51
|
Lajqi T, Köstlin-Gille N, Gille C. Trained Innate Immunity in Pediatric Infectious Diseases. Pediatr Infect Dis J 2024; 43:e57-e59. [PMID: 38190648 DOI: 10.1097/inf.0000000000004157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Affiliation(s)
- Trim Lajqi
- From the Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany
| | - Natascha Köstlin-Gille
- From the Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany
| | - Christian Gille
- From the Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany
| |
Collapse
|
52
|
Pulendran B. Integrated organ immunity: a path to a universal vaccine. Nat Rev Immunol 2024; 24:81-82. [PMID: 38212452 PMCID: PMC11706691 DOI: 10.1038/s41577-024-00990-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2024]
Abstract
Here, I introduce the concept of ‘integrated organ immunity’ to explain how the innate and adaptive immune systems and non-haematopoietic cells can interact in tissues to generate enduring protective immunity against diverse pathogens in an antigen-agnostic manner. Considering immune responses through this framework could enable the design of a new class of vaccines termed ‘universal vaccines’ that are not pathogen specific.
Collapse
Affiliation(s)
- Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
53
|
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
|
54
|
Rescigno M. BCG-mediated viral protection goes biphasic. Nat Immunol 2024; 25:13-14. [PMID: 38168959 DOI: 10.1038/s41590-023-01713-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Affiliation(s)
- Maria Rescigno
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.
- IRCCS Humanitas Research Hospital, Milan, Italy.
| |
Collapse
|
55
|
Pontiroli AE, Scovenna F, Carlini V, Tagliabue E, Martin-Delgado J, Sala LL, Tanzi E, Zanoni I. Vaccination against influenza viruses reduces infection, not hospitalization or death, from respiratory COVID-19: A systematic review and meta-analysis. J Med Virol 2024; 96:e29343. [PMID: 38163281 PMCID: PMC10924223 DOI: 10.1002/jmv.29343] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/01/2023] [Accepted: 12/13/2023] [Indexed: 01/03/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes COVID-19 and has brought a huge burden in terms of human lives. Strict social distance and influenza vaccination have been recommended to avoid co-infections between influenza viruses and SARS-CoV-2. Scattered reports suggested a protective effect of influenza vaccine on COVID-19 development and severity. We analyzed 51 studies on the capacity of influenza vaccination to affect infection with SARS-CoV-2, hospitalization, admission to Intensive Care Units (ICU), and mortality. All subjects taken into consideration did not receive any anti-SARS-CoV-2 vaccine, although their status with respect to previous infections with SARS-CoV-2 is not known. Comparison between vaccinated and not-vaccinated subjects for each of the four endpoints was expressed as odds ratio (OR), with 95% confidence intervals (CIs); all analyses were performed by DerSimonian and Laird model, and Hartung-Knapp model when studies were less than 10. In a total of 61 029 936 subjects from 33 studies, influenza vaccination reduced frequency of SARS-CoV-2 infection [OR plus 95% CI = 0.70 (0.65-0.77)]. The effect was significant in all studies together, in health care workers and in the general population; distance from influenza vaccination and the type of vaccine were also of importance. In 98 174 subjects from 11 studies, frequency of ICU admission was reduced with influenza vaccination [OR (95% CI) = 0.71 (0.54-0.94)]; the effect was significant in all studies together, in pregnant women and in hospitalized subjects. In contrast, in 4 737 328 subjects from 14 studies hospitalization was not modified [OR (95% CI) = 1.05 (0.82-1.35)], and in 4 139 660 subjects from 19 studies, mortality was not modified [OR (95% CI) = 0.76 (0.26-2.20)]. Our study emphasizes the importance of influenza vaccination in the protection against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Antonio E. Pontiroli
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, 20142 Milan, Italy
| | - Francesco Scovenna
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, 20142 Milan, Italy
| | - Valentina Carlini
- IRCCS MultiMedica, Laboratory of Cardiovascular and Dysmetabolic Disease, 20138 Milan, Italy
| | - Elena Tagliabue
- IRCCS MultiMedica, Value-Based Healthcare Unit, 20099 Milan, Italy
| | - Jimmy Martin-Delgado
- Hospital Luis Vernaza, Junta de Beneficiencia de Guayaquil 090603, Ecuador
- Instituto de Investigacion e Innovacion en Salud Integral, Universidad Catolica de Santiago de Guayaquil, Guayaquil 090603, Ecuador
| | - Lucia La Sala
- IRCCS MultiMedica, Laboratory of Cardiovascular and Dysmetabolic Disease, 20138 Milan, Italy
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan, Italy
| | - Elisabetta Tanzi
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, 20142 Milan, Italy
| | - Ivan Zanoni
- Harvard Medical School, Boston Children’s Hospital, Division of Immunology and Division of Gastroenterology, Boston, MA 02115, USA
| |
Collapse
|
56
|
Lee A, Floyd K, Wu S, Fang Z, Tan TK, Froggatt HM, Powers JM, Leist SR, Gully KL, Hubbard ML, Li C, Hui H, Scoville D, Ruggiero AD, Liang Y, Pavenko A, Lujan V, Baric RS, Nolan GP, Arunachalam PS, Suthar MS, Pulendran B. BCG vaccination stimulates integrated organ immunity by feedback of the adaptive immune response to imprint prolonged innate antiviral resistance. Nat Immunol 2024; 25:41-53. [PMID: 38036767 PMCID: PMC10932731 DOI: 10.1038/s41590-023-01700-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 11/03/2023] [Indexed: 12/02/2023]
Abstract
Bacille Calmette-Guérin (BCG) vaccination can confer nonspecific protection against heterologous pathogens. However, the underlying mechanisms remain mysterious. We show that mice vaccinated intravenously with BCG exhibited reduced weight loss and/or improved viral clearance when challenged with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2 B.1.351) or PR8 influenza. Protection was first evident between 14 and 21 d post-vaccination and lasted ∼3 months. Notably, BCG induced a biphasic innate response and robust antigen-specific type 1 helper T cell (TH1 cell) responses in the lungs. MyD88 signaling was essential for innate and TH1 cell responses, and protection against SARS-CoV-2. Depletion of CD4+ T cells or interferon (IFN)-γ activity before infection obliterated innate activation and protection. Single-cell and spatial transcriptomics revealed CD4-dependent expression of IFN-stimulated genes in lung myeloid and epithelial cells. Notably, BCG also induced protection against weight loss after mouse-adapted SARS-CoV-2 BA.5, SARS-CoV and SHC014 coronavirus infections. Thus, BCG elicits integrated organ immunity, where CD4+ T cells feed back on tissue myeloid and epithelial cells to imprint prolonged and broad innate antiviral resistance.
Collapse
Affiliation(s)
- Audrey Lee
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Katharine Floyd
- Department of Pediatrics, Emory Vaccine Center, Emory National Primate Research Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Shengyang Wu
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Zhuoqing Fang
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Tze Kai Tan
- Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Heather M Froggatt
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - John M Powers
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sarah R Leist
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kendra L Gully
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Miranda L Hubbard
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Chunfeng Li
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Harold Hui
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | | | | | - Yan Liang
- NanoString Technologies, Seattle, WA, USA
| | | | - Victor Lujan
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Garry P Nolan
- Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Prabhu S Arunachalam
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Mehul S Suthar
- Department of Pediatrics, Emory Vaccine Center, Emory National Primate Research Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
- Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
57
|
Zhang H, Lin J, Wu J, Zhang J, Zhang L, Yuan S, Chen J, Tang Q, Zhang A, Cui Y, Xu X, Dai H, Shi H, Hu X, Xie D, Chen J, He F, Yin Y. Allergic diseases aggravate the symptoms of SARS-CoV-2 infection in China. Front Immunol 2023; 14:1284047. [PMID: 38204754 PMCID: PMC10777727 DOI: 10.3389/fimmu.2023.1284047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 12/08/2023] [Indexed: 01/12/2024] Open
Abstract
Background The relationship between allergic diseases and the adverse outcomes of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has been a subject of controversy. This study aimed to investigate the association between allergic diseases and the incidence and severity of symptoms in SARS-CoV-2 infection. Methods Clinical data of individuals, including children and their parents, infected with SARS-CoV-2 from December 2022 to January 2023 in China were retrospectively analyzed. The data were collected through questionnaires. Statistical analysis, including chi-squared tests, nonparametric analysis, one-way ANOVA, and logistic regression analysis, was used to examine the relationship between allergic diseases, prior medication, and the symptoms of SARS-CoV-2 infection. Results There were 3,517 adults and 3,372 children with SARS-CoV-2 infection included in the study. Fever was found to occur at similar rates in children (86.5%) and adults (86.8%). However, other symptoms related to respiratory issues (such as cough and sore throat), neurological symptoms (headache, loss of smell, and loss of taste), and systemic symptoms (muscle soreness and weakness) were observed more frequently in adults (P < 0.001). Additionally, adults exhibited higher overall symptom scores, indicating greater severity. Allergic diseases were found to be associated with the incidence of certain SARS-CoV-2 infection symptoms in both children and adults. Specifically, children with allergic rhinitis (AR) were observed to be more susceptible to upper respiratory symptoms (OR: 1.320, 95% CI: 1.081-1.611, P = 0.006), while asthma patients were found to be more susceptible to severe respiratory symptoms (OR: 1.736, 95% CI: 1.250-2.411, P = 0.001). Similar patterns were identified in adults. Furthermore, AR was also suggested to be a risk factor for symptom severity in both children (OR: 1.704, 95% CI: 1.314-2.209, P < 0.001) and adults (OR: 1.736, 95% CI: 1.250-2.411, P = 0.001). However, prior medication for allergic diseases did not exhibit a preventive effect on SARS-CoV-2 infection symptoms. Conclusions Both children and adults with allergic diseases were found to be more prone to experiencing symptoms of SARS-CoV-2 infection, and these symptoms tended to be more severe.
Collapse
Affiliation(s)
- Huishan Zhang
- Department of Respiratory Medicine, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jilei Lin
- Department of Respiratory Medicine, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jinhong Wu
- Department of Respiratory Medicine, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Zhang
- Department of Respiratory Medicine, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lei Zhang
- Department of Respiratory Medicine, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shuhua Yuan
- Department of Respiratory Medicine, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiande Chen
- Department of Respiratory Medicine, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qiuyu Tang
- Department of Respiratory Medicine, Shanghai Children’s Medical Centre Affiliated to Shanghai Jiaotong University School of Medicine, Fujian Children’s Hospital, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology And Pediatrics, Fujian Medical University, Fuzhou, Fujian, China
| | - Ailian Zhang
- Department of Respiratory Medicine, The Second Hospital of Jiaxing, Jiaxing, Zhejiang, China
| | - Yuxia Cui
- Department of Respiratory Medicine, Guizhou Provincial People’s Hospital, Shanghai Children’s Medical Center, Shanghai JiaoTong University School of Medicine, Guiyang, Guizhou, China
| | - Xiaojuan Xu
- Department of Respiratory Medicine, Shaoxing Central Hospital, Shaoxing, Zhejiang, China
| | - Hongxie Dai
- Department of Respiratory Medicine, Zhoupu Hospital Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Hongbo Shi
- Department of Respiratory Medicine, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Xiaowei Hu
- Department of Respiratory Medicine, Shanghai Children’s Medical Centre Affiliated to Shanghai Jiaotong University School of Medicine, Fujian Children’s Hospital, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology And Pediatrics, Fujian Medical University, Fuzhou, Fujian, China
| | - Dan Xie
- Department of Respiratory Medicine, Sanya Women and Children’s Hospital Affiliated to Hainan Medical College, Shanghai Children’s Medical Center, Sanya, Hainan, China
| | - Jing Chen
- Department of Respiratory Medicine, Linyi Maternal and Child Healthcare Hospital, Linyi, Shandong, China
| | - Fengquan He
- HongHe MCH (HongHe Hani and Yi Autonomous Prefecture Maternal and Child Health Hospital), Honghe, Yunnan, China
| | - Yong Yin
- Department of Respiratory Medicine, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Respiratory Medicine, Sanya Women and Children’s Hospital Affiliated to Hainan Medical College, Shanghai Children’s Medical Center, Sanya, Hainan, China
- Department of Respiratory Medicine, Linyi Maternal and Child Healthcare Hospital, Linyi, Shandong, China
- Department of Respiratory Medicine, Shanghai Children’s Medical Center Pediatric Medical Complex (Pudong), Shanghai, China
- Pediatric Artificial Intelligence Clinical Application and Research Center, Shanghai Children’s Medical Center, Shanghai, China
| |
Collapse
|
58
|
van Zutphen M, Kiemeney LA, Oldenhof UT, Maurits JS, Witjes JA, Joosten LA, Netea MG, Aben KK, Vrieling A, Vermeulen SH. No Association Between BCG Instillations and COVID-19 Incidence in a Dutch Non-Muscle Invasive Bladder Cancer Cohort. Bladder Cancer 2023; 9:355-363. [PMID: 38994242 PMCID: PMC11165912 DOI: 10.3233/blc-230088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 11/22/2023] [Indexed: 07/13/2024]
Affiliation(s)
| | | | | | | | | | - Leo A.B. Joosten
- Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Mihai G. Netea
- Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Katja K.H. Aben
- Radboud University Medical Center, Nijmegen, The Netherlands
- Netherlands Comprehensive Cancer Organisation, Utrecht, The Netherlands
| | - Alina Vrieling
- Radboud University Medical Center, Nijmegen, The Netherlands
| | | |
Collapse
|
59
|
Hilligan KL, Namasivayam S, Clancy CS, Baker PJ, Old SI, Peluf V, Amaral EP, Oland SD, O'Mard D, Laux J, Cohen M, Garza NL, Lafont BAP, Johnson RF, Feng CG, Jankovic D, Lamiable O, Mayer-Barber KD, Sher A. Bacterial-induced or passively administered interferon gamma conditions the lung for early control of SARS-CoV-2. Nat Commun 2023; 14:8229. [PMID: 38086794 PMCID: PMC10716133 DOI: 10.1038/s41467-023-43447-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 11/09/2023] [Indexed: 12/18/2023] Open
Abstract
Type-1 and type-3 interferons (IFNs) are important for control of viral replication; however, less is known about the role of Type-2 IFN (IFNγ) in anti-viral immunity. We previously observed that lung infection with Mycobacterium bovis BCG achieved though intravenous (iv) administration provides strong protection against SARS-CoV-2 in mice yet drives low levels of type-1 IFNs but robust IFNγ. Here we examine the role of ongoing IFNγ responses to pre-established bacterial infection on SARS-CoV-2 disease outcomes in two murine models. We report that IFNγ is required for iv BCG induced reduction in pulmonary viral loads, an outcome dependent on IFNγ receptor expression by non-hematopoietic cells. Importantly, we show that BCG infection prompts pulmonary epithelial cells to upregulate IFN-stimulated genes with reported anti-viral activity in an IFNγ-dependent manner, suggesting a possible mechanism for the observed protection. Finally, we confirm the anti-viral properties of IFNγ by demonstrating that the recombinant cytokine itself provides strong protection against SARS-CoV-2 challenge when administered intranasally. Together, our data show that a pre-established IFNγ response within the lung is protective against SARS-CoV-2 infection, suggesting that concurrent or recent infections that drive IFNγ may limit the pathogenesis of SARS-CoV-2 and supporting possible prophylactic uses of IFNγ in COVID-19 management.
Collapse
Affiliation(s)
- Kerry L Hilligan
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
- Malaghan Institute of Medical Research, Wellington, 6012, New Zealand.
| | - Sivaranjani Namasivayam
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Chad S Clancy
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Paul J Baker
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Samuel I Old
- Malaghan Institute of Medical Research, Wellington, 6012, New Zealand
| | - Victoria Peluf
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
- Immunoparasitology Unit, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Eduardo P Amaral
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Sandra D Oland
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Danielle O'Mard
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Julie Laux
- Flow Cytometry Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Melanie Cohen
- Flow Cytometry Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Nicole L Garza
- SARS-CoV2- Virology Core, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Bernard A P Lafont
- SARS-CoV2- Virology Core, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Reed F Johnson
- SARS-CoV2- Virology Core, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Carl G Feng
- Immunology and Host Defense Group, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2006, Australia
- Centenary Institute, The University of Sydney, Camperdown, NSW, 2050, Australia
| | - Dragana Jankovic
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
- Immunoparasitology Unit, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Olivier Lamiable
- Malaghan Institute of Medical Research, Wellington, 6012, New Zealand
| | - Katrin D Mayer-Barber
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Alan Sher
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
60
|
Cavalcante-Silva LHA, Leite EG, Almeida FS, de Andrade AG, Comberlang FC, Lucena CKR, Pachá ASC, Csordas BG, Keesen TSL. T Cell Response in Tuberculosis-Infected Patients Vaccinated against COVID-19. Microorganisms 2023; 11:2810. [PMID: 38004820 PMCID: PMC10673403 DOI: 10.3390/microorganisms11112810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/01/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Many studies have focused on SARS-CoV-2 and Mycobacterium tuberculosis (Mtb) co-infection consequences. However, after a vaccination plan against COVID-19, the cases of severe disease and death are consistently controlled, although cases of asymptomatic and mild COVID-19 still happen together with tuberculosis (TB) cases. Thus, in this context, we sought to compare the T cell response of COVID-19-non-vaccinated and -vaccinated patients with active tuberculosis exposed to SARS-CoV-2 antigens. Flow cytometry was used to analyze activation markers (i.e., CD69 and CD137) and cytokines (IFN-γ, TNFα, IL-17, and IL-10) levels in CD4+ and CD8+ T cells upon exposure to SARS-CoV-2 peptides. The data obtained showed that CD8+ T cells from non-vaccinated TB patients present a high frequency of CD69 and TNF-α after viral challenge compared to vaccinated TB donors. Conversely, CD4+ T cells from vaccinated TB patients show a high frequency of IL-10 after spike peptide stimulus compared to non-vaccinated patients. No differences were observed in the other parameters analyzed. The results suggest that this reduced immune balance in coinfected individuals may have consequences for pathogen control, necessitating further research to understand its impact on clinical outcomes after COVID-19 vaccination in those with concurrent SARS-CoV-2 and Mtb infections.
Collapse
Affiliation(s)
- Luiz Henrique Agra Cavalcante-Silva
- Immunology of Infectious Diseases Laboratory, Department of Cellular and Molecular Biology, Federal University of Paraiba, João Pessoa 58051-900, PB, Brazil; (L.H.A.C.-S.); (E.G.L.); (F.S.A.); (A.G.d.A.); (F.C.C.); (B.G.C.)
| | - Ericka Garcia Leite
- Immunology of Infectious Diseases Laboratory, Department of Cellular and Molecular Biology, Federal University of Paraiba, João Pessoa 58051-900, PB, Brazil; (L.H.A.C.-S.); (E.G.L.); (F.S.A.); (A.G.d.A.); (F.C.C.); (B.G.C.)
| | - Fernanda Silva Almeida
- Immunology of Infectious Diseases Laboratory, Department of Cellular and Molecular Biology, Federal University of Paraiba, João Pessoa 58051-900, PB, Brazil; (L.H.A.C.-S.); (E.G.L.); (F.S.A.); (A.G.d.A.); (F.C.C.); (B.G.C.)
| | - Arthur Gomes de Andrade
- Immunology of Infectious Diseases Laboratory, Department of Cellular and Molecular Biology, Federal University of Paraiba, João Pessoa 58051-900, PB, Brazil; (L.H.A.C.-S.); (E.G.L.); (F.S.A.); (A.G.d.A.); (F.C.C.); (B.G.C.)
| | - Fernando Cézar Comberlang
- Immunology of Infectious Diseases Laboratory, Department of Cellular and Molecular Biology, Federal University of Paraiba, João Pessoa 58051-900, PB, Brazil; (L.H.A.C.-S.); (E.G.L.); (F.S.A.); (A.G.d.A.); (F.C.C.); (B.G.C.)
| | | | | | - Bárbara Guimarães Csordas
- Immunology of Infectious Diseases Laboratory, Department of Cellular and Molecular Biology, Federal University of Paraiba, João Pessoa 58051-900, PB, Brazil; (L.H.A.C.-S.); (E.G.L.); (F.S.A.); (A.G.d.A.); (F.C.C.); (B.G.C.)
| | - Tatjana S. L. Keesen
- Immunology of Infectious Diseases Laboratory, Department of Cellular and Molecular Biology, Federal University of Paraiba, João Pessoa 58051-900, PB, Brazil; (L.H.A.C.-S.); (E.G.L.); (F.S.A.); (A.G.d.A.); (F.C.C.); (B.G.C.)
| |
Collapse
|
61
|
Noble CCA, Messina NL, Pittet LF, Curtis N. Interpreting the Results of Trials of BCG Vaccination for Protection Against COVID-19. J Infect Dis 2023; 228:1467-1478. [PMID: 37558650 PMCID: PMC10640778 DOI: 10.1093/infdis/jiad316] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 07/21/2023] [Accepted: 08/08/2023] [Indexed: 08/11/2023] Open
Abstract
BCG vaccination has beneficial off-target ("nonspecific") effects on nonmycobacterial infections. On this premise, trials set out to investigate whether BCG provides off-target protection against coronavirus disease 2019 (COVID-19). A literature search identified 11 randomized "BCG COVID-19" trials, with conflicting results. These trials and the differences in their study design are discussed using the PICOT (participants, intervention, control, outcome, and timing) framework to highlight the factors that likely explain their inconsistent findings. These include participant age, sex and comorbid conditions, BCG vaccination strain and dose, outcome measure and duration of follow-up. Understanding how to control these factors to best exploit BCG's off-target effects will be important in designing future trials and intervention strategies.
Collapse
Affiliation(s)
- Christie C A Noble
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
- Infectious Diseases Research Group, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Nicole L Messina
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
- Infectious Diseases Research Group, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Laure F Pittet
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
- Infectious Diseases Research Group, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Paediatric Infectious Diseases Unit, Faculty of Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Nigel Curtis
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
- Infectious Diseases Research Group, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Infectious Diseases, The Royal Children's Hospital Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
62
|
Gong W, Du J. Excluding Participants With Mycobacteria Infections From Clinical Trials: A Critical Consideration in Evaluating the Efficacy of BCG Against COVID-19. J Korean Med Sci 2023; 38:e343. [PMID: 37904656 PMCID: PMC10615642 DOI: 10.3346/jkms.2023.38.e343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 09/01/2023] [Indexed: 11/01/2023] Open
Abstract
In the context of the coronavirus disease 2019 (COVID-19) pandemic, Bacillus Calmette-Guérin (BCG), a tuberculosis (TB) vaccine, has been investigated for its potential to prevent COVID-19 with conflicting outcomes. Currently, over 50 clinical trials have been conducted to assess the effectiveness of BCG in preventing COVID-19, but the results have shown considerable variations. After scrutinizing the data, it was discovered that some trials had enrolled individuals with active TB, latent TB infection, or a history of TB. This finding raises concerns about the reliability and validity of the trial outcomes. In this study, we explore the potential consequences of including these participants in clinical trials, including impaired host immunity, immune exhaustion, and the potential masking of the BCG vaccine's protective efficacy against COVID-19 by persistent mycobacterial infections. We also put forth several suggestions for future clinical trials. Our study underscores the criticality of excluding individuals with active or latent TB from clinical trials evaluating the efficacy of BCG in preventing COVID-19.
Collapse
Affiliation(s)
- Wenping Gong
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The 8th Medical Center of PLA General Hospital, Beijing, China.
| | - Jingli Du
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The 8th Medical Center of PLA General Hospital, Beijing, China.
| |
Collapse
|
63
|
Jalalizadeh M, Leme PAF, Buosi K, Dionato FAV, Dal Col LSB, Giacomelli CF, Reis LO. Healthcare Workers (HCWs) and non-HCWs reaction to Bacillus Calmette-Guérin (BCG) in the BATTLE trial. Vaccine 2023; 41:6599-6606. [PMID: 37743116 DOI: 10.1016/j.vaccine.2023.09.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 09/10/2023] [Accepted: 09/18/2023] [Indexed: 09/26/2023]
Abstract
OBJECTIVES Healthcare workers (HCWs) may have different response to Bacillus Calmette-Guérin (BCG) vaccination due to previous occupational exposure to Mycobacterium particles. We report subgroup analysis of the BATTLE trial, comparing BCG effects in HCWs vs non-HCWs. This was a secondary analysis of a trial. METHODS The BATTLE trial was a double-blind placebo-controlled phase III clinical trial that investigated BCG revaccinating adults who were recently infected with SARS-CoV-2 virus. BCG and placebo recipients were sub-grouped based on regular occupational contact with patients into HCWs (48 BCG and 50 placebo) and non-HCWs (124 BCG and 134 placebo). Weekly COVID-19 symptom progression and injection site reactions were compared between subgroups on weeks one, two, three, and six follow-ups. RESULTS HCWs were more likely to complain of itching on the injection site early after injection (OR = 2.5, p = 0.049). They developed peeling and crusting on the site of injection faster than non-HCWs (during the second week, p = 0.033 and 0.040, OR = 3.3 and 2.7, respectively). HCWs were also more likely to maintain their papule or develop a late onset pustule during later weeks (weeks four and six, p = 0.024 and 0.006, OR = 2.2 and 8.6, respectively). In terms of COVID-19 symptom progression, recovery from anosmia was more likely in the non-HCWs who received BCG (week six, pHolm's corrected = 0.002, OR = 3.3). CONCLUSION HCWs' local reaction to BCG injection was slightly more rapid and more intense, possibly due to their occupational exposure. BCG may also ameliorate COVID-19 induced inflammation and anosmia in non-HCWs but not HCWs. Therefore, HCWs might be less likely to benefit from BCG vaccination. CLINICALTRIALS gov register number NCT04369794.
Collapse
Affiliation(s)
- Mehrsa Jalalizadeh
- UroScience, School of Medical Sciences, University of Campinas, UNICAMP, Campinas, Sao Paulo, Brazil
| | - Patrícia A F Leme
- UroScience, School of Medical Sciences, University of Campinas, UNICAMP, Campinas, Sao Paulo, Brazil
| | - Keini Buosi
- UroScience, School of Medical Sciences, University of Campinas, UNICAMP, Campinas, Sao Paulo, Brazil
| | - Franciele A V Dionato
- UroScience, School of Medical Sciences, University of Campinas, UNICAMP, Campinas, Sao Paulo, Brazil
| | - Luciana S B Dal Col
- UroScience, School of Medical Sciences, University of Campinas, UNICAMP, Campinas, Sao Paulo, Brazil
| | - Cristiane F Giacomelli
- UroScience, School of Medical Sciences, University of Campinas, UNICAMP, Campinas, Sao Paulo, Brazil
| | - Leonardo O Reis
- UroScience, School of Medical Sciences, University of Campinas, UNICAMP, Campinas, Sao Paulo, Brazil; Pontifical Catholic University of Campinas, PUC-Campinas, Sao Paulo, Brazil.
| |
Collapse
|
64
|
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: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
65
|
Pittet LF, Moore CL, McDonald E, Barry S, Bonten M, Campbell J, Croda J, Dalcolmo M, Davidson A, Douglas MW, Gardiner K, Gwee A, Jardim B, Lacerda MV, Lucas M, Lynn DJ, Manning L, de Oliveira RD, Perrett KP, Prat-Aymerich C, Richmond PC, Rocha JL, Rodriguez-Baño J, Warris A, Wood NJ, Messina NL, Curtis N. Bacillus Calmette-Guérin vaccination for protection against recurrent herpes labialis: a nested randomised controlled trial. EClinicalMedicine 2023; 64:102203. [PMID: 37719417 PMCID: PMC10500555 DOI: 10.1016/j.eclinm.2023.102203] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/08/2023] [Accepted: 08/21/2023] [Indexed: 09/19/2023] Open
Abstract
Background Recurrences of herpes simplex virus (HSV) in the orofacial region (herpes labialis or cold sores) impact quality-of-life. We aimed to study whether the bacille Calmette-Guérin (BCG) vaccine can attenuate cold sore recurrences through off-target immunomodulatory effects. Methods In this nested randomised controlled trial within the multicentre, phase 3 BRACE trial, 6828 healthcare workers were randomised in 36 sites in Australia, the Netherlands, Spain, the United Kingdom and Brazil, to receive BCG-Denmark or no BCG (1:1 ratio using a web-based procedure) and followed for 12 months with 3-monthly questionnaires. Exclusion criteria included contraindication to BCG vaccine or previous vaccination with BCG within the past year, any other live-attenuated vaccine within the last month, or any COVID-specific vaccine. The intervention group received one intradermal dose of 0.1 mL of BCG-Denmark corresponding to 2-8 x 105 colony forming units of Mycobacterium bovis, Danish strain 1331. The primary outcome was the difference in restricted mean survival time (i.e., time to first cold-sore recurrence), in participants with frequent recurrent herpes labialis (≥4 recurrences/year), analysed by intention-to-treat. Secondary outcomes addressed additional questions, including analyses in other sub-populations. Adverse events were monitored closely during the first 3 months and were reported in all participants who received one dose of study drug according to intervention received. The BRACE trial is registered with ClinicalTrials.gov, NCT04327206. Findings Between March 30, 2020 and February 18, 2021, 84 individuals with frequent recurrent cold sores were randomly assigned to BCG (n = 38) or control (n = 46). The average time to first cold-sore recurrence was 1.55 months longer in the BCG group (95% CI 0.27-2.82, p = 0.02) than the control group (hazard ratio 0.54, 95% CI 0.32-0.91; intention-to-treat). The beneficial effect of BCG was greater in the as-treated population (difference 1.91 months, 95% CI 0.69-3.12, p = 0.003; hazard ratio 0.45, 95% CI 0.26-0.76). In prespecified subgroup analyses, only sex modified the treatment effect (interaction p = 0.007), with benefit restricted to males. Over 12 months, a greater proportion of participants in the BCG group compared with the control group reported a decrease in duration (61% vs 21%), severity (74% vs 21%), frequency (55% vs 21%), and impact on quality of life (42% vs 15%) of cold sore recurrences. In participants who had ever had a cold sore, there was also a decrease in self-reported burden of recurrences in the BCG group. In participants who had never had a cold sore, there was an increased risk of a first episode in the BCG group (risk difference 1.4%; 95% CI 0.3-2.6%, p = 0.02). There were no safety concerns. Interpretation BCG-Denmark vaccination had a beneficial effect on herpes labialis, particularly in males with frequent recurrences, but may increase the risk of a first cold sore. Funding Bill & Melinda Gates Foundation, the 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, 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, Royal Children's Hospital Melbourne, Parkville, Victoria, Australia
- Immunology, Vaccinology, and Infectious Diseases Unit, Department of Paediatrics, Gynaecology and Obsterics, Faculty of Medicine, University of Geneva and University Hospitals of Geneva, Geneva, Switzerland
| | - Cecilia L. Moore
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
- 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
| | - Ellie McDonald
- Infectious Diseases Group, 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
| | - Margareth Dalcolmo
- Helio Fraga Reference Center, Oswaldo Cruz Foundation Ministry of Health, Curicica, Brazil
- Catholic University, Rio de Janeiro, Brazil
| | - Andrew Davidson
- Melbourne Children's Trial Centre, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Mark W. Douglas
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, 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
- 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
| | - 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
- 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
- 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
| | - Roberto D. de Oliveira
- Nursing Course, State University of Mato Grosso do Sul, Dourados, Brazil
- Graduate Program in Health Sciences, Federal University of Grande Dourados, Dourados, Brazil
| | - 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
| | - 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
| | - Nicole L. Messina
- Infectious Diseases Group, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, 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
|
66
|
Booysen P, Wilkinson KA, Sheerin D, Waters R, Coussens AK, Wilkinson RJ. Immune interaction between SARS-CoV-2 and Mycobacterium tuberculosis. Front Immunol 2023; 14:1254206. [PMID: 37841282 PMCID: PMC10569495 DOI: 10.3389/fimmu.2023.1254206] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 09/12/2023] [Indexed: 10/17/2023] Open
Abstract
SARS-CoV-2 and Mycobacterium tuberculosis (Mtb) are major infectious causes of death, with meta-analyses and population-based studies finding increased mortality in co-infected patients simultaneously diagnosed with COVID-19 and tuberculosis (TB). There is a need to understand the immune interaction between SARS-CoV-2 and Mtb which impacts poor outcomes for those co-infected. We performed a PubMed and preprint search using keywords [SARS-CoV-2] AND [tuberculosis] AND [Immune response], including publications after January 2020, excluding reviews or opinions. Abstracts were evaluated by authors for inclusion of data specifically investigating the innate and/or acquired immune responses to SARS-CoV-2 and Mtb in humans and animal models, immunopathological responses in co-infection and both trials and investigations of potential protection against SARS-CoV-2 by Bacille Calmette Guérin (BCG). Of the 248 articles identified, 39 were included. Incidence of co-infection is discussed, considering in areas with a high burden of TB, where reported co-infection is likely underestimated. We evaluated evidence of the clinical association between COVID-19 and TB, discuss differences and similarities in immune responses in humans and in murine studies, and the implications of co-infection. SARS-CoV-2 and Mtb have both been shown to modulate immune responses, particularly of monocytes, macrophages, neutrophils, and T cells. Co-infection may result in impaired immunity to SARS-CoV-2, with an exacerbated inflammatory response, while T cell responses to Mtb may be modulated by SARS-CoV-2. Furthermore, there has been no proven potential COVID-19 clinical benefit of BCG despite numerous large-scale clinical trials.
Collapse
Affiliation(s)
- Petro Booysen
- Centre for Infectious Diseases Research in Africa (CIDRI-Africa), Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Katalin A. Wilkinson
- Centre for Infectious Diseases Research in Africa (CIDRI-Africa), Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Department of Medicine, University of Cape Town, Cape Town, South Africa
- Tuberculosis Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Dylan Sheerin
- Infectious Diseases and Immune Defence Division, The Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Robyn Waters
- Centre for Infectious Diseases Research in Africa (CIDRI-Africa), Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Anna K. Coussens
- Centre for Infectious Diseases Research in Africa (CIDRI-Africa), Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Infectious Diseases and Immune Defence Division, The Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Robert J. Wilkinson
- Centre for Infectious Diseases Research in Africa (CIDRI-Africa), Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Department of Medicine, University of Cape Town, Cape Town, South Africa
- Tuberculosis Laboratory, The Francis Crick Institute, London, United Kingdom
- Department of Infectious Diseases, Imperial College, London, United Kingdom
| |
Collapse
|
67
|
Perera DJ, Domenech P, Babuadze GG, Naghibosadat M, Alvarez F, Koger-Pease C, Labrie L, Stuible M, Durocher Y, Piccirillo CA, Lametti A, Fiset PO, Elahi SM, Kobinger GP, Gilbert R, Olivier M, Kozak R, Reed MB, Ndao M. BCG administration promotes the long-term protection afforded by a single-dose intranasal adenovirus-based SARS-CoV-2 vaccine. iScience 2023; 26:107612. [PMID: 37670783 PMCID: PMC10475483 DOI: 10.1016/j.isci.2023.107612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 07/19/2023] [Accepted: 08/09/2023] [Indexed: 09/07/2023] Open
Abstract
Recent publications have explored intranasal (i.n.) adenovirus-based (Ad) vaccines as an effective strategy for SARS-CoV-2 in pre-clinical models. However, the effects of prior immunizations and infections have yet to be considered. Here, we investigate the immunomodulatory effects of Mycobacterium bovis BCG pre-immunization followed by vaccination with an S-protein-expressing i.n. Ad, termed Ad(Spike). While i.n. Ad(Spike) retains some protective effect after 6 months, a single administration of BCG-Danish prior to Ad(Spike) potentiates its ability to control viral replication of the B.1.351 SARS-CoV-2 variant within the respiratory tract. Though BCG-Danish did not affect Ad(Spike)-generated humoral immunity, it promoted the generation of cytotoxic/Th1 responses over suppressive FoxP3+ TREG cells in the lungs of infected mice. Thus, this vaccination strategy may prove useful in limiting future pandemics by potentiating the long-term efficacy of mucosal vaccines within the context of the widely distributed BCG vaccine.
Collapse
Affiliation(s)
- Dilhan J. Perera
- Division of Experimental Medicine, McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Pilar Domenech
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- McGill International TB Centre, McGill University, Montréal, QC, Canada
| | - George Giorgi Babuadze
- Department of Biological Sciences, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Maedeh Naghibosadat
- Department of Biological Sciences, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Fernando Alvarez
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
| | - Cal Koger-Pease
- Division of Experimental Medicine, McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Lydia Labrie
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
| | - Matthew Stuible
- Department of Production Platforms & Analytics, Human Health Therapeutics Research Center, National Research Council Canada, Montréal, QC, Canada
| | - Yves Durocher
- Department of Production Platforms & Analytics, Human Health Therapeutics Research Center, National Research Council Canada, Montréal, QC, Canada
| | - Ciriaco A. Piccirillo
- Division of Experimental Medicine, McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
| | - André Lametti
- Department of Pathology, McGill University, Montréal, QC, Canada
| | | | - Seyyed Mehdy Elahi
- Department of Production Platforms & Analytics, Human Health Therapeutics Research Center, National Research Council Canada, Montréal, QC, Canada
| | - Gary P. Kobinger
- Département de Microbiologie-Infectiologie et Immunologie, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Rénald Gilbert
- Department of Production Platforms & Analytics, Human Health Therapeutics Research Center, National Research Council Canada, Montréal, QC, Canada
| | - Martin Olivier
- Division of Experimental Medicine, McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
| | - Robert Kozak
- Department of Biological Sciences, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Molecular Diagnostics, Division of Microbiology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Michael B. Reed
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- McGill International TB Centre, McGill University, Montréal, QC, Canada
| | - Momar Ndao
- Division of Experimental Medicine, McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- National Reference Centre for Parasitology, McGill University Health Centre, Montréal, QC, Canada
| |
Collapse
|
68
|
Hilligan KL, Namasivayam S, Sher A. BCG mediated protection of the lung against experimental SARS-CoV-2 infection. Front Immunol 2023; 14:1232764. [PMID: 37744331 PMCID: PMC10514903 DOI: 10.3389/fimmu.2023.1232764] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/18/2023] [Indexed: 09/26/2023] Open
Abstract
The observation of reduced COVID-19 incidence and severity in populations receiving neonatal intradermal BCG vaccination vaccine raised the question of whether BCG can induce non-specific protection against the SARS-CoV-2 (SCV2) virus. Subsequent epidemiologic studies and clinical trials have largely failed to support this hypothesis. Furthermore, in small animal model studies all investigators have failed to observe resistance to viral challenge in response to BCG immunization by the conventional and clinically acceptable intradermal or subcutaneous routes. Nevertheless, BCG administered by the intravenous (IV) route has been shown to strongly protect both hamsters and mice against SCV2 infection and disease. In this Perspective, we review the current data on the effects of BCG vaccination on resistance to COVID-19 as well as summarize recent work in rodent models on the mechanisms by which IV administered BCG promotes resistance to the virus and discuss the translational implications of these findings.
Collapse
Affiliation(s)
- Kerry L. Hilligan
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States
- Immune Cell Biology Programme, Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Sivaranjani Namasivayam
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States
| | - Alan Sher
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States
| |
Collapse
|
69
|
Şimşek-Yavuz S. COVID-19: An Update on Epidemiology, Prevention and Treatment, September-2023. INFECTIOUS DISEASES & CLINICAL MICROBIOLOGY 2023; 5:165-187. [PMID: 38633552 PMCID: PMC10986731 DOI: 10.36519/idcm.2023.251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/19/2023] [Indexed: 04/19/2024]
Abstract
After a downward trend for more than 12 months, the incidence of COVID-19 has increased in the last months. Although COVID-19 is not as frequent as in the first years of the pandemic, case numbers are still very high, and it causes a significant number of deaths. COVID-19 is not seen with a predictable frequency, at least two times more deadly than the flu, continues as an epidemic, and has not reached the endemic level yet. Currently, the Omicron strains EG.5 and XBB.1.16 are dominant worldwide. Although BA.2.86 and FLip variants, including FL.1.5.1 are not widespread at the moment, both were shown to be highly immune-evasive and require close monitoring. Prevention of COVID-19 relies on vaccinations, surveillance, proper ventilation of enclosed spaces, isolation of patients, and mask usage. Currently, monovalent COVID-19 vaccines, including XBB.1.5 Omicron SARS-CoV-2, are recommended for both primary and booster vaccinations against COVID-19. Monovalent vaccines, including only original SARS-CoV-2 strain, and bivalent vaccines, including original virus plus BA4/5 variant, are no longer recommended against COVID-19. Booster vaccination with XBB.1.5 containing vaccine should be prioritized for patients at high risk for severe COVID-19. Bacillus Calmette-Guérin (BCG) vaccination does not seem to be effective in preventing COVID-19. At the current phase of the pandemic, nirmatrelvir/ritonavir, remdesivir, molnupiravir, sotrovimab (for patients from XBB.1.5 variant dominant settings), and convalescent plasma can be considered for the treatment of high-risk early-stage outpatients with COVID-19, while hospitalized patients with more severe disease can be treated with dexamethasone, anti cytokines including tocilizumab, sarilumab, baricitinib, and tofacitinib and antithrombotic agents including enoxaparin. Remdesivir oral analogues and ensitrelvir fumarate are promising agents for treating acute COVID-19, which are in phase trials now; however, ivermectin, fluvoxamine, and metformin were shown to be ineffective.
Collapse
Affiliation(s)
- Serap Şimşek-Yavuz
- Department of Infectious Diseases and Clinical Microbiology, İstanbul University School of Medicine, İstanbul, Türkiye
| |
Collapse
|
70
|
Trauth J. [Respiratory viral infections : Under special consideration of severe acute respiratory syndrome coronavirus 2 and influenza viruses]. Med Klin Intensivmed Notfmed 2023; 118:445-453. [PMID: 37642653 DOI: 10.1007/s00063-023-01050-7] [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: 05/15/2023] [Accepted: 05/22/2023] [Indexed: 08/31/2023]
Abstract
Respiratory viruses cause the highest number of morbidities and deaths annually among all infectious pathogens. This article discusses the current epidemiology, pathogenesis, risk factors, and drug treatment of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), influenza viruses, respiratory syncytial virus (RSV), and other respiratory viruses. The SARS-CoV‑2 and influenza are preventable with vaccines and a first vaccine against RSV is available since 08/2023. For infections with SARS-CoV‑2 and influenza, a stage-specific (antiviral) drug treatment is also recommended. Due to the high and commonly underestimated disease burden caused by RSV, it must be hoped that antiviral substances will be found in the future. In patients at risk, particular attention should be paid to an adequate vaccination status against respiratory pathogens and if there is clinical suspicion of a viral airway infection, the pathogen should be promptly identified and, if necessary, specific treatment should be carried out. Now that effective vaccinations and antiviral drugs are available, the challenge is to use them for all patients at risk and thus really prevent avoidable infections, severe courses and long-term sequelae.
Collapse
Affiliation(s)
- Janina Trauth
- Universitätsklinikum Gießen und Marburg GmbH, Medizinische Klinik V für Innere Medizin m.S. Infektiologie und Krankenhaushygiene, Justus-Liebig-Universität Gießen, Klinikstr 33, 35392, Gießen, Deutschland.
| |
Collapse
|
71
|
Corleis B, Bastian M, Hoffmann D, Beer M, Dorhoi A. Animal models for COVID-19 and tuberculosis. Front Immunol 2023; 14:1223260. [PMID: 37638020 PMCID: PMC10451089 DOI: 10.3389/fimmu.2023.1223260] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/21/2023] [Indexed: 08/29/2023] Open
Abstract
Respiratory infections cause tremendous morbidity and mortality worldwide. Amongst these diseases, tuberculosis (TB), a bacterial illness caused by Mycobacterium tuberculosis which often affects the lung, and coronavirus disease 2019 (COVID-19) caused by the Severe Acute Respiratory Syndrome Coronavirus type 2 (SARS-CoV-2), stand out as major drivers of epidemics of global concern. Despite their unrelated etiology and distinct pathology, these infections affect the same vital organ and share immunopathogenesis traits and an imperative demand to model the diseases at their various progression stages and localizations. Due to the clinical spectrum and heterogeneity of both diseases experimental infections were pursued in a variety of animal models. We summarize mammalian models employed in TB and COVID-19 experimental investigations, highlighting the diversity of rodent models and species peculiarities for each infection. We discuss the utility of non-human primates for translational research and emphasize on the benefits of non-conventional experimental models such as livestock. We epitomize advances facilitated by animal models with regard to understanding disease pathophysiology and immune responses. Finally, we highlight research areas necessitating optimized models and advocate that research of pulmonary infectious diseases could benefit from cross-fertilization between studies of apparently unrelated diseases, such as TB and COVID-19.
Collapse
Affiliation(s)
- Björn Corleis
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Max Bastian
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Donata Hoffmann
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Martin Beer
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Anca Dorhoi
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
- Faculty of Mathematics and Natural Sciences, University of Greifswald, Greifswald, Germany
| |
Collapse
|
72
|
Villanueva P, Crawford NW, Garcia Croda M, Collopy S, Araújo Jardim B, de Almeida Pinto Jardim T, Marshall H, Prat-Aymerich C, Sawka A, Sharma K, Troeman D, Wadia U, Warris A, Wood N, Messina NL, Curtis N, Pittet LF. Safety of BCG vaccination and revaccination in healthcare workers. Hum Vaccin Immunother 2023; 19:2239088. [PMID: 37551885 PMCID: PMC10411308 DOI: 10.1080/21645515.2023.2239088] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/09/2023] [Accepted: 07/16/2023] [Indexed: 08/09/2023] Open
Abstract
BCG vaccination and revaccination are increasingly being considered for the protection of adolescents and adults against tuberculosis and, more broadly, for the off-target protective immunological effects against other infectious and noninfectious diseases. Within an international randomized controlled trial of BCG vaccination in healthcare workers (the BRACE trial), we evaluated the incidence of local and serious adverse events, as well as the impact of previous BCG vaccination on local injection site reactions (BCG revaccination). Prospectively collected data from 99% (5351/5393) of participants in Australia, Brazil, Spain, The Netherlands and the UK was available for analysis. Most BCG recipients experienced the expected self-limiting local injection site reactions (pain, tenderness, erythema, swelling). BCG injection site itch was an additional common initial local symptom reported in 49% of BCG recipients. Compared to BCG vaccination in BCG-naïve individuals, BCG revaccination was associated with increased frequency of mild injection site reactions, as well as earlier onset and shorter duration of erythema and swelling, which were generally self-limiting. Injection site abscess and regional lymphadenopathy were the most common adverse events and had a benign course. Self-resolution occurred within a month in 80% of abscess cases and 100% of lymphadenopathy cases. At a time when BCG is being increasingly considered for its off-target effects, our findings indicate that BCG vaccination and revaccination have an acceptable safety profile in adults.
Collapse
Affiliation(s)
- Paola Villanueva
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
- Infection and Immunity, Murdoch Children’s Research Institute, Parkville, VIC, Australia
- Infectious Diseases, Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
- Department of General Medicine, Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
| | - Nigel W. Crawford
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
- Infection and Immunity, Murdoch Children’s Research Institute, Parkville, VIC, Australia
- Department of General Medicine, Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
- Immunisation Service, Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
| | - Mariana Garcia Croda
- School of Medicine, Federal University of Mato Grosso do Sul, Campo Grande, Brazil
| | - Simone Collopy
- Department of Pediatrics, Universidade Estadual do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bruno Araújo Jardim
- Carlos Borborema Clinical Research Institute, Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Tyane de Almeida Pinto Jardim
- Carlos Borborema Clinical Research Institute, Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Helen Marshall
- The University of Adelaide and the Women’s and Children’s Health Network, Adelaide, SA, Australia
| | - Cristina Prat-Aymerich
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- Institut d’Investigació Germans Trias i Pujol, Departament de Genètica i Microbiologia, CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Alice Sawka
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Ketaki Sharma
- National Centre for Immunisation Research and Surveillance, Westmead, NSW, Australia
- The Children’s Hospital at Westmead, Westmead, NSW, Australia
| | - Darren Troeman
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Ushma Wadia
- Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, WA, Australia
| | - Adilia Warris
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
- Department of Infectious Diseases, Great Ormond Street Hospital, London, UK
| | - Nicholas Wood
- National Centre for Immunisation Research and Surveillance, Westmead, NSW, Australia
- The Children’s Hospital at Westmead, Westmead, NSW, Australia
- The Children’s Hospital at Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Nicole L. Messina
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
- Infection and Immunity, Murdoch Children’s Research Institute, Parkville, VIC, Australia
| | - Nigel Curtis
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
- Infection and Immunity, Murdoch Children’s Research Institute, Parkville, VIC, Australia
- Infectious Diseases, Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
| | - Laure F. Pittet
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
- Infection and Immunity, Murdoch Children’s Research Institute, Parkville, VIC, Australia
- Infectious Diseases, Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
- Infectious Diseases Unit, Department of Paediatrics, Gynaecology and Obstetrics, Faculty of Medicine, University of Geneva, University Hospitals of Geneva, Geneva, Switzerland
| |
Collapse
|
73
|
|
74
|
Netea MG, Ziogas A, Benn CS, Giamarellos-Bourboulis EJ, Joosten LAB, Arditi M, Chumakov K, van Crevel R, Gallo R, Aaby P, van der Meer JWM. The role of trained immunity in COVID-19: Lessons for the next pandemic. Cell Host Microbe 2023; 31:890-901. [PMID: 37321172 PMCID: PMC10265767 DOI: 10.1016/j.chom.2023.05.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 05/03/2023] [Accepted: 05/08/2023] [Indexed: 06/17/2023]
Abstract
Trained immunity is a long-term increase in responsiveness of innate immune cells, induced by certain infections and vaccines. During the last 3 years of the COVID-19 pandemic, vaccines that induce trained immunity, such as BCG, MMR, OPV, and others, have been investigated for their capacity to protect against COVID-19. Further, trained immunity-inducing vaccines have been shown to improve B and T cell responsiveness to both mRNA- and adenovirus-based anti-COVID-19 vaccines. Moreover, SARS-CoV-2 infection itself induces inappropriately strong programs of trained immunity in some individuals, which may contribute to the long-term inflammatory sequelae. In this review, we detail these and other aspects of the role of trained immunity in SARS-CoV-2 infection and COVID-19. We also examine the learnings from the trained immunity studies conducted in the context of this pandemic and discuss how they may help us in preparing for future infectious outbreaks.
Collapse
Affiliation(s)
- Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Immunology and Metabolism, Life & Medical Sciences Institute, University of Bonn, Bonn, Germany.
| | - Athanasios Ziogas
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Christine Stabell Benn
- Bandim Health Project, OPEN, Department of Clinical Research, University of Southern Denmark, Copenhagen, Denmark; Danish Institute for Advanced Study, University of Southern Denmark, Odense, Denmark
| | | | - Leo A B Joosten
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Moshe Arditi
- Departments of Pediatrics and Biomedical Sciences, Guerin Children's and Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, USA
| | - Konstantin Chumakov
- Office of Vaccines Research and Review, Food and Drug Administration, Global Virus Network Center of Excellence, Silver Spring, MD, USA
| | - Reinout van Crevel
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Robert Gallo
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Global Virus Network, Baltimore, MD, USA
| | - Peter Aaby
- Bandim Health Project, OPEN, Department of Clinical Research, University of Southern Denmark, Copenhagen, Denmark
| | - Jos W M van der Meer
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
75
|
Tang C, Kurata S, Fuse N. Re-recognition of innate immune memory as an integrated multidimensional concept. Microbiol Immunol 2023. [PMID: 37311618 DOI: 10.1111/1348-0421.13083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/15/2023]
Abstract
In the past decade, the concept of immunological memory, which has long been considered a phenomenon observed in the adaptive immunity of vertebrates, has been extended to the innate immune system of various organisms. This de novo immunological memory is mainly called "innate immune memory", "immune priming", or "trained immunity" and has received increased attention because of its potential for clinical and agricultural applications. However, research on different species, especially invertebrates and vertebrates, has caused controversy regarding this concept. Here we discuss the current studies focusing on this immunological memory and summarize several mechanisms underlying it. We propose "innate immune memory" as a multidimensional concept as an integration between the seemingly different immunological phenomena.
Collapse
Affiliation(s)
- Chang Tang
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Shoichiro Kurata
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Naoyuki Fuse
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| |
Collapse
|
76
|
Hopes fizzle that a TB vaccine also fends off COVID. Nature 2023; 617:10. [PMID: 37101078 DOI: 10.1038/d41586-023-01426-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
|
77
|
Ng TW, Porcelli SA. Designing Anti-Viral Vaccines that Harness Intrastructural Help from Prior BCG Vaccination. JOURNAL OF CELLULAR IMMUNOLOGY 2023; 5:97-102. [PMID: 37946751 PMCID: PMC10635577 DOI: 10.33696/immunology.5.174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
Vaccines are among the most effective tools for combatting the impact and spread of infectious diseases. However, the effectiveness of a vaccine can be diminished by vaccine inequality, particularly during severe outbreaks of infectious diseases in resource-poor areas. As seen in many developing countries that lack adequate healthcare infrastructure and economic resources, the acquisition and distribution of potentially life-saving vaccines may be limited, leading to prolonged suffering and increased deaths. To improve vaccine equity, vaccine design must take into consideration the logistics needed to implement a successful vaccination drive, particularly among the most vulnerable populations. In the manuscript titled "Exploiting Pre-Existing CD4+ T Cell Help from Bacille Calmette-Guérin Vaccination to Improve Antiviral Antibody Responses" published in the Journal of Immunology, the authors designed a recombinant subunit vaccine against the Ebola virus (EBOV) glycoprotein that can harness the pre-existing T helper cells from prior BCG vaccination. As a recombinant subunit vaccine adjuvanted with alum, this approach has many features that make it well suited for the design of vaccines for developing nations, such as relative ease of production, scalability, and distribution. In addition, the high prevalence of BCG immunization and natural immunity to mycobacteria in many regions of the world endow such vaccines with features that should increase potency and efficacy among populations residing in such regions. As a result of using the helper activity of pre-existing BCG-specific Th cells to drive antibody responses, a lower vaccine dose is needed, which is a major advantage for vaccine manufacture. Furthermore, the BCG-specific Th cells also stimulate immunoglobulin class switching to IgG isotypes that have strong affinities for activating Fc-gamma receptors (FcγRs). Taken together, we propose that the design of subunit vaccines with intrastructural help from BCG-specific Th cells can improve protection against viral infection and represents a vaccine design that can be generally adapted to other emerging viral pathogens for the control and prevention of infection in many developing countries.
Collapse
Affiliation(s)
- Tony W. Ng
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY10461, USA
| | - Steven A. Porcelli
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY10461, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|