1
|
Rhodes P, Parry PI. Pharmaceutical product recall and educated hesitancy towards new drugs and novel vaccines. INTERNATIONAL JOURNAL OF RISK & SAFETY IN MEDICINE 2024; 35:317-333. [PMID: 39973420 DOI: 10.1177/09246479241292008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Background: Of many pharmaceutical products launched for the benefit of humanity, a significant number have had to be recalled from the marketplace due to adverse events. A systematic review found market recalls for 462 pharmaceutical products between 1953 and 2013. In our current and remarkable period of medical history, excess mortality figures are high in many countries. Yet these statistics receive limited attention, often ignored or dismissed by mainstream news outlets. This excess mortality may include adverse effects caused by novel pharmaceutical agents that use gene-code technology.Objective: To examine key pharmaceutical product withdrawals and derive lessons that inform the current use of gene-based COVID-19 vaccines.Methods: Selective narrative review of historical pharmaceutical recalls and comparative issues with recent COVID-19 vaccines.Results: Parallels with past drug withdrawals and gene-based vaccines include distortion of clinical trial data, with critical adverse event data absent from high-impact journal publications. Delayed regulatory action on pharmacovigilance data to trigger market withdrawal occurred with Vioxx (rofecoxib) and is apparent with the gene-based COVID-19 vaccines.Conclusion: Public health requires access to raw clinical trial data, improved transparency from corporations and heightened, active pharmacovigilance worldwide.
Collapse
Affiliation(s)
- Peter Rhodes
- Gonville & Caius College, University of Cambridge, Cambridge, UK
- Anaesthesia and Intensive Care Medicine, Brisbane, QLD, Australia
| | - Peter I Parry
- Childrens Health Queensland Clinical Unit, Faculty of Medicine, University of Queensland, South Brisbane, QLD, Australia
- Department of Psychiatry, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| |
Collapse
|
2
|
Mead MN, Seneff S, Wolfinger R, Rose J, Denhaerynck K, Kirsch S, McCullough PA. COVID-19 mRNA Vaccines: Lessons Learned from the Registrational Trials and Global Vaccination Campaign. Cureus 2024; 16:e52876. [PMID: 38274635 PMCID: PMC10810638 DOI: 10.7759/cureus.52876] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2024] [Indexed: 01/27/2024] Open
Abstract
Our understanding of COVID-19 vaccinations and their impact on health and mortality has evolved substantially since the first vaccine rollouts. Published reports from the original randomized phase 3 trials concluded that the COVID-19 mRNA vaccines could greatly reduce COVID-19 symptoms. In the interim, problems with the methods, execution, and reporting of these pivotal trials have emerged. Re-analysis of the Pfizer trial data identified statistically significant increases in serious adverse events (SAEs) in the vaccine group. Numerous SAEs were identified following the Emergency Use Authorization (EUA), including death, cancer, cardiac events, and various autoimmune, hematological, reproductive, and neurological disorders. Furthermore, these products never underwent adequate safety and toxicological testing in accordance with previously established scientific standards. Among the other major topics addressed in this narrative review are the published analyses of serious harms to humans, quality control issues and process-related impurities, mechanisms underlying adverse events (AEs), the immunologic basis for vaccine inefficacy, and concerning mortality trends based on the registrational trial data. The risk-benefit imbalance substantiated by the evidence to date contraindicates further booster injections and suggests that, at a minimum, the mRNA injections should be removed from the childhood immunization program until proper safety and toxicological studies are conducted. Federal agency approval of the COVID-19 mRNA vaccines on a blanket-coverage population-wide basis had no support from an honest assessment of all relevant registrational data and commensurate consideration of risks versus benefits. Given the extensive, well-documented SAEs and unacceptably high harm-to-reward ratio, we urge governments to endorse a global moratorium on the modified mRNA products until all relevant questions pertaining to causality, residual DNA, and aberrant protein production are answered.
Collapse
Affiliation(s)
- M Nathaniel Mead
- Biology and Nutritional Epidemiology, Independent Research, Copper Hill, USA
| | - Stephanie Seneff
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, USA
| | - Russ Wolfinger
- Biostatistics and Epidemiology, Independent Research, Research Triangle Park, USA
| | - Jessica Rose
- Immunology and Public Health Research, Independent Research, Ottawa, CAN
| | - Kris Denhaerynck
- Epidemiology and Biostatistics, Independent Research, Basel, CHE
| | - Steve Kirsch
- Data Science, Independent Research, Los Angeles, USA
| | - Peter A McCullough
- Cardiology, Epidemiology, and Public Health, McCullough Foundation, Dallas, USA
- Cardiology, Epidemiology, and Public Health, Truth for Health Foundation, Tucson, USA
| |
Collapse
|
3
|
Tseng PT, Zeng BS, Thompson T, Stubbs B, Hsueh PR, Su KP, Chen YW, Chen TY, Wu YC, Lin PY, Carvalho AF, Hsu CW, Li DJ, Yeh TC, Sun CK, Cheng YS, Shiue YL, Liang CS, Tu YK. Placebo effects on all-cause mortality of patients with COVID-19 in randomized controlled trials of interleukin 6 antagonists: A systematic review and network meta-analysis. Psychiatry Clin Neurosci 2023; 77:638-645. [PMID: 37646204 DOI: 10.1111/pcn.13592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/14/2023] [Accepted: 08/23/2023] [Indexed: 09/01/2023]
Abstract
AIM Many randomized controlled trials (RCTs) have investigated the use of interleukin 6 antagonists for the treatment of coronavirus disease 2019 (COVID-19), yielding inconsistent results. This network meta-analysis (NMA) aimed to identify the source of these inconsistent results by reassessing whether participants treated with standard of care (SoC) plus placebo have different all-cause mortality from those treated with SoC alone and to reevaluate the efficacy of interleukin 6 antagonists in the treatment of COVID-19. METHODS We conducted a systematic search for relevant RCTs from the inception of electronic databases through 1 September 2022. The primary outcome was all-cause mortality. The secondary outcomes were the incidences of major medical events, secondary infections, all-cause discontinuation, and serious adverse events. RESULTS The results of NMA of 33 RCTs showed that patients with COVID-19 treated with SoC plus placebo had lower odds of all-cause mortality than those who received SoC alone (OR, 0.75 [95% confidence interval, 0.58-0.97]). This finding remained consistent after excluding studies with no incident deaths. In addition, when we consider the impact of the widely promoted COVID-19 vaccination and newly developed antiviral treatment strategy, the results from the analysis of the RCT published in 2021 and 2022 remained similar. CONCLUSION These findings suggest the potential influence of placebo effects on the treatment outcomes of COVID-19 in RCTs. When evaluating the efficacy of treatment strategies for COVID-19, it is crucial to consider the use of placebo in the design of clinical trials.
Collapse
Affiliation(s)
- Ping-Tao Tseng
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
- Department of Psychology, College of Medical and Health Science, Asia University, Taichung, Taiwan
- Prospect Clinic for Otorhinolaryngology & Neurology, Kaohsiung, Taiwan
- Institute of Precision Medicine, National Sun Yat-sen University, Kaohsiung City, Taiwan
| | - Bing-Syuan Zeng
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
- Department of Internal Medicine, E-Da Cancer Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Trevor Thompson
- Centre for Chronic Illness and Ageing, University of Greenwich, London, UK
| | - Brendon Stubbs
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Physiotherapy Department, South London and Maudsley NHS Foundation Trust, London, UK
- Faculty of Health, Social Care Medicine and Education, Anglia Ruskin University, Chelmsford, UK
| | - Po-Ren Hsueh
- Departments of Laboratory Medicine and Internal Medicine, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Kuan-Pin Su
- College of Medicine, China Medical University, Taichung, Taiwan
- Mind-Body Interface Laboratory (MBI-Lab), China Medical University and Hospital, Taichung, Taiwan
- An-Nan Hospital, China Medical University, Tainan, Taiwan
| | - Yen-Wen Chen
- Prospect Clinic for Otorhinolaryngology & Neurology, Kaohsiung, Taiwan
| | - Tien-Yu Chen
- Department of Psychiatry, Tri-Service General Hospital; School of Medicine, National Defense Medical Center, Taipei, Taiwan
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Cheng Wu
- Department of Sports Medicine, Landseed International Hospital, Taoyuan, Taiwan
| | - Pao-Yen Lin
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Institute for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Andre F Carvalho
- Innovation in Mental and Physical Health and Clinical Treatment (IMPACT) Strategic Research Centre, School of Medicine, Barwon Health, Deakin University, Geelong, Victoria, Australia
| | - Chih-Wei Hsu
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Dian-Jeng Li
- Department of Addiction Science, Kaohsiung Municipal Kai-Syuan Psychiatric Hospital, Kaohsiung City, Taiwan
| | - Ta-Chuan Yeh
- Department of Psychiatry, Tri-Service General Hospital; School of Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Cheuk-Kwan Sun
- Department of Emergency Medicine, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
- School of Medicine for International Students, College of Medicine, I-Shou University Kaohsiung, Kaohsiung, Taiwan
| | - Yu-Shian Cheng
- Department of Psychology, College of Medical and Health Science, Asia University, Taichung, Taiwan
- Department of Psychiatry, Tsyr-Huey Mental Hospital, Kaohsiung Jen-Ai's Home, Kaohsiung, Taiwan
| | - Yow-Ling Shiue
- Department of Internal Medicine, E-Da Cancer Hospital, I-Shou University, Kaohsiung, Taiwan
- Institute for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Chih-Sung Liang
- Department of Psychiatry, Beitou Branch, Tri-Service General Hospital; School of Medicine, National Defense Medical Center, Taipei, Taiwan
- Department of Psychiatry, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Kang Tu
- Institute of Health Data Analytics & Statistics, College of Public Health, National Taiwan University, Taipei, Taiwan
- Department of Dentistry, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
4
|
Dayan GH, Rouphael N, Walsh SR, Chen A, Grunenberg N, Allen M, Antony J, Asante KP, Bhate AS, Beresnev T, Bonaparte MI, Celle M, Ceregido MA, Corey L, Dobrianskyi D, Fu B, Grillet MH, Keshtkar-Jahromi M, Juraska M, Kee JJ, Kibuuka H, Koutsoukos M, Masotti R, Michael NL, Neuzil KM, Reynales H, Robb ML, Villagómez Martínez SM, Sawe F, Schuerman L, Tong T, Treanor J, Wartel TA, Diazgranados CA, Chicz RM, Gurunathan S, Savarino S, Sridhar S. Efficacy of a bivalent (D614 + B.1.351) SARS-CoV-2 recombinant protein vaccine with AS03 adjuvant in adults: a phase 3, parallel, randomised, modified double-blind, placebo-controlled trial. THE LANCET. RESPIRATORY MEDICINE 2023; 11:975-990. [PMID: 37716365 PMCID: PMC10872639 DOI: 10.1016/s2213-2600(23)00263-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/16/2023] [Accepted: 07/04/2023] [Indexed: 09/18/2023]
Abstract
BACKGROUND COVID-19 vaccines with alternative strain compositions are needed to provide broad protection against newly emergent SARS-CoV-2 variants of concern. This study aimed to describe the clinical efficacy and safety of a bivalent SARS-CoV-2 recombinant protein vaccine as a two-injection primary series during a period of circulation of the omicron (B.1.1.529) variant. METHODS We conducted a phase 3, parallel, randomised, modified double-blind, placebo-controlled trial in adults aged 18 years or older at 54 clinical research centres in eight countries (Colombia, Ghana, India, Kenya, Mexico, Nepal, Uganda, and Ukraine). Participants were recruited from the community and randomly assigned (1:1) by use of an interactive response technology system to receive two intramuscular 0·5 mL injections, 21 days apart, of the bivalent vaccine (5 μg of ancestral [D614] and 5 μg of beta [B.1.351] variant spike protein, with AS03 adjuvant) or placebo (0·9% normal saline). All participants, outcome assessors, and laboratory staff performing assays were masked to group assignments; those involved in the preparation and administration of the vaccines were unmasked. Participants were stratified by age (18-59 years and ≥60 years) and baseline SARS-CoV-2 rapid serodiagnostic test positivity. Symptomatic COVID-19 was defined as laboratory-confirmed (via nucleic acid amplification test or PCR test) COVID-19 with COVID-19-like illness symptoms. The primary efficacy endpoint was the clinical efficacy of the bivalent vaccine for prevention of symptomatic COVID-19 at least 14 days after the second injection (dose 2). Safety was assessed in all participants receiving at least one injection of the study vaccine or placebo. This trial is registered with ClinicalTrials.gov (NCT04904549) and is closed to recruitment. FINDINGS Between Oct 19, 2021, and Feb 15, 2022, 13 002 participants were enrolled and randomly assigned to receive the first dose of the study vaccine (n=6512) or placebo (n=6490). 12 924 participants (6472 in the vaccine group and 6452 in the placebo group) received at least one study injection, of whom 7542 (58·4%) were male and 9693 (75·0%) were SARS-CoV-2 non-naive. Of these 12 924 participants, 11 543 (89·3%) received both study injections (5788 in the vaccine group and 5755 in the placebo group). The efficacy-evaluable population after dose 2 comprised 11 416 participants (5736 in the vaccine group and 5680 in the placebo group). The median duration of follow-up was 85 days (IQR 50-95) after dose 1 and 58 days (29-70) after dose 2. 121 symptomatic COVID-19 cases were reported at least 14 days after dose 2 (32 in the vaccine group and 89 in the placebo group), with an overall vaccine efficacy of 64·7% (95% CI 46·6 to 77·2). Vaccine efficacy against symptomatic COVID-19 was 75·1% (95% CI 56·3 to 86·6) in SARS-CoV-2 non-naive participants and 30·9% (-39·3 to 66·7) in SARS-CoV-2-naive participants. Viral genome sequencing identified the infecting strain in 68 (56·2%) of 121 cases (omicron [BA.1 and BA.2] in 63; delta in four; and both omicron and delta in one). Immediate unsolicited adverse events were reported by four (<0·1%) participants in the vaccine group and seven (0·1%) participants in the placebo group. Immediate unsolicited adverse reactions within 30 min after any injection were reported by four (<0·1%) participants in the vaccine group and six (<0·1%) participants in the placebo group. In the reactogenicity subset with available data, solicited reactions (solicited injection-site reactions and solicited systemic reactions) within 7 days after any injection occurred in 1398 (57·8%) of 2420 vaccine recipients and 983 (40·9%) of 2403 placebo recipients. Grade 3 solicited reactions were reported by 196 (8·1%; 95% CI 7·0 to 9·3) of 2420 vaccine recipients and 118 (4·9%; 4·1 to 5·9) of 2403 placebo recipients within 7 days after any injection, with comparable frequencies after dose 1 and dose 2 in the vaccine group. At least one serious adverse event occurred in 30 (0·5%) participants in the vaccine group and 26 (0·4%) in the placebo group. The proportion of adverse events of special interest and deaths was less than 0·1% in both study groups. No adverse event of special interest, serious adverse event, or death was deemed to be treatment related. There were no reported cases of thrombosis with thrombocytopenia syndrome, myocarditis, pericarditis, Bell's Palsy, or Guillain-Barré syndrome, or other immune-mediated diseases. INTERPRETATION The bivalent variant vaccine conferred heterologous protection against symptomatic SARS-CoV-2 infection in the epidemiological context of the circulating contemporary omicron variant. These findings suggest that vaccines developed with an antigen from a non-predominant strain could confer cross-protection against newly emergent SARS-CoV-2 variants, although further investigation is warranted. FUNDING Sanofi, US Biomedical Advanced Research and Development Authority, and the US National Institute of Allergy and Infectious Diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Mary Allen
- National Institute of Allergy and Infectious Diseases / National Institutes of Health, Bethesda, MD, USA
| | | | - Kwaku Poku Asante
- Research and Development Division, Ghana Health Service, Kintampo North Municipality, Ghana
| | | | - Tatiana Beresnev
- National Institute of Allergy and Infectious Diseases / National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | | | - Bo Fu
- National Institute of Allergy and Infectious Diseases / National Institutes of Health, Bethesda, MD, USA
| | | | - Maryam Keshtkar-Jahromi
- National Institutes of Health, Rockville, MD, USA; John Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Jia Jin Kee
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Hannah Kibuuka
- Makerere University Walter Reed Project, Kampala, Uganda
| | | | | | | | | | - Humberto Reynales
- Centro de Attencion e Investigation Medica S.A.S. - Caimed Chía, Chía, Colombia
| | - Merlin L Robb
- The Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MA, USA
| | | | - Fredrick Sawe
- Kenya Medical Research Institute - US Army Medical Research, Kericho, Kenya
| | | | - Tina Tong
- National Institute of Allergy and Infectious Diseases / National Institutes of Health, Bethesda, MD, USA
| | - John Treanor
- Tunnell Government Services in support of Biomedical Advanced Research and Development Authority, Administration for Strategic Preparedness and Response, Department of Health and Human Services, Washington, DC, USA
| | - T Anh Wartel
- International Vaccine Institute, Seoul, South Korea
| | | | | | | | | | | |
Collapse
|
5
|
Parry PI, Lefringhausen A, Turni C, Neil CJ, Cosford R, Hudson NJ, Gillespie J. 'Spikeopathy': COVID-19 Spike Protein Is Pathogenic, from Both Virus and Vaccine mRNA. Biomedicines 2023; 11:2287. [PMID: 37626783 PMCID: PMC10452662 DOI: 10.3390/biomedicines11082287] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 07/17/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
The COVID-19 pandemic caused much illness, many deaths, and profound disruption to society. The production of 'safe and effective' vaccines was a key public health target. Sadly, unprecedented high rates of adverse events have overshadowed the benefits. This two-part narrative review presents evidence for the widespread harms of novel product COVID-19 mRNA and adenovectorDNA vaccines and is novel in attempting to provide a thorough overview of harms arising from the new technology in vaccines that relied on human cells producing a foreign antigen that has evidence of pathogenicity. This first paper explores peer-reviewed data counter to the 'safe and effective' narrative attached to these new technologies. Spike protein pathogenicity, termed 'spikeopathy', whether from the SARS-CoV-2 virus or produced by vaccine gene codes, akin to a 'synthetic virus', is increasingly understood in terms of molecular biology and pathophysiology. Pharmacokinetic transfection through body tissues distant from the injection site by lipid-nanoparticles or viral-vector carriers means that 'spikeopathy' can affect many organs. The inflammatory properties of the nanoparticles used to ferry mRNA; N1-methylpseudouridine employed to prolong synthetic mRNA function; the widespread biodistribution of the mRNA and DNA codes and translated spike proteins, and autoimmunity via human production of foreign proteins, contribute to harmful effects. This paper reviews autoimmune, cardiovascular, neurological, potential oncological effects, and autopsy evidence for spikeopathy. With many gene-based therapeutic technologies planned, a re-evaluation is necessary and timely.
Collapse
Affiliation(s)
- Peter I. Parry
- Children’s Health Research Clinical Unit, Faculty of Medicine, The University of Queensland, South Brisbane, QLD 4101, Australia
- Department of Psychiatry, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia
| | - Astrid Lefringhausen
- Children’s Health Defence (Australia Chapter), Huskisson, NSW 2540, Australia; (A.L.); (R.C.); (J.G.)
| | - Conny Turni
- Microbiology Research, QAAFI (Queensland Alliance for Agriculture and Food Innovation), The University of Queensland, St. Lucia, QLD 4072, Australia;
| | - Christopher J. Neil
- Department of Medicine, University of Melbourne, Melbourne, VIC 3010, Australia;
| | - Robyn Cosford
- Children’s Health Defence (Australia Chapter), Huskisson, NSW 2540, Australia; (A.L.); (R.C.); (J.G.)
| | - Nicholas J. Hudson
- School of Agriculture and Food Science, The University of Queensland, Brisbane, QLD 4072, Australia;
| | - Julian Gillespie
- Children’s Health Defence (Australia Chapter), Huskisson, NSW 2540, Australia; (A.L.); (R.C.); (J.G.)
| |
Collapse
|
6
|
Bannick MS, Gao F, Brown ER, Janes HE. Retrospective, Observational Studies for Estimating Vaccine Effects on the Secondary Attack Rate of SARS-CoV-2. Am J Epidemiol 2023; 192:1016-1028. [PMID: 36883907 PMCID: PMC10505422 DOI: 10.1093/aje/kwad046] [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: 05/31/2022] [Revised: 11/21/2022] [Accepted: 02/23/2023] [Indexed: 03/09/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) vaccines are highly efficacious at preventing symptomatic infection, severe disease, and death. Most of the evidence that COVID-19 vaccines also reduce transmission of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is based on retrospective, observational studies. Specifically, an increasing number of studies are evaluating vaccine effectiveness against the secondary attack rate of SARS-CoV-2 using data available in existing health-care databases or contact-tracing databases. Since these types of databases were designed for clinical diagnosis or management of COVID-19, they are limited in their ability to provide accurate information on infection, infection timing, and transmission events. We highlight challenges with using existing databases to identify transmission units and confirm potential SARS-CoV-2 transmission events. We discuss the impact of common diagnostic testing strategies, including event-prompted and infrequent testing, and illustrate their potential biases in estimating vaccine effectiveness against the secondary attack rate of SARS-CoV-2. We articulate the need for prospective observational studies of vaccine effectiveness against the SARS-CoV-2 secondary attack rate, and we provide design and reporting considerations for studies using retrospective databases.
Collapse
Affiliation(s)
- Marlena S Bannick
- Correspondence to Marlena Bannick, Department of Biostatistics, Hans Rosling Center for Population Health, Box 357232, University of Washington, Seattle, WA 98195 (e-mail: )
| | | | | | | |
Collapse
|
7
|
Akhter N, Sana S, Adnan Ahsan M, Siddique Z, Huraira A, Sana S. Advances in Diagnosis and Treatment for SARS-CoV-2 Variants. Infect Dis (Lond) 2023. [DOI: 10.5772/intechopen.107846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/31/2023] Open
Abstract
The COVID-19 pandemic’s epidemiological and clinical characteristics have been affected in recent months by the introduction of SARS-CoV-2 variants with unique spikes of protein alterations. These variations can lessen the protection provided by suppressing monoclonal antibodies and vaccines, as well as enhance the frequencies of transmission of the virus and/or the risk of contracting the disease. Due to these mutations, SARS-CoV-2 may be able to proliferate despite increasing levels of vaccination coverage while preserving and enhancing its reproduction efficiency. This is one of the main strategies in tackling the COVID-19 epidemics, the accessibility of precise and trustworthy biomarkers for the SARS-CoV-2 genetic material and also its nucleic acids is important to investigate the disease in suspect communities, start making diagnoses and management in symptomatic or asymptomatic persons, and evaluate authorization of the pathogen after infection. Quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) for virus nucleic acid identification is still the most effective method for such uses due to its sensitivity, quickness, high-throughput sequencing capacity, and trustworthiness. It is essential to update the primer and probe sequences to maintain the recognition of recently emerging variations. Concerning viral variations could develop that are dangerously resistant to the immunization induced by the present vaccinations in coronavirus disease 2019. Additionally, the significance of effective public health interventions and vaccination programs will grow if some variations of concern exhibit an increased risk of transmission or toxicity. The international reaction must’ve been immediate and established in science. These results supported ongoing efforts to prevent and identify infection, as well as to describe mutations in vaccine recipients, and they suggest a potential risk of illness following effective immunization and transmission of pathogens with a mutant viral.
Collapse
|
8
|
Benkeser D, Fong Y, Janes HE, Kelly EJ, Hirsch I, Sproule S, Stanley AM, Maaske J, Villafana T, Houchens CR, Martins K, Jayashankar L, Castellino F, Ayala V, Petropoulos CJ, Leith A, Haugaard D, Webb B, Lu Y, Yu C, Borate B, van der Laan LWP, Hejazi NS, Carpp LN, Randhawa AK, Andrasik MP, Kublin JG, Isaacs MB, Makhene M, Tong T, Robb ML, Corey L, Neuzil KM, Follmann D, Hoffman C, Falsey AR, Sobieszczyk M, Koup RA, Donis RO, Gilbert PB. Immune correlates analysis of a phase 3 trial of the AZD1222 (ChAdOx1 nCoV-19) vaccine. NPJ Vaccines 2023; 8:36. [PMID: 36899062 PMCID: PMC10005913 DOI: 10.1038/s41541-023-00630-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 02/20/2023] [Indexed: 03/12/2023] Open
Abstract
In the phase 3 trial of the AZD1222 (ChAdOx1 nCoV-19) vaccine conducted in the U.S., Chile, and Peru, anti-spike binding IgG concentration (spike IgG) and pseudovirus 50% neutralizing antibody titer (nAb ID50) measured four weeks after two doses were assessed as correlates of risk and protection against PCR-confirmed symptomatic SARS-CoV-2 infection (COVID-19). These analyses of SARS-CoV-2 negative participants were based on case-cohort sampling of vaccine recipients (33 COVID-19 cases by 4 months post dose two, 463 non-cases). The adjusted hazard ratio of COVID-19 was 0.32 (95% CI: 0.14, 0.76) per 10-fold increase in spike IgG concentration and 0.28 (0.10, 0.77) per 10-fold increase in nAb ID50 titer. At nAb ID50 below the limit of detection (< 2.612 IU50/ml), 10, 100, and 270 IU50/ml, vaccine efficacy was -5.8% (-651%, 75.6%), 64.9% (56.4%, 86.9%), 90.0% (55.8%, 97.6%) and 94.2% (69.4%, 99.1%). These findings provide further evidence towards defining an immune marker correlate of protection to help guide regulatory/approval decisions for COVID-19 vaccines.
Collapse
Affiliation(s)
- David Benkeser
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Youyi Fong
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Holly E Janes
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Elizabeth J Kelly
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Ian Hirsch
- Biometrics, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Stephanie Sproule
- Biometrics, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Ann Marie Stanley
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Jill Maaske
- Clinical Development, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Tonya Villafana
- Clinical Development, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Christopher R Houchens
- Biomedical Advanced Research and Development Authority, Administration for Strategic Preparedness and Response, Department of Health and Human Services, Washington, DC, USA
| | - Karen Martins
- Biomedical Advanced Research and Development Authority, Administration for Strategic Preparedness and Response, Department of Health and Human Services, Washington, DC, USA
| | - Lakshmi Jayashankar
- Biomedical Advanced Research and Development Authority, Administration for Strategic Preparedness and Response, Department of Health and Human Services, Washington, DC, USA
| | - Flora Castellino
- Biomedical Advanced Research and Development Authority, Administration for Strategic Preparedness and Response, Department of Health and Human Services, Washington, DC, USA
| | - Victor Ayala
- Biomedical Advanced Research and Development Authority, Administration for Strategic Preparedness and Response, Department of Health and Human Services, Washington, DC, USA
| | | | | | | | | | - Yiwen Lu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Chenchen Yu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Bhavesh Borate
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Lars W P van der Laan
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Statistics, University of Washington, Seattle, WA, USA
| | - Nima S Hejazi
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Biostatistics, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Lindsay N Carpp
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - April K Randhawa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Michele P Andrasik
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - James G Kublin
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Mamodikoe Makhene
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Tina Tong
- Vaccine Translational Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Merlin L Robb
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Lawrence Corey
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Kathleen M Neuzil
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dean Follmann
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Corey Hoffman
- Biomedical Advanced Research and Development Authority, Administration for Strategic Preparedness and Response, Department of Health and Human Services, Washington, DC, USA
| | - Ann R Falsey
- Division of Infectious Diseases, Department of Medicine, University of Rochester, Rochester, NY, USA
| | - Magdalena Sobieszczyk
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center and New York-Presbyterian Hospital, New York, NY, USA
| | - Richard A Koup
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ruben O Donis
- Biomedical Advanced Research and Development Authority, Administration for Strategic Preparedness and Response, Department of Health and Human Services, Washington, DC, USA
| | - Peter B Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA.
| |
Collapse
|
9
|
Fong Y, Huang Y, Benkeser D, Carpp LN, Áñez G, Woo W, McGarry A, Dunkle LM, Cho I, Houchens CR, Martins K, Jayashankar L, Castellino F, Petropoulos CJ, Leith A, Haugaard D, Webb B, Lu Y, Yu C, Borate B, van der Laan LWP, Hejazi NS, Randhawa AK, Andrasik MP, Kublin JG, Hutter J, Keshtkar-Jahromi M, Beresnev TH, Corey L, Neuzil KM, Follmann D, Ake JA, Gay CL, Kotloff KL, Koup RA, Donis RO, Gilbert PB. Immune correlates analysis of the PREVENT-19 COVID-19 vaccine efficacy clinical trial. Nat Commun 2023; 14:331. [PMID: 36658109 PMCID: PMC9851580 DOI: 10.1038/s41467-022-35768-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 12/28/2022] [Indexed: 01/21/2023] Open
Abstract
In the PREVENT-19 phase 3 trial of the NVX-CoV2373 vaccine (NCT04611802), anti-spike binding IgG concentration (spike IgG), anti-RBD binding IgG concentration (RBD IgG), and pseudovirus 50% neutralizing antibody titer (nAb ID50) measured two weeks post-dose two are assessed as correlates of risk and as correlates of protection against COVID-19. Analyses are conducted in the U.S. cohort of baseline SARS-CoV-2 negative per-protocol participants using a case-cohort design that measures the markers from all 12 vaccine recipient breakthrough COVID-19 cases starting 7 days post antibody measurement and from 639 vaccine recipient non-cases. All markers are inversely associated with COVID-19 risk and directly associated with vaccine efficacy. In vaccine recipients with nAb ID50 titers of 50, 100, and 7230 international units (IU50)/ml, vaccine efficacy estimates are 75.7% (49.8%, 93.2%), 81.7% (66.3%, 93.2%), and 96.8% (88.3%, 99.3%). The results support potential cross-vaccine platform applications of these markers for guiding decisions about vaccine approval and use.
Collapse
Affiliation(s)
- Youyi Fong
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Yunda Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Global Health, University of Washington, Seattle, WA, 98195, USA
| | - David Benkeser
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Lindsay N Carpp
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Wayne Woo
- Novavax, Inc., Gaithersburg, MD, USA
| | | | | | | | | | - Karen Martins
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| | | | - Flora Castellino
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| | | | | | | | | | - Yiwen Lu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Chenchen Yu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Bhavesh Borate
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Lars W P van der Laan
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Statistics, University of Washington, Seattle, WA, USA
| | - Nima S Hejazi
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Biostatistics, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - April K Randhawa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Michele P Andrasik
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - James G Kublin
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Julia Hutter
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD, USA
| | - Maryam Keshtkar-Jahromi
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD, USA
| | - Tatiana H Beresnev
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD, USA
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Kathleen M Neuzil
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dean Follmann
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Julie A Ake
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Cynthia L Gay
- University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Karen L Kotloff
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Richard A Koup
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ruben O Donis
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| | - Peter B Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA.
| |
Collapse
|
10
|
Graña C, Ghosn L, Evrenoglou T, Jarde A, Minozzi S, Bergman H, Buckley BS, Probyn K, Villanueva G, Henschke N, Bonnet H, Assi R, Menon S, Marti M, Devane D, Mallon P, Lelievre JD, Askie LM, Kredo T, Ferrand G, Davidson M, Riveros C, Tovey D, Meerpohl JJ, Grasselli G, Rada G, Hróbjartsson A, Ravaud P, Chaimani A, Boutron I. Efficacy and safety of COVID-19 vaccines. Cochrane Database Syst Rev 2022; 12:CD015477. [PMID: 36473651 PMCID: PMC9726273 DOI: 10.1002/14651858.cd015477] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Different forms of vaccines have been developed to prevent the SARS-CoV-2 virus and subsequent COVID-19 disease. Several are in widespread use globally. OBJECTIVES: To assess the efficacy and safety of COVID-19 vaccines (as a full primary vaccination series or a booster dose) against SARS-CoV-2. SEARCH METHODS We searched the Cochrane COVID-19 Study Register and the COVID-19 L·OVE platform (last search date 5 November 2021). We also searched the WHO International Clinical Trials Registry Platform, regulatory agency websites, and Retraction Watch. SELECTION CRITERIA We included randomized controlled trials (RCTs) comparing COVID-19 vaccines to placebo, no vaccine, other active vaccines, or other vaccine schedules. DATA COLLECTION AND ANALYSIS We used standard Cochrane methods. We used GRADE to assess the certainty of evidence for all except immunogenicity outcomes. We synthesized data for each vaccine separately and presented summary effect estimates with 95% confidence intervals (CIs). MAIN RESULTS: We included and analyzed 41 RCTs assessing 12 different vaccines, including homologous and heterologous vaccine schedules and the effect of booster doses. Thirty-two RCTs were multicentre and five were multinational. The sample sizes of RCTs were 60 to 44,325 participants. Participants were aged: 18 years or older in 36 RCTs; 12 years or older in one RCT; 12 to 17 years in two RCTs; and three to 17 years in two RCTs. Twenty-nine RCTs provided results for individuals aged over 60 years, and three RCTs included immunocompromized patients. No trials included pregnant women. Sixteen RCTs had two-month follow-up or less, 20 RCTs had two to six months, and five RCTs had greater than six to 12 months or less. Eighteen reports were based on preplanned interim analyses. Overall risk of bias was low for all outcomes in eight RCTs, while 33 had concerns for at least one outcome. We identified 343 registered RCTs with results not yet available. This abstract reports results for the critical outcomes of confirmed symptomatic COVID-19, severe and critical COVID-19, and serious adverse events only for the 10 WHO-approved vaccines. For remaining outcomes and vaccines, see main text. The evidence for mortality was generally sparse and of low or very low certainty for all WHO-approved vaccines, except AD26.COV2.S (Janssen), which probably reduces the risk of all-cause mortality (risk ratio (RR) 0.25, 95% CI 0.09 to 0.67; 1 RCT, 43,783 participants; high-certainty evidence). Confirmed symptomatic COVID-19 High-certainty evidence found that BNT162b2 (BioNtech/Fosun Pharma/Pfizer), mRNA-1273 (ModernaTx), ChAdOx1 (Oxford/AstraZeneca), Ad26.COV2.S, BBIBP-CorV (Sinopharm-Beijing), and BBV152 (Bharat Biotect) reduce the incidence of symptomatic COVID-19 compared to placebo (vaccine efficacy (VE): BNT162b2: 97.84%, 95% CI 44.25% to 99.92%; 2 RCTs, 44,077 participants; mRNA-1273: 93.20%, 95% CI 91.06% to 94.83%; 2 RCTs, 31,632 participants; ChAdOx1: 70.23%, 95% CI 62.10% to 76.62%; 2 RCTs, 43,390 participants; Ad26.COV2.S: 66.90%, 95% CI 59.10% to 73.40%; 1 RCT, 39,058 participants; BBIBP-CorV: 78.10%, 95% CI 64.80% to 86.30%; 1 RCT, 25,463 participants; BBV152: 77.80%, 95% CI 65.20% to 86.40%; 1 RCT, 16,973 participants). Moderate-certainty evidence found that NVX-CoV2373 (Novavax) probably reduces the incidence of symptomatic COVID-19 compared to placebo (VE 82.91%, 95% CI 50.49% to 94.10%; 3 RCTs, 42,175 participants). There is low-certainty evidence for CoronaVac (Sinovac) for this outcome (VE 69.81%, 95% CI 12.27% to 89.61%; 2 RCTs, 19,852 participants). Severe or critical COVID-19 High-certainty evidence found that BNT162b2, mRNA-1273, Ad26.COV2.S, and BBV152 result in a large reduction in incidence of severe or critical disease due to COVID-19 compared to placebo (VE: BNT162b2: 95.70%, 95% CI 73.90% to 99.90%; 1 RCT, 46,077 participants; mRNA-1273: 98.20%, 95% CI 92.80% to 99.60%; 1 RCT, 28,451 participants; AD26.COV2.S: 76.30%, 95% CI 57.90% to 87.50%; 1 RCT, 39,058 participants; BBV152: 93.40%, 95% CI 57.10% to 99.80%; 1 RCT, 16,976 participants). Moderate-certainty evidence found that NVX-CoV2373 probably reduces the incidence of severe or critical COVID-19 (VE 100.00%, 95% CI 86.99% to 100.00%; 1 RCT, 25,452 participants). Two trials reported high efficacy of CoronaVac for severe or critical disease with wide CIs, but these results could not be pooled. Serious adverse events (SAEs) mRNA-1273, ChAdOx1 (Oxford-AstraZeneca)/SII-ChAdOx1 (Serum Institute of India), Ad26.COV2.S, and BBV152 probably result in little or no difference in SAEs compared to placebo (RR: mRNA-1273: 0.92, 95% CI 0.78 to 1.08; 2 RCTs, 34,072 participants; ChAdOx1/SII-ChAdOx1: 0.88, 95% CI 0.72 to 1.07; 7 RCTs, 58,182 participants; Ad26.COV2.S: 0.92, 95% CI 0.69 to 1.22; 1 RCT, 43,783 participants); BBV152: 0.65, 95% CI 0.43 to 0.97; 1 RCT, 25,928 participants). In each of these, the likely absolute difference in effects was fewer than 5/1000 participants. Evidence for SAEs is uncertain for BNT162b2, CoronaVac, BBIBP-CorV, and NVX-CoV2373 compared to placebo (RR: BNT162b2: 1.30, 95% CI 0.55 to 3.07; 2 RCTs, 46,107 participants; CoronaVac: 0.97, 95% CI 0.62 to 1.51; 4 RCTs, 23,139 participants; BBIBP-CorV: 0.76, 95% CI 0.54 to 1.06; 1 RCT, 26,924 participants; NVX-CoV2373: 0.92, 95% CI 0.74 to 1.14; 4 RCTs, 38,802 participants). For the evaluation of heterologous schedules, booster doses, and efficacy against variants of concern, see main text of review. AUTHORS' CONCLUSIONS Compared to placebo, most vaccines reduce, or likely reduce, the proportion of participants with confirmed symptomatic COVID-19, and for some, there is high-certainty evidence that they reduce severe or critical disease. There is probably little or no difference between most vaccines and placebo for serious adverse events. Over 300 registered RCTs are evaluating the efficacy of COVID-19 vaccines, and this review is updated regularly on the COVID-NMA platform (covid-nma.com). Implications for practice Due to the trial exclusions, these results cannot be generalized to pregnant women, individuals with a history of SARS-CoV-2 infection, or immunocompromized people. Most trials had a short follow-up and were conducted before the emergence of variants of concern. Implications for research Future research should evaluate the long-term effect of vaccines, compare different vaccines and vaccine schedules, assess vaccine efficacy and safety in specific populations, and include outcomes such as preventing long COVID-19. Ongoing evaluation of vaccine efficacy and effectiveness against emerging variants of concern is also vital.
Collapse
Affiliation(s)
- Carolina Graña
- Cochrane France, Paris, France
- Centre of Research in Epidemiology and Statistics (CRESS), INSERM, INRAE, Université de Paris, Paris, France
| | - Lina Ghosn
- Cochrane France, Paris, France
- Centre of Research in Epidemiology and Statistics (CRESS), INSERM, INRAE, Université de Paris, Paris, France
| | - Theodoros Evrenoglou
- Centre of Research in Epidemiology and Statistics (CRESS), INSERM, INRAE, Université de Paris, Paris, France
| | - Alexander Jarde
- Cochrane France, Paris, France
- Centre of Research in Epidemiology and Statistics (CRESS), INSERM, INRAE, Université de Paris, Paris, France
| | | | | | | | | | | | | | - Hillary Bonnet
- Cochrane France, Paris, France
- Centre of Research in Epidemiology and Statistics (CRESS), INSERM, INRAE, Université de Paris, Paris, France
| | - Rouba Assi
- Cochrane France, Paris, France
- Centre of Research in Epidemiology and Statistics (CRESS), INSERM, INRAE, Université de Paris, Paris, France
| | | | - Melanie Marti
- Department of Immunization, Vaccines and Biologicals, World Health Organization, Geneva, Switzerland
| | - Declan Devane
- Evidence Synthesis Ireland, Cochrane Ireland and HRB-Trials Methodology Research Network, National University of Ireland, Galway, Ireland
| | - Patrick Mallon
- UCD Centre for Experimental Pathogen Host Research and UCD School of Medicine, University College Dublin, Dublin, Ireland
| | - Jean-Daniel Lelievre
- Department of Clinical Immunology and Infectious Diseases, Henri Mondor Hospital, Vaccine Research Institute, Université Paris Est Créteil, Paris, France
| | - Lisa M Askie
- Quality Assurance Norms and Standards Department, World Health Organization, Geneva, Switzerland
| | - Tamara Kredo
- Cochrane South Africa, South African Medical Research Council, Cape Town, South Africa
| | | | - Mauricia Davidson
- Cochrane France, Paris, France
- Centre of Research in Epidemiology and Statistics (CRESS), INSERM, INRAE, Université de Paris, Paris, France
| | - Carolina Riveros
- Cochrane France, Paris, France
- Centre of Research in Epidemiology and Statistics (CRESS), INSERM, INRAE, Université de Paris, Paris, France
| | | | - Joerg J Meerpohl
- Institute for Evidence in Medicine, Medical Center & Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Cochrane Germany, Cochrane Germany Foundation, Freiburg, Germany
| | - Giacomo Grasselli
- Department of Anesthesia, Intensive Care and Emergency, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Gabriel Rada
- Epistemonikos Foundation, Santiago, Chile
- UC Evidence Center, Cochrane Chile Associated Center, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Asbjørn Hróbjartsson
- Centre for Evidence Based Medicine Odense (CEBMO) and Cochrane Denmark, University of Southern Denmark, Odense, Denmark
- Open Patient data Explorative Network (OPEN), Odense University Hospital, Odense, Denmark
| | - Philippe Ravaud
- Cochrane France, Paris, France
- Centre of Research in Epidemiology and Statistics (CRESS), INSERM, INRAE, Université de Paris, Paris, France
| | - Anna Chaimani
- Cochrane France, Paris, France
- Centre of Research in Epidemiology and Statistics (CRESS), INSERM, INRAE, Université de Paris, Paris, France
| | - Isabelle Boutron
- Cochrane France, Paris, France
- Centre of Research in Epidemiology and Statistics (CRESS), INSERM, INRAE, Université de Paris, Paris, France
| |
Collapse
|
11
|
Fong Y, McDermott AB, Benkeser D, Roels S, Stieh DJ, Vandebosch A, Le Gars M, Van Roey GA, Houchens CR, Martins K, Jayashankar L, Castellino F, Amoa-Awua O, Basappa M, Flach B, Lin BC, Moore C, Naisan M, Naqvi M, Narpala S, O'Connell S, Mueller A, Serebryannyy L, Castro M, Wang J, Petropoulos CJ, Luedtke A, Hyrien O, Lu Y, Yu C, Borate B, van der Laan LWP, Hejazi NS, Kenny A, Carone M, Wolfe DN, Sadoff J, Gray GE, Grinsztejn B, Goepfert PA, Little SJ, Paiva de Sousa L, Maboa R, Randhawa AK, Andrasik MP, Hendriks J, Truyers C, Struyf F, Schuitemaker H, Douoguih M, Kublin JG, Corey L, Neuzil KM, Carpp LN, Follmann D, Gilbert PB, Koup RA, Donis RO. Immune correlates analysis of the ENSEMBLE single Ad26.COV2.S dose vaccine efficacy clinical trial. Nat Microbiol 2022; 7:1996-2010. [PMID: 36357712 PMCID: PMC10166187 DOI: 10.1038/s41564-022-01262-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 10/04/2022] [Indexed: 11/12/2022]
Abstract
Measuring immune correlates of disease acquisition and protection in the context of a clinical trial is a prerequisite for improved vaccine design. We analysed binding and neutralizing antibody measurements 4 weeks post vaccination as correlates of risk of moderate to severe-critical COVID-19 through 83 d post vaccination in the phase 3, double-blind placebo-controlled phase of ENSEMBLE, an international randomized efficacy trial of a single dose of Ad26.COV2.S. We also evaluated correlates of protection in the trial cohort. Of the three antibody immune markers we measured, we found most support for 50% inhibitory dilution (ID50) neutralizing antibody titre as a correlate of risk and of protection. The outcome hazard ratio was 0.49 (95% confidence interval 0.29, 0.81; P = 0.006) per 10-fold increase in ID50; vaccine efficacy was 60% (43%, 72%) at non-quantifiable ID50 (<2.7 IU50 ml-1) and increased to 89% (78%, 96%) at ID50 = 96.3 IU50 ml-1. Comparison of the vaccine efficacy by ID50 titre curves for ENSEMBLE-US, the COVE trial of the mRNA-1273 vaccine and the COV002-UK trial of the AZD1222 vaccine supported the ID50 titre as a correlate of protection across trials and vaccine types.
Collapse
Affiliation(s)
- Youyi Fong
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Adrian B McDermott
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - David Benkeser
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Sanne Roels
- Janssen R&D, a division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Daniel J Stieh
- Janssen Vaccines and Prevention, Leiden, the Netherlands
| | - An Vandebosch
- Janssen R&D, a division of Janssen Pharmaceutica NV, Beerse, Belgium
| | | | | | | | - Karen Martins
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| | | | - Flora Castellino
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| | - Obrimpong Amoa-Awua
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Manjula Basappa
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Britta Flach
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Bob C Lin
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Christopher Moore
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mursal Naisan
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Muhammed Naqvi
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sandeep Narpala
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sarah O'Connell
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Allen Mueller
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Leo Serebryannyy
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mike Castro
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jennifer Wang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Alex Luedtke
- Department of Statistics, University of Washington, Seattle, WA, USA
| | - Ollivier Hyrien
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Yiwen Lu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Chenchen Yu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Bhavesh Borate
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Lars W P van der Laan
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Statistics, University of Washington, Seattle, WA, USA
| | - Nima S Hejazi
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Biostatistics, School of Public Health, Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Avi Kenny
- Department of Biostatistics, University of Washington School of Public Health, Seattle, WA, USA
| | - Marco Carone
- Department of Biostatistics, University of Washington School of Public Health, Seattle, WA, USA
| | - Daniel N Wolfe
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| | - Jerald Sadoff
- Janssen Vaccines and Prevention, Leiden, the Netherlands
| | - Glenda E Gray
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- South African Medical Research Council, Cape Town, South Africa
| | - Beatriz Grinsztejn
- Evandro Chagas National Institute of Infectious Diseases-Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Paul A Goepfert
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Susan J Little
- Division of Infectious Diseases, University of California San Diego, La Jolla, CA, USA
| | - Leonardo Paiva de Sousa
- Evandro Chagas National Institute of Infectious Diseases-Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Rebone Maboa
- Ndlovu Elandsdoorn Site, Dennilton, Limpopo, South Africa
| | - April K Randhawa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Michele P Andrasik
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jenny Hendriks
- Janssen Vaccines and Prevention, Leiden, the Netherlands
| | - Carla Truyers
- Janssen R&D, a division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Frank Struyf
- Janssen R&D, a division of Janssen Pharmaceutica NV, Beerse, Belgium
| | | | | | - James G Kublin
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Kathleen M Neuzil
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Lindsay N Carpp
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Dean Follmann
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Peter B Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Department of Biostatistics, University of Washington School of Public Health, Seattle, WA, USA.
| | - Richard A Koup
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ruben O Donis
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| |
Collapse
|
12
|
Deen J, Clemens JD. Vaccine clinical trials in low- and middle-income countries: a brief review of standard, newer and proposed approaches. Expert Rev Vaccines 2022; 21:1595-1602. [DOI: 10.1080/14760584.2022.2126357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Jacqueline Deen
- Institute of Child Health and Human Development, National Institutes of Health, University of the Philippines, Pedro Gil Street, Ermita, Manila 1000, Philippines
| | - John D Clemens
- International Vaccine Institute, SNU Research Park, Gwanak-gu, Seoul, 08826 Korea
- UCLA Fielding School of Public Health, 650 Charles E Young Drive South, Los Angeles, California 90095-1772, USA
| |
Collapse
|
13
|
Tanveer S, Rowhani-Farid A, Hong K, Jefferson T, Doshi P. Transparency of COVID-19 vaccine trials: decisions without data. BMJ Evid Based Med 2022; 27:199-205. [PMID: 34373256 DOI: 10.1136/bmjebm-2021-111735] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/15/2021] [Indexed: 12/11/2022]
Affiliation(s)
- Sarah Tanveer
- Department of Pharmaceutical Health Services Research, University of Maryland Baltimore, Baltimore, Maryland, USA
| | - Anisa Rowhani-Farid
- Department of Pharmaceutical Health Services Research, University of Maryland Baltimore, Baltimore, Maryland, USA
| | - Kyungwan Hong
- Department of Pharmaceutical Health Services Research, University of Maryland Baltimore, Baltimore, Maryland, USA
| | - Tom Jefferson
- Department for Continuing Education, University of Oxford, Oxford, UK
| | - Peter Doshi
- Department of Pharmaceutical Health Services Research, University of Maryland Baltimore, Baltimore, Maryland, USA
| |
Collapse
|
14
|
Burgess L, Johannes Jordaan J, Wilson M. Perspective Chapter: Ethics of Using Placebo Controlled Trials for Covid-19 Vaccine Development in Vulnerable Populations. Infect Dis (Lond) 2022. [DOI: 10.5772/intechopen.104776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
When clinical trials are conducted in vulnerable communities such as those found within low-to-middle-income-countries (LMICs), there is always the risk of exploitation or harm to these communities during the course of biomedical research. Historically, there have been multiple instances where significant harm was caused. Various organisations have proposed guidelines to minimise the risk of this occurring, however, questionable clinical trials are still conducted. Research Ethics Committees have an additional duty of care to protect these vulnerable populations. During the Covid-19 pandemic the ongoing use of placebo-controlled trials (PCTs), even after approval of a safe and efficacious vaccine, is a topic of great debate and is discussed from an ethical and moral perspective.
Collapse
|
15
|
Sherman AC, Rouphael N, Baden LR. Coronavirus Disease 2019 Vaccine Trials (and Tribulations): How to Improve the Process of Clinical Trials in a Pandemic. Clin Infect Dis 2022; 75:S5-S10. [PMID: 35436331 PMCID: PMC9047247 DOI: 10.1093/cid/ciac301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Indexed: 01/19/2023] Open
Abstract
Vaccine clinical trials have been essential to developing effective severe acute respiratory syndrome coronavirus 2 vaccines. The challenges of supply chain disruptions, infection control, study designs, and participant factors that affect trial procedures are reviewed, with specific solutions to streamline the clinical trial process.
Collapse
Affiliation(s)
- Amy C. Sherman
- Division of Infectious Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States,Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States,Corresponding Author: Amy C. Sherman, MD, Brigham and Women’s Hospital, 15 Francis Street, PBB-A-4, Boston, MA 02115, USA,
| | - Nadine Rouphael
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, GA, United States
| | - Lindsey R. Baden
- Division of Infectious Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States,Alternate Corresponding Author: Lindsey R. Baden, MD, Brigham and Women’s Hospital, 15 Francis Street, PBB-A-4, Boston, MA 02115, USA,
| |
Collapse
|
16
|
Fong Y, McDermott AB, Benkeser D, Roels S, Stieh DJ, Vandebosch A, Gars ML, Van Roey GA, Houchens CR, Martins K, Jayashankar L, Castellino F, Amoa-Awua O, Basappa M, Flach B, Lin BC, Moore C, Naisan M, Naqvi M, Narpala S, O’Connell S, Mueller A, Serebryannyy L, Castro M, Wang J, Petropoulos CJ, Luedtke A, Hyrien O, Lu Y, Yu C, Borate B, van der Laan LWP, Hejazi NS, Kenny A, Carone M, Wolfe DN, Sadoff J, Gray GE, Grinsztejn B, Goepfert PA, Little SJ, de Sousa LP, Maboa R, Randhawa AK, Andrasik MP, Hendriks J, Truyers C, Struyf F, Schuitemaker H, Douoguih M, Kublin JG, Corey L, Neuzil KM, Carpp LN, Follmann D, Gilbert PB, Koup RA, Donis RO. Immune Correlates Analysis of a Single Ad26.COV2.S Dose in the ENSEMBLE COVID-19 Vaccine Efficacy Clinical Trial. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.04.06.22272763. [PMID: 35441174 PMCID: PMC9016647 DOI: 10.1101/2022.04.06.22272763] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Anti-spike IgG binding antibody, anti-receptor binding domain IgG antibody, and pseudovirus neutralizing antibody measurements four weeks post-vaccination were assessed as correlates of risk of moderate to severe-critical COVID-19 outcomes through 83 days post-vaccination and as correlates of protection following a single dose of Ad26.COV2.S COVID-19 vaccine in the placebo-controlled phase of ENSEMBLE, an international, randomized efficacy trial. Each marker had evidence as a correlate of risk and of protection, with strongest evidence for 50% inhibitory dilution (ID50) neutralizing antibody titer. The outcome hazard ratio was 0.49 (95% confidence interval 0.29, 0.81; p=0.006) per 10-fold increase in ID50; vaccine efficacy was 60% (43, 72%) at nonquantifiable ID50 (< 2.7 IU50/ml) and rose to 89% (78, 96%) at ID50 = 96.3 IU50/ml. Comparison of the vaccine efficacy by ID50 titer curves for ENSEMBLE-US, the COVE trial of the mRNA-1273 vaccine, and the COV002-UK trial of the AZD1222 vaccine supported consistency of the ID50 titer correlate of protection across trials and vaccine types.
Collapse
Affiliation(s)
- Youyi Fong
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Adrian B. McDermott
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - David Benkeser
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Sanne Roels
- Janssen R&D, a division of Janssen Pharmaceutica NV, Beerse, Belgium
| | | | - An Vandebosch
- Janssen R&D, a division of Janssen Pharmaceutica NV, Beerse, Belgium
| | | | | | | | - Karen Martins
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| | | | - Flora Castellino
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| | - Obrimpong Amoa-Awua
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Manjula Basappa
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Britta Flach
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Bob C. Lin
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Christopher Moore
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mursal Naisan
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Muhammed Naqvi
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sandeep Narpala
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sarah O’Connell
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Allen Mueller
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Leo Serebryannyy
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mike Castro
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jennifer Wang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Alex Luedtke
- Department of Statistics, University of Washington, Seattle, WA, USA
| | - Ollivier Hyrien
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Yiwen Lu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Chenchen Yu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Bhavesh Borate
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Lars W. P. van der Laan
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Statistics, University of Washington, Seattle, WA, USA
| | - Nima S. Hejazi
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Biostatistics, School of Public Health, Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Avi Kenny
- Department of Biostatistics, University of Washington School of Public Health, Seattle, WA, USA
| | - Marco Carone
- Department of Biostatistics, University of Washington School of Public Health, Seattle, WA, USA
| | - Daniel N. Wolfe
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| | - Jerald Sadoff
- Janssen Vaccines and Prevention, Leiden, the Netherlands
| | - Glenda E. Gray
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- South African Medical Research Council, Cape Town, South Africa
| | - Beatriz Grinsztejn
- Evandro Chagas National Institute of Infectious Diseases-Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Paul A. Goepfert
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Susan J. Little
- Division of Infectious Diseases, University of California San Diego, La Jolla, CA, USA
| | - Leonardo Paiva de Sousa
- Evandro Chagas National Institute of Infectious Diseases-Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Rebone Maboa
- Ndlovu Elandsdoorn Site, Limpopo, Dennilton, South Africa
| | - April K. Randhawa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Michele P. Andrasik
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jenny Hendriks
- Janssen Vaccines and Prevention, Leiden, the Netherlands
| | - Carla Truyers
- Janssen R&D, a division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Frank Struyf
- Janssen R&D, a division of Janssen Pharmaceutica NV, Beerse, Belgium
| | | | | | - James G. Kublin
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Kathleen M. Neuzil
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Lindsay N. Carpp
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Dean Follmann
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Peter B. Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Biostatistics, University of Washington School of Public Health, Seattle, WA, USA
| | - Richard A. Koup
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ruben O. Donis
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| |
Collapse
|
17
|
Han X, Ye Q. The variants of SARS-CoV-2 and the challenges of vaccines. J Med Virol 2022; 94:1366-1372. [PMID: 34890492 PMCID: PMC9015306 DOI: 10.1002/jmv.27513] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 11/13/2021] [Accepted: 12/07/2021] [Indexed: 12/15/2022]
Abstract
Since the outbreak of coronavirus disease 2019 (COVID-19), countries all over the world have suffered severe losses. It affects not only human life and health but also the economy. In response to COVID-19, countries have made tremendous efforts to vaccine development. The newly discovered variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have brought major challenges to the effectiveness and research of vaccines. This article reviews the existing literature and summarizes the main variants of the SARS-CoV-2 and its impact on vaccines, and provides new ideas for the later development of vaccines. An excellent job in developing and applying vaccines will be an important measure for epidemic prevention and control.
Collapse
Affiliation(s)
- Xiucui Han
- Department of Clinical Laboratory, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child HealthNational Children's Regional Medical CenterHangzhouZhejiangChina
| | - Qing Ye
- Department of Clinical Laboratory, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child HealthNational Children's Regional Medical CenterHangzhouZhejiangChina
| |
Collapse
|
18
|
Singh JA, Kochhar S, Wolff J, Atuire C, Bhan A, Emanuel E, Faden R, Ghimire P, Greco D, Ho C, Moon S, Shamsi-Gooshki E, Touré A, Thomé B, Smith MJ, Upshur REG. WHO guidance on COVID-19 vaccine trial designs in the context of authorized COVID-19 vaccines and expanding global access: Ethical considerations. Vaccine 2022; 40:2140-2149. [PMID: 35248422 PMCID: PMC8882397 DOI: 10.1016/j.vaccine.2022.02.038] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 01/04/2023]
Abstract
While the degree of COVID-19 vaccine accessibility and uptake varies at both national and global levels, increasing vaccination coverage raises questions regarding the standard of prevention that ought to apply to different settings where COVID-19 vaccine trials are hosted. A WHO Expert Group has developed guidance on the ethical implications of conducting placebo-controlled trials in the context of expanding global COVID-19 vaccine coverage. The guidance also considers alternative trial designs to placebo controlled trials in the context of prototype vaccines, modified vaccines, and next generation vaccines.
Collapse
Affiliation(s)
- Jerome Amir Singh
- Howard College School of Law, University of KwaZulu-Natal, Durban, South Africa; Scientific Advisory Group on Emergencies (SAGE), Academy of Science of South Africa (ASSAf), Pretoria, South Africa; Department of Clinical Public Health, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada.
| | - Sonali Kochhar
- Department of Global Health, University of Washington, Seattle, WA, USA; Global Healthcare Consulting, Delhi, India
| | - Jonathan Wolff
- Blavatnik School of Government, University of Oxford, Oxford, UK
| | - Caesar Atuire
- Department of Philosophy and Classics, University of Ghana, Accra, Ghana
| | - Anant Bhan
- Department of Community Medicine and Centre for Ethics, Yenepoya University, Mangalore, India
| | - Ezekiel Emanuel
- Global Initiatives and Healthcare Transformation Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Ruth Faden
- Johns Hopkins Berman Institute of Bioethics, Johns Hopkins University, Baltimore, MD, USA
| | | | - Dirceu Greco
- School of Medicine, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Calvin Ho
- Faculty of Law and Centre for Medical Ethics, University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Suerie Moon
- International Relations and Political Science Department & Interdisciplinary Programmes, Global Health Centre, Graduate Institute of International and Development Studies, Geneva, Switzerland
| | - Ehsan Shamsi-Gooshki
- Department of Medical Ethics, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Beatriz Thomé
- Departamento de Medicina Preventiva, Universidade Federal de São Paulo, Rua Botucatu,Sao Paolo, Brazil
| | - Maxwell J Smith
- Faculty of Health Sciences, Western University, London, Ontario, Canada
| | - Ross E G Upshur
- Department of Family and Community Medicine and Dalla Lana School of Public Health University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
19
|
Ceruti S, Cosentino M, Picozzi M. How to continue COVID-19 vaccine clinical trials? The ethics of vaccine research in a time of pandemic. CLINICAL ETHICS 2022; 17:32-40. [PMID: 35250387 PMCID: PMC8819550 DOI: 10.1177/14777509211052467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Between December 2020 and March 2021, the US Food and Drug Administration and the European Medicines Agency issued Emergency Use Authorizations and Conditional Marketing Authorizations for the distribution of the first COVID-19 vaccines. Although these vaccines were thoroughly assessed before their approval, regulators required companies to continue ongoing placebo-controlled clinical trials in order to gather further reliable scientific information on their safety and efficacy, as well as to start new studies to evaluate additional candidates. The aim of this paper is to present and discuss the ethical issues raised by the tension between the need to continue these types of clinical trials and the obligations related to the protection of the rights and well-being of research participants. Specifically, we question whether—how, and to what extent—fundamental principles governing research involving human beings can be applied to the current pandemic situation. We argue that continuing ongoing placebo-controlled clinical trials can be considered ethically justifiable only if all participants are adequately informed of any developments that may affect their willingness to remain enrolled, including the current situation of resource scarcity and the prioritization criteria established for vaccination. However, we also argue that currently approved vaccines, which are considered safe and effective enough to be administered to millions of people as part of the vaccination campaign, necessarily represent the “best proven intervention” currently available and, therefore, should be used as comparators in future studies instead of placebo.
Collapse
Affiliation(s)
- Silvia Ceruti
- Department of Medicine and Surgery, University of Insubria, Italy
| | - Marco Cosentino
- Department of Medicine and Surgery, University of Insubria, Italy
| | - Mario Picozzi
- Department of Biotechnologies and Life Sciences, University of Insubria, Italy
| |
Collapse
|
20
|
Huang Z, Su Y, Zhang T, Xia N. A review of the safety and efficacy of current COVID-19 vaccines. Front Med 2022; 16:39-55. [PMID: 35122210 PMCID: PMC8815389 DOI: 10.1007/s11684-021-0893-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 08/24/2021] [Indexed: 01/06/2023]
Abstract
Vaccination is the most effective and feasible way to contain the Coronavirus disease 2019 (COVID-19) pandemic. The rapid development of effective COVID-19 vaccines is an extraordinary achievement. This study reviewed the efficacy/effectiveness, immunogenicity, and safety profile of the 12 most progressed COVID-19 vaccines and discussed the challenges and prospects of the vaccine-based approaches in a global crisis. Overall, most of the current vaccines have shown safety and efficacy/effectiveness during actual clinical trials or in the real-world studies, indicating a development of pandemic control. However, many challenges are faced by pandemic control in terms of maximizing the effect of vaccines, such as rapid vaccine coverage, strategies to address variants with immune escape capability, and surveillance of vaccine safety in the medium- and long-terms.
Collapse
Affiliation(s)
- Zehong Huang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Strait Collaborative Innovation Center of Biomedicine and Pharmaceutics, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Yingying Su
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Strait Collaborative Innovation Center of Biomedicine and Pharmaceutics, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Tianying Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Strait Collaborative Innovation Center of Biomedicine and Pharmaceutics, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Ningshao Xia
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Strait Collaborative Innovation Center of Biomedicine and Pharmaceutics, School of Public Health, Xiamen University, Xiamen, 361102, China.
| |
Collapse
|
21
|
Lynch HF, Caplan A, Furlong P, Bateman-House A. Helpful Lessons and Cautionary Tales: How Should COVID-19 Drug Development and Access Inform Approaches to Non-Pandemic Diseases? THE AMERICAN JOURNAL OF BIOETHICS : AJOB 2021; 21:4-19. [PMID: 34665689 DOI: 10.1080/15265161.2021.1974975] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
After witnessing extraordinary scientific and regulatory efforts to speed development of and access to new COVID-19 interventions, patients facing other serious diseases have begun to ask "where's our Operation Warp Speed?" and "why isn't Emergency Use Authorization an option for our health crises?" Although this pandemic bears a number of unique features, the response to COVID-19 offers translatable lessons, in both its successes and failures, for non-pandemic diseases. These include the importance of collaborating across sectors, supporting the highest-priority research efforts, adopting rigorous and innovative trial designs, and sharing reliable information quickly. In addition, the regulatory response to the pandemic demonstrates that lowering standards for marketing authorization can result in increased safety concerns, missed opportunities for research and treatment, and delays in determining what works. Accordingly, policymakers and patient advocates seeking to build on the COVID-19 experience for non-pandemic diseases with unmet treatment needs should focus their efforts on promoting robust and efficient research designs, improving access to clinical trials, and facilitating use of the Food and Drug Administration's existing Expanded Access pathway.
Collapse
|
22
|
Habibzadeh F, Sajadi MM, Chumakov K, Yadollahie M, Kottilil S, Simi A, Stafford K, Saeidimehr S, Rafiei M, Gallo RC. COVID-19 Infection Among Women in Iran Exposed vs Unexposed to Children Who Received Attenuated Poliovirus Used in Oral Polio Vaccine. JAMA Netw Open 2021; 4:e2135044. [PMID: 34817583 PMCID: PMC8613592 DOI: 10.1001/jamanetworkopen.2021.35044] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 09/22/2021] [Indexed: 01/01/2023] Open
Abstract
Importance Live attenuated vaccines may provide short-term protection against infectious diseases through stimulation of the innate immune system. Objective To evaluate whether passive exposure to live attenuated poliovirus is associated with diminished symptomatic infection with SARS-CoV-2. Design, Setting, and Participants In a longitudinal cohort study involving 87 923 people conducted between March 20 and December 20, 2020, the incidence of COVID-19 was compared between 2 groups of aged-matched women with and without exposure to live attenuated poliovirus in the oral polio vaccine (OPV). Participants were people receiving health care services from the Petroleum Industry Health Organization and residing in 2 cities in Iran (ie, Ahwaz and Shiraz). Participants were women aged 18 to 48 years whose children were aged 18 months or younger and a group of age-matched women from the same residence who had had no potential exposure to OPV. Exposures Indirect exposure to live attenuated poliovirus in OPV. Main Outcomes and Measures Symptomatic COVID-19, diagnosed by reverse transcription-polymerase chain reaction. Results After applying the inclusion and exclusion criteria, 419 mothers (mean [SD] age, 35.5 [4.9] years) indirectly exposed to the OPV and 3771 age-matched women (mean [SD] age, 35.7 [5.3] years) who had no exposure to OPV were available for analysis. COVID-19 was diagnosed in 1319 of the 87 923 individuals in the study population (151 per 10 000 population) during the study period. None of the mothers whose children received OPV developed COVID-19 after a median follow-up of 141 days (IQR, 92-188 days; range, 1-270 days); 28 women (0.74%; 95% CI, 0.47%-1.02%) in the unexposed group were diagnosed with COVID-19 during the 9 months of the study. Point-by-point comparison of the survival curves of the exposed and unexposed groups found that indirect exposure to OPV was significantly associated with decreased COVID-19 acquisition; probability of remaining without infection was 1.000 (95% CI, 1.000-1.000) in the exposed group vs 0.993 (95% CI, 0.990-0.995) in the unexposed group after 9 months (P < .001). Conclusions and Relevance In this cohort study, indirect exposure to live attenuated poliovirus was associated with decreased symptomatic infection with COVID-19. Further study of the potential protective effect of OPV should be conducted, especially in nations where OPV is already in use for polio prevention and specific COVID-19 vaccines are delayed, less affordable, or fail to meet demand.
Collapse
Affiliation(s)
- Farrokh Habibzadeh
- Global Virus Network, Middle East Region, Shiraz, Iran
- R&D Headquarters, Petroleum Industry Health Organization, Shiraz, Iran
| | - Mohammad M. Sajadi
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore
- Global Virus Network, Baltimore, Maryland
| | - Konstantin Chumakov
- Office of Vaccines Research and Review, Food and Drug Administration, Global Virus Network Center of Excellence, Silver Spring, Maryland
| | | | - Shyamasundaran Kottilil
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore
- Global Virus Network, Baltimore, Maryland
| | - Ashraf Simi
- R&D Headquarters, Petroleum Industry Health Organization, Shiraz, Iran
| | - Kristen Stafford
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore
- Global Virus Network, Baltimore, Maryland
| | | | - Mohammad Rafiei
- Petroleum Industry Health Organization Headquarters, Tehran, Iran
| | - Robert C. Gallo
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore
- Global Virus Network, Baltimore, Maryland
| |
Collapse
|
23
|
Snowden JA, McGrath E, Orchard K, Kröger N, Sureda A, Gratwohl A. Visions for a JACIE Quality Management System 4.0. Bone Marrow Transplant 2021; 56:2876-2881. [PMID: 34588628 PMCID: PMC8479268 DOI: 10.1038/s41409-021-01467-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 08/30/2021] [Accepted: 09/09/2021] [Indexed: 11/13/2022]
Abstract
Quality management has been part of hematopoietic stem cell transplantation (HSCT) from the very beginning. It evolved step-wise from open data exchange up to the introduction of the FACT/JACIE-based quality management system (QMS) 2 decades ago. This formal step has eased cooperation, and improved outcome for patients. Today’s expansion of cellular and targeted therapies and new drugs, and the regulatory requirements for advanced therapeutic medicinal products have touched the limits of the current system. Based on the Medicine 4.0 concept, the next step should integrate novel views of QMS. The old definition “Best Quality Transplant” will be replaced by “Optimal Treatment,” and encompass the entire health care journey. “Best outcome” will refer to overall survival, quality of life and costs, with or without HSCT, and will be compatible with all requirements by competent authorities. Decisions will be based on high-level evidence, supported by real-time digitized data collection, data analysis, incorporated into artificial-intelligence systems. To reach this goal, EBMT/JACIE will be challenged to start the process by further fostering harmonization within and between organizations at institutional, national, and European levels. Acceleration in information technology and modifications to working practices during the pandemic should facilitate this development to the next stage.
Collapse
Affiliation(s)
- John A Snowden
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | | | - Kim Orchard
- Department of Haematology, University Hospital Southampton, NHS Foundation Trust, Southampton, UK
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Hospital Eppendorf, Hamburg, Germany
| | - Anna Sureda
- Catalan Institute of Oncology, Barcelona, Spain
| | - Alois Gratwohl
- Hematology, Medical Faculty, University of Basel, Basel, Switzerland.
| |
Collapse
|
24
|
Alqahtani M, Mallah SI, Stevenson N, Doherty S. Vaccine trials during a pandemic: potential approaches to ethical dilemmas. Trials 2021; 22:628. [PMID: 34526086 PMCID: PMC8443109 DOI: 10.1186/s13063-021-05597-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 09/01/2021] [Indexed: 01/10/2023] Open
Abstract
Ever since the emergence of the coronavirus disease 2019 (COVID-19), global public health infrastructures and systems, along with community-wide collaboration and service, have risen to an unprecedented challenge. Vaccine development was immediately propelled to the centre of all our scientific, public health and community efforts. Despite the development of SARS-CoV-2 vaccines arguably being the greatest and most palpable achievements of the past 12 months, they have also been one of the most contentious and debated issues during the pandemic. However, what uniquely differentiates vaccine development is its intimate relationship with the community it seeks to serve; both in its clinical trial testing as an efficacious and safe prophylactic, and its post-developmental 'roll-out' success, as an effective public health tool. These relationships have birthed a myriad of complexities, from community-based mistrust, to academically contended ethical dilemmas. Indeed, the accelerated advances in the COVID-19 vaccine race have further exacerbated this phenomenon, bringing with it new ethical dilemmas that need to be examined to ensure the continued clinical success of these therapeutics and a renewed societal trust in clinical medicine.In this paper, we discuss two major ethical dilemmas: (1) the equipoise of continuing new vaccine trials in the advent of successful candidates and (2) the maleficence of blinded placebo arms. Accordingly, we discuss six different potential approaches to these ethical dilemmas: (1) continuing with placebo-controlled trials, (2) transitioning from placebo-controlled to open-label, (3) unblinding at-risk priority groups only, (4) transitioning to a blinded stepped-wedge cross-over design, (5) progressing to a blinded active-controlled stepped-wedge cross-over trial, and (6) conducting randomised stepped-wedge community trials. We also propose a decision-making algorithm for relevant stakeholders in advanced stages of vaccine trials.It is important to remember that the emergent nature of the COVID-19 situation does not justify a compromise on core ethical values. In fact, the discourse surrounding this topic and the decisions made will remain a potent case study and a continuously referenced example for all such future scenarios.
Collapse
Affiliation(s)
- Manaf Alqahtani
- The National Taskforce for the Combating of the Coronavirus (COVID-19), Manama, Kingdom of Bahrain.
- The National COVID-19 Research and Ethics Committee, Manama, Kingdom of Bahrain.
- Department of Medicine, Royal College of Surgeons in Ireland - Bahrain, Busaiteen, Kingdom of Bahrain.
| | - Saad I Mallah
- The National Taskforce for the Combating of the Coronavirus (COVID-19), Manama, Kingdom of Bahrain
- School of Medicine, Royal College of Surgeons in Ireland - Bahrain, Busaiteen, Kingdom of Bahrain
| | - Nigel Stevenson
- Department of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Sally Doherty
- Research Ethics Committee, Royal College of Surgeons in Ireland - Bahrain, Busaiteen, Kingdom of Bahrain
| |
Collapse
|
25
|
Bonati M. Restrictions on the use of placebo in new COVID-19 vaccine trials. Eur J Clin Pharmacol 2021; 78:147-148. [PMID: 34462794 PMCID: PMC8404758 DOI: 10.1007/s00228-021-03203-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 08/11/2021] [Indexed: 11/30/2022]
Affiliation(s)
- Maurizio Bonati
- Department of Public Health, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy.
| |
Collapse
|
26
|
Dal-Ré R, Caplan AL. Current COVID-19 vaccine trials in high-income countries: are placebo-controlled trials ethical? Clin Microbiol Infect 2021; 27:1565-1567. [PMID: 34375757 PMCID: PMC8349440 DOI: 10.1016/j.cmi.2021.08.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/27/2021] [Accepted: 08/02/2021] [Indexed: 11/14/2022]
Affiliation(s)
- Rafael Dal-Ré
- Epidemiology Unit, Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid, Madrid, Spain.
| | - Arthur L Caplan
- Division of Medical Ethics, Grossman School of Medicine, NYU Langone Medical Center, New York, NY, USA
| |
Collapse
|
27
|
Fleming TR, Nason M, Krause PR, Longini IM, Henao-Restrepo AM. COVID-19 vaccine trials: The potential for "hybrid" analyses. Clin Trials 2021; 18:391-397. [PMID: 34041932 PMCID: PMC8317268 DOI: 10.1177/17407745211018613] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Although several COVID-19 vaccines have been found to be effective in rigorous evaluation and have emerging availability in parts of the world, their supply will be inadequate to meet international needs for a considerable period of time. There also will be continued interest in vaccines that are more effective or have improved scalability to facilitate mass vaccination campaigns. Ongoing clinical testing of new vaccines also will be needed as variant strains continue to emerge that may elude some aspects of immunity induced by current vaccines. Randomized clinical trials meaningfully enhance the efficiency and reliability of such clinical testing. In clinical settings with limited or no access to known effective vaccines, placebo-controlled randomized trials of new vaccines remain a preferred approach to maximize the reliability, efficiency and interpretability of results. When emerging availability of licensed vaccines makes it no longer possible to use a placebo control, randomized active comparator non-inferiority trials may enable reliable insights. METHODS In this article, "hybrid" methods are proposed to address settings where, during the conduct of a placebo-controlled trial, a judgment is made to replace the placebo arm by a licensed COVID-19 vaccine due to emerging availability of effective vaccines in regions participating in that trial. These hybrid methods are based on proposed statistics that aggregate evidence to formally test as well as to estimate the efficacy of the experimental vaccine, by combining placebo-controlled data during the first period of trial conduct with active-controlled data during the second period. RESULTS Application of the proposed methods is illustrated in two important scenarios where the active control vaccine would become available in regions engaging in the experimental vaccine's placebo-controlled trial: in the first, the active comparator's vaccine efficacy would have been established to be 50%-70% for the 4- to 6-month duration of follow-up of its placebo-controlled trial; in the second, the active comparator's vaccine efficacy would have been established to be 90%-95% during that duration. These two scenarios approximate what has been seen with adenovirus vaccines or mRNA vaccines, respectively, assuming the early estimates of vaccine efficacy for those vaccines would hold over longer-term follow-up. CONCLUSION The proposed hybrid methods could readily play an important role in the near future in the design, conduct and analysis of randomized clinical trials performed to address the need for multiple additional vaccines reliably established to be safe and have worthwhile efficacy in reducing the risk of symptomatic disease from SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Thomas R Fleming
- Department of Biostatistics, University of Washington,
Seattle, WA, USA
| | - Martha Nason
- Biostatistics Research Branch, National Institute of
Allergy and Infectious Diseases (NIAID/NIH), Bethesda, MD, USA
| | - Philip R Krause
- Office of Vaccines Research and Review, FDA/CBER, Silver
Spring, MD, USA
| | - Ira M Longini
- Department of Biostatistics, University of Florida,
Gainesville, USA
| | | |
Collapse
|
28
|
Sadarangani M, Marchant A, Kollmann TR. Immunological mechanisms of vaccine-induced protection against COVID-19 in humans. Nat Rev Immunol 2021; 21:475-484. [PMID: 34211186 PMCID: PMC8246128 DOI: 10.1038/s41577-021-00578-z] [Citation(s) in RCA: 385] [Impact Index Per Article: 96.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2021] [Indexed: 02/06/2023]
Abstract
Most COVID-19 vaccines are designed to elicit immune responses, ideally neutralizing antibodies (NAbs), against the SARS-CoV-2 spike protein. Several vaccines, including mRNA, adenoviral-vectored, protein subunit and whole-cell inactivated virus vaccines, have now reported efficacy in phase III trials and have received emergency approval in many countries. The two mRNA vaccines approved to date show efficacy even after only one dose, when non-NAbs and moderate T helper 1 cell responses are detectable, but almost no NAbs. After a single dose, the adenovirus vaccines elicit polyfunctional antibodies that are capable of mediating virus neutralization and of driving other antibody-dependent effector functions, as well as potent T cell responses. These data suggest that protection may require low levels of NAbs and might involve other immune effector mechanisms including non-NAbs, T cells and innate immune mechanisms. Identifying the mechanisms of protection as well as correlates of protection is crucially important to inform further vaccine development and guide the use of licensed COVID-19 vaccines worldwide.
Collapse
Affiliation(s)
- Manish Sadarangani
- Vaccine Evaluation Center, BC Children's Hospital, Vancouver, British Columbia, Canada.
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Arnaud Marchant
- Institute for Medical Immunology, Université libre de Bruxelles, Charleroi, Belgium
| | - Tobias R Kollmann
- Telethon Kids Institute, Perth Children's Hospital, University of Western Australia, Nedlands, Western Australia, Australia
| |
Collapse
|
29
|
Follmann D, Fintzi J, Fay MP, Janes HE, Baden LR, El Sahly HM, Fleming TR, Mehrotra DV, Carpp LN, Juraska M, Benkeser D, Donnell D, Fong Y, Han S, Hirsch I, Huang Y, Huang Y, Hyrien O, Luedtke A, Carone M, Nason M, Vandebosch A, Zhou H, Cho I, Gabriel E, Kublin JG, Cohen MS, Corey L, Gilbert PB, Neuzil KM. A Deferred-Vaccination Design to Assess Durability of COVID-19 Vaccine Effect After the Placebo Group Is Vaccinated. Ann Intern Med 2021; 174:1118-1125. [PMID: 33844575 PMCID: PMC8099035 DOI: 10.7326/m20-8149] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Multiple candidate vaccines to prevent COVID-19 have entered large-scale phase 3 placebo-controlled randomized clinical trials, and several have demonstrated substantial short-term efficacy. At some point after demonstration of substantial efficacy, placebo recipients should be offered the efficacious vaccine from their trial, which will occur before longer-term efficacy and safety are known. The absence of a placebo group could compromise assessment of longer-term vaccine effects. However, by continuing follow-up after vaccination of the placebo group, this study shows that placebo-controlled vaccine efficacy can be mathematically derived by assuming that the benefit of vaccination over time has the same profile for the original vaccine recipients and the original placebo recipients after their vaccination. Although this derivation provides less precise estimates than would be obtained by a standard trial where the placebo group remains unvaccinated, this proposed approach allows estimation of longer-term effect, including durability of vaccine efficacy and whether the vaccine eventually becomes harmful for some. Deferred vaccination, if done open-label, may lead to riskier behavior in the unblinded original vaccine group, confounding estimates of long-term vaccine efficacy. Hence, deferred vaccination via blinded crossover, where the vaccine group receives placebo and vice versa, would be the preferred way to assess vaccine durability and potential delayed harm. Deferred vaccination allows placebo recipients timely access to the vaccine when it would no longer be proper to maintain them on placebo, yet still allows important insights about immunologic and clinical effectiveness over time.
Collapse
Affiliation(s)
- Dean Follmann
- National Institute of Allergy and Infectious Diseases, Bethesda, Maryland (D.F., J.F., M.P.F., M.N.)
| | - Jonathan Fintzi
- National Institute of Allergy and Infectious Diseases, Bethesda, Maryland (D.F., J.F., M.P.F., M.N.)
| | - Michael P Fay
- National Institute of Allergy and Infectious Diseases, Bethesda, Maryland (D.F., J.F., M.P.F., M.N.)
| | - Holly E Janes
- Fred Hutchinson Cancer Research Center, Seattle, Washington (H.E.J., L.N.C., M.J., D.D., Y.F., Y.H., Y.H., O.H., J.G.K.)
| | - Lindsey R Baden
- Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts (L.R.B.)
| | | | - Thomas R Fleming
- University of Washington, Seattle, Washington (T.R.F., A.L., M.C.)
| | | | - Lindsay N Carpp
- Fred Hutchinson Cancer Research Center, Seattle, Washington (H.E.J., L.N.C., M.J., D.D., Y.F., Y.H., Y.H., O.H., J.G.K.)
| | - Michal Juraska
- Fred Hutchinson Cancer Research Center, Seattle, Washington (H.E.J., L.N.C., M.J., D.D., Y.F., Y.H., Y.H., O.H., J.G.K.)
| | - David Benkeser
- Rollins School of Public Health, Emory University, Atlanta, Georgia (D.B.)
| | - Deborah Donnell
- Fred Hutchinson Cancer Research Center, Seattle, Washington (H.E.J., L.N.C., M.J., D.D., Y.F., Y.H., Y.H., O.H., J.G.K.)
| | - Youyi Fong
- Fred Hutchinson Cancer Research Center, Seattle, Washington (H.E.J., L.N.C., M.J., D.D., Y.F., Y.H., Y.H., O.H., J.G.K.)
| | - Shu Han
- Moderna, Inc., Cambridge, Massachusetts (S.H., H.Z.)
| | - Ian Hirsch
- AstraZeneca, Cambridge, United Kingdom (I.H.)
| | - Ying Huang
- Fred Hutchinson Cancer Research Center, Seattle, Washington (H.E.J., L.N.C., M.J., D.D., Y.F., Y.H., Y.H., O.H., J.G.K.)
| | - Yunda Huang
- Fred Hutchinson Cancer Research Center, Seattle, Washington (H.E.J., L.N.C., M.J., D.D., Y.F., Y.H., Y.H., O.H., J.G.K.)
| | - Ollivier Hyrien
- Fred Hutchinson Cancer Research Center, Seattle, Washington (H.E.J., L.N.C., M.J., D.D., Y.F., Y.H., Y.H., O.H., J.G.K.)
| | - Alex Luedtke
- University of Washington, Seattle, Washington (T.R.F., A.L., M.C.)
| | - Marco Carone
- University of Washington, Seattle, Washington (T.R.F., A.L., M.C.)
| | - Martha Nason
- National Institute of Allergy and Infectious Diseases, Bethesda, Maryland (D.F., J.F., M.P.F., M.N.)
| | | | - Honghong Zhou
- Moderna, Inc., Cambridge, Massachusetts (S.H., H.Z.)
| | - Iksung Cho
- Novavax, Inc., Gaithersburg, Maryland (I.C.)
| | | | - James G Kublin
- Fred Hutchinson Cancer Research Center, Seattle, Washington (H.E.J., L.N.C., M.J., D.D., Y.F., Y.H., Y.H., O.H., J.G.K.)
| | - Myron S Cohen
- Institute for Global Health and Infectious Diseases, University of North Carolina, Chapel Hill, North Carolina (M.S.C.)
| | - Lawrence Corey
- Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington (L.C., P.B.G.)
| | - Peter B Gilbert
- Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington (L.C., P.B.G.)
| | - Kathleen M Neuzil
- University of Maryland School of Medicine, Baltimore, Maryland (K.M.N.)
| |
Collapse
|
30
|
Dal-Ré R. Clinical Equipoise in COVID-19 Vaccine Candidate Trials. J Clin Pharmacol 2021; 61:1249-1250. [PMID: 33826784 PMCID: PMC8250871 DOI: 10.1002/jcph.1868] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 03/30/2021] [Indexed: 11/05/2022]
Affiliation(s)
- Rafael Dal-Ré
- Epidemiology Unit, Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
31
|
Nodosome Inhibition as a Novel Broad-Spectrum Antiviral Strategy against Arboviruses, Enteroviruses, and SARS-CoV-2. Antimicrob Agents Chemother 2021; 65:e0049121. [PMID: 34001511 DOI: 10.1128/aac.00491-21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
In the present report, we describe two small molecules with broad-spectrum antiviral activity. These drugs block the formation of the nodosome. The studies were prompted by the observation that infection of human fetal brain cells with Zika virus (ZIKV) induces the expression of nucleotide-binding oligomerization domain-containing protein 2 (NOD2), a host factor that was found to promote ZIKV replication and spread. A drug that targets NOD2 was shown to have potent broad-spectrum antiviral activity against other flaviviruses, alphaviruses, enteroviruses, and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19). Another drug that inhibits receptor-interacting serine/threonine protein kinase 2 (RIPK2), which functions downstream of NOD2, also decreased the replication of these pathogenic RNA viruses. The antiviral effect of this drug was particularly potent against enteroviruses. The broad-spectrum action of nodosome-targeting drugs is mediated in part by the enhancement of the interferon response. Together, these results suggest that further preclinical investigation of nodosome inhibitors as potential broad-spectrum antivirals is warranted.
Collapse
|
32
|
Dal-Ré R. Placebo-controlled trials with COVID-19 vaccines: participants first. J Clin Epidemiol 2021; 138:211-212. [PMID: 34271109 PMCID: PMC8276569 DOI: 10.1016/j.jclinepi.2021.07.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/18/2021] [Accepted: 07/10/2021] [Indexed: 02/06/2023]
Affiliation(s)
- Rafael Dal-Ré
- Epidemiology Unit, Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid, Madrid, Spain.
| |
Collapse
|
33
|
BULCA ACAR A, NUR EKE R, ÖZEN M. An Analysis of the Attitudes of Family Physicians Towards the COVID-19 Vaccine. KONURALP TIP DERGISI 2021. [DOI: 10.18521/ktd.880762] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
34
|
A flow cytometric assay to detect platelet-activating antibodies in VITT after ChAdOx1 nCov-19 vaccination. Blood 2021; 137:3656-3659. [PMID: 33945605 PMCID: PMC8105122 DOI: 10.1182/blood.2021012064] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 05/03/2021] [Indexed: 01/23/2023] Open
Abstract
Anti-PF4 antibodies as detected by some PF4/heparin enzyme immunoassays are not specific for VITT. We provide a functional whole-blood flow cytometry test for detection of platelet-activating anti-PF4 antibodies in VITT.
Vaccination is crucial in combatting the severe acute respiratory syndrome coronavirus 2 pandemic. The rare complication of thrombocytopenia and thrombotic complications at unusual sites after ChAdOx1 nCov-19 vaccination is caused by platelet-activating antibodies directed against platelet factor 4 (PF4). We present a widely applicable whole-blood standard flow cytometric assay to identify the pathogenic antibodies associated with vaccine-induced immune-mediated thrombotic thrombocytopenia (VITT) after ChAdOx1 nCov-19 vaccination. This assay will enable rapid diagnosis by many laboratories. This trial was registered at www.clinicaltrials.gov as #NCT04370119.
Collapse
|
35
|
Krause PR, Fleming TR, Longini IM, Peto R, Briand S, Heymann DL, Beral V, Snape MD, Rees H, Ropero AM, Balicer RD, Cramer JP, Muñoz-Fontela C, Gruber M, Gaspar R, Singh JA, Subbarao K, Van Kerkhove MD, Swaminathan S, Ryan MJ, Henao-Restrepo AM. SARS-CoV-2 Variants and Vaccines. N Engl J Med 2021; 385:179-186. [PMID: 34161052 PMCID: PMC8262623 DOI: 10.1056/nejmsr2105280] [Citation(s) in RCA: 268] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Viral variants of concern may emerge with dangerous resistance to the immunity generated by the current vaccines to prevent coronavirus disease 2019 (Covid-19). Moreover, if some variants of concern have increased transmissibility or virulence, the importance of efficient public health measures and vaccination programs will increase. The global response must be both timely and science based.
Collapse
Affiliation(s)
- Philip R Krause
- From the Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD (P.R.K., M.G.); the Department of Biostatistics, University of Washington, Seattle (T.R.F.); the Department of Biostatistics, University of Florida, Gainesville (I.M.L.); the Nuffield Department of Population Health, University of Oxford (R.P., V.B.), and the Oxford Vaccine Group, Department of Paediatrics, University of Oxford and National Institute for Health Research Oxford Biomedical Research Centre (M.D.S.), Oxford, and the Global Health Programme, Chatham House (D.L.H.), and the Coalition for Epidemic Preparedness Innovations (J.P.C.), London - all in the United Kingdom; the Howard College School of Law, University of KwaZulu-Natal, Durban (J.A.S.), and the Wits Reproductive Health and HIV Institute, Johannesburg (H.R.) - both in South Africa; the Dalla Lana School of Public Health, University of Toronto, Toronto (J.A.S.); the Clalit Research Institute, Innovation Division, Clalit Health Services, Tel Aviv, Israel (R.D.B.); the Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany (C.M.-F.); the World Health Organization Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia (K.S.); and the World Health Organization, Geneva (S.B., A.-M.R., R.G., M.D.V.K., S.S., M.J.R., A.-M.H.-R.)
| | - Thomas R Fleming
- From the Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD (P.R.K., M.G.); the Department of Biostatistics, University of Washington, Seattle (T.R.F.); the Department of Biostatistics, University of Florida, Gainesville (I.M.L.); the Nuffield Department of Population Health, University of Oxford (R.P., V.B.), and the Oxford Vaccine Group, Department of Paediatrics, University of Oxford and National Institute for Health Research Oxford Biomedical Research Centre (M.D.S.), Oxford, and the Global Health Programme, Chatham House (D.L.H.), and the Coalition for Epidemic Preparedness Innovations (J.P.C.), London - all in the United Kingdom; the Howard College School of Law, University of KwaZulu-Natal, Durban (J.A.S.), and the Wits Reproductive Health and HIV Institute, Johannesburg (H.R.) - both in South Africa; the Dalla Lana School of Public Health, University of Toronto, Toronto (J.A.S.); the Clalit Research Institute, Innovation Division, Clalit Health Services, Tel Aviv, Israel (R.D.B.); the Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany (C.M.-F.); the World Health Organization Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia (K.S.); and the World Health Organization, Geneva (S.B., A.-M.R., R.G., M.D.V.K., S.S., M.J.R., A.-M.H.-R.)
| | - Ira M Longini
- From the Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD (P.R.K., M.G.); the Department of Biostatistics, University of Washington, Seattle (T.R.F.); the Department of Biostatistics, University of Florida, Gainesville (I.M.L.); the Nuffield Department of Population Health, University of Oxford (R.P., V.B.), and the Oxford Vaccine Group, Department of Paediatrics, University of Oxford and National Institute for Health Research Oxford Biomedical Research Centre (M.D.S.), Oxford, and the Global Health Programme, Chatham House (D.L.H.), and the Coalition for Epidemic Preparedness Innovations (J.P.C.), London - all in the United Kingdom; the Howard College School of Law, University of KwaZulu-Natal, Durban (J.A.S.), and the Wits Reproductive Health and HIV Institute, Johannesburg (H.R.) - both in South Africa; the Dalla Lana School of Public Health, University of Toronto, Toronto (J.A.S.); the Clalit Research Institute, Innovation Division, Clalit Health Services, Tel Aviv, Israel (R.D.B.); the Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany (C.M.-F.); the World Health Organization Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia (K.S.); and the World Health Organization, Geneva (S.B., A.-M.R., R.G., M.D.V.K., S.S., M.J.R., A.-M.H.-R.)
| | - Richard Peto
- From the Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD (P.R.K., M.G.); the Department of Biostatistics, University of Washington, Seattle (T.R.F.); the Department of Biostatistics, University of Florida, Gainesville (I.M.L.); the Nuffield Department of Population Health, University of Oxford (R.P., V.B.), and the Oxford Vaccine Group, Department of Paediatrics, University of Oxford and National Institute for Health Research Oxford Biomedical Research Centre (M.D.S.), Oxford, and the Global Health Programme, Chatham House (D.L.H.), and the Coalition for Epidemic Preparedness Innovations (J.P.C.), London - all in the United Kingdom; the Howard College School of Law, University of KwaZulu-Natal, Durban (J.A.S.), and the Wits Reproductive Health and HIV Institute, Johannesburg (H.R.) - both in South Africa; the Dalla Lana School of Public Health, University of Toronto, Toronto (J.A.S.); the Clalit Research Institute, Innovation Division, Clalit Health Services, Tel Aviv, Israel (R.D.B.); the Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany (C.M.-F.); the World Health Organization Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia (K.S.); and the World Health Organization, Geneva (S.B., A.-M.R., R.G., M.D.V.K., S.S., M.J.R., A.-M.H.-R.)
| | - Sylvie Briand
- From the Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD (P.R.K., M.G.); the Department of Biostatistics, University of Washington, Seattle (T.R.F.); the Department of Biostatistics, University of Florida, Gainesville (I.M.L.); the Nuffield Department of Population Health, University of Oxford (R.P., V.B.), and the Oxford Vaccine Group, Department of Paediatrics, University of Oxford and National Institute for Health Research Oxford Biomedical Research Centre (M.D.S.), Oxford, and the Global Health Programme, Chatham House (D.L.H.), and the Coalition for Epidemic Preparedness Innovations (J.P.C.), London - all in the United Kingdom; the Howard College School of Law, University of KwaZulu-Natal, Durban (J.A.S.), and the Wits Reproductive Health and HIV Institute, Johannesburg (H.R.) - both in South Africa; the Dalla Lana School of Public Health, University of Toronto, Toronto (J.A.S.); the Clalit Research Institute, Innovation Division, Clalit Health Services, Tel Aviv, Israel (R.D.B.); the Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany (C.M.-F.); the World Health Organization Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia (K.S.); and the World Health Organization, Geneva (S.B., A.-M.R., R.G., M.D.V.K., S.S., M.J.R., A.-M.H.-R.)
| | - David L Heymann
- From the Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD (P.R.K., M.G.); the Department of Biostatistics, University of Washington, Seattle (T.R.F.); the Department of Biostatistics, University of Florida, Gainesville (I.M.L.); the Nuffield Department of Population Health, University of Oxford (R.P., V.B.), and the Oxford Vaccine Group, Department of Paediatrics, University of Oxford and National Institute for Health Research Oxford Biomedical Research Centre (M.D.S.), Oxford, and the Global Health Programme, Chatham House (D.L.H.), and the Coalition for Epidemic Preparedness Innovations (J.P.C.), London - all in the United Kingdom; the Howard College School of Law, University of KwaZulu-Natal, Durban (J.A.S.), and the Wits Reproductive Health and HIV Institute, Johannesburg (H.R.) - both in South Africa; the Dalla Lana School of Public Health, University of Toronto, Toronto (J.A.S.); the Clalit Research Institute, Innovation Division, Clalit Health Services, Tel Aviv, Israel (R.D.B.); the Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany (C.M.-F.); the World Health Organization Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia (K.S.); and the World Health Organization, Geneva (S.B., A.-M.R., R.G., M.D.V.K., S.S., M.J.R., A.-M.H.-R.)
| | - Valerie Beral
- From the Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD (P.R.K., M.G.); the Department of Biostatistics, University of Washington, Seattle (T.R.F.); the Department of Biostatistics, University of Florida, Gainesville (I.M.L.); the Nuffield Department of Population Health, University of Oxford (R.P., V.B.), and the Oxford Vaccine Group, Department of Paediatrics, University of Oxford and National Institute for Health Research Oxford Biomedical Research Centre (M.D.S.), Oxford, and the Global Health Programme, Chatham House (D.L.H.), and the Coalition for Epidemic Preparedness Innovations (J.P.C.), London - all in the United Kingdom; the Howard College School of Law, University of KwaZulu-Natal, Durban (J.A.S.), and the Wits Reproductive Health and HIV Institute, Johannesburg (H.R.) - both in South Africa; the Dalla Lana School of Public Health, University of Toronto, Toronto (J.A.S.); the Clalit Research Institute, Innovation Division, Clalit Health Services, Tel Aviv, Israel (R.D.B.); the Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany (C.M.-F.); the World Health Organization Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia (K.S.); and the World Health Organization, Geneva (S.B., A.-M.R., R.G., M.D.V.K., S.S., M.J.R., A.-M.H.-R.)
| | - Matthew D Snape
- From the Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD (P.R.K., M.G.); the Department of Biostatistics, University of Washington, Seattle (T.R.F.); the Department of Biostatistics, University of Florida, Gainesville (I.M.L.); the Nuffield Department of Population Health, University of Oxford (R.P., V.B.), and the Oxford Vaccine Group, Department of Paediatrics, University of Oxford and National Institute for Health Research Oxford Biomedical Research Centre (M.D.S.), Oxford, and the Global Health Programme, Chatham House (D.L.H.), and the Coalition for Epidemic Preparedness Innovations (J.P.C.), London - all in the United Kingdom; the Howard College School of Law, University of KwaZulu-Natal, Durban (J.A.S.), and the Wits Reproductive Health and HIV Institute, Johannesburg (H.R.) - both in South Africa; the Dalla Lana School of Public Health, University of Toronto, Toronto (J.A.S.); the Clalit Research Institute, Innovation Division, Clalit Health Services, Tel Aviv, Israel (R.D.B.); the Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany (C.M.-F.); the World Health Organization Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia (K.S.); and the World Health Organization, Geneva (S.B., A.-M.R., R.G., M.D.V.K., S.S., M.J.R., A.-M.H.-R.)
| | - Helen Rees
- From the Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD (P.R.K., M.G.); the Department of Biostatistics, University of Washington, Seattle (T.R.F.); the Department of Biostatistics, University of Florida, Gainesville (I.M.L.); the Nuffield Department of Population Health, University of Oxford (R.P., V.B.), and the Oxford Vaccine Group, Department of Paediatrics, University of Oxford and National Institute for Health Research Oxford Biomedical Research Centre (M.D.S.), Oxford, and the Global Health Programme, Chatham House (D.L.H.), and the Coalition for Epidemic Preparedness Innovations (J.P.C.), London - all in the United Kingdom; the Howard College School of Law, University of KwaZulu-Natal, Durban (J.A.S.), and the Wits Reproductive Health and HIV Institute, Johannesburg (H.R.) - both in South Africa; the Dalla Lana School of Public Health, University of Toronto, Toronto (J.A.S.); the Clalit Research Institute, Innovation Division, Clalit Health Services, Tel Aviv, Israel (R.D.B.); the Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany (C.M.-F.); the World Health Organization Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia (K.S.); and the World Health Organization, Geneva (S.B., A.-M.R., R.G., M.D.V.K., S.S., M.J.R., A.-M.H.-R.)
| | - Alba-Maria Ropero
- From the Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD (P.R.K., M.G.); the Department of Biostatistics, University of Washington, Seattle (T.R.F.); the Department of Biostatistics, University of Florida, Gainesville (I.M.L.); the Nuffield Department of Population Health, University of Oxford (R.P., V.B.), and the Oxford Vaccine Group, Department of Paediatrics, University of Oxford and National Institute for Health Research Oxford Biomedical Research Centre (M.D.S.), Oxford, and the Global Health Programme, Chatham House (D.L.H.), and the Coalition for Epidemic Preparedness Innovations (J.P.C.), London - all in the United Kingdom; the Howard College School of Law, University of KwaZulu-Natal, Durban (J.A.S.), and the Wits Reproductive Health and HIV Institute, Johannesburg (H.R.) - both in South Africa; the Dalla Lana School of Public Health, University of Toronto, Toronto (J.A.S.); the Clalit Research Institute, Innovation Division, Clalit Health Services, Tel Aviv, Israel (R.D.B.); the Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany (C.M.-F.); the World Health Organization Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia (K.S.); and the World Health Organization, Geneva (S.B., A.-M.R., R.G., M.D.V.K., S.S., M.J.R., A.-M.H.-R.)
| | - Ran D Balicer
- From the Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD (P.R.K., M.G.); the Department of Biostatistics, University of Washington, Seattle (T.R.F.); the Department of Biostatistics, University of Florida, Gainesville (I.M.L.); the Nuffield Department of Population Health, University of Oxford (R.P., V.B.), and the Oxford Vaccine Group, Department of Paediatrics, University of Oxford and National Institute for Health Research Oxford Biomedical Research Centre (M.D.S.), Oxford, and the Global Health Programme, Chatham House (D.L.H.), and the Coalition for Epidemic Preparedness Innovations (J.P.C.), London - all in the United Kingdom; the Howard College School of Law, University of KwaZulu-Natal, Durban (J.A.S.), and the Wits Reproductive Health and HIV Institute, Johannesburg (H.R.) - both in South Africa; the Dalla Lana School of Public Health, University of Toronto, Toronto (J.A.S.); the Clalit Research Institute, Innovation Division, Clalit Health Services, Tel Aviv, Israel (R.D.B.); the Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany (C.M.-F.); the World Health Organization Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia (K.S.); and the World Health Organization, Geneva (S.B., A.-M.R., R.G., M.D.V.K., S.S., M.J.R., A.-M.H.-R.)
| | - Jakob P Cramer
- From the Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD (P.R.K., M.G.); the Department of Biostatistics, University of Washington, Seattle (T.R.F.); the Department of Biostatistics, University of Florida, Gainesville (I.M.L.); the Nuffield Department of Population Health, University of Oxford (R.P., V.B.), and the Oxford Vaccine Group, Department of Paediatrics, University of Oxford and National Institute for Health Research Oxford Biomedical Research Centre (M.D.S.), Oxford, and the Global Health Programme, Chatham House (D.L.H.), and the Coalition for Epidemic Preparedness Innovations (J.P.C.), London - all in the United Kingdom; the Howard College School of Law, University of KwaZulu-Natal, Durban (J.A.S.), and the Wits Reproductive Health and HIV Institute, Johannesburg (H.R.) - both in South Africa; the Dalla Lana School of Public Health, University of Toronto, Toronto (J.A.S.); the Clalit Research Institute, Innovation Division, Clalit Health Services, Tel Aviv, Israel (R.D.B.); the Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany (C.M.-F.); the World Health Organization Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia (K.S.); and the World Health Organization, Geneva (S.B., A.-M.R., R.G., M.D.V.K., S.S., M.J.R., A.-M.H.-R.)
| | - César Muñoz-Fontela
- From the Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD (P.R.K., M.G.); the Department of Biostatistics, University of Washington, Seattle (T.R.F.); the Department of Biostatistics, University of Florida, Gainesville (I.M.L.); the Nuffield Department of Population Health, University of Oxford (R.P., V.B.), and the Oxford Vaccine Group, Department of Paediatrics, University of Oxford and National Institute for Health Research Oxford Biomedical Research Centre (M.D.S.), Oxford, and the Global Health Programme, Chatham House (D.L.H.), and the Coalition for Epidemic Preparedness Innovations (J.P.C.), London - all in the United Kingdom; the Howard College School of Law, University of KwaZulu-Natal, Durban (J.A.S.), and the Wits Reproductive Health and HIV Institute, Johannesburg (H.R.) - both in South Africa; the Dalla Lana School of Public Health, University of Toronto, Toronto (J.A.S.); the Clalit Research Institute, Innovation Division, Clalit Health Services, Tel Aviv, Israel (R.D.B.); the Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany (C.M.-F.); the World Health Organization Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia (K.S.); and the World Health Organization, Geneva (S.B., A.-M.R., R.G., M.D.V.K., S.S., M.J.R., A.-M.H.-R.)
| | - Marion Gruber
- From the Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD (P.R.K., M.G.); the Department of Biostatistics, University of Washington, Seattle (T.R.F.); the Department of Biostatistics, University of Florida, Gainesville (I.M.L.); the Nuffield Department of Population Health, University of Oxford (R.P., V.B.), and the Oxford Vaccine Group, Department of Paediatrics, University of Oxford and National Institute for Health Research Oxford Biomedical Research Centre (M.D.S.), Oxford, and the Global Health Programme, Chatham House (D.L.H.), and the Coalition for Epidemic Preparedness Innovations (J.P.C.), London - all in the United Kingdom; the Howard College School of Law, University of KwaZulu-Natal, Durban (J.A.S.), and the Wits Reproductive Health and HIV Institute, Johannesburg (H.R.) - both in South Africa; the Dalla Lana School of Public Health, University of Toronto, Toronto (J.A.S.); the Clalit Research Institute, Innovation Division, Clalit Health Services, Tel Aviv, Israel (R.D.B.); the Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany (C.M.-F.); the World Health Organization Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia (K.S.); and the World Health Organization, Geneva (S.B., A.-M.R., R.G., M.D.V.K., S.S., M.J.R., A.-M.H.-R.)
| | - Rogerio Gaspar
- From the Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD (P.R.K., M.G.); the Department of Biostatistics, University of Washington, Seattle (T.R.F.); the Department of Biostatistics, University of Florida, Gainesville (I.M.L.); the Nuffield Department of Population Health, University of Oxford (R.P., V.B.), and the Oxford Vaccine Group, Department of Paediatrics, University of Oxford and National Institute for Health Research Oxford Biomedical Research Centre (M.D.S.), Oxford, and the Global Health Programme, Chatham House (D.L.H.), and the Coalition for Epidemic Preparedness Innovations (J.P.C.), London - all in the United Kingdom; the Howard College School of Law, University of KwaZulu-Natal, Durban (J.A.S.), and the Wits Reproductive Health and HIV Institute, Johannesburg (H.R.) - both in South Africa; the Dalla Lana School of Public Health, University of Toronto, Toronto (J.A.S.); the Clalit Research Institute, Innovation Division, Clalit Health Services, Tel Aviv, Israel (R.D.B.); the Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany (C.M.-F.); the World Health Organization Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia (K.S.); and the World Health Organization, Geneva (S.B., A.-M.R., R.G., M.D.V.K., S.S., M.J.R., A.-M.H.-R.)
| | - Jerome A Singh
- From the Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD (P.R.K., M.G.); the Department of Biostatistics, University of Washington, Seattle (T.R.F.); the Department of Biostatistics, University of Florida, Gainesville (I.M.L.); the Nuffield Department of Population Health, University of Oxford (R.P., V.B.), and the Oxford Vaccine Group, Department of Paediatrics, University of Oxford and National Institute for Health Research Oxford Biomedical Research Centre (M.D.S.), Oxford, and the Global Health Programme, Chatham House (D.L.H.), and the Coalition for Epidemic Preparedness Innovations (J.P.C.), London - all in the United Kingdom; the Howard College School of Law, University of KwaZulu-Natal, Durban (J.A.S.), and the Wits Reproductive Health and HIV Institute, Johannesburg (H.R.) - both in South Africa; the Dalla Lana School of Public Health, University of Toronto, Toronto (J.A.S.); the Clalit Research Institute, Innovation Division, Clalit Health Services, Tel Aviv, Israel (R.D.B.); the Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany (C.M.-F.); the World Health Organization Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia (K.S.); and the World Health Organization, Geneva (S.B., A.-M.R., R.G., M.D.V.K., S.S., M.J.R., A.-M.H.-R.)
| | - Kanta Subbarao
- From the Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD (P.R.K., M.G.); the Department of Biostatistics, University of Washington, Seattle (T.R.F.); the Department of Biostatistics, University of Florida, Gainesville (I.M.L.); the Nuffield Department of Population Health, University of Oxford (R.P., V.B.), and the Oxford Vaccine Group, Department of Paediatrics, University of Oxford and National Institute for Health Research Oxford Biomedical Research Centre (M.D.S.), Oxford, and the Global Health Programme, Chatham House (D.L.H.), and the Coalition for Epidemic Preparedness Innovations (J.P.C.), London - all in the United Kingdom; the Howard College School of Law, University of KwaZulu-Natal, Durban (J.A.S.), and the Wits Reproductive Health and HIV Institute, Johannesburg (H.R.) - both in South Africa; the Dalla Lana School of Public Health, University of Toronto, Toronto (J.A.S.); the Clalit Research Institute, Innovation Division, Clalit Health Services, Tel Aviv, Israel (R.D.B.); the Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany (C.M.-F.); the World Health Organization Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia (K.S.); and the World Health Organization, Geneva (S.B., A.-M.R., R.G., M.D.V.K., S.S., M.J.R., A.-M.H.-R.)
| | - Maria D Van Kerkhove
- From the Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD (P.R.K., M.G.); the Department of Biostatistics, University of Washington, Seattle (T.R.F.); the Department of Biostatistics, University of Florida, Gainesville (I.M.L.); the Nuffield Department of Population Health, University of Oxford (R.P., V.B.), and the Oxford Vaccine Group, Department of Paediatrics, University of Oxford and National Institute for Health Research Oxford Biomedical Research Centre (M.D.S.), Oxford, and the Global Health Programme, Chatham House (D.L.H.), and the Coalition for Epidemic Preparedness Innovations (J.P.C.), London - all in the United Kingdom; the Howard College School of Law, University of KwaZulu-Natal, Durban (J.A.S.), and the Wits Reproductive Health and HIV Institute, Johannesburg (H.R.) - both in South Africa; the Dalla Lana School of Public Health, University of Toronto, Toronto (J.A.S.); the Clalit Research Institute, Innovation Division, Clalit Health Services, Tel Aviv, Israel (R.D.B.); the Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany (C.M.-F.); the World Health Organization Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia (K.S.); and the World Health Organization, Geneva (S.B., A.-M.R., R.G., M.D.V.K., S.S., M.J.R., A.-M.H.-R.)
| | - Soumya Swaminathan
- From the Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD (P.R.K., M.G.); the Department of Biostatistics, University of Washington, Seattle (T.R.F.); the Department of Biostatistics, University of Florida, Gainesville (I.M.L.); the Nuffield Department of Population Health, University of Oxford (R.P., V.B.), and the Oxford Vaccine Group, Department of Paediatrics, University of Oxford and National Institute for Health Research Oxford Biomedical Research Centre (M.D.S.), Oxford, and the Global Health Programme, Chatham House (D.L.H.), and the Coalition for Epidemic Preparedness Innovations (J.P.C.), London - all in the United Kingdom; the Howard College School of Law, University of KwaZulu-Natal, Durban (J.A.S.), and the Wits Reproductive Health and HIV Institute, Johannesburg (H.R.) - both in South Africa; the Dalla Lana School of Public Health, University of Toronto, Toronto (J.A.S.); the Clalit Research Institute, Innovation Division, Clalit Health Services, Tel Aviv, Israel (R.D.B.); the Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany (C.M.-F.); the World Health Organization Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia (K.S.); and the World Health Organization, Geneva (S.B., A.-M.R., R.G., M.D.V.K., S.S., M.J.R., A.-M.H.-R.)
| | - Michael J Ryan
- From the Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD (P.R.K., M.G.); the Department of Biostatistics, University of Washington, Seattle (T.R.F.); the Department of Biostatistics, University of Florida, Gainesville (I.M.L.); the Nuffield Department of Population Health, University of Oxford (R.P., V.B.), and the Oxford Vaccine Group, Department of Paediatrics, University of Oxford and National Institute for Health Research Oxford Biomedical Research Centre (M.D.S.), Oxford, and the Global Health Programme, Chatham House (D.L.H.), and the Coalition for Epidemic Preparedness Innovations (J.P.C.), London - all in the United Kingdom; the Howard College School of Law, University of KwaZulu-Natal, Durban (J.A.S.), and the Wits Reproductive Health and HIV Institute, Johannesburg (H.R.) - both in South Africa; the Dalla Lana School of Public Health, University of Toronto, Toronto (J.A.S.); the Clalit Research Institute, Innovation Division, Clalit Health Services, Tel Aviv, Israel (R.D.B.); the Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany (C.M.-F.); the World Health Organization Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia (K.S.); and the World Health Organization, Geneva (S.B., A.-M.R., R.G., M.D.V.K., S.S., M.J.R., A.-M.H.-R.)
| | - Ana-Maria Henao-Restrepo
- From the Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD (P.R.K., M.G.); the Department of Biostatistics, University of Washington, Seattle (T.R.F.); the Department of Biostatistics, University of Florida, Gainesville (I.M.L.); the Nuffield Department of Population Health, University of Oxford (R.P., V.B.), and the Oxford Vaccine Group, Department of Paediatrics, University of Oxford and National Institute for Health Research Oxford Biomedical Research Centre (M.D.S.), Oxford, and the Global Health Programme, Chatham House (D.L.H.), and the Coalition for Epidemic Preparedness Innovations (J.P.C.), London - all in the United Kingdom; the Howard College School of Law, University of KwaZulu-Natal, Durban (J.A.S.), and the Wits Reproductive Health and HIV Institute, Johannesburg (H.R.) - both in South Africa; the Dalla Lana School of Public Health, University of Toronto, Toronto (J.A.S.); the Clalit Research Institute, Innovation Division, Clalit Health Services, Tel Aviv, Israel (R.D.B.); the Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany (C.M.-F.); the World Health Organization Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia (K.S.); and the World Health Organization, Geneva (S.B., A.-M.R., R.G., M.D.V.K., S.S., M.J.R., A.-M.H.-R.)
| |
Collapse
|
36
|
Stoehr JR, Hamidian Jahromi A, Thomason C. Ethical Considerations for Unblinding and Vaccinating COVID-19 Vaccine Trial Placebo Group Participants. Front Public Health 2021; 9:702960. [PMID: 34249853 PMCID: PMC8264198 DOI: 10.3389/fpubh.2021.702960] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 05/28/2021] [Indexed: 11/24/2022] Open
Affiliation(s)
- Jenna Rose Stoehr
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Alireza Hamidian Jahromi
- Plastic and Reconstructive Surgery Department, Rush University Medical Center, Chicago, IL, United States
- The Center for Gender Confirmation Surgery, Weiss Memorial Hospital, The University of Illinois at Chicago, Chicago, IL, United States
| | - Clayton Thomason
- Department of Religion, Health and Human Values, Rush University Medical Center, Chicago, IL, United States
| |
Collapse
|
37
|
Lefebvre M, Vignier N, Pitard B, Botelho-Nevers E, Wyplosz B, Cohen R, Epaulard O. COVID-19 vaccines: Frequently asked questions and updated answers. Infect Dis Now 2021; 51:319-333. [PMID: 33681861 PMCID: PMC7910656 DOI: 10.1016/j.idnow.2021.02.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 02/23/2021] [Indexed: 12/29/2022]
Abstract
At the end of December 2019, China notified the World Health Organization about a viral pneumonia epidemic soon to be named COVID-19, of which the infectious agent, SARS-CoV-2, was rapidly identified. Less than one year later, published phase 3 clinical trials underlined the effectiveness of vaccines utilizing hitherto unusual technology consisting in injection of the messenger RNA (m-RNA) of a viral protein. In the meantime, numerous clinical trials had failed to identify a maximally effective antiviral treatment, and mass vaccination came to be considered as the strategy most likely to put an end to the pandemic. The objective of this text is to address and hopefully answer the questions being put forward by healthcare professionals on the different anti-SARS-CoV-2 vaccines as regards their development, their modes of action, their effectiveness, their limits, and their utilization in different situations; we are proposing a report on both today's state of knowledge, and the 14 February 2021 recommendations of the French health authorities.
Collapse
Affiliation(s)
- M Lefebvre
- Service des maladies infectieuses et tropicales, centre de prévention des maladies infectieuses et transmissibles, centre hospitalo-universitaire Hôtel-Dieu, Inserm CIC1413, 1, place Alexis-Ricordeau, 44000 Nantes, France.
| | - N Vignier
- Centre d'investigation clinique Antilles Guyane, CIC Inserm 1424, DRISP, centre hospitalier Andrée-Rosemon, Cayenne, French Guyana; Inserm, Sorbonne université, institut Pierre-Louis d'épidémiologie et de santé publique, IPLESP, 75012 Paris, France; Department of infectious disease, Groupe hospitalier Sud Ile-de-France, 77000 Melun, France
| | - B Pitard
- Université de Nantes, CNRS ERL6001, Inserm 1232, CRCINA, Nantes, France
| | - E Botelho-Nevers
- Service d'infectiologie, centre hospitalo-universitaire de Saint-Étienne, CIC 1408 Inserm, 42055 Saint-Étienne, France; Centre international de recherche en infectiologie (CIRI), Team GIMAP, université Lyon, université Jean-Monnet, université Claude-Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, 42023 Saint-Étienne, France
| | - B Wyplosz
- Service des maladies infectieuses et tropicales, Assistance publique-hôpitaux de Paris, Centre hospitalier universitaire Bicêtre, Paris, France
| | - R Cohen
- InfoVac, centre hospitalier intercommunal de Créteil, service de pédiatrie, 40, avenue de Verdun, 94000 Créteil, France
| | - O Epaulard
- Service des maladies infectieuses, centre hospitalo-universitaire Grenoble Alpes, Grenoble, France, CIC 1406 Inserm, Grenoble, France
| |
Collapse
|
38
|
Dal-Ré R, Bekker LG, Gluud C, Holm S, Jha V, Poland GA, Rosendaal FR, Schwarzer-Daum B, Sevene E, Tinto H, Voo TC, Sreeharan N. Ongoing and future COVID-19 vaccine clinical trials: challenges and opportunities. THE LANCET. INFECTIOUS DISEASES 2021; 21:e342-e347. [PMID: 34019801 PMCID: PMC8131060 DOI: 10.1016/s1473-3099(21)00263-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 12/18/2022]
Abstract
Large-scale deployment of COVID-19 vaccines will seriously affect the ongoing phases 2 and 3 randomised placebo-controlled trials assessing SARS-CoV-2 vaccine candidates. The effect will be particularly acute in high-income countries where the entire adult or older population could be vaccinated by late 2021. Regrettably, only a small proportion of the population in many low-income and middle-income countries will have access to available vaccines. Sponsors of COVID-19 vaccine candidates currently in phase 2 or initiating phase 3 trials in 2021 should consider continuing the research in countries with limited affordability and availability of COVID-19 vaccines. Several ethical principles must be implemented to ensure the equitable, non-exploitative, and respectful conduct of trials in resource-poor settings. Once sufficient knowledge on the immunogenicity response to COVID-19 vaccines is acquired, non-inferiority immunogenicity trials—comparing the immune response of a vaccine candidate to that of an authorised vaccine—would probably be the most common trial design. Until then, placebo-controlled, double-blind, crossover trials will continue to play a role in the development of new vaccine candidates. WHO or the Council for International Organizations of Medical Sciences should define an ethical framework for the requirements and benefits for trial participants and host communities in resource-poor settings that should require commitment from all vaccine candidate sponsors from high-income countries.
Collapse
Affiliation(s)
- Rafael Dal-Ré
- Epidemiology Unit, Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid, Madrid, Spain.
| | - Linda-Gail Bekker
- The Desmond Tutu HIV Centre, University of Cape Town, Cape Town, South Africa
| | - Christian Gluud
- The Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region, Rigshospitalet, Copenhagen, Denmark; Institute for Regional Health Research, The Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Søren Holm
- Centre for Social Ethics and Policy, Department of Law, University of Manchester, Manchester, UK
| | - Vivekanand Jha
- George Institute for Global Health, UNSW, New Delhi, India; Manipal Academy of Higher Education, Manipal, India
| | - Gregory A Poland
- Mayo Clinic Vaccine Research Group and Division of General Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Frits R Rosendaal
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Esperança Sevene
- Department of Physiological Sciences, Faculty of Medicine, Eduardo Mondlane University, Maputo, Mozambique; Manhiça Health Research Centre, Maputo, Mozambique
| | - Halidou Tinto
- Clinical Research Unit of Nanoro, Institut de Recherche en Sciences de La Santé, Nanoro, Burkina Faso
| | - Teck Chuan Voo
- Centre for Biomedical Ethics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | |
Collapse
|
39
|
Torres I, Lopez-Cevallos D, Artaza O, Profeta B, Kang J, Machado CV. Vaccine scarcity in LMICs is a failure of global solidarity and multilateral instruments. Lancet 2021; 397:1804. [PMID: 33992137 PMCID: PMC8118613 DOI: 10.1016/s0140-6736(21)00893-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/09/2021] [Indexed: 11/05/2022]
Affiliation(s)
| | - Daniel Lopez-Cevallos
- School of Language, Culture, and Society, Oregon State University, Corvallis, OR, USA
| | - Osvaldo Artaza
- Facultad de Ciencias de la Salud, Universidad de las Américas, Santiago de Chile, Chile
| | | | - JaHyun Kang
- College of Nursing and Research Institute of Nursing Science, Seoul National University, Seoul, South Korea
| | | |
Collapse
|
40
|
Abstract
This minireview addresses problems of financing the vaccine development, regulatory questions, the ethics and efficacy of vaccine prioritization strategies and the coverage of variant viruses by current vaccines. Serious adverse effects observed with adenovirus vectored vaccines and mRNA vaccines in mass vaccination campaigns are reported. The ethical problems of continuing with placebo controlled vaccine trials and alternative clinical trial protocols are discussed as well as concrete vaccination issues such as the splitting of doses, the delaying of the second dose, the immunization with two different vaccine types and the need of vaccinating seropositive subjects. Strategies to increase vaccine acceptance in the population are shortly mentioned.
Collapse
Affiliation(s)
- Harald Brüssow
- Department of Biosystems, Laboratory of Gene Technology, KU Leuven, Leuven, Belgium
| |
Collapse
|
41
|
Excler JL, Saville M, Berkley S, Kim JH. Vaccine development for emerging infectious diseases. Nat Med 2021; 27:591-600. [PMID: 33846611 DOI: 10.1038/s41591-021-01301-0] [Citation(s) in RCA: 189] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 03/01/2021] [Indexed: 01/19/2023]
Abstract
Examination of the vaccine strategies and technical platforms used for the COVID-19 pandemic in the context of those used for previous emerging and reemerging infectious diseases and pandemics may offer some mutually beneficial lessons. The unprecedented scale and rapidity of dissemination of recent emerging infectious diseases pose new challenges for vaccine developers, regulators, health authorities and political constituencies. Vaccine manufacturing and distribution are complex and challenging. While speed is essential, clinical development to emergency use authorization and licensure, pharmacovigilance of vaccine safety and surveillance of virus variants are also critical. Access to vaccines and vaccination needs to be prioritized in low- and middle-income countries. The combination of these factors will weigh heavily on the ultimate success of efforts to bring the current and any future emerging infectious disease pandemics to a close.
Collapse
Affiliation(s)
| | - Melanie Saville
- Coalition for Epidemic Preparedness Innovations (CEPI), London, UK
| | | | - Jerome H Kim
- International Vaccine Institute, Seoul, Republic of Korea.
| |
Collapse
|
42
|
Caplan AI. Placebo Controls: Now??? Arch Immunol Ther Exp (Warsz) 2021; 69:9. [PMID: 33782781 PMCID: PMC8006883 DOI: 10.1007/s00005-021-00612-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 03/22/2021] [Indexed: 11/05/2022]
Affiliation(s)
- Arnold I Caplan
- Biology, Skeletal Research Center, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
43
|
Friesen P, Caplan AL, Miller JE. COVID-19 vaccine research and the trouble with clinical equipoise. Lancet 2021; 397:576. [PMID: 33539728 PMCID: PMC7906638 DOI: 10.1016/s0140-6736(21)00198-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 01/12/2021] [Indexed: 01/05/2023]
Affiliation(s)
- Phoebe Friesen
- Biomedical Ethics Unit, McGill University, Montreal, QC, Canada
| | - Arthur L Caplan
- Division of Medical Ethics, New York University, New York, NY, USA
| | - Jennifer E Miller
- Yale School of Medicine, Yale University, New Haven, CT 06520, USA; Bioethics International, New York, NY, USA.
| |
Collapse
|
44
|
Bell KJL, Glasziou P, Stanaway F, Bossuyt P, Irwig L. Equity and evidence during vaccine rollout: stepped wedge cluster randomised trials could help. BMJ 2021; 372:n435. [PMID: 33579675 DOI: 10.1136/bmj.n435] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Katy J L Bell
- Edward Ford Building (A27), School of Public Health, University of Sydney, NSW 2006, Australia
| | - Paul Glasziou
- Institute for Evidence-Based Healthcare, Faculty of Health Sciences and Medicine, Bond University, Robina, QLD 4226, Australia
| | - Fiona Stanaway
- Edward Ford Building (A27), School of Public Health, University of Sydney, NSW 2006, Australia
| | - Patrick Bossuyt
- Department of Epidemiology and Data Science, Amsterdam Public Health, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Les Irwig
- Edward Ford Building (A27), School of Public Health, University of Sydney, NSW 2006, Australia
| |
Collapse
|
45
|
Fleming TR, Krause PR, Nason M, Longini IM, Henao-Restrepo AMM. COVID-19 vaccine trials: The use of active controls and non-inferiority studies. Clin Trials 2021; 18:335-342. [PMID: 33535811 PMCID: PMC8172418 DOI: 10.1177/1740774520988244] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Background: Recently emerging results from a few placebo-controlled randomized trials of COVID-19 vaccines revealed estimates of 62%–95% relative reductions in risk of virologically confirmed symptomatic COVID-19 disease, over approximately 2-month average follow-up period. Additional safe and effective COVID-19 vaccines are needed in a timely manner to adequately address the pandemic on an international scale. Such safe and effective vaccines would be especially appealing for international deployment if they also have favorable stability, supply, and potential for implementation in mass vaccination campaigns. Randomized trials provide particularly reliable insights about vaccine efficacy and safety. While enhanced efficiency and interpretability can be obtained from placebo-controlled trials, in settings where their conduct is no longer possible, randomized non-inferiority trials may enable obtaining reliable evaluations of experimental vaccines through direct comparison with active comparator vaccines established to have worthwhile efficacy. Methods: The usual objective of non-inferiority trials is to reliably assess whether the efficacy of an experimental vaccine is not unacceptably worse than that of an active control vaccine previously established to be effective, likely in a placebo-controlled trial. This is formally achieved by ruling out a non-inferiority margin identified to be the minimum threshold for what would constitute an unacceptable loss of efficacy. This article not only investigates non-inferiority margins, denoted by δ, that address the usual objective of determining whether the experimental vaccine is “at least similarly effective to” the active comparator vaccine in the non-inferiority trial, but also develops non-inferiority margins, denoted by δo, intended to address the worldwide need for multiple safe and effective vaccines by satisfying the less stringent requirement that the experimental vaccine be “at least similarly effective to” an active comparator vaccine having efficacy that satisfies the widely accepted World Health Organization–Food and Drug Administration criteria for “worthwhile” vaccine efficacy. Results: Using the margin δ enables non-inferiority trials to reliably evaluate experimental vaccines that truly are similarly effective to an active comparator vaccine having any level of “worthwhile” efficacy. When active comparator vaccines have efficacy in the range of 50%–70%, non-inferiority trials designed to use the margin δo have appealing properties, especially for experimental vaccines having true efficacy of approximately 60%. Conclusion: Non-inferiority trials using the proposed margins may enable reliable randomized evaluations of efficacy and safety of experimental COVID-19 vaccines. Such trials often require approximately two- to three-fold the person-years follow-up than a placebo-controlled trial. This could be achieved, without substantive increases in sample size, by increasing the average duration of follow-up from 2 months to approximately 4–6 months, assuming efficacy of the active comparator vaccine has been reliably evaluated over that longer duration.
Collapse
Affiliation(s)
- Thomas R Fleming
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Philip R Krause
- Office of Vaccines Research and Review, FDA/CBER, Silver Spring, MD, USA
| | - Martha Nason
- National Institute of Allergy and Infectious Diseases (NIAID/NIH), Bethesda, MD, USA
| | - Ira M Longini
- Department of Biostatistics, University of Florida, Gainesville, FL, USA
| | | |
Collapse
|
46
|
Fiocchi A, Jensen-Jarolim E. SARS-COV-2, can you be over it? World Allergy Organ J 2021; 14:100514. [PMID: 33552379 PMCID: PMC7846213 DOI: 10.1016/j.waojou.2021.100514] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVES Uncertainty has surrounded the duration of immunity against SARS CoV-2. This concerns both the duration of vaccine immunity and the duration of natural immunity. We aim to critically review the information available today, and draw practical conclusions. METHODS This is a narrative review of the recently published information on the topic, compared with the knowledge we already have of the behavior of various viral infectious agents. RESULTS It is too early to have any meaningful information on the duration of vaccine immunity against SARS CoV-2. For those who already had the infeciton, the rate of reinfection is very low. Most reinfections are due to laboratory errors, to incomplete cure of the primary infection, to the supervening immunodeficiency of the host, or to pre-existing immunodeficiency made evident by the SARS CoV-2 infection. The available studies on the immunology of the infection converge in indicating that it generates a robust and persistent immunity. This behavior does not differ from that of respiratory viruses known to date: in naturally occurring viral respiratory infections, reinfections are exceptional. CONCLUSIONS AND IMPLICATIONS The civil community awaits suggestions from scientists not only to protect susceptible people, but to be able to safely resume activities made uncertain by the pandemic. From the information we have to-date, we suggest that, in principle, patients who have already overcome the infection should not be prioritized to the SARS CoV-2 vaccine. Instead, they could be provided with an immunological passport that allows them to resume a normal social life.
Collapse
Affiliation(s)
- Alessandro Fiocchi
- Allergy Unit, Bambino Gesù Children's Hospital, IRCCS, Piazza Sant'Onofrio 4, 00165, Rome, Italy
| | - Erika Jensen-Jarolim
- The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Vienna, Austria
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectivology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
47
|
Trial participants' rights after authorisation of COVID-19 vaccines. THE LANCET RESPIRATORY MEDICINE 2021; 9:e30-e31. [PMID: 33476582 PMCID: PMC7816575 DOI: 10.1016/s2213-2600(21)00044-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 01/08/2021] [Indexed: 11/22/2022]
|
48
|
Akbar SM, Mahtab MA, Aguilar JC, Uddin MH, Khan MSI, Yoshida O, Penton E, Gerardo GN, Hiasa Y. Role of Pegylated Interferon in Patients with Chronic Liver Diseases in the Context of SARS-CoV-2 Infection. Euroasian J Hepatogastroenterol 2021; 11:27-31. [PMID: 34316461 PMCID: PMC8286362 DOI: 10.5005/jp-journals-10018-1341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The coronavirus 2019 (COVID-19) pandemic has resulted in 168 million cases and about 3.5 million deaths (as of May 26, 2021) during the last 18 months. These 18 months of the COVID-19 pandemic have been characterized by phases or waves of new cases, the emergence of new variants of the deadly virus, and several new complications. After providing emergency approval to several drugs and adherence to several public health measures with frequent full and partial lockdowns, the incidence of new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) could not be contained till now on a global basis. Although prophylactic vaccines have inspired optimism, the scarcity of vaccines and several vaccine-related regulations indicate that the vaccine's benefit would not be reaching the people of developing countries anytime soon. In the course of our clinical practice, we used pegylated interferon (Peg-IFN) in 35 patients with chronic liver diseases (CLD), and we found that only two of them were infected with SARS-CoV-2 that was mild in nature. These two patients with CLD have a mild course of disease cured without any specific therapy. Patients with CLD are usually immune-compromised. However, three CLD patients remained free of SARS-CoV-2 although they had COVID-19 patients among their family members. Next, we accomplished two studies for assessing the immune-modulatory capacities of Peg-IFN, 1 and 12 injections following administration of Peg-IFN. The data revealed that peripheral blood mononuclear cells (PBMCs) of Peg-IFN-administered CLD patients produced significantly higher levels of some cytokines of innate immunity in comparison with the cytokines produced by PBMC of CLD patients before Peg-IFN intake. The pattern of cytokine responses and absence of infection of SARS-CoV-2 in 33 of 35 CLD patients represent some preliminary observations indicating a possible role of Peg-IFN in patients with CLD. The study may be extended to other chronic infections and cancers in which patients receive Peg-IFN. The role of Peg-IFN for pre- or postexposure prophylaxis in the acquisition of SARS-CoV-2 infection and influencing the natural course of COVID-19 remains to be clarified.
Collapse
Affiliation(s)
- Sheikh Mf Akbar
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Mamun A Mahtab
- Department of Hepatology, Bangabandhu Sheikh Mujib Medical University, BSMMU, Dhaka, Bangladesh
| | - Julio C Aguilar
- Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Md H Uddin
- Specialized Liver Center, Dhaka, Bangladesh
| | - Md Sakirul I Khan
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Osamu Yoshida
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Eduardo Penton
- Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | | | - Yoichi Hiasa
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Ehime, Japan
| |
Collapse
|
49
|
|
50
|
Follmann D, Fintzi J, Fay MP, Janes HE, Baden L, Sahly HE, Fleming TR, Mehrotra DV, Carpp LN, Juraska M, Benkeser D, Donnell D, Fong Y, Han S, Hirsch I, Huang Y, Huang Y, Hyrien O, Luedtke A, Carone M, Nason M, Vandebosch A, Zhou H, Cho I, Gabriel E, Kublin JG, Cohen MS, Corey L, Gilbert PB, Neuzil KM. Assessing Durability of Vaccine Effect Following Blinded Crossover in COVID-19 Vaccine Efficacy Trials. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020:2020.12.14.20248137. [PMID: 33336213 PMCID: PMC7745130 DOI: 10.1101/2020.12.14.20248137] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Several candidate vaccines to prevent COVID-19 disease have entered large-scale phase 3 placebo-controlled randomized clinical trials and some have demonstrated substantial short-term efficacy. Efficacious vaccines should, at some point, be offered to placebo participants, which will occur before long-term efficacy and safety are known. METHODS Following vaccination of the placebo group, we show that placebo-controlled vaccine efficacy can be derived by assuming the benefit of vaccination over time has the same profile for the original vaccine recipients and the placebo crossovers. This reconstruction allows estimation of both vaccine durability and potential vaccine-associated enhanced disease. RESULTS Post-crossover estimates of vaccine efficacy can provide insights about durability, identify waning efficacy, and identify late enhancement of disease, but are less reliable estimates than those obtained by a standard trial where the placebo cohort is maintained. As vaccine efficacy estimates for post-crossover periods depend on prior vaccine efficacy estimates, longer pre-crossover periods with higher case counts provide better estimates of late vaccine efficacy. Further, open-label crossover may lead to riskier behavior in the immediate crossover period for the unblinded vaccine arm, confounding vaccine efficacy estimates for all post-crossover periods. CONCLUSIONS We advocate blinded crossover and continued follow-up of trial participants to best assess vaccine durability and potential delayed enhancement of disease. This approach allows placebo recipients timely access to the vaccine when it would no longer be proper to maintain participants on placebo, yet still allows important insights about immunological and clinical effectiveness over time.
Collapse
Affiliation(s)
- Dean Follmann
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Jonathan Fintzi
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Michael P Fay
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Holly E Janes
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | | | - Thomas R Fleming
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Devan V Mehrotra
- Biostatistics and Research Decision Sciences, Merck & Co., Inc., North Wales, PA, USA
| | - Lindsay N Carpp
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Michal Juraska
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - David Benkeser
- Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Deborah Donnell
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Youyi Fong
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Shu Han
- Moderna, Inc., Cambridge, MA, USA
| | - Ian Hirsch
- Biometrics, Late-stage Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Ying Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Yunda Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Ollivier Hyrien
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Alex Luedtke
- Department of Statistics, University of Washington, Seattle, WA, USA
| | - Marco Carone
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Martha Nason
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - An Vandebosch
- Janssen R&D, Janssen Pharmaceuticals NV, Beerse, Belgium
| | | | - Iksung Cho
- Biostatistics, Novavax, Inc., Gaithersburg, MD, USA
| | - Erin Gabriel
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Solna, Sweden
| | - James G Kublin
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Myron S Cohen
- Institute for Global Health and Infectious Diseases, University of North Carolina, Chapel Hill, NC, USA
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Peter B Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | |
Collapse
|