51
|
Chen S, Tikhonovsky N, Dhanji N, Ramagopalan S. Emulating Trials and Quantifying Bias: The Convergence of Health Technology Assessment Agency Real-World Evidence Guidance. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2024; 27:265-267. [PMID: 38135213 DOI: 10.1016/j.jval.2023.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 11/17/2023] [Indexed: 12/24/2023]
Affiliation(s)
- Simon Chen
- Health Analytics, Lane Clark & Peacock LLP, London, England, UK
| | | | - Nishit Dhanji
- Health Analytics, Lane Clark & Peacock LLP, London, England, UK
| | - Sreeram Ramagopalan
- Health Analytics, Lane Clark & Peacock LLP, London, England, UK; Centre for Pharmaceutical Medicine Research, King's College London, England, UK.
| |
Collapse
|
52
|
Aebersold H, Foster-Witassek F, Aeschbacher S, Beer JH, Blozik E, Blum M, Bonati L, Conte G, Coslovsky M, De Perna ML, Di Valentino M, Felder S, Huber CA, Moschovitis G, Mueller A, Paladini RE, Reichlin T, Rodondi N, Stauber A, Sticherling C, Szucs TD, Conen D, Kuhne M, Osswald S, Schwenkglenks M, Serra-Burriel M. Patients on vitamin K treatment: is switching to direct-acting oral anticoagulation cost-effective? A target trial on a prospective cohort. Open Heart 2024; 11:e002567. [PMID: 38302139 PMCID: PMC10831440 DOI: 10.1136/openhrt-2023-002567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 01/18/2024] [Indexed: 02/03/2024] Open
Abstract
AIMS Direct-acting oral anticoagulants (DOACs) have, to a substantial degree, replaced vitamin K antagonists (VKA) as treatments for stroke prevention in atrial fibrillation (AF) patients. However, evidence on the real-world causal effects of switching patients from VKA to DOAC is lacking. We aimed to assess the empirical incremental cost-effectiveness of switching patients to DOAC compared with maintaining VKA treatment. METHODS The target trial approach was applied to the prospective observational Swiss-AF cohort, which enrolled 2415 AF patients from 2014 to 2017. Clinical data, healthcare resource utilisation and EQ-5D-based utilities representing quality of life were collected in yearly follow-ups. Health insurance claims were available for 1024 patients (42.4%). Overall survival, quality-of-life, costs from the Swiss statutory health insurance perspective and cost-effectiveness were estimated by emulating a target trial in which patients were randomly assigned to switch to DOAC or maintain VKA treatment. RESULTS 228 patients switching from VKA to DOAC compared with 563 patients maintaining VKA treatment had no overall survival advantage over a 5-year observation period (HR 0.99, 95% CI 0.45, 1.55). The estimated gain in quality-adjusted life years (QALYs) was 0.003 over the 5-year period at an incremental costs of CHF 23 033 (€ 20 940). The estimated incremental cost-effectiveness ratio was CHF 425 852 (€ 387 138) per QALY gained. CONCLUSIONS Applying a causal inference method to real-world data, we could not demonstrate switching to DOACs to be cost-effective for AF patients with at least 1 year of VKA treatment. Our estimates align with results from a previous randomised trial.
Collapse
Affiliation(s)
- Helena Aebersold
- Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland
| | | | - Stefanie Aeschbacher
- Cardiology Division, Department of Medicine, University Hospital Basel, Basel, Switzerland
- Cardiovascular Research Institute Basel, University Hospital Basel, Basel, Switzerland
| | - Juerg H Beer
- Department of Medicine, Cantonal Hospital of Baden, Baden, Switzerland
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | - Eva Blozik
- Institute of Primary Care, University of Zurich, Zurich, Switzerland
| | - Manuel Blum
- Institute of Primary Health Care (BIHAM), University of Bern, Bern, Switzerland
- Department of General Internal Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Leo Bonati
- Department of Neurology, University Hospital Basel, Basel, Switzerland
- Research Department, Reha Rheinfelden, Rheinfelden, Switzerland
| | - Giulio Conte
- Division of Cardiology, Cardiocentro Ticino (CCT), Lugano, Switzerland
| | - Michael Coslovsky
- Cardiovascular Research Institute Basel, University Hospital Basel, Basel, Switzerland
- Clinical Trial Unit Basel, Department of Clinical Research, University Hospital Basel, Basel, Switzerland
| | - Maria Luisa De Perna
- Division of Cardiology, Istituto Cardiocentro Ticino, Ospedale Regionale di Bellinzona, Bellinzona, Switzerland
| | - Marcello Di Valentino
- Division of Cardiology, Istituto Cardiocentro Ticino, Ospedale Regionale di Bellinzona, Bellinzona, Switzerland
| | - Stefan Felder
- Faculty of Business and Economics, University of Basel, Basel, Switzerland
| | - Carola A Huber
- Department of Health Sciences, Helsana Group, Zurich, Switzerland
| | - Giorgio Moschovitis
- Division of Cardiology, Ente Ospedaliero Cantonale (EOC), Istituto Cardiocentro Ticino, Ospedale Regionale di Lugano, Lugano, Switzerland
| | - Andreas Mueller
- Department of Cardiology, Triemli Hospital Zurich, Zurich, Switzerland
| | - Rebecca E Paladini
- Cardiology Division, Department of Medicine, University Hospital Basel, Basel, Switzerland
- Cardiovascular Research Institute Basel, University Hospital Basel, Basel, Switzerland
| | - Tobias Reichlin
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Nicolas Rodondi
- Institute of Primary Health Care (BIHAM), University of Bern, Bern, Switzerland
- Department of General Internal Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Annina Stauber
- Department of Cardiology, Triemli Hospital Zurich, Zurich, Switzerland
| | - Christian Sticherling
- Cardiology Division, Department of Medicine, University Hospital Basel, Basel, Switzerland
- Cardiovascular Research Institute Basel, University Hospital Basel, Basel, Switzerland
| | - Thomas D Szucs
- Health Economics Facility, Department of Public Health, University of Basel, Basel, Switzerland
| | - David Conen
- Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Michael Kuhne
- Cardiology Division, Department of Medicine, University Hospital Basel, Basel, Switzerland
- Cardiovascular Research Institute Basel, University Hospital Basel, Basel, Switzerland
| | - Stefan Osswald
- Cardiology Division, Department of Medicine, University Hospital Basel, Basel, Switzerland
- Cardiovascular Research Institute Basel, University Hospital Basel, Basel, Switzerland
| | - Matthias Schwenkglenks
- Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland
- Health Economics Facility, Department of Public Health, University of Basel, Basel, Switzerland
| | - Miquel Serra-Burriel
- Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland
| |
Collapse
|
53
|
Ramagopalan SV, Diaz J, Mitchell G, Garrison LP, Kolchinsky P. Is the price right? Paying for value today to get more value tomorrow. BMC Med 2024; 22:45. [PMID: 38287326 PMCID: PMC10826180 DOI: 10.1186/s12916-024-03262-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 01/18/2024] [Indexed: 01/31/2024] Open
Abstract
BACKGROUND Contemporary debates about drug pricing feature several widely held misconceptions, including the relationship between incentives and innovation, the proportion of total healthcare spending on pharmaceuticals, and whether the economic evaluation of a medicine can be influenced by things other than clinical efficacy. MAIN BODY All citizens should have access to timely, equitable, and cost-effective care covered by public funds, private insurance, or a combination of both. Better managing the collective burden of diseases borne by today's and future generations depends in part on developing better technologies, including better medicines. As in any innovative industry, the expectation of adequate financial returns incentivizes innovators and their investors to develop new medicines. Estimating expected returns requires that they forecast revenues, based on the future price trajectory and volume of use over time. How market participants decide what price to set or accept can be complicated, and some observers and stakeholders want to confirm whether the net prices society pays for novel medicines, whether as a reward for past innovation or an incentive for future innovation, are commensurate with those medicines' incremental value. But we must also ask "value to whom?"; medicines not only bring immediate clinical benefits to patients treated today, but also can provide a broad spectrum of short- and long-term benefits to patients, their families, and society. Spending across all facets of healthcare has grown over the last 25 years, but both inpatient and outpatient spending has outpaced drug spending growth even as our drug armamentarium is constantly improving with safer and more effective medicines. In large part, this is because, unlike hospitals, drugs typically go generic, thus making room in our budgets for new and better ones, even as they often keep patients out of hospitals, driving further savings. CONCLUSION A thorough evaluation of drug spending and value can help to promote a better allocation of healthcare resources for both the healthy and the sick, both of whom must pay for healthcare. Taking a holistic approach to assessing drug value makes it clear that a branded drug's value to a patient is often only a small fraction of the drug's total value to society. Societal value merits consideration when determining whether and how to make a medicine affordable and accessible to patients: a drug that is worth its price to society should not be rendered inaccessible to ill patients by imposing high out-of-pocket costs or restricting coverage based on narrow health technology assessments (HTAs). Furthermore, recognizing the total societal cost of un- or undertreated conditions is crucial to gaining a thorough understanding of what guides the biomedical innovation ecosystem to create value for society. It would be unwise to discourage the development of new solutions without first appreciating the cost of leaving the problems unsolved.
Collapse
Affiliation(s)
- Sreeram V Ramagopalan
- Lane Clark & Peacock LLP, London, UK.
- Centre for Pharmaceutical Medicine Research, King's College London, London, UK.
| | - Jose Diaz
- Global Health Economics and Outcomes Research, Bristol Myers Squibb, Uxbridge, UK
| | | | - Louis P Garrison
- The Comparative Health Outcomes, Policy, and Economics (CHOICE) Institute, University of Washington, Seattle, USA
| | | |
Collapse
|
54
|
Kensler KH, Johnson R, Morley F, Albrair M, Dickerman BA, Gulati R, Holt SK, Iyer HS, Kibel AS, Lee JR, Preston MA, Vassy JL, Wolff EM, Nyame YA, Etzioni R, Rebbeck TR. Prostate cancer screening in African American men: a review of the evidence. J Natl Cancer Inst 2024; 116:34-52. [PMID: 37713266 PMCID: PMC10777677 DOI: 10.1093/jnci/djad193] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/25/2023] [Accepted: 08/30/2023] [Indexed: 09/16/2023] Open
Abstract
BACKGROUND Prostate cancer is the most diagnosed cancer in African American men, yet prostate cancer screening regimens in this group are poorly guided by existing evidence, given underrepresentation of African American men in prostate cancer screening trials. It is critical to optimize prostate cancer screening and early detection in this high-risk group because underdiagnosis may lead to later-stage cancers at diagnosis and higher mortality while overdiagnosis may lead to unnecessary treatment. METHODS We performed a review of the literature related to prostate cancer screening and early detection specific to African American men to summarize the existing evidence available to guide health-care practice. RESULTS Limited evidence from observational and modeling studies suggests that African American men should be screened for prostate cancer. Consideration should be given to initiating screening of African American men at younger ages (eg, 45-50 years) and at more frequent intervals relative to other racial groups in the United States. Screening intervals can be optimized by using a baseline prostate-specific antigen measurement in midlife. Finally, no evidence has indicated that African American men would benefit from screening beyond 75 years of age; in fact, this group may experience higher rates of overdiagnosis at older ages. CONCLUSIONS The evidence base for prostate cancer screening in African American men is limited by the lack of large, randomized studies. Our literature search supported the need for African American men to be screened for prostate cancer, for initiating screening at younger ages (45-50 years), and perhaps screening at more frequent intervals relative to men of other racial groups in the United States.
Collapse
Affiliation(s)
- Kevin H Kensler
- Department of Population Health Sciences, Weill Cornell Medical Center, New York, NY, USA
| | - Roman Johnson
- Center for Global Health, Massachusetts General Hospital, Boston, MA, USA
| | - Faith Morley
- Department of Population Health Sciences, Weill Cornell Medical Center, New York, NY, USA
| | - Mohamed Albrair
- Department of Global Health, University of Washington, Seattle, WA, USA
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Barbra A Dickerman
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Roman Gulati
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Sarah K Holt
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Hari S Iyer
- Section of Cancer Epidemiology and Health Outcomes, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Adam S Kibel
- Department of Urology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Jenney R Lee
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Mark A Preston
- Department of Urology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Jason L Vassy
- VA Boston Healthcare System, Boston, MA, USA
- Division of General Internal Medicine and Primary Care, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Erika M Wolff
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Yaw A Nyame
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Ruth Etzioni
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Timothy R Rebbeck
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Division of Population Sciences, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
55
|
Platt A, Wilson J, Hall R, Ephraim PL, Morton S, Shafi T, Weiner DE, Boulware LE, Pendergast J, Scialla JJ. Comparative Effectiveness of Alternative Treatment Approaches to Secondary Hyperparathyroidism in Patients Receiving Maintenance Hemodialysis: An Observational Trial Emulation. Am J Kidney Dis 2024; 83:58-70. [PMID: 37690631 PMCID: PMC10919553 DOI: 10.1053/j.ajkd.2023.05.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 05/15/2023] [Accepted: 05/23/2023] [Indexed: 09/12/2023]
Abstract
RATIONALE & OBJECTIVE Optimal approaches to treat secondary hyperparathyroidism (SHPT) in patients on maintenance hemodialysis (HD) have yet to be established in randomized controlled trials (RCTs). STUDY DESIGN Two observational clinical trial emulations. SETTING & PARTICIPANTS Both emulations included adults receiving in-center HD from a national dialysis organization. The patients who had SHPT in the period between 2009 and 2014, were insured for≥180 days by Medicare as primary payer, and did not have contraindications or poor health status limiting theoretical trial participation. EXPOSURE The parathyroid hormone (PTH) Target Trial emulation included patients with new-onset SHPT (first PTH 300-600pg/mL), with 2 arms defined as up-titration of either vitamin D sterols or cinacalcet within 30 days (lower target) or no up-titration (higher target). The Agent Trial emulation included patients with a PTH≥300 pg/mL while on≥6μg weekly of vitamin D sterol (paricalcitol equivalent dose) and no prior history of cinacalcet. The 2 arms were defined by the first dose or agent change within 30 days (vitamin D-favoring [vitamin-D was up-titrated] vs cinacalcet-favoring [cinacalcet was added] vs nondefined [neither applies]). Multiple trials per patient were allowed in trial 2. OUTCOME The primary outcome was all-cause death over 24 months; secondary outcomes included cardiovascular (CV) hospitalization or the composite of CV hospitalization or death. ANALYTICAL APPROACH Pooled logistic regression. RESULTS There were 1,152 patients in the PTH Target Trial (635 lower target and 517 higher target). There were 2,726 unique patients with 6,727 patient trials in the Agent Trial (6,268 vitamin D-favoring trials and 459 cinacalcet-favoring trials). The lower PTH target approach was associated with reduced adjusted hazard of death (HR, 0.71 [95% CI, 0.52-0.93]), CV hospitalization (HR, 0.78 [95% CI, 0.63-0.98]), and their composite (HR, 0.74 [95% CI, 0.61-0.89]). The cinacalcet-favoring approach demonstrated lower adjusted hazard of death compared to the vitamin D-favoring approach (HR, 0.79 [95% CI, 0.62-0.99]), but not of CV hospitalization or the composite outcome. LIMITATIONS Potential for residual confounding; low use of cinacalcet with low power. CONCLUSIONS SHPT management that is focused on lower PTH targets may lower mortality and CV disease in patients receiving HD. These findings should be confirmed in a pragmatic randomized trial. PLAIN-LANGUAGE SUMMARY Optimal approaches to treat secondary hyperparathyroidism (SHPT) have not been established in randomized controlled trials. Data from a national dialysis organization was used to identify patients with SHPT in whom escalated treatment may be indicated. The approach to treatment was defined based on observed upward titration of SHPT-controlling medications: earlier titration (lower target) versus delayed titration (higher target); and the choice of medication (cinacalcet vs vitamin D sterols). In the first trial emulation, we estimated a 29% lower rate of death and 26% lower rate of cardiovascular disease or death for patients managed with a lower versus higher target approach. Cinacalcet versus vitamin D-favoring approaches were not consistently associated with outcomes in the second trial emulation. This observational study suggests the need for additional clinical trials of SHPT treatment intensity.
Collapse
Affiliation(s)
- Alyssa Platt
- Department of Biostatistics and Bioinformatics, School of Medicine, Duke University, Durham, North Carolina
| | - Jonathan Wilson
- Department of Biostatistics and Bioinformatics, School of Medicine, Duke University, Durham, North Carolina
| | - Rasheeda Hall
- Department of Medicine, School of Medicine, Duke University, Durham, North Carolina
| | - Patti L Ephraim
- Feinstein Institute for Medical Research, Northwell Health, New York, New York
| | - Sarah Morton
- Department of Biostatistics and Bioinformatics, School of Medicine, Duke University, Durham, North Carolina
| | - Tariq Shafi
- Department of Medicine, Houston Methodist Hospital, Houston, Texas
| | - Daniel E Weiner
- Department of Medicine, Tufts Medical Center, Boston, Massachusetts
| | - L Ebony Boulware
- School of Medicine, Wake Forest University, Winston-Salem, North Carolina
| | - Jane Pendergast
- Department of Biostatistics and Bioinformatics, School of Medicine, Duke University, Durham, North Carolina; Department of Medicine, School of Medicine, Duke University, Durham, North Carolina
| | - Julia J Scialla
- Departments of Medicine and Public Health Sciences, University of Virginia School of Medicine, Charlottesville, Virginia.
| |
Collapse
|
56
|
Zang C, Zhang H, Xu J, Zhang H, Fouladvand S, Havaldar S, Cheng F, Chen K, Chen Y, Glicksberg BS, Chen J, Bian J, Wang F. High-throughput target trial emulation for Alzheimer's disease drug repurposing with real-world data. Nat Commun 2023; 14:8180. [PMID: 38081829 PMCID: PMC10713627 DOI: 10.1038/s41467-023-43929-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
Target trial emulation is the process of mimicking target randomized trials using real-world data, where effective confounding control for unbiased treatment effect estimation remains a main challenge. Although various approaches have been proposed for this challenge, a systematic evaluation is still lacking. Here we emulated trials for thousands of medications from two large-scale real-world data warehouses, covering over 10 years of clinical records for over 170 million patients, aiming to identify new indications of approved drugs for Alzheimer's disease. We assessed different propensity score models under the inverse probability of treatment weighting framework and suggested a model selection strategy for improved baseline covariate balancing. We also found that the deep learning-based propensity score model did not necessarily outperform logistic regression-based methods in covariate balancing. Finally, we highlighted five top-ranked drugs (pantoprazole, gabapentin, atorvastatin, fluticasone, and omeprazole) originally intended for other indications with potential benefits for Alzheimer's patients.
Collapse
Affiliation(s)
- Chengxi Zang
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
- Institute of Artificial Intelligence for Digital Health, Weill Cornell Medicine, New York, NY, USA
| | - Hao Zhang
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Jie Xu
- Department of Health Outcomes & Biomedical Informatics, University of Florida, Gainesville, FL, USA
| | - Hansi Zhang
- Department of Health Outcomes & Biomedical Informatics, University of Florida, Gainesville, FL, USA
| | - Sajjad Fouladvand
- Institude for Biomedical Informatics (IBI) and Department of Computer Science, University of Kentucky, Lexington, KY, USA
| | - Shreyas Havaldar
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Kun Chen
- Department of Statistics, University of Connecticut, Storrs, CT, USA
| | - Yong Chen
- Department of Biostatistics, Epidemiology and Informatics (DBEI), the Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Benjamin S Glicksberg
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jin Chen
- Institude for Biomedical Informatics (IBI) and Department of Computer Science, University of Kentucky, Lexington, KY, USA
| | - Jiang Bian
- Department of Health Outcomes & Biomedical Informatics, University of Florida, Gainesville, FL, USA
| | - Fei Wang
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA.
- Institute of Artificial Intelligence for Digital Health, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
57
|
Barcellini L, Bresesti I, Folgori L. Neglected Populations Not to Be Forgotten: Tackling Antimicrobial Resistance in Neonatal Infections. Antibiotics (Basel) 2023; 12:1688. [PMID: 38136722 PMCID: PMC10741198 DOI: 10.3390/antibiotics12121688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 11/24/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023] Open
Abstract
Making further progress in reducing child mortality hinges on lowering the annual count of neonatal deaths; currently, this stands at 2 [...].
Collapse
Affiliation(s)
- Lucia Barcellini
- Department of Pediatrics, “V. Buzzi” Children’s Hospital, ASST FBF Sacco, 20154 Milan, Italy;
| | - Ilia Bresesti
- Neonatal Intensive Care Unit, “Filippo del Ponte” Hospital, ASST Settelaghi, 21100 Varese, Italy;
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy
| | - Laura Folgori
- Department of Pediatrics, “V. Buzzi” Children’s Hospital, ASST FBF Sacco, 20154 Milan, Italy;
| |
Collapse
|
58
|
Matthews AA, Young JC, Kurth T. The target trial framework in clinical epidemiology: principles and applications. J Clin Epidemiol 2023; 164:112-115. [PMID: 37863299 DOI: 10.1016/j.jclinepi.2023.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 10/12/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023]
Affiliation(s)
- Anthony A Matthews
- CAUSALab, Unit of Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jessica C Young
- CAUSALab, Unit of Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Tobias Kurth
- Institute of Public Health, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
59
|
Katsoulis M, Lai AG, Kipourou DK, Gomes M, Banerjee A, Denaxas S, Lumbers RT, Tsilidis K, Kostara M, Belot A, Dale C, Sofat R, Leyrat C, Hemingway H, Diaz-Ordaz K. On the estimation of the effect of weight change on a health outcome using observational data, by utilising the target trial emulation framework. Int J Obes (Lond) 2023; 47:1309-1317. [PMID: 37884665 PMCID: PMC10663146 DOI: 10.1038/s41366-023-01396-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 09/17/2023] [Accepted: 10/10/2023] [Indexed: 10/28/2023]
Abstract
BACKGROUND/OBJECTIVES When studying the effect of weight change between two time points on a health outcome using observational data, two main problems arise initially (i) 'when is time zero?' and (ii) 'which confounders should we account for?' From the baseline date or the 1st follow-up (when the weight change can be measured)? Different methods have been previously used in the literature that carry different sources of bias and hence produce different results. METHODS We utilised the target trial emulation framework and considered weight change as a hypothetical intervention. First, we used a simplified example from a hypothetical randomised trial where no modelling is required. Then we simulated data from an observational study where modelling is needed. We demonstrate the problems of each of these methods and suggest a strategy. INTERVENTIONS weight loss/gain vs maintenance. RESULTS The recommended method defines time-zero at enrolment, but adjustment for confounders (or exclusion of individuals based on levels of confounders) should be performed both at enrolment and the 1st follow-up. CONCLUSIONS The implementation of our suggested method [adjusting for (or excluding based on) confounders measured both at baseline and the 1st follow-up] can help researchers attenuate bias by avoiding some common pitfalls. Other methods that have been widely used in the past to estimate the effect of weight change on a health outcome are more biased. However, two issues remain (i) the exposure is not well-defined as there are different ways of changing weight (however we tried to reduce this problem by excluding individuals who develop a chronic disease); and (ii) immortal time bias, which may be small if the time to first follow up is short.
Collapse
Affiliation(s)
- M Katsoulis
- MRC Unit for Lifelong Health and Ageing, Institute of Cardiovascular Science, University College London, London, UK.
| | - A G Lai
- Institute of Health Informatics, University College London, London, UK
| | - D K Kipourou
- Inequalities in Cancer Outcomes Network, Department of Non-communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
- AstraZeneca, London, UK
| | - M Gomes
- Department of Applied Health Research, University College London, London, UK
| | - A Banerjee
- Institute of Health Informatics, University College London, London, UK
- University College London Hospitals NHS Trust, London, UK
- Barts Health NHS Trust, The Royal London Hospital, London, UK
| | - S Denaxas
- Institute of Health Informatics, University College London, London, UK
- Alan Turing Institute, London, UK
| | - R T Lumbers
- Institute of Health Informatics, University College London, London, UK
| | - K Tsilidis
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Department of Hygiene and Epidemiology, University of Ioannina School of Medicine, Ioannina, Greece
| | - Maria Kostara
- Department of Pediatrics, University Hospital of Ioannina, Ioannina, Greece
| | - A Belot
- Inequalities in Cancer Outcomes Network, Department of Non-communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - C Dale
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, UK
| | - R Sofat
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, UK
| | - C Leyrat
- Department of Medical Statistics, London School of Hygiene and Tropical Medicine, London, UK
| | - H Hemingway
- Institute of Health Informatics, University College London, London, UK
| | - K Diaz-Ordaz
- Dept of Statistical Science, Faculty of Maths & Physical Sciences, University College London, London, UK
| |
Collapse
|
60
|
Serra-Burriel M, Aebersold H, Foster-Witassek F, Coslovsky M, Rodondi N, Blum MR, Sticherling C, Moschovitis G, Beer JH, Reichlin T, Krisai P, Aeschbacher S, Paladini RE, Kühne M, Osswald S, Conen D, Felder S, Schwenkglenks M. Real-World Cost-Effectiveness of Pulmonary Vein Isolation for Atrial Fibrillation: A Target Trial Approach. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2023; 26:1721-1729. [PMID: 37741443 DOI: 10.1016/j.jval.2023.08.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/27/2023] [Accepted: 08/05/2023] [Indexed: 09/25/2023]
Abstract
OBJECTIVES Randomized controlled trials of pulmonary vein isolation (PVI) for treating atrial fibrillation (AF) have proven the procedure's efficacy. Studies assessing its empirical cost-effectiveness outside randomized trial settings are lacking. We aimed to evaluate the effectiveness and cost-effectiveness of PVI versus medical therapy for AF. METHODS We followed a target trial approach using the Swiss-AF cohort, a prospective observational cohort study that enrolled patients with AF between 2014 and 2017. Resource utilization and cost information were collected through claims data. Quality of life was measured with EQ-5D-3L utilities. We estimated incremental cost-effectiveness ratios (ICERs) from the perspective of the Swiss statutory health insurance system. RESULTS Patients undergoing PVI compared with medical therapy had a 5-year overall survival advantage with a hazard ratio of 0.75 (95% CI 0.46-1.21; P = .69) and a 19.8% SD improvement in quality of life (95% CI 15.5-22.9; P < .001), at an incremental cost of 29 604 Swiss francs (CHF) (95% CI 16 354-42 855; P < .001). The estimated ICER was CHF 158 612 per quality-adjusted life-year (QALY) gained within a 5-year time horizon. Assuming similar health effects and costs over 5 additional years changed the ICER to CHF 82 195 per QALY gained. Results were robust to the sensitivity analyses performed. CONCLUSIONS Our results show that PVI might be a cost-effective intervention within the Swiss healthcare context in a 10-year time horizon, but unlikely to be so at 5 years, if a willingness-to-pay threshold of CHF 100 000 per QALY gained is assumed. Given data availability, we find target trial designs are a valuable tool for assessing the cost-effectiveness of healthcare interventions outside of randomized controlled trial settings.
Collapse
Affiliation(s)
- Miquel Serra-Burriel
- Epidemiology, Biostatistics, and Prevention Institute, University of Zurich, Zurich, Switzerland.
| | - Helena Aebersold
- Epidemiology, Biostatistics, and Prevention Institute, University of Zurich, Zurich, Switzerland
| | - Fabienne Foster-Witassek
- Epidemiology, Biostatistics, and Prevention Institute, University of Zurich, Zurich, Switzerland
| | - Michael Coslovsky
- Department of Clinical Research, University of Basel, University Hospital, Basel, Switzerland; Cardiovascular Research Institute Basel, University of Basel, University Hospital, Basel, Switzerland
| | - Nicolas Rodondi
- Department of General Internal Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Institute of Primary Health Care (BIHAM), University of Bern, Bern, Switzerland
| | - Manuel R Blum
- Department of General Internal Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Institute of Primary Health Care (BIHAM), University of Bern, Bern, Switzerland
| | - Christian Sticherling
- Cardiovascular Research Institute Basel, University of Basel, University Hospital, Basel, Switzerland; Department of Cardiology, University Hospital Basel, University of Basel, Basel, Switzerland; Department of Medicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Giorgio Moschovitis
- Division of Cardiology, Ente Ospedaliero Cantonale, Regional Hospital of Lugano, Lugano, Switzerland
| | - Jürg H Beer
- Department of Medicine, Cantonal Hospital of Baden and Molecular Cardiology, University Hospital of Zürich, Zurich, Switzerland
| | - Tobias Reichlin
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Philipp Krisai
- Cardiovascular Research Institute Basel, University of Basel, University Hospital, Basel, Switzerland; Department of Cardiology, University Hospital Basel, University of Basel, Basel, Switzerland; Department of Medicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Stefanie Aeschbacher
- Cardiovascular Research Institute Basel, University of Basel, University Hospital, Basel, Switzerland; Department of Cardiology, University Hospital Basel, University of Basel, Basel, Switzerland; Department of Medicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Rebecca E Paladini
- Cardiovascular Research Institute Basel, University of Basel, University Hospital, Basel, Switzerland; Department of Cardiology, University Hospital Basel, University of Basel, Basel, Switzerland; Department of Medicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Michael Kühne
- Cardiovascular Research Institute Basel, University of Basel, University Hospital, Basel, Switzerland; Department of Cardiology, University Hospital Basel, University of Basel, Basel, Switzerland; Department of Medicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Stefan Osswald
- Cardiovascular Research Institute Basel, University of Basel, University Hospital, Basel, Switzerland; Department of Cardiology, University Hospital Basel, University of Basel, Basel, Switzerland; Department of Medicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - David Conen
- Population Health Research Institute, McMaster University, Hamilton, Canada
| | - Stefan Felder
- Department of Business and Economics, University of Basel, Basel, Switzerland
| | - Matthias Schwenkglenks
- Epidemiology, Biostatistics, and Prevention Institute, University of Zurich, Zurich, Switzerland; Health Economics Facility, Department of Public Health, University of Basel, Basel, Switzerland
| |
Collapse
|
61
|
Smit JM, Krijthe JH, Kant WMR, Labrecque JA, Komorowski M, Gommers DAMPJ, van Bommel J, Reinders MJT, van Genderen ME. Causal inference using observational intensive care unit data: a scoping review and recommendations for future practice. NPJ Digit Med 2023; 6:221. [PMID: 38012221 PMCID: PMC10682453 DOI: 10.1038/s41746-023-00961-1] [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: 06/23/2023] [Accepted: 11/05/2023] [Indexed: 11/29/2023] Open
Abstract
This scoping review focuses on the essential role of models for causal inference in shaping actionable artificial intelligence (AI) designed to aid clinicians in decision-making. The objective was to identify and evaluate the reporting quality of studies introducing models for causal inference in intensive care units (ICUs), and to provide recommendations to improve the future landscape of research practices in this domain. To achieve this, we searched various databases including Embase, MEDLINE ALL, Web of Science Core Collection, Google Scholar, medRxiv, bioRxiv, arXiv, and the ACM Digital Library. Studies involving models for causal inference addressing time-varying treatments in the adult ICU were reviewed. Data extraction encompassed the study settings and methodologies applied. Furthermore, we assessed reporting quality of target trial components (i.e., eligibility criteria, treatment strategies, follow-up period, outcome, and analysis plan) and main causal assumptions (i.e., conditional exchangeability, positivity, and consistency). Among the 2184 titles screened, 79 studies met the inclusion criteria. The methodologies used were G methods (61%) and reinforcement learning methods (39%). Studies considered both static (51%) and dynamic treatment regimes (49%). Only 30 (38%) of the studies reported all five target trial components, and only seven (9%) studies mentioned all three causal assumptions. To achieve actionable AI in the ICU, we advocate careful consideration of the causal question of interest, describing this research question as a target trial emulation, usage of appropriate causal inference methods, and acknowledgement (and examination of potential violations of) the causal assumptions.
Collapse
Affiliation(s)
- J M Smit
- Department of Intensive Care, Erasmus University Medical Center, Rotterdam, The Netherlands.
- Pattern Recognition & Bioinformatics group, EEMCS, Delft University of Technology, Delft, The Netherlands.
| | - J H Krijthe
- Pattern Recognition & Bioinformatics group, EEMCS, Delft University of Technology, Delft, The Netherlands
| | - W M R Kant
- Data Science group, Institute for Computing and Information Sciences, Radboud University, Nijmegen, The Netherlands
| | - J A Labrecque
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - M Komorowski
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
- Intensive Care Unit, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - D A M P J Gommers
- Department of Intensive Care, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - J van Bommel
- Department of Intensive Care, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - M J T Reinders
- Pattern Recognition & Bioinformatics group, EEMCS, Delft University of Technology, Delft, The Netherlands
| | - M E van Genderen
- Department of Intensive Care, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
62
|
Christiaens A, Simon-Tillaux N, Thompson W, Sinclair AJ, Henrard S, Boland BB, Slaouti-Jégou Y, Lekens B, Bonnet-Zamponi D, Tubach F, Zerah L. Impact of deintensifying hypoglycaemic drugs in older adults with type 2 diabetes: protocol for an emulation of a target trial. BMJ Open 2023; 13:e073081. [PMID: 37984943 PMCID: PMC10660441 DOI: 10.1136/bmjopen-2023-073081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 11/07/2023] [Indexed: 11/22/2023] Open
Abstract
INTRODUCTION In older adults with type 2 diabetes (T2D), overtreatment with hypoglycaemic drugs (HDs: sulfonylureas, glinides and/or insulins) is frequent and associated with increased 1-year mortality. Deintensification of HD is thus a key issue, for which evidence is though limited. The primary objective of this study will be to estimate the effect of deintensifying HD on clinical outcomes (hospital admission or death) within 3 months in older adults (≥75 years) with T2D. METHODS We will emulate with real-world data a target trial, within The Health Improvement Network cohort, a large-scale database of data collected from electronic medical records of 2000 general practitioners in France. From 1 January 2010 to 28 February 2019, we will include eligible patients ≥75 years who will have T2D, a stable dose of HDs, glycated haemoglobin A1c (HbA1c) value <75 mmol/mol (9.0%) and no deintensification in the past year. The target trial will be sequentially emulated (ie, eligibility assessed) every month in the database. Patients will be classified at baseline of each sequential trial in the intervention arm (deintensification of HDs: decrease of ≥50% in the total dose of HDs, including complete cessation) or control arm (no deintensification of HDs). The pooled dataset for all sequential emulated trials will be analysed. The primary outcome will be time to first occurrence of hospital admission or death, within 3 months. Secondary outcomes will be hospitalisation, death, appropriateness of glycaemic control and occurrence of HbA1c >75 mmol/mol within 1 year. Participants will be followed from baseline to 12 months after randomisation, administrative censoring, or death, whichever occurs first. A pooled logistic regression will be used to estimate the treatment effect on the incidence of the outcomes. DISSEMINATION AND ETHICS No ethical approval is needed for using retrospectively this fully anonymised database. The results will be disseminated during conferences and through publications in scientific journals.
Collapse
Affiliation(s)
- Antoine Christiaens
- FNRS, Fund for Scientific Research, Bruxelles, Belgium
- Clinical Pharmacy research group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
- Institut Pierre Louis d'Epidémiologie et de Santé Publique, INSERM, Sorbonne Universite, Paris, Île-de-France, France
| | - Noémie Simon-Tillaux
- Institut Pierre Louis d'Epidémiologie et de Santé Publique, INSERM, Sorbonne Universite, Paris, Île-de-France, France
- Hôpital Pitié Salpêtrière, Département de Santé Publique, Centre de Pharmacoépidémiologie (Cephepi), Unité́ de Recherche Clinique PSL-CFX, CIC-1901, Assistance Publique - Hôpitaux de Paris, Paris, Île-de-France, France
| | - Wade Thompson
- Department of Anesthesiology, Pharmacology, and Therapeutics, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
- Research Unit of General Practice, University of Southern Denmark, Odense, Denmark
| | - Alan J Sinclair
- Care for long term conditions - Diabetes research group, King's College London, London, UK
- Foundation for Diabetes Research in Older People, Taplow, UK
| | - Séverine Henrard
- Clinical Pharmacy research group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
- Institute of Health and Society, Université catholique de Louvain, Brussels, Belgium
| | - Benoit B Boland
- Institute of Health and Society, Université catholique de Louvain, Brussels, Belgium
- Geriatric Department, Cliniques universitaires Saint-Luc, Bruxelles, Belgium
| | | | | | - Dominique Bonnet-Zamponi
- Institut Pierre Louis d'Epidémiologie et de Santé Publique, INSERM, Sorbonne Universite, Paris, Île-de-France, France
- Observatoire des médicaments, dispositifs médicaux, innovations thérapeutiques d'Île-de-France, Paris, France
| | - Florence Tubach
- Institut Pierre Louis d'Epidémiologie et de Santé Publique, INSERM, Sorbonne Universite, Paris, Île-de-France, France
- Hôpital Pitié Salpêtrière, Département de Santé Publique, Centre de Pharmacoépidémiologie (Cephepi), Unité́ de Recherche Clinique PSL-CFX, CIC-1901, Assistance Publique - Hôpitaux de Paris, Paris, Île-de-France, France
| | - Lorène Zerah
- Institut Pierre Louis d'Epidémiologie et de Santé Publique, INSERM, Sorbonne Universite, Paris, Île-de-France, France
- Hôpital Pitié Salpêtrière, Département de gériatrie, Assistance Publique - Hôpitaux de Paris, Paris, Île-de-France, France
| |
Collapse
|
63
|
Lagerberg T, Matthews AA, Zhu N, Fazel S, Carrero JJ, Chang Z. Effect of selective serotonin reuptake inhibitor treatment following diagnosis of depression on suicidal behaviour risk: a target trial emulation. Neuropsychopharmacology 2023; 48:1760-1768. [PMID: 37507510 PMCID: PMC10579366 DOI: 10.1038/s41386-023-01676-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 07/12/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023]
Abstract
There is concern regarding the impact of selective serotonin reuptake inhibitors (SSRIs) on suicidal behaviour. Using the target trial framework, we investigated the effect on suicidal behaviour of SSRI treatment following a depression diagnosis. We identified 162,267 individuals receiving a depression diagnosis aged 6-59 years during 2006-2018 in Stockholm County, Sweden, after at least 1 year without antidepressant dispensation. Individuals who initiated an SSRI within 28 days of the diagnosis were assigned as SSRI initiators, others as non-initiators. Intention-to-treat and per-protocol effects were estimated; for the latter, individuals were censored when they ceased adhering to their assigned treatment strategy. We applied inverse probability weighting (IPW) to account for baseline confounding in the intention-to-treat analysis, and additionally for treatment non-adherence and time-varying confounding in the per-protocol analysis. The suicidal behaviour risk difference (RD), and risk ratio (RR) between SSRI initiators and non-initiators were estimated at 12 weeks. In the overall cohort, we found an increased risk of suicidal behaviour among SSRI initiators (intention-to-treat RR = 1.50, 95% CI = 1.25, 1.80; per-protocol RR = 1.69, 95% CI = 1.20, 2.36). In age strata, we only found evidence of an increased risk among individuals under age 25, with the greatest risk among 6-17-year-olds (intention-to-treat RR = 2.90, 95% CI = 1.72, 4.91; per-protocol RR = 3.34, 95% CI = 1.59, 7.00). Our finding of an increased suicidal behaviour risk among individuals under age 25 reflects evidence from RCTs. We found no evidence of an effect in the high-risk group of individuals with past suicidal behaviour. Further studies with information on a wider array of confounders are called for.
Collapse
Affiliation(s)
- Tyra Lagerberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, UK.
| | - Anthony A Matthews
- Unit of Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Nanbo Zhu
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Seena Fazel
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, UK
- Oxford Health NHS Foundation Trust, Oxford, UK
| | - Juan-Jesus Carrero
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Zheng Chang
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
64
|
Colacci M, Fralick M. Methodological progress note: Utilizing inpatient data sources to minimize common biases when emulating target trials. J Hosp Med 2023; 18:1017-1020. [PMID: 37469234 DOI: 10.1002/jhm.13171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 07/06/2023] [Accepted: 07/08/2023] [Indexed: 07/21/2023]
Affiliation(s)
- Michael Colacci
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Institute of Health Policy, Management, and Evaluation, University of Toronto, Toronto, Ontario, Canada
| | - Michael Fralick
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, Mount Sinai Hospital, Sinai Health, Toronto, Ontario, Canada
| |
Collapse
|
65
|
Matilla-Santander N, Matthews AA, Gunn V, Muntaner C, Kreshpaj B, Wegman DH, Sánchez-Martínez N, Hernando-Rodriguez JC, Albin M, Balogh R, Davis L, Bodin T. Causal effect of shifting from precarious to standard employment on all-cause mortality in Sweden: an emulation of a target trial. J Epidemiol Community Health 2023; 77:736-743. [PMID: 37620008 PMCID: PMC10579471 DOI: 10.1136/jech-2023-220734] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 08/05/2023] [Indexed: 08/26/2023]
Abstract
BACKGROUND We aimed at estimating the causal effect of switching from precarious to standard employment on the 6-year and 12-year risk of all-cause mortality among workers aged 20-55 years in Sweden. METHODS We emulated a series of 12 target trials starting every year between 2005 and 2016 using Swedish register data (n=251 273). We classified precariously employed individuals using a multidimensional approach at baseline as (1) remaining in precarious employment (PE) (73.8%) and (2) shifting to standard employment (26.2%). All-cause mortality was measured from 2006 to 2017. We pooled data for all 12 emulated trials and used covariate-adjusted pooled logistic regression to estimate intention-to-treat and per-protocol effects via risk ratios (RRs) and standardised risk curves (the parametric g-formula). RESULTS Shifting from precarious to standard employment decreases the 12-year risk of death by 20% on the relative scale (RR: 0.82, 95% CI: 0.73; 0.93), regardless of what happens after the initial shift. However, we estimated a 12-year risk reduction of 30% on the relative scale for workers shifting from precarious to standard employment and staying within this employment category for the full 12 years (RR: 0.71, 95% CI: 0.54; 0.95). CONCLUSIONS This study finds that shifting from low to higher-quality employment conditions (ie, stable employment, sufficient income levels and high coverage by collective agreements) decreases the risk of death. Remaining in PE increases the risk of premature mortality. Our results emphasise the necessity of ensuring decent work for the entire working population to accomplish the 2030 Agenda for Sustainable Development.
Collapse
Affiliation(s)
| | - Anthony A Matthews
- Unit of Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Virginia Gunn
- Unit of Occupational Medicine, Karolinska Institute, Stockholm, Sweden
- MAP Centre for Urban Health Solutions, Li Ka Shing Knowledge Institute, Unity Health Toronto, Toronto, Ontario, Canada
- School of Nursing, Cape Breton University, Sydney, New South Wales, Canada
| | - Carles Muntaner
- Bllomberg Faculty of Nursing, Division of Social and Behavioural Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Bertina Kreshpaj
- Unit of Occupational Medicine, Karolinska Institute, Stockholm, Sweden
- Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - David H Wegman
- Public Health, University of Massachusetts Lowell, Lowell, Massachusetts, USA
| | - Néstor Sánchez-Martínez
- Unit of Occupational Medicine, Karolinska Institute, Stockholm, Sweden
- Public Health, Universitat Internacional de Catalunya, Barcelona, Spain
| | | | - Maria Albin
- Unit of Occupational Medicine, Karolinska Institute, Stockholm, Sweden
- Centre for Occupational and Environmental Medicine, Stockholm Region, Stockholm, Sweden
| | - Rebeka Balogh
- Interface Demography, Vrije Universiteit Brussel, Brussels, Belgium
- Institute for Employment Research, University of Warwick, Coventry, UK
| | | | - Theo Bodin
- Unit of Occupational Medicine, Karolinska Institute, Stockholm, Sweden
- Centre for Occupational and Environmental Medicine, Stockholm Region, Stockholm, Sweden
| |
Collapse
|
66
|
Robertson DS, Wason JM, Ramdas A. Online multiple hypothesis testing. Stat Sci 2023; 38:557-575. [PMID: 38223302 PMCID: PMC7615519 DOI: 10.1214/23-sts901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
Modern data analysis frequently involves large-scale hypothesis testing, which naturally gives rise to the problem of maintaining control of a suitable type I error rate, such as the false discovery rate (FDR). In many biomedical and technological applications, an additional complexity is that hypotheses are tested in an online manner, one-by-one over time. However, traditional procedures that control the FDR, such as the Benjamini-Hochberg procedure, assume that all p-values are available to be tested at a single time point. To address these challenges, a new field of methodology has developed over the past 15 years showing how to control error rates for online multiple hypothesis testing. In this framework, hypotheses arrive in a stream, and at each time point the analyst decides whether to reject the current hypothesis based both on the evidence against it, and on the previous rejection decisions. In this paper, we present a comprehensive exposition of the literature on online error rate control, with a review of key theory as well as a focus on applied examples. We also provide simulation results comparing different online testing algorithms and an up-to-date overview of the many methodological extensions that have been proposed.
Collapse
Affiliation(s)
| | - James M.S. Wason
- Population Health Sciences Institute, Newcastle University, Newcastle, UK
| | - Aaditya Ramdas
- Departments of Statistics and Machine Learning, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
67
|
Birnie K, Tomson C, Caskey FJ, Ben-Shlomo Y, Nitsch D, Casula A, Murray EJ, Sterne JAC. Comparative Effectiveness of Dynamic Treatment Strategies for Medication Use and Dosage: Emulating a Target Trial Using Observational Data. Epidemiology 2023; 34:879-887. [PMID: 37757876 PMCID: PMC7615288 DOI: 10.1097/ede.0000000000001649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2023]
Abstract
BACKGROUND Availability of detailed data from electronic health records (EHRs) has increased the potential to examine the comparative effectiveness of dynamic treatment strategies using observational data. Inverse probability (IP) weighting of dynamic marginal structural models can control for time-varying confounders. However, IP weights for continuous treatments may be sensitive to model choice. METHODS We describe a target trial comparing strategies for treating anemia with darbepoetin in hemodialysis patients using EHR data from the UK Renal Registry 2004 to 2016. Patients received a specified dose (microgram/week) or did not receive darbepoetin. We compared 4 methods for modeling time-varying treatment: (A) logistic regression for zero dose, standard linear regression for log dose; (B) logistic regression for zero dose, heteroscedastic linear regression for log dose; (C) logistic regression for zero dose, heteroscedastic linear regression for log dose, multinomial regression for patients who recently received very low or high doses; and (D) ordinal logistic regression. RESULTS For this dataset, method (C) was the only approach that provided a robust estimate of the mortality hazard ratio (HR), with less-extreme weights in a fully weighted analysis and no substantial change of the HR point estimate after weight truncation. After truncating IP weights at the 95th percentile, estimates were similar across the methods. CONCLUSIONS EHR data can be used to emulate target trials estimating the comparative effectiveness of dynamic strategies adjusting treatment to evolving patient characteristics. However, model checking, monitoring of large weights, and adaptation of model strategies to account for these is essential if an aspect of treatment is continuous.
Collapse
Affiliation(s)
- Kate Birnie
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Charles Tomson
- Department of Renal Medicine, Freeman Hospital, Newcastle upon Tyne, UK
| | - Fergus J Caskey
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Department of Renal Medicine, North Bristol NHS Trust, Bristol, UK
| | - Yoav Ben-Shlomo
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Dorothea Nitsch
- Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
- Department of Nephrology, Royal Free London NHS Foundation Trust, London, UK
| | - Anna Casula
- UK Renal Registry, UK Kidney Association, Bristol, UK
| | - Eleanor J Murray
- Department of Epidemiology, School of Public Health, Boston University, Boston, MA, USA
| | - Jonathan AC Sterne
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Health Data Research UK South-West
- NIHR Bristol Biomedical Research Centre, Bristol, UK
| |
Collapse
|
68
|
Sheridan P, Chen C, Thompson CA, Benmarhnia T. Immortal Time Bias With Time-Varying Exposures in Environmental Epidemiology: A Case Study in Lung Cancer Survival. Am J Epidemiol 2023; 192:1754-1762. [PMID: 37400995 PMCID: PMC10558188 DOI: 10.1093/aje/kwad135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 01/19/2023] [Accepted: 06/04/2023] [Indexed: 07/05/2023] Open
Abstract
Immortal time bias is a well-recognized bias in clinical epidemiology but is rarely discussed in environmental epidemiology. Under the target trial framework, this bias is formally conceptualized as a misalignment between the start of study follow-up (time 0) and treatment assignment. This misalignment can occur when attained duration of follow-up is encoded into treatment assignment using minimums, maximums, or averages. The bias can be exacerbated in the presence of time trends commonly found in environmental exposures. Using lung cancer cases from the California Cancer Registry (2000-2010) linked with estimated concentrations of particulate matter less than or equal to 2.5 μm in aerodynamic diameter (PM2.5), we replicated previous studies that averaged PM2.5 exposure over follow-up in a time-to-event model. We compared this approach with one that ensures alignment between time 0 and treatment assignment, a discrete-time approach. In the former approach, the estimated overall hazard ratio for a 5-μg/m3 increase in PM2.5 was 1.38 (95% confidence interval: 1.36, 1.40). Under the discrete-time approach, the estimated pooled odds ratio was 0.99 (95% confidence interval: 0.98, 1.00). We conclude that the strong estimated effect in the former approach was likely driven by immortal time bias, due to misalignment at time 0. Our findings highlight the importance of appropriately conceptualizing a time-varying environmental exposure under the target trial framework to avoid introducing preventable systematic errors.
Collapse
Affiliation(s)
- Paige Sheridan
- Correspondence to Dr. Paige Sheridan, Herbert Wertheim School of Public Health, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093 (e-mail: )
| | | | | | | |
Collapse
|
69
|
Pegram C, Diaz-Ordaz K, Brodbelt DC, Chang YM, Tayler S, Allerton F, Prisk L, Church DB, O’Neill DG. Target trial emulation: Do antimicrobials or gastrointestinal nutraceuticals prescribed at first presentation for acute diarrhoea cause a better clinical outcome in dogs under primary veterinary care in the UK? PLoS One 2023; 18:e0291057. [PMID: 37792702 PMCID: PMC10550114 DOI: 10.1371/journal.pone.0291057] [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: 06/06/2023] [Accepted: 08/21/2023] [Indexed: 10/06/2023] Open
Abstract
Target trial emulation applies design principles from randomised controlled trials to the analysis of observational data for causal inference and is increasingly used within human epidemiology. Veterinary electronic clinical records represent a potentially valuable source of information to estimate real-world causal effects for companion animal species. This study employed the target trial framework to evaluate the usefulness on veterinary observational data. Acute diarrhoea in dogs was used as a clinical exemplar. Inclusion required dogs aged ≥ 3 months and < 10 years, presenting for veterinary primary care with acute diarrhoea during 2019. Treatment strategies were: 1. antimicrobial prescription compared to no antimicrobial prescription and 2. gastrointestinal nutraceutical prescription compared to no gastrointestinal nutraceutical prescription. The primary outcome was clinical resolution (defined as no revisit with ongoing diarrhoea within 30 days from the date of first presentation). Informed from a directed acyclic graph, data on the following covariates were collected: age, breed, bodyweight, insurance status, comorbidities, vomiting, reduced appetite, haematochezia, pyrexia, duration, additional treatment prescription and veterinary group. Inverse probability of treatment weighting was used to balance covariates between the treatment groups for each of the two target trials. The risk difference (RD) of 0.4% (95% CI -4.5% to 5.3%) was non-significant for clinical resolution in dogs treated with antimicrobials compared with dogs not treated with antimicrobials. The risk difference (RD) of 0.3% (95% CI -4.5% to 5.0%) was non-significant for clinical resolution in dogs treated with gastrointestinal nutraceuticals compared with dogs not treated with gastrointestinal nutraceuticals. This study successfully applied the target trial framework to veterinary observational data. The findings show that antimicrobial or gastrointestinal prescription at first presentation of acute diarrhoea in dogs causes no difference in clinical resolution. The findings support the recommendation for veterinary professionals to limit antimicrobial use for acute diarrhoea in dogs.
Collapse
Affiliation(s)
- Camilla Pegram
- Pathobiology and Population Sciences, The Royal Veterinary College, Hatfield, Herts, United Kingdom
| | - Karla Diaz-Ordaz
- Department of Statistical Science, University College London, London, United Kingdom
| | - Dave C. Brodbelt
- Pathobiology and Population Sciences, The Royal Veterinary College, Hatfield, Herts, United Kingdom
| | - Yu-Mei Chang
- Research Support Office, The Royal Veterinary College, Hatfield, Herts, United Kingdom
| | - Sarah Tayler
- Clinical Sciences and Services, The Royal Veterinary College, Hatfield, Herts, United Kingdom
| | - Fergus Allerton
- Willows Veterinary Centre & Referral Centre, Solihull, United Kingdom
| | - Lauren Prisk
- Pathobiology and Population Sciences, The Royal Veterinary College, Hatfield, Herts, United Kingdom
| | - David B. Church
- Clinical Sciences and Services, The Royal Veterinary College, Hatfield, Herts, United Kingdom
| | - Dan G. O’Neill
- Pathobiology and Population Sciences, The Royal Veterinary College, Hatfield, Herts, United Kingdom
| |
Collapse
|
70
|
Nicolajsen CW, Søgaard M, Jensen M, Eldrup N, Larsen TB, Goldhaber SZ, Behrendt CA, Nielsen PB. Antiplatelet Therapy in Patients With Abdominal Aortic Aneurysm Without Symptomatic Atherosclerotic Disease. JAMA Netw Open 2023; 6:e2339715. [PMID: 37878310 PMCID: PMC10600585 DOI: 10.1001/jamanetworkopen.2023.39715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 09/13/2023] [Indexed: 10/26/2023] Open
Abstract
Importance Patients with abdominal aortic aneurysm have a high risk of ischemic events associated with concomitant atherosclerotic cardiovascular disease, and current clinical practice guidelines recommend antiplatelet therapy to mitigate this risk. However, in patients with aneurysms without symptomatic atherosclerosis, the benefit of antiplatelet therapy has been sparsely investigated. Objective To estimate the effect of antiplatelets on the risk of ischemic events and bleeding in individuals with abdominal aneurysms with no symptomatic atherosclerotic vascular disease. Design, Setting, and Participants A comparative effectiveness research study using a target trial emulation framework was performed. Population-based, cross-linked observational data from Danish national health registries containing comprehensive, individual-level information on all Danish citizens were used to evaluate patients who were antiplatelet-naive and diagnosed with abdominal aortic aneurysms, with no record of symptomatic atherosclerotic vascular disease, from January 1, 2010, through August 21, 2021. Exposure Prescription filled for aspirin or clopidogrel. Main Outcomes and Measures Risk of ischemic events (myocardial infarction and/or ischemic stroke) and risk of major bleeding. For target trial emulation, trials were emulated as sequential, contingent on patient eligibility at the time of inclusion, and were evaluated by means of pooled logistic regression models to estimate the intention-to-treat and as-treated effects, expressed as hazard ratio (HR) and event-free survival. Results A total of 6344 patients (65.2% men; age, 72 [IQR, 64-78] years) provided 131 047 trial cases; 3363 of these cases involved initiation of antiplatelet therapy and 127 684 did not. A total of 182 ischemic events occurred among initiators and 5602 ischemic events occurred among noninitiators, corresponding to an intention-to-treat HR of 0.91 (95% CI, 0.73-1.17) and an estimated absolute event-free survival difference of -0.6% (95% CI, -1.7% to 0.5%). After censoring nonadherent person-time, the treatment HR was 0.90 (95% CI, 0.68-1.20), with similar risk difference. For bleeding, the intention-to-treat HR was 1.26 (95% CI, 0.97-1.58) and the event-free survival difference was 1.0%. The treatment HR was 1.21 (95% CI, 0.82-1.72); the risk difference was similar. Conclusions and Relevance In this study, no evidence of effectiveness of antiplatelet therapy to lower the risk of ischemic events and a trend toward higher bleeding risk was noted. The observed differences between the treatment groups were minimal, suggesting limited clinical relevance of antiplatelet treatment.
Collapse
Affiliation(s)
- Chalotte W. Nicolajsen
- Department of Cardiology, Aalborg University Hospital, Aalborg, Denmark
- Aalborg Thrombosis Research Unit, Department of Clinical Medicine, Faculty of Health, Aalborg University, Aalborg, Denmark
- Department of Vascular Surgery, Viborg Regional Hospital, Viborg, Denmark
| | - Mette Søgaard
- Department of Cardiology, Aalborg University Hospital, Aalborg, Denmark
- Aalborg Thrombosis Research Unit, Department of Clinical Medicine, Faculty of Health, Aalborg University, Aalborg, Denmark
| | - Martin Jensen
- Department of Cardiology, Aalborg University Hospital, Aalborg, Denmark
| | - Nikolaj Eldrup
- Department of Vascular Surgery, Rigshospitalet, Copenhagen, Denmark
- Institute of Clinical Medicine, Copenhagen University, Copenhagen, Denmark
| | - Torben B. Larsen
- Department of Cardiology, Aalborg University Hospital, Aalborg, Denmark
- Aalborg Thrombosis Research Unit, Department of Clinical Medicine, Faculty of Health, Aalborg University, Aalborg, Denmark
| | - Samuel Z. Goldhaber
- Thrombosis Research Group, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Christian-Alexander Behrendt
- Department of Vascular and Endovascular Surgery, Asklepios Medical School, Asklepios Clinic Wandsbek, Hamburg, Germany
- Division of Cardiovascular Medicine, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
| | - Peter B. Nielsen
- Department of Cardiology, Aalborg University Hospital, Aalborg, Denmark
- Aalborg Thrombosis Research Unit, Department of Clinical Medicine, Faculty of Health, Aalborg University, Aalborg, Denmark
| |
Collapse
|
71
|
Dehaene I, Steen J, Dukes O, Olarte Parra C, De Coen K, Smets K, Roelens K, Decruyenaere J. On optimal timing of antenatal corticosteroids: time to reformulate the question. Arch Gynecol Obstet 2023; 308:1085-1091. [PMID: 36738316 DOI: 10.1007/s00404-023-06941-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 01/18/2023] [Indexed: 02/05/2023]
Abstract
Administration of antenatal corticosteroids (ACS) for accelerating foetal lung maturation in threatened preterm birth is one of the cornerstones of prevention of neonatal mortality and morbidity. To identify the optimal timing of ACS administration, most studies have compared subgroups based on treatment-to-delivery intervals. Such subgroup analysis of the first placebo-controlled randomised controlled trial indicated that a one to seven day interval between ACS administration and birth resulted in the lowest rates of neonatal respiratory distress syndrome. This efficacy window was largely confirmed by a series of subgroup analyses of subsequent trials and observational studies and strongly influenced obstetric management. However, these subgroup analyses suffer from a methodological flaw that often seems to be overlooked and potentially has important consequences for drawing valid conclusions. In this commentary, we point out that studies comparing treatment outcomes between subgroups that are retrospectively identified at birth (i.e. after randomisation) may not only be plagued by post-randomisation confounding bias but, more importantly, may not adequately inform decision making before birth, when the projected duration of the interval is still unknown. We suggest two more formal interpretations of these subgroup analyses, using a counterfactual framework for causal inference, and demonstrate that each of these interpretations can be linked to a different hypothetical trial. However, given the infeasibility of these trials, we argue that none of these rescue interpretations are helpful for clinical decision making. As a result, guidelines based on these subgroup analyses may have led to suboptimal clinical practice. As an alternative to these flawed subgroup analyses, we suggest a more principled approach that clearly formulates the question about optimal timing of ACS treatment in terms of the protocol of a future randomised study. Even if this 'target trial' would never be conducted, its protocol may still provide important guidance to avoid repeating common design flaws when conducting observational 'real world' studies using statistical methods for causal inference.
Collapse
Affiliation(s)
- Isabelle Dehaene
- Obstetrics and Gynaecology, Ghent University Hospital, Corneel Heymanslaan 10, Ghent, Belgium.
| | - Johan Steen
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Renal Division, Ghent University Hospital, Ghent, Belgium
- Department of Intensive Care Medicine, Ghent University Hospital, Ghent, Belgium
| | - Oliver Dukes
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Camila Olarte Parra
- Department of Medical Statistics, London School of Hygiene and Tropical Medicine, London, UK
| | - Kris De Coen
- Neonatal Intensive Care Unit, Ghent University Hospital, Ghent, Belgium
| | - Koenraad Smets
- Neonatal Intensive Care Unit, Ghent University Hospital, Ghent, Belgium
| | - Kristien Roelens
- Obstetrics and Gynaecology, Ghent University Hospital, Corneel Heymanslaan 10, Ghent, Belgium
| | | |
Collapse
|
72
|
Granger E, Davies G, Keogh RH. Emulated trial investigating effects of multiple treatments: estimating combined effects of mucoactive nebulisers in cystic fibrosis using registry data. Thorax 2023; 78:1011-1018. [PMID: 37451864 PMCID: PMC10511967 DOI: 10.1136/thorax-2023-220031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 06/14/2023] [Indexed: 07/18/2023]
Abstract
INTRODUCTION People with cystic fibrosis (CF) are often on multiple long-term treatments, including mucoactive nebulisers. In the UK, the most common mucoactive nebuliser is dornase alfa (DNase). A common therapeutic approach for people already on DNase is to add hypertonic saline (HS). The effects of DNase and HS used alone have been studied in randomised trials, but their effects in combination have not. This study investigates whether, for people already prescribed DNase, adding HS has additional benefit for lung function or use of intravenous antibiotics. METHODS Using UK CF Registry data from 2007 to 2018, we emulated a target trial. We included people aged 6 years and over who were prescribed DNase without HS for 2 years. We investigated the effects of combinations of DNase and HS over 5 years of follow-up. Inverse-probability-of-treatment weighting was used to control confounding. The period predated triple combination CF transmembrane conductance regulator modulators in routine care. RESULTS 4498 individuals were included. At baseline, average age and forced expiratory volume in 1 s (FEV1%) predicted were 21.1 years and 69.7 respectively. During first year of follow-up, 3799 individuals were prescribed DNase alone; 426 added HS; 57 switched to HS alone and 216 were prescribed neither. We found no evidence that adding HS improved FEV1% at 1-5 years, or use of intravenous antibiotics at 1-4 years, compared with DNase alone. CONCLUSION For individuals with CF prescribed DNase, we found no evidence that adding HS had an effect on FEV1% or prescription of intravenous antibiotics. Our study illustrates the emulated target trial approach using CF Registry data.
Collapse
Affiliation(s)
- Emily Granger
- Department of Medical Statistics, London School of Hygiene & Tropical Medicine, London, UK
| | - Gwyneth Davies
- UCL Great Ormond Street Institute of Child Health, UCL, London, UK
- Respiratory Medicine, Great Ormond Street Hospital For Children NHS Foundation Trust, London, UK
| | - Ruth H Keogh
- Department of Medical Statistics, London School of Hygiene & Tropical Medicine, London, UK
| |
Collapse
|
73
|
Wanis KN, Madenci AL, Hao S, Moukheiber M, Moukheiber L, Moukheiber D, Moukheiber S, Young JG, Celi LA. Emulating Target Trials Comparing Early and Delayed Intubation Strategies. Chest 2023; 164:885-891. [PMID: 37150505 PMCID: PMC10567927 DOI: 10.1016/j.chest.2023.04.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 04/15/2023] [Accepted: 04/30/2023] [Indexed: 05/09/2023] Open
Abstract
BACKGROUND Whether intubation should be initiated early in the clinical course of critically ill patients remains a matter of debate. Results from prior observational studies are difficult to interpret because of avoidable flaws including immortal time bias, inappropriate eligibility criteria, and unrealistic treatment strategies. RESEARCH QUESTION Do treatment strategies that intubate patients early in the critical care admission improve 30-day survival compared with strategies that delay intubation? STUDY DESIGN AND METHODS We estimated the effect of strategies that require early intubation of critically ill patients compared with those that delay intubation. With data extracted from the Medical Information Mart for Intensive Care-IV database, we emulated three target trials, varying the flexibility of the treatment strategies and the baseline eligibility criteria. RESULTS Under unrealistically strict treatment strategies with broad eligibility criteria, the 30-day mortality risk was 7.1 percentage points higher for intubating early compared with delaying intubation (95% CI, 6.2-7.9). Risk differences were 0.4 (95% CI, -0.1 to 0.9) and -0.9 (95% CI, -2.5 to 0.7) percentage points in subsequent target trial emulations that included more realistic treatment strategies and eligibility criteria. INTERPRETATION When realistic treatment strategies and eligibility criteria are used, strategies that delay intubation result in similar 30-day mortality risks compared with those that intubate early. Delaying intubation ultimately avoids intubation in most patients.
Collapse
Affiliation(s)
- Kerollos Nashat Wanis
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA; Division of General Surgery, Department of Surgery, Western University, London, ON, Canada.
| | - Arin L Madenci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA; Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Sicheng Hao
- Laboratory for Computational Physiology, Massachusetts Institute of Technology, Cambridge, MA
| | - Mira Moukheiber
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA
| | - Lama Moukheiber
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA
| | - Dana Moukheiber
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA
| | - Sulaiman Moukheiber
- Department of Computer Science, Worcester Polytechnic Institute, Worcester, MA
| | - Jessica G Young
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA
| | - Leo Anthony Celi
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA; Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA; Laboratory for Computational Physiology, Massachusetts Institute of Technology, Cambridge, MA
| |
Collapse
|
74
|
Zuo H, Yu L, Campbell SM, Yamamoto SS, Yuan Y. The implementation of target trial emulation for causal inference: a scoping review. J Clin Epidemiol 2023; 162:29-37. [PMID: 37562726 DOI: 10.1016/j.jclinepi.2023.08.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 07/25/2023] [Accepted: 08/02/2023] [Indexed: 08/12/2023]
Abstract
OBJECTIVES We aim to investigate the implementation of Target Trial Emulation (TTE) for causal inference, involving research topics, frequently used strategies, and issues indicating the need for future improvements. STUDY DESIGN AND SETTING We performed a scoping review by following the Joanna Briggs Institute (JBI) guidance and Preferred Reporting Items for Systematic Reviews and Meta-Analyses extension for Scoping Reviews (PRISMA-ScR) checklist. A health research-focused librarian searched multiple medical databases, and two independent reviewers completed screening and extraction within covidence review management software. RESULTS Our search resulted in 1,240 papers, of which 96 papers were eligible for data extraction. Results show a significant increase in the use of TTE in 2018 and 2021. The study topics varied and focused primarily on cancer, cardiovascular and cerebrovascular diseases, and infectious diseases. However, not all papers specified well all three critical components for generating robust causal evidence: time-zero, random assignment simulation, and comparison strategy. Some common issues were observed from retrieved papers, and key limitations include residual confounding, limited generalizability, and a lack of reporting guidance that need to be improved. CONCLUSION Uneven adherence to the TTE framework exists, and future improvements are needed to progress applications using causal inference with observational data.
Collapse
Affiliation(s)
- Hanxiao Zuo
- School of Public Health, University of Alberta, Edmonton, Alberta T6G 1C9, Canada.
| | - Lin Yu
- School of Public Health, University of Alberta, Edmonton, Alberta T6G 1C9, Canada
| | - Sandra M Campbell
- John W. Scott Health Sciences Library, University of Alberta, Edmonton, Alberta T6G 2R7, Canada
| | - Shelby S Yamamoto
- School of Public Health, University of Alberta, Edmonton, Alberta T6G 1C9, Canada
| | - Yan Yuan
- School of Public Health, University of Alberta, Edmonton, Alberta T6G 1C9, Canada
| |
Collapse
|
75
|
Zaky MY, Fan C, Zhang H, Sun XF. Unraveling the Anticancer Potential of Statins: Mechanisms and Clinical Significance. Cancers (Basel) 2023; 15:4787. [PMID: 37835481 PMCID: PMC10572000 DOI: 10.3390/cancers15194787] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Statins are an essential medication class in the treatment of lipid diseases because they inhibit 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase. They reduce cholesterol levels and reduce the risk of cardiovascular disease in both primary and secondary prevention. In addition to their powerful pharmacologic suppression of cholesterol production, statins appear to have pleitropic effects in a wide variety of other diseases by modulating signaling pathways. In recent years, statins have seen a large increase in interest due to their putative anticancer effects. Statins appear to cause upregulation or inhibition in key pathways involved in cancer such as inhibition of proliferation, angiogenesis, and metastasis as well as reducing cancer stemness. Further, statins have been found to induce oxidative stress, cell cycle arrest, autophagy, and apoptosis of cancer cells. Interestingly, clinical studies have shown that statin use is associated with a decreased risk of cancer formation, lower cancer grade at diagnosis, reduction in the risk of local reoccurrence, and increasing survival in patients. Therefore, our objective in the present review is to summarize the findings of the publications on the underlying mechanisms of statins' anticancer effects and their clinical implications.
Collapse
Affiliation(s)
- Mohamed Y. Zaky
- Department of Oncology, Linköping University, 581 83 Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, 581 83 Linköping, Sweden
- Molecular Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef 62521, Egypt
| | - Chuanwen Fan
- Department of Oncology, Linköping University, 581 83 Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, 581 83 Linköping, Sweden
| | - Huan Zhang
- Department of Oncology, Linköping University, 581 83 Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, 581 83 Linköping, Sweden
| | - Xiao-Feng Sun
- Department of Oncology, Linköping University, 581 83 Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, 581 83 Linköping, Sweden
| |
Collapse
|
76
|
Hansford HJ, Cashin AG, Jones MD, Swanson SA, Islam N, Dahabreh IJ, Dickerman BA, Egger M, Garcia-Albeniz X, Golub RM, Lodi S, Moreno-Betancur M, Pearson SA, Schneeweiss S, Sterne J, Sharp MK, Stuart EA, Hernan MA, Lee H, McAuley JH. Development of the TrAnsparent ReportinG of observational studies Emulating a Target trial (TARGET) guideline. BMJ Open 2023; 13:e074626. [PMID: 37699620 PMCID: PMC10503363 DOI: 10.1136/bmjopen-2023-074626] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 08/25/2023] [Indexed: 09/14/2023] Open
Abstract
BACKGROUND Observational studies are increasingly used to inform health decision-making when randomised trials are not feasible, ethical or timely. The target trial approach provides a framework to help minimise common biases in observational studies that aim to estimate the causal effect of interventions. Incomplete reporting of studies using the target trial framework limits the ability for clinicians, researchers, patients and other decision-makers to appraise, synthesise and interpret findings to inform clinical and public health practice and policy. This paper describes the methods that we will use to develop the TrAnsparent ReportinG of observational studies Emulating a Target trial (TARGET) reporting guideline. METHODS/DESIGN The TARGET reporting guideline will be developed in five stages following recommended guidance. The first stage will identify target trial reporting practices by systematically reviewing published studies that explicitly emulated a target trial. The second stage will identify and refine items to be considered for inclusion in the TARGET guideline by consulting content experts using sequential online surveys. The third stage will prioritise and consolidate key items to be included in the TARGET guideline at an in-person consensus meeting of TARGET investigators. The fourth stage will produce and pilot-test both the TARGET guideline and explanation and elaboration document with relevant stakeholders. The fifth stage will disseminate the TARGET guideline and resources via journals, conferences and courses. ETHICS AND DISSEMINATION Ethical approval for the survey has been attained (HC220536). The TARGET guideline will be disseminated widely in partnership with stakeholders to maximise adoption and improve reporting of these studies.
Collapse
Affiliation(s)
- Harrison J Hansford
- School of Health Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
- Centre for Pain IMPACT, Neuroscience Research Australia, Randwick, New South Wales, Australia
| | - Aidan G Cashin
- School of Health Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
- Centre for Pain IMPACT, Neuroscience Research Australia, Randwick, New South Wales, Australia
| | - Matthew D Jones
- School of Health Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
- Centre for Pain IMPACT, Neuroscience Research Australia, Randwick, New South Wales, Australia
| | - Sonja A Swanson
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- CAUSALab, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Nazrul Islam
- Oxford Population Health, Big Data Institute, University of Oxford, Oxford, UK
- Faculty of Medicine, University of Southampton, Southampton, UK
| | - Issa J Dahabreh
- CAUSALab, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Barbra A Dickerman
- CAUSALab, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Matthias Egger
- Institute of Social & Preventive Medicine, University of Bern, Bern, Switzerland
- Centre for Infectious Disease Epidemiology and Research, University of Cape Town Faculty of Health Sciences, Observatory, Western Cape, South Africa
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Xavier Garcia-Albeniz
- CAUSALab, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- RTI Health Solutions Barcelona, Barcelona, Catalunya, Spain
| | - Robert M Golub
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Sara Lodi
- CAUSALab, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Margarita Moreno-Betancur
- Clinical Epidemiology & Biostatistics Unit, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
| | - Sallie-Anne Pearson
- School of Population Health, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Sebastian Schneeweiss
- Division of Pharmacoepidemiology, Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jonathan Sterne
- Department of Population Health Sciences, University of Bristol, Bristol, UK
- NIHR Bristol Biomedical Research Centre, Bristol, UK
- Health Data Research UK South-West, Bristol, UK
| | - Melissa K Sharp
- Department of Public Health and Epidemiology, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Elizabeth A Stuart
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Miguel A Hernan
- CAUSALab, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Hopin Lee
- University of Exeter Medical School, Exeter, UK
- EMEA Methods and Evidence Generation, IQVIA London, London, UK
| | - James H McAuley
- School of Health Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
- Centre for Pain IMPACT, Neuroscience Research Australia, Randwick, New South Wales, Australia
| |
Collapse
|
77
|
Hansford HJ, Cashin AG, Jones MD, Swanson SA, Islam N, Douglas SRG, Rizzo RRN, Devonshire JJ, Williams SA, Dahabreh IJ, Dickerman BA, Egger M, Garcia-Albeniz X, Golub RM, Lodi S, Moreno-Betancur M, Pearson SA, Schneeweiss S, Sterne JAC, Sharp MK, Stuart EA, Hernán MA, Lee H, McAuley JH. Reporting of Observational Studies Explicitly Aiming to Emulate Randomized Trials: A Systematic Review. JAMA Netw Open 2023; 6:e2336023. [PMID: 37755828 PMCID: PMC10534275 DOI: 10.1001/jamanetworkopen.2023.36023] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 08/22/2023] [Indexed: 09/28/2023] Open
Abstract
Importance Observational (nonexperimental) studies that aim to emulate a randomized trial (ie, the target trial) are increasingly informing medical and policy decision-making, but it is unclear how these studies are reported in the literature. Consistent reporting is essential for quality appraisal, evidence synthesis, and translation of evidence to policy and practice. Objective To assess the reporting of observational studies that explicitly aimed to emulate a target trial. Evidence Review We searched Medline, Embase, PsycINFO, and Web of Science for observational studies published between March 2012 and October 2022 that explicitly aimed to emulate a target trial of a health or medical intervention. Two reviewers double-screened and -extracted data on study characteristics, key predefined components of the target trial protocol and its emulation (eligibility criteria, treatment strategies, treatment assignment, outcome[s], follow-up, causal contrast[s], and analysis plan), and other items related to the target trial emulation. Findings A total of 200 studies that explicitly aimed to emulate a target trial were included. These studies included 26 subfields of medicine, and 168 (84%) were published from January 2020 to October 2022. The aim to emulate a target trial was explicit in 70 study titles (35%). Forty-three studies (22%) reported use of a published reporting guideline (eg, Strengthening the Reporting of Observational Studies in Epidemiology). Eighty-five studies (43%) did not describe all key items of how the target trial was emulated and 113 (57%) did not describe the protocol of the target trial and its emulation. Conclusion and Relevance In this systematic review of 200 studies that explicitly aimed to emulate a target trial, reporting of how the target trial was emulated was inconsistent. A reporting guideline for studies explicitly aiming to emulate a target trial may improve the reporting of the target trial protocols and other aspects of these emulation attempts.
Collapse
Affiliation(s)
- Harrison J. Hansford
- School of Health Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, Australia
- Centre for Pain IMPACT, Neuroscience Research Australia, Sydney, Australia
| | - Aidan G. Cashin
- School of Health Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, Australia
- Centre for Pain IMPACT, Neuroscience Research Australia, Sydney, Australia
| | - Matthew D. Jones
- School of Health Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, Australia
- Centre for Pain IMPACT, Neuroscience Research Australia, Sydney, Australia
| | - Sonja A. Swanson
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, Pennsylvania
- CAUSALab, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Nazrul Islam
- Oxford Population Health, Big Data Institute, University of Oxford, Oxford, United Kingdom
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Susan R. G. Douglas
- School of Health Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, Australia
| | - Rodrigo R. N. Rizzo
- School of Health Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, Australia
- Centre for Pain IMPACT, Neuroscience Research Australia, Sydney, Australia
| | - Jack J. Devonshire
- Centre for Pain IMPACT, Neuroscience Research Australia, Sydney, Australia
| | - Sam A. Williams
- Centre for Pain IMPACT, Neuroscience Research Australia, Sydney, Australia
| | - Issa J. Dahabreh
- CAUSALab, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Barbra A. Dickerman
- CAUSALab, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Matthias Egger
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
- Centre for Infectious Disease Epidemiology and Research, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Xabier Garcia-Albeniz
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- RTI Health Solutions, Barcelona, Spain
| | - Robert M. Golub
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Sara Lodi
- CAUSALab, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Margarita Moreno-Betancur
- Clinical Epidemiology & Biostatistics Unit, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - Sallie-Anne Pearson
- School of Population Health, Faculty of Medicine and Health, UNSW Sydney, New South Wales, Australia
| | - Sebastian Schneeweiss
- Division of Pharmacoepidemiology, Department of Medicine, Brigham & Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jonathan A. C. Sterne
- Department of Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- NIHR Bristol Biomedical Research Centre, Bristol, United Kingdom
- Health Data Research UK South-West, Bristol, United Kingdom
| | - Melissa K. Sharp
- Department of Public Health and Epidemiology, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Elizabeth A. Stuart
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Miguel A. Hernán
- CAUSALab, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Hopin Lee
- University of Exeter Medical School, Exeter, United Kingdom
| | - James H. McAuley
- School of Health Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, Australia
- Centre for Pain IMPACT, Neuroscience Research Australia, Sydney, Australia
| |
Collapse
|
78
|
Martinuka O, Hazard D, Marateb HR, Maringe C, Mansourian M, Rubio-Rivas M, Wolkewitz M. Target trial emulation with multi-state model analysis to assess treatment effectiveness using clinical COVID-19 data. BMC Med Res Methodol 2023; 23:197. [PMID: 37660025 PMCID: PMC10474639 DOI: 10.1186/s12874-023-02001-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 07/25/2023] [Indexed: 09/04/2023] Open
Abstract
BACKGROUND Real-world observational data are an important source of evidence on the treatment effectiveness for patients hospitalized with coronavirus disease 2019 (COVID-19). However, observational studies evaluating treatment effectiveness based on longitudinal data are often prone to methodological biases such as immortal time bias, confounding bias, and competing risks. METHODS For exemplary target trial emulation, we used a cohort of patients hospitalized with COVID-19 (n = 501) in a single centre. We described the methodology for evaluating the effectiveness of a single-dose treatment, emulated a trial using real-world data, and drafted a hypothetical study protocol describing the main components. To avoid immortal time and time-fixed confounding biases, we applied the clone-censor-weight technique. We set a 5-day grace period as a period of time when treatment could be initiated. We used the inverse probability of censoring weights to account for the selection bias introduced by artificial censoring. To estimate the treatment effects, we took the multi-state model approach. We considered a multi-state model with five states. The primary endpoint was defined as clinical severity status, assessed by a 5-point ordinal scale on day 30. Differences between the treatment group and standard of care treatment group were calculated using a proportional odds model and shown as odds ratios. Additionally, the weighted cause-specific hazards and transition probabilities for each treatment arm were presented. RESULTS Our study demonstrates that trial emulation with a multi-state model analysis is a suitable approach to address observational data limitations, evaluate treatment effects on clinically heterogeneous in-hospital death and discharge alive endpoints, and consider the intermediate state of admission to ICU. The multi-state model analysis allows us to summarize results using stacked probability plots that make it easier to interpret results. CONCLUSIONS Extending the emulated target trial approach to multi-state model analysis complements treatment effectiveness analysis by gaining information on competing events. Combining two methodologies offers an option to address immortal time bias, confounding bias, and competing risk events. This methodological approach can provide additional insight for decision-making, particularly when data from randomized controlled trials (RCTs) are unavailable.
Collapse
Affiliation(s)
- Oksana Martinuka
- Institute of Medical Biometry and Statistics, Faculty of Medicine and Medical Centre, University of Freiburg, Freiburg, Germany.
| | - Derek Hazard
- Institute of Medical Biometry and Statistics, Faculty of Medicine and Medical Centre, University of Freiburg, Freiburg, Germany
| | - Hamid Reza Marateb
- Biomedical Engineering Research Centre (CREB), Automatic Control Department (ESAII), Universitat Politècnica de Catalunya-Barcelona Tech (UPC), Barcelona, Spain
| | - Camille Maringe
- Inequalities in Cancer Outcomes Network (ICON), Department of Non-Communicable Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Marjan Mansourian
- Biomedical Engineering Research Centre (CREB), Automatic Control Department (ESAII), Universitat Politècnica de Catalunya-Barcelona Tech (UPC), Barcelona, Spain
- Department of Epidemiology and Biostatistics, School of Health, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Manuel Rubio-Rivas
- Department of Internal Medicine, Bellvitge University Hospital, Bellvitge Biomedical Research Institute-IDIBELL, University of Barcelona, Barcelona, Spain
| | - Martin Wolkewitz
- Institute of Medical Biometry and Statistics, Faculty of Medicine and Medical Centre, University of Freiburg, Freiburg, Germany
| |
Collapse
|
79
|
Dickerman BA, García-Albéniz X, Logan RW, Denaxas S, Hernán MA. Evaluating Metformin Strategies for Cancer Prevention: A Target Trial Emulation Using Electronic Health Records. Epidemiology 2023; 34:690-699. [PMID: 37227368 PMCID: PMC10524586 DOI: 10.1097/ede.0000000000001626] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
BACKGROUND Metformin users appear to have a substantially lower risk of cancer than nonusers in many observational studies. These inverse associations may be explained by common flaws in observational analyses that can be avoided by explicitly emulating a target trial. METHODS We emulated target trials of metformin therapy and cancer risk using population-based linked electronic health records from the UK (2009-2016). We included individuals with diabetes, no history of cancer, no recent prescription for metformin or other glucose-lowering medication, and hemoglobin A1c (HbA1c) <64 mmol/mol (<8.0%). Outcomes included total cancer and 4 site-specific cancers (breast, colorectal, lung, and prostate). We estimated risks using pooled logistic regression with adjustment for risk factors via inverse-probability weighting. We emulated a second target trial among individuals regardless of diabetes status. We compared our estimates with those obtained using previously applied analytic approaches. RESULTS Among individuals with diabetes, the estimated 6-year risk differences (metformin - no metformin) were -0.2% (95% CI = -1.6%, 1.3%) in the intention-to-treat analysis and 0.0% (95% CI = -2.1%, 2.3%) in the per-protocol analysis. The corresponding estimates for all site-specific cancers were close to zero. Among individuals regardless of diabetes status, these estimates were also close to zero and more precise. By contrast, previous analytic approaches yielded estimates that appeared strongly protective. CONCLUSIONS Our findings are consistent with the hypothesis that metformin therapy does not meaningfully influence cancer incidence. The findings highlight the importance of explicitly emulating a target trial to reduce bias in the effect estimates derived from observational analyses.
Collapse
Affiliation(s)
- Barbra A. Dickerman
- CAUSALab, Harvard T.H. Chan School of Public Health,
Boston, Massachusetts, US
- Department of Epidemiology, Harvard T.H. Chan School of
Public Health, Boston, Massachusetts, US
| | - Xabier García-Albéniz
- CAUSALab, Harvard T.H. Chan School of Public Health,
Boston, Massachusetts, US
- Department of Epidemiology, Harvard T.H. Chan School of
Public Health, Boston, Massachusetts, US
- RTI Health Solutions, Barcelona, Spain
| | - Roger W. Logan
- CAUSALab, Harvard T.H. Chan School of Public Health,
Boston, Massachusetts, US
- Department of Epidemiology, Harvard T.H. Chan School of
Public Health, Boston, Massachusetts, US
| | - Spiros Denaxas
- Institute of Health Informatics Research, University
College London, London, UK
- Health Data Research UK (HDR UK) London, University College
London, London, UK
- The Alan Turing Institute, London, UK
| | - Miguel A. Hernán
- CAUSALab, Harvard T.H. Chan School of Public Health,
Boston, Massachusetts, US
- Department of Epidemiology, Harvard T.H. Chan School of
Public Health, Boston, Massachusetts, US
- Department of Biostatistics, Harvard T.H. Chan School of
Public Health, Boston, Massachusetts, US
| |
Collapse
|
80
|
Sun J, Halfvarson J, Bergman D, Ebrahimi F, Roelstraete B, Lochhead P, Song M, Olén O, Ludvigsson JF. Statin use and risk of colorectal cancer in patients with inflammatory bowel disease. EClinicalMedicine 2023; 63:102182. [PMID: 37662517 PMCID: PMC10474364 DOI: 10.1016/j.eclinm.2023.102182] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/10/2023] [Accepted: 08/11/2023] [Indexed: 09/05/2023] Open
Abstract
Background Statin use has been linked to a reduced risk of advanced colorectal adenomas, but its association with colorectal cancer (CRC) in patients with inflammatory bowel disease (IBD) - a high risk population for CRC - remains inconclusive. Methods From a nationwide IBD cohort in Sweden, we identified 5273 statin users and 5273 non-statin users (1:1 propensity score matching) from July 2006 to December 2018. Statin use was defined as the first filled prescription for ≥30 cumulative defined daily doses and followed until December 2019. Primary outcome was incident CRC. Secondary outcomes were CRC-related mortality and all-cause mortality. Cox regression estimated adjusted hazard ratios (aHRs) and 95% confidence intervals (CIs). Findings During a median follow-up of 5.6 years, 70 statin users (incidence rate (IR): 21.2 per 10,000 person-years) versus 90 non-statin users (IR: 29.2) were diagnosed with incident CRC (rate difference (RD), -8.0 (95% CIs: -15.8 to -0.2 per 10,000 person-years); aHR = 0.76 (95% CIs: 0.61 to 0.96)). The benefit for incident CRC was duration-dependent in a nested case-control design: as compared to short-term use (30 days to <1 year), the adjusted odd ratios were 0.59 (0.25 to 1.43) for 1 to <2 years of use, 0.46 (0.21 to 0.98) for 2 to <5 years of use, and 0.38 (0.16 to 0.86) for ≥5 years of use (Pfor tread = 0.016). Compared with non-statin users, statin users also had a decreased risk for CRC-related mortality (IR: 6.0 vs. 11.9; RD, -5.9 (-10.5 to -1.2); aHR, 0.56 (0.37 to 0.83)) and all-cause mortality (IR: 156.4 vs. 231.4; RD, -75.0 (-96.6 to -53.4); aHR, 0.63 (0.57 to 0.69)). Interpretation Statin use was associated with a lower risk of incident CRC, CRC-related mortality, and all-cause mortality. The benefit for incident CRC was duration-dependent, with a significantly lower risk after ≥2 years of statin use. Funding This research was supported by Forte (i.e., the Swedish Research Council for Health, Working Life and Welfare).
Collapse
Affiliation(s)
- Jiangwei Sun
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Jonas Halfvarson
- Department of Gastroenterology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - David Bergman
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Fahim Ebrahimi
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Gastroenterology and Hepatology, Clarunis - University Center for Gastrointestinal and Liver Diseases, Basel, Switzerland
| | - Bjorn Roelstraete
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | | | - Mingyang Song
- Departments of Epidemiology and Nutrition, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
- Clinical and Translational Epidemiology Unit and Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ola Olén
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Sachs’ Children and Youth Hospital, Stockholm South General Hospital, Stockholm, Sweden
- Department of Clinical Science and Education Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Jonas F. Ludvigsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Pediatrics, Örebro University Hospital, Örebro, Sweden
- Division of Digestive and Liver Disease, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
81
|
Scola G, Chis Ster A, Bean D, Pareek N, Emsley R, Landau S. Implementation of the trial emulation approach in medical research: a scoping review. BMC Med Res Methodol 2023; 23:186. [PMID: 37587484 PMCID: PMC10428565 DOI: 10.1186/s12874-023-02000-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 07/25/2023] [Indexed: 08/18/2023] Open
Abstract
BACKGROUND When conducting randomised controlled trials is impractical, an alternative is to carry out an observational study. However, making valid causal inferences from observational data is challenging because of the risk of several statistical biases. In 2016 Hernán and Robins put forward the 'target trial framework' as a guide to best design and analyse observational studies whilst preventing the most common biases. This framework consists of (1) clearly defining a causal question about an intervention, (2) specifying the protocol of the hypothetical trial, and (3) explaining how the observational data will be used to emulate it. METHODS The aim of this scoping review was to identify and review all explicit attempts of trial emulation studies across all medical fields. Embase, Medline and Web of Science were searched for trial emulation studies published in English from database inception to February 25, 2021. The following information was extracted from studies that were deemed eligible for review: the subject area, the type of observational data that they leveraged, and the statistical methods they used to address the following biases: (A) confounding bias, (B) immortal time bias, and (C) selection bias. RESULTS The search resulted in 617 studies, 38 of which we deemed eligible for review. Of those 38 studies, most focused on cardiology, infectious diseases or oncology and the majority used electronic health records/electronic medical records data and cohort studies data. Different statistical methods were used to address confounding at baseline and selection bias, predominantly conditioning on the confounders (N = 18/49, 37%) and inverse probability of censoring weighting (N = 7/20, 35%) respectively. Different approaches were used to address immortal time bias, assigning individuals to treatment strategies at start of follow-up based on their data available at that specific time (N = 21, 55%), using the sequential trial emulations approach (N = 11, 29%) or the cloning approach (N = 6, 16%). CONCLUSION Different methods can be leveraged to address (A) confounding bias, (B) immortal time bias, and (C) selection bias. When working with observational data, and if possible, the 'target trial' framework should be used as it provides a structured conceptual approach to observational research.
Collapse
Affiliation(s)
- Giulio Scola
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | - Anca Chis Ster
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Daniel Bean
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Health Data Research UK London, Institute of Health Informatics, University College London, London, UK
| | - Nilesh Pareek
- King's College Hospital NHS Foundation Trust, London, UK
- School of Cardiovascular and Metabolic Medicine & Sciences, BHF Centre of Excellence, King's College London, London, UK
| | - Richard Emsley
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Sabine Landau
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
82
|
Moler-Zapata S, Hutchings A, O'Neill S, Silverwood RJ, Grieve R. Emulating Target Trials With Real-World Data to Inform Health Technology Assessment: Findings and Lessons From an Application to Emergency Surgery. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2023; 26:1164-1174. [PMID: 37164043 DOI: 10.1016/j.jval.2023.04.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 04/05/2023] [Accepted: 04/26/2023] [Indexed: 05/12/2023]
Abstract
OBJECTIVES International health technology assessment (HTA) agencies recommend that real-world data (RWD) are used in some circumstances to add to the evidence base about the effectiveness and cost-effectiveness of health interventions. The target trial framework applies the design principles of randomized-controlled trials to RWD and can help alleviate inevitable concerns about bias and design flaws with nonrandomized studies. This article aimed to tackle the lack of guidance and exemplar applications on how this methodology can be applied to RWD to inform HTA decision making. METHODS We use Hospital Episode Statistics data from England on emergency hospital admissions from 2010 to 2019 to evaluate the cost-effectiveness of emergency surgery for 2 acute gastrointestinal conditions. We draw on the case study to describe the main challenges in applying the target trial framework alongside RWD and provide recommendations for how these can be addressed in practice. RESULTS The 4 main challenges when applying the target trial framework to RWD are (1) defining the study population, (2) defining the treatment strategies, (3) establishing time zero (baseline), and (4) adjusting for unmeasured confounding. The recommendations for how to address these challenges, mainly around the incorporation of expert judgment and use of appropriate methods for handling unmeasured confounding, are illustrated within the case study. CONCLUSIONS The recommendations outlined in this study could help future studies seeking to inform HTA decision processes. These recommendations can complement checklists for economic evaluations and design tools for estimating treatment effectiveness in nonrandomized studies.
Collapse
Affiliation(s)
- Silvia Moler-Zapata
- Department of Health Services Research and Policy, Faculty of Public Health and Policy, London School of Hygiene and Tropical Medicine, London, England, UK.
| | - Andrew Hutchings
- Department of Health Services Research and Policy, Faculty of Public Health and Policy, London School of Hygiene and Tropical Medicine, London, England, UK
| | - Stephen O'Neill
- Department of Health Services Research and Policy, Faculty of Public Health and Policy, London School of Hygiene and Tropical Medicine, London, England, UK
| | - Richard J Silverwood
- Centre for Longitudinal Studies, UCL Social Research Institute, University College London, London, England, UK
| | - Richard Grieve
- Department of Health Services Research and Policy, Faculty of Public Health and Policy, London School of Hygiene and Tropical Medicine, London, England, UK
| |
Collapse
|
83
|
Fu EL. Target Trial Emulation to Improve Causal Inference from Observational Data: What, Why, and How? J Am Soc Nephrol 2023; 34:1305-1314. [PMID: 37131279 PMCID: PMC10400102 DOI: 10.1681/asn.0000000000000152] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 04/17/2023] [Indexed: 05/04/2023] Open
Abstract
ABSTRACT Target trial emulation has drastically improved the quality of observational studies investigating the effects of interventions. Its ability to prevent avoidable biases that have plagued many observational analyses has contributed to its recent popularity. This review explains what target trial emulation is, why it should be the standard approach for causal observational studies that investigate interventions, and how to do a target trial emulation analysis. We discuss the merits of target trial emulation compared with often used, but biased analyses, as well as potential caveats, and provide clinicians and researchers with the tools to better interpret results from observational studies investigating the effects of interventions.
Collapse
Affiliation(s)
- Edouard L Fu
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
84
|
Bigirumurame T, Hiu SKW, Teare MD, Wason JMS, Bryant A, Breckons M. Current practices in studies applying the target trial emulation framework: a protocol for a systematic review. BMJ Open 2023; 13:e070963. [PMID: 37369393 PMCID: PMC10410979 DOI: 10.1136/bmjopen-2022-070963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
INTRODUCTION Observational studies represent an alternative to estimate real-world causal effects in the absence of available randomised controlled trials (RCTs). Target trial emulation is a framework for the application of RCT design principles to emulate a hypothetical open-label RCT (the hypothetical target trial) using existing observational data as the primary data source as opposed to the prospective recruitment and measurement of randomised units. The aim of this systematic review is to investigate the practices of studies applying the target trial emulation framework to evaluate the effectiveness of interventions. METHODS AND ANALYSIS We will systematically search in Medline (via Ovid), Embase (via Ovid, entries from medRxiv are included), PsycINFO (via Ovid), SCOPUS, Web of Science, Cochrane Library, the ISRCTN registry and ClinicalTrials.gov for all study reports and protocols which used the trial emulation framework (without time restriction). We will extract information concerning study design, data source, analysis, results, interpretation and dissemination. Two reviewers will perform study selection, data extraction and quality assessment. Disagreements between reviewers will be resolved by a third reviewer. A narrative approach will be used to synthesise and report qualitative and quantitative data. Reporting of the review will be informed by Preferred Reporting Items for Systematic Review and Meta-Analysis guidance (PRISMA). ETHICS AND DISSEMINATION Ethical approval is not required as it is a protocol for a systematic review. Findings will be disseminated through peer-reviewed publications and conference presentations.
Collapse
Affiliation(s)
| | - Shaun Kuan Wei Hiu
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - M Dawn Teare
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - James M S Wason
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Andrew Bryant
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Matthew Breckons
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
85
|
Vell MS, Loomba R, Krishnan A, Wangensteen KJ, Trebicka J, Creasy KT, Trautwein C, Scorletti E, Seeling KS, Hehl L, Rendel MD, Zandvakili I, Li T, Chen J, Vujkovic M, Alqahtani S, Rader DJ, Schneider KM, Schneider CV. Association of Statin Use With Risk of Liver Disease, Hepatocellular Carcinoma, and Liver-Related Mortality. JAMA Netw Open 2023; 6:e2320222. [PMID: 37358849 PMCID: PMC10293910 DOI: 10.1001/jamanetworkopen.2023.20222] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/11/2023] [Indexed: 06/27/2023] Open
Abstract
IMPORTANCE Given the burden of chronic liver disease on the health care system, more information on the hepatoprotective association of statins in the general population is needed. OBJECTIVE To examine whether regular statin use is associated with a reduction in liver disease, particularly hepatocellular carcinoma (HCC) and liver-related deaths, in the general population. DESIGN, SETTING, AND PARTICIPANTS This cohort study used data from the UK Biobank (UKB) (individuals aged 37-73 years) collected from baseline (2006-2010) to the end of follow-up in May 2021, from the TriNetX cohort (individuals aged 18-90 years) enrolled from baseline (2011-2020) until end of follow-up in September 2022, and from the Penn Medicine Biobank (PMBB) (individuals aged 18-102 years) with ongoing enrollment starting in 2013 to the end of follow-up in December 2020. Individuals were matched using propensity score matching according to the following criteria: age, sex, body mass index, ethnicity, diabetes with or without insulin or biguanide use, hypertension, ischemic heart disease, dyslipidemia, aspirin use, and number of medications taken (UKB only). Data analysis was performed from April 2021 to April 2023. EXPOSURE Regular statin use. MAIN OUTCOMES AND MEASURES Primary outcomes were liver disease and HCC development as well as liver-associated death. RESULTS A total of 1 785 491 individuals were evaluated after matching (aged 55 to 61 years on average, up to 56% men, and up to 49% women). A total of 581 cases of liver-associated death, 472 cases of incident HCC, and 98 497 new liver diseases were registered during the follow-up period. Individuals were aged 55-61 years on average, with a slightly higher proportion of men (up to 56%). In UKB individuals (n = 205 057) without previously diagnosed liver disease, statin users (n = 56 109) had a 15% lower hazard ratio (HR) for the association of developing a new liver disease (HR, 0.85; 95% CI, 0.78-0.92; P < .001). In addition, statin users demonstrated a 28% lower HR for the association with liver-related death (HR, 0.72; 95% CI, 0.59-0.88; P = .001) and a 42% lower HR for the development of HCC (HR, 0.58; 95% CI, 0.35-0.96; P = .04). In TriNetX individuals (n = 1 568 794), the HR for the association of HCC was reduced even further for statin users (HR, 0.26; 95% CI, 0.22-0.31; P = .003). The hepatoprotective association of statins was time and dose dependent, with a significant association in PMBB individuals (n = 11 640) for incident liver diseases after 1 year of statin use (HR, 0.76; 95% CI, 0.59-0.98; P = .03). Taking statins was particularly beneficial in men, individuals with diabetes, and individuals with a high Fibrosis-4 index at baseline. Carriers of the heterozygous minor allele of PNPLA3 rs738409 benefited from statin use and had a 69% lower HR for the association with HCC (UKB HR, 0.31; 95% CI, 0.11-0.85; P = .02). CONCLUSIONS AND RELEVANCE This cohort study indicates substantial preventive associations of statins against liver disease, with an association with duration and dose of intake.
Collapse
Affiliation(s)
- Mara Sophie Vell
- Gastroenterology, Metabolic Diseases, and Intensive Care, Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Rohit Loomba
- Division of Gastroenterology, University of California, San Diego, La Jolla
| | - Arunkumar Krishnan
- Section of Gastroenterology and Hepatology, West Virginia University School of Medicine, Morgantown
| | - Kirk J. Wangensteen
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Jonel Trebicka
- Medical Clinic B, Gastroenterology, Hepatology, Endocrinology, Clinical Infectiology, University Hospital Münster, Münster, Germany
| | - Kate Townsend Creasy
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia
| | - Christian Trautwein
- Gastroenterology, Metabolic Diseases, and Intensive Care, Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Eleonora Scorletti
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Katharina Sophie Seeling
- Gastroenterology, Metabolic Diseases, and Intensive Care, Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Leonida Hehl
- Gastroenterology, Metabolic Diseases, and Intensive Care, Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Miriam Daphne Rendel
- Gastroenterology, Metabolic Diseases, and Intensive Care, Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Inuk Zandvakili
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Tang Li
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Jinbo Chen
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Marijana Vujkovic
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Saleh Alqahtani
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Liver Transplant Center, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Daniel James Rader
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Kai Markus Schneider
- Gastroenterology, Metabolic Diseases, and Intensive Care, Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Carolin Victoria Schneider
- Gastroenterology, Metabolic Diseases, and Intensive Care, Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| |
Collapse
|
86
|
Bjornevik K, Ascherio A. The intricate connection between diabetes mellitus and Parkinson's disease. Eur J Epidemiol 2023; 38:587-589. [PMID: 37145289 DOI: 10.1007/s10654-023-01013-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 04/18/2023] [Indexed: 05/06/2023]
Affiliation(s)
- Kjetil Bjornevik
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Departments of Epidemiology and Nutrition, Harvard T.H. Chan School of Public Health, 665 Huntington Ave, Boston, MA, 02115, USA.
| | - Alberto Ascherio
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
87
|
Bhatia A, Preiss AJ, Xiao X, Brannock MD, Alexander GC, Chew RF, Fitzgerald M, Hill E, Kelly EP, Mehta HB, Madlock-Brown C, Wilkins KJ, Chute CG, Haendel M, Moffitt R, Pfaff ER. Effect of Nirmatrelvir/Ritonavir (Paxlovid) on Hospitalization among Adults with COVID-19: an EHR-based Target Trial Emulation from N3C. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.05.03.23289084. [PMID: 37205340 PMCID: PMC10187454 DOI: 10.1101/2023.05.03.23289084] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
This study leverages electronic health record data in the National COVID Cohort Collaborative's (N3C) repository to investigate disparities in Paxlovid treatment and to emulate a target trial assessing its effectiveness in reducing COVID-19 hospitalization rates. From an eligible population of 632,822 COVID-19 patients seen at 33 clinical sites across the United States between December 23, 2021 and December 31, 2022, patients were matched across observed treatment groups, yielding an analytical sample of 410,642 patients. We estimate a 65% reduced odds of hospitalization among Paxlovid-treated patients within a 28-day follow-up period, and this effect did not vary by patient vaccination status. Notably, we observe disparities in Paxlovid treatment, with lower rates among Black and Hispanic or Latino patients, and within socially vulnerable communities. Ours is the largest study of Paxlovid's real-world effectiveness to date, and our primary findings are consistent with previous randomized control trials and real-world studies.
Collapse
Affiliation(s)
- Abhishek Bhatia
- University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Xuya Xiao
- School of Medicine, Public Health, and Nursing, Johns Hopkins University, Baltimore, MD, USA
| | | | - G Caleb Alexander
- School of Medicine, Public Health, and Nursing, Johns Hopkins University, Baltimore, MD, USA
| | | | | | - Elaine Hill
- University of Rochester, Department of Public Health Sciences and Department of Economics, Rochester, NY, USA
| | | | - Hemalkumar B Mehta
- School of Medicine, Public Health, and Nursing, Johns Hopkins University, Baltimore, MD, USA
| | | | - Kenneth J Wilkins
- National Institute of Diabetes & Digestive & Kidney Diseases, Office of the Director, National Institutes of Health, Bethesda, MD, USA
- F. Edward Hébert School of Medicine, Department of Preventive Medicine & Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Christopher G Chute
- School of Medicine, Public Health, and Nursing, Johns Hopkins University, Baltimore, MD, USA
| | - Melissa Haendel
- University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Emily R Pfaff
- University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
88
|
Caniglia EC, Zash R, Fennell C, Diseko M, Mayondi G, Heintz J, Mmalane M, Makhema J, Lockman S, Mumford SL, Murray EJ, Hernández-Díaz S, Shapiro R. Emulating Target Trials to Avoid Immortal Time Bias - An Application to Antibiotic Initiation and Preterm Delivery. Epidemiology 2023; 34:430-438. [PMID: 36805380 PMCID: PMC10263190 DOI: 10.1097/ede.0000000000001601] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
BACKGROUND Randomized trials in pregnancy are extremely challenging, and observational studies are often the only option to evaluate medication safety during pregnancy. However, such studies are often susceptible to immortal time bias if treatment initiation occurs after time zero of follow-up. We describe how emulating a sequence of target trials avoids immortal time bias and apply the approach to estimate the safety of antibiotic initiation between 24 and 37 weeks gestation on preterm delivery. METHODS The Tsepamo Study captured birth outcomes at hospitals throughout Botswana from 2014 to 2021. We emulated 13 sequential target trials of antibiotic initiation versus no initiation among individuals presenting to care <24 weeks, one for each week from 24 to 37 weeks. For each trial, eligible individuals had not previously initiated antibiotics. We also conducted an analysis susceptible to immortal time bias by defining time zero as 24 weeks and exposure as antibiotic initiation between 24 and 37 weeks. We calculated adjusted risk ratios (RR) and 95% confidence intervals (CI) for preterm delivery. RESULTS Of 111,403 eligible individuals, 17,009 (15.3%) initiated antibiotics between 24 and 37 weeks. In the sequence of target trials, RRs (95% CIs) ranged from 1.04 (0.90, 1.19) to 1.24 (1.11, 1.39) (pooled RR: 1.11 [1.06, 1.15]). In the analysis susceptible to immortal time bias, the RR was 0.90 (0.86, 0.94). CONCLUSIONS Defining exposure as antibiotic initiation at any time during follow-up after time zero resulted in substantial immortal time bias, making antibiotics appear protective against preterm delivery. Conducting a sequence of target trials can avoid immortal time bias in pregnancy studies.
Collapse
Affiliation(s)
- Ellen C. Caniglia
- University of Pennsylvania Perelman School of Medicine
- Botswana-Harvard AIDS Institute Partnership
| | - Rebecca Zash
- Botswana-Harvard AIDS Institute Partnership
- Beth Israel Deaconess Medical Centerss
| | | | | | | | | | | | | | - Shahin Lockman
- Botswana-Harvard AIDS Institute Partnership
- Harvard T.H. Chan School of Public Health
- Brigham and Women’s Hospital
| | | | | | | | - Roger Shapiro
- Botswana-Harvard AIDS Institute Partnership
- Harvard T.H. Chan School of Public Health
| |
Collapse
|
89
|
Gottschau M, Rosthøj S, Settnes A, Aalborg GL, Viuff JH, Munk C, Jensen A, Kjær SK, Mellemkjær L. Long-Term Health Consequences After Ovarian Removal at Benign Hysterectomy : A Nationwide Cohort Study. Ann Intern Med 2023; 176:596-604. [PMID: 37068275 DOI: 10.7326/m22-1628] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/19/2023] Open
Abstract
BACKGROUND More evidence is needed to substantiate current recommendations about removing ovaries during hysterectomy for benign conditions. OBJECTIVE To compare long-term outcomes in women with and without bilateral salpingo-oophorectomy (BSO) during hysterectomy for benign conditions. DESIGN Emulated target trial using data from a population-based cohort. SETTING Women in Denmark aged 20 years or older during 1977 to 2017. PARTICIPANTS 142 985 women with hysterectomy for a benign condition, 22 974 with BSO and 120 011 without. INTERVENTION Benign hysterectomy with or without BSO. MEASUREMENTS The primary outcomes were overall hospitalization for cardiovascular disease (CVD), overall cancer incidence, and all-cause mortality through December 2018. RESULTS Compared with women without BSO, women with BSO who were younger than 45 years at surgery had a higher 10-year cumulative risk for hospitalization for CVD (risk difference [RD], 1.19 percentage points [95% CI, 0.09 to 2.43 percentage points]). Women with BSO had a higher 10-year cumulative risk for cancer for ages 45 to 54 years (RD, 0.73 percentage point [CI, 0.05 to 1.38 percentage points]), 55 to 64 years (RD, 1.92 percentage points [CI, 0.69 to 3.25 percentage points]), and 65 years or older (RD, 2.54 percentage points [CI, 0.91 to 4.25 percentage points]). Women with BSO had higher 10-year mortality in all age groups, although the differences were statistically significant only for ages 45 to 54 years (RD, 0.79 percentage point [CI, 0.27 to 1.30 percentage points]). The mortality at 20 years was inconsistent with that at 10 years in women aged 65 years or older. LIMITATION Age was a proxy for menopausal status. CONCLUSION The authors find that these results support current recommendations for conserving ovaries in premenopausal women without a high risk for ovarian cancer and suggest a cautious approach in postmenopausal women. PRIMARY FUNDING SOURCE The Danish Cancer Society's Scientific Committee and the Mermaid Project.
Collapse
Affiliation(s)
- Mathilde Gottschau
- Diet, Cancer and Health, Danish Cancer Society Research Center, Copenhagen, Denmark (M.G., J.H.V., L.M.)
| | - Susanne Rosthøj
- Statistics and Data Analysis, Danish Cancer Society Research Center, Copenhagen, Denmark (S.R., G.L.A.)
| | - Annette Settnes
- Department of Obstetrics and Gynecology, North Zealand Hospital, University of Copenhagen, Hillerød, Denmark (A.S.)
| | - Gitte Lerche Aalborg
- Statistics and Data Analysis, Danish Cancer Society Research Center, Copenhagen, Denmark (S.R., G.L.A.)
| | - Jakob Hansen Viuff
- Diet, Cancer and Health, Danish Cancer Society Research Center, Copenhagen, Denmark (M.G., J.H.V., L.M.)
| | - Christian Munk
- Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark (C.M.)
| | - Allan Jensen
- Lifestyle, Reproduction and Cancer, Danish Cancer Society Research Center, Copenhagen, Denmark (A.J.)
| | - Susanne K Kjær
- Virus, Lifestyle and Genes, Danish Cancer Society Research Center, and Department of Gynecology, Rigshospitalet University Hospital, University of Copenhagen, Copenhagen, Denmark (S.K.K.)
| | - Lene Mellemkjær
- Diet, Cancer and Health, Danish Cancer Society Research Center, Copenhagen, Denmark (M.G., J.H.V., L.M.)
| |
Collapse
|
90
|
Zhang N, Wang C, Li Y, Chen F, Yan P. Hypothetical interventions on risk factors for depression among middle-aged and older community-dwellers in China: An application of the parametric g-formula in a longitudinal study. J Affect Disord 2023; 327:355-361. [PMID: 36754095 DOI: 10.1016/j.jad.2023.01.113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 01/22/2023] [Accepted: 01/30/2023] [Indexed: 02/09/2023]
Abstract
BACKGROUND Sleep disturbances, frailty, and body pain are widespread in middle-aged and older adults with depression, and have been identified as depression risk factors. However, there is a scarcity of research on the benefits of sleep improvement, frailty amelioration, and pain management on incident depression. METHODS A total of 8895 respondents aged above 45 years were derived from the China Health and Retirement Longitudinal Study (CHARLS) between 2011 and 2018. The parametric g-formula was used to estimate the 7-year risks of depression under independent hypothetical interventions on nighttime sleep duration (NSD), daytime napping duration (DND), perceived sleep quality (PSQ), frailty, and pain, as well as their various combinations. RESULTS The observed depression risk was 41.77 %. The independent intervention on frailty was the most effective in lowering incident depression, with a risk ratio (RR) of 0.61 (95 % CI: 0.57-0.64), followed by PSQ (RR: 0.75, 95 % CI: 0.73-0.78), pain (RR: 0.90, 95 % CI: 0.87-0.91), and NSD (RR: 0.96, 95 % CI: 0.93-0.98). In subgroup analysis, intervention on NSD was more effective in men, PSQ was more effective in middle-aged individuals, and frailty and pain were more effective in older persons. The combined intervention of NSD, PSQ, frailty, and pain lowered the risk the greatest (RR: 0.35, 95 % CI: 0.32-0.37). LIMITATIONS Generalizing our results to other populations should be possible if they have the same distribution of effect modifiers and interference patterns because of the calculation principle of the parametric g-formula. CONCLUSIONS Interventions for sleep disturbances, frailty, and body pain can minimize the risk of depression.
Collapse
Affiliation(s)
- Nan Zhang
- School of Nursing, Xinjiang Medical University, Urumqi, China
| | - Cui Wang
- School of Nursing, Peking University, Beijing, China
| | - Yuli Li
- School of Nursing and Rehabilitation, Shandong University, Jinan, China
| | - Fenghui Chen
- School of Nursing, Xinjiang Medical University, Urumqi, China
| | - Ping Yan
- School of Nursing, Xinjiang Medical University, Urumqi, China.
| |
Collapse
|
91
|
Xie Y, Bowe B, Al-Aly Z. Nirmatrelvir and risk of hospital admission or death in adults with covid-19: emulation of a randomized target trial using electronic health records. BMJ 2023; 381:e073312. [PMID: 37041016 PMCID: PMC10086514 DOI: 10.1136/bmj-2022-073312] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
OBJECTIVE To estimate the effectiveness of nirmatrelvir, compared with no treatment, in reducing admission to hospital or death at 30 days in people infected with the SARS-CoV-2 virus and at risk of developing severe disease, according to vaccination status and history of previous SARS-CoV-2 infection. DESIGN Emulation of a randomized target trial with electronic health records. SETTING Healthcare databases of the US Department of Veterans Affairs PARTICIPANTS: 256 288 participants with a SARS-CoV-2 positive test result and at least one risk factor for developing severe covid-19 disease, between 3 January and 30 November 2022. 31 524 were treated with nirmatrelvir within five days of testing positive for SARS-CoV-2 and 224 764 received no treatment. MAIN OUTCOME MEASURES The effectiveness of starting nirmatrelvir within five days of a positive SARS-CoV-2 test result versus no treatment in reducing the risk of admission to hospital or death at 30 days was estimated in those who were not vaccinated, in those who received one or two doses of vaccine, and those who received a vaccine booster and, separately, in participants with a primary SARS-CoV-2 infection or reinfection. The inverse probability weighting method was used to balance personal and health characteristics between the groups. Relative risk and absolute risk reduction were computed from cumulative incidence at 30 days, estimated by weighted Kaplan-Meier estimator. RESULTS Among people who were not vaccinated (n=76 763; 5338 nirmatrelvir and 71 425 no treatment), compared with no treatment, the relative risk of nirmatrelvir in reducing admission to hospital or death at 30 days was 0.60 (95% confidence interval 0.50 to 0.71); the absolute risk reduction was 1.83% (95% confidence interval 1.29% to 2.49%). The relative risk and absolute risk reduction, compared with no treatment, were 0.65 (0.57 to 0.74) and 1.27% (0.90% to 1.61%), respectively, in people who received one or two doses of vaccine (n=84 620; 7989 nirmatrelvir and 76 631 no treatment); 0.64 (0.58 to 0.71) and 1.05% (0.85% to 1.27%) in individuals who received a booster dose of vaccine (n=94 905; 18 197 nirmatrelvir and 76 708 no treatment); 0.61 (0.57 to 0.65) and 1.36% (1.19% to 1.53%) in participants with a primary SARS-CoV-2 infection (n=228 081; 26 350 nirmatrelvir and 201 731 no treatment); and 0.74 (0.63 to 0.87) and 0.79% (0.36% to 1.18%) in participants who were reinfected with the SARS-CoV-2 virus (n=28 207; 5174 nirmatrelvir and 23 033 no treatment). Nirmatrelvir was associated with a reduced risk of admission to hospital or death in those aged ≤65 years and > 65 years; in men and women; in black and white participants; in those with 1-2, 3-4, and ≥5 risk factors for progression to severe covid-19 illness; and in those infected during the omicron BA.1 or BA.2 predominant era, and the BA.5 predominant era. CONCLUSIONS In people with SARS-CoV-2 infection who were at risk of developing severe disease, compared with no treatment, nirmatrelvir was associated with a reduced risk of admission to hospital or death at 30 days in people who were not vaccinated, vaccinated, and had received a booster vaccine, and in those with a primary SARS-CoV-2 infection and reinfection.
Collapse
Affiliation(s)
- Yan Xie
- Clinical Epidemiology Center, Research and Development Service, VA Saint Louis Health Care System, Saint Louis, Missouri, USA
- Veterans Research and Education Foundation of Saint Louis, Saint Louis, Missouri, USA
- Department of Epidemiology and Biostatistics, College for Public Health and Social Justice, Saint Louis University, Saint Louis, Missouri, USA
| | - Benjamin Bowe
- Clinical Epidemiology Center, Research and Development Service, VA Saint Louis Health Care System, Saint Louis, Missouri, USA
- Veterans Research and Education Foundation of Saint Louis, Saint Louis, Missouri, USA
| | - Ziyad Al-Aly
- Clinical Epidemiology Center, Research and Development Service, VA Saint Louis Health Care System, Saint Louis, Missouri, USA
- Veterans Research and Education Foundation of Saint Louis, Saint Louis, Missouri, USA
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
- Nephrology Section, Medicine Service, VA Saint Louis Health Care System, Saint Louis, Missouri, USA
- Institute for Public Health, Washington University in Saint Louis, Saint Louis, Missouri, USA
| |
Collapse
|
92
|
Zeleznik OA, Irvin SR, Samimi G, Trabert B. The Role of Statins in the Prevention of Ovarian and Endometrial Cancers. Cancer Prev Res (Phila) 2023; 16:191-197. [PMID: 37009709 PMCID: PMC10405632 DOI: 10.1158/1940-6207.capr-22-0374] [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: 09/22/2022] [Revised: 12/05/2022] [Accepted: 02/13/2023] [Indexed: 04/04/2023]
Abstract
Ovarian and endometrial cancers are the most common gynecologic malignancies and emerging evidence suggests that lipid metabolism and subsequent inflammation are important etiologic factors for both tumors. Statins (HMG-CoA reductase inhibitors) are the most widely prescribed lipid-lowering drugs in the United States and are used by 25% of adults aged 40+ years. In addition to their cardio-protective actions, statins have anti-inflammatory effects and have demonstrated antiproliferative and apoptotic properties in cancer cell lines, supporting a potential role in cancer prevention. To appropriately quantify potential public health impact of statin use for cancer prevention, there is a great need to understand the potential risk reduction among individuals at a higher risk of gynecologic cancers, the group that will likely need to be targeted to effectively balance risk/benefit of medications repurposed for cancer prevention. In this commentary, we focus on summarizing emerging evidence suggesting that the anti-inflammatory and lipid-lowering mechanisms of statins may provide important cancer-preventive benefits for gynecologic cancers as well as outline important unanswered questions and future research directions.
Collapse
Affiliation(s)
- Oana A Zeleznik
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Sarah R Irvin
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD
| | - Goli Samimi
- Division of Cancer Prevention, National Cancer Institute, Rockville, MD
| | - Britton Trabert
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD
- Department of Obstetrics and Gynecology, University of Utah, Huntsman Cancer Institute at the University of Utah, Salt Lake City, UT
| |
Collapse
|
93
|
Xie Y, Bowe B, Al-Aly Z. Molnupiravir and risk of hospital admission or death in adults with covid-19: emulation of a randomized target trial using electronic health records. BMJ 2023; 380:e072705. [PMID: 36882199 PMCID: PMC9989554 DOI: 10.1136/bmj-2022-072705] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/08/2023] [Indexed: 03/09/2023]
Abstract
OBJECTIVE To emulate a randomized target trial to estimate the association between the antiviral drug molnupiravir and hospital admission or death in adults with SARS-CoV-2 infection in the community during the omicron predominant era who were at high risk of progression to severe covid-19. DESIGN Emulation of a randomized target trial using electronic health records. SETTING US Department of Veterans Affairs. PARTICIPANTS 85 998 adults with SARS-CoV-2 infection between 5 January and 30 September 2022 and at least one risk factor for progression to severe covid-19: 7818 participants were eligible for and treated with molnupiravir and 78 180 received no treatment. MAIN OUTCOMES MEASURE The primary outcome was a composite of hospital admission or death at 30 days. The clone method with inverse probability of censoring weighting was used to adjust for informative censoring and balance baseline characteristics between the groups. The cumulative incidence function was used to estimate the relative risk and the absolute risk reduction at 30 days. RESULTS Molnupiravir was associated with a reduction in hospital admissions or death at 30 days (relative risk 0.72 (95% confidence interval 0.64 to 0.79)) compared with no treatment; the event rates for hospital admission or death at 30 days were 2.7% (95% confidence interval 2.5% to 3.0%) for molnupiravir and 3.8% (3.7% to 3.9%) for no treatment; the absolute risk reduction was 1.1% (95% confidence interval 0.8% to 1.4%). Molnupiravir appeared to be effective in those who had not been vaccinated against covid-19 (relative risk 0.83 (0.70 to 0.97) and absolute risk reduction 0.9% (0.2% to 1.9%)), had received one or two vaccine doses (0.69 (0.56 to 0.83) and 1.3% (0.7% to 1.9%)), and had received a booster dose (0.71 (0.58 to 0.83) and 1.0% (0.5% to 1.4%)); in those infected during the era when the omicron subvariant BA.1 or BA.2 was predominant (0.72 (0.62 to 0.83) and 1.2% (0.7% to 1.6%)) and when BA.5 was predominant (0.75 (0.66 to 0.86) and 0.9% (0.5% to 1.3%)); and in those with no history of SARS-CoV-2 infection (0.72 (0.64 to 0.81) and 1.1% (0.8% to 1.4%)) and with a history of SARS-CoV-2 infection (0.75 (0.58 to 0.97) and 1.1% (0.1% to 1.8%)). CONCLUSIONS The findings of this emulation of a randomized target trial suggest that molnupiravir might have reduced hospital admission or death at 30 days in adults with SARS-CoV-2 infection in the community during the recent omicron predominant era who were at high risk of progression to severe covid-19 and eligible for treatment with molnupiravir.
Collapse
Affiliation(s)
- Yan Xie
- Clinical Epidemiology Center, Research and Development Service, VA Saint Louis Health Care System, Saint Louis, Missouri, USA
- Veterans Research and Education Foundation of Saint Louis, Saint Louis, Missouri, USA
- Department of Epidemiology and Biostatistics, College for Public Health and Social Justice, Saint Louis University, Saint Louis, Missouri, USA
| | - Benjamin Bowe
- Clinical Epidemiology Center, Research and Development Service, VA Saint Louis Health Care System, Saint Louis, Missouri, USA
- Veterans Research and Education Foundation of Saint Louis, Saint Louis, Missouri, USA
| | - Ziyad Al-Aly
- Clinical Epidemiology Center, Research and Development Service, VA Saint Louis Health Care System, Saint Louis, Missouri, USA
- Veterans Research and Education Foundation of Saint Louis, Saint Louis, Missouri, USA
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
- Nephrology Section, Medicine Service, VA Saint Louis Health Care System, Saint Louis, Missouri, USA
- Institute for Public Health, Washington University in Saint Louis, Saint Louis, Missouri, USA
| |
Collapse
|
94
|
Moctezuma-Velázquez C, Abraldes JG. How strong is the evidence to support statins repurposing for the treatment of cirrhosis? REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2023; 115:107-109. [PMID: 36353965 DOI: 10.17235/reed.2022.9281/2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
In this editorial, we briefly mention the studies that support the use of statins to change the natural history of liver cirrhosis, alongside potential biases and flaws that need to be considered when analyzing data. The key message that we want to communicate is that even if current evidence is somehow compelling, it is limited, mostly from observational studies, and in general not enough to formally recommend the prescription of statins in patients with cirrhosis as disease-modifying agents. Finally, we also mention some important facts about the safety of statins in the context of patients with underlying liver disease.
Collapse
Affiliation(s)
| | - Juan G Abraldes
- Gastroenterology (Liver Unit), University of Alberta, Canada
| |
Collapse
|
95
|
Rivera AS, Al-Heeti O, Petito LC, Feinstein MJ, Achenbach CJ, Williams J, Taiwo B. Association of statin use with outcomes of patients admitted with COVID-19: an analysis of electronic health records using superlearner. BMC Infect Dis 2023; 23:115. [PMID: 36829115 PMCID: PMC9951166 DOI: 10.1186/s12879-023-08026-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 01/23/2023] [Indexed: 02/26/2023] Open
Abstract
IMPORTANCE Statin use prior to hospitalization for Coronavirus Disease 2019 (COVID-19) is hypothesized to improve inpatient outcomes including mortality, but prior findings from large observational studies have been inconsistent, due in part to confounding. Recent advances in statistics, including incorporation of machine learning techniques into augmented inverse probability weighting with targeted maximum likelihood estimation, address baseline covariate imbalance while maximizing statistical efficiency. OBJECTIVE To estimate the association of antecedent statin use with progression to severe inpatient outcomes among patients admitted for COVD-19. DESIGN, SETTING AND PARTICIPANTS We retrospectively analyzed electronic health records (EHR) from individuals ≥ 40-years-old who were admitted between March 2020 and September 2022 for ≥ 24 h and tested positive for SARS-CoV-2 infection in the 30 days before to 7 days after admission. EXPOSURE Antecedent statin use-statin prescription ≥ 30 days prior to COVID-19 admission. MAIN OUTCOME Composite end point of in-hospital death, intubation, and intensive care unit (ICU) admission. RESULTS Of 15,524 eligible COVID-19 patients, 4412 (20%) were antecedent statin users. Compared with non-users, statin users were older (72.9 (SD: 12.6) versus 65.6 (SD: 14.5) years) and more likely to be male (54% vs. 51%), White (76% vs. 71%), and have ≥ 1 medical comorbidity (99% vs. 86%). Unadjusted analysis demonstrated that a lower proportion of antecedent users experienced the composite outcome (14.8% vs 19.3%), ICU admission (13.9% vs 18.3%), intubation (5.1% vs 8.3%) and inpatient deaths (4.4% vs 5.2%) compared with non-users. Risk differences adjusted for labs and demographics were estimated using augmented inverse probability weighting with targeted maximum likelihood estimation using Super Learner. Statin users still had lower rates of the composite outcome (adjusted risk difference: - 3.4%; 95% CI: - 4.6% to - 2.1%), ICU admissions (- 3.3%; - 4.5% to - 2.1%), and intubation (- 1.9%; - 2.8% to - 1.0%) but comparable inpatient deaths (0.6%; - 1.3% to 0.1%). CONCLUSIONS AND RELEVANCE After controlling for confounding using doubly robust methods, antecedent statin use was associated with minimally lower risk of severe COVID-19-related outcomes, ICU admission and intubation, however, we were not able to corroborate a statin-associated mortality benefit.
Collapse
Affiliation(s)
- Adovich S Rivera
- Institute for Public Health and Management, Feinberg School of Medicine, Chicago, IL, 60611, USA
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA, 91101, USA
| | - Omar Al-Heeti
- Division of Infectious Diseases, Department of Medicine, Northwestern University Feinberg School of Medicine, 645 N. Michigan Ave, Suite 900, Chicago, IL, 60611, USA
| | - Lucia C Petito
- Division of Biostatistics, Department of Preventive Medicine, Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Mathew J Feinstein
- Division of Cardiology, Department of Medicine, Feinberg School of Medicine, Chicago, IL, 60611, USA
- Division of Epidemiology, Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Chad J Achenbach
- Division of Infectious Diseases, Department of Medicine, Northwestern University Feinberg School of Medicine, 645 N. Michigan Ave, Suite 900, Chicago, IL, 60611, USA
- Havey Institute for Global Health, Northwestern University Feinberg School of Medicine, Chicago, IL, 606011, USA
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Janna Williams
- Division of Infectious Diseases, Department of Medicine, Northwestern University Feinberg School of Medicine, 645 N. Michigan Ave, Suite 900, Chicago, IL, 60611, USA
| | - Babafemi Taiwo
- Division of Infectious Diseases, Department of Medicine, Northwestern University Feinberg School of Medicine, 645 N. Michigan Ave, Suite 900, Chicago, IL, 60611, USA.
- Havey Institute for Global Health, Northwestern University Feinberg School of Medicine, Chicago, IL, 606011, USA.
| |
Collapse
|
96
|
Yarnell CJ, Angriman F, Ferreyro BL, Liu K, De Grooth HJ, Burry L, Munshi L, Mehta S, Celi L, Elbers P, Thoral P, Brochard L, Wunsch H, Fowler RA, Sung L, Tomlinson G. Oxygenation thresholds for invasive ventilation in hypoxemic respiratory failure: a target trial emulation in two cohorts. Crit Care 2023; 27:67. [PMID: 36814287 PMCID: PMC9944781 DOI: 10.1186/s13054-023-04307-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 01/06/2023] [Indexed: 02/24/2023] Open
Abstract
BACKGROUND The optimal thresholds for the initiation of invasive ventilation in patients with hypoxemic respiratory failure are unknown. Using the saturation-to-inspired oxygen ratio (SF), we compared lower versus higher hypoxemia severity thresholds for initiating invasive ventilation. METHODS This target trial emulation included patients from the Medical Information Mart for Intensive Care (MIMIC-IV, 2008-2019) and the Amsterdam University Medical Centers (AmsterdamUMCdb, 2003-2016) databases admitted to intensive care and receiving inspired oxygen fraction ≥ 0.4 via non-rebreather mask, noninvasive ventilation, or high-flow nasal cannula. We compared the effect of using invasive ventilation initiation thresholds of SF < 110, < 98, and < 88 on 28-day mortality. MIMIC-IV was used for the primary analysis and AmsterdamUMCdb for the secondary analysis. We obtained posterior means and 95% credible intervals (CrI) with nonparametric Bayesian G-computation. RESULTS We studied 3,357 patients in the primary analysis. For invasive ventilation initiation thresholds SF < 110, SF < 98, and SF < 88, the predicted 28-day probabilities of invasive ventilation were 72%, 47%, and 19%. Predicted 28-day mortality was lowest with threshold SF < 110 (22.2%, CrI 19.2 to 25.0), compared to SF < 98 (absolute risk increase 1.6%, CrI 0.6 to 2.6) or SF < 88 (absolute risk increase 3.5%, CrI 1.4 to 5.4). In the secondary analysis (1,279 patients), the predicted 28-day probability of invasive ventilation was 50% for initiation threshold SF < 110, 28% for SF < 98, and 19% for SF < 88. In contrast with the primary analysis, predicted mortality was highest with threshold SF < 110 (14.6%, CrI 7.7 to 22.3), compared to SF < 98 (absolute risk decrease 0.5%, CrI 0.0 to 0.9) or SF < 88 (absolute risk decrease 1.9%, CrI 0.9 to 2.8). CONCLUSION Initiating invasive ventilation at lower hypoxemia severity will increase the rate of invasive ventilation, but this can either increase or decrease the expected mortality, with the direction of effect likely depending on baseline mortality risk and clinical context.
Collapse
Affiliation(s)
- Christopher J. Yarnell
- grid.17063.330000 0001 2157 2938Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Canada ,grid.231844.80000 0004 0474 0428Department of Medicine, Division of Respirology, University Health Network and Sinai Health System, Toronto, Canada ,grid.17063.330000 0001 2157 2938Institute of Health Policy, Management and Evaluation, University of Toronto, Medical-Surgical ICU, 10th floor, 585 University Avenue, Toronto, ON M5G 1X5 Canada
| | - Federico Angriman
- grid.17063.330000 0001 2157 2938Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Canada ,grid.17063.330000 0001 2157 2938Institute of Health Policy, Management and Evaluation, University of Toronto, Medical-Surgical ICU, 10th floor, 585 University Avenue, Toronto, ON M5G 1X5 Canada ,grid.413104.30000 0000 9743 1587Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Bruno L. Ferreyro
- grid.17063.330000 0001 2157 2938Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Canada ,grid.231844.80000 0004 0474 0428Department of Medicine, Division of Respirology, University Health Network and Sinai Health System, Toronto, Canada ,grid.17063.330000 0001 2157 2938Institute of Health Policy, Management and Evaluation, University of Toronto, Medical-Surgical ICU, 10th floor, 585 University Avenue, Toronto, ON M5G 1X5 Canada
| | - Kuan Liu
- grid.17063.330000 0001 2157 2938Institute of Health Policy, Management and Evaluation, University of Toronto, Medical-Surgical ICU, 10th floor, 585 University Avenue, Toronto, ON M5G 1X5 Canada
| | - Harm Jan De Grooth
- grid.12380.380000 0004 1754 9227Department of Intensive Care Medicine, Laboratory for Critical Care Computational Intelligence, Amsterdam Medical Data Science, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | - Lisa Burry
- grid.17063.330000 0001 2157 2938Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Canada ,grid.492573.e0000 0004 6477 6457Department of Pharmacy and Medicine, Sinai Health System, Toronto, Canada ,grid.17063.330000 0001 2157 2938Leslie Dan Faculty of Pharmacy and Interdepartmental Division of Critical Care, University of Toronto, Toronto, ON Canada
| | - Laveena Munshi
- grid.17063.330000 0001 2157 2938Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Canada ,grid.231844.80000 0004 0474 0428Department of Medicine, Division of Respirology, University Health Network and Sinai Health System, Toronto, Canada
| | - Sangeeta Mehta
- grid.17063.330000 0001 2157 2938Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Canada ,grid.231844.80000 0004 0474 0428Department of Medicine, Division of Respirology, University Health Network and Sinai Health System, Toronto, Canada
| | - Leo Celi
- grid.116068.80000 0001 2341 2786Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02142 USA ,grid.239395.70000 0000 9011 8547Division of Pulmonary, Critical Care and Sleep Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215 USA ,grid.38142.3c000000041936754XDepartment of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115 USA
| | - Paul Elbers
- grid.12380.380000 0004 1754 9227Department of Intensive Care Medicine, Laboratory for Critical Care Computational Intelligence, Amsterdam Medical Data Science, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | - Patrick Thoral
- grid.12380.380000 0004 1754 9227Department of Intensive Care Medicine, Laboratory for Critical Care Computational Intelligence, Amsterdam Medical Data Science, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | - Laurent Brochard
- grid.415502.7Keenan Research Centre for Biomedical Research, Li Ka Shing Knowledge Institute, St Michael’s Hospital, Unity Health Toronto, Toronto, Canada ,grid.17063.330000 0001 2157 2938Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Canada
| | - Hannah Wunsch
- grid.418647.80000 0000 8849 1617Institute for Clinical Evaluative Sciences, Toronto, Canada ,grid.17063.330000 0001 2157 2938Institute of Health Policy, Management and Evaluation, University of Toronto, Medical-Surgical ICU, 10th floor, 585 University Avenue, Toronto, ON M5G 1X5 Canada ,grid.413104.30000 0000 9743 1587Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Robert A. Fowler
- grid.17063.330000 0001 2157 2938Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Canada ,grid.17063.330000 0001 2157 2938Department of Medicine, University of Toronto, Toronto, Canada ,grid.418647.80000 0000 8849 1617Institute for Clinical Evaluative Sciences, Toronto, Canada ,grid.17063.330000 0001 2157 2938Institute of Health Policy, Management and Evaluation, University of Toronto, Medical-Surgical ICU, 10th floor, 585 University Avenue, Toronto, ON M5G 1X5 Canada ,grid.413104.30000 0000 9743 1587Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Lillian Sung
- grid.17063.330000 0001 2157 2938Institute of Health Policy, Management and Evaluation, University of Toronto, Medical-Surgical ICU, 10th floor, 585 University Avenue, Toronto, ON M5G 1X5 Canada ,grid.42327.300000 0004 0473 9646Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Canada
| | - George Tomlinson
- grid.231844.80000 0004 0474 0428Department of Medicine, University Health Network and Sinai Health System, Toronto, Canada ,grid.17063.330000 0001 2157 2938Institute of Health Policy, Management and Evaluation, University of Toronto, Medical-Surgical ICU, 10th floor, 585 University Avenue, Toronto, ON M5G 1X5 Canada
| |
Collapse
|
97
|
Massol J, Simon-Tillaux N, Tohme J, Hariri G, Dureau P, Duceau B, Belin L, Hajage D, De Rycke Y, Charfeddine A, Lebreton G, Combes A, Bouglé A. Levosimendan in patients undergoing extracorporeal membrane oxygenation after cardiac surgery: an emulated target trial using observational data. Crit Care 2023; 27:51. [PMID: 36750852 PMCID: PMC9906922 DOI: 10.1186/s13054-023-04328-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 01/21/2023] [Indexed: 02/09/2023] Open
Abstract
BACKGROUND Retrospective cohorts have suggested that levosimendan may facilitate the weaning of veno-arterial extracorporeal membrane oxygenation (VA-ECMO). We therefore studied this clinical question by emulating a randomized trial with observational data. METHODS All patients with refractory postcardiotomy cardiogenic shock and assisted with VA-ECMO, admitted to a surgical intensive care unit at La Pitié-Salpêtrière Hospital between 2016 and 2019, were eligible. To avoid immortal-time bias, we emulated a target trial sequentially comparing levosimendan administration versus no levosimendan administration in patients treated with VA-ECMO. The primary outcome was time to successful ECMO weaning. The secondary outcomes were 30-day and 1-year mortality. We performed a multivariable analysis to adjust for confounding at baseline. RESULTS Two hundred and thirty-nine patients were included in the study allowing building a nested trials cohort of 1434 copies of patients. No association of levosimendan treatment and VA-ECMO weaning was found (HR = 0.91, [0.57; 1.45], p = 0.659 in multivariable analysis), or 30-day mortality (OR = 1.03, [0.52; 2.03], p = 0.940) and 1-year mortality (OR = 1.00, [0.53; 1.89], p = 0.999). CONCLUSIONS Using the emulated target trial framework, this study did not find any association of levosimendan treatment and ECMO weaning success after postcardiotomy cardiogenic shock. However, the population of interest remains heterogeneous and subgroups might benefit from levosimendan.
Collapse
Affiliation(s)
- Julien Massol
- grid.411439.a0000 0001 2150 9058Department of Anesthesiology and Critical Care Medicine, La Pitié-Salpêtrière University Hospital, 47-83 Boulevard de L’Hôpital, 75013 Paris, France
| | - Noémie Simon-Tillaux
- grid.411439.a0000 0001 2150 9058Department of Anesthesiology and Critical Care Medicine, La Pitié-Salpêtrière University Hospital, 47-83 Boulevard de L’Hôpital, 75013 Paris, France
| | - Joanna Tohme
- grid.411439.a0000 0001 2150 9058Department of Anesthesiology and Critical Care Medicine, La Pitié-Salpêtrière University Hospital, 47-83 Boulevard de L’Hôpital, 75013 Paris, France
| | - Geoffroy Hariri
- grid.411439.a0000 0001 2150 9058Department of Anesthesiology and Critical Care Medicine, La Pitié-Salpêtrière University Hospital, 47-83 Boulevard de L’Hôpital, 75013 Paris, France
| | - Pauline Dureau
- grid.411439.a0000 0001 2150 9058Department of Anesthesiology and Critical Care Medicine, La Pitié-Salpêtrière University Hospital, 47-83 Boulevard de L’Hôpital, 75013 Paris, France
| | - Baptiste Duceau
- grid.411439.a0000 0001 2150 9058Department of Anesthesiology and Critical Care Medicine, La Pitié-Salpêtrière University Hospital, 47-83 Boulevard de L’Hôpital, 75013 Paris, France
| | - Lisa Belin
- grid.411439.a0000 0001 2150 9058Department of Anesthesiology and Critical Care Medicine, La Pitié-Salpêtrière University Hospital, 47-83 Boulevard de L’Hôpital, 75013 Paris, France
| | - David Hajage
- grid.411439.a0000 0001 2150 9058Department of Anesthesiology and Critical Care Medicine, La Pitié-Salpêtrière University Hospital, 47-83 Boulevard de L’Hôpital, 75013 Paris, France
| | - Yann De Rycke
- grid.411439.a0000 0001 2150 9058Department of Anesthesiology and Critical Care Medicine, La Pitié-Salpêtrière University Hospital, 47-83 Boulevard de L’Hôpital, 75013 Paris, France
| | - Ahmed Charfeddine
- grid.411439.a0000 0001 2150 9058Department of Anesthesiology and Critical Care Medicine, La Pitié-Salpêtrière University Hospital, 47-83 Boulevard de L’Hôpital, 75013 Paris, France
| | - Guillaume Lebreton
- grid.411439.a0000 0001 2150 9058Department of Anesthesiology and Critical Care Medicine, La Pitié-Salpêtrière University Hospital, 47-83 Boulevard de L’Hôpital, 75013 Paris, France
| | - Alain Combes
- grid.411439.a0000 0001 2150 9058Department of Anesthesiology and Critical Care Medicine, La Pitié-Salpêtrière University Hospital, 47-83 Boulevard de L’Hôpital, 75013 Paris, France
| | - Adrien Bouglé
- Department of Anesthesiology and Critical Care Medicine, La Pitié-Salpêtrière University Hospital, 47-83 Boulevard de L'Hôpital, 75013, Paris, France.
| |
Collapse
|
98
|
Crown W, Dahabreh IJ, Li X, Toh S, Bierer B. Can Observational Analyses of Routinely Collected Data Emulate Randomized Trials? Design and Feasibility of the Observational Patient Evidence for Regulatory Approval Science and Understanding Disease Project. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2023; 26:176-184. [PMID: 35970705 DOI: 10.1016/j.jval.2022.07.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 06/28/2022] [Accepted: 07/10/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVES The Observational Patient Evidence for Regulatory Approval Science and Understanding Disease (OPERAND) project examines whether real-world data (RWD) can be used to inform regulatory decision making. METHODS OPERAND evaluates whether observational analyses using RWD to emulate index trials can produce effect estimates similar to those of the trials and examines the impact of relaxing the eligibility criteria of the observational analyses to obtain samples that more closely match the real-world populations receiving the treatments. In OPERAND, 2 research teams independently attempt to emulate the ROCKET Atrial Fibrillation and LEAD-2 trials using OptumLabs data. This article describes the design of the project, summarizes the approaches of the 2 research teams, and presents feasibility results for 2 emulations using new-user designs. RESULTS There were differences in the teams' conceptualizations of the emulation, design decisions for cohort identification, and resulting RWD cohorts. These differences occurred even though both teams were guided by the same index trials and had access to the same source of RWD. CONCLUSIONS Reasonable alternative design and analysis approaches may be taken to answer the same research question, even when attempting to emulate the same index trial. Researcher decision making is an understudied and potentially important source of variability across RWD analyses.
Collapse
Affiliation(s)
- William Crown
- OptumLabs, Eden Prairie, MN, USA; Brandeis University, Waltham, MA, USA.
| | - Issa J Dahabreh
- OptumLabs Visiting Fellow, Eden Prairie, MN, USA; Departments of Epidemiology and Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Xiaojuan Li
- OptumLabs Visiting Fellow, Eden Prairie, MN, USA; Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Sengwee Toh
- OptumLabs Visiting Fellow, Eden Prairie, MN, USA; Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Barbara Bierer
- OptumLabs Visiting Fellow, Eden Prairie, MN, USA; Department of Medicine, Brigham and Women's Hospital, Harvard University, Boston, MA, USA
| |
Collapse
|
99
|
Dusingize JC, Olsen CM, Law MH, Pandeya N, Neale RE, MacGregor S, Whiteman DC, Ong JS. Cholesterol-lowering genetic variants are not associated with the risk of skin cancer. J Eur Acad Dermatol Venereol 2023; 37:e792-e795. [PMID: 36662597 DOI: 10.1111/jdv.18886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 01/13/2023] [Indexed: 01/21/2023]
Affiliation(s)
- Jean Claude Dusingize
- Departments of Population Health and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Catherine M Olsen
- Departments of Population Health and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Matthew H Law
- Departments of Population Health and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, Australia.,School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Nirmala Pandeya
- Departments of Population Health and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,School of Public Health, University of Queensland, Brisbane, Australia
| | - Rachel E Neale
- Departments of Population Health and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,School of Public Health, University of Queensland, Brisbane, Australia
| | - Stuart MacGregor
- Departments of Population Health and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - David C Whiteman
- Departments of Population Health and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,School of Public Health, University of Queensland, Brisbane, Australia
| | - Jue-Sheng Ong
- Departments of Population Health and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
100
|
Wirth KE, Edwards JK, Feinstein L, Breskin A. When Emulating a Trial, Do as the Trialists Do: Missteps in Estimating Relative Effectiveness of a Severe Acute Respiratory Syndrome Coronavirus 2 Vaccine Booster Dose. Clin Infect Dis 2023; 76:176-177. [PMID: 36041013 PMCID: PMC9452123 DOI: 10.1093/cid/ciac700] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 08/24/2022] [Indexed: 01/11/2023] Open
Affiliation(s)
- Kathleen E Wirth
- NoviSci, a division of Target RWE Health Evidence Solutions, Durham, NC, USA
| | - Jessie K Edwards
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lydia Feinstein
- NoviSci, a division of Target RWE Health Evidence Solutions, Durham, NC, USA
| | - Alexander Breskin
- NoviSci, a division of Target RWE Health Evidence Solutions, Durham, NC, USA
| |
Collapse
|