Abstract
Background Whether ivermectin, with a maximum targeted dose of 600 μg/kg, shortens symptom duration or prevents hospitalization among outpatients with mild to moderate coronavirus disease 2019 (COVID-19) remains unknown. Our objective was to evaluate the effectiveness of ivermectin, dosed at 600 μg/kg, daily for 6 days compared with placebo for the treatment of early mild to moderate COVID-19.
Methods ACTIV-6, an ongoing, decentralized, randomized, double-blind, placebo-controlled, platform trial, was designed to evaluate repurposed therapies in outpatients with mild to moderate COVID-19. A total of 1206 participants age ≥30 years with confirmed COVID-19, experiencing ≥2 symptoms of acute infection for ≤7 days, were enrolled from February 16, 2022, through July 22, 2022, with follow-up data through November 10, 2022, at 93 sites in the US. Participants were randomized to ivermectin, with a maximum targeted dose of 600 μg/kg (n=602), daily vs. placebo daily (n=604) for 6 days. The primary outcome was time to sustained recovery, defined as at least 3 consecutive days without symptoms. The 7 secondary outcomes included a composite of hospitalization, death, or urgent/emergent care utilization by day 28.
Results Among 1206 randomized participants who received study medication or placebo, median (interquartile range) age was 48 (38–58) years; 713 (59%) were women; and 1008 (84%) reported ≥2 SARS-CoV-2 vaccine doses. Median time to recovery was 11 (11–12) days in the ivermectin group and 11 (11–12) days in the placebo group. The hazard ratio (HR) (95% credible interval [CrI], posterior probability of benefit) for improvement in time to recovery was 1.02 (0.92–1.13; P[HR>1]=0.68). In those receiving ivermectin, 34 (5.7%) were hospitalized, died, or had urgent or emergency care visits compared with 36 (6.0%) receiving placebo (HR 1.0, 0.6– 1.5; P[HR<1]=0.53). In the ivermectin group, 1 participant died and 4 were hospitalized (0.8%); 2 participants (0.3%) were hospitalized in the placebo group and there were no deaths. Adverse events were uncommon in both groups.
Conclusions Among outpatients with mild to moderate COVID-19, treatment with ivermectin, with a maximum targeted dose of 600 μg/kg daily for 6 days, compared with placebo did not improve time to recovery. These findings do not support the use of ivermectin in patients with mild to moderate COVID-19.
Trial registration ClinicalTrials.gov Identifier: NCT04885530.
INTRODUCTION
Despite treatment advances for coronavirus disease 2019 (COVID-19), the evolution of SARS-CoV-2 variants and subvariants has shifted therapeutic options with loss of effectiveness of monoclonal antibodies. Novel oral antivirals have been authorized for high-risk individuals in high-income countries.1,2 However, efficacy of these antivirals in those vaccinated or with prior infection remains unclear. Interest remains for the potential of repurposed drugs to improve symptoms and clinical outcomes in patients with COVID-19.
Numerous repurposed drugs have been investigated for COVID-19, with several large, randomized outpatient trials published.3–5 Trial results have been mixed. Some suggest promise to reduce emergency department (ED) visits or hospitalizations for drugs including fluvoxamine at 100 mg twice daily3 and immediate-release metformin.6 Others have failed to show a reduction in emergency department visits or hospitalizations such as fluvoxamine 50 mg twice daily.6 While recently completed trials benefit from increasing representation of vaccinated people, which is more relevant to the pandemic’s current state, the results have not impacted treatment guidelines due to study design and regulatory requirements.7–9
Ivermectin, an anti-parasitic drug used worldwide for onchocerciasis and strongyloidiasis, emerged in 2020 as a potential repurposed drug for COVID-19 informed by an in vitro study suggesting possible anti-viral activity.10 The interest for ivermectin as a COVID-19 therapeutic has remained high and, while there have been numerous ivermectin studies, its use has become controversial due to a lack of high-quality, adequately powered randomized trials and article retractions of some of the earlier and most positive studies.11–14 Three large randomized outpatient trials in people with symptomatic mild or moderate COVID-19 failed to identify a clinical benefit of ivermectin when dosed at 400 μg/kg daily for 3 days.15–17 One possibility is that the dose and duration studied was too low and too short, missing the therapeutic window for ivermectin. A proof-of-concept randomized controlled clinical trial in hospitalized patients showed that ivermectin, 600 μg/kg, for 5 days suggested possible anti-viral activity with increasing dose.17,18 For this reason we tested ivermectin, with a maximum targeted dose of 600 μg/kg, daily for 6 days on the Accelerating COVID-19 Therapeutic Interventions and Vaccines (ACTIV-6) platform from February 16, 2022 through July 22, 2022.
ACTIV-6 is an ongoing, fully remote (decentralized), double-blind, randomized, placebo-controlled, platform trial investigating repurposed drugs for the treatment of mild to moderate COVID-19 in the outpatient setting. This report describes the effectiveness of this dose and duration of ivermectin compared with blinded placebo for the treatment of early mild to moderate COVID-19.
METHODS
Trial Design and Oversight
This is a double-blind, randomized, placebo-controlled platform protocol designed to be flexible, allowing enrollment across a wide range of settings within healthcare systems, the community, and virtually. The platform enrolls outpatients with mild to moderate COVID-19 with a confirmed positive SARS-CoV-2 test. The full trial protocol and statistical analysis plan are available in Supplement 1 and Supplement 2, respectively.
The protocol was approved by each site’s institutional review board. Participants provided informed consent either via written consent or an electronic consent process. An independent data monitoring committee oversaw safety and trial performance.
Participants
Recruitment into the platform trial opened on June 11, 2021. Ivermectin 600 μg/kg was included on the platform beginning February 16, 2022. Enrollment into the ivermectin 600 μg/kg group was stopped on July 22, 2022, when 1206 participants had received their study drug, identical matched-placebo, or contributing-placebo. Participants were either identified by sites or self-identified by contacting a central study telephone hotline or website.
Study staff verified eligibility criteria including age ≥30 years, SARS-CoV-2 infection within 10 days (positive polymerase chain reaction [PCR] or antigen, including home-based test), and experiencing >2 symptoms of acute COVID-19 for ≤7 days from enrollment. Symptoms could include fatigue, dyspnea, fever, cough, nausea, vomiting, diarrhea, body aches, chills, headache, sore throat, nasal symptoms, and loss of sense of taste or smell. Exclusion criteria included hospitalization, study drug use within 14 days, or known allergy or contraindication to study drug (Supplement 1). Vaccination was allowable, as were standard of care therapies for COVID-19 available under US Food and Drug Administration (FDA) authorization or approval.
Randomization
Participants were randomized, using a random number generator, in a 2-step process (Figure 1). First, participants were randomized to active agent or placebo in a ratio of m:1, where m is the number of study drugs for which the participant was eligible; the other study drug under investigation during this period was fluvoxamine, 50 mg twice daily for 10 days. Participants could choose to opt out of specific study drug groups if they or the site investigator did not feel there was equipoise, or a contraindication existed. After randomization to active agent versus placebo, participants were randomized with equal probability among the study drugs for which they were eligible. The more study drugs a participant was eligible for, the greater the chance of receiving an active agent. Participants eligible for both ivermectin and fluvoxamine 50 mg but randomized to fluvoxamine-matched placebo were included and contributed to the placebo group for ivermectin.
CONSORT diagram
Interventions
A central pharmacy supplied ivermectin or placebo to participants via direct home delivery. Ivermectin was supplied as a bottle of 7-mg tablets. Participants were instructed to take a pre-specified number of tablets for 6 consecutive days based on their weight for a maximum daily dose of approximately 600 μg/kg. The dosing schedule was based on weight (kg) ranges as follows: 35-52, 53-69, 70-89, 90-109, 110-129, and >129. This schedule resulted in a range of doses from 400-600 μg/kg (eFigure 1). Packaging for matched placebo was identical to ivermectin while packaging for the contributing placebos was identical to that of the associated study drug, which in this case was fluvoxamine 50mg.
Outcome Measures
The primary measure of effectiveness was time to sustained recovery, defined as the number of days between study drug receipt and the third of 3 consecutive days without symptoms. This was selected a priori from among the 2 co-primary endpoints that remain available to other study drugs in the platform (Supplement 2). The key secondary outcome was the composite of hospitalization or death by day 28. Other secondary outcomes included mean time unwell estimated from a longitudinal ordinal model; COVID-19 Clinical Progression Scale on days 7, 14, and 28; mortality through day 28; and the composite of urgent or emergency care visit, hospitalization, or death through day 28.
Trial Procedures
The study was designed as a fully remote, or decentralized, trial. Screening and eligibility confirmation were participant-reported and site confirmed. A positive SARS-CoV-2 PCR or antigen test result was verified prior to randomization. At screening, participant-reported demographic information was collected and included race and ethnicity, eligibility criteria, medical history, concomitant medications, symptom reporting, and quality of life questionnaires.
A central investigational pharmacy distributed study drug (either active or placebo) using a next-day priority shipping service. Delivery was tracked, delayed deliveries followed-up on, and participants must have received study drug within 7 days of enrollment to be included. Confirmation that the study drug was delivered to the participant’s address was required for the participant to be included in the analysis. Receipt of study drug was defined as study day 1.
We asked participants to complete daily assessments and report adverse events through day 14. Assessments included symptoms and severity, health care visits, and medications. If symptoms were still ongoing at day 14, daily surveys continued until participants experienced 3 consecutive days without symptoms or until day 28. At days 28 and 90, all participants completed assessments. Supplement 1 presents survey details. Additional details are available in Supplement 3.
Statistical Analysis Plan
This platform trial was designed to be analyzed accepting the possibility of adding and dropping groups as the trial progresses. The general analytical approach was regression modelling. We utilized proportional hazard regression for time-to-event analysis and cumulative probability ordinal regression models for ordinal outcomes. In addition, we estimated mean time spent unwell using a longitudinal ordinal regression model as a quantification of benefit (Supplement 2).
The planned primary endpoint analysis was a Bayesian proportional hazards model for time to sustained recovery. The primary inferential (decision-making) quantity was the posterior distribution for the treatment assignment hazard ratio (HR), with HR>1 indicating faster recovery. If at any of the interim or final analyses, the posterior probability of benefit exceeded 0.95 (i.e. ≥95% probability of benefit), the trial would conclude efficacy of ivermectin. To preserve type 1 error <0.05, the prior for the treatment effect parameter (on the loge relative hazard scale) was a normal distribution centered at 0 and scaled to a standard deviation of 0.1. All other parameter priors were non-informative, using the software default of 2.5 times the ratio of the standard deviation of the outcome divided by the standard deviation of the predictor variable. The study design was estimated to have 80% power to detect a HR of 1.2 in the primary endpoint with approximately 1200 participants. To achieve this sample size in an ongoing platform trial, once 1200 participants had been randomized to the study group or matching placebo and had received study drug, the study arm became unavailable for new participants expressing interest in the platform. Some participants had already consented to participate but had not yet been randomized or received study drug at the time of arm closure, and these participants were allowed to continue as assigned.
The primary endpoint adjusted model included the following predictor variables in addition to randomization assignment: age (as restricted cubic spline), sex, duration of symptoms at study drug receipt, calendar time (as restricted cubic spline), vaccination status, geographic region (Northeast, Midwest, South, West), call center enrollment, and day 1 symptom severity.
This adjusted model was pre-specified. The proportional hazards assumption of the primary endpoint was evaluated by generating visual diagnostics such as the log-log plot and plots of time-dependent regression coefficients for each predictor in the model, a diagnostic which indicates deviations from proportionality if the time-dependent coefficients are not constant in time.
Secondary endpoints were analyzed with Bayesian regression models (either proportional hazards or proportional odds) using non-informative priors for all parameters. Secondary endpoints were not used for formal decision making, and no decision threshold was selected. Due to potential for inflated type 1 error due to multiple comparisons, secondary endpoints should be interpreted as exploratory. The same covariates used in the primary endpoint model were used in the adjusted analysis of secondary endpoints, provided the endpoint accrued enough events to be analyzed with covariate adjustment.
As a platform trial, the primary analysis is implemented separately for each study drug, where the placebo group consists of contemporaneously randomized participants who met the eligibility criteria for that study drug; this includes both matched and contributing placebo. From other remote trials,3,19 we recognized that study drug delivery may not always occur (e.g., failure of delivery, participant withdrawal, or interval hospitalization). For ACTIV-6, the modified-intent-to-treat (mITT) analysis set for the primary analyses included all participants who received study drug, and participants were analyzed as assigned. We used all available data to compare ivermectin versus placebo, regardless of post-randomization adherence. In both the primary and secondary endpoint analyses, missing data among covariates used for adjustment was addressed with conditional mean imputation as the amount of missing covariate data was minimal (<4%).
A pre-specified analysis tested for differential treatment effects as a function of pre-existing participant characteristics. Analysis of heterogeneity of treatment effect included: age, symptom duration, body mass index, day 1 symptom severity, calendar time (surrogate for SARS-CoV-2 variant), sex, and vaccination status; continuous variables were modelled as such without creating subgroups.
Analyses were performed with R 4.1 with primary packages of: rstanarm, rmsb, and survival.20
Additional details are available in Supplement 3.
RESULTS
Study Population
Of the 2212 participants who consented to be evaluated for inclusion in the ivermectin group, 1206 were eligible for ivermectin, randomized to ivermectin, with a targeted maximum dose of 600 μg/kg, (n=602) or placebo (n=604); and received study drug (Figure 1). Of participants receiving placebo, 543 (90%) received matching placebo and 61 (10%) received placebo as part of the contributing placebo group.
The median age of the participants was 48 years (IQR 38–58), and 46% were aged 50 years or older (Table 1). The population was 59% female, 7.7% identified as Black/African American, 75% identified as White, and 21.6% reported being of Latino/Hispanic ethnicity. Although not required for enrollment, high-risk comorbidities included body mass index >30 kg/m2 (38%), diabetes (9%), hypertension (27%), asthma (14%), and chronic obstructive pulmonary disease (2%). Overall, 84% of participants reported receiving ≥2 COVID-19 vaccine doses. Median time from symptom onset to enrollment was 3 days (IQR 2–5) and to study drug receipt was 5 days (IQR 3–7) with 60% receiving study drug within 5 days of symptom onset (eFigure 2). eTable 1 presents baseline symptom prevalence and severity. Receipt of FDA-authorized therapies was uncommon (remdesivir 0%, monoclonal antibody 3.9%, nirmatrelvir/ritonavir 3.4%, molnupiravir 0.5%).
Baseline characteristics
Primary Outcome
The median time to recovery was 11 days (IQR 11–12) in the ivermectin group and 11 days (IQR 11–12) in the placebo group. The posterior probability for benefit was 0.68 for the primary outcome of time to recovery with a HR of 1.02 (95% credible interval [CrI], 0.92–1.13) where HR>1 is for faster symptom resolution with ivermectin (Table 2, Figure 2A). This posterior probability for the primary outcome was below the prespecified threshold of 0.95 probability (Supplement 2). The data do not provide evidence of a treatment benefit even when using a Bayesian non-informative prior, no prior, with various approaches to imputing missing symptom data, or when restricting the analysis to participants who received drug within 2 or 3 days of symptom onset and across severity of symptoms reported on Day 1 (Table 2, Figure 3, eFigures 3 and 4). The probability that ivermectin reduced symptom duration by 24 hours was less than 0.1%.
Posterior Distributions of Effects of (A) Time to Sustained Recovery (n=1206); (B) Hospitalization, Urgent or Emergency Care Visits, or Death (n=1206); and (C) Mean Time Unwell (n=1205).
Thick vertical lines denote the estimated mean of the posterior distribution. Density is the relative likelihood of posterior probability distribution. Outcomes with higher density are more likely than outcomes with lower density.
Primary and secondary outcomes
Kaplan-Meier for Primary Outcome of Time to Sustained Recovery.
Recovery occurs on the third of 3 consecutive days without symptoms. Four participants were censored for nonresponse, and all others were followed up until recovery, death, or the end of short-term 28-day follow-up. Median (IQR) time to recovery was 11 days (11, 12) in the ivermectin group and 11 days (11, 12) in the placebo group.
Secondary Outcomes
Hospitalizations and deaths were uncommon, with 5 events (1 death not attributable to Covid-19 or treatment) in the ivermectin group and 2 events (no deaths) in the placebo group (eFigure 5A). Statistical comparisons were not done as they would be uninformative with so few events. The composite secondary outcome of urgent care or ED visits, hospitalizations, or death was not shown to differ with ivermectin (5.6% [34/602]) compared with placebo (6.0% [36/604]) (HR 1.0, 95% CrI 0.6–1.5, P(HR<1=0.53) (Table 2, Figure 2B, eFigure 5B). The difference in the amount of time spent feeling unwell with COVID-19 was estimated as 3 hours and 20 minutes faster with ivermectin (95% CrI, 12 hours better to 6 hours worse) than placebo (Figure 2C). The COVID Clinical Progression Scale at days 7, 14, and 28 did not meet prespecified thresholds for beneficial treatment effect (Supplement 3). For example, by day 7, 88% (532/602) of the ivermectin group and 91% (549/604) of the placebo group were not hospitalized and did not report limitation of activities (eFigure 6).
Heterogeneity of Treatment Effect Analyses
Interaction tests for heterogeneity of treatment effect showed no overall influence of the putative treatment effect modifiers even when all subgroup analyses across symptom severity were not adjusted for multiple comparisons (eFigure 7). The overall effect of timing from symptom onset to receipt of study drug was not significant (p=0.15 for heterogeneity). Similarly, no evidence existed for a different treatment effect of ivermectin compared with placebo for severity of symptoms, sex, age, body mass index, calendar time, or vaccination status.
Adverse Events
Among participants who reported taking study drug at least once, adverse events were similar in both groups (9.2% [52/566] with ivermectin and 7.1% [41/576] with placebo) (eTable 2). Adverse events reported >2 times only in the ivermectin group included cognitive impairment (n=4), blurred vision (n=5), light sensitivity to eye (n=5), photophobia (n=4), dizziness (n=5), and asthma (n=3). Serious adverse events were rare: 5 with ivermectin and 3 with placebo. The death in the ivermectin arm was accidental.
DISCUSSION
Among a largely vaccinated outpatient population with COVID-19, treatment with ivermectin, with a targeted maximum dose of 600 μg/kg, daily for 6 days compared with placebo was not shown to improve time to recovery in over 1200 participants in the US during omicron variant/subvariant circulation. No evidence of benefit was observed for secondary clinical outcomes including the composite of hospitalization, death, or acute care visits. Hospitalization and death were uncommon in this largely vaccinated population. These findings do not support the use of ivermectin in outpatients with COVID-19.
Multiple large, double-blind randomized controlled trials have failed to identify a clinically meaningful benefit of ivermectin when used at a targeted dose of 400 μg/kg daily for 3 days.6,16 This large clinical trial addresses a potential gap in knowledge by testing 1) a higher daily dose (targeted maximum dose of 600 μg/kg) and 2) longer 6-day duration of ivermectin,. Due to the lack of early phase studies or animal model studies to determine optimal dosing for a therapeutic drug, the appropriate dosing of ivermectin for COVID-19 was never determined. A combination of modeling studies and a proof-of-concept clinical study have suggested doses up to 600 μg/kg daily may achieve system levels sufficient for in vitro antiviral activity.17,18 Although a phase 2 trial testing ivermectin 600 μg/kg daily for 7 days and assessing a virologic endpoint of oropharyngeal SARS-CoV-2 PCR did not show measurable antiviral activity and was stopped for futility.21 This dose was safe and generally well tolerated, with a higher prevalence of the known self-resolving visual disturbances in the intervention arm previously reported with similar doses of ivermectin for parasitic infections.17,18
Recent results from the current platform trial reported participants receiving ivermectin, with a 400 μg/kg, daily for 3 days had a an average ∼12-hour shorter time spent unwell when compared with placebo, a secondary outcome for the trial.15 This finding was not replicated here. The notable difference in baseline characteristics between these 2 cohorts is the completed vaccination rate, which is 84% for this study and was 47% for the prior ivermectin 400 μg/kg group.15 Hospitalizations and COVID-19-related clinical events were less common in this largely vaccinated cohort. The incidence of acute care visits, hospitalizations, or death was similar with ivermectin (5.7%) and placebo (6.0%); a result also observed in the two prior ivermectin 400 μg/kg randomized trials in the US.6,15
This trial has several strengths. This was a double-blind, randomized, placebo-controlled nationwide trial with 93 enrolling sites and a call center that recruited participants from all 50 US states. The ivermectin 600 μg/kg group of the platform trial enrolled rapidly due to ongoing omicron variant/subvariant surges and largely included vaccinated people, thus representing a highly relevant study population that also addresses a weakness of many other studies that excluded vaccinated people.
Limitations
This study has limitations. Due to infrequent hospitalization, we cannot draw definitive inferences on whether ivermectin impacts hospitalization without much larger trials. Also, due to the remote nature of the trial, 60% of participants received study drug within 5 days of symptom onset. Most outpatient COVID-19 antiviral trials have limited enrollment to participants within 5 days of symptom onset.1,2 In this trial, we observed no evidence of a differential treatment effect based on shorter time to study drug receipt.
Conclusions
Among outpatients with mild or moderate COVID-19, treatment with ivermectin, with a targeted maximum dose of 600 μg/kg daily for 6 days, was not shown to improve time to recovery compared with placebo. These findings do not support the use of ivermectin in outpatients with COVID-19.
Data Availability
ACTIV-6 is a platform trial using shared placebos. On completion of the platform trial when there is no risk of unblinding across study arms the data will be made publicly available by depositing it in an approved data repository such as NHLBI BioData Catalyst.
Author Contributions
Drs Naggie and Hernandez had full access to all of the blinded data in the study. Drs Lindsell and Stewart directly accessed and verified the underlying study data and take responsibility for the integrity of the data and the accuracy of the data analysis. All authors contributed to the drafting and review of the manuscript and agreed to submit for publication.
Disclosures
Naggie: Reports grants from NIH, the sponsor for this study, during the conduct of the study; Institutional research grants from Gilead Sciences, AbbVie; Consulting fees from Pardes Biosciences; Scientific advisor/Stock options from Vir Biotechnology; Consulting with no financial payment from Silverback Therapeutics; DSMB fees from Personal Health Insights, Inc; Event adjudication committee fees from BMS/PRA outside the submitted work.
Boulware: Reports grants from NIH during the conduct of the study.
Lindsell: Reports institutional grants from NCATS during the conduct of the study; Institutional grants from NIH, CDC, and DoD; Contract with institution for research services from Endpoint Health, bioMerieux, Entegrion Inc, Abbvie, and Astra Zeneca outside the submitted work; Dr Lindsell has a patent for risk stratification in sepsis and septic shock issued to Cincinnati Children’s Hospital Medical Center.
Stewart: Reports grants from NIH NCATS during the conduct of the study; Grants from NIH outside the submitted work.
Lim: NIH funding for this study, CDC funding for other studies, Integrated Neurologics for phase 2 device trial.
Cohen: Nothing to report.
Kavtaradze: Nothing to report.
Amon: Nothing to report.
Gabriel: Nothing to report.
Gentile: Reports personal fees from Duke University for protocol development and oversight during the conduct of the study; grants from NIH outside the submitted work.
Felker: Reports institutional research grants from NIH during the conduct of the study and from Novartis outside the submitted work.
Rothman: Reports institutional research grants from NIH, PCORI, AHRQ, and CDC during the conduct of the study. Spouse owns stock in Moderna unrelated to the current work.
Jayaweera: Reports grants from NCATS PI-Ralph Sacco during the conduct of the study; Grants from Gilead, Pfizer, Janssen, and Viiv; Consulting fees from Theratechnologies outside the submitted work.
McCarthy: Nothing to report.
Sulkowski: Reports advisory board fees from AbbVie, Gilead, GSK, Atea, Antios, Precision Bio, Viiv, and Virion; Institutional grants from Janssen outside the submitted work.
Wilson: Reports institutional research funding from the National Center for Advancing Translational Sciences (3U24TR001608) during the conduct of the study.
DeLong: Reports institutional research funding from the National Center for Advancing Translational Sciences (3U24TR001608) during the conduct of the study.
Remaly: Reports institutional research funding from the National Center for Advancing Translational Sciences (3U24TR001608) during the conduct of the study.
Wilder: Reports institutional research funding from the National Center for Advancing Translational Sciences (3U24TR001608) during the conduct of the study.
Collins: Reports grant funding from NHLBI and personal fees from Vir Biotechnology during the conduct of the study.
Dunsmore: Nothing to report.
Adam: Reports other from US Government Funding through Operation Warp Speed during the conduct of the study.
Thicklin: Nothing to report.
Hanna: Reports grants from US Biomedical Advanced Research & Development Authority contract to Tunnell Government Services for consulting services during the conduct of the study; Personal fees from Merck & Co. and AbPro outside the submitted work.
Ginde: Reports grants from NIH during the conduct of the study; Grants from NIH, CDC, DoD, AbbVie (investigator-initiated), and Faron Pharmaceuticals (investigator-initiated) outside the submitted work.
Castro: Reports institutional grant funding from NIH, ALA, PCORI, AstraZeneca, GSK, Novartis, Pulmatrix, Sanofi-Aventis, Shionogi; Speaker/Consultant fees from Grant Funding, Genentech, Teva, Sanofi-Aventis; Consultant fees from Merck, Novartis, Arrowhead, OM Pharma, Allakos; Speaker honorarium from Amgen, AstraZeneca, GSK, Regeneron; Royalties from Elsevier all outside the submitted work.
McTigue: Reports grants from NIH Research Subcontract to the University of Pittsburgh during the conduct of the study; Research contract to the University of Pittsburgh from Pfizer, and Janssen outside the submitted work.
Shenkman: Nothing to report.
Hernandez: Reports grants from American Regent, Amgen, Boehringer Ingelheim, Merck, Verily, Somologic, and Pfizer; Personal fees from AstraZeneca, Boston Scientific, Cytokinetics, Bristol Myers Squibb, and Merck outside the submitted work.
Data Sharing Statement
Available in the Supplement to the ACTIV-6 Statistical Analysis Plan (Supplement 4).
Funding
ACTIV-6 is funded by the National Center for Advancing Translational Sciences (NCATS) (3U24TR001608-06S1). Additional support for this study was provided by the Office of the Assistant Secretary for Preparedness and Response, Biomedical Advanced Research and Development Authority (Contract No.75A50122C00037). The Vanderbilt University Medical Center Clinical and Translational Science Award from NCATS (UL1TR002243) supported the REDCap infrastructure.
Role of the Sponsor
NCATS participated in the design and conduct of the study; collection, management, analysis, and interpretation of the data; preparation, review, or approval of the manuscript; and decision to submit the manuscript for publication.
Acknowledgments
We thank Samuel Bozzette, MD, PhD, and Eugene Passamani, MD, both of the National Center for Advancing Translational Sciences (NCATS), for their roles in the trial design and protocol development. We also thank the ACTIV-6 Data Monitoring Committee and Clinical Events Committee Members (listed below) for their contributions.
Data Monitoring Committee: Clyde Yancy, MD, MSc, Northwestern University Feinberg School of Medicine; Adaora Adimora, MD, University of North Carolina, Chapel Hill; Susan Ellenberg, PhD, University of Pennsylvania; Kaleab Abebe, PhD, University of Pittsburgh; Arthur Kim, MD, Massachusetts General Hospital; John D. Lantos, MD, Children’s Mercy Hospital; Jennifer Silvey-Cason, Participant representative; Frank Rockhold, PhD, Duke Clinical Research Institute; Sean O’Brien, PhD, Duke Clinical Research Institute; Frank Harrell, PhD, Vanderbilt University Medical Center; Zhen Huang, MS, Duke Clinical Research Institute.
Clinical Events Committee: Renato Lopes, MD, PhD, MHS, W. Schuyler Jones, MD, Antonio Gutierrez, MD, Robert Harrison, MD, David Kong, MD, Robert McGarrah, MD, Michelle Kelsey, MD, Konstantin Krychtiuk, MD, Vishal Rao, MD all of the Duke Clinical Research Institute, Duke University School of Medicine.