant, including major studies launched in the National Patient-Centered Clinical Research Network ("PCORnet"), where OneFlorida is 1 of 9 clinical research networks. The Data Trust's robust, centralized, statewide data are a valuable and relatively unique research resource.
Collapse
Affiliation(s)
- William R Hogan
- Department of Health Outcomes & Biomedical Informatics, University of Florida, Gainesville, Florida, USA,Corresponding Author: William R. Hogan, MD, MS, FACMI, Clinical & Translational Research Building, 2004 Mowry Road, PO Box 100219, Gainesville, FL 32610, USA;
| | - Elizabeth A Shenkman
- Department of Health Outcomes & Biomedical Informatics, University of Florida, Gainesville, Florida, USA
| | | | | | | | | | - Jiang Bian
- Department of Health Outcomes & Biomedical Informatics, University of Florida, Gainesville, Florida, USA
| | | | - Christopher Harle
- Department of Health Outcomes & Biomedical Informatics, University of Florida, Gainesville, Florida, USA,UF Health, Gainesville, Florida, USA
| | | | - Lizabeth Manini
- Department of Health Outcomes & Biomedical Informatics, University of Florida, Gainesville, Florida, USA
| | - Tona Mendoza
- Department of Health Outcomes & Biomedical Informatics, University of Florida, Gainesville, Florida, USA
| | - Sonya White
- Department of Health Outcomes & Biomedical Informatics, University of Florida, Gainesville, Florida, USA
| | - Alex Loiacono
- Department of Health Outcomes & Biomedical Informatics, University of Florida, Gainesville, Florida, USA
| | - Jackie Hall
- Department of Health Outcomes & Biomedical Informatics, University of Florida, Gainesville, Florida, USA
| | | |
Collapse
|
22
|
Wedlund L, Kvedar J. Simulated trials: in silico approach adds depth and nuance to the RCT gold-standard. NPJ Digit Med 2021; 4:121. [PMID: 34381148 PMCID: PMC8357951 DOI: 10.1038/s41746-021-00492-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 07/20/2021] [Indexed: 11/09/2022] Open
|