1
|
Xu S, Cobzaru R, Finkelstein SN, Welsch RE, Ng K, Middleton L. Foundational model aided automatic high-throughput drug screening using self-controlled cohort study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.04.24311480. [PMID: 39148849 PMCID: PMC11326319 DOI: 10.1101/2024.08.04.24311480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Background Developing medicine from scratch to governmental authorization and detecting adverse drug reactions (ADR) have barely been economical, expeditious, and risk-averse investments. The availability of large-scale observational healthcare databases and the popularity of large language models offer an unparalleled opportunity to enable automatic high-throughput drug screening for both repurposing and pharmacovigilance. Objectives To demonstrate a general workflow for automatic high-throughput drug screening with the following advantages: (i) the association of various exposure on diseases can be estimated; (ii) both repurposing and pharmacovigilance are integrated; (iii) accurate exposure length for each prescription is parsed from clinical texts; (iv) intrinsic relationship between drugs and diseases are removed jointly by bioinformatic mapping and large language model - ChatGPT; (v) causal-wise interpretations for incidence rate contrasts are provided. Methods Using a self-controlled cohort study design where subjects serve as their own control group, we tested the intention-to-treat association between medications on the incidence of diseases. Exposure length for each prescription is determined by parsing common dosages in English free text into a structured format. Exposure period starts from initial prescription to treatment discontinuation. A same exposure length preceding initial treatment is the control period. Clinical outcomes and categories are identified using existing phenotyping algorithms. Incident rate ratios (IRR) are tested using uniformly most powerful (UMP) unbiased tests. Results We assessed 3,444 medications on 276 diseases on 6,613,198 patients from the Clinical Practice Research Datalink (CPRD), an UK primary care electronic health records (EHR) spanning from 1987 to 2018. Due to the built-in selection bias of self-controlled cohort studies, ingredients-disease pairs confounded by deterministic medical relationships are removed by existing map from RxNorm and nonexistent maps by calling ChatGPT. A total of 16,901 drug-disease pairs reveals significant risk reduction, which can be considered as candidates for repurposing, while a total of 11,089 pairs showed significant risk increase, where drug safety might be of a concern instead. Conclusions This work developed a data-driven, nonparametric, hypothesis generating, and automatic high-throughput workflow, which reveals the potential of natural language processing in pharmacoepidemiology. We demonstrate the paradigm to a large observational health dataset to help discover potential novel therapies and adverse drug effects. The framework of this study can be extended to other observational medical databases.
Collapse
Affiliation(s)
- Shenbo Xu
- Institute for Data, Systems, and Society, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Raluca Cobzaru
- Institute for Data, Systems, and Society, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Stan N. Finkelstein
- Institute for Data, Systems, and Society, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Roy E. Welsch
- Institute for Data, Systems, and Society, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Kenney Ng
- Institute for Data, Systems, and Society, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Lefkos Middleton
- Institute for Data, Systems, and Society, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| |
Collapse
|
2
|
Ali M, Tabassum H, Alam MM, Alothaim AS, Al-Malki ES, Jamal A, Parvez S. Valsartan: An Angiotensin Receptor Blocker Modulates BDNF Expression and Provides Neuroprotection Against Cerebral Ischemic Reperfusion Injury. Mol Neurobiol 2024:10.1007/s12035-024-04237-x. [PMID: 38789895 DOI: 10.1007/s12035-024-04237-x] [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: 07/04/2023] [Accepted: 05/08/2024] [Indexed: 05/26/2024]
Abstract
AT1 receptor blockers (ARBs) are commonly used drugs to treat cardiovascular disease and hypertension, but research on their impact on brain disorders is unattainable. Valsartan (VAL) is a drug that specifically blocks AT1 receptor. Despite the previous evidence for VAL to provide neuroprotection in case of ischemic reperfusion injury, evaluation of their potential in mitigating mitochondrial dysfunction that causes neuronal cell death and neurobehavioral impairment remains unknown. The aim of this study was to evaluate the therapeutic effect of repurposed drug VAL against ischemic reperfusion injury-induced neuronal alternation. tMCAO surgery was performed to induce focal cerebral ischemic reperfusion injury. Following ischemic reperfusion injury, we analyzed the therapeutic efficacy of VAL by measuring the infarct volume, brain water content, mitochondrial oxidative stress, mitochondrial membrane potential, histopathological architecture, and apoptotic marker protein. Our results showed that VAL administrations (5 and 10 mg/kg b.wt.) mitigated the brain damage, enhanced neurobehavioral outcomes, and alleviated mitochondrial-mediated oxidative damage. In addition to this, our findings demonstrated that VAL administration inhibits neuronal apoptosis by restoring the mitochondrial membrane potential. A follow-up investigation demonstrated that VAL induces BDNF expression and promoted ischemic tolerance via modulating the Akt/p-Creb signaling pathway. In summary, our results suggested that VAL administration provided neuroprotection, ameliorated mitochondrial dysfunction, preserved the integrity of neurons, and lead to functional improvement after ischemic reperfusion injury.
Collapse
Affiliation(s)
- Mubashshir Ali
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
- USF Health Byrd Alzheimer's Center and Neuroscience Institute, Department of Molecular Medicine, Morsani College of Medicine, Tampa, FL, 33613, USA
| | - Heena Tabassum
- Division of Basic Medical Sciences, Indian Council of Medical Research, Ministry of Health and Family Welfare, Government of India, V. Ramalingaswami Bhawan, New Delhi, 110029, India
| | - Mohammad Mumtaz Alam
- Drug Design and Medicinal Chemistry Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Abdulaziz S Alothaim
- Department of Biology, College of Science, Al-Zulfi, Majmaah University, Riyadh Region, 11952, Majmaah, Saudi Arabia
| | - Esam S Al-Malki
- Department of Biology, College of Science, Al-Zulfi, Majmaah University, Riyadh Region, 11952, Majmaah, Saudi Arabia
| | - Azfar Jamal
- Department of Biology, College of Science, Al-Zulfi, Majmaah University, Riyadh Region, 11952, Majmaah, Saudi Arabia.
- Health and Basic Science Research Centre, Majmaah University, 11952, Majmaah, Saudi Arabia.
| | - Suhel Parvez
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
3
|
Kan-Tor Y, Ness L, Szlak L, Benninger F, Ravid S, Chorev M, Rosen-Zvi M, Shimoni Y, Fisher RS. Comparing the efficacy of anti-seizure medications using matched cohorts on a large insurance claims database. Epilepsy Res 2024; 201:107313. [PMID: 38417192 DOI: 10.1016/j.eplepsyres.2024.107313] [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: 10/26/2023] [Revised: 01/22/2024] [Accepted: 01/25/2024] [Indexed: 03/01/2024]
Abstract
Epilepsy is a severe chronic neurological disease affecting 60 million people worldwide. Primary treatment is with anti-seizure medicines (ASMs), but many patients continue to experience seizures. We used retrospective insurance claims data on 280,587 patients with uncontrolled epilepsy (UE), defined as status epilepticus, need for a rescue medicine, or admission or emergency visit for an epilepsy code. We conducted a computational risk ratio analysis between pairs of ASMs using a causal inference method, in order to match 1034 clinical factors and simulate randomization. Data was extracted from the MarketScan insurance claims Research Database records from 2011 to 2015. The cohort consisted of individuals over 18 years old with a diagnosis of epilepsy who took one of eight ASMs and had more than a year of history prior to the filling of the drug prescription. Seven ASM exposures were analyzed: topiramate, phenytoin, levetiracetam, gabapentin, lamotrigine, valproate, and carbamazepine or oxcarbazepine (treated as the same exposure). We calculated the risk ratio of UE between pairs of ASM after controlling for bias with inverse propensity weighting applied to 1034 factors, such as demographics, confounding illnesses, non-epileptic conditions treated by ASMs, etc. All ASMs exhibited a significant reduction in the prevalence of UE, but three drugs showed pair-wise differences compared to other ASMs. Topiramate consistently was associated with a lower risk of UE, with a mean risk ratio range of 0.68-0.93 (average 0.82, CI: 0.56-1.08). Phenytoin and levetiracetam were consistently associated with a higher risk of UE with mean risk ratio ranges of 1.11 to 1.47 (average 1.13, CI 0.98-1.65) and 1.15 to 1.43 (average 1.2, CI 0.72-1.69), respectively. Large-scale retrospective insurance claims data - combined with causal inference analysis - provides an opportunity to compare the effect of treatments in real-world data in populations 1,000-fold larger than those in typical randomized trials. Our causal analysis identified the clinically unexpected finding of topiramate as being associated with a lower risk of UE; and phenytoin and levetiracetam as associated with a higher risk of UE (compared to other studied drugs, not to baseline). However, we note that our data set for this study only used insurance claims events, which does not comprise actual seizure frequencies, nor a clear picture of side effects. Our results do not advocate for any change in practice but demonstrate that conclusions from large databases may differ from and supplement those of randomized trials and clinical practice and therefore may guide further investigation.
Collapse
Affiliation(s)
- Yoav Kan-Tor
- AI for Healthcare and Life Sciences Department, IBM Research, Haifa, Israel
| | - Lior Ness
- AI for Healthcare and Life Sciences Department, IBM Research, Haifa, Israel
| | - Liran Szlak
- AI for Healthcare and Life Sciences Department, IBM Research, Haifa, Israel
| | - Felix Benninger
- Department of Neurology, Rabin Medical Center, Petach Tikva, Israel; School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sivan Ravid
- AI for Healthcare and Life Sciences Department, IBM Research, Haifa, Israel
| | - Michal Chorev
- AI for Healthcare and Life Sciences Department, IBM Research, Haifa, Israel; Centre for Applied Research, IBM Australia, Melbourne, Australia
| | - Michal Rosen-Zvi
- AI for Healthcare and Life Sciences Department, IBM Research, Haifa, Israel; Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Yishai Shimoni
- AI for Healthcare and Life Sciences Department, IBM Research, Haifa, Israel
| | | |
Collapse
|
4
|
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
|
5
|
Bieber T. Disease modification in inflammatory skin disorders: opportunities and challenges. Nat Rev Drug Discov 2023; 22:662-680. [PMID: 37443275 DOI: 10.1038/s41573-023-00735-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/05/2023] [Indexed: 07/15/2023]
Abstract
Progress in understanding of the mechanisms underlying chronic inflammatory skin disorders, such as atopic dermatitis and psoriasis vulgaris, has led to new treatment options with the primary goal of alleviating symptoms. In addition, this knowledge has the potential to inform on new strategies aimed at inducing deep and therapy-free remission, that is, disease modification, potentially impacting on associated comorbidities. However, to reach this goal, key areas require further exploration, including the definitions of disease modification and disease activity index, further understanding of disease mechanisms and systemic spillover effects, potential windows of opportunity, biomarkers for patient stratification and successful intervention, as well as appropriate study design. This Perspective article assesses the opportunities and challenges in the discovery and development of disease-modifying therapies for chronic inflammatory skin disorders.
Collapse
Affiliation(s)
- Thomas Bieber
- Department of Dermatology and Allergy, University Hospital, Bonn, Germany.
- Christine Kühne - Center for Allergy Research and Education, Davos, Switzerland.
- Davos Biosciences, Davos, Switzerland.
| |
Collapse
|
6
|
Kosmowska B, Paleczna M, Biała D, Kadłuczka J, Wardas J, Witkin JM, Cook JM, Sharmin D, Marcinkowska M, Kuter KZ. GABA-A Alpha 2/3 but Not Alpha 1 Receptor Subunit Ligand Inhibits Harmaline and Pimozide-Induced Tremor in Rats. Biomolecules 2023; 13:biom13020197. [PMID: 36830567 PMCID: PMC9953228 DOI: 10.3390/biom13020197] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/11/2023] [Accepted: 01/15/2023] [Indexed: 01/20/2023] Open
Abstract
Treatment of tremors, such as in essential tremor (ET) and Parkinson's disease (PD) is mostly ineffective. Exact tremor pathomechanisms are unknown and relevant animal models are missing. GABA-A receptor is a target for tremorolytic medications, but current non-selective drugs produce side effects and have safety liabilities. The aim of this study was a search for GABA-A subunit-specific tremorolytics using different tremor-generating mechanisms. Two selective positive allosteric modulators (PAMs) were tested. Zolpidem, targeting GABA-A α1, was not effective in models of harmaline-induced ET, pimozide- or tetrabenazine-induced tremulous jaw movements (TJMs), while the novel GABA-A α2/3 selective MP-III-024 significantly reduced both the harmaline-induced ET tremor and pimozide-induced TJMs. While zolpidem decreased the locomotor activity of the rats, MP-III-024 produced small increases. These results provide important new clues into tremor suppression mechanisms initiated by the enhancement of GABA-driven inhibition in pathways controlled by α2/3 but not α1 containing GABA-A receptors. Tremor suppression by MP-III-024 provides a compelling reason to consider selective PAMs targeting α2/3-containing GABA-A receptors as novel therapeutic drug targets for ET and PD-associated tremor. The possibility of the improved tolerability and safety of this mechanism over non-selective GABA potentiation provides an additional rationale to further pursue the selective α2/3 hypothesis.
Collapse
Affiliation(s)
- Barbara Kosmowska
- Department of Neuropsychopharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna St., 31-343 Krakow, Poland
| | - Martyna Paleczna
- Department of Neuropsychopharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna St., 31-343 Krakow, Poland
| | - Dominika Biała
- Department of Neuropsychopharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna St., 31-343 Krakow, Poland
| | - Justyna Kadłuczka
- Department of Neuropsychopharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna St., 31-343 Krakow, Poland
| | - Jadwiga Wardas
- Department of Neuropsychopharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna St., 31-343 Krakow, Poland
| | - Jeffrey M. Witkin
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA
- RespireRx Pharmaceuticals Inc., Glen Rock, NJ 07452, USA
| | - James M. Cook
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA
- RespireRx Pharmaceuticals Inc., Glen Rock, NJ 07452, USA
| | - Dishary Sharmin
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA
| | - Monika Marcinkowska
- Department of Pharmaceutical Chemistry, Jagiellonian University, Medical College, 9 Medyczna St., 30-688 Krakow, Poland
| | - Katarzyna Z. Kuter
- Department of Neuropsychopharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna St., 31-343 Krakow, Poland
- Correspondence: ; Tel.: +48-12-662-32-26
| |
Collapse
|
7
|
Courtois É, Nguyen TTH, Fournier A, Carcaillon-Bentata L, Moutengou É, Escolano S, Tubert-Bitter P, Elbaz A, Thiébaut ACM, Ahmed I. Identifying Protective Drugs for Parkinson's Disease in Health-Care Databases Using Machine Learning. Mov Disord 2022; 37:2376-2385. [PMID: 36054665 PMCID: PMC10087353 DOI: 10.1002/mds.29205] [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: 05/23/2022] [Revised: 07/29/2022] [Accepted: 08/12/2022] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Available treatments for Parkinson's disease (PD) are only partially or transiently effective. Identifying existing molecules that may present a therapeutic or preventive benefit for PD (drug repositioning) is thus of utmost interest. OBJECTIVE We aimed at detecting potentially protective associations between marketed drugs and PD through a large-scale automated screening strategy. METHODS We implemented a machine learning (ML) algorithm combining subsampling and lasso logistic regression in a case-control study nested in the French national health data system. Our study population comprised 40,760 incident PD patients identified by a validated algorithm during 2016 to 2018 and 176,395 controls of similar age, sex, and region of residence, all followed since 2006. Drug exposure was defined at the chemical subgroup level, then at the substance level of the Anatomical Therapeutic Chemical (ATC) classification considering the frequency of prescriptions over a 2-year period starting 10 years before the index date to limit reverse causation bias. Sensitivity analyses were conducted using a more specific definition of PD status. RESULTS Six drug subgroups were detected by our algorithm among the 374 screened. Sulfonamide diuretics (ATC-C03CA), in particular furosemide (C03CA01), showed the most robust signal. Other signals included adrenergics in combination with anticholinergics (R03AL) and insulins and analogues (A10AD). CONCLUSIONS We identified several signals that deserve to be confirmed in large studies with appropriate consideration of the potential for reverse causation. Our results illustrate the value of ML-based signal detection algorithms for identifying drugs inversely associated with PD risk in health-care databases. © 2022 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Émeline Courtois
- High-Dimensional Biostatistics for Drug Safety and Genomics, Université Paris-Saclay, UVSQ, Inserm, CESP, Villejuif, France
| | - Thi Thu Ha Nguyen
- Exposome, Heredity, Cancer and Health, Université Paris-Saclay, UVSQ, Inserm, CESP, Villejuif, France
| | - Agnès Fournier
- Exposome, Heredity, Cancer and Health, Université Paris-Saclay, UVSQ, Inserm, CESP, Villejuif, France
| | | | | | - Sylvie Escolano
- High-Dimensional Biostatistics for Drug Safety and Genomics, Université Paris-Saclay, UVSQ, Inserm, CESP, Villejuif, France
| | - Pascale Tubert-Bitter
- High-Dimensional Biostatistics for Drug Safety and Genomics, Université Paris-Saclay, UVSQ, Inserm, CESP, Villejuif, France
| | - Alexis Elbaz
- Exposome, Heredity, Cancer and Health, Université Paris-Saclay, UVSQ, Inserm, CESP, Villejuif, France
| | - Anne C M Thiébaut
- High-Dimensional Biostatistics for Drug Safety and Genomics, Université Paris-Saclay, UVSQ, Inserm, CESP, Villejuif, France
| | - Ismaïl Ahmed
- High-Dimensional Biostatistics for Drug Safety and Genomics, Université Paris-Saclay, UVSQ, Inserm, CESP, Villejuif, France
| |
Collapse
|
8
|
Shahn Z, Spear P, Lu H, Jiang S, Zhang S, Deshmukh N, Xu S, Ng K, Welsch R, Finkelstein S. Systematically exploring repurposing effects of antihypertensives. Pharmacoepidemiol Drug Saf 2022; 31:944-952. [PMID: 35689299 PMCID: PMC9545793 DOI: 10.1002/pds.5491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 05/20/2022] [Accepted: 06/06/2022] [Indexed: 11/29/2022]
Abstract
With availability of voluminous sets of observational data, an empirical paradigm to screen for drug repurposing opportunities (i.e., beneficial effects of drugs on nonindicated outcomes) is feasible. In this article, we use a linked claims and electronic health record database to comprehensively explore repurposing effects of antihypertensive drugs. We follow a target trial emulation framework for causal inference to emulate randomized controlled trials estimating confounding adjusted effects of antihypertensives on each of 262 outcomes of interest. We then fit hierarchical models to the results as a form of postprocessing to account for multiple comparisons and to sift through the results in a principled way. Our motivation is twofold. We seek both to surface genuinely intriguing drug repurposing opportunities and to elucidate through a real application some study design decisions and potential biases that arise in this context.
Collapse
Affiliation(s)
- Zach Shahn
- Division of Healthcare and Life Sciences, IBM Research, Armonk, New York, USA.,MIT-IBM Watson AI Lab, Cambridge, Massachusetts, USA.,Department of Epidemiology and Biostatistics, CUNY School of Public Health, New York City, New York, USA
| | - Phoebe Spear
- Department of Electrical Engineering and Computer Science, MIT, Cambridge, Massachusetts, USA
| | - Helen Lu
- Department of Electrical Engineering and Computer Science, MIT, Cambridge, Massachusetts, USA
| | - Sharon Jiang
- Department of Electrical Engineering and Computer Science, MIT, Cambridge, Massachusetts, USA
| | - Suki Zhang
- Department of Electrical Engineering and Computer Science, MIT, Cambridge, Massachusetts, USA
| | - Neil Deshmukh
- Department of Electrical Engineering and Computer Science, MIT, Cambridge, Massachusetts, USA
| | - Shenbo Xu
- Department of Electrical Engineering and Computer Science, MIT, Cambridge, Massachusetts, USA.,Engineering Systems, MIT Institute for Data Systems, and Society, Cambridge, Massachusetts, USA
| | - Kenney Ng
- Division of Healthcare and Life Sciences, IBM Research, Armonk, New York, USA.,MIT-IBM Watson AI Lab, Cambridge, Massachusetts, USA
| | - Roy Welsch
- MIT-IBM Watson AI Lab, Cambridge, Massachusetts, USA.,Operations Research and Statistics, MIT Sloan School of Management, Cambridge, Massachusetts, USA
| | - Stan Finkelstein
- MIT-IBM Watson AI Lab, Cambridge, Massachusetts, USA.,Engineering Systems, MIT Institute for Data Systems, and Society, Cambridge, Massachusetts, USA.,Division of Clinical Informatics, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
9
|
Park K. The use of real-world data in drug repurposing. Transl Clin Pharmacol 2021; 29:117-124. [PMID: 34621704 PMCID: PMC8492393 DOI: 10.12793/tcp.2021.29.e18] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 09/24/2021] [Accepted: 09/24/2021] [Indexed: 02/02/2023] Open
Abstract
Drug repurposing, or repositioning, is to identify new uses for existing drugs. Significantly reducing the costs and time-to-market of a medication, drug repurposing has been an alternative tool to accelerate drug development process. On the other hand, 'real world data (RWD)' has been also increasingly used to support drug development process owing to its better representing actual pattern of drug treatment and outcome in real world. In the healthcare domain, RWD refers to data collected from sources other than traditional clinical trials; for example, in electronic health records or claims and billing data. With the enactment of the 21st Century Cures Act, which encourages the use of RWD in drug development and repurposing as well, such increasing trend in RWD use will be expedited. In this context, this review provides an overview of recent progresses in the area of drug repurposing where RWD was used by firstly introducing the increasing trend and regulatory change in the use of RWD in drug development, secondly reviewing published works using RWD in drug repurposing, classifying them in the repurposing strategy, and lastly addressing limitations and advantages of RWDs.
Collapse
Affiliation(s)
- Kyungsoo Park
- Department of Pharmacology, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
10
|
Ypinga JHL, Van Halteren AD, Henderson EJ, Bloem BR, Smink AJ, Tenison E, Munneke M, Ben-Shlomo Y, Darweesh SKL. Rationale and design to evaluate the PRIME Parkinson care model: a prospective observational evaluation of proactive, integrated and patient-centred Parkinson care in The Netherlands (PRIME-NL). BMC Neurol 2021; 21:286. [PMID: 34294077 PMCID: PMC8298196 DOI: 10.1186/s12883-021-02308-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 07/01/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Culminating evidence shows that current care does not optimally meet the needs of persons with parkinsonism, their carers and healthcare professionals. Recently, a new model of care was developed to address the limitations of usual care: Proactive and Integrated Management and Empowerment in Parkinson's Disease (PRIME Parkinson). From 2021 onwards, PRIME Parkinson care will replace usual care in a well-defined region in The Netherlands. The utility of PRIME Parkinson care will be evaluated on a single primary endpoint (parkinsonism-related complications), which reflects the health of people with parkinsonism. Furthermore, several secondary endpoints will be measured for four dimensions: health, patient and carer experience, healthcare professional experience, and cost of healthcare. The reference will be usual care, which will be continued in other regions in The Netherlands. METHODS This is a prospective observational study which will run from January 1, 2020 until December 31, 2023. Before the new model of care will replace the usual care in the PRIME Parkinson care region all baseline assessments will take place. Outcomes will be informed by two data sources. We will use healthcare claims-based data to evaluate the primary endpoint, and costs of healthcare, in all persons with parkinsonism receiving PRIME Parkinson care (estimated number: 2,000) and all persons with parkinsonism receiving usual care in the other parts of The Netherlands (estimated number: 48,000). We will also evaluate secondary endpoints by performing annual questionnaire-based assessments. These assessments will be administered to a subsample across both regions (estimated numbers: 1,200 persons with parkinsonism, 600 carers and 250 healthcare professionals). DISCUSSION This prospective cohort study will evaluate the utility of a novel integrated model of care for persons with parkinsonism in The Netherlands. We anticipate that the results of this study will also provide insight for the delivery of care to persons with parkinsonism in other regions and may inform the design of a similar model for other chronic health conditions.
Collapse
Affiliation(s)
- Jan H L Ypinga
- Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behaviour, Department of Neurology, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
| | - Angelika D Van Halteren
- Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behaviour, Department of Neurology, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Emily J Henderson
- Department of Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 1NU, UK
- Older People's Unit, Royal United Hospitals Bath NHS Foundation Trust, Combe Park, Bath, UK
| | - Bastiaan R Bloem
- Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behaviour, Department of Neurology, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Agnes J Smink
- Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behaviour, Department of Neurology, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Emma Tenison
- Department of Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 1NU, UK
| | - Marten Munneke
- Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behaviour, Department of Neurology, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Yoav Ben-Shlomo
- Department of Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 1NU, UK
| | - Sirwan K L Darweesh
- Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behaviour, Department of Neurology, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| |
Collapse
|