1
|
Messina NL, Germano S, Chung AW, van de Sandt CE, Stevens NE, Allen LF, Bonnici R, Croda J, Counoupas C, Grubor‐Bauk B, Haycroft ER, Kedzierska K, McDonald E, McElroy R, Netea MG, Novakovic B, Perrett KP, Pittet LF, Purcell RA, Subbarao K, Triccas JA, Lynn DJ, Curtis N. Effect of Bacille Calmette-Guérin vaccination on immune responses to SARS-CoV-2 and COVID-19 vaccination. Clin Transl Immunology 2025; 14:e70023. [PMID: 39872402 PMCID: PMC11761716 DOI: 10.1002/cti2.70023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 12/23/2024] [Accepted: 01/01/2025] [Indexed: 01/30/2025] Open
Abstract
Objectives Bacille Calmette-Guérin (BCG) vaccination has off-target effects on disease risk for unrelated infections and immune responses to vaccines. This study aimed to determine the immunomodulatory effects of BCG vaccination on immune responses to vaccines against SARS-CoV-2. Methods Blood samples, from a subset of 275 SARS-CoV-2-naïve healthcare workers randomised to BCG vaccination (BCG group) or no BCG vaccination (Control group) in the BRACE trial, were collected before and 28 days after the primary course (two doses) of ChAdOx1-S (Oxford-AstraZeneca) or BNT162b2 (Pfizer-BioNTech) vaccination. SARS-CoV-2-specific antibodies were measured using ELISA and multiplex bead array, whole blood cytokine responses to γ-irradiated SARS-CoV-2 (iSARS) stimulation were measured by multiplex bead array, and SARS-CoV-2-specific T-cell responses were measured by activation-induced marker and intracellular cytokine staining assays. Results After randomisation (mean 11 months) but prior to COVID-19 vaccination, the BCG group had lower cytokine responses to iSARS stimulation than the Control group. After two doses of ChAdOx1-S, differences in iSARS-induced cytokine responses between the BCG group and Control group were found for three cytokines (CTACK, TRAIL and VEGF). No differences were found between the groups after BNT162b2 vaccination. There were also no differences between the BCG and Control groups in COVID-19 vaccine-induced antigen-specific antibody responses, T-cell activation or T-cell cytokine production. Conclusion BCG vaccination induced a broad and persistent reduction in ex vivo cytokine responses to SARS-CoV-2. Following COVID-19 vaccination, this effect was abrogated, and BCG vaccination did not influence adaptive immune responses to COVID-19 vaccine antigens.
Collapse
Affiliation(s)
- Nicole L Messina
- Infectious Diseases Group, Infection, Immunity and Global Health ThemeMurdoch Children's Research InstituteParkvilleVICAustralia
- Department of PaediatricsThe University of MelbourneParkvilleVICAustralia
| | - Susie Germano
- Infectious Diseases Group, Infection, Immunity and Global Health ThemeMurdoch Children's Research InstituteParkvilleVICAustralia
| | - Amy W Chung
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and ImmunityThe University of MelbourneParkvilleVICAustralia
| | - Carolien E van de Sandt
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and ImmunityThe University of MelbourneParkvilleVICAustralia
| | - Natalie E Stevens
- Precision Medicine ThemeSouth Australian Health and Medical Research InstituteAdelaideSAAustralia
- Flinders Health and Medical Research InstituteFlinders UniversityBedford ParkSAAustralia
| | - Lilith F Allen
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and ImmunityThe University of MelbourneParkvilleVICAustralia
| | - Rhian Bonnici
- Infectious Diseases Group, Infection, Immunity and Global Health ThemeMurdoch Children's Research InstituteParkvilleVICAustralia
| | - Julio Croda
- Universidade Federal de Mato Grosso do Sul‐UFMSCampo GrandeMSBrazil
- Fiocruz Mato Grosso do SulFundação Oswaldo CruzCampo GrandeMSBrazil
- Department of Epidemiology of Microbial DiseasesYale School of Public HealthNew HavenCTUSA
| | - Claudio Counoupas
- School of Medical Sciences, Faculty of Medicine and HealthThe University of SydneyCamperdownNSWAustralia
- Sydney Institute for Infectious Diseases and the Charles Perkins CentreThe University of SydneyCamperdownNSWAustralia
- Centre for Infection and ImmunityCentenary InstituteCamperdownNSWAustralia
| | - Branka Grubor‐Bauk
- Viral Immunology Group, Adelaide Medical School, Basil Hetzel Institute for Translational Health ResearchUniversity of AdelaideAdelaideSAAustralia
| | - Ebene R Haycroft
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and ImmunityThe University of MelbourneParkvilleVICAustralia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and ImmunityThe University of MelbourneParkvilleVICAustralia
| | - Ellie McDonald
- Infectious Diseases Group, Infection, Immunity and Global Health ThemeMurdoch Children's Research InstituteParkvilleVICAustralia
| | - Rebecca McElroy
- Infectious Diseases Group, Infection, Immunity and Global Health ThemeMurdoch Children's Research InstituteParkvilleVICAustralia
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenThe Netherlands
- Department for Immunology and Metabolism, Life and Medical Sciences InstituteUniversity of BonnBonnGermany
| | - Boris Novakovic
- Department of PaediatricsThe University of MelbourneParkvilleVICAustralia
- Molecular Immunity Group, Infection and Immunity ThemeMurdoch Children's Research InstituteParkvilleVICAustralia
| | - Kirsten P Perrett
- Department of PaediatricsThe University of MelbourneParkvilleVICAustralia
- Population Allergy GroupMurdoch Children's Research InstituteParkvilleVICAustralia
- Department of Allergy and ImmunologyThe Royal Children's Hospital MelbourneParkvilleVICAustralia
| | - Laure F Pittet
- Infectious Diseases Group, Infection, Immunity and Global Health ThemeMurdoch Children's Research InstituteParkvilleVICAustralia
- Department of PaediatricsThe University of MelbourneParkvilleVICAustralia
- Immunology, Vaccinology, Rheumatology and Infectious Diseases UnitGeneva University Hospitals and Faculty of MedicineGenevaSwitzerland
| | - Ruth A Purcell
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and ImmunityThe University of MelbourneParkvilleVICAustralia
| | - Kanta Subbarao
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and ImmunityThe University of MelbourneParkvilleVICAustralia
| | - James A Triccas
- School of Medical Sciences, Faculty of Medicine and HealthThe University of SydneyCamperdownNSWAustralia
- Sydney Institute for Infectious Diseases and the Charles Perkins CentreThe University of SydneyCamperdownNSWAustralia
- Centre for Infection and ImmunityCentenary InstituteCamperdownNSWAustralia
| | - David J Lynn
- Precision Medicine ThemeSouth Australian Health and Medical Research InstituteAdelaideSAAustralia
- Flinders Health and Medical Research InstituteFlinders UniversityBedford ParkSAAustralia
| | - Nigel Curtis
- Infectious Diseases Group, Infection, Immunity and Global Health ThemeMurdoch Children's Research InstituteParkvilleVICAustralia
- Department of PaediatricsThe University of MelbourneParkvilleVICAustralia
- Department of Infectious DiseasesThe Royal Children's Hospital MelbourneParkvilleVICAustralia
| |
Collapse
|
2
|
Mambelli F, de Araujo ACVSC, Farias JP, de Andrade KQ, Ferreira LCS, Minoprio P, Leite LCC, Oliveira SC. An Update on Anti-COVID-19 Vaccines and the Challenges to Protect Against New SARS-CoV-2 Variants. Pathogens 2025; 14:23. [PMID: 39860984 PMCID: PMC11768231 DOI: 10.3390/pathogens14010023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/11/2024] [Accepted: 12/20/2024] [Indexed: 01/27/2025] Open
Abstract
The COVID-19 pandemic has posed a significant threat to global health systems, with extensive impacts across many sectors of society. The pandemic has been responsible for millions of deaths worldwide since its first identification in late 2019. Several actions have been taken to prevent the disease, including the unprecedented fast development and global vaccination campaigns, which were pivotal in reducing symptoms and deaths. Given the impact of the pandemic, the continuous changes of the virus, and present vaccine technologies, this review analyzes how, so far, we have met the challenge posed by the emergence of new variants and discusses how next-generation pan-coronavirus vaccines, with enhanced longevity and breadth of immune responses, may be tackled with alternative administration routes and antigen delivery platforms. By addressing these critical aspects, this review aims to contribute to the ongoing efforts to achieve long-term control of COVID-19, stimulating the discussion and work on next-generation vaccines capable of facing future waves of infection.
Collapse
Affiliation(s)
- Fábio Mambelli
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil; (F.M.); (A.C.V.S.C.d.A.); (K.Q.d.A.)
- Institut Pasteur de São Paulo, São Paulo 05508-020, Brazil; (L.C.S.F.); (P.M.)
| | - Ana Carolina V. S. C. de Araujo
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil; (F.M.); (A.C.V.S.C.d.A.); (K.Q.d.A.)
| | - Jéssica P. Farias
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil;
| | - Kivia Q. de Andrade
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil; (F.M.); (A.C.V.S.C.d.A.); (K.Q.d.A.)
| | - Luis C. S. Ferreira
- Institut Pasteur de São Paulo, São Paulo 05508-020, Brazil; (L.C.S.F.); (P.M.)
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil;
| | - Paola Minoprio
- Institut Pasteur de São Paulo, São Paulo 05508-020, Brazil; (L.C.S.F.); (P.M.)
| | - Luciana C. C. Leite
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo 05503-900, Brazil;
| | - Sergio C. Oliveira
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil; (F.M.); (A.C.V.S.C.d.A.); (K.Q.d.A.)
- Institut Pasteur de São Paulo, São Paulo 05508-020, Brazil; (L.C.S.F.); (P.M.)
| |
Collapse
|
3
|
Jalalizadeh M, Buosi K, Giacomelli CF, Leme PAF, Ferrari KL, Dionato FAV, Brito WRS, Brunetti NS, Maia AR, Morari J, Pagliarone AC, Farias AS, Velloso LA, Queiroz MAF, Vallinoto ACR, Bajgelman MC, Reis LO. Therapeutic BCG vaccine protects against long COVID: The BATTLE randomized clinical trial. J Intern Med 2025; 297:60-78. [PMID: 39560319 DOI: 10.1111/joim.20033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2024]
Abstract
BACKGROUND Bacillus Calmette-Guérin (BCG) injected during the COVID-19 convalescence period was safe and enhanced recovery from anosmia and dysgeusia in the acute phase. OBJECTIVES To report the long-term results of the BATTLE trial, BCG vaccine in adults with mild COVID-19. METHODS Design: Double-blind, placebo-controlled, randomized (1:1) clinical trial. INTERVENTION BCG intradermal vaccine and placebo. PATIENTS A total of 157 BCG and 142 placebo recipients participated in the 6-month follow-up, and 97 BCG and 95 placebo recipients participated in the 12-month follow-up. MEASUREMENTS Long COVID symptoms and mechanistic analyses. RESULTS BCG reduced hearing problems at 6 months (odds ratio [OR] = 0.26) and sleeping, concentration, memory, and vision problems at 12 months (OR = 0.45, 0.36, 0.38, and 0.36, respectively). Sensitivity analyses confirmed that long COVID-19 symptoms were reduced at the 6- and 12-month follow-ups (p = 0.010 and 0.031, respectively). BCG's crossover interaction paradoxically increased hair loss in women and decreased it in men at 6 months (p = 0.032). BCG immunomodulation is likely mediated through inhibition of Fas ligand expression in the blood and increased induction of IL6, IL10, interferon-induced transmembrane protein 3, and angiotensin-converting enzyme 2 in cultured human macrophages. CONCLUSION Long-term follow-up of the BATTLE trial participants revealed that BCG protects against long COVID development if administered within the COVID-19 convalescence period. The response to BCG was subject-specific, including a paradoxical crossover interaction based on sex. LIMITATIONS Not tested for previous mycobacterial exposure; loss to follow-up, particularly at 12 months.
Collapse
Grants
- 88887.506617/2020-00 Coordination for the Improvement of Higher Education Personnel, CAPES, Federal Government, Brazil
- 88887.657670/2021-00 Coordination for the Improvement of Higher Education Personnel, CAPES, Federal Government, Brazil
- General Coordination of the National Immunization Program - CGPNI/DEIDT/SVS/MS
- 465/2020 Ministry of Health, Brazil
- 304747/2018-1 National Council for Scientific and Technological Development-CNPq, Research Productivity
- 310135/2022-2 National Council for Scientific and Technological Development-CNPq, Research Productivity
- 302935/2021-5 National Council for Scientific and Technological Development-CNPq, Research Productivity
Collapse
Affiliation(s)
- Mehrsa Jalalizadeh
- UroScience, State University of Campinas, Unicamp, Campinas, São Paulo, Brazil
| | - Keini Buosi
- UroScience, State University of Campinas, Unicamp, Campinas, São Paulo, Brazil
| | | | - Patricia A F Leme
- UroScience, State University of Campinas, Unicamp, Campinas, São Paulo, Brazil
| | - Karen L Ferrari
- UroScience, State University of Campinas, Unicamp, Campinas, São Paulo, Brazil
| | | | - Wandrey R S Brito
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém, Pará, Brazil
| | - Natália S Brunetti
- Biology Institute, State University of Campinas, Unicamp, Campinas, São Paulo, Brazil
| | - Aline R Maia
- Obesity and Comorbidities Research Center, University of Campinas, Unicamp, Campinas, São Paulo, Brazil
| | - Joseane Morari
- Obesity and Comorbidities Research Center, University of Campinas, Unicamp, Campinas, São Paulo, Brazil
| | - Ana C Pagliarone
- UroScience, State University of Campinas, Unicamp, Campinas, São Paulo, Brazil
| | - Alessandro S Farias
- Biology Institute, State University of Campinas, Unicamp, Campinas, São Paulo, Brazil
| | - Licio A Velloso
- Obesity and Comorbidities Research Center, University of Campinas, Unicamp, Campinas, São Paulo, Brazil
| | - Maria A F Queiroz
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém, Pará, Brazil
| | - Antonio C R Vallinoto
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém, Pará, Brazil
| | - Marcio C Bajgelman
- Brazilian Biosciences National Laboratory, LNBio, CNPEM, Campinas, São Paulo, Brazil
| | - Leonardo O Reis
- UroScience, State University of Campinas, Unicamp, Campinas, São Paulo, Brazil
- ImmunOncology, Pontifical Catholic University of Campinas, PUC-Campinas, Campinas, São Paulo, Brazil
| |
Collapse
|
4
|
Angulo M, Angulo C. Trained immunity-based vaccines: A vision from the one health initiative. Vaccine 2025; 43:126505. [PMID: 39520776 DOI: 10.1016/j.vaccine.2024.126505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/29/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Trained immunity-based vaccines (TIbV or TRIMbV) represent a novel approach to combating infectious diseases. The innate immune system in animals, including humans, exhibits "memory-like" functions. Remarkably, the immunological mechanisms -both epigenetic and metabolic-) underlying this memory enables immune cells to develop defensive and protective outcomes against unspecific pathogenic infections. Under this context, the One Health initiative promotes integrative efforts to combat zoonotic (and anthropozoonotic) diseases, which is critical because 3 of 4 animal infections are transmitted to humans. Therefore, TIbV constitutes a potential affordable approach to control zoonotic pathologies, especially under pandemic scenarios. This review describes the state-of-the-art TIbV and their hurdles, opportunities, and prospects for the One Health initiative to prevent, control, and treat infectious diseases.
Collapse
Affiliation(s)
- Miriam Angulo
- Immunology & Vaccinology Group, Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S. 23096, Mexico.; Laboratorio Nacional CONAHCYT de Generación de Vacunas Veterinarias y Servicios de Diagnóstico (LNC-GVD), Centro de Investigaciones Biológicas del Noroeste, S.C., Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C. S., C.P. 23096, Mexico
| | - Carlos Angulo
- Immunology & Vaccinology Group, Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S. 23096, Mexico.; Laboratorio Nacional CONAHCYT de Generación de Vacunas Veterinarias y Servicios de Diagnóstico (LNC-GVD), Centro de Investigaciones Biológicas del Noroeste, S.C., Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C. S., C.P. 23096, Mexico.
| |
Collapse
|
5
|
Messina NL, Pittet LF, McDonald E, Moore C, Barry S, Bonten M, Byrne A, Campbell J, Croda J, Croda MG, Dalcolmo M, de Almeida E Val FF, de Oliveira RD, Dos Santos G, Douglas MW, Gardiner K, Gwee A, Jardim BA, Kollmann T, Lacerda MV, Lucas M, Lynn DJ, Manning L, Marshall H, O'Connell A, Perrett KP, Post JJ, Prat-Aymerich C, Rocha JL, Rodriguez-Baño J, Wadia U, Warris A, Davidson A, Curtis N. BCG vaccination of healthcare workers for protection against COVID-19: 12-month outcomes from an international randomised controlled trial. J Infect 2024; 89:106245. [PMID: 39127450 PMCID: PMC11409612 DOI: 10.1016/j.jinf.2024.106245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
OBJECTIVES Bacille Calmette-Guérin (BCG) vaccine has immunomodulatory effects that may provide protection against unrelated infectious diseases. We aimed to determine whether BCG vaccination protects adults against COVID-19. DESIGN Phase III double-blind randomised controlled trial. SETTING Healthcare centres in Australia, Brazil, the Netherlands, Spain, and the United Kingdom during the COVID-19 pandemic. PARTICIPANTS 3988 healthcare workers with no prior COVID-19 and no contraindication to BCG. INTERVENTION Randomised 1:1 using a web-based procedure to receive a single 0.1 mL intradermal dose of BCG-Denmark (BCG group, n = 1999) or saline (placebo group, n = 1989). MAIN OUTCOME MEASURES Difference in incidence of (i) symptomatic and (ii) severe COVID-19 during the 12 months following randomisation in the modified intention to treat (mITT) population (confirmed SARS-CoV-2 naïve at inclusion). RESULTS Of the 3988 participants randomised, 3386 had a negative baseline SARS-CoV-2 test and were included in the mITT population. The 12-month adjusted estimated risk of symptomatic COVID-19 was higher in the BCG group (22.6%; 95% confidence interval [CI] 20.6 to 24.5%) compared with the placebo group (19.6%; 95% CI 17.6 to 21.5%); adjusted difference +3.0% points (95% CI 0.2 to 5.8%; p = 0.04). The 12-month adjusted estimated risk of severe COVID-19 (mainly comprising those reporting being unable to work for ≥3 consecutive days) was 11.0% in the BCG group (95% CI 9.5 to 12.4%) compared with 9.6% in the placebo group (95% CI 8.3 to 11.1%); adjusted difference +1.3% points (95% CI -0.7 to 3.3%, p = 0.2). Breakthrough COVID-19 (post COVID-19 vaccination) and asymptomatic SARS-CoV-2 infections were similar in the two groups. There were 18 hospitalisations due to COVID-19 (11 in BCG group, 7 in placebo group; adjusted hazard ratio 1.56, 95% CI 0.60 to 4.02, p = 0.4) and two deaths due to COVID-19, both in the placebo group. CONCLUSIONS Compared to placebo, vaccination with BCG-Denmark increased the risk of symptomatic COVID-19 over 12 months among healthcare workers and did not decrease the risk of severe COVID-19 or post-vaccination breakthrough COVID-19. TRIAL REGISTRATION ClinicalTrials.gov NCT04327206.
Collapse
Affiliation(s)
- Nicole L Messina
- Infectious Diseases Group, Murdoch Children's Research Institute, Parkville, Victoria, Australia; Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - Laure F Pittet
- Infectious Diseases Group, Murdoch Children's Research Institute, Parkville, Victoria, Australia; Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia; Immunology, Vaccinology, Rheumatology and Infectious Diseases Unit, Geneva and University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Ellie McDonald
- Infectious Diseases Group, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Cecilia Moore
- Clinical Epidemiology and Biostatistics Unit, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Simone Barry
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia; Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Marc Bonten
- Julius Center for Health Sciences and Primary Care, University Medical Centre Utrecht, Utrecht University, the Netherlands; European Clinical Research Alliance on Infectious Diseases, Utrecht, Netherlands
| | - Anthony Byrne
- St Vincent's Hospitals, Darlinghurst, New South Wales, Australia; Partners In Health, Socios En Salud, Peru; Thoracic Society of Australia & New Zealand (NSW/ACT Branch), Australia
| | - John Campbell
- Exeter Collaboration for Academic Primary Care, University of Exeter Medical School, Exeter, United Kingdom
| | - Julio Croda
- Fiocruz Mato Grosso do Sul, Fundação Oswaldo Cruz, Campo Grande, Mato Grosso do Sul, Brazil; Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA; Universidade Federal de Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Mariana G Croda
- Universidade Federal de Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Margareth Dalcolmo
- Centro de Referência Professor Hélio Fraga, ENSP/FIOCRUZ (Fundação Oswaldo Cruz), Rio de Janeiro, Brazil
| | | | - Roberto D de Oliveira
- State University of Mato Grosso do Sul, Dourados, Brazil; Post Graduate Program in Health Sciences, Federal University of Grande Dourados, Dourados, Brazil
| | - Glauce Dos Santos
- Centro de Referência Professor Hélio Fraga, ENSP/FIOCRUZ (Fundação Oswaldo Cruz), Rio de Janeiro, Brazil
| | - Mark W Douglas
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Syndey at Westmead Hospital, Westmead, New South Wales, Australia; Centre for Infectious Diseases and Microbiology, Sydney Infectious Diseases Institute, The University of Sydney at Westmead Hospital, Westmead, New South Wales, Australia
| | - Kaya Gardiner
- Infectious Diseases Group, Murdoch Children's Research Institute, Parkville, Victoria, Australia; Research Operations, The Royal Children's Hospital Melbourne, Parkville, Victoria, Australia
| | - Amanda Gwee
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia; Infectious Diseases, Royal Children's Hospital Melbourne, Parkville, Victoria, Australia; Antimicrobials Group, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Bruno A Jardim
- Institute of Clinical Research Carlos Borborema, Doctor Heitor Vieira Dourado Tropical Medicine Foundation, Manaus, Brazil
| | - Tobias Kollmann
- Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, Nedlands, Western Australia, Australia
| | - Marcus Vg Lacerda
- Institute of Clinical Research Carlos Borborema, Doctor Heitor Vieira Dourado Tropical Medicine Foundation, Manaus, Brazil; Instituto Leônidas & Maria Deane, Oswaldo Cruz Foundation Ministry of Health, Manaus, Brazil; University of Texas Medical Branch, Galveston, TX, USA
| | - Michaela Lucas
- Department of Immunology, Pathwest, Queen Elizabeth II Medical Centre, Nedlands, Western Australia, Australia; Department of Immunology, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia; Department of Immunology, Perth Children's Hospital, Nedlands, Western Australia, Australia; School of Medicine, University of Western Australia, Perth, Western Australia, Australia
| | - David J Lynn
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia; Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia, Australia
| | - Laurens Manning
- Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, Nedlands, Western Australia, Australia; School of Medicine, University of Western Australia, Perth, Western Australia, Australia; Department of Infectious Diseases, Fiona Stanley Hospital, Murdoch, Western Australia, Australia
| | - Helen Marshall
- The University of Adelaide and the Women's and Children's Health Network, Adelaide, SA, Australia
| | - Abby O'Connell
- Exeter Clinical Trials Unit, University of Exeter, Exeter, United Kingdom
| | - Kirsten P Perrett
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia; Population Allergy Group, Murdoch Children's Research Institute, Parkville, Victoria, Australia; Department of Allergy and Immunology, Royal Children's Hospital Melbourne, Parkville, Victoria, Australia
| | - Jeffrey J Post
- Department of Infectious Diseases, Prince of Wales Hospital, Randwick, New South Wales, Australia; School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Cristina Prat-Aymerich
- Julius Center for Health Sciences and Primary Care, University Medical Centre Utrecht, Utrecht University, the Netherlands; European Clinical Research Alliance on Infectious Diseases, Utrecht, Netherlands
| | - Jorge L Rocha
- Helio Fraga Reference Center, Oswaldo Cruz Foundation Ministry of Health, Curicica, Brazil
| | - Jesus Rodriguez-Baño
- Division of Infectious Diseases and Microbiology, Department of Medicine, Hospital Universitario Virgen Macarena, University of Seville, Biomedicines Institute of Seville-Consejo Superior de Investigaciones Científicas, Seville, Spain; CIBER de Enfermedades Infecciosas, Instituto de Salud Carloss III, Madrid, Spain
| | - Ushma Wadia
- Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, Nedlands, Western Australia, Australia; School of Medicine, University of Western Australia, Perth, Western Australia, Australia
| | - Adilia Warris
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom; Department of Infectious Diseases, Great Ormond Street Hospital, London, United Kingdom
| | - Andrew Davidson
- Melbourne Children's Trial Centre, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Nigel Curtis
- Infectious Diseases Group, Murdoch Children's Research Institute, Parkville, Victoria, Australia; Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia; Infectious Diseases, Royal Children's Hospital Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
6
|
Maytum A, Porter D, de Whalley P, Thompson A, Plested E, Kerridge S, Liu X, Smits G, van der Klis F, Snape MD, Clutterbuck E, Pollard AJ. The Impact of Infant Bacille Calmette-Guérin Vaccination on the Immunogenicity of Other Vaccines: A Randomized Exploratory Study. Pediatr Infect Dis J 2024; 43:809-812. [PMID: 38717982 DOI: 10.1097/inf.0000000000004373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/19/2024]
Abstract
The effect of the Bacille Calmette-Guérin (BCG) vaccine on the immunogenicity of separately administered serogroup C meningococcal vaccine and other vaccinations was examined in 28 infants randomized to receive BCG at age ≤7 days, at 3 months or after study completion. Immunogenicity of the serogroup C meningococcal vaccine and other routine vaccines might be improved when BCG is administered in early infancy.
Collapse
Affiliation(s)
- Alexander Maytum
- From the Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - David Porter
- From the Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Philip de Whalley
- From the Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Amber Thompson
- From the Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Emma Plested
- From the Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Simon Kerridge
- From the Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Xinxue Liu
- From the Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Gaby Smits
- National Institute of Public Health and the Environment, Centre for Infectious Disease Control, Bilthoven, The Netherlands
| | - Fiona van der Klis
- National Institute of Public Health and the Environment, Centre for Infectious Disease Control, Bilthoven, The Netherlands
| | - Matthew D Snape
- From the Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Elizabeth Clutterbuck
- From the Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Andrew J Pollard
- From the Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
7
|
Buosi K, Jalalizadeh M, Maia AR, Morari J, Velloso LA, Reis LO. Modulation of Human Macrophages by Plasma from COVID-19 Patients Following BCG Vaccination: BATTLE Trial. Int J Gen Med 2024; 17:3107-3117. [PMID: 39049828 PMCID: PMC11268781 DOI: 10.2147/ijgm.s468047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024] Open
Abstract
Purpose To analyze the interfering effect of plasma from COVID-19 convalescent adults vaccinated or not with intradermal Bacillus Calmette-Guérin (BCG) on human macrophages. Methods The BATTLE clinical trial (NCT04369794) was initiated in the 2020 SARS-CoV-2 pandemic to study the safety and efficacy of BCG revaccination of COVID-19 convalescent adults. We measured the expression induction of eleven COVID-19-related genes in human macrophages cultured in plasma taken from 22 BCG vaccinated and 17 placebo patients at baseline and 45 days post-intervention. Subgroup analysis was based on gender, age, job type (healthcare worker [HCW] vs non-HCW), and the presence of anosmia/dysgeusia. Results Compared to plasma from placebo counterparts, the plasma of BCG vaccinated patients increased the expression induction of interferon (IFN)β-1b (p = 0.042) in human macrophages. This increase was more pronounced in females and in healthcare workers (HCW) (p = 0.007 and 0.001, respectively). Interferon-induced transmembrane protein 3 (IFITM3) expression induction was increased by plasma from BCG vaccinated females, young age group, and HCWs (p = 0.004, 0.011, and 0.040, respectively). Interleukin (IL)-10 induction increased by the plasma of young BCG recipients (p = 0.008). Induction of IL-6 expression increased by non-HCW BCG recipients plasma but decreased by HCW BCG recipients plasma (p = 0.005). Baseline plasma of patients who presented with anosmia/dysgeusia at the time of admission induced lower angiotensin-converting enzyme 2 (ACE2) compared to those without the symptom (0.76 vs 0.97, p = 0.004). ACE2 expression induction significantly increased by plasma of BCG recipients if they had anosmia/dysgeusia on admission (p = 0.028). Conclusion The expressions of IFNβ-1b, IFITM3, IL-6, and IL-10 in human macrophages incubated with the plasma of COVID-19 convalescent patients were modulated by BCG. These modulations depended on subject-specific characteristics, including gender, age, clinical presentation (anosmia/dysgeusia), job type, and previous exposure to mycobacteria.
Collapse
Affiliation(s)
- Keini Buosi
- Uroscience, School of Medical Sciences, University of Campinas, UNICAMP, Campinas, Sao Paulo, 13083-872, Brazil
| | - Mehrsa Jalalizadeh
- Uroscience, School of Medical Sciences, University of Campinas, UNICAMP, Campinas, Sao Paulo, 13083-872, Brazil
| | - Aline Rosa Maia
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of Campinas, Campinas, 13083-864, Brazil
| | - Joseane Morari
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of Campinas, Campinas, 13083-864, Brazil
| | - Licio Augusto Velloso
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of Campinas, Campinas, 13083-864, Brazil
| | - Leonardo Oliveira Reis
- Uroscience, School of Medical Sciences, University of Campinas, UNICAMP, Campinas, Sao Paulo, 13083-872, Brazil
- Immunoncology, Pontifical Catholic University of Campinas, PUC-Campinas, Campinas, Sao Paulo, 13087-571, Brazil
| |
Collapse
|
8
|
Kühtreiber WM, Hostetter ER, Wolfe GE, Vaishnaw MS, Goldstein R, Bulczynski ER, Hullavarad NS, Braley JE, Zheng H, Faustman DL. Late in the US pandemic, multi-dose BCG vaccines protect against COVID-19 and infectious diseases. iScience 2024; 27:109881. [PMID: 39055605 PMCID: PMC11270028 DOI: 10.1016/j.isci.2024.109881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/13/2024] [Accepted: 04/29/2024] [Indexed: 07/27/2024] Open
Abstract
The Bacillus Calmette-Guérin vaccine has many off-target benefits, including protection from diverse infectious diseases. As SARS-CoV-2 evolved, COVID-19 disease became more transmissible and less lethal. In this Phase III double-blinded, placebo-controlled trial conducted late in the pandemic, we tested at-risk US adults with type 1 diabetes if multi-dose BCG protected against COVID-19 and other infectious disease, co-primary outcomes. From April 2021 to November 2022, Tokyo-strain BCG vaccines provided significant protection against COVID-19 disease (p = 0.023) and strong platform protection against all infectious diseases (p < 0.0001). Over the course of the study, commercial COVID-19 vaccines were rolled out, most of which were mRNA-based. In contrast to the protection afforded by BCG, as reported by others, COVID-19 mRNA vaccine alone provided no protection from COVID-19 disease (p = 0.43). BCG vaccination efficacy was unaffected by concurrent COVID-19 vaccinations; COVID-19 vaccines neither helped nor hindered BCG protection.
Collapse
Affiliation(s)
- Willem M. Kühtreiber
- Massachusetts General Hospital and Harvard Medical School, Boston, MA 02192, USA
| | | | | | | | | | | | | | | | - Hui Zheng
- Massachusetts General Hospital, Boston, MA 02192, USA
| | - Denise L. Faustman
- Massachusetts General Hospital and Harvard Medical School, Boston, MA 02192, USA
| |
Collapse
|
9
|
Pittet LF, Messina NL, McDonald E, Orsini F, Barry S, Bonten M, Campbell J, Croda J, Croda MG, Dalcolmo M, Gardiner K, Gwee A, Jardim B, Lacerda MV, Lucas M, Lynn DJ, Manning L, Perrett KP, Post JJ, Prat-Aymerich C, Richmond PC, Rocha JL, Rodriguez-Baño J, Warris A, Wood NJ, Davidson A, Curtis N. Bacille Calmette-Guérin vaccination to prevent febrile and respiratory illness in adults (BRACE): secondary outcomes of a randomised controlled phase 3 trial. EClinicalMedicine 2024; 72:102616. [PMID: 38774675 PMCID: PMC11106519 DOI: 10.1016/j.eclinm.2024.102616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 04/03/2024] [Accepted: 04/11/2024] [Indexed: 05/24/2024] Open
Abstract
Background Bacille Calmette-Guérin (BCG) vaccination has off-target (non-specific) effects that are associated with protection against unrelated infections and decreased all-cause mortality in infants. We aimed to determine whether BCG vaccination prevents febrile and respiratory infections in adults. Methods This randomised controlled phase 3 trial was done in 36 healthcare centres in Australia, Brazil, the Netherlands, Spain, and the United Kingdom. Healthcare workers were randomised to receive BCG-Denmark (single 0.1 ml intradermal injection) or no BCG in a 1:1 ratio using a web-based procedure, stratified by stage, site, age, and presence of co-morbidity. The difference in occurrence of febrile or respiratory illness were measured over 12 months (prespecified secondary outcome) using the intention-to-treat (ITT) population. This trial is registered with ClinicalTrials.gov, NCT04327206. Findings Between March 30, 2020, and April 1, 2021, 6828 healthcare workers were randomised to BCG-Denmark (n = 3417) or control (n = 3411; no intervention or placebo) groups. The 12-month adjusted estimated risk of ≥1 episode of febrile or respiratory illness was 66.8% in the BCG group (95% CI 65.3%-68.2%), compared with 63.4% in the control group (95% CI 61.8%-65.0%), a difference of +3.4 percentage points (95% CI +1.3% to +5.5%; p 0.002). The adjusted estimated risk of a severe episode (defined as being incapacitated for ≥3 consecutive days or hospitalised) was 19.4% in the BCG group (95% CI 18.0%-20.7%), compared with 18.8% in the control group (95% CI 17.4%-20.2%) a difference of +0.6 percentage points (95% CI -1.3% to +2.5%; p 0.6). Both groups had a similar number of episodes of illness, pneumonia, and hospitalisation. There were three deaths, all in the control group. There were no safety concerns following BCG vaccination. Interpretation In contrast to the beneficial off-target effects reported following neonatal BCG in infants, a small increased risk of symptomatic febrile or respiratory illness was observed in the 12 months following BCG vaccination in adults. There was no evidence of a difference in the risk of severe disease. Funding Bill & Melinda Gates Foundation, Minderoo Foundation, Sarah and Lachlan Murdoch, the Royal Children's Hospital Foundation, Health Services Union NSW, the Peter Sowerby Foundation, SA Health, the Insurance Advisernet Foundation, the NAB Foundation, the Calvert-Jones Foundation, the Modara Pines Charitable Foundation, the UHG Foundation Pty Ltd, Epworth Healthcare, the National Health and Medical Research Council, the Swiss National Science Foundation and individual donors.
Collapse
Affiliation(s)
- Laure F. Pittet
- Infectious Diseases Group, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
- Infectious Diseases, The Royal Children's Hospital Melbourne, Parkville, Victoria, Australia
- Immunology, Vaccinology, Rheumatology, and Infectious Diseases Unit, Department of Paediatrics, Gynaecology and Obsterics, Faculty of Medicine, University of Geneva and University Hospitals of Geneva, Geneva, Switzerland
| | - Nicole L. Messina
- Infectious Diseases Group, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - Ellie McDonald
- Infectious Diseases Group, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Francesca Orsini
- Clinical Epidemiology and Biostatistics Unit, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Melbourne Children's Trial Centre, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Simone Barry
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Marc Bonten
- Julius Center for Health Sciences and Primary Care, University Medical Centre Utrecht, Utrecht University, the Netherlands
| | - John Campbell
- Exeter Collaboration for Academic Primary Care, University of Exeter Medical School, Exeter, United Kingdom
| | - Julio Croda
- Fiocruz Mato Grosso do Sul, Fundação Oswaldo Cruz, Campo Grande, Brazil
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
- Universidade Federal de Mato Grosso do Sul, Campo Grande, Brazil
| | - Mariana G. Croda
- Universidade Federal de Mato Grosso do Sul, Campo Grande, Brazil
| | - Margareth Dalcolmo
- Helio Fraga Reference Center, Oswaldo Cruz Foundation Ministry of Health, Curicica, Brazil
- Catholic University, Rio de Janeiro, Brazil
| | - Kaya Gardiner
- Infectious Diseases Group, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Research Operations, The Royal Children's Hospital Melbourne, Parkville, Victoria, Australia
| | - Amanda Gwee
- Infectious Diseases Group, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
- Infectious Diseases, The Royal Children's Hospital Melbourne, Parkville, Victoria, Australia
| | - Bruno Jardim
- Institute of Clinical Research Carlos Borborema, Doctor Heitor Vieira Dourado Tropical Medicine Foundation, Manaus, Brazil
| | - Marcus V.G. Lacerda
- Institute of Clinical Research Carlos Borborema, Doctor Heitor Vieira Dourado Tropical Medicine Foundation, Manaus, Brazil
- Instituto Leônidas & Maria Deane, Oswaldo Cruz Foundation Ministry of Health, Manaus, Brazil
- University of Texas Medical Branch, Galveston, TX, USA
| | - Michaela Lucas
- Department of Immunology, Pathwest, Queen Elizabeth II Medical Centre, Nedlands, Western Australia, Australia
- Department of Immunology, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
- Department of Immunology and General Paediatrics, Perth Children's Hospital, Nedlands, Western Australia, Australia
- School of Medicine, University of Western Australia, Perth, Western Australia, Australia
| | - David J. Lynn
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia, Australia
| | - Laurens Manning
- School of Medicine, University of Western Australia, Perth, Western Australia, Australia
- Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, Nedlands, Western Australia, Australia
- Department of Infectious Diseases, Fiona Stanley Hospital, Murdoch, Western Australia, Australia
| | - Kirsten P. Perrett
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
- Melbourne Children's Trial Centre, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Allergy and Immunology, Royal Children's Hospital Melbourne, Parkville, Victoria, Australia
| | - Jeffrey J. Post
- Department of Infectious Diseases, Prince of Wales Hospital, Randwick, New South Wales, Australia
- School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Cristina Prat-Aymerich
- Julius Center for Health Sciences and Primary Care, University Medical Centre Utrecht, Utrecht University, the Netherlands
- Institut d'Investigació Germans Trias i Pujol, Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Respiratorias, Instituto de Salud Carlos III, Barcelona, Spain
| | - Peter C. Richmond
- Department of Immunology and General Paediatrics, Perth Children's Hospital, Nedlands, Western Australia, Australia
- School of Medicine, University of Western Australia, Perth, Western Australia, Australia
- Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, Nedlands, Western Australia, Australia
| | - Jorge L. Rocha
- Helio Fraga Reference Center, Oswaldo Cruz Foundation Ministry of Health, Curicica, Brazil
| | - Jesus Rodriguez-Baño
- Division of Infectious Diseases and Microbiology, Department of Medicine, Hospital Universitario Virgen Macarena, University of Seville, Biomedicines Institute of Seville-Consejo Superior de Investigaciones Científicas, Seville, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carloss III, Madrid, Spain
| | - Adilia Warris
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Nicholas J. Wood
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Sydney Children's Hospital Network, Westmead, New South Wales, Australia
- National Centre for Immunisation Research and Surveillance of Vaccine Preventable Disease, Westmead, New South Wales, Australia
| | - Andrew Davidson
- Melbourne Children's Trial Centre, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Nigel Curtis
- Infectious Diseases Group, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
- Infectious Diseases, The Royal Children's Hospital Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
10
|
Specht AG, Ginese M, Kurtz SL, Elkins KL, Specht H, Beamer G. Host Genetic Background Influences BCG-Induced Antibodies Cross-Reactive to SARS-CoV-2 Spike Protein. Vaccines (Basel) 2024; 12:242. [PMID: 38543876 PMCID: PMC10975245 DOI: 10.3390/vaccines12030242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/16/2024] [Accepted: 02/20/2024] [Indexed: 04/01/2024] Open
Abstract
Mycobacterium bovis Bacillus Calmette-Guérin (BCG) protects against childhood tuberculosis; and unlike most vaccines, BCG broadly impacts immunity to other pathogens and even some cancers. Early in the COVID-19 pandemic, epidemiological studies identified a protective association between BCG vaccination and outcomes of SARS-CoV-2, but the associations in later studies were inconsistent. We sought possible reasons and noticed the study populations often lived in the same country. Since individuals from the same regions can share common ancestors, we hypothesized that genetic background could influence associations between BCG and SARS-CoV-2. To explore this hypothesis in a controlled environment, we performed a pilot study using Diversity Outbred mice. First, we identified amino acid sequences shared by BCG and SARS-CoV-2 spike protein. Next, we tested for IgG reactive to spike protein from BCG-vaccinated mice. Sera from some, but not all, BCG-vaccinated Diversity Outbred mice contained higher levels of IgG cross-reactive to SARS-CoV-2 spike protein than sera from BCG-vaccinated C57BL/6J inbred mice and unvaccinated mice. Although larger experimental studies are needed to obtain mechanistic insight, these findings suggest that genetic background may be an important variable contributing to different associations observed in human randomized clinical trials evaluating BCG vaccination on SARS-CoV-2 and COVID-19.
Collapse
Affiliation(s)
- Aubrey G. Specht
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA 01536, USA; (A.G.S.); (M.G.)
| | - Melanie Ginese
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA 01536, USA; (A.G.S.); (M.G.)
| | - Sherry L. Kurtz
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA; (S.L.K.); (K.L.E.)
| | - Karen L. Elkins
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA; (S.L.K.); (K.L.E.)
| | - Harrison Specht
- Department of Bioengineering and Barnett Institute, Northeastern University, Boston, MA 02115, USA;
| | - Gillian Beamer
- Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| |
Collapse
|
11
|
Pittet LF, Noble CCA, Messina NL, Curtis N. Using BCG vaccination to protect against COVID-19: when reality fails to meet expectation. Nat Rev Immunol 2024; 24:83-84. [PMID: 38238441 DOI: 10.1038/s41577-024-00992-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2024]
Affiliation(s)
- Laure F Pittet
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia.
- Murdoch Children's Research Institute, Parkville, Victoria, Australia.
- Immunology-Vaccinology and Paediatric Infectious Diseases Unit, Faculty of Medicine, Geneva University Hospitals, Geneva, Switzerland.
| | - Christie C A Noble
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Infectious Diseases, The Royal Children's Hospital Melbourne, Parkville, Victoria, Australia
| | - Nicole L Messina
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Nigel Curtis
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia.
- Murdoch Children's Research Institute, Parkville, Victoria, Australia.
- Infectious Diseases, The Royal Children's Hospital Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
12
|
DiNardo AR, Arditi M, Kamat AM, Koster KJ, Carrero S, Nishiguchi T, Lebedev M, Benjamin AB, Avalos P, Lozano M, Moule MG, McCune B, Herron B, Ladki M, Sheikh D, Spears M, Herrejon IA, Dodge C, Kumar S, Hutchison RW, Ofili TU, Opperman LA, Bernard JA, Lerner SP, Udeani G, Neal G, Netea MG, Cirillo JD. Bacillus Calmette-Guérin vaccination as defense against SARS-CoV-2 (BADAS): a randomized controlled trial to protect healthcare workers in the USA by enhanced trained immune responses. Trials 2023; 24:636. [PMID: 37794431 PMCID: PMC10548680 DOI: 10.1186/s13063-023-07662-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 09/19/2023] [Indexed: 10/06/2023] Open
Abstract
BACKGROUND A large epidemic, such as that observed with SARS-CoV-2, seriously challenges available hospital capacity, and this would be augmented by infection of healthcare workers (HCW). Bacillus Calmette-Guérin (BCG) is a vaccine against tuberculosis, with protective non-specific effects against other respiratory tract infections in vitro and in vivo. Preliminary analyses suggest that regions of the world with existing BCG vaccination programs have lower incidence and mortality from COVID-19. We hypothesize that BCG vaccination can reduce SARS-CoV-2 infection and disease severity. METHODS This will be a placebo-controlled adaptive multi-center randomized controlled trial. A total of 1800 individuals considered to be at high risk, including those with comorbidities (hypertension, diabetes, obesity, reactive airway disease, smokers), racial and ethnic minorities, elderly, teachers, police, restaurant wait-staff, delivery personnel, health care workers who are defined as personnel working in a healthcare setting, at a hospital, medical center or clinic (veterinary, dental, ophthalmology), and first responders (paramedics, firefighters, or law enforcement), will be randomly assigned to two treatment groups. The treatment groups will receive intradermal administration of BCG vaccine or placebo (saline) with groups at a 1:1 ratio. Individuals will be tracked for evidence of SARS-CoV-2 infection and severity as well as obtaining whole blood to track immunological markers, and a sub-study will include cognitive function and brain imaging. The majority of individuals will be followed for 6 months, with an option to extend for another 6 months, and the cognitive sub-study duration is 2 years. We will plot Kaplan-Meier curves that will be plotted comparing groups and hazard ratios and p-values reported using Cox proportional hazard models. DISCUSSION It is expected this trial will allow evaluation of the effects of BCG vaccination at a population level in high-risk healthcare individuals through a mitigated clinical course of SARS-CoV-2 infection and inform policy making during the ongoing epidemic. TRIAL REGISTRATION ClinicalTrials.gov NCT04348370. Registered on April 16, 2020.
Collapse
Affiliation(s)
- Andrew R DiNardo
- Global and Immigrant Health, Baylor College of Medicine, Houston, TX, 77030, USA
- Radboud Center for Infectious Diseases, Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Moshe Arditi
- Departments of Pediatrics and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Ashish M Kamat
- Department of Urology, UT MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Kent J Koster
- Center for Airborne Pathogen Research and Imaging, Texas A&M School of Medicine, Bryan, TX, 77807, USA
| | - Santiago Carrero
- Global and Immigrant Health, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Tomoki Nishiguchi
- Global and Immigrant Health, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Maxim Lebedev
- Center for Airborne Pathogen Research and Imaging, Texas A&M School of Medicine, Bryan, TX, 77807, USA
| | - Aaron B Benjamin
- Center for Airborne Pathogen Research and Imaging, Texas A&M School of Medicine, Bryan, TX, 77807, USA
| | - Pablo Avalos
- Cedars-Sinai Medical Center, Regenerative Medicine Institute, Los Angeles, CA, 90048, USA
| | - Marisa Lozano
- Department of Urology, UT MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Madeleine G Moule
- Institute of Immunology & Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | | | | | - Malik Ladki
- Global and Immigrant Health, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Daanish Sheikh
- Global and Immigrant Health, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Matthew Spears
- College of Osteopathic Medicine, Sam Houston State University, Conroe, TX, 77304, USA
| | - Ivan A Herrejon
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, TX, 77843, USA
| | - Courtney Dodge
- Texas A&M School of Medicine, Round Rock, TX, 78665, USA
| | - Sathish Kumar
- Center for Airborne Pathogen Research and Imaging, Texas A&M School of Medicine, Bryan, TX, 77807, USA
| | - Robert W Hutchison
- Department of Pharmacy Practice, Texas A&M School of Pharmacy, College Station, TX, 77843, USA
| | - Theresa U Ofili
- Department of Pharmacy Practice, Texas A&M School of Pharmacy, College Station, TX, 77843, USA
| | - Lynne A Opperman
- Center for Craniofacial Research and Diagnosis, Texas A&M School of Dentistry, Dallas, TX, 75246, USA
| | - Jessica A Bernard
- Department of Psychological and Brain Sciences, Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX, 77843, USA
| | - Seth P Lerner
- Scott Department of Urology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - George Udeani
- Department of Pharmacy Practice, Texas A&M School of Pharmacy, Kingsville, TX, 78363, USA
| | - Gabriel Neal
- Primary Care and Rural Medicine, Texas A&M School of Medicine, Bryan, TX, 77807, USA
| | - Mihai G Netea
- Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jeffrey D Cirillo
- Center for Airborne Pathogen Research and Imaging, Texas A&M School of Medicine, Bryan, TX, 77807, USA.
| |
Collapse
|