1
|
Jeng AC, Sibley IJ, Bale TL. A global perspective on AI innovation and effective use in the research lab. Neuroscience 2024; 560:106-108. [PMID: 39307414 DOI: 10.1016/j.neuroscience.2024.09.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024]
Affiliation(s)
- Alyssa C Jeng
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Isabelle J Sibley
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Tracy L Bale
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
2
|
Jafari E, Blackman MH, Karnes JH, Van Driest SL, Crawford DC, Choi L, McDonough CW. Using electronic health records for clinical pharmacology research: Challenges and considerations. Clin Transl Sci 2024; 17:e13871. [PMID: 38943244 PMCID: PMC11213823 DOI: 10.1111/cts.13871] [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: 03/19/2024] [Revised: 05/21/2024] [Accepted: 05/24/2024] [Indexed: 07/01/2024] Open
Abstract
Electronic health records (EHRs) contain a vast array of phenotypic data on large numbers of individuals, often collected over decades. Due to the wealth of information, EHR data have emerged as a powerful resource to make first discoveries and identify disparities in our healthcare system. While the number of EHR-based studies has exploded in recent years, most of these studies are directed at associations with disease rather than pharmacotherapeutic outcomes, such as drug response or adverse drug reactions. This is largely due to challenges specific to deriving drug-related phenotypes from the EHR. There is great potential for EHR-based discovery in clinical pharmacology research, and there is a critical need to address specific challenges related to accurate and reproducible derivation of drug-related phenotypes from the EHR. This review provides a detailed evaluation of challenges and considerations for deriving drug-related data from EHRs. We provide an examination of EHR-based computable phenotypes and discuss cutting-edge approaches to map medication information for clinical pharmacology research, including medication-based computable phenotypes and natural language processing. We also discuss additional considerations such as data structure, heterogeneity and missing data, rare phenotypes, and diversity within the EHR. By further understanding the complexities associated with conducting clinical pharmacology research using EHR-based data, investigators will be better equipped to design thoughtful studies with more reproducible results. Progress in utilizing EHRs for clinical pharmacology research should lead to significant advances in our ability to understand differential drug response and predict adverse drug reactions.
Collapse
Affiliation(s)
- Eissa Jafari
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics and Precision Medicine, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
- Department of Pharmacy Practice, College of PharmacyJazan UniversityJazanSaudi Arabia
| | - Marisa H. Blackman
- Department of BiostatisticsVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Jason H. Karnes
- Department of Pharmacy Practice and ScienceUniversity of Arizona R. Ken Coit College of PharmacyTucsonArizonaUSA
| | - Sara L. Van Driest
- Department of PediatricsVanderbilt University Medical Center (VUMC)NashvilleTennesseeUSA
- Present address:
All of US Research Program, National Institutes of HealthBethesdaMarylandUSA
| | - Dana C. Crawford
- Department of Population and Quantitative Health Sciences, Cleveland Institute for Computational BiologyCase Western Reserve UniversityClevelandOhioUSA
- Department of Genetics and Genome Sciences, Cleveland Institute for Computational BiologyCase Western Reserve UniversityClevelandOhioUSA
| | - Leena Choi
- Department of Biostatistics and Biomedical InformaticsVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Caitrin W. McDonough
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics and Precision Medicine, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| |
Collapse
|
3
|
Salem AM, Smith T, Wilkes J, Bailly DK, Heyrend C, Profsky M, Yellepeddi VK, Gopalakrishnan M. Pharmacokinetic Modeling Using Real-World Data to Optimize Unfractionated Heparin Dosing in Pediatric Patients on Extracorporeal Membrane Oxygenation and Evaluate Target Achievement-Clinical Outcomes Relationship. J Clin Pharmacol 2024; 64:30-44. [PMID: 37565528 DOI: 10.1002/jcph.2333] [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: 04/19/2023] [Accepted: 08/07/2023] [Indexed: 08/12/2023]
Abstract
Unfractionated heparin (UFH) is a commonly used anticoagulant for pediatric patients undergoing extracorporeal membrane oxygenation (ECMO), but evidence is lacking on the ideal dosing. We aimed to (1) develop a population pharmacokinetic (PK) model for UFH, measured through anti-factor Xa assay; (2) optimize UFH starting infusions and dose titrations through simulations; and (3) explore UFH exposure-clinical outcomes relationship. Data from 218 patients admitted to Utah's Primary Children's Hospital were retrospectively collected. A 1-compartment PK model with time-varying clearance (CL) adequately described UFH PK. Weight on CL and volume of distribution and ECMO circuit change on CL were significant covariates. The typical estimates for initial CL and first-order rate constant to reach steady-state CL were 0.57 L/(h·10 kg) and 0.02/h. Comparable to non-ECMO patients, the typical steady-state CL was 0.81 L/(h·10 kg). Simulations showed that a 75 IU/kg UFH bolus dose followed by starting infusions of 25 and 20 IU/h/kg for patients aged younger than 6 years and 6 years or older, respectively, achieved the therapeutic target in 56.6% of all patients, whereas only 3.1% exceeded the target. The proposed UFH titration schemes achieved the target in more than 90% of patients while less than 0.63% were above the target after 24 and 48 hours of treatment. The median intensive care unit survival time in patients within and below the target at 24 hours was 136 and 66 hours, respectively. In conclusion, PK model of UFH was developed for pediatric patients on ECMO. The proposed UFH dosing scheme attained the anti-factor Xa target rapidly and safely.
Collapse
Affiliation(s)
- Ahmed M Salem
- Center for Translational Medicine, Department of Pharmacy Practice, University of Maryland School of Pharmacy, Baltimore, MD, USA
| | - Trey Smith
- Department of Pharmacy, Primary Children's Hospital, Intermountain Healthcare, Salt Lake City, UT, USA
| | - Jacob Wilkes
- Pediatric Analytics, Primary Children's Hospital, Intermountain Healthcare, Salt Lake City, UT, USA
| | - David K Bailly
- Division of Pediatric Critical Care, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Caroline Heyrend
- Department of Pharmacy, Primary Children's Hospital, Intermountain Healthcare, Salt Lake City, UT, USA
| | - Michael Profsky
- Mechanical Circulatory Support, Primary Children's Hospital, Intermountain Healthcare, Salt Lake City, UT, USA
| | - Venkata K Yellepeddi
- Division of Clinical Pharmacology, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT, USA
- Department of Molecular Pharmaceutics, College of Pharmacy, University of Utah, Salt Lake City, UT, USA
| | - Mathangi Gopalakrishnan
- Center for Translational Medicine, Department of Pharmacy Practice, University of Maryland School of Pharmacy, Baltimore, MD, USA
| |
Collapse
|
4
|
Thompson EJ, Foote HP, Hill KD, Hornik CP. A point-of-care pharmacokinetic/pharmacodynamic trial in critically ill children: Study design and feasibility. Contemp Clin Trials Commun 2023; 35:101182. [PMID: 37485397 PMCID: PMC10362170 DOI: 10.1016/j.conctc.2023.101182] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 06/01/2023] [Accepted: 07/02/2023] [Indexed: 07/25/2023] Open
Abstract
Background High-quality, efficient, pharmacokinetic (PK), pharmacodynamic (PD), and safety studies in children are needed. Point-of-care trials in adults have facilitated clinical trial participation for patients and providers, minimized the disruption of clinical workflow, and capitalized on routine data collection. The feasibility and value of point-of-care trials to study PK/PD in children are unknown, but appear promising. The Opportunistic PK/PD Trial in Critically Ill Children with Heart Disease (OPTIC) is a programmatic point-of-care approach to PK/PD trials in critically ill children that seeks to overcome barriers of traditional pediatric PK/PD studies to generate safety, efficacy, PK, and PD data across multiple medications, ages, and disease processes. Methods This prospective, open-label, non-randomized point-of-care trial will characterize the PK/PD and safety of multiple drugs given per routine care to critically ill children with heart disease using opportunistic and scavenged biospecimen samples and data collected from the electronic health record. OPTIC has one informed consent form with drug-specific appendices, streamlining study structure and institutional review board approval. OPTIC capitalizes on routine data collection through multiple data sources that automatically capture demographics, medications, laboratory values, vital signs, flowsheets, and other clinical data. This innovative automatic data collection minimizes the burden of data collection and facilitates trial conduct. Data will be validated across sources to ensure accuracy of dataset variables. Discussion OPTIC's point-of-care trial design and automated data acquisition via the electronic health record may provide a mechanism for conducting minimal risk, minimal burden, high efficiency trials and support drug development in historically understudied patient populations. Trial registration clinicaltrials.gov number: NCT05055830. Registered on September 24, 2021.
Collapse
Affiliation(s)
| | - Henry P. Foote
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Kevin D. Hill
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
- Duke Clinical Research Institute, Durham, NC, USA
| | - Christoph P. Hornik
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
- Duke Clinical Research Institute, Durham, NC, USA
| |
Collapse
|
5
|
Shannon ML, Muhammad A, James NT, Williams ML, Breeyear J, Edwards T, Mosley JD, Choi L, Kannankeril P, Van Driest S. Variant-based heritability assessment of dexmedetomidine and fentanyl clearance in pediatric patients. Clin Transl Sci 2023; 16:1628-1638. [PMID: 37353859 PMCID: PMC10499425 DOI: 10.1111/cts.13574] [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: 02/09/2023] [Revised: 04/12/2023] [Accepted: 06/01/2023] [Indexed: 06/25/2023] Open
Abstract
Despite complex pathways of drug disposition, clinical pharmacogenetic predictors currently rely on only a few high effect variants. Quantification of the polygenic contribution to variability in drug disposition is necessary to prioritize target drugs for pharmacogenomic approaches and guide analytic methods. Dexmedetomidine and fentanyl, often used in postoperative care of pediatric patients, have high rates of inter-individual variability in dosing requirements. Analyzing previously generated population pharmacokinetic parameters, we used Bayesian hierarchical mixed modeling to measure narrow-sense (additive) heritability (h SNP 2 ) of dexmedetomidine and fentanyl clearance in children and identify relative contributions of small, moderate, and large effect-size variants toh SNP 2 . We used genome-wide association studies (GWAS) to identify variants contributing to variation in dexmedetomidine and fentanyl clearance, followed by functional analyses to identify associated pathways. For dexmedetomidine, median clearance was 33.0 L/h (interquartile range [IQR] 23.8-47.9 L/h) andh SNP 2 was estimated to be 0.35 (90% credible interval 0.00-0.90), with 45% ofh SNP 2 attributed to large-, 32% to moderate-, and 23% to small-effect variants. The fentanyl cohort had median clearance of 8.2 L/h (IQR 4.7-16.7 L/h), with estimatedh SNP 2 of 0.30 (90% credible interval 0.00-0.84). Large-effect variants accounted for 30% ofh SNP 2 , whereas moderate- and small-effect variants accounted for 37% and 33%, respectively. As expected, given small sample sizes, no individual variants or pathways were significantly associated with dexmedetomidine or fentanyl clearance by GWAS. We conclude that clearance of both drugs is highly polygenic, motivating the future use of polygenic risk scores to guide appropriate dosing of dexmedetomidine and fentanyl.
Collapse
Affiliation(s)
| | - Ayesha Muhammad
- School of MedicineVanderbilt UniversityNashvilleTennesseeUSA
| | - Nathan T. James
- Department of BiostatisticsVanderbilt University Medical CenterNashvilleTennesseeUSA
- Present address:
Berry Consultants, LLCAustinTexasUSA
| | - Michael L. Williams
- Department of BiostatisticsVanderbilt University Medical CenterNashvilleTennesseeUSA
- Present address:
Department of Clinical Pharmacology and Quantitative PharmacologyAstraZenecaGothenburgSweden
| | - Joseph Breeyear
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Todd Edwards
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Jonathan D. Mosley
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
- Department of Biomedical InformaticsVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Leena Choi
- Department of BiostatisticsVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Prince Kannankeril
- Center for Pediatric Precision Medicine, Department of PediatricsVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Sara Van Driest
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
- Center for Pediatric Precision Medicine, Department of PediatricsVanderbilt University Medical CenterNashvilleTennesseeUSA
- Present address:
All of Us Research ProgramNational Institutes of HealthWashingtonDCUSA
| |
Collapse
|
6
|
Salem AM, Dvergsten E, Karovic S, Maitland ML, Gopalakrishnan M. Model-based approach to identify predictors of paclitaxel-induced myelosuppression in "real-world" administration. CPT Pharmacometrics Syst Pharmacol 2023; 12:929-940. [PMID: 37101403 PMCID: PMC10349185 DOI: 10.1002/psp4.12963] [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: 11/19/2022] [Revised: 03/11/2023] [Accepted: 03/22/2023] [Indexed: 04/28/2023] Open
Abstract
Taxanes are currently the most frequently used chemotherapeutic agents in cancer care, where real-world use has focused on minimizing adverse events and standardizing the delivery. Myelosuppression is a well-characterized, adverse pharmacodynamic effect of taxanes. Electronic health records (EHRs) comprise data collected during routine clinical care that include patients with heterogeneous demographic, clinical, and treatment characteristics. Application of pharmacokinetic/pharmacodynamic (PK/PD) modeling to EHR data promises new insights on the real-world use of taxanes and strategies to improve therapeutic outcomes especially for populations who are typically excluded from clinical trials, including the elderly. This investigation: (i) leveraged previously published PK/PD models developed with clinical trial data and addressed challenges to fit EHR data, and (ii) evaluated predictors of paclitaxel-induced myelosuppression. Relevant EHR data were collected from patients treated with paclitaxel-containing chemotherapy at Inova Schar Cancer Institute between 2015 and 2019 (n = 405). Published PK models were used to simulate mean individual exposures of paclitaxel and carboplatin, which were linearly linked to absolute neutrophil count (ANC) using a published semiphysiologic myelosuppression model. Elderly patients (≥70 years) constituted 21.2% of the dataset and 2274 ANC measurements were included in the analysis. The PD parameters were estimated and matched previously reported values. The baseline ANC and chemotherapy regimen were significant predictors of paclitaxel-induced myelosuppression. The nadir ANC and use of supportive treatments, such as growth factors and antimicrobials, were consistent across age quantiles suggesting age had no effect on paclitaxel-induced myelosuppression. In conclusion, EHR data could complement clinical trial data in answering key therapeutic questions.
Collapse
Affiliation(s)
- Ahmed M. Salem
- Center for Translational MedicineUniversity of Maryland School of PharmacyBaltimoreMarylandUSA
| | | | | | - Michael L. Maitland
- Inova Schar Cancer InstituteFairfaxVirginiaUSA
- University of Virginia Comprehensive Cancer CenterCharlottesvilleVirginiaUSA
| | - Mathangi Gopalakrishnan
- Center for Translational MedicineUniversity of Maryland School of PharmacyBaltimoreMarylandUSA
| |
Collapse
|
7
|
Abler D, Courlet P, Dietz M, Gatta R, Girard P, Munafo A, Wicky A, Jreige M, Guidi M, Latifyan S, De Micheli R, Csajka C, Prior JO, Michielin O, Terranova N, Cuendet MA. Semiautomated Pipeline to Quantify Tumor Evolution From Real-World Positron Emission Tomography/Computed Tomography Imaging. JCO Clin Cancer Inform 2023; 7:e2200126. [PMID: 37146261 PMCID: PMC10281365 DOI: 10.1200/cci.22.00126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 11/04/2022] [Accepted: 02/03/2023] [Indexed: 05/07/2023] Open
Abstract
PURPOSE A semiautomated pipeline for the collection and curation of free-text and imaging real-world data (RWD) was developed to quantify cancer treatment outcomes in large-scale retrospective real-world studies. The objectives of this article are to illustrate the challenges of RWD extraction, to demonstrate approaches for quality assurance, and to showcase the potential of RWD for precision oncology. METHODS We collected data from patients with advanced melanoma receiving immune checkpoint inhibitors at the Lausanne University Hospital. Cohort selection relied on semantically annotated electronic health records and was validated using process mining. The selected imaging examinations were segmented using an automatic commercial software prototype. A postprocessing algorithm enabled longitudinal lesion identification across imaging time points and consensus malignancy status prediction. Resulting data quality was evaluated against expert-annotated ground-truth and clinical outcomes obtained from radiology reports. RESULTS The cohort included 108 patients with melanoma and 465 imaging examinations (median, 3; range, 1-15 per patient). Process mining was used to assess clinical data quality and revealed the diversity of care pathways encountered in a real-world setting. Longitudinal postprocessing greatly improved the consistency of image-derived data compared with single time point segmentation results (classification precision increased from 53% to 86%). Image-derived progression-free survival resulting from postprocessing was comparable with the manually curated clinical reference (median survival of 286 v 336 days, P = .89). CONCLUSION We presented a general pipeline for the collection and curation of text- and image-based RWD, together with specific strategies to improve reliability. We showed that the resulting disease progression measures match reference clinical assessments at the cohort level, indicating that this strategy has the potential to unlock large amounts of actionable retrospective real-world evidence from clinical records.
Collapse
Affiliation(s)
- Daniel Abler
- Department of Oncology, Precision Oncology Center, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Institute of Informatics, School of Management, University of Applied Sciences Western Switzerland (HES-SO), Sierre, Switzerland
| | - Perrine Courlet
- Department of Oncology, Precision Oncology Center, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Centre for Research and Innovation in Clinical Pharmaceutical Sciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Matthieu Dietz
- Nuclear Medicine and Molecular Imaging Department, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- INSERM U1060, CarMeN Laboratory, University of Lyon, Lyon, France
| | - Roberto Gatta
- Department of Oncology, Precision Oncology Center, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Pascal Girard
- Translational Medicine, Merck Institute of Pharmacometrics, Lausanne, Switzerland, an Affiliate of Merck KGaA, Darmstadt, Germany
| | - Alain Munafo
- Translational Medicine, Merck Institute of Pharmacometrics, Lausanne, Switzerland, an Affiliate of Merck KGaA, Darmstadt, Germany
| | - Alexandre Wicky
- Department of Oncology, Precision Oncology Center, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Mario Jreige
- Nuclear Medicine and Molecular Imaging Department, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Monia Guidi
- Centre for Research and Innovation in Clinical Pharmaceutical Sciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Service of Clinical Pharmacology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Sofiya Latifyan
- Service of Medical Oncology, Department of Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Rita De Micheli
- Service of Medical Oncology, Department of Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Chantal Csajka
- Centre for Research and Innovation in Clinical Pharmaceutical Sciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, University of Lausanne, Geneva, Switzerland
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
| | - John O. Prior
- Nuclear Medicine and Molecular Imaging Department, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Olivier Michielin
- Department of Oncology, Precision Oncology Center, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Nadia Terranova
- Translational Medicine, Merck Institute of Pharmacometrics, Lausanne, Switzerland, an Affiliate of Merck KGaA, Darmstadt, Germany
| | - Michel A. Cuendet
- Department of Oncology, Precision Oncology Center, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, University of Lausanne, Lausanne, Switzerland
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY
| |
Collapse
|
8
|
Williams ML, Weeks HL, Beck C, Birdwell KA, Van Driest SL, Choi L. Sensitivity of estimated tacrolimus population pharmacokinetic profile to assumed dose timing and absorption in real-world data and simulated data. Br J Clin Pharmacol 2022; 88:2863-2874. [PMID: 34997625 PMCID: PMC9106813 DOI: 10.1111/bcp.15218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 12/21/2021] [Accepted: 12/22/2021] [Indexed: 11/28/2022] Open
Abstract
AIMS Use of electronic health record (EHR) data to estimate population pharmacokinetic (PK) profiles necessitates several assumptions. We sought to investigate sensitivity to some of these assumptions about dose timing and absorption rates. METHODS A population PK study with 363 subjects was performed using real-world data extracted from EHRs to estimate the tacrolimus population PK profile. Data were extracted and built using our automated system, EHR2PKPD, suitable for quickly constructing large PK datasets from the EHR. Population PK studies for oral medications performed using EHR data often assume a regular dosing schedule as prescribed without incorporating exact dosing time. We assessed the sensitivity of the PK parameter estimates to assumptions about dose timing using last-dose times extracted by our own natural language processing system, medExtractR. We also investigated the sensitivity of estimates to absorption rate constants that are often fixed at a published value in tacrolimus population PK analyses. We conducted simulation studies to investigate how drug PK profiles and experimental designs such as concentration measurements design affect sensitivity to incorrect assumptions about dose timing and absorption rates. RESULTS There was no appreciable difference in parameter estimates with assumed versus extracted last-dose time, and our sensitivity analysis revealed little difference between parameters estimated across a range of assumed absorption rate constants. CONCLUSION Our findings suggest that drugs with a slower elimination rate (or a longer half-life) are less sensitive to dose timing errors and that experimental designs which only allow for trough blood concentrations are usually insensitive to deviation in absorption rate.
Collapse
Affiliation(s)
- Michael L. Williams
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN
| | - Hannah L. Weeks
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN
| | - Cole Beck
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN
| | - Kelly A. Birdwell
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Sara L. Van Driest
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| | - Leena Choi
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
9
|
James NT, Breeyear JH, Caprioli R, Edwards T, Hachey B, Kannankeril PJ, Keaton JM, Marshall MD, Van Driest SL, Choi L. Population pharmacokinetic analysis of dexmedetomidine in children using real-world data from electronic health records and remnant specimens. Br J Clin Pharmacol 2022; 88:2885-2898. [PMID: 34957589 PMCID: PMC9106818 DOI: 10.1111/bcp.15194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 11/18/2021] [Accepted: 12/14/2021] [Indexed: 11/30/2022] Open
Abstract
AIMS Our objectives were to perform a population pharmacokinetic analysis of dexmedetomidine in children using remnant specimens and electronic health records (EHRs) and explore the impact of patient's characteristics and pharmacogenetics on dexmedetomidine clearance. METHODS Dexmedetomidine dosing and patient data were gathered from EHRs and combined with opportunistically sampled remnant specimens. Population pharmacokinetic models were developed using nonlinear mixed-effects modelling. Stage 1 developed a model without genotype variables; Stage 2 added pharmacogenetic effects. RESULTS Our final study population included 354 post-cardiac surgery patients aged 0-22 years (median 16 mo). The data were best described with a 2-compartment model with allometric scaling for weight and Hill maturation function for age. Population parameter estimates and 95% confidence intervals were 27.3 L/h (24.0-31.1 L/h) for total clearance, 161 L (139-187 L) for central compartment volume of distribution, 26.0 L/h (22.5-30.0 L/h) for intercompartmental clearance and 7903 L (5617-11 119 L) for peripheral compartment volume of distribution. The estimate for postmenstrual age when 50% of adult clearance is achieved was 42.0 weeks (41.5-42.5 weeks) and the Hill coefficient estimate was 7.04 (6.99-7.08). Genotype was not statistically or clinically significant. CONCLUSION Our study demonstrates the use of real-world EHR data and remnant specimens to perform a population pharmacokinetic analysis and investigate covariate effects in a large paediatric population. Weight and age were important predictors of clearance. We did not find evidence for pharmacogenetic effects of UGT1A4 or UGT2B10 genotype or CYP2A6 risk score.
Collapse
Affiliation(s)
- Nathan T. James
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN
| | - Joseph H. Breeyear
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Richard Caprioli
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Todd Edwards
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Brian Hachey
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Prince J. Kannankeril
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
- Center for Pediatric Precision Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Jacob M. Keaton
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Matthew D. Marshall
- Department of Pharmaceutical Services, Vanderbilt University Medical Center, Nashville, TN
| | - Sara L. Van Driest
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
- Center for Pediatric Precision Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Leena Choi
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
10
|
Bhatnagar R, Sardar S, Beheshti M, Podichetty JT. How can natural language processing help model informed drug development?: a review. JAMIA Open 2022; 5:ooac043. [PMID: 35702625 PMCID: PMC9188322 DOI: 10.1093/jamiaopen/ooac043] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/28/2022] [Accepted: 05/26/2022] [Indexed: 01/20/2023] Open
Abstract
Objective To summarize applications of natural language processing (NLP) in model informed drug development (MIDD) and identify potential areas of improvement. Materials and Methods Publications found on PubMed and Google Scholar, websites and GitHub repositories for NLP libraries and models. Publications describing applications of NLP in MIDD were reviewed. The applications were stratified into 3 stages: drug discovery, clinical trials, and pharmacovigilance. Key NLP functionalities used for these applications were assessed. Programming libraries and open-source resources for the implementation of NLP functionalities in MIDD were identified. Results NLP has been utilized to aid various processes in drug development lifecycle such as gene-disease mapping, biomarker discovery, patient-trial matching, adverse drug events detection, etc. These applications commonly use NLP functionalities of named entity recognition, word embeddings, entity resolution, assertion status detection, relation extraction, and topic modeling. The current state-of-the-art for implementing these functionalities in MIDD applications are transformer models that utilize transfer learning for enhanced performance. Various libraries in python, R, and Java like huggingface, sparkNLP, and KoRpus as well as open-source platforms such as DisGeNet, DeepEnroll, and Transmol have enabled convenient implementation of NLP models to MIDD applications. Discussion Challenges such as reproducibility, explainability, fairness, limited data, limited language-support, and security need to be overcome to ensure wider adoption of NLP in MIDD landscape. There are opportunities to improve the performance of existing models and expand the use of NLP in newer areas of MIDD. Conclusions This review provides an overview of the potential and pitfalls of current NLP approaches in MIDD.
Collapse
Affiliation(s)
- Roopal Bhatnagar
- Data Science, Data Collaboration Center, Critical Path Institute , Tucson, Arizona, USA
| | - Sakshi Sardar
- Quantitative Medicine, Critical Path Institute , Tucson, Arizona, USA
| | - Maedeh Beheshti
- Quantitative Medicine, Critical Path Institute , Tucson, Arizona, USA
| | | |
Collapse
|
11
|
Martínez-García M, Hernández-Lemus E. Data Integration Challenges for Machine Learning in Precision Medicine. Front Med (Lausanne) 2022; 8:784455. [PMID: 35145977 PMCID: PMC8821900 DOI: 10.3389/fmed.2021.784455] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/28/2021] [Indexed: 12/19/2022] Open
Abstract
A main goal of Precision Medicine is that of incorporating and integrating the vast corpora on different databases about the molecular and environmental origins of disease, into analytic frameworks, allowing the development of individualized, context-dependent diagnostics, and therapeutic approaches. In this regard, artificial intelligence and machine learning approaches can be used to build analytical models of complex disease aimed at prediction of personalized health conditions and outcomes. Such models must handle the wide heterogeneity of individuals in both their genetic predisposition and their social and environmental determinants. Computational approaches to medicine need to be able to efficiently manage, visualize and integrate, large datasets combining structure, and unstructured formats. This needs to be done while constrained by different levels of confidentiality, ideally doing so within a unified analytical architecture. Efficient data integration and management is key to the successful application of computational intelligence approaches to medicine. A number of challenges arise in the design of successful designs to medical data analytics under currently demanding conditions of performance in personalized medicine, while also subject to time, computational power, and bioethical constraints. Here, we will review some of these constraints and discuss possible avenues to overcome current challenges.
Collapse
Affiliation(s)
- Mireya Martínez-García
- Clinical Research Division, National Institute of Cardiology ‘Ignacio Chávez’, Mexico City, Mexico
| | - Enrique Hernández-Lemus
- Computational Genomics Division, National Institute of Genomic Medicine (INMEGEN), Mexico City, Mexico
- Center for Complexity Sciences, Universidad Nacional Autnoma de Mexico, Mexico City, Mexico
| |
Collapse
|
12
|
Crema C, Attardi G, Sartiano D, Redolfi A. Natural language processing in clinical neuroscience and psychiatry: A review. Front Psychiatry 2022; 13:946387. [PMID: 36186874 PMCID: PMC9515453 DOI: 10.3389/fpsyt.2022.946387] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
Natural language processing (NLP) is rapidly becoming an important topic in the medical community. The ability to automatically analyze any type of medical document could be the key factor to fully exploit the data it contains. Cutting-edge artificial intelligence (AI) architectures, particularly machine learning and deep learning, have begun to be applied to this topic and have yielded promising results. We conducted a literature search for 1,024 papers that used NLP technology in neuroscience and psychiatry from 2010 to early 2022. After a selection process, 115 papers were evaluated. Each publication was classified into one of three categories: information extraction, classification, and data inference. Automated understanding of clinical reports in electronic health records has the potential to improve healthcare delivery. Overall, the performance of NLP applications is high, with an average F1-score and AUC above 85%. We also derived a composite measure in the form of Z-scores to better compare the performance of NLP models and their different classes as a whole. No statistical differences were found in the unbiased comparison. Strong asymmetry between English and non-English models, difficulty in obtaining high-quality annotated data, and train biases causing low generalizability are the main limitations. This review suggests that NLP could be an effective tool to help clinicians gain insights from medical reports, clinical research forms, and more, making NLP an effective tool to improve the quality of healthcare services.
Collapse
Affiliation(s)
- Claudio Crema
- Laboratory of Neuroinformatics, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | | | - Daniele Sartiano
- Istituto di Informatica e Telematica, Consiglio Nazionale delle Ricerche, Pisa, Italy
| | - Alberto Redolfi
- Laboratory of Neuroinformatics, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| |
Collapse
|
13
|
Williams ML, Kannankeril PJ, Breeyear JH, Edwards TL, Van Driest SL, Choi L. Effect of CYP3A5 and CYP3A4 Genetic Variants on Fentanyl Pharmacokinetics in a Pediatric Population. Clin Pharmacol Ther 2021; 111:896-908. [PMID: 34877660 DOI: 10.1002/cpt.2506] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 12/03/2021] [Indexed: 12/30/2022]
Abstract
Fentanyl is an anesthetic/analgesic commonly used in surgical and recovery settings. CYP3A4 and CYP3A5 encode enzymes, which metabolize fentanyl; genetic variants in these genes impact fentanyl pharmacokinetics in adults. Pharmacokinetic (PK) studies are difficult to replicate in children due to the burden of additional blood taken solely for research purposes. The aim of this study is to test the effect of CYP3A5 and CYP3A4 genetic variants on fentanyl PKs in children using opportunistically collected samples. Fentanyl concentrations were measured from remnant blood specimens and dosing data were extracted from electronic health records. Variant data defining CYP3A4*1G and CYP3A5*3 and *6 alleles were available from prior genotyping; alleles with no variant were defined as *1. The study cohort included 434 individuals (median age 9 months, 52% male subjects) and 1,937 fentanyl concentrations were available. A two-compartment model was selected as the base model, and the final covariate model included age, weight, and surgical severity score. Clearance was significantly associated with either CYP3A5*3 or CYP3A5*6 alleles, but not the CYP3A4*1G allele. A genotype of CYP3A5*1/*3 or CYP3A5*1/*6 (i.e., intermediate metabolizer status) was associated with a 0.84-fold (95% confidence interval (CI): 0.71-1.00) reduction in clearance vs. CYP3A5*1/*1 (i.e., normal metabolizer status). CYP3A5*3/*3, CYP3A5*3/*6, or CYP3A5*6/*6 (i.e., poor metabolizer status) was associated with a 0.76-fold (95% CI: 0.58-0.99) reduction in clearance. In the final model, expected clearance was 8.9 and 6.8 L/hour for a normal and poor metabolizer, respectively, with median population covariates (9 months old, 7.7 kg, low surgical severity).
Collapse
Affiliation(s)
- Michael L Williams
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Prince J Kannankeril
- Center for Pediatric Precision Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Joseph H Breeyear
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Todd L Edwards
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Sara L Van Driest
- Center for Pediatric Precision Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Leena Choi
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
14
|
Soeorg H, Sverrisdóttir E, Andersen M, Lund TM, Sessa M. The PHARMACOM-EPI Framework for Integrating Pharmacometric Modelling Into Pharmacoepidemiological Research Using Real-World Data: Application to Assess Death Associated With Valproate. Clin Pharmacol Ther 2021; 111:840-856. [PMID: 34860420 DOI: 10.1002/cpt.2502] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 11/17/2021] [Indexed: 01/14/2023]
Abstract
In pharmacoepidemiology, it is usually expected that the observed association should be directly or indirectly related to the pharmacological effects of the drug/s under investigation. Pharmacological effects are, in turn, strongly connected to the pharmacokinetic and pharmacodynamic properties of a drug, which can be characterized and investigated using pharmacometric models. Recently, the use of pharmacometrics has been proposed to provide pharmacological substantiation of pharmacoepidemiological findings derived from real-world data. However, validated frameworks suggesting how to combine these two disciplines for the aforementioned purpose are missing. Therefore, we propose PHARMACOM-EPI, a framework that provides a structured approach on how to identify, characterize, and apply pharmacometric models with practical details on how to choose software, format dataset, handle missing covariates/dosing data, how to perform the external evaluation of pharmacometric models in real-world data, and how to provide pharmacological substantiation of pharmacoepidemiological findings. PHARMACOM-EPI was tested in a proof-of-concept study to pharmacologically substantiate death associated with valproate use in the Danish population aged ≥ 65 years. Pharmacological substantiation of death during a follow-up period of 1 year showed that in all individuals who died (n = 169) individual predictions were within the subtherapeutic range compared with 52.8% of those who did not die (n = 1,084). Of individuals who died, 66.3% (n = 112) had a cause of death possibly related to valproate and 33.7% (n = 57) with well-defined cause of death unlikely related to valproate. This proof-of-concept study showed that PHARMACOM-EPI was able to provide pharmacological substantiation for death associated with valproate use in the study population.
Collapse
Affiliation(s)
- Hiie Soeorg
- Department of Drug Design and Pharmacology, Pharmacovigilance Research Center, University of Copenhagen, Copenhagen, Denmark.,Department of Drug Design and Pharmacology, Pharmacometrics Research Group, University of Copenhagen, Copenhagen, Denmark
| | - Eva Sverrisdóttir
- Department of Drug Design and Pharmacology, Pharmacometrics Research Group, University of Copenhagen, Copenhagen, Denmark
| | - Morten Andersen
- Department of Drug Design and Pharmacology, Pharmacovigilance Research Center, University of Copenhagen, Copenhagen, Denmark
| | - Trine Meldgaard Lund
- Department of Drug Design and Pharmacology, Pharmacometrics Research Group, University of Copenhagen, Copenhagen, Denmark
| | - Maurizio Sessa
- Department of Drug Design and Pharmacology, Pharmacovigilance Research Center, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
15
|
Bejan CA, Cahill KN, Staso PJ, Choi L, Peterson JF, Phillips EJ. DrugWAS: Drug-wide Association Studies for COVID-19 Drug Repurposing. Clin Pharmacol Ther 2021; 110:1537-1546. [PMID: 34314511 PMCID: PMC8426999 DOI: 10.1002/cpt.2376] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 07/21/2021] [Indexed: 11/16/2022]
Abstract
This study aimed to systematically investigate if any of the available drugs in the electronic health record (EHR) can be repurposed as potential treatment for coronavirus disease 2019 (COVID-19). Based on a retrospective cohort analysis of EHR data, drug-wide association studies (DrugWAS) were performed on 9,748 patients with COVID-19 at Vanderbilt University Medical Center (VUMC). For each drug study, multivariable logistic regression with overlap weighting using propensity score was applied to estimate the effect of drug exposure on COVID-19 disease outcomes. Patient exposure to a drug between 3-months prior to the pandemic and the COVID-19 diagnosis was chosen as the exposure of interest. All-cause of death was selected as the primary outcome. Hospitalization, admission to the intensive care unit, and need for mechanical ventilation were identified as secondary outcomes. Overall, 17 drugs were significantly associated with decreased COVID-19 severity. Previous exposure to two types of 13-valent pneumococcal conjugate vaccines, PCV13 (odds ratio (OR), 0.31, 95% confidence interval (CI), 0.12-0.81 and OR, 0.33, 95% CI, 0.15-0.73), diphtheria toxoid and tetanus toxoid vaccine (OR, 0.38, 95% CI, 0.15-0.93) were significantly associated with a decreased risk of death (primary outcome). Secondary analyses identified several other significant associations showing lower risk for COVID-19 outcomes: acellular pertussis vaccine, 23-valent pneumococcal polysaccharide vaccine (PPSV23), flaxseed extract, ethinyl estradiol, estradiol, turmeric extract, ubidecarenone, azelastine, pseudoephedrine, dextromethorphan, omega-3 fatty acids, fluticasone, and ibuprofen. In conclusion, this cohort study leveraged EHR data to identify a list of drugs that could be repurposed to improve COVID-19 outcomes. Further randomized clinical trials are needed to investigate the efficacy of the proposed drugs.
Collapse
Affiliation(s)
- Cosmin A. Bejan
- Department of Biomedical InformaticsVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Katherine N. Cahill
- Department of MedicineDivision of Allergy, Pulmonary and Critical Care MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Patrick J. Staso
- Department of MedicineDivision of Allergy, Pulmonary and Critical Care MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Leena Choi
- Department of BiostatisticsVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Josh F. Peterson
- Department of Biomedical InformaticsVanderbilt University Medical CenterNashvilleTennesseeUSA
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Elizabeth J. Phillips
- Department of Pathology, Microbiology and ImmunologyVanderbilt University Medical CenterNashvilleTennesseeUSA
- Department of MedicineDivision of Infectious DiseasesVanderbilt University Medical CenterNashvilleTennesseeUSA
- Department of PharmacologyVanderbilt University Medical CenterNashvilleTennesseeUSA
| |
Collapse
|
16
|
Franchetti Y. Use of Propensity Scoring and Its Application to Real-World Data: Advantages, Disadvantages, and Methodological Objectives Explained to Researchers Without Using Mathematical Equations. J Clin Pharmacol 2021; 62:304-319. [PMID: 34671990 DOI: 10.1002/jcph.1989] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 10/17/2021] [Indexed: 12/28/2022]
Abstract
Real-time data collection of patient health status and medications is sped up with modern electronic devices and technologies. As real-world data provide enormous research opportunities, propensity score (PS) methods have been getting attention due to their theoretical grounds in a nonrandomized study setting. In contrast to randomized clinical trials, observational clinical data obtained from a real-world database may not have balanced distributions of patient characteristics between treatment and control groups at the beginning of the respective study. These imbalanced distributions may cause a bias in an estimated treatment effect, which needs to be eliminated. Propensity scoring is one class of statistical methods to address the imbalance issue of real-world data sets. This article provides basic concepts and assesses advantages, disadvantages, and methodological objectives of propensity scoring. Targeting clinical pharmacology researchers with limited statistical background, 5 representative methods are reviewed and visualized: matching, stratification, covariate modeling, inverse probability of treatment weighting, and doubly robust methods. Examples of applications of PS methods were selected from the literature of outcomes research and drug development, nephrology, and pediatrics. Opportunities of applications related to these examples are described. Furthermore, potential future applications of PS methods in clinical pharmacology are discussed. The 21st Century Cures Act signed in 2016 encourages scientists to find opportunities to apply propensity scoring to real-world data. This article underscores that scientists need to justify their choice of statistical methods, whether a PS method or an alternative method, based on their clinical study design, statistical assumptions, and research objectives.
Collapse
|
17
|
Rogers JR, Lee J, Zhou Z, Cheung YK, Hripcsak G, Weng C. Contemporary use of real-world data for clinical trial conduct in the United States: a scoping review. J Am Med Inform Assoc 2021; 28:144-154. [PMID: 33164065 DOI: 10.1093/jamia/ocaa224] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/11/2020] [Accepted: 09/02/2020] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVE Real-world data (RWD), defined as routinely collected healthcare data, can be a potential catalyst for addressing challenges faced in clinical trials. We performed a scoping review of database-specific RWD applications within clinical trial contexts, synthesizing prominent uses and themes. MATERIALS AND METHODS Querying 3 biomedical literature databases, research articles using electronic health records, administrative claims databases, or clinical registries either within a clinical trial or in tandem with methodology related to clinical trials were included. Articles were required to use at least 1 US RWD source. All abstract screening, full-text screening, and data extraction was performed by 1 reviewer. Two reviewers independently verified all decisions. RESULTS Of 2020 screened articles, 89 qualified: 59 articles used electronic health records, 29 used administrative claims, and 26 used registries. Our synthesis was driven by the general life cycle of a clinical trial, culminating into 3 major themes: trial process tasks (51 articles); dissemination strategies (6); and generalizability assessments (34). Despite a diverse set of diseases studied, <10% of trials using RWD for trial process tasks evaluated medications or procedures (5/51). All articles highlighted data-related challenges, such as missing values. DISCUSSION Database-specific RWD have been occasionally leveraged for various clinical trial tasks. We observed underuse of RWD within conducted medication or procedure trials, though it is subject to the confounder of implicit report of RWD use. CONCLUSION Enhanced incorporation of RWD should be further explored for medication or procedure trials, including better understanding of how to handle related data quality issues to facilitate RWD use.
Collapse
Affiliation(s)
- James R Rogers
- Department of Biomedical Informatics, Columbia University, New York, New York, USA
| | - Junghwan Lee
- Department of Biomedical Informatics, Columbia University, New York, New York, USA
| | - Ziheng Zhou
- Institute of Human Nutrition, Columbia University, New York, New York, USA
| | - Ying Kuen Cheung
- Department of Biostatistics, Columbia University, New York, New York, USA, and
| | - George Hripcsak
- Department of Biomedical Informatics, Columbia University, New York, New York, USA.,Medical Informatics Services, New York-Presbyterian Hospital, New York, New York, USA
| | - Chunhua Weng
- Department of Biomedical Informatics, Columbia University, New York, New York, USA
| |
Collapse
|
18
|
Hui KHM, Lam HS, Chow CHT, Li YSJ, Leung PHT, Chan LYB, Lee CP, Ewig CLY, Cheung YT, Lam TNT. Personalized Dosing of Intravenous Vancomycin Among Critically Ill Neonates in Hong Kong: Harnessing Electronic Health Records to Develop a Web-Based Dosing Interface (Preprint). JMIR Med Inform 2021; 10:e29458. [PMID: 35099393 PMCID: PMC8844994 DOI: 10.2196/29458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 08/06/2021] [Accepted: 01/02/2022] [Indexed: 11/15/2022] Open
Abstract
Background Intravenous (IV) vancomycin is used in the treatment of severe infection in neonates. However, its efficacy is compromised by elevated risks of acute kidney injury. The risk is even higher among neonates admitted to the neonatal intensive care unit (NICU), in whom the pharmacokinetics of vancomycin vary widely. Therapeutic drug monitoring is an integral part of vancomycin treatment to balance efficacy against toxicity. It involves individual dose adjustments based on the observed serum vancomycin concentration (VCs). However, the existing trough-based approach shows poor evidence for clinical benefits. The updated clinical practice guideline recommends population pharmacokinetic (popPK) model–based approaches, targeting area under curve, preferably through the Bayesian approach. Since Bayesian methods cannot be performed manually and require specialized computer programs, there is a need to provide clinicians with a user-friendly interface to facilitate accurate personalized dosing recommendations for vancomycin in critically ill neonates. Objective We used medical data from electronic health records (EHRs) to develop a popPK model and subsequently build a web-based interface to perform model-based individual dose optimization of IV vancomycin for NICU patients in local medical institutions. Methods Medical data of subjects prescribed IV vancomycin in the NICUs of Prince of Wales Hospital and Queen Elizabeth Hospital in Hong Kong were extracted from EHRs, namely the Clinical Information System, In-Patient Medication Order Entry, and electronic Patient Record. Patient demographics, such as body weight and postmenstrual age (PMA), serum creatinine (SCr), vancomycin administration records, and VCs were collected. The popPK model employed a 2-compartment infusion model. Various covariate models were tested against body weight, PMA, and SCr, and were evaluated for the best goodness of fit. A previously published web-based dosing interface was adapted to develop the interface in this study. Results The final data set included EHR data extracted from 207 subjects, with a total of 689 VCs measurements. The final model chosen explained 82% of the variability in vancomycin clearance. All parameter estimates were within the bootstrapping CIs. Predictive plots, residual plots, and visual predictive checks demonstrated good model predictability. Model approximations showed that the model-based Bayesian approach consistently promoted a probability of target attainment (PTA) above 75% for all subjects, while only half of the subjects could achieve a PTA over 50% with the trough-based approach. The dosing interface was developed with the capability to optimize individual doses with the model-based empirical or Bayesian approach. Conclusions Using EHRs, a satisfactory popPK model was verified and adopted to develop a web-based individual dose optimization interface. The interface is expected to improve treatment outcomes of IV vancomycin for severe infections among critically ill neonates. This study provides the foundation for a cohort study to demonstrate the utility of the new approach compared with previous dosing methods.
Collapse
Affiliation(s)
- Ka Ho Matthew Hui
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Hugh Simon Lam
- Department of Paediatrics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Cheuk Hin Twinny Chow
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Yuen Shun Janice Li
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Pok Him Tom Leung
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Long Yin Brian Chan
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Chui Ping Lee
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
- Department of Pharmacy, Prince of Wales Hospital, Hospital Authority, Hong Kong, Hong Kong
| | - Celeste Lom Ying Ewig
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
- Department of Pharmacy, Prince of Wales Hospital, Hospital Authority, Hong Kong, Hong Kong
| | - Yin Ting Cheung
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Tai Ning Teddy Lam
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
- Department of Pharmacy, Prince of Wales Hospital, Hospital Authority, Hong Kong, Hong Kong
| |
Collapse
|
19
|
McNeer E, Beck C, Weeks HL, Williams ML, James NT, Bejan CA, Choi L. Building longitudinal medication dose data using medication information extracted from clinical notes in electronic health records. J Am Med Inform Assoc 2021; 28:782-790. [PMID: 33338223 DOI: 10.1093/jamia/ocaa291] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 12/08/2020] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE To develop an algorithm for building longitudinal medication dose datasets using information extracted from clinical notes in electronic health records (EHRs). MATERIALS AND METHODS We developed an algorithm that converts medication information extracted using natural language processing (NLP) into a usable format and builds longitudinal medication dose datasets. We evaluated the algorithm on 2 medications extracted from clinical notes of Vanderbilt's EHR and externally validated the algorithm using clinical notes from the MIMIC-III clinical care database. RESULTS For the evaluation using Vanderbilt's EHR data, the performance of our algorithm was excellent; F1-measures were ≥0.98 for both dose intake and daily dose. For the external validation using MIMIC-III, the algorithm achieved F1-measures ≥0.85 for dose intake and ≥0.82 for daily dose. DISCUSSION Our algorithm addresses the challenge of building longitudinal medication dose data using information extracted from clinical notes. Overall performance was excellent, but the algorithm can perform poorly when incorrect information is extracted by NLP systems. Although it performed reasonably well when applied to the external data source, its performance was worse due to differences in the way the drug information was written. The algorithm is implemented in the R package, "EHR," and the extracted data from Vanderbilt's EHRs along with the gold standards are provided so that users can reproduce the results and help improve the algorithm. CONCLUSION Our algorithm for building longitudinal dose data provides a straightforward way to use EHR data for medication-based studies. The external validation results suggest its potential for applicability to other systems.
Collapse
Affiliation(s)
- Elizabeth McNeer
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cole Beck
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hannah L Weeks
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michael L Williams
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nathan T James
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cosmin A Bejan
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Leena Choi
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
20
|
Kreutzer FP, Meinecke A, Schmidt K, Fiedler J, Thum T. Alternative strategies in cardiac preclinical research and new clinical trial formats. Cardiovasc Res 2021; 118:746-762. [PMID: 33693475 PMCID: PMC7989574 DOI: 10.1093/cvr/cvab075] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 03/03/2021] [Indexed: 02/07/2023] Open
Abstract
An efficient and safe drug development process is crucial for the establishment of new drugs on the market aiming to increase quality of life and life-span of our patients. Despite technological advances in the past decade, successful launches of drug candidates per year remain low. We here give an overview about some of these advances and suggest improvements for implementation to boost preclinical and clinical drug development with a focus on the cardiovascular field. We highlight advantages and disadvantages of animal experimentation and thoroughly review alternatives in the field of three-dimensional cell culture as well as preclinical use of spheroids and organoids. Microfluidic devices and their potential as organ-on-a-chip systems, as well as the use of living animal and human cardiac tissues are additionally introduced. In the second part, we examine recent gold standard randomized clinical trials and present possible modifications to increase lead candidate throughput: adaptive designs, master protocols, and drug repurposing. In silico and N-of-1 trials have the potential to redefine clinical drug candidate evaluation. Finally, we briefly discuss clinical trial designs during pandemic times.
Collapse
Affiliation(s)
- Fabian Philipp Kreutzer
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Anna Meinecke
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Kevin Schmidt
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Jan Fiedler
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany.,REBIRTH Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany.,Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany.,REBIRTH Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany.,Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| |
Collapse
|
21
|
Bejan CA, Cahill KN, Staso PJ, Choi L, Peterson JF, Phillips EJ. DrugWAS: Leveraging drug-wide association studies to facilitate drug repurposing for COVID-19. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.02.04.21251169. [PMID: 33564788 PMCID: PMC7872383 DOI: 10.1101/2021.02.04.21251169] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Importance: There is an unprecedented need to rapidly identify safe and effective treatments for the novel coronavirus disease 2019 (COVID-19). Objective: To systematically investigate if any of the available drugs in Electronic Health Record (EHR), including prescription drugs and dietary supplements, can be repurposed as potential treatment for COVID-19. Design, Setting, and Participants: Based on a retrospective cohort analysis of EHR data, drug-wide association studies (DrugWAS) were performed on COVID-19 patients at Vanderbilt University Medical Center (VUMC). For each drug study, multivariable logistic regression with overlap weighting using propensity score was applied to estimate the effect of drug exposure on COVID-19 disease outcomes. Exposures: Patient exposure to a drug during 1-year prior to the pandemic and COVID-19 diagnosis was chosen as exposure of interest. Natural language processing was employed to extract drug information from clinical notes, in addition to the prescription drug data available in structured format. Main Outcomes and Measures: All-cause of death was selected as primary outcome. Hospitalization, admission to the intensive care unit (ICU), and need for mechanical ventilation were identified as secondary outcomes. Results: The study included 7,768 COVID-19 patients, of which 509 (6.55%) were hospitalized, 82 (1.06%) were admitted to ICU, 64 (0.82%) received mechanical ventilation, and 90 (1.16%) died. Overall, 15 drugs were significantly associated with decreased COVID-19 severity. Previous exposure to either Streptococcus pneumoniae vaccines (adjusted odds ratio [OR], 0.38; 95% CI, 0.14-0.98), diphtheria toxoid vaccine (OR, 0.39; 95% CI, 0.15-0.98), and tetanus toxoid vaccine (OR, 0.39; 95% CI, 0.15-0.98) were significantly associated with a decreased risk of death (primary outcome). Secondary analyses identified several other significant associations showing lower risk for COVID-19 outcomes: 2 vaccines (acellular pertussis, Streptococcus pneumoniae), 3 dietary supplements (turmeric extract, flaxseed extract, omega-3 fatty acids), methylprednisolone acetate, pseudoephedrine, ethinyl estradiol, estradiol, ibuprofen, and fluticasone. Conclusions and Relevance: This cohort study leveraged EHR data to identify a list of drugs that could be repurposed to improve COVID-19 outcomes. Further randomized clinical trials are needed to investigate the efficacy of the proposed drugs.
Collapse
Affiliation(s)
- Cosmin A. Bejan
- Department of Biomedical Informatics; Vanderbilt University Medical Center; Nashville, USA
| | - Katherine N. Cahill
- Department of Medicine; Division of Allergy, Pulmonary and Critical Care Medicine; Vanderbilt University Medical Center; Nashville, USA
| | - Patrick J. Staso
- Department of Medicine; Division of Allergy, Pulmonary and Critical Care Medicine; Vanderbilt University Medical Center; Nashville, USA
| | - Leena Choi
- Department of Biostatistics; Vanderbilt University Medical Center; Nashville, USA
| | - Josh F. Peterson
- Department of Biomedical Informatics; Vanderbilt University Medical Center; Nashville, USA
- Department of Medicine; Vanderbilt University Medical Center; Nashville, USA
| | - Elizabeth J. Phillips
- Department of Pathology, Microbiology and Immunology; Vanderbilt University Medical Center; Nashville, USA
- Department of Medicine; Division of Infectious Diseases; Vanderbilt University Medical Center; Nashville, USA
- Department of Pharmacology; Vanderbilt University Medical Center; Nashville, USA
| |
Collapse
|
22
|
Liau SJ, Lalic S, Sluggett JK, Cesari M, Onder G, Vetrano DL, Morin L, Hartikainen S, Hamina A, Johnell K, Tan ECK, Visvanathan R, Bell JS. Medication Management in Frail Older People: Consensus Principles for Clinical Practice, Research, and Education. J Am Med Dir Assoc 2020; 22:43-49. [PMID: 32669236 DOI: 10.1016/j.jamda.2020.05.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/25/2020] [Accepted: 05/02/2020] [Indexed: 12/18/2022]
Abstract
Frailty is a geriatric condition associated with increased vulnerability to adverse drug events and medication-related harm. Existing clinical practice guidelines rarely provide medication management recommendations specific to frail older people. This report presents international consensus principles, generated by the Optimizing Geriatric Pharmacotherapy through Pharmacoepidemiology Network, related to medication management in frail older people. This consensus comprises 7 principles for clinical practice, 6 principles for research, and 4 principles for education. Principles for clinical practice include (1) perform medication reconciliation and maintain an up-to-date medication list; (2) assess and plan based on individual's capacity to self-manage medications; (3) ensure appropriate prescribing and deprescribing; (4) simplify medication regimens when appropriate to reduce unnecessary burden; (5) be alert to the contribution of medications to geriatric syndromes; (6) regularly review medication regimens to align with changing goals of care; and (7) facilitate multidisciplinary communication among patients, caregivers, and healthcare teams. Principles for research include (1) include frail older people in randomized controlled trials; (2) consider frailty status as an effect modifier; (3) ensure collection and reporting of outcome measures important in frailty; (4) assess impact of frailty on pharmacokinetics and pharmacodynamics; (5) encourage frailty research in under-researched settings; and (6) utilize routinely collected linked health data. Principles for education include (1) provide undergraduate and postgraduate education on frailty; (2) minimize low-value care related to medication management; (3) improve health and medication literacy; and (4) incorporate evidence in relation to frailty into clinical practice guidelines. These principles for clinical practice, research and education highlight different considerations for optimizing medication management in frail older people. These principles can be used in conjunction with existing best practice guidelines to help achieve optimal health outcomes for this vulnerable population. Implementation of the principles will require multidisciplinary collaboration between healthcare professionals, researchers, educators, organizational leaders, and policymakers.
Collapse
Affiliation(s)
- Shin J Liau
- Centre for Medicine Use and Safety, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Melbourne, Australia; National Health and Medical Research Council (NHMRC) Centre of Research Excellence in Frailty and Healthy Ageing, Adelaide, Australia
| | - Samanta Lalic
- Centre for Medicine Use and Safety, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Melbourne, Australia; Pharmacy Department, Monash Health, Melbourne, Australia
| | - Janet K Sluggett
- Centre for Medicine Use and Safety, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Melbourne, Australia; School of Health Sciences, Division of Health Sciences, University of South Australia, Adelaide, Australia; NHMRC Cognitive Decline Partnership Centre, Hornsby Ku-ring-gai Hospital, Hornsby, New South Wales, Australia
| | - Matteo Cesari
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Graziano Onder
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy
| | - Davide L Vetrano
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden; Centro Medicina dell'Invecchiamento, IRCCS Fondazione Policlinico Universitario A. Gemelli, and Università Cattolica del Sacro Cuore, Rome, Italy
| | - Lucas Morin
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden; Inserm CIC 1431, University Hospital of Besançon, Besançon, France
| | - Sirpa Hartikainen
- Kuopio Research Centre of Geriatric Care, School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Aleksi Hamina
- Kuopio Research Centre of Geriatric Care, School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland; Norwegian Centre for Addiction Research (SERAF), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Kristina Johnell
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Edwin C K Tan
- The University of Sydney School of Pharmacy, Faculty of Medicine and Health, Sydney, Australia
| | - Renuka Visvanathan
- National Health and Medical Research Council (NHMRC) Centre of Research Excellence in Frailty and Healthy Ageing, Adelaide, Australia; Adelaide Geriatrics Training and Research with Aged Care (GTRAC) Centre, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
| | - J Simon Bell
- Centre for Medicine Use and Safety, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Melbourne, Australia; National Health and Medical Research Council (NHMRC) Centre of Research Excellence in Frailty and Healthy Ageing, Adelaide, Australia; Kuopio Research Centre of Geriatric Care, School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland.
| | | |
Collapse
|
23
|
Peck RW, Shah P, Vamvakas S, van der Graaf PH. Data Science in Clinical Pharmacology and Drug Development for Improving Health Outcomes in Patients. Clin Pharmacol Ther 2020; 107:683-686. [PMID: 32202650 DOI: 10.1002/cpt.1803] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 01/30/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Richard W Peck
- Pharma Research and Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Pratik Shah
- Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | | | | |
Collapse
|