1
|
Nagai A, Lemes RB, Mill JG, Pereira AC, Marques RE, Hünemeier T. The seroconversion history to SARS-CoV-2 in Indigenous people from Brazil - the interplay between exposure, vaccination, and tuberculosis. Front Immunol 2024; 15:1359066. [PMID: 39081316 PMCID: PMC11286483 DOI: 10.3389/fimmu.2024.1359066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 06/24/2024] [Indexed: 08/02/2024] Open
Abstract
The COVID-19 pandemic caused a significant loss of human lives and a worldwide decline in quality of life. Although our understanding of the pandemic has improved significantly since the beginning, the natural history of COVID-19 and its impacts on under-represented populations, such as Indigenous people from America, remain largely unknown. We performed a retrospective serological survey with two Brazilian Indigenous populations (n=624), Tupiniquim and Guarani-Mbyá. Samples were collected between September 2020 and July 2021: a period comprising the dissemination of SARS-CoV-2 variants and the beginning of COVID-19 vaccination in Brazil. Seroconversions against S and N antigens were assessed using three different commercially available ELISA kits. Samples were also used to assess the prevalence of tuberculosis (TB) in the same population (n=529). Seroconversion against SARS-CoV-2 antigens was considered positive if at least one of the three ELISA kits detected levels of specific antibodies above the threshold specified by the manufacturer. In this sense, we report 56.0% (n=349/623) of seroconverted individuals. Relative seroconversion peaked after introduction of the Coronavac vaccine in February 2021. Vaccination increased the production of anti-S IgG from 3.9% to 48.6%. Our results also indicated that 11.0% (n=46/417) of all individuals were positive for TB. Seroconversion to SARS-CoV-2 was similar between individuals with positive tuberculosis test results to those with negative test results. Most vaccinated individuals seroconverted to SARS-CoV-2, indicating that Coronavac may be as protective in individuals from these indigenous groups as observed in the general Brazilian population. COVID-19 severity was minimal regardless of incomplete vaccine coverage, suggesting that vaccination may not be the only factor protecting individuals from severe COVID-19. Tuberculosis is highly prevalent and not associated with increased seroconversion to SARS-CoV-2.
Collapse
Affiliation(s)
- Alice Nagai
- Laboratory of Pathogen Manipulation, Brazilian Biosciences National Laboratory (LNBio), Department of Virology, CNPEM (Brazilian Center for Research in Energy and Materials), Campinas, Brazil
| | - Renan Barbosa Lemes
- Human Population Genomics Laboratory, Biosciences Institute, Department of Genetics and Evolutionary Biology, University of São Paulo, São Paulo, Brazil
| | - José Geraldo Mill
- Health Sciences Center, Department of Physiological Sciences, Federal University of Espírito Santo, Vitória, Brazil
| | - Alexandre Costa Pereira
- Laboratory of Genetics and Molecular Cardiology, Instituto do Coração, Hospital das Clínicas, University of São Paulo, São Paulo, Brazil
- Department of Genetics, Harvard Medical School, Boston, MA, United States
| | - Rafael Elias Marques
- Laboratory of Pathogen Manipulation, Brazilian Biosciences National Laboratory (LNBio), Department of Virology, CNPEM (Brazilian Center for Research in Energy and Materials), Campinas, Brazil
| | - Tábita Hünemeier
- Human Population Genomics Laboratory, Biosciences Institute, Department of Genetics and Evolutionary Biology, University of São Paulo, São Paulo, Brazil
- Department of Population Genetics, Institut de Biologia Evolutiva (CSIC/Universitat Pompeu Fabra), Barcelona, Spain
| |
Collapse
|
2
|
Juste RA, Vrotsou K, Mateo-Abad M, Gutiérrez-Stampa MA, Rotaeche R, Vergara I, Bujanda L. Non-specific protection against severe COVID-19 associated to typhoid fever and DTP vaccination. Heliyon 2024; 10:e29935. [PMID: 38707311 PMCID: PMC11068531 DOI: 10.1016/j.heliyon.2024.e29935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 05/07/2024] Open
Abstract
Trained immunity (TRAIM) or the enhanced non-specific immune response after primary stimulation by infection or vaccination is a recent but well-recognized concept. To verify its predictions, our objective was to determine the effects of two bacterial vaccines, typhoid fever (TFV) and diphtheria-tetanus-pertussis (DTP) on the infection, hospitalization and death frequencies associated to COVID-19 in a retrospective study on subjects vaccinated or not with TFV and DTP in the 4 years prior to the start of COVID-19 pandemia in the Basque Country (Spain). The studied outcome records were split into two periods according to COVID-19 vaccination, the pre-vaccination (ACV) from March to December 2020 and the post-vaccination (PCV) from September 2021 to June 2022). In total, 13,673 subjects were vaccinated against TFV and 42,997 against DTP. A total of 2,005,084 individual records were studied in the ACV period and 1,436,693 in the PCV period. The proportion of infection, hospitalization and death associated to COVID-19 among controls in ACV was 4.97 %, 7.14 % and 3.54 %, respectively vs. 7.20 %, 2.24 % and 0.10 % among TFV subjects. Regarding DTP, the proportions were 4.97 %, 7.12 % and 3.58 % for controls and 5.79 %, 5.79 % and 0.80 % for vaccinees. In the PCV period, the proportion of infection, hospitalization and death among controls was 21.89 %, 2.62 % and 0.92 %, respectively vs. 31.19 %, 0.76 %, 0.00 % among TFV. For DTP, infection, hospitalization and death proportions were 21.89 %, 2.62 % and 0.92 %, respectively, among controls vs. 32.03 %, 1.85 % and 0.24 % among vaccinated subjects. The corresponding combined ACV and PCV odds ratios (OR) for SARS-CoV2 infection were 1.505 (95%CI 1.455-1.558; p < 0.0001; reduction -41.85 %) and 1.633 (95%CI 1.603-1.662; p < 0.0001; reduction -51.74 %), for TFV and DTP, respectively. Regarding COVID-19 associated hospitalization, the OR were 0.295 (95%CI 0.220-0.396; p = 0.0001; reduction 69.74 %) and 0.667 (95%CI 0.601-0.741; p = 0.0001; reduction 32.44 %), for TFV and DTP, respectively). COVID-19 associated death OR were 0.016 (95%CI 0.002-0.113, p < 0.0001; reduction 98.38 %) and 0.212 (95%CI 0.161-0.280; p = 0.0001; reduction 78.52 %), for TFV and DTP, respectively. We conclude that TRAIM effects by TFV and DTP vaccination in the four years prior to the pandemic SARS-CoV2 were supported by slightly increased infection rates, but strongly reduced COVID-19 associated hospitalization and death rates.
Collapse
Affiliation(s)
- Ramon A. Juste
- NEIKER-Basque Institute for Agricultural Research and Development, Basque Research and Technology Alliance (BRTA), Parque Científico y Tecnológico de Bizkaia, P812, E-48160 Derio, Spain
| | - Kalliopi Vrotsou
- Osakidetza Health Care Directorate, PC-IHO Research Unit of Gipuzkoa, Donostia-San Sebastián, Spain, P° Dr. Beguiristain, s/n 20014 Donostia-San Sebastian, Spain
- Primary Care Group, Biogipuzkoa Institute for Health Research, Donostia-San Sebastián, Spain
- Red de Investigación en Cronicidad, Atención Primaria y Promoción de la Salud (RICAPPS) , Spain
| | - Maider Mateo-Abad
- Primary Care Group, Biogipuzkoa Institute for Health Research, Donostia-San Sebastián, Spain
- Red de Investigación en Cronicidad, Atención Primaria y Promoción de la Salud (RICAPPS) , Spain
| | - Maria A. Gutiérrez-Stampa
- Primary Care Group, Biogipuzkoa Institute for Health Research, Donostia-San Sebastián, Spain
- Red de Investigación en Cronicidad, Atención Primaria y Promoción de la Salud (RICAPPS) , Spain
- Altza Primary Care Health Center, Donostialdea IHO, Biodonostia Health Research Institute, 20014 Donostia-San Sebastián, Spain
| | - Rafael Rotaeche
- Osakidetza Health Care Directorate, PC-IHO Research Unit of Gipuzkoa, Donostia-San Sebastián, Spain, P° Dr. Beguiristain, s/n 20014 Donostia-San Sebastian, Spain
- Primary Care Group, Biogipuzkoa Institute for Health Research, Donostia-San Sebastián, Spain
- Red de Investigación en Cronicidad, Atención Primaria y Promoción de la Salud (RICAPPS) , Spain
- Altza Primary Care Health Center, Donostialdea IHO, Biodonostia Health Research Institute, 20014 Donostia-San Sebastián, Spain
| | - Itziar Vergara
- Osakidetza Health Care Directorate, PC-IHO Research Unit of Gipuzkoa, Donostia-San Sebastián, Spain, P° Dr. Beguiristain, s/n 20014 Donostia-San Sebastian, Spain
- Primary Care Group, Biogipuzkoa Institute for Health Research, Donostia-San Sebastián, Spain
- Red de Investigación en Cronicidad, Atención Primaria y Promoción de la Salud (RICAPPS) , Spain
| | - Luis Bujanda
- Department of Gastroenterology, Biodonostia Health Research Institute, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Universidad del País Vasco (UPV/EHU), Donostia-San Sebastian, Spain
| |
Collapse
|
3
|
Nakamura N, Kobashi Y, Kim KS, Park H, Tani Y, Shimazu Y, Zhao T, Nishikawa Y, Omata F, Kawashima M, Yoshida M, Abe T, Saito Y, Senoo Y, Nonaka S, Takita M, Yamamoto C, Kawamura T, Sugiyama A, Nakayama A, Kaneko Y, Jeong YD, Tatematsu D, Akao M, Sato Y, Iwanami S, Fujita Y, Wakui M, Aihara K, Kodama T, Shibuya K, Iwami S, Tsubokura M. Modeling and predicting individual variation in COVID-19 vaccine-elicited antibody response in the general population. PLOS DIGITAL HEALTH 2024; 3:e0000497. [PMID: 38701055 PMCID: PMC11068210 DOI: 10.1371/journal.pdig.0000497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 02/14/2024] [Indexed: 05/05/2024]
Abstract
As we learned during the COVID-19 pandemic, vaccines are one of the most important tools in infectious disease control. To date, an unprecedentedly large volume of high-quality data on COVID-19 vaccinations have been accumulated. For preparedness in future pandemics beyond COVID-19, these valuable datasets should be analyzed to best shape an effective vaccination strategy. We are collecting longitudinal data from a community-based cohort in Fukushima, Japan, that consists of 2,407 individuals who underwent serum sampling two or three times after a two-dose vaccination with either BNT162b2 or mRNA-1273. Using the individually reconstructed time courses of the vaccine-elicited antibody response based on mathematical modeling, we first identified basic demographic and health information that contributed to the main features of the antibody dynamics, i.e., the peak, the duration, and the area under the curve. We showed that these three features of antibody dynamics were partially explained by underlying medical conditions, adverse reactions to vaccinations, and medications, consistent with the findings of previous studies. We then applied to these factors a recently proposed computational method to optimally fit an "antibody score", which resulted in an integer-based score that can be used as a basis for identifying individuals with higher or lower antibody titers from basic demographic and health information. The score can be easily calculated by individuals themselves or by medical practitioners. Although the sensitivity of this score is currently not very high, in the future, as more data become available, it has the potential to identify vulnerable populations and encourage them to get booster vaccinations. Our mathematical model can be extended to any kind of vaccination and therefore can form a basis for policy decisions regarding the distribution of booster vaccines to strengthen immunity in future pandemics.
Collapse
Affiliation(s)
- Naotoshi Nakamura
- interdisciplinary Biology Laboratory (iBLab), Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Yurie Kobashi
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima, Japan
- Department of General Internal Medicine, Hirata Central Hospital, Fukushima, Japan
| | - Kwang Su Kim
- interdisciplinary Biology Laboratory (iBLab), Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya, Japan
- Department of Science System Simulation, Pukyong National University, Busan, South Korea
- Department of Mathematics, Pusan National University, Busan, South Korea
| | - Hyeongki Park
- interdisciplinary Biology Laboratory (iBLab), Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Yuta Tani
- Medical Governance Research Institute, Tokyo, Japan
| | - Yuzo Shimazu
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Tianchen Zhao
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Yoshitaka Nishikawa
- Department of General Internal Medicine, Hirata Central Hospital, Fukushima, Japan
| | - Fumiya Omata
- Department of General Internal Medicine, Hirata Central Hospital, Fukushima, Japan
| | - Moe Kawashima
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima, Japan
| | | | - Toshiki Abe
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima, Japan
| | | | - Yuki Senoo
- Medical Governance Research Institute, Tokyo, Japan
| | - Saori Nonaka
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Morihito Takita
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Chika Yamamoto
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Takeshi Kawamura
- Proteomics Laboratory, Isotope Science Center, The University of Tokyo, Tokyo, Japan
- Laboratory for Systems Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Akira Sugiyama
- Proteomics Laboratory, Isotope Science Center, The University of Tokyo, Tokyo, Japan
| | - Aya Nakayama
- Proteomics Laboratory, Isotope Science Center, The University of Tokyo, Tokyo, Japan
| | - Yudai Kaneko
- Laboratory for Systems Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
- Medical & Biological Laboratories Co., Ltd, Tokyo, Japan
| | - Yong Dam Jeong
- interdisciplinary Biology Laboratory (iBLab), Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya, Japan
- Department of Mathematics, Pusan National University, Busan, South Korea
| | - Daiki Tatematsu
- interdisciplinary Biology Laboratory (iBLab), Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Marwa Akao
- interdisciplinary Biology Laboratory (iBLab), Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Yoshitaka Sato
- Department of Virology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shoya Iwanami
- interdisciplinary Biology Laboratory (iBLab), Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Yasuhisa Fujita
- interdisciplinary Biology Laboratory (iBLab), Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Masatoshi Wakui
- Department of Laboratory Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Kazuyuki Aihara
- International Research Center for Neurointelligence, The University of Tokyo Institutes for Advanced Study, The University of Tokyo, Tokyo, Japan
| | - Tatsuhiko Kodama
- Laboratory for Systems Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Kenji Shibuya
- Soma Medical Center of Vaccination for COVID-19, Fukushima, Japan
- Tokyo Foundation for Policy Research, Tokyo, Japan
| | - Shingo Iwami
- interdisciplinary Biology Laboratory (iBLab), Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya, Japan
- Institute of Mathematics for Industry, Kyushu University, Fukuoka, Japan
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
- Interdisciplinary Theoretical and Mathematical Sciences Program (iTHEMS), RIKEN, Saitama, Japan
- NEXT-Ganken Program, Japanese Foundation for Cancer Research (JFCR), Tokyo, Japan
- Science Groove Inc., Fukuoka, Japan
| | - Masaharu Tsubokura
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima, Japan
- Department of General Internal Medicine, Hirata Central Hospital, Fukushima, Japan
- Medical Governance Research Institute, Tokyo, Japan
- Minamisoma Municipal General Hospital, Fukushima, Japan
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Guthrie CM, Tan X, Meeker AC, Self AE, Liu L, Cheng Y. Engineering a dual vaccine against COVID-19 and tuberculosis. Front Cell Infect Microbiol 2023; 13:1273019. [PMID: 37965265 PMCID: PMC10641007 DOI: 10.3389/fcimb.2023.1273019] [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: 08/05/2023] [Accepted: 10/12/2023] [Indexed: 11/16/2023] Open
Abstract
The COVID-19 pandemic, caused by SARS-CoV-2 virus, has been one of the top public health threats across the world over the past three years. Mycobacterium bovis BCG is currently the only licensed vaccine for tuberculosis, one of the deadliest infectious diseases in the world, that is caused by Mycobacterium tuberculosis. In the past decades, recombinant M.bovis BCG has been studied as a novel vaccine vector for other infectious diseases in humans besides tuberculosis, such as viral infections. In the current study, we generated a recombinant M. bovis BCG strain AspikeRBD that expresses a fusion protein consisting of M. tb Ag85A protein and the receptor-binding domain (RBD) of the SARS-CoV-2 spike protein using synthetic biology technique. Our results show that the recombinant M. bovis BCG strain successfully expressed this fusion protein. Interestingly, the recombinant M. bovis BCG strain AspikeRBD significantly induced SARS-CoV-2 spike-specific T cell activation and IgG production in mice when compared to the parental M.bovis BCG strain, and was more potent than the recombinant M.bovis BCG strain expressing SARS-CoV-2 spike RBD alone. As expected, the recombinant M. bovis BCG strain AspikeRBD activated an increased number of M. tb Ag85A-specific IFNγ-releasing T cells and enhanced IgG production in mice when compared to the parental M.bovis BCG strain or the BCG strain expressing SARS-CoV-2 spike RBD alone. Taken together, our results indicate a potential application of the recombinant M. bovis BCG strain AspikeRBD as a novel dual vaccine against SARS-CoV-2 and M. tb in humans.
Collapse
Affiliation(s)
- Carlyn Monèt Guthrie
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, OK, United States
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK, United States
| | - Xuejuan Tan
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, OK, United States
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK, United States
| | - Amber Cherry Meeker
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, OK, United States
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK, United States
| | - Ashton Elisabeth Self
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, OK, United States
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK, United States
| | - Lin Liu
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK, United States
- Department of Physiological Sciences, Oklahoma State University, Stillwater, OK, United States
| | - Yong Cheng
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, OK, United States
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK, United States
| |
Collapse
|
7
|
Noé A, Dang TD, Axelrad C, Burrell E, Germano S, Elia S, Burgner D, Perrett KP, Curtis N, Messina NL. BNT162b2 COVID-19 vaccination in children alters cytokine responses to heterologous pathogens and Toll-like receptor agonists. Front Immunol 2023; 14:1242380. [PMID: 37691937 PMCID: PMC10485613 DOI: 10.3389/fimmu.2023.1242380] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 07/24/2023] [Indexed: 09/12/2023] Open
Abstract
Background Vaccines can have beneficial off-target (heterologous) effects that alter immune responses to, and protect against, unrelated infections. The heterologous effects of COVID-19 vaccines have not been investigated in children. Aim To investigate heterologous and specific immunological effects of BNT162b2 COVID-19 vaccination in children. Methods A whole blood stimulation assay was used to investigate in vitro cytokine responses to heterologous stimulants (killed pathogens, Toll-like receptor ligands) and SARS-CoV-2 antigens. Samples from 29 children, aged 5-11 years, before and 28 days after a second BNT162b2 vaccination were analysed (V2 + 28). Samples from eight children were analysed six months after BNT162b2 vaccination. Results At V2 + 28, interferon-γ and monocyte chemoattractant protein-1 responses to S. aureus, E. coli, L. monocytogenes, BCG vaccine, H. influenzae, hepatitis B antigen, poly(I:C) and R848 stimulations were decreased compared to pre-vaccination. For most of these heterologous stimulants, IL-6, IL-15 and IL-17 responses were also decreased. There were sustained decreases in cytokine responses to viral, but not bacterial, stimulants six months after BNT162b2 vaccination. Cytokine responses to irradiated SARS-CoV-2, and spike glycoprotein subunits (S1 and S2) were increased at V2 + 28 for most cytokines and remained higher than pre-vaccination responses 6 months after BNT162b2 vaccination for irradiated SARS-CoV-2 and S1. There was no correlation between BNT162b2 vaccination-induced anti-SARS-CoV2-receptor binding domain IgG antibody titre at V2 + 28 and cytokine responses. Conclusions BNT162b2 vaccination in children alters cytokine responses to heterologous stimulants, particularly one month after vaccination. This study is the first to report the immunological heterologous effects of COVID-19 vaccination in children.
Collapse
Affiliation(s)
- Andrés Noé
- Infection, Immunity and Global Health, Murdoch Children’s Research Institute, Parkville, VIC, Australia
- Infectious Diseases Unit, The Royal Children’s Hospital, Melbourne, Parkville, VIC, Australia
| | - Thanh D. Dang
- Infection, Immunity and Global Health, Murdoch Children’s Research Institute, Parkville, VIC, Australia
| | - Christine Axelrad
- Infection, Immunity and Global Health, Murdoch Children’s Research Institute, Parkville, VIC, Australia
| | - Emma Burrell
- Infection, Immunity and Global Health, Murdoch Children’s Research Institute, Parkville, VIC, Australia
| | - Susie Germano
- Infection, Immunity and Global Health, Murdoch Children’s Research Institute, Parkville, VIC, Australia
| | - Sonja Elia
- Infectious Diseases Unit, The Royal Children’s Hospital, Melbourne, Parkville, VIC, Australia
| | - David Burgner
- Infection, Immunity and Global Health, Murdoch Children’s Research Institute, Parkville, VIC, Australia
- Infectious Diseases Unit, The Royal Children’s Hospital, Melbourne, Parkville, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
| | - Kirsten P. Perrett
- Infectious Diseases Unit, The Royal Children’s Hospital, Melbourne, Parkville, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
- Population Health, Murdoch Children’s Research Institute, Parkville, VIC, Australia
| | - Nigel Curtis
- Infection, Immunity and Global Health, Murdoch Children’s Research Institute, Parkville, VIC, Australia
- Infectious Diseases Unit, The Royal Children’s Hospital, Melbourne, Parkville, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
| | - Nicole L. Messina
- Infection, Immunity and Global Health, Murdoch Children’s Research Institute, Parkville, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
8
|
Messina NL, Sperotto MG, Puga MAM, da Silva PV, de Oliveira RD, Moore CL, Pittet LF, Jamieson T, Dalcolmo M, dos Santos G, Jardim B, Lacerda MVG, Curtis N, Croda J. Impact of vaccine platform and BCG vaccination on antibody responses to COVID-19 vaccination. Front Immunol 2023; 14:1172851. [PMID: 37465688 PMCID: PMC10352084 DOI: 10.3389/fimmu.2023.1172851] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 06/12/2023] [Indexed: 07/20/2023] Open
Abstract
Multiple factors, including vaccine platform and prior vaccinations, influence vaccine responses. We compared antibody responses to CoronaVac (Sinovac) and ChAdOx1-S (AstraZeneca-Oxford) vaccination in 874 healthcare workers in Brazil. As participants were randomised to BCG vaccination or placebo in the preceding 0-6 months as part of the BCG vaccination to reduce the impact of COVID-19 in healthcare workers (BRACE) trial, we also investigated the influence of recent BCG vaccination on antibody responses to these COVID-19 vaccines. Twenty-eight days after the second dose of each vaccine, ChAdOx1-S induced a stronger anti-spike IgG response than CoronaVac vaccination. Recent BCG vaccination did not impact IgG antibody responses to ChAdOx1-S or CoronaVac.
Collapse
Affiliation(s)
- Nicole L. Messina
- Infectious Diseases Group, Infection and Immunity Theme, Murdoch Children’s Research Institute, Parkville, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
| | - Mariana G. Sperotto
- School of Medicine, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
- Oswaldo Cruz Foundation Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Marco A. M. Puga
- School of Medicine, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
- Oswaldo Cruz Foundation Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Patricia V. da Silva
- School of Medicine, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
- Oswaldo Cruz Foundation Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Roberto D. de Oliveira
- State University of Mato Grosso do Sul, Dourados-Mato Grosso do Sul, Brazil
- Federal University of Grande Dourados, Dourados, Brazil
| | - Cecilia L. Moore
- Clinical Epidemiology and Biostatistics Unit, Murdoch Children’s Research Institute, Parkville, VIC, Australia
| | - Laure F. Pittet
- Infectious Diseases Group, Infection and Immunity Theme, Murdoch Children’s Research Institute, Parkville, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
- Infectious Diseases, The Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
| | - Tenaya Jamieson
- Infectious Diseases Group, Infection and Immunity Theme, Murdoch Children’s Research Institute, Parkville, VIC, Australia
| | - Margareth Dalcolmo
- Helio Fraga Reference Center, Oswaldo Cruz Foundation Ministry of Health, Rio de Janeiro, Rio de Janeiro, Brazil
- Pontifical Catholic University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Glauce dos Santos
- National School of Public Health, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bruno Jardim
- Institute of Clinical Research Carlos Borborema, Fundação de Medicina Tropical Doutor Heitor Vieira Dourado, Manaus, Amazonas, Brazil
| | - Marcus V. G. Lacerda
- Institute of Clinical Research Carlos Borborema, Fundação de Medicina Tropical Doutor Heitor Vieira Dourado, Manaus, Amazonas, Brazil
| | - Nigel Curtis
- Infectious Diseases Group, Infection and Immunity Theme, Murdoch Children’s Research Institute, Parkville, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
- Infectious Diseases, The Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
| | - Julio Croda
- School of Medicine, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
- Oswaldo Cruz Foundation Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
- Yale School of Public Health, New Haven, CT, United States
| |
Collapse
|
9
|
Mambelli F, Marinho FV, Andrade JM, de Araujo ACVSC, Abuna RPF, Fabri VMR, Santos BPO, da Silva JS, de Magalhães MTQ, Homan EJ, Leite LCC, Dias GB, Heck N, Mendes DAGB, Mansur DS, Báfica A, Oliveira SC. Recombinant Bacillus Calmette-Guérin Expressing SARS-CoV-2 Chimeric Protein Protects K18-hACE2 Mice against Viral Challenge. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1925-1937. [PMID: 37098890 PMCID: PMC10247535 DOI: 10.4049/jimmunol.2200731] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 03/24/2023] [Indexed: 04/27/2023]
Abstract
COVID-19 has accounted for more than 6 million deaths worldwide. Bacillus Calmette-Guérin (BCG), the existing tuberculosis vaccine, is known to induce heterologous effects over other infections due to trained immunity and has been proposed to be a potential strategy against SARS-CoV-2 infection. In this report, we constructed a recombinant BCG (rBCG) expressing domains of the SARS-CoV-2 nucleocapsid and spike proteins (termed rBCG-ChD6), recognized as major candidates for vaccine development. We investigated whether rBCG-ChD6 immunization followed by a boost with the recombinant nucleocapsid and spike chimera (rChimera), together with alum, provided protection against SARS-CoV-2 infection in K18-hACE2 mice. A single dose of rBCG-ChD6 boosted with rChimera associated with alum elicited the highest anti-Chimera total IgG and IgG2c Ab titers with neutralizing activity against SARS-CoV-2 Wuhan strain when compared with control groups. Importantly, following SARS-CoV-2 challenge, this vaccination regimen induced IFN-γ and IL-6 production in spleen cells and reduced viral load in the lungs. In addition, no viable virus was detected in mice immunized with rBCG-ChD6 boosted with rChimera, which was associated with decreased lung pathology when compared with BCG WT-rChimera/alum or rChimera/alum control groups. Overall, our study demonstrates the potential of a prime-boost immunization system based on an rBCG expressing a chimeric protein derived from SARS-CoV-2 to protect mice against viral challenge.
Collapse
Affiliation(s)
- Fábio Mambelli
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Fábio V. Marinho
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Juvana M. Andrade
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ana C. V. S. C. de Araujo
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Rodrigo P. F. Abuna
- Platform of Bi-Institutional Research in Translational Medicine, Oswaldo Cruz Foundation-Fiocruz, Ribeirão Preto, São Paulo, Brazil
| | - Victor M. R. Fabri
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Bruno P. O. Santos
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - João S. da Silva
- Platform of Bi-Institutional Research in Translational Medicine, Oswaldo Cruz Foundation-Fiocruz, Ribeirão Preto, São Paulo, Brazil
| | - Mariana T. Q. de Magalhães
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - E. Jane Homan
- ioGenetics LLC, Madison, Wisconsin, United States of America
| | | | - Greicy B.M. Dias
- Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Nicoli Heck
- Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Daniel A. G. B. Mendes
- Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Daniel S. Mansur
- Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - André Báfica
- Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Sergio C. Oliveira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Department of Immunology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
10
|
Anwardeen NR, Cyprian FS, Yassine HM, Al-Thani AA, Abdallah AM, Emara MM, Elrayess MA. The retrospective study of the metabolic patterns of BCG-vaccination in type-2 diabetic individuals in COVID-19 infection. Front Immunol 2023; 14:1146443. [PMID: 37122708 PMCID: PMC10131282 DOI: 10.3389/fimmu.2023.1146443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 03/27/2023] [Indexed: 04/08/2023] Open
Abstract
BackgroundThe cross-protective nature of Bacillus Calmette-Guerin (BCG) vaccine against SARS-CoV-2 virus was previously suggested, however its effect in COVID-19 patients with type 2 diabetes (T2D) and the underlying metabolic pathways has not been addressed. This study aims to investigate the difference in the metabolomic patterns of type 2 diabetic patients with BCG vaccination showing different severity levels of COVID-19 infection.MethodsSixty-seven COVID-19 patients were categorized into diabetic and non-diabetic individuals who had been previously vaccinated or not with BCG vaccination. Targeted metabolomics were performed from serum samples from all patients using tandem mass spectrometry. Statistical analysis included multivariate and univariate models.ResultsData suggested that while BCG vaccination may provide protection for individuals who do not have diabetes, it appears to be linked to more severe COVID-19 symptoms in T2D patients (p = 0.02). Comparing the metabolic signature of BCG vaccinated T2D individuals to non-vaccinated counterparts revealed that amino acid (sarcosine), cholesterol esters (CE 20:0, 20:1, 22:2), carboxylic acid (Aconitic acid) were enriched in BCG vaccinated T2D patients, whereas spermidine, glycosylceramides (Hex3Cer(d18:1_22:0), Hex2Cer(d18:1/22:0), HexCer(d18:1/26:1), Hex2Cer(d18:1/24:0), HexCer(d18:1/22:0) were higher in BCG vaccinated non- T2D patients. Furthermore, data indicated a decrease in sarcosine synthesis from glycine and choline and increase in spermidine synthesis in the BCG vaccinated cohort in T2D and non-T2D groups, respectively.ConclusionThis pilot study suggests increased severity of COVID-19 in BCG vaccinated T2D patients, which was marked by decreased sarcosine synthesis, perhaps via lower sarcosine-mediated removal of viral antigens.
Collapse
Affiliation(s)
| | - Farhan S. Cyprian
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Hadi M. Yassine
- Biomedical Research Center (BRC), QU Health, Qatar University, Doha, Qatar
- College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - Asmaa A. Al-Thani
- Biomedical Research Center (BRC), QU Health, Qatar University, Doha, Qatar
- College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - Abdallah M. Abdallah
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Mohamed M. Emara
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Mohamed A. Elrayess
- Biomedical Research Center (BRC), QU Health, Qatar University, Doha, Qatar
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
- *Correspondence: Mohamed A. Elrayess,
| |
Collapse
|
11
|
Murdaca G, Paladin F, Mangini G, Tiso D, Gangemi S. TBC and COVID: an interplay between two infections. Expert Opin Drug Saf 2023; 22:303-311. [PMID: 37079022 DOI: 10.1080/14740338.2023.2205638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 04/18/2023] [Indexed: 04/21/2023]
Abstract
INTRODUCTION In a historical era dominated by the SARS-CoV-2 pandemic, a fact of growing interest emerges regarding co-infection with Mycobacterium tuberculosis (M. tuberculosis). This represents today an important clinical and diagnostic challenge, as the two pathogens are capable, through specific immunopathological mechanisms, of interacting with each other, determining a severe respiratory condition with a severe prognosis. AREAS COVERED With this review, we wanted to collect and analyze the latest scientific evidence concerning the main immunopathogenetic mechanisms shared by these two respiratory pathogens, with particular interest in the possible iatrogenic factors favoring coinfection and the need to define multidisciplinary and standardized screening tools aimed to identify coinfection early, ensuring the best clinical and therapeutic management. EXPERT OPINION The existence of a direct immunopathogenetic link between COVID-19 and TB indirectly contributes to mutual morbidity and mortality. The identification and application of early and standardized screening tools aimed at the identification of this condition is essential, in addition to vaccine prevention.
Collapse
Affiliation(s)
- Giuseppe Murdaca
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- Department of Internal Medicine (DIMI), Ospedale Policlinico San Martino IRCCS, Genoa, Italy
| | - Francesca Paladin
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- Department of Internal Medicine (DIMI), Ospedale Policlinico San Martino IRCCS, Genoa, Italy
| | - Gloria Mangini
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- Department of Internal Medicine (DIMI), Ospedale Policlinico San Martino IRCCS, Genoa, Italy
| | - Debora Tiso
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- Department of Internal Medicine (DIMI), Ospedale Policlinico San Martino IRCCS, Genoa, Italy
| | - Sebastiano Gangemi
- School and Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| |
Collapse
|
12
|
Antibody dependent disease enhancement (ADE) after COVID-19 vaccination and beta glucans as a safer strategy in management. Vaccine 2023; 41:2427-2429. [PMID: 36906407 PMCID: PMC9992059 DOI: 10.1016/j.vaccine.2023.03.005] [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: 06/05/2021] [Revised: 01/30/2023] [Accepted: 03/02/2023] [Indexed: 03/11/2023]
Abstract
A potential risk associated with vaccines for COVID-19 is antibody-dependent disease enhancement (ADE) in which vaccine induced antibody mediated immune responses may lead to enhanced SARS CoV- 2 acquisition or increased disease severity. Though ADE has not been clinically demonstrated with any of the COVID-19 vaccines so far, when neutralizing antibodies are suboptimal, the severity of COVID-19 has been reported to greater. ADE is presumed to occur via abnormal macrophages induced by the vaccine based immune response by antibody-mediated virus uptake into Fc gamma receptor IIa (FcγRIIa) or by the formation of Fc-mediated excessive antibody effector functions. Beta-glucans which are naturally occurring polysaccharides known for unique immunomodulation by capability to interact with macrophages, eliciting a specific beneficial immune-response and enhancing all arms of the immune system, importantly without over-activation are suggested as safer nutritional supplement-based vaccine adjuvants for COVID-19.
Collapse
|
13
|
Dutta D, Ghosh A, Dutta C, Sukla S, Biswas S. Cross-reactivity of SARS-CoV-2 with other pathogens, especially dengue virus: A historical perspective. J Med Virol 2023; 95:e28557. [PMID: 36755367 DOI: 10.1002/jmv.28557] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/20/2023] [Accepted: 02/04/2023] [Indexed: 02/10/2023]
Abstract
Dengue is a vector-borne viral disease caused by a Flavivirus whereas the COVID-19 pandemic was caused by a highly contagious virus, SARS-CoV-2 belonging to the family Coronaviridae. However, COVID-19 severity was observably less in dengue-endemic countries and vice versa especially during the active years of the pandemic (2019-2021). We observed that dengue virus (DENV) antibodies (Abs) could cross-react with SARS-CoV-2 spike antigen. This resulted in SARS-CoV-2 false positivity by rapid Ab test kits. DENV Abs binding to SARS-CoV-2 receptor-binding domain (and the reverse scenario), as revealed by docking studies further validated DENV and SARS-CoV-2 cross-reactivity. Finally, SARS-CoV-2 Abs were found to cross-neutralize DENV1 and DENV2 in virus neutralization test (VNT). Abs to other pathogens like Plasmodium were also cross-reactive but non-neutralizing for SARS-CoV-2. Here, we analyze the existing data on SARS-CoV-2 cross-reactivity with other pathogens, especially dengue to assess its impact on health (cross-protection?) and differential sero-diagnosis/surveillance.
Collapse
Affiliation(s)
- Debrupa Dutta
- National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, India
| | - Anisa Ghosh
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal, India
| | - Chiroshri Dutta
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal, India
| | - Soumi Sukla
- National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, India
| | - Subhajit Biswas
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
14
|
Gupta MM, Gilhotra R, Deopa D, Bhat AA, Thapa R, Singla N, Kulshrestha R, Gupta G. Epigenetics of Pulmonary Tuberculosis. TARGETING EPIGENETICS IN INFLAMMATORY LUNG DISEASES 2023:127-144. [DOI: 10.1007/978-981-99-4780-5_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
15
|
Rawat S, Gilhotra R, Singh SK, Bhat AA, Ojha A, Dhaundhiyal K, Dhramshaktu IS, Gupta G. Epigenetics of SARS-CoV2 (COVID-19). TARGETING EPIGENETICS IN INFLAMMATORY LUNG DISEASES 2023:199-208. [DOI: 10.1007/978-981-99-4780-5_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
16
|
Nobre JG, Delgadinho M, Silva C, Mendes J, Mateus V, Ribeiro E, Costa DA, Lopes M, Pedroso AI, Trigueiros F, Rodrigues MI, de Sousa CL, Brito M. Gut microbiota profile of COVID-19 patients: Prognosis and risk stratification (MicroCOVID-19 study). Front Microbiol 2022; 13:1035422. [PMID: 36483197 PMCID: PMC9723140 DOI: 10.3389/fmicb.2022.1035422] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 09/27/2022] [Indexed: 03/10/2024] Open
Abstract
Background Gut microbiota is intrinsically associated with the immune system and can promote or suppress infectious diseases, especially viral infections. This study aims to characterize and compare the microbiota profile of infected patients with SARS-CoV-2 (milder or severe symptoms), non-infected people, and recovered patients. This is a national, transversal, observational, multicenter, and case-control study that analyzed the microbiota of COVID-19 patients with mild or severe symptoms at home, at the hospital, or in the intensive care unit, patients already recovered, and healthy volunteers cohabiting with COVID-19 patients. DNA was isolated from stool samples and sequenced in a NGS platform. A demographic questionnaire was also applied. Statistical analysis was performed in SPSS. Results Firmicutes/Bacteroidetes ratios were found to be significantly lower in infected patients (1.61 and 2.57) compared to healthy volunteers (3.23) and recovered patients (3.89). Furthermore, the microbiota composition differed significantly between healthy volunteers, mild and severe COVID-19 patients, and recovered patients. Furthermore, Escherichia coli, Actinomyces naeslundii, and Dorea longicatena were shown to be more frequent in severe cases. The most common COVID-19 symptoms were linked to certain microbiome groups. Conclusion We can conclude that microbiota composition is significantly affected by SARS-CoV-2 infection and may be used to predict COVID-19 clinical evolution. Therefore, it will be possible to better allocate healthcare resources and better tackle future pandemics.
Collapse
Affiliation(s)
- José Guilherme Nobre
- Faculty of Medicine, Lisbon University, Lisbon, Portugal
- Faculdade de Medicina, Instituto de Saúde Ambiental, Universidade de Lisboa, Lisboa, Portugal
- PTSurg – Portuguese Surgical Research Collaborative, Lisbon, Portugal
| | - Mariana Delgadinho
- H&TRC- Health and Technology Research Center, ESTeSL- Escola Superior de Tecnologia da Saúde de Lisboa, Instituto Politécnico de Lisboa, Lisbon, Portugal
| | - Carina Silva
- H&TRC- Health and Technology Research Center, ESTeSL- Escola Superior de Tecnologia da Saúde de Lisboa, Instituto Politécnico de Lisboa, Lisbon, Portugal
- Centro de Estatística e Aplicações, Universidade de Lisboa, Lisbon, Portugal
| | - Joana Mendes
- H&TRC- Health and Technology Research Center, ESTeSL- Escola Superior de Tecnologia da Saúde de Lisboa, Instituto Politécnico de Lisboa, Lisbon, Portugal
| | - Vanessa Mateus
- H&TRC- Health and Technology Research Center, ESTeSL- Escola Superior de Tecnologia da Saúde de Lisboa, Instituto Politécnico de Lisboa, Lisbon, Portugal
| | - Edna Ribeiro
- H&TRC- Health and Technology Research Center, ESTeSL- Escola Superior de Tecnologia da Saúde de Lisboa, Instituto Politécnico de Lisboa, Lisbon, Portugal
| | - Diogo Alpuim Costa
- Breast Cancer Unit, CUF Oncologia, Lisbon, Portugal
- Faculdade de Ciências Médicas, NOVA Medical School, Lisbon, Portugal
| | - Miguel Lopes
- Departamento de Pneumologia, Hospital Garcia de Orta, Almada, Portugal
| | - Ana Isabel Pedroso
- Serviço de Medicina Intensiva, Hospital de Cascais Dr. José de Almeida, Cascais, Portugal
| | - Frederico Trigueiros
- Departamento de Medicina Interna I, Centro Hospitalar Lisboa Norte – Hospital de Santa Maria, Lisbon, Portugal
| | - Maria Inês Rodrigues
- Departamento de Medicina Interna I, Centro Hospitalar Lisboa Norte – Hospital de Santa Maria, Lisbon, Portugal
| | | | - Miguel Brito
- H&TRC- Health and Technology Research Center, ESTeSL- Escola Superior de Tecnologia da Saúde de Lisboa, Instituto Politécnico de Lisboa, Lisbon, Portugal
| |
Collapse
|
17
|
BCG Vaccine-associated Complications in a Large Cohort of Children With Combined Immunodeficiencies Affecting Cellular and Humoral Immunity. Pediatr Infect Dis J 2022; 41:933-937. [PMID: 36102730 DOI: 10.1097/inf.0000000000003678] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
AIMS To present the details of Bacillus Calmette-Guérin (BCG)-vaccine associated complications (VACs) in combined immunodeficiencies (CID) patients. METHODS Five centers participated in this retrospective study and completed a data form, which included general patients' information, clinical and laboratory data. RESULTS Among 236 CID patients, 127 were BCG vaccinated. 41.9% of patients with family history of CID and 17.1% who were diagnosed by screening were BCG vaccinated. Twenty-three patients (18.1%) developed BCG-VACs. The median age of VACs was 6 months and the median time from vaccination to complications was 6 months. The highest rate of BCG-VACs was recorded in patients receiving the Russian BCG strain compared to the Tokyo and Danish strains. Univariate analysis of T-lymphocyte subsets showed increased odds of BCG complications in patients with CD3+, CD4+, and CD8+ counts of ≤250 cells/µL. Only CD8 + count ≤250 cells/µL had increased such odds on multivariate analysis. VACs were disseminated in 13 and localized in 10 patients. Localized complication occurred earlier after vaccination (median: 4 months) compared with disseminated ones (median: 7 months). There were no significant associations between sex, administered vaccine strain, serum immunoglobulins levels, lymphocyte subsets counts, and the chance of having either localized or disseminated BCG-related complications. COCLUSIONS Although contraindicated, many patients with CID continue to be vaccinated with BCG. Low CD8 + count is a risk factor for BCG-related complications and localized complications occurred earlier than disseminated ones. Considerations should be undertaken by health care authorities especially in countries with high incidence of CID to implement newborn screening, delay the time of BCG vaccine administration beyond 6 months of age and to use the relatively safer strains like the Danish and Tokyo ones.
Collapse
|
18
|
Rakshit S, Adiga V, Ahmed A, Parthiban C, Chetan Kumar N, Dwarkanath P, Shivalingaiah S, Rao S, D’Souza G, Dias M, Maguire TJA, Doores KJ, Zoodsma M, Geckin B, Dasgupta P, Babji S, van Meijgaarden KE, Joosten SA, Ottenhoff THM, Li Y, Netea MG, Stuart KD, De Rosa SC, McElrath MJ, Vyakarnam A. Evidence for the heterologous benefits of prior BCG vaccination on COVISHIELD™ vaccine-induced immune responses in SARS-CoV-2 seronegative young Indian adults. Front Immunol 2022; 13:985938. [PMID: 36268023 PMCID: PMC9577398 DOI: 10.3389/fimmu.2022.985938] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/26/2022] [Indexed: 11/15/2022] Open
Abstract
This proof-of-concept study tested if prior BCG revaccination can qualitatively and quantitively enhance antibody and T-cell responses induced by Oxford/AstraZeneca ChAdOx1nCoV-19 or COVISHIELD™, an efficacious and the most widely distributed vaccine in India. We compared COVISHIELD™ induced longitudinal immune responses in 21 BCG re-vaccinees (BCG-RV) and 13 BCG-non-revaccinees (BCG-NRV), all of whom were BCG vaccinated at birth; latent tuberculosis negative and SARS-CoV-2 seronegative prior to COVISHIELD™ vaccination. Compared to BCG-NRV, BCG-RV displayed significantly higher and persistent spike-specific neutralizing (n) Ab titers and polyfunctional CD4+ and CD8+ T-cells for eight months post COVISHIELD™ booster, including distinct CD4+IFN-γ+ and CD4+IFN-γ- effector memory (EM) subsets co-expressing IL-2, TNF-α and activation induced markers (AIM) CD154/CD137 as well as CD8+IFN-γ+ EM,TEMRA (T cell EM expressing RA) subset combinations co-expressing TNF-α and AIM CD137/CD69. Additionally, elevated nAb and T-cell responses to the Delta mutant in BCG-RV highlighted greater immune response breadth. Mechanistically, these BCG adjuvant effects were associated with elevated markers of trained immunity, including higher IL-1β and TNF-α expression in CD14+HLA-DR+monocytes and changes in chromatin accessibility highlighting BCG-induced epigenetic changes. This study provides first in-depth analysis of both antibody and memory T-cell responses induced by COVISHIELD™ in SARS-CoV-2 seronegative young adults in India with strong evidence of a BCG-induced booster effect and therefore a rational basis to validate BCG, a low-cost and globally available vaccine, as an adjuvant to enhance heterologous adaptive immune responses to current and emerging COVID-19 vaccines.
Collapse
Affiliation(s)
- Srabanti Rakshit
- Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
- Infectious Disease Unit, St. John’s Research Institute, Bangalore, India
| | - Vasista Adiga
- Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
- Infectious Disease Unit, St. John’s Research Institute, Bangalore, India
- Department of Biotechnology, PES University, Bangalore, India
| | - Asma Ahmed
- Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
- Infectious Disease Unit, St. John’s Research Institute, Bangalore, India
| | - Chaitra Parthiban
- Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
- Infectious Disease Unit, St. John’s Research Institute, Bangalore, India
| | - Nirutha Chetan Kumar
- Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
- Infectious Disease Unit, St. John’s Research Institute, Bangalore, India
| | | | | | - Srishti Rao
- Infectious Disease Unit, St. John’s Research Institute, Bangalore, India
| | - George D’Souza
- Division of Nutrition, St. John’s Research Institute, Bangalore, India
| | - Mary Dias
- Infectious Disease Unit, St. John’s Research Institute, Bangalore, India
| | | | - Katie J. Doores
- Department of Pulmonary Medicine, St. John’s Medical College, Bangalore, India
| | - Martijn Zoodsma
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
- Department of Computational Biology for Individualized Infection Medicine, Centre for Individualized Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Busranur Geckin
- TWINCORE, a joint venture between the Helmholtz Centre for Infection Research, (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Prokar Dasgupta
- Department of Internal Medicine and Radboud Center for infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Sudhir Babji
- Peter Gorer Department of Immunobiology, Liver Renal Urology Transplant Gastro/Gastrointestinal Surgery, Inflammation Biology, King’s College London, London, United Kingdom
| | | | - Simone A. Joosten
- The Wellcome Trust Research Laboratory, Christian Medical College, Vellore, India
| | - Tom H. M. Ottenhoff
- The Wellcome Trust Research Laboratory, Christian Medical College, Vellore, India
| | - Yang Li
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
- Department of Computational Biology for Individualized Infection Medicine, Centre for Individualized Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Mihai G. Netea
- TWINCORE, a joint venture between the Helmholtz Centre for Infection Research, (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Kenneth D. Stuart
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Stephen C. De Rosa
- Centre for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States
| | - M. Juliana McElrath
- Centre for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Centre, Seattle, WA, United States
| | - Annapurna Vyakarnam
- Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
- Infectious Disease Unit, St. John’s Research Institute, Bangalore, India
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, United States
- *Correspondence: Annapurna Vyakarnam, ;
| |
Collapse
|
19
|
Jalalizadeh M, Buosi K, Dionato FAV, Dal Col LSB, Giacomelli CF, Ferrari KL, Pagliarone AC, Leme PAF, Maia CL, Yadollahvandmiandoab R, Trinh QD, Franchini KG, Bajgelman MC, Reis LO. Randomized clinical trial of BCG vaccine in patients with convalescent COVID-19: Clinical evolution, adverse events, and humoral immune response. J Intern Med 2022; 292:654-666. [PMID: 35599154 PMCID: PMC9347570 DOI: 10.1111/joim.13523] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND The Bacillus Calmette-Guérin (BCG) vaccine may confer cross-protection against viral diseases in adults. This study evaluated BCG vaccine cross-protection in adults with convalescent coronavirus disease 2019 (COVID-19). METHOD This was a multicenter, prospective, randomized, placebo-controlled, double-blind phase III study (ClinicalTrials.gov: NCT04369794). SETTING University Community Health Center and Municipal Outpatient Center in South America. PATIENTS a total of 378 adult patients with convalescent COVID-19 were included. INTERVENTION single intradermal BCG vaccine (n = 183) and placebo (n = 195). MEASUREMENTS the primary outcome was clinical evolution. Other outcomes included adverse events and humoral immune responses for up to 6 months. RESULTS A significantly higher proportion of BCG patients with anosmia and ageusia recovered at the 6-week follow-up visit than placebo (anosmia: 83.1% vs. 68.7% healed, p = 0.043, number needed to treat [NNT] = 6.9; ageusia: 81.2% vs. 63.4% healed, p = 0.032, NNT = 5.6). BCG also prevented the appearance of ageusia in the following weeks: seven in 113 (6.2%) BCG recipients versus 19 in 126 (15.1%) placebos, p = 0.036, NNT = 11.2. BCG did not induce any severe or systemic adverse effects. The most common and expected adverse effects were local vaccine lesions, erythema (n = 152; 86.4%), and papules (n = 111; 63.1%). Anti-severe acute respiratory syndrome coronavirus 2 humoral response measured by N protein immunoglobulin G titer and seroneutralization by interacting with the angiotensin-converting enzyme 2 receptor suggest that the serum of BCG-injected patients may neutralize the virus at lower specificity; however, the results were not statistically significant. CONCLUSION BCG vaccine is safe and offers cross-protection against COVID-19 with potential humoral response modulation. LIMITATIONS No severely ill patients were included.
Collapse
Affiliation(s)
- Mehrsa Jalalizadeh
- Department of UroScience, School of Medical Sciences, State University of Campinas-UNICAMP, Campinas, Brazil
| | - Keini Buosi
- Department of UroScience, School of Medical Sciences, State University of Campinas-UNICAMP, Campinas, Brazil
| | - Franciele A V Dionato
- Department of UroScience, School of Medical Sciences, State University of Campinas-UNICAMP, Campinas, Brazil
| | - Luciana S B Dal Col
- Department of UroScience, School of Medical Sciences, State University of Campinas-UNICAMP, Campinas, Brazil
| | - Cristiane F Giacomelli
- Department of UroScience, School of Medical Sciences, State University of Campinas-UNICAMP, Campinas, Brazil
| | - Karen L Ferrari
- Department of UroScience, School of Medical Sciences, State University of Campinas-UNICAMP, Campinas, Brazil
| | - Ana Carolina Pagliarone
- Department of UroScience, School of Medical Sciences, State University of Campinas-UNICAMP, Campinas, Brazil
| | - Patrícia A F Leme
- Department of UroScience, School of Medical Sciences, State University of Campinas-UNICAMP, Campinas, Brazil
| | - Cristiane L Maia
- Department of UroScience, School of Medical Sciences, State University of Campinas-UNICAMP, Campinas, Brazil
| | - Reza Yadollahvandmiandoab
- Department of UroScience, School of Medical Sciences, State University of Campinas-UNICAMP, Campinas, Brazil
| | - Quoc-Dien Trinh
- Brigham and Women's Center for Surgery and Public Health, Harvard Medical School, Boston, Massachusetts, USA
| | - Kleber G Franchini
- Brazilian Center for Research in Energy and Materials, CNPEM, Brazilian Biosciences National Laboratory-LNBio, Campinas, Brazil
| | - Marcio C Bajgelman
- Brazilian Center for Research in Energy and Materials, CNPEM, Brazilian Biosciences National Laboratory-LNBio, Campinas, Brazil
| | - Leonardo O Reis
- Department of UroScience, School of Medical Sciences, State University of Campinas-UNICAMP, Campinas, Brazil.,Center for Life Sciences, Pontifical Catholic University of Campinas, Campinas, Brazil
| |
Collapse
|
20
|
Kulesza J, Kulesza E, Koziński P, Karpik W, Broncel M, Fol M. BCG and SARS-CoV-2-What Have We Learned? Vaccines (Basel) 2022; 10:1641. [PMID: 36298506 PMCID: PMC9610589 DOI: 10.3390/vaccines10101641] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/22/2022] [Accepted: 09/24/2022] [Indexed: 11/19/2022] Open
Abstract
Despite controversy over the protective effect of the BCG (Bacille Calmette-Guérin) vaccine in preventing pulmonary tuberculosis (TB) in adults, it has been used worldwide since 1921. Although the first reports in the 1930s had noted a remarkable decrease in child mortality after BCG immunization, this could not be explained solely by a decrease in mortality from TB. These observations gave rise to the suggestion of nonspecific beneficial effects of BCG vaccination, beyond the desired protection against M. tuberculosis. The existence of an innate immunity-training mechanism based on epigenetic changes was demonstrated several years ago. The emergence of the pandemic caused by the severe acute respiratory syndrome coronavirus (SARS-CoV-2) in 2019 revived the debate about whether the BCG vaccine can affect the immune response against the virus or other unrelated pathogens. Due to the mortality of the coronavirus disease (COVID-19), it is important to verify each factor that may have a potential protective value against the severe course of COVID-19, complications, and death. This paper reviews the results of numerous retrospective studies and prospective trials which shed light on the potential of a century-old vaccine to mitigate the pandemic impact of the new virus. It should be noted, however, that although there are numerous studies intending to verify the hypothesis that the BCG vaccine may have a beneficial effect on COVID-19, there is no definitive evidence on the efficacy of the BCG vaccine against SARS-CoV-2.
Collapse
Affiliation(s)
- Jakub Kulesza
- Department of Internal Diseases and Clinical Pharmacology, Medical University of Lodz, Kniaziewicza 1/5, 91-347 Lodz, Poland
| | - Ewelina Kulesza
- Department of Rheumatology and Internal Diseases, Medical University of Lodz, Żeromskiego 113, 90-549 Lodz, Poland
| | - Piotr Koziński
- Tuberculosis and Lung Diseases Outpatient Clinic, Health Facility Unit in Łęczyca, Zachodnia 6, 99-100 Łęczyca, Poland
| | - Wojciech Karpik
- Department of Immunology and Infectious Biology, Faculty of Biology and Environmental Protection, University of Lodz, Banacha 12/16, 90-237 Lodz, Poland
| | - Marlena Broncel
- Department of Internal Diseases and Clinical Pharmacology, Medical University of Lodz, Kniaziewicza 1/5, 91-347 Lodz, Poland
| | - Marek Fol
- Department of Immunology and Infectious Biology, Faculty of Biology and Environmental Protection, University of Lodz, Banacha 12/16, 90-237 Lodz, Poland
| |
Collapse
|
21
|
Miteva D, Peshevska-Sekulovska M, Snegarova V, Batselova H, Alexandrova R, Velikova T. Mucosal COVID-19 vaccines: Risks, benefits and control of the pandemic. World J Virol 2022; 11:221-236. [PMID: 36188733 PMCID: PMC9523321 DOI: 10.5501/wjv.v11.i5.221] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 05/14/2022] [Accepted: 08/10/2022] [Indexed: 02/05/2023] Open
Abstract
Based on mucosal immunization to promote both mucosal and systemic immune responses, next-generation coronavirus disease 2019 (COVID-19) vaccines would be administered intranasally or orally. The goal of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines is to provide adequate immune protection and avoid severe disease and death. Mucosal vaccine candidates for COVID-19 including vector vaccines, recombinant subunit vaccines and live attenuated vaccines are under development. Furthermore, subunit protein vac-cines and virus-vectored vaccines have made substantial progress in preclinical and clinical settings, resulting in SARS-CoV-2 intranasal vaccines based on the previously successfully used nasal vaccines. Additional to their ability to trigger stable, protective immune responses at the sites of pathogenic infection, the development of 'specific' mucosal vaccines targeting coronavirus antigens could be an excellent option for preventing future pandemics. However, their efficacy and safety should be confirmed.
Collapse
Affiliation(s)
- Dimitrina Miteva
- Department of Genetics, Sofia University “St. Kliment Ohridski,” Faculty of Biology, Sofia 1164, Bulgaria
| | - Monika Peshevska-Sekulovska
- Department of Gastroenterology, University Hospital Lozenetz, Sofia 1407, Bulgaria
- Medical Faculty, Sofia University St. Kliment Ohridski, Sofia 1407, Bulgaria
| | - Violeta Snegarova
- Clinic of Internal Diseases, Naval Hospital - Varna, Military Medical Academy, Medical Faculty, Medical University, Varna 9000, Bulgaria
| | - Hristiana Batselova
- Department of Epidemiology and Disaster Medicine, Medical University, Plovdiv, University Hospital “St George”, Plovdiv 6000, Bulgaria
| | - Radostina Alexandrova
- Department of Pathology, Institute of Experimental Morphology, Pathology and Anthropology with Museum, Bulgarian Academy of Sciences, Sofia 1000, Bulgaria
| | - Tsvetelina Velikova
- Department of Clinical Immunology, University Hospital Lozenetz, Sofia 1407, Bulgaria
- Medical Faculty, Sofia University St. Kliment Ohridski, Sofia 1407, Bulgaria
| |
Collapse
|
22
|
Flores-Lovon K, Ortiz-Saavedra B, Cueva-Chicaña LA, Aperrigue-Lira S, Montes-Madariaga ES, Soriano-Moreno DR, Bell B, Macedo R. Immune responses in COVID-19 and tuberculosis coinfection: A scoping review. Front Immunol 2022; 13:992743. [PMID: 36090983 PMCID: PMC9459402 DOI: 10.3389/fimmu.2022.992743] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Background and aim Patients with COVID-19 and tuberculosis coinfection are at an increased risk of severe disease and death. We therefore sought to evaluate the current evidence which assessed the immune response in COVID-19 and tuberculosis coinfection. Methods We searched Pubmed/MEDLINE, EMBASE, Scopus, and Web of Science to identify articles published between 2020 and 2021. We included observational studies evaluating the immune response in patients with tuberculosis and COVID-19 compared to patients with COVID-19 alone. Results Four cross-sectional studies (372 participants) were identified. In patients with asymptomatic COVID-19 and latent tuberculosis (LTBI), increased cytokines, chemokines, growth factors and humoral responses were found. In addition, patients with symptomatic COVID-19 and LTBI had higher leukocytes counts and less inflammation. Regarding patients with COVID-19 and active tuberculosis (aTB), they exhibited decreased total lymphocyte counts, CD4 T cells specific against SARS-CoV-2 and responsiveness to SARS-CoV-2 antigens compared to patients with only COVID-19. Conclusion Although the evidence is limited, an apparent positive immunomodulation is observed in patients with COVID-19 and LTBI. On the other hand, patients with COVID-19 and aTB present a dysregulated immune response. Longitudinal studies are needed to confirm these findings and expand knowledge.
Collapse
Affiliation(s)
- Kevin Flores-Lovon
- Universidad Nacional de San Agustín de Arequipa, Arequipa, Peru
- Grupo de Investigación en Inmunología – GII, UNSA, Arequipa, Peru
| | - Brando Ortiz-Saavedra
- Universidad Nacional de San Agustín de Arequipa, Arequipa, Peru
- Grupo de Investigación en Inmunología – GII, UNSA, Arequipa, Peru
| | - Luis A. Cueva-Chicaña
- Universidad Nacional de San Agustín de Arequipa, Arequipa, Peru
- Grupo de Investigación en Inmunología – GII, UNSA, Arequipa, Peru
| | - Shalom Aperrigue-Lira
- Universidad Nacional de San Agustín de Arequipa, Arequipa, Peru
- Grupo de Investigación en Inmunología – GII, UNSA, Arequipa, Peru
| | - Elizbet S. Montes-Madariaga
- Universidad Nacional de San Agustín de Arequipa, Arequipa, Peru
- Grupo de Investigación en Inmunología – GII, UNSA, Arequipa, Peru
| | | | - Brett Bell
- Department of Radiation Oncology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Rodney Macedo
- Grupo de Investigación en Inmunología – GII, UNSA, Arequipa, Peru
- Department of Radiation Oncology, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
23
|
Cardoso Pinto AM, Ranasinghe L, Dodd PJ, Budhathoki SS, Seddon JA, Whittaker E. Disruptions to routine childhood vaccinations in low- and middle-income countries during the COVID-19 pandemic: A systematic review. Front Pediatr 2022; 10:979769. [PMID: 36034563 PMCID: PMC9403570 DOI: 10.3389/fped.2022.979769] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 07/22/2022] [Indexed: 11/16/2022] Open
Abstract
Background The COVID-19 pandemic has disrupted routine childhood vaccinations worldwide with low- and middle-income countries (LMICs) most affected. This study aims to quantify levels of disruption to routine vaccinations in LMICs. Methods A systematic review (PROSPERO CRD42021286386) was conducted of MEDLINE, Embase, Global Health, CINAHL, Scopus and MedRxiv, on the 11th of February 2022. Primary research studies published from January 2020 onwards were included if they reported levels of routine pediatrics vaccinations before and after March 2020. Study appraisal was performed using NHLBI tool for cross-sectional studies. Levels of disruption were summarized using medians and interquartile ranges. Results A total of 39 cross-sectional studies were identified. These showed an overall relative median decline of -10.8% [interquartile range (IQR) -27.6%, -1.4%] across all vaccines. Upper-middle-income countries (upper-MICs) (-14.3%; IQR -24.3%, -2.4%) and lower-MICs (-18.0%; IQR -48.6%, -4.1%) showed greater declines than low-income countries (-3.1%; IQR -12.8%, 2.9%), as did vaccines administered at birth (-11.8%; IQR -27.7%, -3.5%) compared to those given after birth (-8.0%; IQR -28.6%, -0.4%). Declines during the first 3 months of the pandemic (-8.1%; IQR -35.1%, -1.4%) were greater than during the remainder of 2020 (-3.9%; IQR -13.0%, 11.4%) compared to baseline. Conclusion There has been a decline in routine pediatric vaccination, greatest in MICs and for vaccines administered at birth. Nations must prioritize catch-up programs alongside public health messaging to encourage vaccine uptake. Systematic review registration Identifier: CRD42021286386.
Collapse
Affiliation(s)
| | - Lasith Ranasinghe
- Academic Foundation Doctor, Imperial College London, London, United Kingdom
| | - Peter J. Dodd
- School of Health and Related Research, University of Sheffield, Sheffield, United Kingdom
| | - Shyam Sundar Budhathoki
- Department of Primary Care and Public Health, School of Public Health, Imperial College London, London, United Kingdom
| | - James A. Seddon
- Department of Infectious Disease, Imperial College London, London, United Kingdom
- Desmond Tutu TB Centre, Department of Paediatrics and Child Health, Stellenbosch University, Cape Town, South Africa
| | - Elizabeth Whittaker
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| |
Collapse
|
24
|
Negi K, Bhaskar A, Dwivedi VP. Progressive Host-Directed Strategies to Potentiate BCG Vaccination Against Tuberculosis. Front Immunol 2022; 13:944183. [PMID: 35967410 PMCID: PMC9365942 DOI: 10.3389/fimmu.2022.944183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 06/21/2022] [Indexed: 11/13/2022] Open
Abstract
The pursuit to improve the TB control program comprising one approved vaccine, M. bovis Bacille Calmette-Guerin (BCG) has directed researchers to explore progressive approaches to halt the eternal TB pandemic. Mycobacterium tuberculosis (M.tb) was first identified as the causative agent of TB in 1882 by Dr. Robert Koch. However, TB has plagued living beings since ancient times and continues to endure as an eternal scourge ravaging even with existing chemoprophylaxis and preventive therapy. We have scientifically come a long way since then, but despite accessibility to the standard antimycobacterial antibiotics and prophylactic vaccine, almost one-fourth of humankind is infected latently with M.tb. Existing therapeutics fail to control TB, due to the upsurge of drug-resistant strains and increasing incidents of co-infections in immune-compromised individuals. Unresponsiveness to established antibiotics leaves patients with no therapeutic possibilities. Hence the search for an efficacious TB immunization strategy is a global health priority. Researchers are paving the course for efficient vaccination strategies with the radically advanced operation of core principles of protective immune responses against M.tb. In this review; we have reassessed the progression of the TB vaccination program comprising BCG immunization in children and potential stratagems to reinforce BCG-induced protection in adults.
Collapse
Affiliation(s)
| | | | - Ved Prakash Dwivedi
- Immunobiology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| |
Collapse
|
25
|
Dheda K, Perumal T, Moultrie H, Perumal R, Esmail A, Scott AJ, Udwadia Z, Chang KC, Peter J, Pooran A, von Delft A, von Delft D, Martinson N, Loveday M, Charalambous S, Kachingwe E, Jassat W, Cohen C, Tempia S, Fennelly K, Pai M. The intersecting pandemics of tuberculosis and COVID-19: population-level and patient-level impact, clinical presentation, and corrective interventions. THE LANCET. RESPIRATORY MEDICINE 2022; 10:603-622. [PMID: 35338841 PMCID: PMC8942481 DOI: 10.1016/s2213-2600(22)00092-3] [Citation(s) in RCA: 142] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 02/17/2022] [Accepted: 03/03/2022] [Indexed: 01/19/2023]
Abstract
The global tuberculosis burden remains substantial, with more than 10 million people newly ill per year. Nevertheless, tuberculosis incidence has slowly declined over the past decade, and mortality has decreased by almost a third in tandem. This positive trend was abruptly reversed by the COVID-19 pandemic, which in many parts of the world has resulted in a substantial reduction in tuberculosis testing and case notifications, with an associated increase in mortality, taking global tuberculosis control back by roughly 10 years. Here, we consider points of intersection between the tuberculosis and COVID-19 pandemics, identifying wide-ranging approaches that could be taken to reverse the devastating effects of COVID-19 on tuberculosis control. We review the impact of COVID-19 at the population level on tuberculosis case detection, morbidity and mortality, and the patient-level impact, including susceptibility to disease, clinical presentation, diagnosis, management, and prognosis. We propose strategies to reverse or mitigate the deleterious effects of COVID-19 and restore tuberculosis services. Finally, we highlight research priorities and major challenges and controversies that need to be addressed to restore and advance the global response to tuberculosis.
Collapse
Affiliation(s)
- Keertan Dheda
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine and University of Cape Town Lung Institute, University of Cape Town, Cape Town, South Africa; South African Medical Research Council (SAMRC) Centre for the Study of Antimicrobial Resistance, University of Cape Town, Cape Town, South Africa; Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK.
| | - Tahlia Perumal
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine and University of Cape Town Lung Institute, University of Cape Town, Cape Town, South Africa; South African Medical Research Council (SAMRC) Centre for the Study of Antimicrobial Resistance, University of Cape Town, Cape Town, South Africa
| | - Harry Moultrie
- Centre for TB, National Institute for Communicable Diseases, Division of the National Health Laboratory Services, Johannesburg, South Africa; School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Rubeshan Perumal
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine and University of Cape Town Lung Institute, University of Cape Town, Cape Town, South Africa; SAMRC-CAPRISA HIV-TB Pathogenesis and Treatment Research Unit, Durban, South Africa
| | - Aliasgar Esmail
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine and University of Cape Town Lung Institute, University of Cape Town, Cape Town, South Africa; South African Medical Research Council (SAMRC) Centre for the Study of Antimicrobial Resistance, University of Cape Town, Cape Town, South Africa
| | - Alex J Scott
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine and University of Cape Town Lung Institute, University of Cape Town, Cape Town, South Africa; South African Medical Research Council (SAMRC) Centre for the Study of Antimicrobial Resistance, University of Cape Town, Cape Town, South Africa
| | - Zarir Udwadia
- Department of Pulmonology, P D Hinduja Hospital and Medical Research Centre, Mumbai, India
| | - Kwok Chiu Chang
- Tuberculosis and Chest Service, Department of Health, Hong Kong Special Administrative Region, China
| | - Jonathan Peter
- Allergy and Immunology unit, Division of Allergy and Clinical Immunology, University of Cape Town Lung Institute, University of Cape Town, Cape Town, South Africa
| | - Anil Pooran
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine and University of Cape Town Lung Institute, University of Cape Town, Cape Town, South Africa; South African Medical Research Council (SAMRC) Centre for the Study of Antimicrobial Resistance, University of Cape Town, Cape Town, South Africa
| | - Arne von Delft
- School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa; TB Proof, Cape Town, South Africa
| | | | - Neil Martinson
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa; Johns Hopkins University Center for TB Research, Baltimore, MD, USA
| | - Marian Loveday
- HIV Prevention Research Unit, South African Medical Research Council, Durban, South Africa
| | - Salome Charalambous
- School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa; The Aurum Institute, Johannesburg, South Africa
| | - Elizabeth Kachingwe
- Centre for TB, National Institute for Communicable Diseases, Division of the National Health Laboratory Services, Johannesburg, South Africa
| | - Waasila Jassat
- Division of Public Health Surveillance and Response, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
| | - Cheryl Cohen
- School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa; Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
| | - Stefano Tempia
- School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa; Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
| | - Kevin Fennelly
- Pulmonary Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Madhukar Pai
- McGill International TB Centre, McGill University, Montreal, QC, Canada
| |
Collapse
|
26
|
Raqib R, Sarker P, Akhtar E, Nurul Huda TM, Haq MA, Roy AK, Hosen MB, Haque F, Chowdhury MR, Reidpath DD, Emdadul Hoque DM, Islam Z, Ahmed S, Ahmed T, Tofail F, Razzaque A. Seroprevalence of SARS-CoV-2 infection and associated factors among Bangladeshi slum and non-slum dwellers in pre-COVID-19 vaccination era: October 2020 to February 2021. PLoS One 2022; 17:e0268093. [PMID: 35604947 PMCID: PMC9126397 DOI: 10.1371/journal.pone.0268093] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 04/21/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Seroprevalence studies have been carried out in many developed and developing countries to evaluate ongoing and past infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Data on this infection in marginalized populations in urban slums are limited, which may offer crucial information to update prevention and mitigation policies and strategies. We aimed to determine the seroprevalence of SARS-CoV-2 infection and factors associated with seropositivity in slum and non-slum communities in two large cities in Bangladesh. METHODS A cross-sectional study was carried out among the target population in Dhaka and Chattogram cities between October 2020 and February 2021. Questionnaire-based data, anthropometric and blood pressure measurements and blood were obtained. SARS-CoV-2 serology was assessed by Roche Elecsys® Anti-SARS-CoV-2 immunoassay. RESULTS Among the 3220 participants (2444 adults, ≥18 years; 776 children, 10-17 years), the overall weighted seroprevalence was 67.3% (95% confidence intervals (CI) = 65.2, 69.3) with 71.0% in slum (95% CI = 68.7, 72.2) and 62.2% in non-slum (95% CI = 58.5, 65.8). The weighted seroprevalence was 72.9% in Dhaka and 54.2% in Chattogram. Seroprevalence was positively associated with limited years of formal education (adjusted odds ratio [aOR] = 1.61; 95% CI = 1.43, 1.82), lower income (aOR = 1.23; 95% CI = 1.03, 1.46), overweight (aOR = 1.2835; 95% CI = 1.26, 1.97), diabetes (aOR = 1.67; 95% CI = 1.21, 2.32) and heart disease (aOR = 1.38; 95% CI = 1.03, 1.86). Contrarily, negative associations were found between seropositivity and regular wearing of masks and washing hands, and prior BCG vaccination. About 63% of the population had asymptomatic infection; only 33% slum and 49% non-slum population showed symptomatic infection. CONCLUSION The estimated seroprevalence of SARS-CoV-2 was more prominent in impoverished informal settlements than in the adjacent middle-income non-slum areas. Additional factors associated with seropositivity included limited education, low income, overweight and pre-existing chronic conditions. Behavioral factors such as regular wearing of masks and washing hands were associated with lower probability of seropositivity.
Collapse
Affiliation(s)
- Rubhana Raqib
- Infectious Diseases Division, icddrb, Dhaka, Bangladesh
| | - Protim Sarker
- Infectious Diseases Division, icddrb, Dhaka, Bangladesh
| | - Evana Akhtar
- Infectious Diseases Division, icddrb, Dhaka, Bangladesh
| | | | | | | | | | - Farjana Haque
- Infectious Diseases Division, icddrb, Dhaka, Bangladesh
| | | | - Daniel D. Reidpath
- Health Systems and Population Studies Division, icddrb, Dhaka, Bangladesh
| | | | | | - Shehlina Ahmed
- Foreign, Commonwealth & Development Office (FCDO), Dhaka, Bangladesh
| | - Tahmeed Ahmed
- Nutrition and Clinical Services Division, icddrb, Dhaka, Bangladesh
| | - Fahmida Tofail
- Nutrition and Clinical Services Division, icddrb, Dhaka, Bangladesh
| | - Abdur Razzaque
- Health Systems and Population Studies Division, icddrb, Dhaka, Bangladesh
| |
Collapse
|
27
|
Functional in-vitro evaluation of the non-specific effects of BCG vaccination in a randomised controlled clinical study. Sci Rep 2022; 12:7808. [PMID: 35552463 PMCID: PMC9096342 DOI: 10.1038/s41598-022-11748-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 04/19/2022] [Indexed: 11/09/2022] Open
Abstract
Bacille Calmette-Guérin (BCG), the only currently licenced tuberculosis vaccine, may exert beneficial non-specific effects (NSE) in reducing infant mortality. We conducted a randomised controlled clinical study in healthy UK adults to evaluate potential NSE using functional in-vitro growth inhibition assays (GIAs) as a surrogate of protection from four bacteria implicated in infant mortality. Volunteers were randomised to receive BCG intradermally (n = 27) or to be unvaccinated (n = 8) and were followed up for 84 days; laboratory staff were blinded until completion of the final visit. Using GIAs based on peripheral blood mononuclear cells, we observed a significant reduction in the growth of the Gram-negative bacteria Escherichia coli and Klebsiella pneumonia following BCG vaccination, but no effect for the Gram-positive bacteria Staphylococcus aureus and Streptococcus agalactiae. There was a modest association between S. aureus nasal carriage and growth of S. aureus in the GIA. Our findings support a causal link between BCG vaccination and improved ability to control growth of heterologous bacteria. Unbiased assays such as GIAs are potentially useful tools for the assessment of non-specific as well as specific effects of TB vaccines. This study was funded by the Bill and Melinda Gates Foundation and registered with ClinicalTrials.gov (NCT02380508, 05/03/2015; completed).
Collapse
|
28
|
Trained immunity-related vaccines: innate immune memory and heterologous protection against infections. Trends Mol Med 2022; 28:497-512. [DOI: 10.1016/j.molmed.2022.03.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/25/2022] [Accepted: 03/25/2022] [Indexed: 11/21/2022]
|
29
|
Mohta A, Kumar Sharma M, Chand Ghiya B, Dutt Mehta R. Clearance of Recalcitrant Verruca Plana following COVID-19 Vaccination. JAAD Int 2022; 8:1-2. [PMID: 35308145 PMCID: PMC8923995 DOI: 10.1016/j.jdin.2022.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Alpana Mohta
- senior resident, Department of Dermatology, Venereology and Leprology, Sardar Patel Medical College, Bikaner , Rajasthan
| | | | - Bhikam Chand Ghiya
- professor and head, Department of Dermatology, Venereology and Leprology, Sardar Patel Medical College, Bikaner , Rajasthan
| | - Rajesh Dutt Mehta
- senior professor, Department of Dermatology, Venereology and Leprology, Sardar Patel Medical College, Bikaner , Rajasthan.,Department(s) and institution(s): Dermatology, venereology and leprosy. Sardar Patel Medical College, Bikaner
| |
Collapse
|
30
|
White AD, Sibley L, Sarfas C, Morrison AL, Bewley K, Churchward C, Fotheringham S, Gkolfinos K, Gooch K, Handley A, Humphries HE, Hunter L, Kennard C, Longet S, Mabbutt A, Moffatt M, Rayner E, Tipton T, Watson R, Hall Y, Bodman-Smith M, Gleeson F, Dennis M, Salguero FJ, Carroll M, McShane H, Cookson W, Hopkin J, Sharpe S. Influence of Aerosol Delivered BCG Vaccination on Immunological and Disease Parameters Following SARS-CoV-2 Challenge in Rhesus Macaques. Front Immunol 2022; 12:801799. [PMID: 35222355 PMCID: PMC8863871 DOI: 10.3389/fimmu.2021.801799] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/24/2021] [Indexed: 12/19/2022] Open
Abstract
The tuberculosis vaccine, Bacille Calmette-Guerin (BCG), also affords protection against non-tuberculous diseases attributable to heterologous immune mechanisms such as trained innate immunity, activation of non-conventional T-cells, and cross-reactive adaptive immunity. Aerosol vaccine delivery can target immune responses toward the primary site of infection for a respiratory pathogen. Therefore, we hypothesised that aerosol delivery of BCG would enhance cross-protective action against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection and be a deployable intervention against coronavirus disease 2019 (COVID-19). Immune parameters were monitored in vaccinated and unvaccinated rhesus macaques for 28 days following aerosol BCG vaccination. High-dose SARS-CoV-2 challenge was applied by intranasal and intrabronchial instillation and animals culled 6–8 days later for assessment of viral, disease, and immunological parameters. Mycobacteria-specific cell-mediated immune responses were detected following aerosol BCG vaccination, but SARS-CoV-2-specific cellular- and antibody-mediated immunity was only measured following challenge. Early secretion of cytokine and chemokine markers associated with the innate cellular and adaptive antiviral immune response was detected following SARS-CoV-2 challenge in vaccinated animals, at concentrations that exceeded titres measured in unvaccinated macaques. Classical CD14+ monocytes and Vδ2 γδ T-cells quantified by whole-blood immunophenotyping increased rapidly in vaccinated animals following SARS-CoV-2 challenge, indicating a priming of innate immune cells and non-conventional T-cell populations. However, viral RNA quantified in nasal and pharyngeal swabs, bronchoalveolar lavage (BAL), and tissue samples collected at necropsy was equivalent in vaccinated and unvaccinated animals, and in-life CT imaging and histopathology scoring applied to pulmonary tissue sections indicated that the disease induced by SARS-CoV-2 challenge was comparable between vaccinated and unvaccinated groups. Hence, aerosol BCG vaccination did not induce, or enhance the induction of, SARS-CoV-2 cross-reactive adaptive cellular or humoral immunity, although an influence of BCG vaccination on the subsequent immune response to SARS-CoV-2 challenge was apparent in immune signatures indicative of trained innate immune mechanisms and primed unconventional T-cell populations. Nevertheless, aerosol BCG vaccination did not enhance the initial clearance of virus, nor reduce the occurrence of early disease pathology after high dose SARS-CoV-2 challenge. However, the heterologous immune mechanisms primed by BCG vaccination could contribute to the moderation of COVID-19 disease severity in more susceptible species following natural infection.
Collapse
Affiliation(s)
- Andrew D White
- Research and Evaluation, United Kingdom Health Security Agency, Salisbury, United Kingdom
| | - Laura Sibley
- Research and Evaluation, United Kingdom Health Security Agency, Salisbury, United Kingdom
| | - Charlotte Sarfas
- Research and Evaluation, United Kingdom Health Security Agency, Salisbury, United Kingdom
| | - Alexandra L Morrison
- Research and Evaluation, United Kingdom Health Security Agency, Salisbury, United Kingdom
| | - Kevin Bewley
- Research and Evaluation, United Kingdom Health Security Agency, Salisbury, United Kingdom
| | - Colin Churchward
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Susan Fotheringham
- Research and Evaluation, United Kingdom Health Security Agency, Salisbury, United Kingdom
| | - Konstantinos Gkolfinos
- Research and Evaluation, United Kingdom Health Security Agency, Salisbury, United Kingdom
| | - Karen Gooch
- Research and Evaluation, United Kingdom Health Security Agency, Salisbury, United Kingdom
| | - Alastair Handley
- Research and Evaluation, United Kingdom Health Security Agency, Salisbury, United Kingdom
| | - Holly E Humphries
- Research and Evaluation, United Kingdom Health Security Agency, Salisbury, United Kingdom
| | - Laura Hunter
- Research and Evaluation, United Kingdom Health Security Agency, Salisbury, United Kingdom
| | - Chelsea Kennard
- Research and Evaluation, United Kingdom Health Security Agency, Salisbury, United Kingdom
| | - Stephanie Longet
- Research and Evaluation, United Kingdom Health Security Agency, Salisbury, United Kingdom
| | - Adam Mabbutt
- Research and Evaluation, United Kingdom Health Security Agency, Salisbury, United Kingdom
| | - Miriam Moffatt
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Emma Rayner
- Research and Evaluation, United Kingdom Health Security Agency, Salisbury, United Kingdom
| | - Tom Tipton
- Research and Evaluation, United Kingdom Health Security Agency, Salisbury, United Kingdom
| | - Robert Watson
- Research and Evaluation, United Kingdom Health Security Agency, Salisbury, United Kingdom
| | - Yper Hall
- Research and Evaluation, United Kingdom Health Security Agency, Salisbury, United Kingdom
| | - Mark Bodman-Smith
- Infection and Immunity Research Institute, St George's University of London, London, United Kingdom
| | - Fergus Gleeson
- Department of Oncology, Churchill Hospital, Oxford, United Kingdom
| | - Mike Dennis
- Research and Evaluation, United Kingdom Health Security Agency, Salisbury, United Kingdom
| | - Francisco J Salguero
- Research and Evaluation, United Kingdom Health Security Agency, Salisbury, United Kingdom
| | - Miles Carroll
- Research and Evaluation, United Kingdom Health Security Agency, Salisbury, United Kingdom
| | - Helen McShane
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - William Cookson
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Julian Hopkin
- College of Medicine, Institute of Life Science, Swansea University, Swansea, United Kingdom
| | - Sally Sharpe
- Research and Evaluation, United Kingdom Health Security Agency, Salisbury, United Kingdom
| |
Collapse
|
31
|
Hu Z, Lu S, Lowrie DB, Fan X. Trained immunity: A Yin-Yang balance. MedComm (Beijing) 2022; 3:e121. [PMID: 35281787 PMCID: PMC8906449 DOI: 10.1002/mco2.121] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/16/2022] [Accepted: 02/18/2022] [Indexed: 12/17/2022] Open
Abstract
Traditionally, immune memory is regarded as an exclusive hallmark of adaptive immunity. However, a growing body of evidence suggesting that innate immune cells show adaptive characteristics has challenged this dogma. In the past decade, trained immunity, a de facto innate immune memory, has been defined as a long-term functional reprogramming of cells of the innate immune system: the reprogramming is evoked by endogenous or exogenous insults, the cells return to a nonactivated state and subsequently show altered inflammatory responses against a second challenge. Trained immunity became regarded as a mechanism selected in evolution to protect against infection; however, a maladaptive effect might result in hyperinflammation. This dual effect is consistent with the Yin-Yang theory in traditional Chinese philosophy, in which Yang represents active, positive, and aggressive factors, whereas Yin represents passive, negative, and inhibitory factors. In this review, we give a brief overview of history and latest progress about trained immunity, including experimental models, inductors, molecular mechanisms, clinical application and so on. Moreover, this is the first time to put forward the theory of Yin-Yang balance to understand trained immunity. We envision that more efforts will be focused on developing novel immunotherapies targeting trained immunity in the coming years.
Collapse
Affiliation(s)
- Zhidong Hu
- Shanghai Public Health Clinical CenterKey Laboratory of Medical Molecular Virology of MOE/MOHFudan UniversityShanghaiChina
| | - Shui‐Hua Lu
- Shanghai Public Health Clinical CenterKey Laboratory of Medical Molecular Virology of MOE/MOHFudan UniversityShanghaiChina
- National Medical Center for Infectious Diseases of ChinaShenzhen Third People Hospital, South Science & Technology UniversityShenzhenChina
| | - Douglas B. Lowrie
- National Medical Center for Infectious Diseases of ChinaShenzhen Third People Hospital, South Science & Technology UniversityShenzhenChina
| | - Xiao‐Yong Fan
- Shanghai Public Health Clinical CenterKey Laboratory of Medical Molecular Virology of MOE/MOHFudan UniversityShanghaiChina
| |
Collapse
|
32
|
Dass SA, Balakrishnan V, Arifin N, Lim CSY, Nordin F, Tye GJ. The COVID-19/Tuberculosis Syndemic and Potential Antibody Therapy for TB Based on the Lessons Learnt From the Pandemic. Front Immunol 2022; 13:833715. [PMID: 35242137 PMCID: PMC8886238 DOI: 10.3389/fimmu.2022.833715] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 01/25/2022] [Indexed: 12/19/2022] Open
Abstract
2020 will be marked in history for the dreadful implications of the COVID-19 pandemic that shook the world globally. The pandemic has reshaped the normality of life and affected mankind in the aspects of mental and physical health, financial, economy, growth, and development. The focus shift to COVID-19 has indirectly impacted an existing air-borne disease, Tuberculosis. In addition to the decrease in TB diagnosis, the emergence of the TB/COVID-19 syndemic and its serious implications (possible reactivation of latent TB post-COVID-19, aggravation of an existing active TB condition, or escalation of the severity of a COVID-19 during TB-COVID-19 coinfection), serve as primary reasons to equally prioritize TB. On a different note, the valuable lessons learnt for the COVID-19 pandemic provide useful knowledge for enhancing TB diagnostics and therapeutics. In this review, the crucial need to focus on TB amid the COVID-19 pandemic has been discussed. Besides, a general comparison between COVID-19 and TB in the aspects of pathogenesis, diagnostics, symptoms, and treatment options with importance given to antibody therapy were presented. Lastly, the lessons learnt from the COVID-19 pandemic and how it is applicable to enhance the antibody-based immunotherapy for TB have been presented.
Collapse
Affiliation(s)
- Sylvia Annabel Dass
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Minden, Malaysia
| | - Venugopal Balakrishnan
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Minden, Malaysia
| | - Norsyahida Arifin
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Minden, Malaysia
| | - Crystale Siew Ying Lim
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Fazlina Nordin
- Tissue Engineering Centre (TEC), Universiti Kebangsaan Malaysia Medical Centre (UKMMC), Kuala Lumpur, Malaysia
| | - Gee Jun Tye
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Minden, Malaysia
| |
Collapse
|
33
|
Contopoulos-Ioannidis DG, Altamirano J, Maldonado Y. Infectious Diseases-Related Hospitalizations During Oral Polio Vaccine(OPV) and non-OPV immunization periods: An Empirical Evaluation of all Hospital Discharges in California(1985-2010). Clin Infect Dis 2022; 75:1123-1130. [PMID: 35139187 DOI: 10.1093/cid/ciac114] [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: 08/16/2021] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Live attenuated vaccines such as oral polio vaccine (OPV) can stimulate innate immunity and may have off-target protective effects on other pathogens. We aimed to address this hypothesis by examining changes in infectious diseases (ID)-related hospitalizations in all hospital discharges in California during OPV-(1985-1996) and non-OPV-immunization periods (2000-2010). METHODS We searched the OSHPD (Office of Statewide Health Planning and Development) database for all hospital discharges with any ID-related discharge diagnosis code during 1985-2010. We compared the proportion of ID-related hospitalizations (with at least one ID-related discharge diagnosis) among total hospitalizations during OPV immunization (1985-1996) vs non-OPV immunization (2000-2010) periods. RESULTS There were 19,281,039 ID-related hospitalizations (8,464,037 with an ID-related discharge-diagnosis as the principal discharge diagnosis for the hospitalization) among 98,117,475 hospitalizations in 1985-2010; 9,520,810 ID-hospitalizations/43,456,484 total hospitalizations in 2000-2010 vs 7,526,957/43,472,796 in 1985-1996. The RR for ID-related hospitalizations in 2000-2010 vs 1985-1996 was 1.27(95% CI: 1.26-1.27) for all diagnoses and 1.15(95% CI: 1.15-1.16) for principal diagnoses. Increases also existed in the proportion of lower respiratory and gastrointestinal infections. DISCUSSION The proportion of ID-related hospitalizations was lower in the OPV-immunization period compared to the period after OPV was discontinued. When focused only on hospitalizations with ID as the principal discharge diagnosis the signal remained significant but was smaller. These findings require replication in additional studies.
Collapse
Affiliation(s)
- Despina G Contopoulos-Ioannidis
- Division of Infectious Diseases, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Jonathan Altamirano
- Division of Infectious Diseases, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States of America.,Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Yvonne Maldonado
- Division of Infectious Diseases, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States of America.,Senior Associate Dean for Faculty Development and Diversity, Stanford University, Stanford, CA, United States of America.,Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, United States of America
| |
Collapse
|
34
|
Zhang B, Moorlag SJ, Domínguez-Andrés J, Bulut Ö, Kilic G, Liu Z, van Crevel R, Xu CJ, Joosten LA, Netea MG, Li Y. Single-cell RNA sequencing reveals induction of distinct trained immunity programs in human monocytes. J Clin Invest 2022; 132:147719. [PMID: 35133977 PMCID: PMC8970681 DOI: 10.1172/jci147719] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 02/02/2022] [Indexed: 11/17/2022] Open
Abstract
Trained immunity refers to the long-lasting memory traits of innate immunity. Recent studies have shown that trained immunity is orchestrated by sustained changes in epigenetic marks and metabolic pathways, leading to an altered transcriptional response towards a second challenge. However, the potential heterogeneity of trained immunity induction in innate immune cells has not been explored. In this study, we demonstrate cellular transcriptional programs to four different inducers of trained immunity in monocyte populations at single-cell resolution. Specifically, we identified three monocyte subpopulations upon the induction of trained immunity, and replicated these findings in an in vivo study. In addition, we found gene signatures consistent with these functional programs in ulcerative colitis, sepsis and COVID-19 patients, suggesting the impact of trained immunity programs in immune-mediated diseases.
Collapse
Affiliation(s)
- Bowen Zhang
- Department of Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine (CiiM), Hannover, Germany
| | - Simone Jcfm Moorlag
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Jorge Domínguez-Andrés
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Özlem Bulut
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Gizem Kilic
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Zhaoli Liu
- Centre for Experimental and Clinical Infection Research, Helmholtz-Centre for Infection Research (HZI), Hannover, Germany
| | - Reinout van Crevel
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Cheng-Jian Xu
- Department of Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine (CiiM), Hannover, Germany
| | - Leo Ab Joosten
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Yang Li
- Department of Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine (CiiM), Hannover, Germany
| |
Collapse
|
35
|
Soto JA, Gálvez NMS, Andrade CA, Ramírez MA, Riedel CA, Kalergis AM, Bueno SM. BCG vaccination induces cross-protective immunity against pathogenic microorganisms. Trends Immunol 2022; 43:322-335. [PMID: 35074254 DOI: 10.1016/j.it.2021.12.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/04/2021] [Accepted: 12/06/2021] [Indexed: 01/03/2023]
Abstract
Bacillus Calmette-Guérin (BCG) is an attenuated Mycobacterium bovis strain used as a vaccine to prevent Mycobacterium tuberculosis (M. tb) infection. Its ability to potentiate the immune response induced by other vaccines and to promote nonspecific immunomodulatory effects has been described. These effects can be triggered by epigenetic reprogramming and metabolic shifts on innate immune cells, a phenomenon known as trained immunity. The induction of trained immunity may contribute to explain why BCG vaccination effectively decreases disease symptoms caused by pathogens different from M. tb. This article explains the importance of BCG immunization and the possible mechanisms associated with the induction of trained immunity, which might be used as a strategy for rapid activation of the immune system against unrelated pathogens.
Collapse
Affiliation(s)
- Jorge A Soto
- Millennium Institute of Immunology and Immunotherapy, Santiago 8330025, Chile; Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile
| | - Nicolás M S Gálvez
- Millennium Institute of Immunology and Immunotherapy, Santiago 8330025, Chile; Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile
| | - Catalina A Andrade
- Millennium Institute of Immunology and Immunotherapy, Santiago 8330025, Chile; Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile
| | - Mario A Ramírez
- Millennium Institute of Immunology and Immunotherapy, Santiago 8330025, Chile; Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile
| | - Claudia A Riedel
- Millennium Institute of Immunology and Immunotherapy, Santiago 8330025, Chile; Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute of Immunology and Immunotherapy, Santiago 8330025, Chile; Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile; Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile.
| | - Susan M Bueno
- Millennium Institute of Immunology and Immunotherapy, Santiago 8330025, Chile; Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile.
| |
Collapse
|
36
|
Hupert N, Marín-Hernández D, Gao B, Águas R, Nixon DF. Heterologous vaccination interventions to reduce pandemic morbidity and mortality: Modeling the US winter 2020 COVID-19 wave. Proc Natl Acad Sci U S A 2022; 119:e2025448119. [PMID: 35012976 PMCID: PMC8784160 DOI: 10.1073/pnas.2025448119] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 12/02/2021] [Indexed: 12/12/2022] Open
Abstract
COVID-19 remains a stark health threat worldwide, in part because of minimal levels of targeted vaccination outside high-income countries and highly transmissible variants causing infection in vaccinated individuals. Decades of theoretical and experimental data suggest that nonspecific effects of non-COVID-19 vaccines may help bolster population immunological resilience to new pathogens. These routine vaccinations can stimulate heterologous cross-protective effects, which modulate nontargeted infections. For example, immunization with Bacillus Calmette-Guérin, inactivated influenza vaccine, oral polio vaccine, and other vaccines have been associated with some protection from SARS-CoV-2 infection and amelioration of COVID-19 disease. If heterologous vaccine interventions (HVIs) are to be seriously considered by policy makers as bridging or boosting interventions in pandemic settings to augment nonpharmaceutical interventions and specific vaccination efforts, evidence is needed to determine their optimal implementation. Using the COVID-19 International Modeling Consortium mathematical model, we show that logistically realistic HVIs with low (5 to 15%) effectiveness could have reduced COVID-19 cases, hospitalization, and mortality in the United States fall/winter 2020 wave. Similar to other mass drug administration campaigns (e.g., for malaria), HVI impact is highly dependent on both age targeting and intervention timing in relation to incidence, with maximal benefit accruing from implementation across the widest age cohort when the pandemic reproduction number is >1.0. Optimal HVI logistics therefore differ from optimal rollout parameters for specific COVID-19 immunizations. These results may be generalizable beyond COVID-19 and the US to indicate how even minimally effective heterologous immunization campaigns could reduce the burden of future viral pandemics.
Collapse
Affiliation(s)
- Nathaniel Hupert
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY 10065;
- Division of General Internal Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY 10065
- Cornell Institute for Disease and Disaster Preparedness, Cornell University, New York, NY 10065
| | - Daniela Marín-Hernández
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065
| | - Bo Gao
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - Ricardo Águas
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - Douglas F Nixon
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065
| |
Collapse
|
37
|
Setiabudiawan TP, Reurink RK, Hill PC, Netea MG, van Crevel R, Koeken VACM. Protection against tuberculosis by Bacillus Calmette-Guérin (BCG) vaccination: A historical perspective. MED 2022; 3:6-24. [PMID: 35590145 DOI: 10.1016/j.medj.2021.11.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 01/23/2023]
Abstract
Bacillus Calmette-Guérin (BCG) was developed exactly 100 years ago, and it is still the only licensed tuberculosis (TB) vaccine and the most frequently administered of all vaccines worldwide. Despite universal vaccination policies in TB-endemic settings, the burden of TB remains high. Although BCG protects against Mycobacterium tuberculosis infection and TB disease, the level of protection varies greatly between age groups and settings. In this review, we present a historical perspective and describe the evidence for BCG's ability to protect against TB as well as the factors that influence protection. We also present the immunological mechanisms through which BCG vaccination induces protection, focusing on T cell, B cell, and innate immunity. Finally, we discuss several possibilities to boost BCG's efficacy, including alternative vaccination routes, BCG revaccination, and use of recombinant BCG vaccines, and describe the knowledge gaps that exist with respect to BCG's protection against TB.
Collapse
Affiliation(s)
- Todia P Setiabudiawan
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Gelderland 6525 GA, the Netherlands
| | - Remi K Reurink
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Gelderland 6525 GA, the Netherlands
| | - Philip C Hill
- Centre for International Health, University of Otago, Dunedin, North Dunedin 9016, New Zealand
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Gelderland 6525 GA, the Netherlands; Human Genomics Laboratory, Craiova University of Medicine and Pharmacy, Craiova, Județul Dolj 200349, Romania
| | - Reinout van Crevel
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Gelderland 6525 GA, the Netherlands; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Valerie A C M Koeken
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Gelderland 6525 GA, the Netherlands; Department of Computational Biology for Individualised Infection Medicine, Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover 30625, Germany; TWINCORE, a joint venture between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover 30625, Germany.
| |
Collapse
|
38
|
Tarabini RF, Rigo MM, Faustino Fonseca A, Rubin F, Bellé R, Kavraki LE, Ferreto TC, Amaral Antunes D, de Souza APD. Large-Scale Structure-Based Screening of Potential T Cell Cross-Reactivities Involving Peptide-Targets From BCG Vaccine and SARS-CoV-2. Front Immunol 2022; 12:812176. [PMID: 35095907 PMCID: PMC8793865 DOI: 10.3389/fimmu.2021.812176] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/21/2021] [Indexed: 12/22/2022] Open
Abstract
Although not being the first viral pandemic to affect humankind, we are now for the first time faced with a pandemic caused by a coronavirus. The Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has been responsible for the COVID-19 pandemic, which caused more than 4.5 million deaths worldwide. Despite unprecedented efforts, with vaccines being developed in a record time, SARS-CoV-2 continues to spread worldwide with new variants arising in different countries. Such persistent spread is in part enabled by public resistance to vaccination in some countries, and limited access to vaccines in other countries. The limited vaccination coverage, the continued risk for resistant variants, and the existence of natural reservoirs for coronaviruses, highlight the importance of developing additional therapeutic strategies against SARS-CoV-2 and other coronaviruses. At the beginning of the pandemic it was suggested that countries with Bacillus Calmette-Guérin (BCG) vaccination programs could be associated with a reduced number and/or severity of COVID-19 cases. Preliminary studies have provided evidence for this relationship and further investigation is being conducted in ongoing clinical trials. The protection against SARS-CoV-2 induced by BCG vaccination may be mediated by cross-reactive T cell lymphocytes, which recognize peptides displayed by class I Human Leukocyte Antigens (HLA-I) on the surface of infected cells. In order to identify potential targets of T cell cross-reactivity, we implemented an in silico strategy combining sequence-based and structure-based methods to screen over 13,5 million possible cross-reactive peptide pairs from BCG and SARS-CoV-2. Our study produced (i) a list of immunogenic BCG-derived peptides that may prime T cell cross-reactivity against SARS-CoV-2, (ii) a large dataset of modeled peptide-HLA structures for the screened targets, and (iii) new computational methods for structure-based screenings that can be used by others in future studies. Our study expands the list of BCG peptides potentially involved in T cell cross-reactivity with SARS-CoV-2-derived peptides, and identifies multiple high-density "neighborhoods" of cross-reactive peptides which could be driving heterologous immunity induced by BCG vaccination, therefore providing insights for future vaccine development efforts.
Collapse
Affiliation(s)
- Renata Fioravanti Tarabini
- Laboratory of Clinical and Experimental Immunology, Infant Center, School of Health Science, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | | | - André Faustino Fonseca
- Antunes Lab, Department of Biology and Biochemistry, University of Houston, Houston, TX, United States
| | - Felipe Rubin
- School of Technology - Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Rafael Bellé
- Laboratório de alto desempenho – Centro de Apoio ao desenvolvimento cientifico e tecnológico da (IDEIA), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Lydia E Kavraki
- Kavraki Lab, Department of Computer Science, Rice University, Houston, TX, United States
| | - Tiago Coelho Ferreto
- School of Technology - Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil,Laboratório de alto desempenho – Centro de Apoio ao desenvolvimento cientifico e tecnológico da (IDEIA), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Dinler Amaral Antunes
- Antunes Lab, Department of Biology and Biochemistry, University of Houston, Houston, TX, United States,*Correspondence: Ana Paula Duarte de Souza, ; Dinler Amaral Antunes,
| | - Ana Paula Duarte de Souza
- Laboratory of Clinical and Experimental Immunology, Infant Center, School of Health Science, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil,*Correspondence: Ana Paula Duarte de Souza, ; Dinler Amaral Antunes,
| |
Collapse
|
39
|
Thompson KM, Badizadegan K. Health economic analyses of secondary vaccine effects: a systematic review and policy insights. Expert Rev Vaccines 2021; 21:297-312. [PMID: 34927511 DOI: 10.1080/14760584.2022.2017287] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
INTRODUCTION : Numerous analyses demonstrate substantial health economic impacts of primary vaccine effects (preventing or mitigating clinical manifestations of the diseases they target), but vaccines may also be associated with secondary effects, previously known as non-specific, heterologous, or off-target effects. AREAS COVERED : We define key concepts to distinguish primary and secondary vaccine effects for health economic analyses, summarized terminology used in different fields, and perform a systematic review of health economic analyses focused on secondary vaccine effects (SVEs). EXPERT OPINION : Health economists integrate evidence from multiple fields, which often use incomplete or inconsistent definitions. Like regulators and policy makers, health economists require high-quality evidence of specific effects. Consistent with the limited evidence on mechanisms of action for SVEs, the associated health economic literature remains highly limited, with 4 studies identified by our systematic review. The lack of specific and well-controlled evidence that supports quantification of specific SVEs limits the consideration of these effects in vaccine research, development, regulatory, and recommendation decisions and health economic analyses.
Collapse
|
40
|
Chauhan A, Singh M, Agarwal A, Jaiswal N, M Lakshmi PV, Singh M. Exploring the role of bacillus calmette-guerin vaccination in protection against COVID-19. Int J Mycobacteriol 2021; 10:433-436. [PMID: 34916464 DOI: 10.4103/ijmy.ijmy_179_21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Background In recent studies, it has been established that countries having Bacillus Calmette-Guerin (BCG) vaccination programs reported less COVID-19 cases and deaths per population Preliminary studies suggest that BCG vaccination could upregulate the vaccine-induced immunity against SARS-CoV-2. Hence, the recent study was aimed to determine the association of BCG vaccine with the development of COVID-19 in health care workers. Methods The participants included in the study were healthcare workers (doctors, paramedics, and other supporting staff) working in the high and low-risk zones of COVID-19 in the hospital. The Graph Pad statistical analysis tool was used to determine the association of preliminary BCG vaccination with their COVID-19 status. Results A total of 200 health care workers working in different departments of the hospital were enrolled in the study. The relative risk of COVID-19 positivity in BCG vaccinated versus unvaccinated was 0.79 (0.50-1.25). There was also a significant negative correlation between two groups. The relative risk of COVID-19 positivity in those with a BCG scar was 0.63 (0.44-0.92). The relative risk of COVID-19 infection in those with a history of tuberculosis was 1.08 (0.56-2.09). Conclusion The present study did not support the beneficial effect of BCG vaccine in protection against the development of COVID-19 disease.
Collapse
Affiliation(s)
- Anil Chauhan
- Department of Telemedicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Manvi Singh
- Department of Telemedicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Amit Agarwal
- Department of Telemedicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Nishant Jaiswal
- Department of Telemedicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - P V M Lakshmi
- Department of Community Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Meenu Singh
- Department of Telemedicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
41
|
Prentice S, Dockrell HM. BCG Specific and Nonspecific Effects: Different Questions, Similar Challenges. J Infect Dis 2021; 224:1105-1108. [PMID: 34244763 PMCID: PMC8514176 DOI: 10.1093/infdis/jiab307] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 06/10/2021] [Indexed: 12/17/2022] Open
Affiliation(s)
- Sarah Prentice
- Department of Paediatrics and Neonatology, East and North Hertfordshire NHS Trust, Stevenage, United Kingdom
| | - Hazel M Dockrell
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
42
|
COVID-19 and Beyond: Exploring Public Health Benefits from Non-Specific Effects of BCG Vaccination. Microorganisms 2021; 9:microorganisms9102120. [PMID: 34683441 PMCID: PMC8539044 DOI: 10.3390/microorganisms9102120] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 09/29/2021] [Accepted: 10/06/2021] [Indexed: 12/23/2022] Open
Abstract
Bacille Calmette–Guérin (BCG) vaccination, widely used throughout the world to protect against infant tuberculous meningitis and miliary tuberculosis (TB), can provide broad non-specific protection against infectious respiratory diseases in certain groups. Interest in BCG has seen a resurgence within the scientific community as the mechanisms for non-specific protection have begun to be elucidated. The impact of the COVID-19 pandemic on nearly every aspect of society has profoundly illustrated the pressure that respiratory infections can place on a national healthcare system, further renewing interest in BCG vaccination as a public health policy to reduce the burden of those illnesses. However, the United States does not recommend BCG vaccination due to its variable effectiveness against adult TB, the relatively low risk of Mycobacterium tuberculosis infection in most of the United States, and the vaccine’s interference with tuberculin skin test reactivity that complicates TB screening. In this review, we explore the broad immune training effects of BCG vaccination and literature on the effects of BCG vaccination on COVID-19 spread, disease severity, and mortality. We further discuss barriers to scheduled BCG vaccination in the United States and how those barriers could potentially be overcome.
Collapse
|
43
|
Ayele AG, Enyew EF, Kifle ZD. Roles of existing drug and drug targets for COVID-19 management. Metabol Open 2021; 11:100103. [PMID: 34222852 PMCID: PMC8239316 DOI: 10.1016/j.metop.2021.100103] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 06/27/2021] [Indexed: 02/07/2023] Open
Abstract
In December 2019, a highly transmissible, pneumonia epidemic caused by a novel coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), erupted in China and other countries, resulting in devastation and health crisis worldwide currently. The search and using existing drugs support to curb the current highly contagious viral infection is spirally increasing since the pandemic began. This is based on these drugs had against other related RNA-viruses such as MERS-Cov, and SARS-Cov. Moreover, researchers are scrambling to identify novel drug targets and discover novel therapeutic options to vanquish the current pandemic. Since there is no definitive treatment to control Covid-19 vaccines are remain to be a lifeline. Currently, many vaccine candidates are being developed with most of them are reported to have positive results. Therapeutic targets such as helicases, transmembrane serine protease 2, cathepsin L, cyclin G-associated kinase, adaptor-associated kinase 1, two-pore channel, viral virulence factors, 3-chymotrypsin-like protease, suppression of excessive inflammatory response, inhibition of viral membrane, nucleocapsid, envelope, and accessory proteins, and inhibition of endocytosis were identified as a potential target against COVID-19 infection. This review also summarizes plant-based medicines for the treatment of COVID-19 such as saposhnikoviae divaricata, lonicerae japonicae flos, scutellaria baicalensis, lonicera japonicae, and some others. Thus, this review aimed to focus on the most promising therapeutic targets being repurposed against COVID-19 and viral elements that are used in COVID-19 vaccine candidates.
Collapse
Key Words
- 3CLpro, 3-chymotrypsin-like protease
- AAK1, adaptor-associated kinase 1
- ACE-2, Angiotensin-Converting Enzyme-2
- CEF, Cepharanthine
- COVID-19
- COVID-19, coronavirus disease-2019
- Existing drug
- GAK, cyclin G-associated kinase
- MERS-CoV, Middle East respiratory syndrome coronavirus
- Management
- Nsp, non-structure protein
- ORF, open reading frame
- PLpro, papain-like protease
- RdRp, RNA-dependence RNA-polymerase
- SARS-COV-2, severe acute respiratory syndrome coronavirus-2
- TMPRSS2, transmembrane Serine Protease 2
- TPC2, two-pore channel 2
- Therapeutic target
Collapse
Affiliation(s)
- Akeberegn Gorems Ayele
- Department of Pharmacology and Clinical Pharmacy, School of Pharmacy, College of Health Science, Addis Ababa University, Addis Ababa, Ethiopia
| | - Engidaw Fentahun Enyew
- Department of Human Anatomy, School of Medicine, College of Medicine and Health Sciences, Gondar, Ethiopia
| | - Zemene Demelash Kifle
- Department of Pharmacology, School of Pharmacy, College of Medicine and Health Science, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
44
|
Amirlak L, Haddad R, Hardy JD, Khaled NS, Chung MH, Amirlak B. Effectiveness of booster BCG vaccination in preventing Covid-19 infection. Hum Vaccin Immunother 2021; 17:3913-3915. [PMID: 34403297 PMCID: PMC8425429 DOI: 10.1080/21645515.2021.1956228] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The evidence that BCG (bacille Calmette-Guerin) vaccine may increase the ability of the immune system to fight off pathogens other than tuberculosis has been studied in the past. This nonspecific immunity gained our interest, especially after initial reports of less cases in countries with universal BCG vaccination. In hopes of possible protective immunity, all staff of the Emirates International Hospital (United Arab Emirates) were offered a booster BCG vaccine in early March 2020. All the hospital staff were then tested for Covid-19 infection by the end of June 2020. We divided the subjects into two groups: booster vaccinated versus unvaccinated. The rate of Covid-19 infection was compared between the groups. Criteria included all staff who were offered the vaccine. Seventy-one subjects received the booster vaccination. This group had zero cases of positive COVID 19 infection. Two hundred nine subjects did not receive the vaccination, with 18 positive PCR confirmed COVID 19 cases. The infection rate in the unvaccinated group was 8.6% versus zero in the booster vaccinated group (Fisher’s exact test p-value = .004). Our findings demonstrated the potential effectiveness of the booster BCG vaccine, specifically the booster in preventing Covid-19 infections in an elevated-risk healthcare population.
Collapse
Affiliation(s)
- Lradj Amirlak
- Department of Pediatrics, Emirates International Hospital, AL-Ain, United Arab Emirates
| | - Rifat Haddad
- Department of Otolaryngology, Emirates International Hospital, AL-Ain, United Arab Emirates
| | - John Denis Hardy
- Department of Pediatrics, Emirates International Hospital, AL-Ain, United Arab Emirates
| | - Naief Suleiman Khaled
- Department of Surgery, Emigrates International Hospital, AL-Ain, United Arab Emirates
| | - Michael H Chung
- Department of Plastic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bardia Amirlak
- Department of Plastic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
45
|
Koeken VACM. Controlling inflammation in the elderly with BCG vaccination. SCIENCE ADVANCES 2021; 7:eabk1668. [PMID: 34348906 PMCID: PMC8336952 DOI: 10.1126/sciadv.abk1668] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 07/06/2021] [Indexed: 05/04/2023]
Abstract
The tuberculosis vaccine BCG may protect against inflammation in the elderly as well as offer an option for protection from SARS-CoV-2 in developing countries.
Collapse
Affiliation(s)
- Valerie A C M Koeken
- Radboud Center for Infectious Diseases and Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Computational Biology for Individualised Infection Medicine, Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
- TWINCORE, a joint venture between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany.
| |
Collapse
|