1
|
Harkey BA, Distefano S, Pagliaro JA, Heyd L, Chase M, Igne C, Yu H, Sherman AV, Dagostino D, Tustison E, Changkuon G, Hall M, Kittle G, Connolly MR, Giacomelli E, Scirocco E, Berry JD, Babu S, Shefner J, Macklin EA, Chibnik LB, De Mattos A, Drake K, Kamp C, McGarry A, Torti M, Small C, Bulat A, Cudkowicz ME, Paganoni S, HEALEY ALS Platform Trial Study Group. Operational Development and Launch of an Adaptive Platform Trial in Amyotrophic Lateral Sclerosis: Processes and Learnings From the First Four Regimens of the HEALEY ALS Platform Trial. Muscle Nerve 2025. [PMID: 40420561 DOI: 10.1002/mus.28442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Collaborators] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 05/01/2025] [Accepted: 05/10/2025] [Indexed: 05/28/2025]
Abstract
INTRODUCTION/AIMS Platform trials present several advantages over traditional interventional clinical trials. Here, we provide a detailed description of the operational framework of the HEALEY ALS Platform Trial. METHODS Platform-level procedures for regulatory oversight, safety, and site management were developed prior to trial launch. Central vendors and a single Institutional Review Board (sIRB) were used. An Investigational New Drug (IND) application was submitted for the master protocol, and each regimen was added as an amendment. RESULTS The HEALEY ALS Platform Trial was launched in 2020. Fifty-four geographically diverse sites from the Northeast ALS Consortium (NEALS), all highly experienced in ALS care and research, were selected. Three investigational products were selected to launch concurrently at the start of the trial as individual regimens. A fourth investigational product was selected and added to the trial after the initial launch. The Master Protocol and the first three regimens (Regimens A-C) were sIRB approved in 120 days. sIRB amendment for Regimen D was approved in 21 days. Enrollment for regimens A-C was completed in 15 months, whereas Regimen D was completed in 11 months from the start of enrollment. Results of all regimens were available within approximately 2 years from the initial trial launch. DISCUSSION The HEALEY ALS Platform Trial capitalized on the benefits of the platform approach, including an adaptable operational infrastructure, concurrent enrollment into four distinct regimens, and an accelerated start-up time for a new regimen added after initial trial launch.
Collapse
Affiliation(s)
- Brittney A Harkey
- Sean M. Healey and AMG Center for ALS & The Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sofia Distefano
- Sean M. Healey and AMG Center for ALS & The Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jaclyn A Pagliaro
- Sean M. Healey and AMG Center for ALS & The Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Lindsay Heyd
- Sean M. Healey and AMG Center for ALS & The Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Marianne Chase
- Sean M. Healey and AMG Center for ALS & The Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Courtney Igne
- Sean M. Healey and AMG Center for ALS & The Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Hong Yu
- Sean M. Healey and AMG Center for ALS & The Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Alexander V Sherman
- Sean M. Healey and AMG Center for ALS & The Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Derek Dagostino
- Sean M. Healey and AMG Center for ALS & The Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Eric Tustison
- Sean M. Healey and AMG Center for ALS & The Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Genevive Changkuon
- Sean M. Healey and AMG Center for ALS & The Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Meghan Hall
- Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Gale Kittle
- Barrow Neurological Institute, Phoenix, Arizona, USA
| | | | - Elisa Giacomelli
- Sean M. Healey and AMG Center for ALS & The Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Erica Scirocco
- Sean M. Healey and AMG Center for ALS & The Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - James D Berry
- Sean M. Healey and AMG Center for ALS & The Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Suma Babu
- Sean M. Healey and AMG Center for ALS & The Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Eric A Macklin
- Biostatistics, Massachusetts General Hospital, Departments of Medicine and Neurology, Harvard Medical School, Boston, Massachusetts, USA
| | - Lori B Chibnik
- Biostatistics, Massachusetts General Hospital, Departments of Medicine and Neurology, Harvard Medical School, Boston, Massachusetts, USA
| | - Annette De Mattos
- Sean M. Healey and AMG Center for ALS & The Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kristin Drake
- Sean M. Healey and AMG Center for ALS & The Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Cornelia Kamp
- Clintrex Research Corporation, Sarasota, Florida, USA
- University of Rochester, Center for Health and Technology, Clinical Materials Services Unit, Rochester, New York, USA
| | | | | | - Catherine Small
- Sean M. Healey and AMG Center for ALS & The Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Allison Bulat
- Sean M. Healey and AMG Center for ALS & The Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Merit E Cudkowicz
- Sean M. Healey and AMG Center for ALS & The Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sabrina Paganoni
- Sean M. Healey and AMG Center for ALS & The Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
Collaborators
Eric A Macklin, Lori B Chibnik, Douglas Hayden, Po-Ying Lai, Rachel A Donahue, Hao-Wun Chen, Jianing Wang, Melanie Quintana, Benjamin R Saville, Michelle A Detry, Joe Marion, Matteo Vestrucci, Anna McGlothlin, Gustavo Alameda, Nithya Mathai, Eduardo Locatelli, Gabriela Lopes, Sabrina Paganoni, Merit Cudkowicz, Doreen Ho, Alexandra McCaffrey, James D Berry, Suma Babu, Jennifer Scalia, Sarah Luppino, Clotilde Lagier-Tourenne, Ghazaleh Sadri-Vakili, Elisa Giacomelli, Erica Scirocco, Raina Chrobak, Adam Quick, Stephen Kolb, Sarah Heintzman, Senda Ajroud-Driss, Robert Sufit, April Szymanski, Jonathan Katz, Liberty Jenkins, Daragh Heitzman, Alan Martin, Stanley H Appel, Simpson Greene Ericka, Jason R Thonhoff, Sheetal Shroff, Bing Liao, Kevin Felice, Charles Whitaker, Nicholas J Maragakis, Lora L Clawson, Alpa Uchil, Kristen M Riley, JinAe Arneklev, Zachary Simmons, James Grogan, Xiaowei Su, Mansoureh Mamarabadi, Timothy M Miller, Amber Malcolm, Nicholas Olney, Tracy Bazan, Michael D Weiss, Nassim Rad, Leo H Wang, Stephen A Goutman, Eva L Feldman, Joseph Americo Fernandes, Ezequiel Piccione, Pariwat Thaisetthawatkul, Omar Jawdat, Constantine Farmakidis, Duaa Jabari, Jeffrey Statland, Mamatha Pasnoor, Mazen Dimachkie, Margaret Ayo Owegi, Robert H Brown, Mehdi Ghasemi, Hajar Houmani, Catherine Douthwright, Kate Daniello, Laura A Foster, Tuan Vu, Niraja Suresh, Jerrica Farias, I-Hweii A Chen, Hristelina Ilieva, Piera Pasinelli, Daniel S Newman, Ximena Arcila-Londono, Kara Steijlen, Carlayne E Jackson Md, Ratna Bhavaraju-Sanka, Shafeeq Ladha, Bill Jacobsen, Jourdan Milliard, Robert Bowser, Jeremy M Shefner, Terry Heiman-Patterson, Anahita Deboo, James B Caress, Michael S Cartwright, Andrea Swenson, Christopher Nance, Ludwig Gutmann, Jinsy Andrews, Julia Yasek, Matthew Harms, Amanda Peltier, Richard Lewis, Matthew Burford, Frank Diaz, Dominic Fee, David Shrilla, Matthew Elliott, Goran Rakocevic, Sarah Jones, Guillermo Solorzano, Richard Bedlack, Xiaoyan Li, Edward J Kasarskis, Zabeen Mahuwala, Vish Mathur Kumaraswamy Vishakhadatta, M D Lauren Elman, Colin Quinn, Michael Baer, Jeffrey Rosenfeld, David Borg, Karthikeyan Bhuvaneswaran, Jasdeep Kaur, David Walk, Sam Maiser, Seward B Rutkove, Courtney E McIlduff, Paul Twydell, Andrew Mundwiler, Eufrosina Young, Jenny A Meyer, Kristin Johnson, Pooja Rao, Kourosh Rezania, Betty Soliven, Raymond Roos, Namita A Goyal, Ali A Habib, Tahseen Mozaffar, Manisha Kak Korb, Jeffrey Mullen, Jeffrey A Cohen, Elijah Stommel, Nathaniel M Robbins, Michael Benatar, Nathan Carberry, Volkan Granit, Raghav Govindarajan, Vovanti Jones, Christina N Fournier, Jaimin S Shah, Bjorn Oskarsson, Said R Beydoun, Leila Darki, Rodrigo Rodriguez, James P Wymer, Miguel Chuquilin, Whitney McNeely, Lindsay Zilliox, Montserrat Diaz-Abad, Peter H Jin, Chandana Chauhan, Shakti Nayar, Gary L Pattee, James Bobenhouse, Jennifer Martinez-Thompson, P Staff Nathan, Ghazala Hayat, Luisa Arroyave, Abbey Bailey, Jesse Bailey, Victoria Barlow, Allison Bulat, Genevive Changkuon, Marianne Chase, Melissa Cirino, Derek Dagostino, Cristina Deignan, Emma Deirmendjian, Annette De Mattos, Sofia DiStefano, Kristin Drake, Michaela Estes, Kenneth Faulconer, Precious Figueroa-Szostek, Tessa Garozzo, Brittney A Harkey, Meredith Gibbons Hasenoehrl, Jennifer Henrique, Natalie Henrique, Samuel Hurwitz, Courtney Igne, Liam Irwin, Katie Jentoft, Boglarka Jordan, Igor Katsovskiy, Olga Kharakozova, Taylor Kolvek, Alexander Korin, Thuong La, Haining Li, Joey Nguyen, Ilya Novak, Ricardo Ortiz, Joe Ostrow, Jaclyn Pagliaro, Jack Palillo, Payal Patel, Janae Patterson, Minh Phan, Najla Popel, Lindsay Pothier, Serena Proueng, Jesse Rosenthal, Alexander V Sherman, Catherine Small, Natalia Tarasenko, Mirna Thomas, Eric Tustison, Prasha Vigneswaran, Yusra Wahab, Isaac Whitworth, Spencer Wright, Hong Yu, Mariah Connolly, Diana De Santiago, Adrian Felix, Karly Garrett, Meghan Hall, Jenny Hamilton, Kamran Khan, Gale Kittle, Marlee Lovett, Linda Nelson, Marissa Pabon, Diana Rede, Patrick Bolger, Ahmed Fetouh, Joan Woodcook, Cornelia Kamp, Julie Kennedy, Andrew McGarry, Margherita Torti,
Collapse
|
2
|
Massonnaud CR, Schönenberger CM, Chiaborelli M, Ehrenzeller S, Griessbach A, Gillibert A, Briel M, Laouénan C. Characteristics, design, and statistical methods in platform trials: a systematic review. J Clin Epidemiol 2025; 184:111827. [PMID: 40349734 DOI: 10.1016/j.jclinepi.2025.111827] [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: 03/14/2025] [Revised: 04/27/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND AND OBJECTIVE Platform trials (PTs) are gaining popularity in clinical research due to their innovative and flexible methodologies. The objective was to determine the characteristics, methodological, and statistical practices in PTs. METHODS We identified PTs from trial registries and bibliographic databases up to August 2024. Eligible PTs were randomized controlled trials studying multiple interventions within a single population, with flexibility to add or drop arms. Data were extracted on trial status, design, statistical methods, and reporting practices. RESULTS We identified 189 PTs. Most focused on infectious diseases (77, including 57 for COVID-19) and oncology (68). PT initiation peaked during the COVID-19 pandemic but has since stabilized at 84 active PTs, with 25 in planning. A complete master protocol was available for 47% (89/189) of PTs. Bayesian designs featured in 58/189 PTs vs. 56/189 frequentist trials, 20/189 trials utilizing both (unclear in 55/189 PTs). Overall, 25/111 trials (23%) were designed without a predetermined target sample size, all of which were Bayesian. Among these, 16 were explicitly reported as "perpetual" trials. The number of interim analyses was predetermined in 18% (10/57) of Bayesian trials vs. 58% (28/48) of frequentist trials. Simulations to evaluate operating characteristics were used in 93% (39/42) of Bayesian trials. Simulation reports were available in 67% (26/39) of cases, and the procedures were detailed for 62% (24/39) of trials. Only two trials shared the simulation code. CONCLUSION PTs remain popular and increasingly diverse. Efforts to enhance transparency and reporting, especially in complex Bayesian PTs, are essential to ensure reliability and broader acceptance.
Collapse
Affiliation(s)
- Clément R Massonnaud
- Université Paris Cité, Inserm, IAME, Paris F-75018, France; AP-HP, Hôpital Bichat, Département d'Épidémiologie, Biostatistique et Recherche Clinique, Paris F-75018, France.
| | - Christof Manuel Schönenberger
- CLEAR Methods Center, Division of Clinical Epidemiology, Department of Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Malena Chiaborelli
- CLEAR Methods Center, Division of Clinical Epidemiology, Department of Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Selina Ehrenzeller
- CLEAR Methods Center, Division of Clinical Epidemiology, Department of Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Alexandra Griessbach
- CLEAR Methods Center, Division of Clinical Epidemiology, Department of Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
| | | | - Matthias Briel
- CLEAR Methods Center, Division of Clinical Epidemiology, Department of Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland; Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada
| | - Cédric Laouénan
- Université Paris Cité, Inserm, IAME, Paris F-75018, France; AP-HP, Hôpital Bichat, Département d'Épidémiologie, Biostatistique et Recherche Clinique, Paris F-75018, France
| |
Collapse
|
3
|
Medicherla CB, Salem A, Dammavalam V, Spirollari E, Jain A, Sacknovitz A, Mayer SA, Gandhi CD, Al-Mufti F. StrokeNet Thrombectomy Endovascular Platform (STEP): A Catalyst for Stroke Research. Cardiol Rev 2025:00045415-990000000-00471. [PMID: 40262022 DOI: 10.1097/crd.0000000000000920] [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] [Indexed: 04/24/2025]
Abstract
Endovascular thrombectomy is a critical treatment for acute ischemic stroke. Challenges remain in extending treatment windows, refining patient selection, optimizing posttreatment interventions, and expanding endovascular thrombectomy indications. Traditional stroke trials have played a pivotal role in advancing care but are hindered by inefficiencies, rigid designs, high costs, long timelines, and slow enrollment. The StrokeNet Thrombectomy Endovascular Platform (STEP) represents an innovative paradigm shift in stroke research, offering a more agile, cost-effective, and flexible trial design. Here, we explore the architecture of STEP and its implications on stroke research.
Collapse
Affiliation(s)
- Chaitanya B Medicherla
- From the Department of Neurology, New York Medical College, Westchester Medical Center, Valhalla, NY
| | - Amr Salem
- From the Department of Neurology, New York Medical College, Westchester Medical Center, Valhalla, NY
| | - Vikalpa Dammavalam
- From the Department of Neurology, New York Medical College, Westchester Medical Center, Valhalla, NY
| | - Eris Spirollari
- Department of Neurology, Neurosurgery, and Radiology, New York Medical College, Westchester Medical Center, Valhalla, NY
| | - Ankita Jain
- Department of Neurology, Neurosurgery, and Radiology, New York Medical College, Westchester Medical Center, Valhalla, NY
| | - Ariel Sacknovitz
- Department of Neurology, Neurosurgery, and Radiology, New York Medical College, Westchester Medical Center, Valhalla, NY
| | - Stephan A Mayer
- From the Department of Neurology, New York Medical College, Westchester Medical Center, Valhalla, NY
| | - Chirag D Gandhi
- Department of Neurology, Neurosurgery, and Radiology, New York Medical College, Westchester Medical Center, Valhalla, NY
| | - Fawaz Al-Mufti
- Department of Neurology, Neurosurgery, and Radiology, New York Medical College, Westchester Medical Center, Valhalla, NY
| |
Collapse
|
4
|
Angus DC. Effect of hydrocortisone on mortality in patients with severe community-acquired pneumonia : The REMAP-CAP Corticosteroid Domain Randomized Clinical Trial. Intensive Care Med 2025; 51:665-680. [PMID: 40261382 PMCID: PMC12055926 DOI: 10.1007/s00134-025-07861-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 03/09/2025] [Indexed: 04/24/2025]
Abstract
PURPOSE To determine whether hydrocortisone improves mortality in severe community-acquired pneumonia (CAP). METHODS In an international adaptive randomized controlled platform trial testing multiple interventions, adults admitted to the intensive care unit (ICU) with severe CAP were randomized to a 7-day course of intravenous hydrocortisone (50 mg every 6 h) or control (no corticosteroid). The primary end point was 90-day all-cause mortality, analyzed iteratively by a Bayesian hierarchical model estimating distinct treatment effects for patients presenting with influenza (Y/N) and shock (Y/N). RESULTS Fixed 7-day course hydrocortisone enrollment was stopped for futility (< 5% probability of > 20% relative improvement). Of 658 patients enrolled, 536 were randomized to hydrocortisone and 122 to control. Vital status at day 90 was missing for 15 patients. Day 90 mortality was 15% (78/521) and 9.8% (12/122) for the hydrocortisone and control groups. The adjusted odds ratio ranged from 1.52 to 1.63 (with all 95% CrI crossing 1), while the probability of > 20% relative reduction of day 90 mortality ranged from 7.1 to 3.3% across influenza and shock strata. Results were consistent in sensitivity and pre-specified secondary outcomes. In exploratory analyses, the duration of shock appeared lower in the hydrocortisone group compared with control (median (IQR) of 2 (2-5) days compared to control 3 (2-6.75) days, p value = 0.05). CONCLUSIONS Among patients with severe CAP, treatment with a 7-day course of hydrocortisone, compared with no hydrocortisone, appears unlikely to yield a large reduction in mortality. Smaller benefits and possible harm are not excluded. TRIAL REGISTRATION Clinicaltrials.gov identifier: NCT02735707 (registration date: November 4th, 2016).
Collapse
Affiliation(s)
- Derek C Angus
- University of Pittsburgh School of Medicine, Pittsburgh, USA.
| |
Collapse
|
5
|
Adalja AA, Inglesby TV. Immunomodulator Stockpiling as a Means of Broad Defense From Biological Threats. Crit Care Explor 2025; 7:e1244. [PMID: 40126911 PMCID: PMC11936613 DOI: 10.1097/cce.0000000000001244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2025] Open
Affiliation(s)
- Amesh A. Adalja
- Both authors: Johns Hopkins Center for Health Security, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Thomas V. Inglesby
- Both authors: Johns Hopkins Center for Health Security, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| |
Collapse
|
6
|
Delgado C, Nogara PA, Miranda MD, Rosa AS, Ferreira VNS, Batista LT, Oliveira TKF, Omage FB, Motta F, Bastos IM, Orian L, Rocha JBT. In Silico and In Vitro Studies of the Approved Antibiotic Ceftaroline Fosamil and Its Metabolites as Inhibitors of SARS-CoV-2 Replication. Viruses 2025; 17:491. [PMID: 40284934 PMCID: PMC12031345 DOI: 10.3390/v17040491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/14/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
The SARS-CoV-2 proteases Mpro and PLpro are critical targets for antiviral drug development for the treatment of COVID-19. The 1,2,4-thiadiazole functional group is an inhibitor of cysteine proteases, such as papain and cathepsins. This chemical moiety is also present in ceftaroline fosamil (CF), an FDA-approved fifth-generation cephalosporin antibiotic. This study investigates the interactions between CF, its primary metabolites (M1 is dephosphorylated CF and M2 is an opened β-lactam ring) and derivatives (protonated M1H and M2H), and its open 1,2,4-thiadiazole rings derivatives (open-M1H and open-M2H) with SARS-CoV-2 proteases and evaluates CF's effects on in vitro viral replication. In silico analyses (molecular docking and molecular dynamics (MD) simulations) demonstrated that CF and its metabolites are potential inhibitors of PLpro and Mpro. Docking analysis indicated that the majority of the ligands were more stable with Mpro than PLpro; however, in vitro biochemical analysis indicated PLpro as the preferred target for CF. CF inhibited viral replication in the human Calu-3 cell model at submicromolar concentrations when added to cell culture medium at 12 h. Our results suggest that CF should be evaluated as a potential repurposing agent for COVID-19, considering not only viral proteases but also other viral targets and relevant cellular pathways. Additionally, the reactivity of sulfur in the 1,2,4-thiadiazole moiety warrants further exploration for the development of viral protease inhibitors.
Collapse
Affiliation(s)
- Cássia Delgado
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria 97000-000, RS, Brazil; (C.D.); (J.B.T.R.)
| | - Pablo Andrei Nogara
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria 97000-000, RS, Brazil; (C.D.); (J.B.T.R.)
- Instituto Federal de Educação, Ciência e Tecnologia Sul-rio-grandense (IFSul), Bagé 96400-000, RS, Brazil
| | - Milene Dias Miranda
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.S.R.); (V.N.S.F.); (L.T.B.); (T.K.F.O.)
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil
| | - Alice Santos Rosa
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.S.R.); (V.N.S.F.); (L.T.B.); (T.K.F.O.)
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil
| | - Vivian Neuza Santos Ferreira
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.S.R.); (V.N.S.F.); (L.T.B.); (T.K.F.O.)
| | - Luisa Tozatto Batista
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.S.R.); (V.N.S.F.); (L.T.B.); (T.K.F.O.)
| | - Thamara Kelcya Fonseca Oliveira
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.S.R.); (V.N.S.F.); (L.T.B.); (T.K.F.O.)
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil
| | - Folorunsho Bright Omage
- Biological Chemistry Laboratory, Department of Organic Chemistry, Institute of Chemistry, University of Campinas (UNICAMP), Campinas 13000-000, SP, Brazil;
| | - Flávia Motta
- Laboratório de interface patógeno-hospedeiro, Departamento de Biologia Celular, Universidade de Brasília (UnB), Brasília 70910-900, DF, Brazil; (F.M.); (I.M.B.)
| | - Izabela Marques Bastos
- Laboratório de interface patógeno-hospedeiro, Departamento de Biologia Celular, Universidade de Brasília (UnB), Brasília 70910-900, DF, Brazil; (F.M.); (I.M.B.)
| | - Laura Orian
- Dipartimento di Scienze Chimiche, Università degli Studi di Padova, Via Marzolo 1, 35129 Padova, Italy;
| | - João Batista Teixeira Rocha
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria 97000-000, RS, Brazil; (C.D.); (J.B.T.R.)
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90000-000, RS, Brazil
| |
Collapse
|
7
|
Dhaka P, Mahto JK, Singh A, Kumar P, Tomar S. Structural insights into the RNA binding inhibitors of the C-terminal domain of the SARS-CoV-2 nucleocapsid. J Struct Biol 2025; 217:108197. [PMID: 40113149 DOI: 10.1016/j.jsb.2025.108197] [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: 10/09/2024] [Revised: 01/26/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025]
Abstract
The SARS-CoV-2 nucleocapsid (N) protein is an essential structural element of the virion, playing a crucial role in enclosing the viral genome into a ribonucleoprotein (RNP) assembly, as well as viral replication and transmission. The C-terminal domain of the N-protein (N-CTD) is essential for encapsidation, contributing to the stabilization of the RNP complex. In a previous study, three inhibitors (ceftriaxone, cefuroxime, and ampicillin) were screened for their potential to disrupt the RNA packaging process by targeting the N-protein. However, the binding efficacy, mechanism of RNA binding inhibition, and molecular insights of binding with N-CTD remain unclear. In this study, we evaluated the binding efficacy of these inhibitors using isothermal titration calorimetry (ITC), revealing the affinity of ceftriaxone (18 ± 1.3 μM), cefuroxime (55 ± 4.2 μM), and ampicillin (28 ± 1.2 μM) with the N-CTD. Further inhibition assay and fluorescence polarisation assay demonstrated RNA binding inhibition, with IC50 ranging from ∼ 12 to 18 μM and KD values between 24 μM to 32 μM for the inhibitors, respectively. Additionally, we also determined the inhibitor-bound complex crystal structures of N-CTD-Ceftriaxone (2.0 Å) and N-CTD-Ampicillin (2.2 Å), along with the structure of apo N-CTD (1.4 Å). These crystal structures revealed previously unobserved interaction sites involving residues K261, K266, R293, Q294, and W301 at the oligomerization interface and the predicted RNA-binding region of N-CTD. These findings provide valuable molecular insights into the inhibition of N-CTD, highlighting its potential as an underexplored but promising target for the development of novel antiviral agents against coronaviruses.
Collapse
Affiliation(s)
- Preeti Dhaka
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - Jai Krishna Mahto
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - Ankur Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - Pravindra Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India.
| | - Shailly Tomar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India.
| |
Collapse
|
8
|
Daneman N, Rishu AH, Pinto R, Arabi YM, Cook DJ, Hall R, Muscedere J, Parke R, Reynolds S, Rogers B, Shehabi Y, Fowler RA. Investigator-initiated randomized clinical trials in infectious diseases: the BALANCE experience. Clin Microbiol Infect 2025:S1198-743X(25)00094-1. [PMID: 40024529 DOI: 10.1016/j.cmi.2025.02.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 02/19/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025]
Affiliation(s)
- Nick Daneman
- Division of Infectious Diseases, Sunnybrook Health Sciences Centre, University of Toronto and Adjunct Scientist, Institute for Clinical Evaluative Sciences, Toronto, ON, Canada.
| | - Asgar H Rishu
- Department of Critical Care Medicine, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Ruxandra Pinto
- Department of Critical Care Medicine, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Yaseen M Arabi
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Centre, Intensive Care Department, Ministry of National Guard Health Affairs, Riyadh, Kingdom of Saudi Arabia
| | - Deborah J Cook
- Division of Critical Care Medicine, Departments of Medicine, Clinical Epidemiology & Biostatistics, McMaster University, Hamilton, ON, Canada
| | - Richard Hall
- Departments of Critical Care Medicine and Anesthesiology, Pain Management and Perioperative Medicine, Dalhousie University, Halifax NS, Canada
| | - John Muscedere
- Department of Critical Care Medicine, Queen's University, Kingston, ON, Canada
| | - Rachael Parke
- School of Nursing, The University of Auckland, Auckland, New Zealand
| | - Steven Reynolds
- Department of Biophysiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Benjamin Rogers
- Centre for Inflammatory Diseases, Monash University School of Clinical Sciences, Clayton, Victoria, Australia; Monash Infectious Diseases, Monash Health, Clayton, Victoria, Australia
| | - Yahya Shehabi
- Critical Care and Perioperative Medicine, School of Clinical Sciences, Monash University and Monash Health, Clayton, Victoria, Australia; The Clinical School of Medicine, University of New South Wales, Randwick, New South Wales, Australia
| | - Robert A Fowler
- Departments of Medicine and Critical Care Medicine, Adjunct Scientist, Institute for Clinical Evaluative Sciences, Sunnybrook Health Sciences Centre, Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
9
|
Mahar RK, McGlothlin A, Dymock M, Barina L, Bonten M, Bowen A, Cheng MP, Daneman N, Goodman AL, Lee TC, Lewis RJ, Lumley T, McLean ARD, McQuilten Z, Mora J, Paterson DL, Price DJ, Roberts J, Snelling T, Tverring J, Webb SA, Yahav D, Davis JS, Tong SYC, Marsh JA. Statistical documentation for multi-disease, multi-domain platform trials: our experience with the Staphylococcus aureus Network Adaptive Platform trial. Trials 2025; 26:49. [PMID: 39934879 PMCID: PMC11817021 DOI: 10.1186/s13063-024-08684-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 12/04/2024] [Indexed: 02/13/2025] Open
Abstract
Platform trials have become widely adopted across multiple disease areas over recent years, however, guidelines for operationalising these trials have not kept pace. We outline a series of documents that summarise the statistical components, and implicit processes, of the Staphylococcus aureus Network Adaptive Platform (SNAP) trial to provide an informal template for other researchers and reviewers of platform trials. We briefly summarise the content and role of the core protocol, statistical appendix, domain-specific appendices, simulation report, statistical implementation guides, data safety and monitoring committee (DSMC) reports, and domain-specific statistical analysis plans and final reports, and a transparent governance structure that ensures separate blinded and unblinded statistical teams. In the absence of guidelines or checklists for platform trial statistical documents, we hope to provide useful guidance to others in terms of what has worked so far for the SNAP trial, stimulate discussion, and inform a future consensus.Trial registration NCT05137119 . Registered on 30 November 2021.
Collapse
Affiliation(s)
- Robert K Mahar
- Centre for Epidemiology, Melbourne School of Population and Global Health, University of Melbourne, Parkville, Australia.
- Clinical Epidemiology and Biostatistics Unit, Murdoch Children's Research Institute, Parkville, Australia.
- Methods and Implementation Support for Clinical Research Hub, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia.
| | | | - Michael Dymock
- Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, Nedlands, WA, Australia
- School of Population and Global Health, University of Western Australia, Perth, WA, Australia
| | - Lauren Barina
- Department of Infectious Diseases, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Marc Bonten
- Julius Centre for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- European Clinical Research Alliance On Infectious Diseases, Utrecht, the Netherlands
| | - Asha Bowen
- Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, Nedlands, WA, Australia
- Perth Children's Hospital, Nedlands, WA, Australia
| | - Matthew P Cheng
- Division of Infectious Diseases and Medical Microbiology, McGill University Health Centre, Montreal, Canada
- McGill University, Montreal, Canada
| | - Nick Daneman
- Division of Infectious Diseases, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | - Anna L Goodman
- MRC Clinical Trials Unit at University College London, London, UK
- Centre for Infection Diagnostics Research at King's College, London, UK
- Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Todd C Lee
- Division of Infectious Diseases and Medical Microbiology, McGill University Health Centre, Montreal, Canada
| | - Roger J Lewis
- Berry Consultants, LLC, Austin, TX, USA
- David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Thomas Lumley
- Department of Statistics, University of Auckland, Auckland, New Zealand
| | - Alistair R D McLean
- Methods and Implementation Support for Clinical Research Hub, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia
| | - Zoe McQuilten
- School of Public Health and Preventative Medicine, Monash University, Melbourne, Australia
- Department of Haematology, Alfred Health, Melbourne, Australia
| | - Jocelyn Mora
- Department of Infectious Diseases, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - David L Paterson
- ADVANCE-ID, Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| | - David J Price
- Centre for Epidemiology, Melbourne School of Population and Global Health, University of Melbourne, Parkville, Australia
- Department of Infectious Diseases, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Jason Roberts
- Faculty of Medicine, University of Queensland Centre for Clinical Research, University of Queensland, Brisbane, Australia
- Herston Infectious Diseases Institute, Metro North Health, Brisbane, Australia
- Departments of Pharmacy and Intensive Care Medicine, Royal Brisbane and Women's Hospital, Brisbane, Australia
- Division of Anaesthesiology, Critical Care, Emergency and Pain Medicine, Nîmes University Hospital, University of Montpellier, Montpellier, France
| | - Tom Snelling
- Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, Nedlands, WA, Australia
- Department of Infectious Diseases, Perth Children's Hospital, Perth, WA, Australia
- Sydney School of Public Health, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Jonas Tverring
- Department of Infectious Diseases, Helsingborg Hospital, Helsingborg, Sweden
- Department of Clinical Sciences Helsingborg, Lund University, Lund, Sweden
| | - Steve A Webb
- Australian and New Zealand Intensive Care Research Centre, Monash University, Melbourne, Australia
| | - Dafna Yahav
- Infectious Diseases Unit, Sheba Medical Center, Ramat-Gan, Israel
| | - Joshua S Davis
- Infection Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Sydney, Australia
- School of Medicine and Public Health, University of Newcastle, Newcastle, Australia
| | - Steven Y C Tong
- Department of Infectious Diseases, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Victorian Infectious Diseases Service, Royal Melbourne Hospital, at the, Peter Doherty Institute for Infection and Immunity , Melbourne, Australia
| | - Julie A Marsh
- Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, Nedlands, WA, Australia
- Medical School, Centre for Child Health Research, University of Western Australia, Perth, Australia
| |
Collapse
|
10
|
Schellong P, Joean O, Pletz MW, Hagel S, Weis S. Treatment of Complicated Gram-Positive Bacteremia and Infective Endocarditis. Drugs 2025; 85:193-214. [PMID: 39720961 PMCID: PMC11802659 DOI: 10.1007/s40265-024-02135-z] [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] [Accepted: 11/28/2024] [Indexed: 12/26/2024]
Abstract
The Gram-positive cocci Staphylococcus aureus, Streptococcus spp., and Enterococcus spp. are the most frequent causative organisms of bloodstream infections and infective endocarditis. "Complicated bacteremia" is a term used in S. aureus bloodstream infections and originally implied the presence of metastatic infectious foci (i.e. complications of S. aureus bacteremia). These complications demand longer antimicrobial treatment durations and, frequently, interventional source control. Several risk factors for the incidence of bacteremia complications have been identified and are often used for the definition of complicated bacteremia. Here, we discuss management and diagnostic approaches and treatment options for patients with complicated bacteremia, with particular focus on infective endocarditis. We also summarize the available evidence regarding imaging modalities and the choice of antimicrobial mono- or combination therapy according to resistance patterns for these pathogens as well as treatment durations and optimized application routes. Finally, we synopsize current and future areas of research in complicated bacteremia and infective endocarditis.
Collapse
Affiliation(s)
- Paul Schellong
- Institute for Infectious Disease and Infection Control, Jena University Hospital, Friedrich-Schiller-University, Am Klinikum 1, 07749, Jena, Germany.
- Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-Institute, Jena, Germany.
| | - Oana Joean
- Institute for Infectious Disease and Infection Control, Jena University Hospital, Friedrich-Schiller-University, Am Klinikum 1, 07749, Jena, Germany
| | - Mathias W Pletz
- Institute for Infectious Disease and Infection Control, Jena University Hospital, Friedrich-Schiller-University, Am Klinikum 1, 07749, Jena, Germany
| | - Stefan Hagel
- Institute for Infectious Disease and Infection Control, Jena University Hospital, Friedrich-Schiller-University, Am Klinikum 1, 07749, Jena, Germany
| | - Sebastian Weis
- Institute for Infectious Disease and Infection Control, Jena University Hospital, Friedrich-Schiller-University, Am Klinikum 1, 07749, Jena, Germany
- Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-Institute, Jena, Germany
- Department of Anaesthesiology and Intensive Care Medicine, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany
| |
Collapse
|
11
|
Pattison NA, O’Gara G, Cuthbertson BH, Rose L. The legacy of the COVID-19 pandemic on critical care research: A descriptive interview study. J Intensive Care Soc 2025; 26:53-60. [PMID: 39660273 PMCID: PMC11626551 DOI: 10.1177/17511437241301921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2024] Open
Abstract
Background The COVID-19 pandemic challenged both research and clinical teams in critical care to collaborate on research solutions to new clinical problems. Although an effective, nationally coordinated response helped facilitate critical care research, reprioritisation of research efforts towards COVID-19 studies had significant consequences for existing and planned research activity in critical care. Aims Our aim was to explore the impact of the COVID-19 pandemic research prioritisation policies and practices on critical care research funded prior to the pandemic, the conduct of pandemic research, and implications for ongoing and future critical care research. Methods We undertook a descriptive qualitative study recruiting research-active clinician researchers and research delivery team members working in critical care. We conducted digitally recorded, semi-structured interviews in 2021-2022. Framework Analysis was used to analyse the data. Results We interviewed 22 participants comprising principal investigators, senior trial coordinators and research delivery nurses from across the UK. Six themes were identified: Unit, organisational and national factors; Study specific factors; Resources; Individual/clinician factors; Family/patient factors; Contextual factors. These themes explained how a nationally coordinated response during the pandemic affected individuals, studies and wider organisations in managing the research response in critical care, highlighting future implications for critical care research. Conclusion Harnessing the collective response seen in the COVID-19 pandemic in critical care could better support integration of research activity into routine critical care activities. Future endeavours should focus on workforce preparations, contingency planning, strategies for study prioritisation and integration of research as part of the continuum of clinical care.
Collapse
Affiliation(s)
- Natalie A. Pattison
- School of Health and Social Work, University of Hertfordshire, Hatfield, Hertfordshire, UK
- East and North Herts NHS Trust, Stevenage, Hertfordshire, UK
- Imperial College London, Imperial College Healthcare NHS Trust, London, UK
| | - Geraldine O’Gara
- Kings College London, London, UK
- The Royal Marsden NHS Foundation Trust, London, UK
| | - Brian H Cuthbertson
- Department of Critical Care Medicine, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Anesthesiology and Pain Medicine, University of Toronto. Toronto, ON, Canada
| | - Louise Rose
- Florence Nightingale Faculty of Nursing, Midwifery and Palliative Care, King’s College London, London, UK
| |
Collapse
|
12
|
Martins PN, Lourenço MHT, Mota GPS, Cavalcanti AB, Peçanha Antonio AC, Diaz-Quijano FA. Composite endpoints in COVID-19 randomized controlled trials: a systematic review. Clin Trials 2025; 22:77-87. [PMID: 39390767 DOI: 10.1177/17407745241276130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
BACKGROUND/AIMS This study aimed to determine the prevalence of ordinal, binary, and numerical composite endpoints among coronavirus disease 2019 trials and the potential bias attributable to their use. METHODS We systematically reviewed the Cochrane COVID-19 Study Register to assess the prevalence, characteristics, and bias associated with using composite endpoints in coronavirus disease 2019 randomized clinical trials. We compared the effect measure (relative risk) of composite outcomes and that of its most critical component (i.e. death) by estimating the Bias Attributable to Composite Outcomes index [ln(relative risk for the composite outcome)/ln(relative risk for death)]. RESULTS Composite endpoints accounted for 152 out of 417 primary endpoints in coronavirus disease 2019 randomized trials, being more frequent among studies published in high-impact journals. Ordinal endpoints were the most common (54% of all composites), followed by binary or time-to-event (34%), numerical (11%), and hierarchical (1%). Composites predominated among trials enrolling patients with severe disease when compared to trials with a mild or moderate case mix (odds ratio = 1.72). Adaptations of the seven-point World Health Organization scale occurred in 40% of the ordinal primary endpoints, which frequently underwent dichotomization for the statistical analyses. Mortality accounted for a median of 24% (interquartile range: 6%-48%) of all events when included in the composite. The median point estimate of the Bias Attributable to Composite Outcomes index was 0.3 (interquartile range: -0.1 to 0.7), being significantly lower than 1 in 5 of 24 comparisons. DISCUSSION Composite endpoints were used in a significant proportion of coronavirus disease 2019 trials, especially those involving severely ill patients. This is likely due to the higher anticipated rates of competing events, such as death, in such studies. Ordinal composites were common but often not fully appreciated, reducing the potential gains in information and statistical efficiency. For studies with binary composites, death was the most frequent component, and, unexpectedly, composite outcome estimates were often closer to the null when compared to those for mortality death. Numerical composites were less common, and only two trials used hierarchical endpoints. These newer approaches may offer advantages over traditional binary and ordinal composites; however, their potential benefits warrant further scrutiny. CONCLUSION Composite endpoints accounted for more than a third of coronavirus disease 2019 trials' primary endpoints; their use was more common among studies that included patients with severe disease and their point effect estimates tended to underestimate those for mortality.
Collapse
Affiliation(s)
- Pedro Nascimento Martins
- Federal University of Juiz de Fora, Juiz de Fora, Brazil
- Department of Epidemiology, School of Public Health, University of São Paulo, São Paulo, Brazil
| | | | | | | | | | | |
Collapse
|
13
|
Glasbey J, Webb SA, Peel T, Pinkney TD, Myles PS. Global collaboration between platform trials in surgery and anaesthesia. Br J Anaesth 2025; 134:259-262. [PMID: 39706702 DOI: 10.1016/j.bja.2024.09.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 12/23/2024] Open
Abstract
Large, randomised trials are the bedrock of evidence-based medicine, but the resources required to complete such trials greatly limit the number of important clinical questions that can be addressed within a reasonable period of time. Adaptive platform trials can identify effective, ineffective, or harmful treatments faster. These trials have been shown to deliver rapid evidence through the COVID-19 pandemic and are now being adopted across surgery and anaesthesia, with many opportunities for surgeons, anaesthetists, and other perioperative physicians to conduct and collaborate in platform trials.
Collapse
Affiliation(s)
- James Glasbey
- Academic Department of Surgery, University of Birmingham, Birmingham, UK.
| | - Steve A Webb
- Australian and New Zealand Intensive Care Research Centre, School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia; St John of God Healthcare, Melbourne, VIC, Australia
| | - Trisha Peel
- Department of Infectious Diseases, Monash University, Melbourne, VIC, Australia; Department of Infectious Diseases, Alfred Hospital, Melbourne, VIC, Australia
| | - Thomas D Pinkney
- Academic Department of Surgery, University of Birmingham, Birmingham, UK; University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Paul S Myles
- Department of Anaesthesiology and Perioperative Medicine, Monash University, Melbourne, VIC, Australia; Department of Anaesthesiology and Perioperative Medicine, Alfred Hospital, Melbourne, VIC, Australia.
| |
Collapse
|
14
|
Lee KJ, Middleton M, Mahar RK. Review article: Primer for clinical researchers on innovative trial designs for emergency medicine. Emerg Med Australas 2025; 37:e14532. [PMID: 39552367 PMCID: PMC11744427 DOI: 10.1111/1742-6723.14532] [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: 09/03/2024] [Accepted: 10/27/2024] [Indexed: 11/19/2024]
Abstract
Randomised trials have long been recognised as the gold standard research tool for evidence-based medicine. The past decade has seen the emergence of several innovative trial designs that are revolutionising how trials are conducted. These innovative designs enable more efficient, pragmatic trials that can address complex research questions which were previously not possible. In this paper, we provide an overview of the key innovative designs that are likely to be useful in the emergency medicine context, namely cluster crossover and stepped wedge designs, sequential multiple assignment randomised trial (SMART) designs, and platform trials. We describe the main features of each design, outline their pros and cons, and describe when they may or may not be useful. We also provide examples of these innovative designs in contexts that are relevant to emergency medicine.
Collapse
Affiliation(s)
- Katherine J Lee
- Clinical Epidemiology and Biostatistics UnitMurdoch Children's Research InstituteMelbourneVictoriaAustralia
- Department of PaediatricsUniversity of MelbourneMelbourneVictoriaAustralia
| | - Melissa Middleton
- Clinical Epidemiology and Biostatistics UnitMurdoch Children's Research InstituteMelbourneVictoriaAustralia
| | - Robert K Mahar
- Clinical Epidemiology and Biostatistics UnitMurdoch Children's Research InstituteMelbourneVictoriaAustralia
- Biostatistics Unit, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, Faculty of Medicine, Dentistry, and Health SciencesUniversity of MelbourneMelbourneVictoriaAustralia
- Methods and Implementation Support for Clinical and Health Sciences Research HubUniversity of MelbourneMelbourneVictoriaAustralia
| |
Collapse
|
15
|
Manley BJ, McKinlay CJD, Lee KJ, Groom KM, Whitehead CL. Adapt to survive and thrive: the time is now for adaptive platform trials for preterm birth. THE LANCET. CHILD & ADOLESCENT HEALTH 2025; 9:131-137. [PMID: 39855752 DOI: 10.1016/s2352-4642(24)00328-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 01/27/2025]
Abstract
In this Viewpoint, we discuss the challenges facing perinatal clinical researchers, many of which are unique to this field, and how traditional two-arm randomised trials using frequentist analysis might no longer be fit for purpose for perinatology. We propose a solution: the adoption of adaptive platform trials (APTs) with Bayesian methodology to address perinatal research questions to improve outcomes of preterm birth. APTs use a master protocol as a foundation to efficiently assess multiple interventions simultaneously for a particular disease. APTs can study these interventions in a perpetual manner, with interventions allowed to enter or leave the platform on the basis of preplanned decision algorithms. In this Viewpoint, we outline the ways in which APTs can overcome some of the issues facing perinatal clinical research, and the challenges and essential requirements for the design and implementation of perinatal APTs that should be considered.
Collapse
Affiliation(s)
- Brett J Manley
- Newborn Research, Melbourne, VIC, Australia; Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne, Melbourne, VIC, Australia; Murdoch Children's Research Institute, Melbourne, VIC, Australia.
| | - Christopher J D McKinlay
- Department of Paediatrics: Child and Youth Health, University of Auckland, Auckland, Aotearoa New Zealand
| | - Katherine J Lee
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia; Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | - Katie M Groom
- Liggins Institute, University of Auckland and Te Whatu Ora Te Toka Tumai Auckland, Auckland, Aotearoa New Zealand
| | - Clare L Whitehead
- The Royal Women's Hospital, Melbourne, VIC, Australia; Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne, Melbourne, VIC, Australia; Murdoch Children's Research Institute, Melbourne, VIC, Australia
| |
Collapse
|
16
|
Meyer EL, Mielke T, Bofill Roig M, Freitag MM, Jacko P, Krotka P, Mesenbrink P, Parke T, Zehetmayer S, Zocholl D, König F. Why and how should we simulate platform trials? Learnings from EU-PEARL. BMC Med Res Methodol 2025; 25:12. [PMID: 39819305 PMCID: PMC11740366 DOI: 10.1186/s12874-024-02453-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 12/20/2024] [Indexed: 01/19/2025] Open
Abstract
BACKGROUND Platform trials are innovative clinical trials governed by a master protocol that allows for the evaluation of multiple investigational treatments that enter and leave the trial over time. Interest in platform trials has been steadily increasing over the last decade. Due to their highly adaptive nature, platform trials provide sufficient flexibility to customize important trial design aspects to the requirements of both the specific disease under investigation and the different stakeholders. The flexibility of platform trials, however, comes with complexities when designing such trials. In the past, we reviewed existing software for simulating clinical trials and found that none of them were suitable for simulating platform trials as they do not accommodate the design features and flexibility inherent to platform trials, such as staggered entry of treatments over time. RESULTS We argued that simulation studies are crucial for the design of efficient platform trials. We developed and proposed an iterative, simulation-guided "vanilla and sprinkles" framework, i.e. from a basic to a more complex design, for designing platform trials. We addressed the functionality limitations of existing software as well as the unavailability of the coding therein by developing a suite of open-source software to use in simulating platform trials based on the R programming language. To give some examples, the newly developed software supports simulating staggered entry of treatments throughout the trial, choosing different options for control data sharing, specifying different platform stopping rules and platform-level operating characteristics. The software we developed is available through open-source licensing to enable users to access and modify the code. The separate use of two of these software packages to implement the same platform design by independent teams obtained the same results. CONCLUSION We provide a framework, as well as open-source software for the design and simulation of platform trials. The software tools provide the flexibility necessary to capture the complexity of platform trials.
Collapse
Affiliation(s)
- Elias Laurin Meyer
- Center for Medical Data Science, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Austria
- Berry Consultants, Vienna, Austria
| | | | - Marta Bofill Roig
- Center for Medical Data Science, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Austria
| | - Michaela Maria Freitag
- Institute of Biometry and Clinical Epidemiology, Charité - Universitätsmedizin, Berlin, Germany
| | - Peter Jacko
- Berry Consultants, Abingdon, UK
- Lancaster University, Lancaster, UK
| | - Pavla Krotka
- Center for Medical Data Science, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Austria
| | | | | | - Sonja Zehetmayer
- Center for Medical Data Science, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Austria
| | - Dario Zocholl
- Institute of Biometry and Clinical Epidemiology, Charité - Universitätsmedizin, Berlin, Germany
| | - Franz König
- Center for Medical Data Science, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Austria.
| |
Collapse
|
17
|
Su L, Chen X, Zhang J, Yan F. MIDAS-2: an enhanced Bayesian platform design for immunotherapy combinations with subgroup efficacy exploration. J Biopharm Stat 2025; 35:37-57. [PMID: 38131109 DOI: 10.1080/10543406.2023.2292211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 12/02/2023] [Indexed: 12/23/2023]
Abstract
Although immunotherapy combinations have revolutionised cancer treatment, the rapid screening of effective and optimal therapies from large numbers of candidate combinations, as well as exploring subgroup efficacy, remains challenging. This necessitates innovative, integrated, and efficient trial designs. In this study, we extend the MIDAS design to include subgroup exploration and propose an enhanced Bayesian information borrowing platform design called MIDAS-2. MIDAS-2 enables quick and continuous screening of promising combination strategies and exploration of their subgroup effects within a unified platform design framework. We use a regression model to characterize the efficacy pattern in subgroups. Information borrowing is applied through Bayesian hierarchical modelling to improve trial efficiency considering the limited sample size in subgroups. Time trend calibration is also employed to avoid potential baseline drifts. Simulation results demonstrate that MIDAS-2 yields high probabilities for identifying the effective drug combinations as well as promising subgroups, facilitating appropriate selection of the best treatments for each subgroup. The proposed design is robust against small time trend drifts, and the type I error is successfully controlled after calibration when a large drift is expected. Overall, MIDAS-2 provides an adaptive drug screening and subgroup exploring framework to accelerate immunotherapy development in an efficient, accurate, and integrated fashion.
Collapse
Affiliation(s)
- Liwen Su
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xin Chen
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jingyi Zhang
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Fangrong Yan
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
18
|
Lorenzi E, Crawford AM, Anderson CS, Menon B, Chen X, Mistry E, Khatri P, Elm JJ, Beall J, Saville BR, Berry SM, Lewis RJ. Adaptive Platform Trials in Stroke. Stroke 2025; 56:198-208. [PMID: 39705391 DOI: 10.1161/strokeaha.124.045754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2024]
Abstract
Clinical trials of treatments for stroke have generally utilized 2-arm, randomized designs to evaluate a single intervention against a control. Running separate clinical trials, with each addressing a single therapeutic question, is resource intensive and slows evidence generation, especially in a field with rapidly expanding treatment options and evolving practices. Platform trials-randomized clinical trials designed to evaluate multiple interventions that may enter and exit the ongoing platform based on a master protocol-accelerate the investigation of multiple therapeutic options within a single infrastructure. This in turn has the potential to accelerate access to new interventions for patients with stroke that can save lives and improve outcomes. In the context of acute ischemic stroke, 2 new platform trials have been established, the STEP trial (StrokeNet Thrombectomy Endovascular Platform) and ACT-GLOBAL (A Multi-Factorial, Multi-Arm, Multi-Stage, Randomised, Global Adaptive Platform Trial for Stroke), to address multiple therapeutic questions simultaneously using a multifactorial design including Bayesian modeling and other adaptive features. These trials are designed to maximize the information obtained from each participant, to align clinical research more closely with the complexities of clinical care, and to accelerate the identification of effective therapies. This article explores conceptual, practical, and statistical considerations in the design and implementation of adaptive platform trials and highlights their potential to accelerate the identification of new therapies, management, and rehabilitation in stroke.
Collapse
Affiliation(s)
| | - Amy M Crawford
- Berry Consultants LLC, Austin, TX (E.L., A.M.C., S.M.B., R.J.L.)
| | - Craig S Anderson
- George Institute for Global Health, University of New South Wales, Sydney, Australia (C.S.A., X.C.)
- Institute for Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China (C.S.A.)
| | - Bijoy Menon
- Department of Clinical Neurosciences, Radiology and Community Health Sciences, Cumming School of Medicine, University of Calgary, Alberta, Canada (B.M.)
| | - Xiaoying Chen
- George Institute for Global Health, University of New South Wales, Sydney, Australia (C.S.A., X.C.)
| | - Eva Mistry
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (E.M., P.K.)
| | - Pooja Khatri
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (E.M., P.K.)
| | - Jordan J Elm
- Department of Public Health Sciences, Medical University of South Carolina, Charleston (J.J.E., J.B.)
| | - Jonathan Beall
- Department of Public Health Sciences, Medical University of South Carolina, Charleston (J.J.E., J.B.)
| | - Benjamin R Saville
- Adaptix Trials LLC, Austin, TX (B.R.S.)
- Department of Biostatistics, Vanderbilt University School of Medicine, Nashville, TN (B.R.S.)
| | - Scott M Berry
- Berry Consultants LLC, Austin, TX (E.L., A.M.C., S.M.B., R.J.L.)
| | - Roger J Lewis
- Berry Consultants LLC, Austin, TX (E.L., A.M.C., S.M.B., R.J.L.)
- Department of Emergency Medicine, David Geffen School of Medicine at University of California Los Angeles (UCLA) (R.J.L.)
| |
Collapse
|
19
|
van Steenwijk MPJ, van Rosmalen J, Elzo Kraemer CV, Donker DW, Hermens JAJM, Kraaijeveld AO, Maas JJ, Akin S, Montenij LJ, Vlaar APJ, van den Bergh WM, Oude Lansink-Hartgring A, de Metz J, Voesten N, Boersma E, Scholten E, Beishuizen A, Lexis CPH, Peperstraete H, Schiettekatte S, Lorusso R, Gommers DAMPJ, Tibboel D, de Boer RA, Van Mieghem NMDA, Meuwese CL. A randomized embedded multifactorial adaptive platform for extra corporeal membrane oxygenation (REMAP ECMO) - design and rationale of the left ventricular unloading trial domain. Am Heart J 2025; 279:81-93. [PMID: 39447716 DOI: 10.1016/j.ahj.2024.10.010] [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: 05/29/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND The use of Extracorporeal Membrane Oxygenation (ECMO) remains associated with high rates of complications, weaning failure and mortality which can be partly explained by a knowledge gap on how to properly manage patients on ECMO support. To address relevant patient management issues, we designed a "Randomized Embedded Multifactorial Adaptive Platform (REMAP)" in the setting of ECMO (REMAP ECMO) and a first embedded randomized controlled trial (RCT) investigating the effects of routine early left ventricular (LV) unloading through intra-aortic balloon pumping (IABP). METHODS REMAP ECMO describes a registry-based platform allowing for the embedding of multiple response adaptive RCTs (trial domains) which can perpetually address the effect of relevant patient management issues on ECMO weaning success. A first trial domain studies the effects of LV unloading by means of an IABP as an adjunct to veno-arterial (V-A) ECMO versus V-A ECMO alone on ECMO weaning success at 30 days in adult cardiogenic shock patients admitted to the Intensive Care Unit (ICU). The primary outcome of this trial is "successful weaning from ECMO" being defined as a composite of survival without the need for mechanical circulatory support, heart transplantation, or left ventricular assist device (LVAD) at 30 days after initiation of ECMO. Secondary outcomes include the need for interventional escalation of LV unloading strategy, mechanistic endpoints, survival characteristics until 1 year after ECMO initiation, and quality of life. Trial data will be analysed using a Bayesian statistical framework. The adaptive design allows for a high degree of flexibility, such as response adaptive randomization and early stopping of the trial for efficacy or futility. The REMAP ECMO LV unloading study is approved by the Medical Ethical Committee of the Erasmus Medical Center and is publicly registered. CONCLUSION This REMAP ECMO trial platform enables the efficient roll-out of multiple RCTs on relevant patient management issues. A first embedded trial domain will compare routine LV unloading by means of an IABP as an adjunct to V-A ECMO versus V-A ECMO alone. TRIAL REGISTRATION ClinicalTrials.gov, NCT05913622.
Collapse
Affiliation(s)
- Myrthe P J van Steenwijk
- Department of Intensive Care, Erasmus Medical Center, Rotterdam, the Netherlands; Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Joost van Rosmalen
- Departments of Biostatistics, Erasmus Medical Center, Rotterdam, the Netherlands; Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Carlos V Elzo Kraemer
- Department of Intensive Care, Leiden University Medical Center, Leiden, the Netherlands
| | - Dirk W Donker
- Department of Intensive Care, University Medical Center Utrecht, Utrecht, the Netherlands; Cardiovascular and Respiratory Physiology, University of Twente, Enschede, the Netherlands
| | - Jeannine A J M Hermens
- Department of Intensive Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Adriaan O Kraaijeveld
- Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jacinta J Maas
- Department of Intensive Care, Leiden University Medical Center, Leiden, the Netherlands
| | - Sakir Akin
- Department of Intensive Care, Haga Hospital, The Hague, the Netherlands
| | - Leon J Montenij
- Department of Intensive Care, Catharina Hospital Eindhoven, Eindhoven, the Netherlands
| | - Alexander P J Vlaar
- Department of Intensive Care, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Walter M van den Bergh
- Department of Critical Care, University Medical Center Groningen, Groningen, the Netherlands
| | | | - Jesse de Metz
- Department of Intensive Care, OLVG Amsterdam, Amsterdam, the Netherlands
| | - Niek Voesten
- Department of Intensive Care, Amphia Hospital Breda, Breda, the Netherlands
| | - Eric Boersma
- Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Erik Scholten
- Department of Intensive Care, Sint Antonius Hospital, Nieuwegein, the Netherlands
| | - Albertus Beishuizen
- Department of Intensive Care, Medisch Spectrum Twente, Enschede, the Netherlands
| | - Chris P H Lexis
- Department of Intensive Care and Cardiology, Maastricht UMC, Maastricht, the Netherlands
| | | | | | - Roberto Lorusso
- Department of Cardiothoracic Surgery and Cardiovascular Research Center, Maastricht UMC, Maastricht, the Netherlands
| | | | - Dick Tibboel
- Department of Intensive Care, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Rudolf A de Boer
- Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Nicolas M D A Van Mieghem
- Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Christiaan L Meuwese
- Department of Intensive Care, Erasmus Medical Center, Rotterdam, the Netherlands; Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
20
|
Krishnasamy R, Jardine MJ. Adaptive Designs for Clinical Trials in Nephrology. J Am Soc Nephrol 2025; 36:147-149. [PMID: 39186385 PMCID: PMC11706552 DOI: 10.1681/asn.0000000000000497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Collaborators] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 08/20/2024] [Indexed: 08/28/2024] Open
Affiliation(s)
- Rathika Krishnasamy
- Department of Renal Medicine, Sunshine Coast University Hospital, Birtinya, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
- NHMRC Clinical Trials Centre, The University of Sydney, Sydney, Australia
| | - Meg J. Jardine
- NHMRC Clinical Trials Centre, The University of Sydney, Sydney, Australia
- Department of Renal Medicine, Concord Repatriation General Hospital, Sydney, Australia
| |
Collapse
Collaborators
Smeeta Sinha, Grahame Elder, Carmel Hawley, Nigel Toussaint, Arlen Wilcox, Tamara Young, Elizabeth Lorenzi, Lindsay Berry, Scott Berry, Brendan J Smyth, Laurent Billot, Jonathan Craig, David Johnson, Stephen McDonald, Janak de Zoysa, David Collister, Soo Kun Lin, Angus Ritchie, Michael Collins, Irene Ruderman,
Collapse
|
21
|
Zhai G, Li Y, Zhang L, Hu F. Group Response-Adaptive Randomization With Delayed and Missing Responses. Stat Med 2024; 43:5047-5059. [PMID: 39285137 DOI: 10.1002/sim.10220] [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: 10/08/2023] [Revised: 06/16/2024] [Accepted: 08/27/2024] [Indexed: 11/24/2024]
Abstract
Response-adaptive randomization (RAR) procedures have been extensively studied in the literature, but most of the procedures rely on updating the randomization after each response, which is impractical in many clinical trials. In this article, we propose a new family of RAR procedures that dynamically update based on the responses of a group of individuals, either when available or at fixed time intervals (weekly or biweekly). We show that the proposed design retains the essential theoretical properties of Hu and Zhang's doubly adaptive biased coin designs (DBCD), and performs well in scenarios involving delayed and randomly missing responses. Numerical studies have been conducted to demonstrate that the new proposed group doubly adaptive biased coin design has similar properties to the Hu and Zhang's DBCDs in different situations. We also apply the new design to a real clinical trial, highlighting its advantages and practicality. Our findings open the door to studying the properties of other group response adaptive designs, such as urn models, and facilitate the application of response-adaptive randomized clinical trials in practice.
Collapse
Affiliation(s)
- Guannan Zhai
- Department of Statistics, George Washington University, Washington, DC
| | - Yang Li
- Center for Applied Statistics, School of Statistics, Renmin University of China, Beijing, People's Republic of China
| | - Lixin Zhang
- School of Statistics and Mathematics, Zhejiang Gongshang University, Zhejiang, People's Republic of China
- School of Mathematical Sciences, Zhejiang University, Zhejiang, People's Republic of China
| | - Feifang Hu
- Department of Statistics, George Washington University, Washington, DC
| |
Collapse
|
22
|
Loo WTW, Tang KJY, Chew SY. Can a Bayesian approach clarify if corticosteroids are beneficial for severe community-acquired pneumonia? ANNALS OF THE ACADEMY OF MEDICINE, SINGAPORE 2024; 53:644-646. [PMID: 39636190 DOI: 10.47102/annals-acadmedsg.2024335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Despite advances in the antimicrobial treatment of sepsis and organ support in the intensive care unit (ICU), community-acquired pneumonia (CAP) remains a leading cause of mortality and disability-adjusted life years lost globally.1 Severe CAP, where CAP becomes complicated by acute hypoxaemic respiratory failure or shock, is also the most common cause of sepsis, where complex and heterogeneous biological mechanisms underlie a dysregulated inflammatory host response that ultimately leads to major organ dysfunction and death. Along with the emerging threats of respiratory pandemics, antimicrobial resistance, ageing populations and the rise of chronic diseases, much research has been conducted to improve the treatment outcomes of CAP via host immunomodulation. These efforts have focused almost exclusively on anti-inflammatory effects of corticosteroids, which have a proven track record of improving outcomes in other forms of sepsis, such as bacterial meningitis, Pneumocystis jirovecii pneumonia and severe COVID-19.
Collapse
Affiliation(s)
- Wesley Teck Wee Loo
- Department of Respiratory & Critical Care Medicine, Singapore General Hospital, Singapore
| | - Kendra Jing Ying Tang
- Department of Respiratory & Critical Care Medicine, Singapore General Hospital, Singapore
| | - Si Yuan Chew
- Department of Respiratory & Critical Care Medicine, Singapore General Hospital, Singapore
| |
Collapse
|
23
|
Lewis NM, Harker EJ, Grant LB, Zhu Y, Grijalva CG, Chappell JD, Rhoads JP, Baughman A, Casey JD, Blair PW, Jones ID, Johnson CA, Lauring AS, Gaglani M, Ghamande S, Columbus C, Steingrub JS, Shapiro NI, Duggal A, Busse LW, Felzer J, Prekker ME, Peltan ID, Brown SM, Hager DN, Gong MN, Mohamed A, Exline MC, Khan A, Hough CL, Wilson JG, Mosier J, Qadir N, Chang SY, Ginde AA, Martinez A, Mohr NM, Mallow C, Harris ES, Johnson NJ, Srinivasan V, Gibbs KW, Kwon JH, Vaughn IA, Ramesh M, Safdar B, Goyal A, DeLamielleure LE, DeCuir J, Surie D, Dawood FS, Tenforde MW, Uyeki TM, Garg S, Ellington S, Self WH. Benefit of early oseltamivir therapy for adults hospitalized with influenza A: an observational study. Clin Infect Dis 2024:ciae584. [PMID: 39607747 DOI: 10.1093/cid/ciae584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/07/2024] [Accepted: 11/22/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND clinical guidelines recommend initiation of antiviral therapy as soon as possible for patients hospitalized with confirmed or suspected influenza. METHODS A multicenter US observational sentinel surveillance network prospectively enrolled adults (aged ≥18 years) hospitalized with laboratory-confirmed influenza at 24 hospitals during October 1, 2022-July 21, 2023. A multivariable proportional odds model was used to compare peak pulmonary disease severity (no oxygen support, standard supplemental oxygen, high-flow oxygen/non-invasive ventilation, invasive mechanical ventilation, or death) after the day of hospital admission among patients starting oseltamivir treatment on the day of admission (early) versus those who did not (late or not treated), adjusting for baseline (admission day) severity, age, sex, site, and vaccination status. Multivariable logistic regression models were used to evaluate the odds of intensive care unit (ICU) admission, acute kidney replacement therapy or vasopressor use, and in-hospital death. RESULTS A total of 840 influenza-positive patients were analyzed, including 415 (49%) who started oseltamivir treatment on the day of admission, and 425 (51%) who did not. Compared with late or not treated patients, those treated early had lower peak pulmonary disease severity (proportional aOR: 0.60, 95% CI: 0.49-0.72), and lower odds of intensive care unit admission (aOR: 0.24, 95% CI: 0.13-0.47), acute kidney replacement therapy or vasopressor use (aOR: 0.40, 95% CI: 0.22-0.67), and in-hospital death (aOR: 0.36, 95% CI: 0.18-0.72). CONCLUSION Among adults hospitalized with influenza, treatment with oseltamivir on day of hospital admission was associated reduced risk of disease progression, including pulmonary and extrapulmonary organ failure and death.
Collapse
Affiliation(s)
- Nathaniel M Lewis
- Influenza Division, National Center for Immunization and Respiratory Diseases, CDC
| | - Elizabeth J Harker
- Influenza Division, National Center for Immunization and Respiratory Diseases, CDC
| | - Lauren B Grant
- Influenza Division, National Center for Immunization and Respiratory Diseases, CDC
| | - Yuwei Zhu
- Vanderbilt University Medical Center, Nashville, Tennessee
| | | | | | | | | | | | - Paul W Blair
- Vanderbilt University Medical Center, Nashville, Tennessee
| | - Ian D Jones
- Vanderbilt University Medical Center, Nashville, Tennessee
| | | | | | - Manju Gaglani
- Baylor Scott & White Health, Texas
- Baylor College of Medicine, Temple, Texas
| | | | - Cristie Columbus
- Baylor College of Medicine, Temple, Texas
- Texas A&M University College of Medicine, Dallas, Texas
| | | | | | | | | | | | | | - Ithan D Peltan
- University of Utah, Salt Lake City, Utah
- Intermountain Medical Center, Murray, Utah; University of Utah, Salt Lake City, Utah
| | - Samuel M Brown
- University of Utah, Salt Lake City, Utah
- Intermountain Medical Center, Murray, Utah; University of Utah, Salt Lake City, Utah
| | - David N Hager
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michelle N Gong
- Montefiore Medical Center, Albert Einstein College of Medicine, New York, New York
| | - Amira Mohamed
- Montefiore Medical Center, Albert Einstein College of Medicine, New York, New York
| | | | - Akram Khan
- Oregon Health & Science University, Portland, Oregon
| | | | | | | | - Nida Qadir
- Ronald Reagan UCLA Medical Center, Los Angeles, California
| | - Steven Y Chang
- Ronald Reagan UCLA Medical Center, Los Angeles, California
| | - Adit A Ginde
- University of Colorado School of Medicine, Aurora, Colorado
| | | | | | | | | | | | | | - Kevin W Gibbs
- Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Jennie H Kwon
- Washington University in St. Louis, St. Louis, Missouri
| | | | | | - Basmah Safdar
- Yale University School of Medicine, New Haven, Connecticut
| | - Anirudh Goyal
- Yale University School of Medicine, New Haven, Connecticut
| | | | - Jennifer DeCuir
- Division of COVID-19 and Other Respiratory Viral Diseases, CDC
| | - Diya Surie
- Division of COVID-19 and Other Respiratory Viral Diseases, CDC
| | | | - Mark W Tenforde
- Influenza Division, National Center for Immunization and Respiratory Diseases, CDC
| | - Timothy M Uyeki
- Influenza Division, National Center for Immunization and Respiratory Diseases, CDC
| | - Shikha Garg
- Influenza Division, National Center for Immunization and Respiratory Diseases, CDC
| | - Sascha Ellington
- Influenza Division, National Center for Immunization and Respiratory Diseases, CDC
| | - Wesley H Self
- Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
24
|
Glasbey J, Webb SA, Peel T, Pinkney TD, Myles PS. Global collaboration between platform trials in surgery and anaesthesia. Br J Surg 2024; 111:znae262. [PMID: 39701591 PMCID: PMC11658830 DOI: 10.1093/bjs/znae262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
Large, randomized trials are the bedrock of evidence-based medicine, but the resources required to complete such trials greatly limit the number of important clinical questions that can be addressed within a reasonable period of time. Adaptive platform trials can identify effective, ineffective, or harmful treatments faster. These trials have been shown to deliver rapid evidence through the COVID-19 pandemic and are now being adopted across surgery and anaesthesia, with many opportunities for surgeons, anaesthetists, and other perioperative physicians to conduct and collaborate in platform trials.
Collapse
Affiliation(s)
- James Glasbey
- Academic Department of Surgery, University of Birmingham, Birmingham, UK
| | - Steve A Webb
- Australian and New Zealand Intensive Care Research Centre, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- Director of Research, St John of God Healthcare, Melbourne, Victoria, Australia
| | - Trisha Peel
- Department of Infectious Diseases, Monash University, Melbourne, Victoria, Australia
- Department of Infectious Diseases, Alfred Hospital, Melbourne, Victoria, Australia
| | - Thomas D Pinkney
- Academic Department of Surgery, University of Birmingham, Birmingham, UK
- Department of General Surgery, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Paul S Myles
- Department of Anaesthesiology and Perioperative Medicine, Monash University, Melbourne, Victoria, Australia
- Department of Anaesthesiology and Perioperative Medicine, Alfred Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
25
|
Lyne T, Camporota L, Montgomery H. Contribution of intrapulmonary shunt to the pathogenesis of profound hypoxaemia in viral infection: a mechanistic discussion with an illustrative case. J Intensive Care Soc 2024; 25:427-431. [PMID: 39524067 PMCID: PMC11549710 DOI: 10.1177/17511437241267745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
Background The formation of anastomoses between the pulmonary arteries and pulmonary veins, or the pulmonary and the bronchial circulation, is part of normal foetal lung development. They persist in approximately 30% of adults at rest, and open in almost all adults during exertion. Blood flowing through these anastomoses bypasses the alveolar surface and increases in such shunting can thus cause hypoxaemia. This is now known to contribute to the pathogenesis of hypoxaemia in COVID-19 disease. We here provide evidence to support a similar role in influenza A infection. Illustrative case presentation We describe a case of influenza A infection associated with severe hypoxaemia, poorly responsive to supplemental oxygen and which worsened following the application of continuous positive airway pressure (CPAP), despite the presence of a normal physical examination, chest radiograph and echocardiogram. This combination suggests a significant intrapulmonary (extra-alveolar) shunt as a cause of the severe hypoxaemia. The shunt fraction was estimated to be approximately 57%. Discussion and conclusion Intrapulmonary vascular shunts can contribute substantially to hypoxaemia in viral infection. Seeking to understand the pathogenesis of observed hypoxaemia can help guide respiratory therapy. Mechanistic research may suggest novel therapeutic targets which could assist in avoiding intubation and mechanical ventilatory support.
Collapse
Affiliation(s)
- Tom Lyne
- Whittington Hospital Intensive Care Unit, Department of Intensive Care, London, UK
| | - Luigi Camporota
- Department of Adult Critical Care, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
- Centre for Human and Applied Physiological Sciences, King’s College London, London, UK
| | - Hugh Montgomery
- Whittington Hospital Intensive Care Unit, Department of Intensive Care, London, UK
- Department of Medicine, University College London, London, UK
| |
Collapse
|
26
|
Stewart AG, Simos P, Sivabalan P, Escolà-Vergé L, Garnham K, Isler B. An Update on Recent Clinical Trial Data in Bloodstream Infection. Antibiotics (Basel) 2024; 13:1035. [PMID: 39596730 PMCID: PMC11591257 DOI: 10.3390/antibiotics13111035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 10/23/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024] Open
Abstract
Bloodstream infections (BSIs) remain a significant source of morbidity and mortality globally, exacerbated by an ageing population and rising antimicrobial resistance (AMR). This review offers an updated evaluation of randomized clinical trials (RCTs) in BSI management from 2018 onwards, focusing on the evolving landscape of diagnostics and treatment. New rapid diagnostic technologies and shorter antimicrobial courses have transformed clinical practice, reducing the time to appropriate therapy and hospital stays. Several RCTs demonstrated that rapid phenotypic and genotypic tests shorten the time to optimal therapy, especially when paired with antimicrobial stewardship. Ongoing trials are investigating novel antimicrobial regimens and the safety of early oral switch strategies, particularly for Gram-positive and Gram-negative BSIs. Recent RCTs on Staphylococcus aureus BSI (SAB) and multidrug-resistant Gram-negative bacteria highlight advances in treatment but emphasize the need for further study into the efficacy of combination therapies and the utility of rapid diagnostics in different healthcare settings. The review also explores challenges in trail design, with adaptive and pragmatic appropriates improving the efficacy of clinical trials. Finally, this paper identifies gaps in the research, including the need for further investigation into oral step-down therapy, optimal durations, and the role of rapid diagnostics in resource-limited settings.
Collapse
Affiliation(s)
- Adam G. Stewart
- Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Royal Brisbane and Women’s Hospital Campus, Brisbane 4006, Australia
| | - Peter Simos
- Department of Infectious Diseases, Gold Coast University Hospital, Gold Coast 4215, Australia; (P.S.); (K.G.)
| | | | - Laura Escolà-Vergé
- Servicio de Enfermedades Infecciosas, Hospital Universitari Vall d’Hebron, Departamento de Medicina, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Katherine Garnham
- Department of Infectious Diseases, Gold Coast University Hospital, Gold Coast 4215, Australia; (P.S.); (K.G.)
- Pathology Queensland, Gold Coast Hospital Campus, Gold Coast 4215, Australia
| | - Burcu Isler
- Infection Management Services, Princess Alexandra Hospital, Brisbane 4102, Australia;
| |
Collapse
|
27
|
Thomas SM, Harrington RA, Yancy CW, Nugent D, Erzurum S, Bernard GR, Cushman M, Hochman JS, Ridker PM, Ortel TL, Collins SP, Callaway CW, Nolen TL, Womack KN, Brown SM, Gelijns A, Geraci M, Ginde AA, Key NS, Krishnan JA, LaVange L, Wisniewski SR, Berdan L, Punturieri A, Goff DC, Patterson AP. Lessons Learned from National Heart, Lung, and Blood Institute Covid-19 Clinical Trials. NEJM EVIDENCE 2024; 3:EVIDctcs2300291. [PMID: 39437132 DOI: 10.1056/evidctcs2300291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
AbstractIn response to the Covid-19 pandemic, the National Heart, Lung, and Blood Institute launched five multisite clinical trials testing candidate host tissue-directed medical interventions to hasten recovery, improve function, and reduce morbidity and mortality. Speed, flexibility, and collaboration were essential. This article from the Steering and Executive committees describes the Collaborating Network of Networks for Evaluating Covid-19 and Therapeutic Strategies (CONNECTS) research program that enrolled 6690 participants and evaluated 18 intervention strategies using 10 molecular agents across the care continuum (outpatient, inpatient, and post discharge), and reports lessons learned from this initiative. Successes include rapid trial execution through collaboration and adaptive platform designs. Challenges that impeded efficiency included time required to execute subcontracts, constraints on clinical research workforce, and limited research infrastructure in nonacademic settings.
Collapse
Affiliation(s)
| | | | - Clyde W Yancy
- Northwestern University Feinberg School of Medicine, Chicago
| | | | | | | | | | - Judith S Hochman
- New York University Grossman School of Medicine, NYU Langone Health, New York
| | | | | | - Sean P Collins
- Vanderbilt University Medical Center and Veterans Affairs Tennessee Valley Healthcare System, Geriatric Research, Education and Clinical Center (GRECC), Nashville
| | | | | | | | | | | | | | - Adit A Ginde
- University of Colorado School of Medicine, Aurora
| | - Nigel S Key
- University of North Carolina School of Medicine, Chapel Hill
| | | | - Lisa LaVange
- University of North Carolina Gillings School of Global Public Health, Chapel Hill
| | | | - Lisa Berdan
- National Heart, Lung, and Blood Institute (NHLBI), Bethesda, MD
| | | | - David C Goff
- National Heart, Lung, and Blood Institute (NHLBI), Bethesda, MD
| | - Amy P Patterson
- National Heart, Lung, and Blood Institute (NHLBI), Bethesda, MD
| |
Collapse
|
28
|
Hasan N, Mehrotra K, Danzig CJ, Eichenbaum DA, Ewald A, Regillo C, Momenaei B, Sheth VS, Lally DR, Chhablani J. Screen Failures in Clinical Trials in Retina. Ophthalmol Retina 2024; 8:1093-1099. [PMID: 38810882 DOI: 10.1016/j.oret.2024.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 05/31/2024]
Abstract
PURPOSE Disparities in clinical trials are a major problem because of significant underrepresentation of certain gender, racial, and ethnic groups. Several factors including stringent eligibility criteria and recruitment strategies hinder our understanding of retinal disease. Thus, we aimed to study the various reasons of screen failures and specific patient and study characteristics among screen failures. DESIGN This is a cross-sectional retrospective study. METHODS Screening data of 87 trials from 6 centers were analyzed. Study characteristics (disease studied, phase of trial, and route of drug administration) and patient demographics (age, gender, race, ethnicity, and employment status) were compared among different causes of screen failures. Screen failures were broadly classified into 6 categories: exclusion because of vision-based criteria, exclusion because of imaging findings, exclusion because of other factors, patient-related criteria, physician-related criteria, and miscellaneous. Descriptive statistics, Pearson chi-square test, and analysis of variance were used for statistical analysis. MAIN OUTCOME MEASURES Prevalence of various reasons for screen failures in multiple trials and its trend among different study and patient characteristics. RESULTS Among 87 trials and 962 patients, 465 (48.2%) patients were successfully randomized and 497 (51.8%) patients were classified as screen failures. The trials were conducted for various retinal diseases. Mean age was 76.50 ± 10.45 years and 59.4% were females. Predominantly White patients (93.4%) and unemployed/retired patients (66.6%) were screened. Of the 497 screen failures, most were because of patients not meeting inclusion criteria of imaging findings (n = 221 [44.5%]) followed by inclusion of vision-based criteria (n = 73 [14.7%]), exclusion because of other factors (n = 75 [15.1%]), patient-related (n = 34 [6.8%]), physician-related (n = 28 [5.6%]), and miscellaneous reasons (n = 39 [7.8%]). Reason for screen failure was not available for 27 (5.4%) patients. A higher proportion of patients screened for surgical trials (15%) declined to participate in the study compared with noninvasive trials involving topical drugs and photobiomodulation (0%) (P = 0.02). CONCLUSIONS Patients not meeting the imaging and vision-cased criteria were the most common reasons for screen failures. White patients and unemployed patients predominantly participated in clinical trials. Patients are more inclined to continue participation in noninvasive clinical trials compared with surgical trials. Better recruitment strategies and careful consideration of study criteria can aid in decreasing the rate of screen failures. FINANCIAL DISCLOSURE(S) Proprietary or commercial disclosure may be found after the references in the Footnotes and Disclosures at the end of this article.
Collapse
Affiliation(s)
- Nasiq Hasan
- Department of Ophthalmology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Kunaal Mehrotra
- Department of Ophthalmology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | | | - David A Eichenbaum
- Retina Vitreous Associates of Florida, Saint Petersburg, Florida; Department of Ophthalmology, Morsani College of Medicine at the University of South Florida, Tampa, Florida
| | - Amy Ewald
- Retina Vitreous Associates of Florida, Saint Petersburg, Florida; Department of Ophthalmology, Morsani College of Medicine at the University of South Florida, Tampa, Florida
| | - Carl Regillo
- Retina Service, Wills Eye Hospital, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Bita Momenaei
- Retina Service, Wills Eye Hospital, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Veeral S Sheth
- University Retina and Macula Associates PC, Oak Forest, Illinois
| | - David R Lally
- New England Retina Associates, Springfield, Massachusetts
| | - Jay Chhablani
- Department of Ophthalmology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania.
| |
Collapse
|
29
|
Hayward KS, Dalton EJ, Campbell BCV, Khatri P, Dukelow SP, Johns H, Walter S, Yogendrakumar V, Pandian JD, Sacco S, Bernhardt J, Parsons MW, Saver JL, Churilov L. Adaptive Trials in Stroke: Current Use and Future Directions. Neurology 2024; 103:e209876. [PMID: 39325999 PMCID: PMC11436319 DOI: 10.1212/wnl.0000000000209876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 07/22/2024] [Indexed: 09/28/2024] Open
Abstract
Inclusion of adaptive design features in a clinical trial provides preplanned flexibility to dynamically modify a trial during its conduct while preserving validity and integrity. Adaptive trials are needed to accelerate the conduct of more efficient, informative, and ethical clinical research in the field of neurology. Stroke is a natural candidate for adoption of these innovative approaches to trial design. This Research Methods in Neurology article is informed by a scoping review that identified 45 completed or ongoing adaptive clinical trials in stroke that were appraised: 15 trials had published results with or without a published protocol and 30 ongoing trials (14 trials had a published protocol, and 16 trials were registered only). Interventions spanned acute (n = 28), rehabilitation (n = 8), prevention (n = 8), and rehabilitation and prevention (n = 1). A subsample of these trials was selected to illustrate the utility of adaptive design features and discuss why each adaptive feature was incorporated in the design to best achieve the aim; whether each individual feature was used and whether it resulted in expected efficiencies; and any learnings during preparation, conduct, or reporting. We then discuss the operational, ethical, and regulatory considerations that warrant careful consideration during adaptive trial planning and reflect on the workforce readiness to deliver adaptive trials in practice. We conclude that adaptive trials can be designed, funded, conducted, and published for a wide range of research questions and offer future directions to support adoption of adaptive trial designs in stroke and neurologic research more broadly.
Collapse
Affiliation(s)
- Kathryn S Hayward
- From the Melbourne School of Health Sciences (K.S.H., E.J.D.), and Melbourne Medical School (K.S.H., H.J., L.C.), University of Melbourne, Parkville; Stroke Theme (K.S.H., B.C.V.C., J.B., L.C.), The Florey Institute, University of Melbourne, Heidelberg; National Health and Medical Research Council Centre of Research Excellence to Accelerate Stroke Trial Innovation and Translation (K.S.H., B.C.V.C., L.C., J.B., H.J.), University of Melbourne, Parkville; Department of Medicine and Neurology (B.C.V.C., V.Y.), Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia; Department of Neurology and Rehabilitation Medicine (P.K.), University of Cincinnati, OH; Department of Clinical Neuroscience (S.P.D.), and Hotchkiss Brain Institute (S.P.D.), University of Calgary, Alberta, Canada; Department of Neurology (S.W.), Saarland University, Saarbrücken; Department of Neurology (S.W.), Martin-Luther-University, Halle, Germany; Department of Neurology (V.Y.), University of Ottawa, Ontario, Canada; Department of Neurology (J.D.P.), Christian Medical College, Ludhiana, Punjab, India; Department of Biotechnological and Applied Clinical Sciences (S.S.), University of L'Aquila, Italy; Department of Neurology (M.W.P.), Liverpool Hospital, UNSW South Western Sydney Clinical School, Warwick Farm, Australia; Comprehensive Stroke Center and Department of Neurology (J.L.S.), University of California Los Angeles; and Australian Stroke Alliance (L.C.), University of Melbourne, Parkville, Victoria, Australia
| | - Emily J Dalton
- From the Melbourne School of Health Sciences (K.S.H., E.J.D.), and Melbourne Medical School (K.S.H., H.J., L.C.), University of Melbourne, Parkville; Stroke Theme (K.S.H., B.C.V.C., J.B., L.C.), The Florey Institute, University of Melbourne, Heidelberg; National Health and Medical Research Council Centre of Research Excellence to Accelerate Stroke Trial Innovation and Translation (K.S.H., B.C.V.C., L.C., J.B., H.J.), University of Melbourne, Parkville; Department of Medicine and Neurology (B.C.V.C., V.Y.), Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia; Department of Neurology and Rehabilitation Medicine (P.K.), University of Cincinnati, OH; Department of Clinical Neuroscience (S.P.D.), and Hotchkiss Brain Institute (S.P.D.), University of Calgary, Alberta, Canada; Department of Neurology (S.W.), Saarland University, Saarbrücken; Department of Neurology (S.W.), Martin-Luther-University, Halle, Germany; Department of Neurology (V.Y.), University of Ottawa, Ontario, Canada; Department of Neurology (J.D.P.), Christian Medical College, Ludhiana, Punjab, India; Department of Biotechnological and Applied Clinical Sciences (S.S.), University of L'Aquila, Italy; Department of Neurology (M.W.P.), Liverpool Hospital, UNSW South Western Sydney Clinical School, Warwick Farm, Australia; Comprehensive Stroke Center and Department of Neurology (J.L.S.), University of California Los Angeles; and Australian Stroke Alliance (L.C.), University of Melbourne, Parkville, Victoria, Australia
| | - Bruce C V Campbell
- From the Melbourne School of Health Sciences (K.S.H., E.J.D.), and Melbourne Medical School (K.S.H., H.J., L.C.), University of Melbourne, Parkville; Stroke Theme (K.S.H., B.C.V.C., J.B., L.C.), The Florey Institute, University of Melbourne, Heidelberg; National Health and Medical Research Council Centre of Research Excellence to Accelerate Stroke Trial Innovation and Translation (K.S.H., B.C.V.C., L.C., J.B., H.J.), University of Melbourne, Parkville; Department of Medicine and Neurology (B.C.V.C., V.Y.), Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia; Department of Neurology and Rehabilitation Medicine (P.K.), University of Cincinnati, OH; Department of Clinical Neuroscience (S.P.D.), and Hotchkiss Brain Institute (S.P.D.), University of Calgary, Alberta, Canada; Department of Neurology (S.W.), Saarland University, Saarbrücken; Department of Neurology (S.W.), Martin-Luther-University, Halle, Germany; Department of Neurology (V.Y.), University of Ottawa, Ontario, Canada; Department of Neurology (J.D.P.), Christian Medical College, Ludhiana, Punjab, India; Department of Biotechnological and Applied Clinical Sciences (S.S.), University of L'Aquila, Italy; Department of Neurology (M.W.P.), Liverpool Hospital, UNSW South Western Sydney Clinical School, Warwick Farm, Australia; Comprehensive Stroke Center and Department of Neurology (J.L.S.), University of California Los Angeles; and Australian Stroke Alliance (L.C.), University of Melbourne, Parkville, Victoria, Australia
| | - Pooja Khatri
- From the Melbourne School of Health Sciences (K.S.H., E.J.D.), and Melbourne Medical School (K.S.H., H.J., L.C.), University of Melbourne, Parkville; Stroke Theme (K.S.H., B.C.V.C., J.B., L.C.), The Florey Institute, University of Melbourne, Heidelberg; National Health and Medical Research Council Centre of Research Excellence to Accelerate Stroke Trial Innovation and Translation (K.S.H., B.C.V.C., L.C., J.B., H.J.), University of Melbourne, Parkville; Department of Medicine and Neurology (B.C.V.C., V.Y.), Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia; Department of Neurology and Rehabilitation Medicine (P.K.), University of Cincinnati, OH; Department of Clinical Neuroscience (S.P.D.), and Hotchkiss Brain Institute (S.P.D.), University of Calgary, Alberta, Canada; Department of Neurology (S.W.), Saarland University, Saarbrücken; Department of Neurology (S.W.), Martin-Luther-University, Halle, Germany; Department of Neurology (V.Y.), University of Ottawa, Ontario, Canada; Department of Neurology (J.D.P.), Christian Medical College, Ludhiana, Punjab, India; Department of Biotechnological and Applied Clinical Sciences (S.S.), University of L'Aquila, Italy; Department of Neurology (M.W.P.), Liverpool Hospital, UNSW South Western Sydney Clinical School, Warwick Farm, Australia; Comprehensive Stroke Center and Department of Neurology (J.L.S.), University of California Los Angeles; and Australian Stroke Alliance (L.C.), University of Melbourne, Parkville, Victoria, Australia
| | - Sean P Dukelow
- From the Melbourne School of Health Sciences (K.S.H., E.J.D.), and Melbourne Medical School (K.S.H., H.J., L.C.), University of Melbourne, Parkville; Stroke Theme (K.S.H., B.C.V.C., J.B., L.C.), The Florey Institute, University of Melbourne, Heidelberg; National Health and Medical Research Council Centre of Research Excellence to Accelerate Stroke Trial Innovation and Translation (K.S.H., B.C.V.C., L.C., J.B., H.J.), University of Melbourne, Parkville; Department of Medicine and Neurology (B.C.V.C., V.Y.), Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia; Department of Neurology and Rehabilitation Medicine (P.K.), University of Cincinnati, OH; Department of Clinical Neuroscience (S.P.D.), and Hotchkiss Brain Institute (S.P.D.), University of Calgary, Alberta, Canada; Department of Neurology (S.W.), Saarland University, Saarbrücken; Department of Neurology (S.W.), Martin-Luther-University, Halle, Germany; Department of Neurology (V.Y.), University of Ottawa, Ontario, Canada; Department of Neurology (J.D.P.), Christian Medical College, Ludhiana, Punjab, India; Department of Biotechnological and Applied Clinical Sciences (S.S.), University of L'Aquila, Italy; Department of Neurology (M.W.P.), Liverpool Hospital, UNSW South Western Sydney Clinical School, Warwick Farm, Australia; Comprehensive Stroke Center and Department of Neurology (J.L.S.), University of California Los Angeles; and Australian Stroke Alliance (L.C.), University of Melbourne, Parkville, Victoria, Australia
| | - Hannah Johns
- From the Melbourne School of Health Sciences (K.S.H., E.J.D.), and Melbourne Medical School (K.S.H., H.J., L.C.), University of Melbourne, Parkville; Stroke Theme (K.S.H., B.C.V.C., J.B., L.C.), The Florey Institute, University of Melbourne, Heidelberg; National Health and Medical Research Council Centre of Research Excellence to Accelerate Stroke Trial Innovation and Translation (K.S.H., B.C.V.C., L.C., J.B., H.J.), University of Melbourne, Parkville; Department of Medicine and Neurology (B.C.V.C., V.Y.), Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia; Department of Neurology and Rehabilitation Medicine (P.K.), University of Cincinnati, OH; Department of Clinical Neuroscience (S.P.D.), and Hotchkiss Brain Institute (S.P.D.), University of Calgary, Alberta, Canada; Department of Neurology (S.W.), Saarland University, Saarbrücken; Department of Neurology (S.W.), Martin-Luther-University, Halle, Germany; Department of Neurology (V.Y.), University of Ottawa, Ontario, Canada; Department of Neurology (J.D.P.), Christian Medical College, Ludhiana, Punjab, India; Department of Biotechnological and Applied Clinical Sciences (S.S.), University of L'Aquila, Italy; Department of Neurology (M.W.P.), Liverpool Hospital, UNSW South Western Sydney Clinical School, Warwick Farm, Australia; Comprehensive Stroke Center and Department of Neurology (J.L.S.), University of California Los Angeles; and Australian Stroke Alliance (L.C.), University of Melbourne, Parkville, Victoria, Australia
| | - Silke Walter
- From the Melbourne School of Health Sciences (K.S.H., E.J.D.), and Melbourne Medical School (K.S.H., H.J., L.C.), University of Melbourne, Parkville; Stroke Theme (K.S.H., B.C.V.C., J.B., L.C.), The Florey Institute, University of Melbourne, Heidelberg; National Health and Medical Research Council Centre of Research Excellence to Accelerate Stroke Trial Innovation and Translation (K.S.H., B.C.V.C., L.C., J.B., H.J.), University of Melbourne, Parkville; Department of Medicine and Neurology (B.C.V.C., V.Y.), Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia; Department of Neurology and Rehabilitation Medicine (P.K.), University of Cincinnati, OH; Department of Clinical Neuroscience (S.P.D.), and Hotchkiss Brain Institute (S.P.D.), University of Calgary, Alberta, Canada; Department of Neurology (S.W.), Saarland University, Saarbrücken; Department of Neurology (S.W.), Martin-Luther-University, Halle, Germany; Department of Neurology (V.Y.), University of Ottawa, Ontario, Canada; Department of Neurology (J.D.P.), Christian Medical College, Ludhiana, Punjab, India; Department of Biotechnological and Applied Clinical Sciences (S.S.), University of L'Aquila, Italy; Department of Neurology (M.W.P.), Liverpool Hospital, UNSW South Western Sydney Clinical School, Warwick Farm, Australia; Comprehensive Stroke Center and Department of Neurology (J.L.S.), University of California Los Angeles; and Australian Stroke Alliance (L.C.), University of Melbourne, Parkville, Victoria, Australia
| | - Vignan Yogendrakumar
- From the Melbourne School of Health Sciences (K.S.H., E.J.D.), and Melbourne Medical School (K.S.H., H.J., L.C.), University of Melbourne, Parkville; Stroke Theme (K.S.H., B.C.V.C., J.B., L.C.), The Florey Institute, University of Melbourne, Heidelberg; National Health and Medical Research Council Centre of Research Excellence to Accelerate Stroke Trial Innovation and Translation (K.S.H., B.C.V.C., L.C., J.B., H.J.), University of Melbourne, Parkville; Department of Medicine and Neurology (B.C.V.C., V.Y.), Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia; Department of Neurology and Rehabilitation Medicine (P.K.), University of Cincinnati, OH; Department of Clinical Neuroscience (S.P.D.), and Hotchkiss Brain Institute (S.P.D.), University of Calgary, Alberta, Canada; Department of Neurology (S.W.), Saarland University, Saarbrücken; Department of Neurology (S.W.), Martin-Luther-University, Halle, Germany; Department of Neurology (V.Y.), University of Ottawa, Ontario, Canada; Department of Neurology (J.D.P.), Christian Medical College, Ludhiana, Punjab, India; Department of Biotechnological and Applied Clinical Sciences (S.S.), University of L'Aquila, Italy; Department of Neurology (M.W.P.), Liverpool Hospital, UNSW South Western Sydney Clinical School, Warwick Farm, Australia; Comprehensive Stroke Center and Department of Neurology (J.L.S.), University of California Los Angeles; and Australian Stroke Alliance (L.C.), University of Melbourne, Parkville, Victoria, Australia
| | - Jeyaraj D Pandian
- From the Melbourne School of Health Sciences (K.S.H., E.J.D.), and Melbourne Medical School (K.S.H., H.J., L.C.), University of Melbourne, Parkville; Stroke Theme (K.S.H., B.C.V.C., J.B., L.C.), The Florey Institute, University of Melbourne, Heidelberg; National Health and Medical Research Council Centre of Research Excellence to Accelerate Stroke Trial Innovation and Translation (K.S.H., B.C.V.C., L.C., J.B., H.J.), University of Melbourne, Parkville; Department of Medicine and Neurology (B.C.V.C., V.Y.), Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia; Department of Neurology and Rehabilitation Medicine (P.K.), University of Cincinnati, OH; Department of Clinical Neuroscience (S.P.D.), and Hotchkiss Brain Institute (S.P.D.), University of Calgary, Alberta, Canada; Department of Neurology (S.W.), Saarland University, Saarbrücken; Department of Neurology (S.W.), Martin-Luther-University, Halle, Germany; Department of Neurology (V.Y.), University of Ottawa, Ontario, Canada; Department of Neurology (J.D.P.), Christian Medical College, Ludhiana, Punjab, India; Department of Biotechnological and Applied Clinical Sciences (S.S.), University of L'Aquila, Italy; Department of Neurology (M.W.P.), Liverpool Hospital, UNSW South Western Sydney Clinical School, Warwick Farm, Australia; Comprehensive Stroke Center and Department of Neurology (J.L.S.), University of California Los Angeles; and Australian Stroke Alliance (L.C.), University of Melbourne, Parkville, Victoria, Australia
| | - Simona Sacco
- From the Melbourne School of Health Sciences (K.S.H., E.J.D.), and Melbourne Medical School (K.S.H., H.J., L.C.), University of Melbourne, Parkville; Stroke Theme (K.S.H., B.C.V.C., J.B., L.C.), The Florey Institute, University of Melbourne, Heidelberg; National Health and Medical Research Council Centre of Research Excellence to Accelerate Stroke Trial Innovation and Translation (K.S.H., B.C.V.C., L.C., J.B., H.J.), University of Melbourne, Parkville; Department of Medicine and Neurology (B.C.V.C., V.Y.), Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia; Department of Neurology and Rehabilitation Medicine (P.K.), University of Cincinnati, OH; Department of Clinical Neuroscience (S.P.D.), and Hotchkiss Brain Institute (S.P.D.), University of Calgary, Alberta, Canada; Department of Neurology (S.W.), Saarland University, Saarbrücken; Department of Neurology (S.W.), Martin-Luther-University, Halle, Germany; Department of Neurology (V.Y.), University of Ottawa, Ontario, Canada; Department of Neurology (J.D.P.), Christian Medical College, Ludhiana, Punjab, India; Department of Biotechnological and Applied Clinical Sciences (S.S.), University of L'Aquila, Italy; Department of Neurology (M.W.P.), Liverpool Hospital, UNSW South Western Sydney Clinical School, Warwick Farm, Australia; Comprehensive Stroke Center and Department of Neurology (J.L.S.), University of California Los Angeles; and Australian Stroke Alliance (L.C.), University of Melbourne, Parkville, Victoria, Australia
| | - Julie Bernhardt
- From the Melbourne School of Health Sciences (K.S.H., E.J.D.), and Melbourne Medical School (K.S.H., H.J., L.C.), University of Melbourne, Parkville; Stroke Theme (K.S.H., B.C.V.C., J.B., L.C.), The Florey Institute, University of Melbourne, Heidelberg; National Health and Medical Research Council Centre of Research Excellence to Accelerate Stroke Trial Innovation and Translation (K.S.H., B.C.V.C., L.C., J.B., H.J.), University of Melbourne, Parkville; Department of Medicine and Neurology (B.C.V.C., V.Y.), Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia; Department of Neurology and Rehabilitation Medicine (P.K.), University of Cincinnati, OH; Department of Clinical Neuroscience (S.P.D.), and Hotchkiss Brain Institute (S.P.D.), University of Calgary, Alberta, Canada; Department of Neurology (S.W.), Saarland University, Saarbrücken; Department of Neurology (S.W.), Martin-Luther-University, Halle, Germany; Department of Neurology (V.Y.), University of Ottawa, Ontario, Canada; Department of Neurology (J.D.P.), Christian Medical College, Ludhiana, Punjab, India; Department of Biotechnological and Applied Clinical Sciences (S.S.), University of L'Aquila, Italy; Department of Neurology (M.W.P.), Liverpool Hospital, UNSW South Western Sydney Clinical School, Warwick Farm, Australia; Comprehensive Stroke Center and Department of Neurology (J.L.S.), University of California Los Angeles; and Australian Stroke Alliance (L.C.), University of Melbourne, Parkville, Victoria, Australia
| | - Mark W Parsons
- From the Melbourne School of Health Sciences (K.S.H., E.J.D.), and Melbourne Medical School (K.S.H., H.J., L.C.), University of Melbourne, Parkville; Stroke Theme (K.S.H., B.C.V.C., J.B., L.C.), The Florey Institute, University of Melbourne, Heidelberg; National Health and Medical Research Council Centre of Research Excellence to Accelerate Stroke Trial Innovation and Translation (K.S.H., B.C.V.C., L.C., J.B., H.J.), University of Melbourne, Parkville; Department of Medicine and Neurology (B.C.V.C., V.Y.), Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia; Department of Neurology and Rehabilitation Medicine (P.K.), University of Cincinnati, OH; Department of Clinical Neuroscience (S.P.D.), and Hotchkiss Brain Institute (S.P.D.), University of Calgary, Alberta, Canada; Department of Neurology (S.W.), Saarland University, Saarbrücken; Department of Neurology (S.W.), Martin-Luther-University, Halle, Germany; Department of Neurology (V.Y.), University of Ottawa, Ontario, Canada; Department of Neurology (J.D.P.), Christian Medical College, Ludhiana, Punjab, India; Department of Biotechnological and Applied Clinical Sciences (S.S.), University of L'Aquila, Italy; Department of Neurology (M.W.P.), Liverpool Hospital, UNSW South Western Sydney Clinical School, Warwick Farm, Australia; Comprehensive Stroke Center and Department of Neurology (J.L.S.), University of California Los Angeles; and Australian Stroke Alliance (L.C.), University of Melbourne, Parkville, Victoria, Australia
| | - Jeffrey L Saver
- From the Melbourne School of Health Sciences (K.S.H., E.J.D.), and Melbourne Medical School (K.S.H., H.J., L.C.), University of Melbourne, Parkville; Stroke Theme (K.S.H., B.C.V.C., J.B., L.C.), The Florey Institute, University of Melbourne, Heidelberg; National Health and Medical Research Council Centre of Research Excellence to Accelerate Stroke Trial Innovation and Translation (K.S.H., B.C.V.C., L.C., J.B., H.J.), University of Melbourne, Parkville; Department of Medicine and Neurology (B.C.V.C., V.Y.), Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia; Department of Neurology and Rehabilitation Medicine (P.K.), University of Cincinnati, OH; Department of Clinical Neuroscience (S.P.D.), and Hotchkiss Brain Institute (S.P.D.), University of Calgary, Alberta, Canada; Department of Neurology (S.W.), Saarland University, Saarbrücken; Department of Neurology (S.W.), Martin-Luther-University, Halle, Germany; Department of Neurology (V.Y.), University of Ottawa, Ontario, Canada; Department of Neurology (J.D.P.), Christian Medical College, Ludhiana, Punjab, India; Department of Biotechnological and Applied Clinical Sciences (S.S.), University of L'Aquila, Italy; Department of Neurology (M.W.P.), Liverpool Hospital, UNSW South Western Sydney Clinical School, Warwick Farm, Australia; Comprehensive Stroke Center and Department of Neurology (J.L.S.), University of California Los Angeles; and Australian Stroke Alliance (L.C.), University of Melbourne, Parkville, Victoria, Australia
| | - Leonid Churilov
- From the Melbourne School of Health Sciences (K.S.H., E.J.D.), and Melbourne Medical School (K.S.H., H.J., L.C.), University of Melbourne, Parkville; Stroke Theme (K.S.H., B.C.V.C., J.B., L.C.), The Florey Institute, University of Melbourne, Heidelberg; National Health and Medical Research Council Centre of Research Excellence to Accelerate Stroke Trial Innovation and Translation (K.S.H., B.C.V.C., L.C., J.B., H.J.), University of Melbourne, Parkville; Department of Medicine and Neurology (B.C.V.C., V.Y.), Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia; Department of Neurology and Rehabilitation Medicine (P.K.), University of Cincinnati, OH; Department of Clinical Neuroscience (S.P.D.), and Hotchkiss Brain Institute (S.P.D.), University of Calgary, Alberta, Canada; Department of Neurology (S.W.), Saarland University, Saarbrücken; Department of Neurology (S.W.), Martin-Luther-University, Halle, Germany; Department of Neurology (V.Y.), University of Ottawa, Ontario, Canada; Department of Neurology (J.D.P.), Christian Medical College, Ludhiana, Punjab, India; Department of Biotechnological and Applied Clinical Sciences (S.S.), University of L'Aquila, Italy; Department of Neurology (M.W.P.), Liverpool Hospital, UNSW South Western Sydney Clinical School, Warwick Farm, Australia; Comprehensive Stroke Center and Department of Neurology (J.L.S.), University of California Los Angeles; and Australian Stroke Alliance (L.C.), University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
30
|
Draghia-Akli R, Read SW, Hughes EA. Overview of ACTIV trial-specific lessons learned. J Clin Transl Sci 2024; 8:e149. [PMID: 39478781 PMCID: PMC11523021 DOI: 10.1017/cts.2023.698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 11/02/2024] Open
Abstract
Accelerating COVID-19 Therapeutic Interventions and Vaccines (ACTIV) was an extraordinary example of a public-private partnership (PPP) that brought together over thirty organizations and hundreds of individuals to address one of the most pressing global health needs in recent decades. In particular, ACTIV provided a key avenue for testing numerous therapeutics for their potential benefit in treating the SARS-CoV-2 virus or the resulting symptoms of acute COVID-19 infection. Given the speed and scale at which ACTIV designed and implemented master protocols across global networks that it was simultaneously working to create, the PPP can provide valuable lessons for best practices and avoiding pitfalls the next time the world is faced with a global pandemic of a novel pathogen. This report provides a general overview of the ACTIV partnership to set the stage and context for the subsequent articles in this issue that will relay these lessons learned.
Collapse
Affiliation(s)
| | - Sarah W. Read
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | | |
Collapse
|
31
|
Adam SJ, Dunsmore SE, Merck LH, Read SW, Rosenberg Y. The future is now: Using the lessons learned from the ACTIV COVID-19 therapeutics trials to create an inclusive and efficient clinical trials enterprise. J Clin Transl Sci 2024; 8:e148. [PMID: 39478787 PMCID: PMC11523011 DOI: 10.1017/cts.2024.622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024] Open
Affiliation(s)
- Stacey J. Adam
- Foundation for the National Institutes of Health, North Bethesda, USA
| | - Sarah E. Dunsmore
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, USA
| | - Lisa H. Merck
- Department of Emergency Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Sarah W. Read
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, USA
| | - Yves Rosenberg
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, USA
| |
Collapse
|
32
|
Wilbourn AC, Tsodikov OV, Garneau-Tsodikova S. Association of COVID-19 risk factors with systemic fungal infections in hospitalized patients. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.10.24315254. [PMID: 39417099 PMCID: PMC11482998 DOI: 10.1101/2024.10.10.24315254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Purpose A new category of systemic co-infections that emerged with the COVID-19 pandemic is known as COVID-19-associated (CA) fungal infections, which include pulmonary aspergillosis (CAPA), candidiasis (CAC), and mucormycosis (CAM). We aimed to study the association between patient characteristics of hospitalized COVID-19 patients, COVID-19 comorbidities, and COVID-19 therapies with secondary non-superficial fungal infections. Methods We performed descriptive and regression analyses of data from 4,999 hospitalized COVID-19 patients from the University of Kentucky Healthcare (UKHC) system. Results The patients with secondary systemic fungal infections had a 6-fold higher risk of death than those without such infections. Generally, the risk factors for severe COVID-19 (age, obesity, cardiovascular disease, diabetes, and lack of COVID-19 vaccination) were strong predictors of a secondary fungal infection. However, several characteristics had much higher risks, suggesting that a causative link may be at play: ICU admission, mechanical ventilation, length of hospital stay, and steroid use. Conclusions In sum, this study found that the known risk factors for severe COVID-19 disease, age, diabetes, cardiovascular disease, obesity, ventilation, and high steroid doses were all predictors of a secondary fungal infection. Steroid therapy may need to be modified to account for a risk or a presence of a fungal infection in vulnerable patients.
Collapse
Affiliation(s)
- Abbygail C. Wilbourn
- University of Kentucky, College of Pharmacy, 789 South Limestone St., Lexington, KY, 40536-0596, USA
| | - Oleg V. Tsodikov
- University of Kentucky, College of Pharmacy, 789 South Limestone St., Lexington, KY, 40536-0596, USA
| | - Sylvie Garneau-Tsodikova
- University of Kentucky, College of Pharmacy, 789 South Limestone St., Lexington, KY, 40536-0596, USA
| |
Collapse
|
33
|
Cocker D, Fitzgerald R, Brown CS, Holmes A. Protecting healthcare and patient pathways from infection and antimicrobial resistance. BMJ 2024; 387:e077927. [PMID: 39374953 PMCID: PMC11450933 DOI: 10.1136/bmj-2023-077927] [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] [Indexed: 10/09/2024]
Affiliation(s)
- Derek Cocker
- David Price Evans Global Health and Infectious Diseases Research Group, University of Liverpool, Liverpool, UK
| | - Richard Fitzgerald
- NIHR Royal Liverpool and Broadgreen Clinical Research Facility, Liverpool, UK
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, UK
| | - Colin S Brown
- UK Health Security Agency, London, UK
- National Institute of Health Research, Health Protection Research Unit in Healthcare Associated Infection and Antimicrobial Resistance, Imperial College London, London, UK
| | - Alison Holmes
- David Price Evans Global Health and Infectious Diseases Research Group, University of Liverpool, Liverpool, UK
- National Institute of Health Research, Health Protection Research Unit in Healthcare Associated Infection and Antimicrobial Resistance, Imperial College London, London, UK
| |
Collapse
|
34
|
Ananth S, Mathioudakis AG, Hansel J. Steroids in severe community-acquired pneumonia. Breathe (Sheff) 2024; 20:240081. [PMID: 39360025 PMCID: PMC11444496 DOI: 10.1183/20734735.0081-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 08/17/2024] [Indexed: 10/04/2024] Open
Abstract
There is conflicting evidence regarding the use of steroids in severe community-acquired pneumonia (CAP), with previous randomised controlled trials limited by small sample sizes. ESCAPe and CAPE COD are two recently published large trials on steroids in severe CAP. ESCAPe assessed the initiation of methylprednisolone within 72-96 h of hospital admission, while CAPE COD studied the use of hydrocortisone within 24 h of the development of severe CAP. ESCAPe did not show any differences in all-cause 60-day mortality or any of its secondary outcomes. CAPE COD showed that hydrocortisone improved all-cause 28-day mortality and reduced the risk of intubation or vasopressor-dependent shock. Important differences between the trials included the steroid regimens used, timing of steroid administration and baseline characteristics, with more diabetic patients included in ESCAPe. The results of CAPE COD support the initiation of hydrocortisone within 24 h of developing severe CAP, but more research is needed to evaluate long-term outcomes and optimum dosing regimens for steroids in severe CAP.
Collapse
Affiliation(s)
- Sachin Ananth
- London North West University Healthcare NHS Trust, London, UK
| | - Alexander G. Mathioudakis
- Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- The North West Lung Centre, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - Jan Hansel
- Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- The North West Lung Centre, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| |
Collapse
|
35
|
Chen Z, Harhay MO, Fan E, Granholm A, McAuley DF, Urner M, Yarnell CJ, Goligher EC, Heath A. Statistical Power and Performance of Strategies to Analyze Composites of Survival and Duration of Ventilation in Clinical Trials. Crit Care Explor 2024; 6:e1152. [PMID: 39302988 PMCID: PMC11419436 DOI: 10.1097/cce.0000000000001152] [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: 09/22/2024] Open
Abstract
BACKGROUND Patients with acute hypoxemic respiratory failure are at high risk of death and prolonged time on the ventilator. Interventions often aim to reduce both mortality and time on the ventilator. Many methods have been proposed for analyzing these endpoints as a single composite outcome (days alive and free of ventilation), but it is unclear which analytical method provides the best performance. Thus, we aimed to determine the analysis method with the highest statistical power for use in clinical trials. METHODS Using statistical simulation, we compared multiple methods for analyzing days alive and free of ventilation: the t, Wilcoxon rank-sum, and Kryger Jensen and Lange tests, as well as the proportional odds, hurdle-Poisson, and competing risk models. We compared 14 scenarios relating to: 1) varying baseline distributions of mortality and duration of ventilation, which were based on data from a registry of patients with acute hypoxemic respiratory failure and 2) the varying effects of treatment on mortality and duration of ventilation. RESULTS AND CONCLUSIONS All methods have good control of type 1 error rates (i.e., avoid false positive findings). When data are simulated using a proportional odds model, the t test and ordinal models have the highest relative power (92% and 90%, respectively), followed by competing risk models. When the data are simulated using survival models, the competing risk models have the highest power (100% and 92%), followed by the t test and a ten-category ordinal model. All models struggled to detect the effect of the intervention when the treatment only affected one of mortality and duration of ventilation. Overall, the best performing analytical strategy depends on the respective effects of treatment on survival and duration of ventilation and the underlying distribution of the outcomes. The evaluated models each provide a different interpretation for the treatment effect, which must be considered alongside the statistical power when selecting analysis models.
Collapse
Affiliation(s)
- Ziming Chen
- Child Health Evaluative Sciences, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
| | - Michael O. Harhay
- Department of Biostatistics, Epidemiology and Informatics Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Eddy Fan
- Department of Medicine, Division of Respirology, University Health Network, Toronto, ON, Canada
| | - Anders Granholm
- Department of Intensive Care, Copenhagen University Hospital–Rigshospitalet, Copenhagen, Denmark
| | - Daniel F. McAuley
- School of Medicine, Dentistry and Biomedical Sciences, Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast, United Kingdom
- Regional Intensive Care Unit, Royal Victoria Hospital, Belfast, United Kingdom
| | - Martin Urner
- Department of Anesthesiology and Pain Medicine, University of Toronto, Toronto, ON, Canada
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, ON, Canada
| | - Christopher J. Yarnell
- Department of Medicine, Division of Respirology, University Health Network, Toronto, ON, Canada
- Department of Critical Care Medicine, Scarborough Health Network, Toronto, ON, Canada
- Institute of Health Policy, Management, and Evaluation, University of Toronto, Toronto, ON, Canada
| | - Ewan C. Goligher
- Department of Biostatistics, Epidemiology and Informatics Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, Toronto, ON, Canada
| | - Anna Heath
- Child Health Evaluative Sciences, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
- Department of Statistical Science, University College London, London, United Kingdom
| |
Collapse
|
36
|
Strongin SR, Stelson E, Soares L, Sukhatme V, Dasher P, Schito M, Challa AP, Geng LN, Walker TA. Using real-world data to accelerate the search for long COVID therapies. Life Sci 2024; 353:122940. [PMID: 39098596 DOI: 10.1016/j.lfs.2024.122940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 07/11/2024] [Accepted: 08/01/2024] [Indexed: 08/06/2024]
Abstract
Long COVID, a spectrum of symptoms and syndromes that can develop after SARS-COV-2 infection, can significantly affect patients' health, quality of life and impact their ability to productively function in society. There is currently no approved therapy for Long COVID and there is an urgent need for rigorous clinical trials to find such treatments. Although research into the pathophysiology of Long COVID is advancing, investigations into treatment for patients remain underfunded and, as a result, understudied. Owing to the urgency of the Long COVID pandemic and as a research collaborative across a diversity of biomedical innovation value propositions, we are calling for a new approach that parallelizes pathophysiologic and therapeutic research into this condition, leveraging patient-centered research and real-world data to generate hypotheses to assess the effectiveness of existing FDA approved drugs. Accelerated discovery of therapeutics for Long COVID can then be confirmed through efficient and cost-effective adaptive platform clinical trials.
Collapse
Affiliation(s)
| | | | - Letícia Soares
- Patient-Led Research Collaborative, United States of America
| | | | | | - Marco Schito
- Critical Path Institute, United States of America
| | | | | | | |
Collapse
|
37
|
Behal ML, Flannery AH, Miano TA. The times are changing: A primer on novel clinical trial designs and endpoints in critical care research. Am J Health Syst Pharm 2024; 81:890-902. [PMID: 38742701 PMCID: PMC11383190 DOI: 10.1093/ajhp/zxae134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Indexed: 05/16/2024] Open
Affiliation(s)
- Michael L Behal
- Department of Pharmacy, University of Tennessee Medical Center, Knoxville, TN, USA
| | - Alexander H Flannery
- Department of Pharmacy Practice and Science, University of Kentucky College of Pharmacy, Lexington, KY, USA
| | - Todd A Miano
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, and Department of Pharmacy, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
38
|
Graham Linck EJ, Goligher EC, Semler MW, Churpek MM. Toward Precision in Critical Care Research: Methods for Observational and Interventional Studies. Crit Care Med 2024; 52:1439-1450. [PMID: 39145702 PMCID: PMC11328956 DOI: 10.1097/ccm.0000000000006371] [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: 08/16/2024]
Abstract
Critical care trials evaluate the effect of interventions in patients with diverse personal histories and causes of illness, often under the umbrella of heterogeneous clinical syndromes, such as sepsis or acute respiratory distress syndrome. Given this variation, it is reasonable to expect that the effect of treatment on outcomes may differ for individuals with variable characteristics. However, in randomized controlled trials, efficacy is typically assessed by the average treatment effect (ATE), which quantifies the average effect of the intervention on the outcome in the study population. Importantly, the ATE may hide variations of the treatment's effect on a clinical outcome across levels of patient characteristics, which may erroneously lead to the conclusion that an intervention does not work overall when it may in fact benefit certain patients. In this review, we describe methodological approaches for assessing heterogeneity of treatment effect (HTE), including expert-derived subgrouping, data-driven subgrouping, baseline risk modeling, treatment effect modeling, and individual treatment rule estimation. Next, we outline how insights from HTE analyses can be incorporated into the design of clinical trials. Finally, we propose a research agenda for advancing the field and bringing HTE approaches to the bedside.
Collapse
Affiliation(s)
- Emma J Graham Linck
- Department of Biostatistics and Medical Informatics, UW-Madison, Madison, WI
| | - Ewan C Goligher
- Interdepartmental Division of Critical Care Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Matthew W Semler
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University Medical Center, Nashville, TN
| | - Matthew M Churpek
- Department of Biostatistics and Medical Informatics, UW-Madison, Madison, WI
- Division of Pulmonary and Critical Care, Department of Medicine, University of Wisconsin-Madison, Madison, WI
| |
Collapse
|
39
|
Sanchez-Pinto LN, Del Pilar Arias López M, Scott H, Gibbons K, Moor M, Watson RS, Wiens MO, Schlapbach LJ, Bennett TD. Digital solutions in paediatric sepsis: current state, challenges, and opportunities to improve care around the world. Lancet Digit Health 2024; 6:e651-e661. [PMID: 39138095 PMCID: PMC11371309 DOI: 10.1016/s2589-7500(24)00141-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 05/17/2024] [Accepted: 06/14/2024] [Indexed: 08/15/2024]
Abstract
The digitisation of health care is offering the promise of transforming the management of paediatric sepsis, which is a major source of morbidity and mortality in children worldwide. Digital technology is already making an impact in paediatric sepsis, but is almost exclusively benefiting patients in high-resource health-care settings. However, digital tools can be highly scalable and cost-effective, and-with the right planning-have the potential to reduce global health disparities. Novel digital solutions, from wearable devices and mobile apps, to electronic health record-embedded decision support tools, have an unprecedented opportunity to transform paediatric sepsis research and care. In this Series paper, we describe the current state of digital solutions in paediatric sepsis around the world, the advances in digital technology that are enabling the development of novel applications, and the potential effect of advances in artificial intelligence in paediatric sepsis research and clinical care.
Collapse
Affiliation(s)
- L Nelson Sanchez-Pinto
- Department of Pediatrics, Northwestern University Feinberg School of Medicine and Ann & Robert H Lurie Children's Hospital of Chicago, Chicago, IL, USA.
| | | | - Halden Scott
- Department of Pediatrics, University of Colorado-Denver and Children's Hospital Colorado, Aurora, CO, USA
| | - Kristen Gibbons
- Children's Intensive Care Research Program, Child Health Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Michael Moor
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - R Scott Watson
- Department of Pediatrics, University of Washington and Seattle Children's Hospital, Seattle, WA, USA
| | - Matthew O Wiens
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada; World Alliance for Lung and Intensive Care Medicine in Uganda, Kampala, Uganda
| | - Luregn J Schlapbach
- Children's Intensive Care Research Program, Child Health Research Centre, The University of Queensland, Brisbane, QLD, Australia; Department of Intensive Care and Neonatology, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Tellen D Bennett
- Department of Pediatrics, University of Colorado-Denver and Children's Hospital Colorado, Aurora, CO, USA
| |
Collapse
|
40
|
Spagnolo P, Maher TM. The future of clinical trials in idiopathic pulmonary fibrosis. Curr Opin Pulm Med 2024; 30:494-499. [PMID: 38963152 PMCID: PMC11377049 DOI: 10.1097/mcp.0000000000001099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
PURPOSE OF REVIEW Idiopathic pulmonary fibrosis (IPF) is a progressive lung disease with a poor prognosis and limited therapeutic options. A multitude of promising compounds are currently being investigated; however, the design and conductance of late-phase clinical trials in IPF has proven particularly challenging. RECENT FINDINGS Despite promising phase 2 data, ziritaxestat, an autotaxin inhibitor, pentraxin-2, an endogenous protein that regulates wound healing and fibrosis, and pamrevlumab, a human monoclonal antibody against connective tissue growth factor, failed to show efficacy in phase 3 trials. Endpoint selection is critical for the design, execution, and success of clinical trials; recently, attention has been paid to the assessment of how patients feel, function, and survive with the aim of aligning scientific objectives and patient needs in IPF. External control arms are control patients that derive from historical randomized controlled trials, registries, or electronic health records. They are increasingly used to assess treatment efficacy in clinical trials owing to their potential to reduce study duration and cost and increase generalizability of findings. SUMMARY Advances in study design, end point selection and statistical analysis, and innovative strategies for more efficient enrolment of study participants have the potential to increase the likelihood of success of late-phase clinical trials in IPF.
Collapse
Affiliation(s)
- Paolo Spagnolo
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Toby M. Maher
- Department of Pulmonary, Critical Care and Sleep Medicine, University of Southern California Keck School of Medicine, Los Angeles, California, USA
- Section of Inflammation, Repair and Development, Imperial College London National Heart and Lung Institute, London, UK
| |
Collapse
|
41
|
Hui DSC. Antiviral treatment and prophylaxis for influenza. Lancet 2024; 404:726-727. [PMID: 39181581 DOI: 10.1016/s0140-6736(24)01698-2] [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: 08/07/2024] [Accepted: 08/13/2024] [Indexed: 08/27/2024]
Affiliation(s)
- David S C Hui
- Department of Medicine and Therapeutics and S H Ho Research Center for Infectious Diseases, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region, China.
| |
Collapse
|
42
|
Kawano-Dourado L, Molina-Molina M, Sellares J, Enghelmayer JI. The Relevance of REMAP-ILD for Ibero-American Countries: A Randomized Embedded Multifactorial Adaptive Platform (REMAP) Trial in the Field of Interstitial Lung Diseases (ILDs). Arch Bronconeumol 2024; 60:463-465. [PMID: 38816284 DOI: 10.1016/j.arbres.2024.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 05/11/2024] [Accepted: 05/13/2024] [Indexed: 06/01/2024]
Affiliation(s)
- Leticia Kawano-Dourado
- Hcor Research Institute, Hcor Hospital, Sao Paulo, Brazil; Pulmonary Division, Heart Institute (InCor), University of Sao Paulo, Sao Paulo, Brazil; MAGIC Evidence Ecosystem Foundation, Oslo, Norway.
| | - Maria Molina-Molina
- ILD Unit, Respiratory Department, University Hospital of Bellvitge, IDIBELL, Hospitalet de Llobregat, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER), Spain
| | - Jacobo Sellares
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER), Spain; Servei de Pneumologia, Hospital Clínic-Universitat de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Facultat de Medicina, Universitat de Vic (UVIC), Vic, Spain
| | - Juan I Enghelmayer
- Hospital de Clínicas, Universidad de Buenos Aires, Argentina; Fundación FUNEF, Buenos Aires, Argentina
| |
Collapse
|
43
|
Lee KM, Emsley R. The impact of heterogeneity on the analysis of platform trials with normally distributed outcomes. BMC Med Res Methodol 2024; 24:163. [PMID: 39080538 PMCID: PMC11290279 DOI: 10.1186/s12874-024-02293-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 07/19/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND A platform trial approach allows adding arms to on-going trials to speed up intervention discovery programs. A control arm remains open for recruitment in a platform trial while intervention arms may be added after the onset of the study and could be terminated early for efficacy and/or futility when early stopping is allowed. The topic of utilising non-concurrent control data in the analysis of platform trials has been explored and discussed extensively. A less familiar issue is the presence of heterogeneity, which may exist for example due to modification of enrolment criteria and recruitment strategy. METHOD We conduct a simulation study to explore the impact of heterogeneity on the analysis of a two-stage platform trial design. We consider heterogeneity in treatment effects and heteroscedasticity in outcome data across stages for a normally distributed endpoint. We examine the performance of some hypothesis testing procedures and modelling strategies. The use of non-concurrent control data is also considered accordingly. Alongside standard regression analysis, we examine the performance of a novel method that was known as the pairwise trials analysis. It is similar to a network meta-analysis approach but adjusts for treatment comparisons instead of individual studies using fixed effects. RESULTS Several testing strategies with concurrent control data seem to control the type I error rate at the required level when there is heteroscedasticity in outcome data across stages and/or a random cohort effect. The main parameter of treatment effects in some analysis models correspond to overall treatment effects weighted by stage wise sample sizes; while others correspond to the effect observed within a single stage. The characteristics of the estimates are not affected significantly by the presence of a random cohort effect and/ or heteroscedasticity. CONCLUSION In view of heterogeneity in treatment effect across stages, the specification of null hypotheses in platform trials may need to be more subtle. We suggest employing testing procedure of adaptive design as opposed to testing the statistics from regression models; comparing the estimates from the pairwise trials analysis method and the regression model with interaction terms may indicate if heterogeneity is negligible.
Collapse
Affiliation(s)
- Kim May Lee
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 16 De Crespigny Park, SE5 8AF, London, UK.
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | - Richard Emsley
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 16 De Crespigny Park, SE5 8AF, London, UK
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
44
|
Dymock M, McLeod C, Richmond P, Snelling T, Marsh JA. Statistical considerations for the platform trial in COVID-19 vaccine priming and boosting. Trials 2024; 25:507. [PMID: 39060943 PMCID: PMC11282703 DOI: 10.1186/s13063-024-08343-y] [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: 12/23/2023] [Accepted: 07/14/2024] [Indexed: 07/28/2024] Open
Abstract
The Platform trial In COVID-19 priming and BOOsting (PICOBOO) is a multi-site, adaptive platform trial designed to generate evidence of the immunogenicity, reactogenicity, and cross-protection of different booster vaccination strategies against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its variants, specific for the Australian context. The PICOBOO trial randomises participants to receive one of three COVID-19 booster vaccine brands (Pfizer, Moderna, Novavax) available for use in Australia, where the vaccine brand subtypes vary over time according to the national vaccine roll out strategy, and employs a Bayesian hierarchical modelling approach to efficiently borrow information across consecutive booster doses, age groups and vaccine brand subtypes. Here, we briefly describe the PICOBOO trial structure and report the statistical considerations for the estimands, statistical models and decision making for trial adaptations. This paper should be read in conjunction with the PICOBOO Core Protocol and PICOBOO Sub-Study Protocol 1: Booster Vaccination. PICOBOO was registered on 10 February 2022 with the Australian and New Zealand Clinical Trials Registry ACTRN12622000238774.
Collapse
Affiliation(s)
- Michael Dymock
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, 15 Hospital Avenue, Nedlands, 6009, Perth, Australia.
| | - Charlie McLeod
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, 15 Hospital Avenue, Nedlands, 6009, Perth, Australia
- Infectious Diseases Department, Perth Children's Hospital, 15 Hospital Avenue, Nedlands, 6009, Perth, Australia
- School of Medicine, The University of Western Australia, 35 Stirling Highway, Crawley, 6009, Perth, Australia
| | - Peter Richmond
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, 15 Hospital Avenue, Nedlands, 6009, Perth, Australia
- School of Medicine, The University of Western Australia, 35 Stirling Highway, Crawley, 6009, Perth, Australia
- Centre for Child Health Research, The University of Western Australia, 35 Stirling Highway, Crawley, 6009, Perth, Australia
- General Paediatrics and Immunology Departments, Perth Children's Hospital, 15 Hospital Avenue, Nedlands, 6009, Perth, Australia
| | - Tom Snelling
- Sydney School of Public Health, Faculty of Medicine and Health, University of Sydney, Camperdown, 2006, Sydney, Australia
| | - Julie A Marsh
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, 15 Hospital Avenue, Nedlands, 6009, Perth, Australia
- Centre for Child Health Research, The University of Western Australia, 35 Stirling Highway, Crawley, 6009, Perth, Australia
| |
Collapse
|
45
|
Kawano-Dourado L, Kulkarni T, Ryerson CJ, Rivera-Ortega P, Baldi BG, Chaudhuri N, Funke-Chambour M, Hoffmann-Vold AM, Johannson KA, Khor YH, Montesi SB, Piccari L, Prosch H, Molina-Molina M, Sellares Torres J, Bauer-Ventura I, Rajan S, Jacob J, Richards D, Spencer LG, Wendelberger B, Jensen T, Quintana M, Kreuter M, Gordon AC, Martinez FJ, Kaminski N, Cornelius V, Lewis R, Adams W, Jenkins G. Adaptive multi-interventional trial platform to improve patient care for fibrotic interstitial lung diseases. Thorax 2024; 79:788-795. [PMID: 38448221 PMCID: PMC11287572 DOI: 10.1136/thorax-2023-221148] [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: 11/02/2023] [Accepted: 02/06/2024] [Indexed: 03/08/2024]
Abstract
BACKGROUND Fibrotic interstitial lung diseases (fILDs) are a heterogeneous group of lung diseases associated with significant morbidity and mortality. Despite a large increase in the number of clinical trials in the last 10 years, current regulatory-approved management approaches are limited to two therapies that prevent the progression of fibrosis. The drug development pipeline is long and there is an urgent need to accelerate this process. This manuscript introduces the concept and design of an innovative research approach to drug development in fILD: a global Randomised Embedded Multifactorial Adaptive Platform in fILD (REMAP-ILD). METHODS Description of the REMAP-ILD concept and design: the specific terminology, design characteristics (multifactorial, adaptive features, statistical approach), target population, interventions, outcomes, mission and values, and organisational structure. RESULTS The target population will be adult patients with fILD, and the primary outcome will be a disease progression model incorporating forced vital capacity and mortality over 12 months. Responsive adaptive randomisation, prespecified thresholds for success and futility will be used to assess the effectiveness and safety of interventions. REMAP-ILD embraces the core values of diversity, equity, and inclusion for patients and researchers, and prioritises an open-science approach to data sharing and dissemination of results. CONCLUSION By using an innovative and efficient adaptive multi-interventional trial platform design, we aim to accelerate and improve care for patients with fILD. Through worldwide collaboration, novel analytical methodology and pragmatic trial delivery, REMAP-ILD aims to overcome major limitations associated with conventional randomised controlled trial approaches to rapidly improve the care of people living with fILD.
Collapse
Affiliation(s)
- Leticia Kawano-Dourado
- Hcor Research Institute, Hcor Hospital, Sao Paulo, Brazil
- Pulmonary Division, Heart Institute (InCor), University of Sao Paulo, Sao Paulo, Brazil
- MAGIC Evidence Ecosystem Foundation, Oslo, Norway
| | - Tejaswini Kulkarni
- The University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama, USA
| | - Christopher J Ryerson
- Department of Medicine and Centre of Heart Lung Innovations, University of British Columbia, Vancouver, British Columbia, Canada
| | - Pilar Rivera-Ortega
- Interstitial Lung Disease Unit, Respiratory Medicine, Manchester University NHS Foundation Trust, Manchester, UK
| | - Bruno Guedes Baldi
- Pulmonary Division, Heart Institute (InCor), University of Sao Paulo, Sao Paulo, Brazil
| | - Nazia Chaudhuri
- Department of Health and Life Sciences, School of Medicine, University of Ulster, Londonderry, UK
| | - Manuela Funke-Chambour
- Department for Pulmonology, Allergology and clinical Immunology, Inselspital, University Hospital Bern, Bern, Switzerland
| | - Anna-Maria Hoffmann-Vold
- Department of Rheumatology, Oslo University Hospital, Oslo, Norway
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Kerri A Johannson
- Department of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Yet Hong Khor
- Respiratory Research@Alfred, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Department of Respiratory and Sleep Medicine, Austin Health, Heidelberg, Victoria, Australia
| | - Sydney B Montesi
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Lucilla Piccari
- Department of Pulmonology, Hospital del Mar, Barcelona, Spain
| | - Helmut Prosch
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - María Molina-Molina
- Servei de Pneumologia, Grup de Recerca Pneumològic, Institut d'Investigacions Biomèdiques de Bellvitge (IDIBELL), Hospital Universitari de Bellvitge, Barcelona, Spain
| | - Jacobo Sellares Torres
- Grup de Treball de Malalties Pulmonars Intersticials. Pneumology Service, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Iazsmin Bauer-Ventura
- Rheumatology Division, University of Chicago Pritzker School of Medicine, Chicago, Illinois, USA
| | - Sujeet Rajan
- Bombay Hospital Institute of Medical Sciences, Mumbai, Maharashtra, India
| | - Joseph Jacob
- Centre for Medical Imaging and Computing, University College London, London, UK
- Department of Respiratory Medicine, University College London, London, UK
| | - Duncan Richards
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Lisa G Spencer
- Liverpool Interstitial Lung Disease Service, Aintree Hospital, Liverpool University Hospitals NHS Foundation Trust Library and Knowledge Service, Liverpool, UK
| | | | | | | | - Michael Kreuter
- Mainz Center for Pulmonary Medicine, Department of Pulmology, Mainz University Medical Center and Department of Pulmonary, Critical Care & Sleep Medicine, Marienhaus Clinic Mainz, Mainz, Germany
| | - Anthony C Gordon
- Division of Anaesthetics, Pain Medicine and Intensive Care, Imperial College London, London, UK
| | - Fernando J Martinez
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine, New York City, New York, USA
| | - Naftali Kaminski
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Roger Lewis
- Berry Consultants, Los Angeles, California, USA
| | - Wendy Adams
- Action for Pulmonary Fibrosis Foundation, London, UK
| | - Gisli Jenkins
- Margaret Turner Warwick Centre for Fibrosing Lung Disease, National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
46
|
Gordon AC, Alipanah-Lechner N, Bos LD, Dianti J, Diaz JV, Finfer S, Fujii T, Giamarellos-Bourboulis EJ, Goligher EC, Gong MN, Karakike E, Liu VX, Lumlertgul N, Marshall JC, Menon DK, Meyer NJ, Munroe ES, Myatra SN, Ostermann M, Prescott HC, Randolph AG, Schenck EJ, Seymour CW, Shankar-Hari M, Singer M, Smit MR, Tanaka A, Taccone FS, Thompson BT, Torres LK, van der Poll T, Vincent JL, Calfee CS. From ICU Syndromes to ICU Subphenotypes: Consensus Report and Recommendations for Developing Precision Medicine in the ICU. Am J Respir Crit Care Med 2024; 210:155-166. [PMID: 38687499 PMCID: PMC11273306 DOI: 10.1164/rccm.202311-2086so] [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: 11/14/2023] [Accepted: 04/29/2024] [Indexed: 05/02/2024] Open
Abstract
Critical care uses syndromic definitions to describe patient groups for clinical practice and research. There is growing recognition that a "precision medicine" approach is required and that integrated biologic and physiologic data identify reproducible subpopulations that may respond differently to treatment. This article reviews the current state of the field and considers how to successfully transition to a precision medicine approach. To impact clinical care, identification of subpopulations must do more than differentiate prognosis. It must differentiate response to treatment, ideally by defining subgroups with distinct functional or pathobiological mechanisms (endotypes). There are now multiple examples of reproducible subpopulations of sepsis, acute respiratory distress syndrome, and acute kidney or brain injury described using clinical, physiological, and/or biological data. Many of these subpopulations have demonstrated the potential to define differential treatment response, largely in retrospective studies, and that the same treatment-responsive subpopulations may cross multiple clinical syndromes (treatable traits). To bring about a change in clinical practice, a precision medicine approach must be evaluated in prospective clinical studies requiring novel adaptive trial designs. Several such studies are underway, but there are multiple challenges to be tackled. Such subpopulations must be readily identifiable and be applicable to all critically ill populations around the world. Subdividing clinical syndromes into subpopulations will require large patient numbers. Global collaboration of investigators, clinicians, industry, and patients over many years will therefore be required to transition to a precision medicine approach and ultimately realize treatment advances seen in other medical fields.
Collapse
Affiliation(s)
| | - Narges Alipanah-Lechner
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, California
| | | | - Jose Dianti
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada
- Departamento de Cuidados Intensivos, Centro de Educación Médica e Investigaciones Clínicas, Buenos Aires, Argentina
| | | | - Simon Finfer
- School of Public Health, Imperial College London, London, United Kingdom
- The George Institute for Global Health, University of New South Wales, Sydney, Australia
| | - Tomoko Fujii
- Jikei University School of Medicine, Jikei University Hospital, Tokyo, Japan
| | | | - Ewan C. Goligher
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Michelle Ng Gong
- Division of Critical Care Medicine and
- Division of Pulmonary Medicine, Department of Medicine and Department of Epidemiology and Population Health, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | - Eleni Karakike
- Second Department of Critical Care Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Vincent X. Liu
- Division of Research, Kaiser Permanente, Oakland, California
| | - Nuttha Lumlertgul
- Excellence Center for Critical Care Nephrology, Division of Nephrology, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - John C. Marshall
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada
| | - David K. Menon
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Nuala J. Meyer
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Elizabeth S. Munroe
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Sheila N. Myatra
- Department of Anaesthesiology, Critical Care and Pain, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Marlies Ostermann
- King’s College London, Guy’s & St Thomas’ Hospital, London, United Kingdom
| | - Hallie C. Prescott
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
- Veterans Affairs Center for Clinical Management Research, Ann Arbor, Michigan
| | - Adrienne G. Randolph
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Boston, Massachusetts
- Department of Anaesthesia and
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - Edward J. Schenck
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Christopher W. Seymour
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Manu Shankar-Hari
- Centre for Inflammation Research, Institute of Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| | - Mervyn Singer
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, United Kingdom
| | | | - Aiko Tanaka
- Department of Intensive Care, University of Fukui Hospital, Yoshida, Fukui, Japan
- Department of Anesthesiology and Intensive Care Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Fabio S. Taccone
- Department des Soins Intensifs, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Brussels, Belgium; and
| | - B. Taylor Thompson
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Lisa K. Torres
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Tom van der Poll
- Center of Experimental and Molecular Medicine, and
- Division of Infectious Diseases, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Jean-Louis Vincent
- Department des Soins Intensifs, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Brussels, Belgium; and
| | - Carolyn S. Calfee
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, California
| |
Collapse
|
47
|
Llitjos JF, Carrol ED, Osuchowski MF, Bonneville M, Scicluna BP, Payen D, Randolph AG, Witte S, Rodriguez-Manzano J, François B. Enhancing sepsis biomarker development: key considerations from public and private perspectives. Crit Care 2024; 28:238. [PMID: 39003476 PMCID: PMC11246589 DOI: 10.1186/s13054-024-05032-9] [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: 05/03/2024] [Accepted: 07/10/2024] [Indexed: 07/15/2024] Open
Abstract
Implementation of biomarkers in sepsis and septic shock in emergency situations, remains highly challenging. This viewpoint arose from a public-private 3-day workshop aiming to facilitate the transition of sepsis biomarkers into clinical practice. The authors consist of international academic researchers and clinician-scientists and industry experts who gathered (i) to identify current obstacles impeding biomarker research in sepsis, (ii) to outline the important milestones of the critical path of biomarker development and (iii) to discuss novel avenues in biomarker discovery and implementation. To define more appropriately the potential place of biomarkers in sepsis, a better understanding of sepsis pathophysiology is mandatory, in particular the sepsis patient's trajectory from the early inflammatory onset to the late persisting immunosuppression phase. This time-varying host response urges to develop time-resolved test to characterize persistence of immunological dysfunctions. Furthermore, age-related difference has to be considered between adult and paediatric septic patients. In this context, numerous barriers to biomarker adoption in practice, such as lack of consensus about diagnostic performances, the absence of strict recommendations for sepsis biomarker development, cost and resources implications, methodological validation challenges or limited awareness and education have been identified. Biomarker-guided interventions for sepsis to identify patients that would benefit more from therapy, such as sTREM-1-guided Nangibotide treatment or Adrenomedullin-guided Enibarcimab treatment, appear promising but require further evaluation. Artificial intelligence also has great potential in the sepsis biomarker discovery field through capability to analyse high volume complex data and identify complex multiparametric patient endotypes or trajectories. To conclude, biomarker development in sepsis requires (i) a comprehensive and multidisciplinary approach employing the most advanced analytical tools, (ii) the creation of a platform that collaboratively merges scientific and commercial needs and (iii) the support of an expedited regulatory approval process.
Collapse
Affiliation(s)
- Jean-Francois Llitjos
- Open Innovation and Partnerships (OI&P), bioMérieux S.A., Marcy l'Etoile, France.
- Anesthesiology and Critical Care Medicine, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France.
| | - Enitan D Carrol
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool Institute of Infection Veterinary and Ecological Sciences, Liverpool, UK
- Department of Paediatric Infectious Diseases and Immunology, Alder Hey Children's NHS Foundation Trust, Liverpool, UK
| | - Marcin F Osuchowski
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria
| | - Marc Bonneville
- Medical and Scientific Affairs, Institut Mérieux, Lyon, France
| | - Brendon P Scicluna
- Department of Applied Biomedical Science, Faculty of Health Sciences, Mater Dei Hospital, University of Malta, Msida, Malta
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | - Didier Payen
- Paris 7 University Denis Diderot, Paris Sorbonne, Cité, France
| | - Adrienne G Randolph
- Departments of Anaesthesia and Pediatrics, Harvard Medical School, Boston, MA, USA
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, USA
| | | | | | - Bruno François
- Medical-Surgical Intensive Care Unit, Réanimation Polyvalente, Dupuytren University Hospital, CHU de Limoges, 2 Avenue Martin Luther King, 87042, Limoges Cedex, France.
- Inserm CIC 1435, Dupuytren University Hospital, Limoges, France.
- Inserm UMR 1092, Medicine Faculty, University of Limoges, Limoges, France.
| |
Collapse
|
48
|
Angus DC, Huang AJ, Lewis RJ, Abernethy AP, Califf RM, Landray M, Kass N, Bibbins-Domingo K. The Integration of Clinical Trials With the Practice of Medicine: Repairing a House Divided. JAMA 2024; 332:153-162. [PMID: 38829654 PMCID: PMC12045079 DOI: 10.1001/jama.2024.4088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
Importance Optimal health care delivery, both now and in the future, requires a continuous loop of knowledge generation, dissemination, and uptake on how best to provide care, not just determining what interventions work but also how best to ensure they are provided to those who need them. The randomized clinical trial (RCT) is the most rigorous instrument to determine what works in health care. However, major issues with both the clinical trials enterprise and the lack of integration of clinical trials with health care delivery compromise medicine's ability to best serve society. Observations In most resource-rich countries, the clinical trials and health care delivery enterprises function as separate entities, with siloed goals, infrastructure, and incentives. Consequently, RCTs are often poorly relevant and responsive to the needs of patients and those responsible for care delivery. At the same time, health care delivery systems are often disengaged from clinical trials and fail to rapidly incorporate knowledge generated from RCTs into practice. Though longstanding, these issues are more pressing given the lessons learned from the COVID-19 pandemic, heightened awareness of the disproportionate impact of poor access to optimal care on vulnerable populations, and the unprecedented opportunity for improvement offered by the digital revolution in health care. Four major areas must be improved. First, especially in the US, greater clarity is required to ensure appropriate regulation and oversight of implementation science, quality improvement, embedded clinical trials, and learning health systems. Second, greater adoption is required of study designs that improve statistical and logistical efficiency and lower the burden on participants and clinicians, allowing trials to be smarter, safer, and faster. Third, RCTs could be considerably more responsive and efficient if they were better integrated with electronic health records. However, this advance first requires greater adoption of standards and processes designed to ensure health data are adequately reliable and accurate and capable of being transferred responsibly and efficiently across platforms and organizations. Fourth, tackling the problems described above requires alignment of stakeholders in the clinical trials and health care delivery enterprises through financial and nonfinancial incentives, which could be enabled by new legislation. Solutions exist for each of these problems, and there are examples of success for each, but there is a failure to implement at adequate scale. Conclusions and Relevance The gulf between current care and that which could be delivered has arguably never been wider. A key contributor is that the 2 limbs of knowledge generation and implementation-the clinical trials and health care delivery enterprises-operate as a house divided. Better integration of these 2 worlds is key to accelerated improvement in health care delivery.
Collapse
Affiliation(s)
- Derek C Angus
- JAMA,Chicago, IL
- University of Pittsburgh Schools of the Health Sciences, Pittsburgh, PA
| | | | - Roger J Lewis
- JAMA,Chicago, IL
- University of California, Los Angeles, CA
| | | | | | - Martin Landray
- Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
- Protas, Manchester, United Kingdom
| | | | | |
Collapse
|
49
|
Cook D, Taneja S, Krewulak K, Zytaruk N, Menon K, Fowler R, Lamontagne F, Kho ME, Rochwerg B, Masse MH, Lauzier F, O’Hearn K, Adhikari NKJ, Burns KEA, Bosma KJ, English S, McNally D, Turgeon AF, Brochard L, Parker M, Clayton L, Rishu A, Tuttle A, Daneman N, Fergusson D, McIntyre L, Kelly L, Orr S, Austin P, Mulligan S, Fiest K. Barriers, Solutions, and Opportunities for Adapting Critical Care Clinical Trials in the COVID-19 Pandemic. JAMA Netw Open 2024; 7:e2420458. [PMID: 38995645 PMCID: PMC11245722 DOI: 10.1001/jamanetworkopen.2024.20458] [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: 04/02/2024] [Accepted: 04/29/2024] [Indexed: 07/13/2024] Open
Abstract
Importance The COVID-19 pandemic created unprecedented challenges for clinical trials worldwide, threatening premature closure and trial integrity. Every phase of research operations was affected, often requiring modifications to protocol design and implementation. Objectives To identify the barriers, solutions, and opportunities associated with continuing critical care trials that were interrupted during the pandemic, and to generate suggestions for future trials. Design, Setting, and Participants This mixed-methods study performed an explanatory sequential analysis involving a self-administered electronic survey and focus groups of principal investigators (PIs) and project coordinators (PCs) conducting adult and pediatric individual-patient randomized trials of the Canadian Critical Care Trials Group during the COVID-19 pandemic. Eligible trials were actively enrolling patients on March 11, 2020. Data were analyzed between September 2023 and January 2024. Main Outcomes and Measures Importance ratings of barriers to trial conduct and completion, solutions employed, opportunities arising, and suggested strategies for future trials. Quantitative data examining barriers were analyzed using descriptive statistics. Data addressing solutions, opportunities, and suggestions were analyzed by qualitative content analysis. Integration involved triangulation of data sources and perspectives about 13 trials, synthesized by an interprofessional team incorporating reflexivity and member-checking. Results A total of 13 trials run by 29 PIs and PCs (100% participation rate) were included. The highest-rated barriers (on a 5-point scale) to ongoing conduct during the pandemic were decisions to pause all clinical research (mean [SD] score, 4.7 [0.8]), focus on COVID-19 studies (mean [SD] score, 4.6 [0.8]), and restricted family presence in hospitals (mean [SD] score, 4.1 [0.8]). Suggestions to enable trial progress and completion included providing scientific leadership, implementing technology for communication and data management, facilitating the informed consent process, adapting the protocol as necessary, fostering site engagement, initiating new sites, streamlining ethics and contract review, and designing nested studies. The pandemic necessitated new funding opportunities to sustain trial enrollment. It increased public awareness of critical illness and the importance of randomized trial evidence. Conclusions and Relevance While underscoring the vital role of research in society and drawing the scientific community together with a common purpose, the pandemic signaled the need for innovation to ensure the rigor and completion of ongoing trials. Lessons learned to optimize research procedures will help to ensure a vibrant clinical trials enterprise in the future.
Collapse
Affiliation(s)
- Deborah Cook
- Departments of Medicine, Health Research Evidence & Impact, McMaster University, Hamilton, Canada
| | - Shipra Taneja
- Department of Health Research Evidence & Impact, McMaster University, Hamilton, Canada
| | - Karla Krewulak
- Department of Critical Care, University of Calgary, Calgary, Alberta, Canada
| | - Nicole Zytaruk
- Department of Health Research Evidence & Impact, McMaster University, Hamilton, Canada
| | - Kusum Menon
- Department of Pediatrics, University of Ottawa, Ottawa, Canada
| | - Rob Fowler
- Interdepartmental Division of Critical Care, University of Toronto, Toronto, Canada
| | | | - Michelle E. Kho
- School of Rehabilitation Science, McMaster University, Hamilton, Canada
| | - Bram Rochwerg
- Departments of Medicine, Health Research Evidence & Impact, McMaster University, Hamilton, Canada
| | - Marie-Hélène Masse
- Centre de recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Canada
| | - François Lauzier
- Department of Anesthesiology and Critical Care Medicine, Division of Critical Care Medicine, Université Laval, Québec City, Canada
- CHU de Québec-Université Laval Research Center, Québec City, Canada
| | - Katie O’Hearn
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, Canada
| | - Neill K. J. Adhikari
- Interdepartmental Division of Critical Care, University of Toronto, Toronto, Canada
| | - Karen E. A. Burns
- Interdepartmental Division of Critical Care, University of Toronto, Toronto, Canada
- Keenan Research Center, Li Ka Shing Knowledge Institute, Unity Health Toronto, Toronto, Ontario
| | - Karen J. Bosma
- Department of Medicine, Division of Critical Care Medicine, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Canada
| | - Shane English
- Department of Medicine (Critical Care), University of Ottawa, Ottawa, Canada and Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Dayre McNally
- Department of Pediatrics, University of Ottawa, Ottawa, Canada
| | - Alexis F. Turgeon
- Department of Anesthesiology and Critical Care Medicine, Division of Critical Care Medicine, Université Laval, Québec City, Canada
- CHU de Québec-Université Laval Research Center, Québec City, Canada
| | - Laurent Brochard
- Interdepartmental Division of Critical Care, University of Toronto, Toronto, Canada
- Keenan Research Center, Li Ka Shing Knowledge Institute, Unity Health Toronto, Toronto, Ontario
| | - Melissa Parker
- Departments of Pediatrics and Emergency Medicine, McMaster University, Hamilton, Canada
| | - Lucy Clayton
- CHU de Québec-Université Laval Research Center, Québec City, Canada
| | - Asgar Rishu
- Interdepartmental Division of Critical Care, University of Toronto, Toronto, Canada
| | - Angie Tuttle
- Ottawa Hospital Research Institute, Ottawa, Canada
| | - Nick Daneman
- Division of Infectious Diseases, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | - Dean Fergusson
- Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Canada
| | | | | | - Sherrie Orr
- Department of Pediatrics, McMaster University, Hamilton, Canada
| | - Peggy Austin
- Departments of Medicine, Health Research Evidence & Impact, McMaster University, Hamilton, Canada
| | - Sorcha Mulligan
- Applied Health Research Centre, Li Ka Shing Knowledge Institute, Unity Health Toronto, St Michael’s Hospital, Toronto, Ontario, Canada
| | - Kirsten Fiest
- Department of Critical Care, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
50
|
Xiong W, Roy J, Liu H, Hu L. Leveraging machine learning: Covariate-adjusted Bayesian adaptive randomization and subgroup discovery in multi-arm survival trials. Contemp Clin Trials 2024; 142:107547. [PMID: 38688389 DOI: 10.1016/j.cct.2024.107547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 04/17/2024] [Accepted: 04/25/2024] [Indexed: 05/02/2024]
Abstract
Clinical trials evaluate the safety and efficacy of treatments for specific diseases. Ensuring these studies are well-powered is crucial for identifying superior treatments. With the rise of personalized medicine, treatment efficacy may vary based on biomarker profiles. However, researchers often lack prior knowledge about which biomarkers are linked to varied treatment effects. Fixed or response-adaptive designs may not sufficiently account for heterogeneous patient characteristics, such as genetic diversity, potentially reducing the chance of selecting the optimal treatment for individuals. Recent advances in Bayesian nonparametric modeling pave the way for innovative trial designs that not only maintain robust power but also offer the flexibility to identify subgroups deriving greater benefits from specific treatments. Building on this inspiration, we introduce a Bayesian adaptive design for multi-arm trials focusing on time-to-event endpoints. We introduce a covariate-adjusted response adaptive randomization, updating treatment allocation probabilities grounded on causal effect estimates using a random intercept accelerated failure time BART model. After the trial concludes, we suggest employing a multi-response decision tree to pinpoint subgroups with varying treatment impacts. The performance of our design is then assessed via comprehensive simulations.
Collapse
Affiliation(s)
- Wenxuan Xiong
- Department of Biostatistics and Epidemiology, Rutgers University School of Public Health, Piscataway, NJ, USA.
| | - Jason Roy
- Department of Biostatistics and Epidemiology, Rutgers University School of Public Health, Piscataway, NJ, USA
| | - Hao Liu
- Department of Biostatistics and Epidemiology, Rutgers University School of Public Health, Piscataway, NJ, USA; Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - Liangyuan Hu
- Department of Biostatistics and Epidemiology, Rutgers University School of Public Health, Piscataway, NJ, USA
| |
Collapse
|