Multiple BCG vaccinations for the prevention of COVID-19 and other infectious diseases in type 1 diabetes

Denise L Faustman, Amanda Lee, Emma R Hostetter, Anna Aristarkhova, Nathan C Ng, Gabriella F Shpilsky, Lisa Tran, Grace Wolfe, Hiroyuki Takahashi, Hans F Dias, Joan Braley, Hui Zheng, David A Schoenfeld, Willem M Kühtreiber, Denise L Faustman, Amanda Lee, Emma R Hostetter, Anna Aristarkhova, Nathan C Ng, Gabriella F Shpilsky, Lisa Tran, Grace Wolfe, Hiroyuki Takahashi, Hans F Dias, Joan Braley, Hui Zheng, David A Schoenfeld, Willem M Kühtreiber

Abstract

There is a need for safe and effective platform vaccines to protect against coronavirus disease 2019 (COVID-19) and other infectious diseases. In this randomized, double-blinded, placebo-controlled phase 2/3 trial, we evaluate the safety and efficacy of a multi-dose Bacillus Calmette-Guérin (BCG) vaccine for the prevention of COVID-19 and other infectious disease in a COVID-19-unvaccinated, at-risk-community-based cohort. The at-risk population is made of up of adults with type 1 diabetes. We enrolled 144 subjects and randomized 96 to BCG and 48 to placebo. There were no dropouts over the 15-month trial. A cumulative incidence of 12.5% of placebo-treated and 1% of BCG-treated participants meets criteria for confirmed COVID-19, yielding an efficacy of 92%. The BCG group also displayed fewer infectious disease symptoms and lesser severity and fewer infectious disease events per patient, including COVID-19. There were no BCG-related systemic adverse events. BCG's broad-based infection protection suggests that it may provide platform protection against new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants and other pathogens.

Trial registration: ClinicalTrials.gov NCT02081326.

Keywords: BCG; Bacillus Calmette-Guérin; COVID-19; Clinicaltrials.gov: NCT02081326; autoimmune; host microbe interactions; hygiene hypothesis; infectious diseases; phase 2/3 trial; randomized double blinded clinical trial; type 1 diabetes; vaccine.

Conflict of interest statement

Declaration of interests No author or author family member owns patents on this work. No author has any ownership rights to the study drug. No authors receive consulting or research support from Japan Laboratories.

Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.

Figures

Graphical abstract
Graphical abstract
Figure 1
Figure 1
Flow diagram and characteristics of participants (A) Flow diagram representing all enrolled participants from January 1, 2020 to April, 2021 for this double-blinded, randomized clinical trial testing repeat Tokyo-172 BCG vaccination versus placebo for COVID-19 protection. All 144 subjects were followed for 15 months with a 2:1 randomization and no dropouts. Data collection for this trial ended on April 2, 2021, the date when subjects started to receive provisionally approved COVID-19-specific vaccines. The geographical locations of the participants within the United States are shown in Figure S5. (B) Table of participant characteristics.
Figure 2
Figure 2
BCG vaccine efficacy and diagnostic confirmation of COVID-19 (A) Shown is the cumulative incidence of confirmed COVID-19 as a primary endpoint. During the 15-month surveillance time, one BCG recipient out of 96 (1.0%) met our criteria for molecularly confirmed COVID-19. In contrast, 6 out of 48 placebo recipients met the criteria (12.5%). Fisher’s exact testing showed a significant difference (two-tailed p = 0.006). Calculated vaccine efficacy was 92%, and the posterior probability (vaccine efficacy >30%) was 0.99. This was calculated using the Monte Carlo method. Vaccine efficacy was defined as (p1 – p2)/p1 × 100, where p1 is the percentage of COVID-positive subjects in the placebo group and p2 is the percentage of COVID-positive subjects in the BCG group. Our criteria for confirmed COVID-19 (see main text) required a combination of COVID-19 symptom(s) and ≥5 of 10 positive antibody assays including PCR, if available. Since many current clinical trials only define confirmed COVID-19 by symptom(s) and positive PCR testing, the PCR-only group was studied separately for vaccine efficacy (A and B). (B) Cumulative findings from each of the 10 diagnostic tests used to confirm COVID-19 (along with positive symptoms). These tests included the presence of COVID-19-specific antibodies to various SARS-CoV-2 virus epitopes through protein display (I–VIII), antibodies to the receptor-binding domain with an ELISA test (IX), and point-of-care testing (X) that included PCR. Our criteria for having confirmed COVID-19 required at least 5 of 10 detection methods to be positive, along with symptom(s). For the antibody assays, a patient was considered positive when the test resulted in a Z score of ≥3. In the cumulative graphs, the x axis data show the time period of the 15-month trial. The y axis shows the cumulative percentage of positive subjects. Except for the point-of-care graph (X), all other graphs represent the percentage of BCG and placebo patients with a Z score of ≥3 for the anti-SARS-CoV-2 antibody binding to a given protein region of the virus, i.e., the average antibody level during the COVID trial period was at least 3 standard deviations greater than the average level in the period preceding the COVID trial (baseline). The percentiles at the top right of each graph represent the calculated vaccine efficacy if this test alone was used to diagnose COVID-19 disease. Respective efficacy and Fisher’s exact p values for each COVID-19 antibody test were as follows: (I) 100%, p = 0.0007; (II) 80%, p = 0.089; (III) 91.7%, p = 0.009; (IV) 93.8%, p = 0.001; (V) 90%, p = 0.024; (VI) 87.5%, p = 0.004; (VII) 94.4%, p = 0.0003; (VIII) 83.3%, p = 0.009, (IX) 90%, p = 0.009; and (X) 91.7%, p = 0.029. For all graphs, BCG n = 96, placebo n = 48. Figure S1 shows the viral protein regions for each anti-COVID antibody tested. Number at risk data for each cumulative graph are shown in Figure S4.
Figure 3
Figure 3
Reduced COVID-19 disease markers after BCG vaccination (A) The heatmaps show Z scores for antibodies against SARS-CoV-2 in BCG- and placebo-treated symptomatic subjects (BCG n = 26, placebo n = 21). Since some of the Z scores in the left map are faint, the right map shows which Z scores are ≥3. The Roman numerals at the bottom of each lane denote antibodies against SARS-CoV-2 protein regions as listed in Figure S1. The maximum Z score in the left heatmap was 126. (B) Cumulative incidence of confirmed COVID-19 defined by symptomatic subjects testing positive solely by PCR. The BCG group (n = 96) had no symptomatic subjects who were PCR positive (0%), whereas the placebo group (n = 48) had 5 symptomatic and PCR-positive subjects (10.4%). This difference was significant (Fisher’s exact p = 0.0036) and translated to a vaccine efficacy of 100% with 0.99 posterior probability. There was low availability of positive PCR tests at point-of-care locations during the first 7 months of the trial. This, together with the need to perform the PCR test in a narrow window of about 2 weeks to be positive, is the reason why we designed the 9 non-PCR methods shown in Figures 2A, 2B, and 3A.
Figure 4
Figure 4
Cumulative infections and infection severity (A) Cumulative total infectious diseases during the 15 months of surveillance. This cumulative figure shows all infections per patient, including all COVID-19 events, within the BCG group (blue) compared with the placebo group (red). Included are the infections for which multiple subject events were documented in both BCG and placebo groups. Comparison by means of a Poisson model yields a significant difference with p = 0.004 (BCG n = 24 out of 96 T1D total, placebo n = 20 out of 48 T1D total). (B) Cumulative infections for two different time periods: pre-COVID-19 pandemic (the 2.5-year period prior to this trial, i.e., the pretrial) and during the COVID-19 pandemic (the current trial period of 15 months). The pretrial time period was when all clinical trial subjects received their ≥3 BCG vaccines or placebo vaccines; the current trial was when the subjects were monitored during the 15-month observation during the COVID-19 pandemic (total patients BCG n = 96, placebo n = 48). The lack of a statistical difference in the number of infections between BCG and placebo groups in the pretrial period suggests that a longer length of time than 2.5 years is necessary to realize BCG’s maximal infectious disease protection. It appears that during the entire period of 15 months, prior BCG vaccinations were protecting from COVID-19 and other infections. ∗∗p < 0.01. (C) Infectious disease index for symptomatic patients. Symptomatic patients in the BCG-treated group had significantly reduced total infectious disease symptom index (placebo 152 ± 70 [n = 20] versus BCG 48 ± 11 [n = 31], p = 0.04, single tail and unpaired) as well as average infectious disease symptom index (placebo 23 ± 7 [n = 20] and BCG 13 ± 2 [n = 31], p = 0.04, single tail and unpaired). We first calculated total and average symptom scores per patient and then calculated average and SEM of each of these for BCG and placebo cohorts separately (∗Student’s t test p < 0.05, one-tailed, unpaired). (D) Patients in the BCG cohort reported significantly fewer days of missed work during infections compared with the placebo group (∗p = 0.02). Missed work was reported by 7 out of 32 BCG patients and by 8 out of 17 placebo patients. (E) The number of patients that reported at least one symptom were 20 out of 48 in the placebo group and 33 out of 96 in the BCG group. The placebo group had more severe average symptoms compared with the BCG group for 12 out of 12 symptoms (left panel). The number of patients in BCG and placebo groups that reported each symptom was then expressed (right panel). For 11 out of 12 symptoms, there was a higher percentage of symptomatic patients in the placebo group compared with the BCG group. Statistical analysis by Student’s t test (one-tailed, unpaired, ∗p < 0.05).
Figure 5
Figure 5
Infection symptoms of trial participants versus adult household members (A) Infection symptoms in BCG and placebo groups compared with non-diabetic adult partners living in the same households. We collected surveys of symptoms of infectious diseases from all trial participants and household members of 13 BCG families and 7 placebo families. The BCG-treated trial participants had comparable or lower total infectious symptoms indexes compared with their partners living in the same household, whereas most placebo-treated trial participants had more severe symptoms compared with their partners. Statistical analysis of the differences by two-sample Wilcoxon testing was significant (two-tailed; p = 0.049; BCG n = 13 and placebo n = 7). Left panel: xy plot of symptom index of trial participants versus their household members. Right panel: the same data are also shown as an xy plot of household members versus the difference of trial participants and their household members. This plot makes it easier to visualize which groups had milder or more severe disease by introducing positive and negative differences. (B) Distribution of individual infectious disease symptoms in BCG- and placebo-treated participants and infected household members across the four scoring possibilities (0: no symptoms; 1: mild; 2: moderate; 3: severe symptoms). Number at risk data are shown in Figure S4.

References

    1. Anonymous BCG vaccines: WHO position paper. Wkly. Epidemiol. Rec. 2018
    1. Aaby P., Roth A., Ravn H., Napirna B.M., Rodrigues A., Lisse I.M., Stensballe L., Diness B.R., Lausch K.R., Lund N., et al. Randomized trial of BCG vaccination at birth to low-birth-weight children: beneficial nonspecific effects in the neonatal period? J. Infect. Dis. 2011;204:245–252. doi: 10.1093/infdis/jir240.
    1. Biering-Sorensen S., Aaby P., Lund N., Monteiro I., Jensen K.J., Eriksen H.B., Schaltz-Buchholzer F., Jorgensen A.S.P., Rodrigues A., Fisker A.B., Benn C.S. Early BCG-Denmark and neonatal mortality among infants weighing <2500 g: a randomized controlled trial. Clin. Infect. Dis. 2017;65:1183–1190. doi: 10.1093/cid/cix525.
    1. Biering-Sorensen S., Aaby P., Napirna B.M., Roth A., Ravn H., Rodrigues A., Whittle H., Benn C.S. Small randomized trial among low-birth-weight children receiving bacillus Calmette-Guerin vaccination at first health center contact. Pediatr. Infect. Dis. J. 2012;31:306–308. doi: 10.1097/INF.0b013e3182458289.
    1. de Castro M.J., Pardo-Seco J., Martinon-Torres F. Nonspecific (heterologous) protection of neonatal BCG vaccination against hospitalization due to respiratory infection and sepsis. Clin. Infect. Dis. 2015;60:1611–1619. doi: 10.1093/cid/civ144.
    1. Garly M.L., Martins C.L., Bale C., Balde M.A., Hedegaard K.L., Gustafson P., Lisse I.M., Whittle H.C., Aaby P. BCG scar and positive tuberculin reaction associated with reduced child mortality in West Africa. A non-specific beneficial effect of BCG? Vaccine. 2003;21:2782–2790. doi: 10.1016/s0264-410x(03)00181-6.
    1. Giamarellos-Bourboulis E.J., Tsilika M., Moorlag S., Antonakos N., Kotsaki A., Dominguez-Andres J., Kyriazopoulou E., Gkavogianni T., Adami M.E., Damoraki G., et al. Activate: randomized clinical trial of BCG vaccination against infection in the elderly. Cell. 2020;183:315–323.e9. doi: 10.1016/j.cell.2020.08.051.
    1. Hawkridge A., Hatherill M., Little F., Goetz M.A., Barker L., Mahomed H., Sadoff J., Hanekom W., Geiter L., Hussey G., South African B.C.G.t.t. Efficacy of percutaneous versus intradermal BCG in the prevention of tuberculosis in South African infants: randomised trial. BMJ. 2008;337:a2052. doi: 10.1136/bmj.a2052.
    1. Kristensen I., Aaby P., Jensen H. Routine vaccinations and child survival: follow up study in Guinea-Bissau, West Africa. BMJ. 2000;321:1435–1438. doi: 10.1136/bmj.321.7274.1435.
    1. Nemes E., Geldenhuys H., Rozot V., Rutkowski K.T., Ratangee F., Bilek N., Mabwe S., Makhethe L., Erasmus M., Toefy A., et al. Prevention of M. tuberculosis infection with H4:IC31 vaccine or BCG revaccination. N. Engl. J. Med. 2018;379:138–149. doi: 10.1056/NEJMoa1714021.
    1. Ponnighaus J.M., Fine P.E., Sterne J.A., Wilson R.J., Msosa E., Gruer P.J., Jenkins P.A., Lucas S.B., Liomba N.G., Bliss L. Efficacy of BCG vaccine against leprosy and tuberculosis in northern Malawi. Lancet. 1992;339:636–639. doi: 10.1016/0140-6736(92)90794-4.
    1. Roth A., Gustafson P., Nhaga A., Djana Q., Poulsen A., Garly M.L., Jensen H., Sodemann M., Rodriques A., Aaby P. BCG vaccination scar associated with better childhood survival in Guinea-Bissau. Int. J. Epidemiol. 2005;34:540–547. doi: 10.1093/ije/dyh392.
    1. Shann F. The non-specific effects of vaccines. Arch. Dis. Child. 2010;95:662–667. doi: 10.1136/adc.2009.157537.
    1. Stensballe L.G., Nante E., Jensen I.P., Kofoed P.E., Poulsen A., Jensen H., Newport M., Marchant A., Aaby P. Acute lower respiratory tract infections and respiratory syncytial virus in infants in Guinea-Bissau: a beneficial effect of BCG vaccination for girls community based case-control study. Vaccine. 2005;23:1251–1257. doi: 10.1016/j.vaccine.2004.09.006.
    1. Walk J., de Bree L.C.J., Graumans W., Stoter R., van Gemert G.J., van de Vegte-Bolmer M., Teelen K., Hermsen C.C., Arts R.J.W., Behet M.C., et al. Outcomes of controlled human malaria infection after BCG vaccination. Nat. Commun. 2019;10:874. doi: 10.1038/s41467-019-08659-3.
    1. Wardhana, Datau E.A., Sultana A., Mandang V.V., Jim E. The efficacy of Bacillus Calmette-Guerin vaccinations for the prevention of acute upper respiratory tract infection in the elderly. Acta Med. Indones. 2011;43:185–190.
    1. Calmette A. Preventive vaccination against tuberculosis with BCG. Proc. R Soc. Med. 1931;24:1481–1490.
    1. Faustman D.L., Wang L., Okubo Y., Burger D., Ban L., Man G., Zheng H., Schoenfeld D., Pompei R., Avruch J., Nathan D.M. Proof-of-concept, randomized, controlled clinical trial of Bacillus-Calmette-Guerin for treatment of long-term type 1 diabetes. PLoS One. 2012;7:e41756. doi: 10.1371/journal.pone.0041756.
    1. Kuhtreiber W.M., Tran L., Kim T., Dybala M., Nguyen B., Plager S., Huang D., Janes S., Defusco A., Baum D., et al. Long-term reduction in hyperglycemia in advanced type 1 diabetes: the value of induced aerobic glycolysis with BCG vaccinations. NPJ Vaccines. 2018;3:23. doi: 10.1038/s41541-018-0062-8.
    1. Paolillo A., Buzzi M.G., Giugni E., Sabatini U., Bastianello S., Pozzilli C., Salvetti M., Ristori G. The effect of Bacille Calmette-Guerin on the evolution of new enhancing lesions to hypointense T1 lesions in relapsing remitting MS. J. Neurol. 2003;250:247–248. doi: 10.1007/s00415-003-0967-6.
    1. Ristori G., Buzzi M.G., Sabatini U., Giugni E., Bastianello S., Viselli F., Buttinelli C., Ruggieri S., Colonnese C., Pozzilli C., Salvetti M. Use of bacille Calmette-Guerin (BCG) in multiple sclerosis. Neurology. 1999;53:1588–1589.
    1. Bagheri N., Montazeri H. On BCG vaccine protection from COVID-19: a review. SN Compr. Clin. Med. 2021;3:1261–1271. doi: 10.1007/s42399-021-00835-1.
    1. Curtis N., Sparrow A., Ghebreyesus T.A., Netea M.G. Considering BCG vaccination to reduce the impact of COVID-19. Lancet. 2020;395:1545–1546. doi: 10.1016/S0140-6736(20)31025-4.
    1. Escobar C.E. Correction for Escobar et al., BCG vaccine protection from severe coronavirus disease 2019 (COVID-19) Proc. Natl. Acad. Sci. USA. 2020;117:27741–27742. doi: 10.1073/pnas.2019438117.
    1. Klinger D., Blass I., Rappoport N., Linial M. Significantly improved COVID-19 outcomes in countries with higher BCG vaccination coverage: a multivariable analysis. Vaccines (Basel) 2020;8 doi: 10.3390/vaccines8030378.
    1. Moorlag S., van Deuren R.C., van Werkhoven C.H., Jaeger M., Debisarun P., Taks E., Mourits V.P., Koeken V., de Bree L.C.J., Ten Doesschate T., et al. Safety and COVID-19 symptoms in individuals recently vaccinated with BCG: a retrospective cohort study. Cell Rep. Med. 2020;1:100073. doi: 10.1016/j.xcrm.2020.100073.
    1. Rivas M.N., Ebinger J.E., Wu M., Sun N., Braun J., Sobhani K., Van Eyk J.E., Cheng S., Arditi M. BCG vaccination history associates with decreased SARS-CoV-2 seroprevalence across a diverse cohort of health care workers. J. Clin. Invest. 2021;131 doi: 10.1172/JCI145157.
    1. Wickramasinghe D., Wickramasinghe N., Kamburugamuwa S., Arambepola C., Samarasekera D. Correlation between immunity from BCG and the morbidity and mortality of COVID-19. Trop. Dis. Travel Med. Vaccines. 2020;5:17. doi: 10.1186/s40794-020-00117-z.
    1. Berg M.K., Yu Q., Salvador C.E., Melani I., Kitayama S. Mandated Bacillus Calmette-Guerin (BCG) vaccination predicts flattened curves for the spread of COVID-19. Sci. Adv. 2020;6:eabc1463. doi: 10.1126/sciadv.abc1463.
    1. Joy M., Malavika B., Asirvatham E.S., Sudarsanam T.D., Jeyaseelan L. Is BCG associated with reduced incidence of COVID-19? A meta-regression of global data from 160 countries. Clin. Epidemiol. Glob. Health. 2021;9:202–203. doi: 10.1016/j.cegh.2020.08.015.
    1. Kinoshita M., Tanaka M. Impact of routine infant BCG vaccination on COVID-19. J. Infect. 2020;81:625–633. doi: 10.1016/j.jinf.2020.08.013.
    1. Hauer J., Fischer U., Auer F., Borkhardt A. Regional BCG vaccination policy in former East- and West Germany may impact on both severity of SARS-CoV-2 and incidence of childhood leukemia. Leukemia. 2020;34:2217–2219. doi: 10.1038/s41375-020-0871-4.
    1. Gursel M., Gursel I. Is global BCG vaccination-induced trained immunity relevant to the progression of SARS-CoV-2 pandemic? Allergy. 2020;75:1815–1819. doi: 10.1111/all.14345.
    1. Brooks N.A., Puri A., Garg S., Nag S., Corbo J., Turabi A.E., Kaka N., Zemmel R.W., Hegarty P.K., Kamat A.M. The association of Coronavirus Disease-19 mortality and prior bacille Calmette-Guerin vaccination: a robust ecological analysis using unsupervised machine learning. Sci. Rep. 2021;11:774. doi: 10.1038/s41598-020-80787-z.
    1. Samrah S.M., Al-Mistarehi A.W., Ibnian A.M., Raffee L.A., Momany S.M., Al-Ali M., Hayajneh W.A., Yusef D.H., Awad S.M., Khassawneh B.Y. COVID-19 outbreak in Jordan: epidemiological features, clinical characteristics, and laboratory findings. Ann. Med. Surg. (Lond) 2020;57:103–108. doi: 10.1016/j.amsu.2020.07.020.
    1. Ozdemir C., Kucuksezer U.C., Tamay Z.U. Is BCG vaccination affecting the spread and severity of COVID-19? Allergy. 2020;75:1824–1827. doi: 10.1111/all.14344.
    1. Kumar A., Misra S., Verma V., Vishwakarma R.K., Kamal V.K., Nath M., Prakash K., Upadhyay A.D., Sahu J.K. Global impact of environmental temperature and BCG vaccination coverage on the transmissibility and fatality rate of COVID-19. PLoS One. 2020;15:e0240710. doi: 10.1371/journal.pone.0240710.
    1. Upton C.M., van Wijk R.C., Mockeliunas L., Simonsson U.S.H., McHarry K., van den Hoogen G., Muller C., von Delft A., van der Westhuizen H.M., van Crevel R., et al. Safety and efficacy of BCG re-vaccination in relation to COVID-19 morbidity in healthcare workers: a double-blind, randomised, controlled, phase 3 trial. EClinicalMedicine. 2022;48:101414. doi: 10.1016/j.eclinm.2022.101414.
    1. Moorlag S., Taks E., Ten Doesschate T., van der Vaart T.W., Janssen A.B., Muller L., Ostermann P., Dijkstra H., Lemmers H., Simonetti E., et al. Efficacy of Bacillus Calmette-Guerin vaccination against respiratory tract infections in the elderly during the Covid-19 pandemic. Clin. Infect. Dis. 2022 doi: 10.1093/cid/ciac182.
    1. Li W.X. Worldwide inverse correlation between Bacille Calmette-Guerin (BCG) immunization and COVID-19 mortality. Infection. 2021;49:463–473. doi: 10.1007/s15010-020-01566-6.
    1. Pepin J., Labbe A.C., Carignan A., Parent M.E., Yu J., Grenier C., Beauchemin S., De Wals P., Valiquette L., Rousseau M.C. Does BCG provide long-term protection against SARS-CoV-2 infection? a case-control study in Quebec, Canada. Vaccine. 2021;39:7300–7307. doi: 10.1016/j.vaccine.2021.08.019.
    1. de Chaisemartin C., de Chaisemartin L. Public Health Emergency Collection. Oxford University Press; 2020. BCG Vaccination in Infancy Does Not Protect against COVID-19. Evidence from a Natural Experiment in Sweden.
    1. Hensel J., McAndrews K.M., McGrail D.J., Dowlatshahi D.P., LeBleu V.S., Kalluri R. Protection against SARS-CoV-2 by BCG vaccination is not supported by epidemiological analyses. Sci. Rep. 2020;10:18377. doi: 10.1038/s41598-020-75491-x.
    1. Hamiel U., Kozer E., Youngster I. SARS-CoV-2 rates in BCG-vaccinated and unvaccinated young adults. JAMA. 2020;323:2340–2341. doi: 10.1001/jama.2020.8189.
    1. Bates M.N., Herron T.J., Lwi S.J., Baldo J.V. BCG vaccination at birth and COVID-19: a case-control study among U.S. military Veterans. Hum. Vaccin. Immunother. 2022;18:1981084. doi: 10.1080/21645515.2021.1981084.
    1. Su W.J., Chang C.H., Wang J.L., Chen S.F., Yang C.H. COVID-19 severity and neonatal BCG vaccination among young population in Taiwan. Int. J. Environ. Res. Public Health. 2021;18 doi: 10.3390/ijerph18084303.
    1. Amirlak L., Haddad R., Hardy J.D., Khaled N.S., Chung M.H., Amirlak B. Effectiveness of booster BCG vaccination in preventing Covid-19 infection. Hum. Vaccin. Immunother. 2021;17:3913–3915. doi: 10.1080/21645515.2021.1956228.
    1. Carey I.M., Critchley J.A., DeWilde S., Harris T., Hosking F.J., Cook D.G. Risk of infection in type 1 and type 2 diabetes compared with the general population: a matched cohort study. Diabetes Care. 2018;41:513–521. doi: 10.2337/dc17-2131.
    1. Barrett C.E., Park J., Kompaniyets L., Baggs J., Cheng Y.J., Zhang P., Imperatore G., Pavkov M.E. Intensive care unit admission, mechanical ventilation, and mortality among patients with type 1 diabetes hospitalized for COVID-19 in the U.S. Diabetes Care. 2021;44:1788–1796. doi: 10.2337/dc21-0604.
    1. Moorlag S., Arts R.J.W., van Crevel R., Netea M.G. Non-specific effects of BCG vaccine on viral infections. Clin. Microbiol. Infect. 2019;25:1473–1478. doi: 10.1016/j.cmi.2019.04.020.
    1. FDA . Center for Biologics Evaluation and Research Center for Drug Evaluation and Research. FDA; 2020. Assessing COVID-19 related symptoms in outpatient adult and adolescent subjects in clinical trials of drugs and biological products for COVID-19 prevention or treatment. Guidance for Industry.
    1. Abdallah M., Mahgoub A., Ahmed H., Chaterji S. Author correction: athena: automated tuning of k-mer based genomic error correction algorithms using language models. Sci. Rep. 2020;10:2390. doi: 10.1038/s41598-020-59141-w.
    1. Angelidou A., Conti M.G., Diray-Arce J., Benn C.S., Shann F., Netea M.G., Liu M., Potluri L.P., Sanchez-Schmitz G., Husson R., et al. Licensed Bacille Calmette-Guerin (BCG) formulations differ markedly in bacterial viability, RNA content and innate immune activation. Vaccine. 2020;38:2229–2240. doi: 10.1016/j.vaccine.2019.11.060.
    1. Hayashi D., Takii T., Fujiwara N., Fujita Y., Yano I., Yamamoto S., Kondo M., Yasuda E., Inagaki E., Kanai K., et al. Comparable studies of immunostimulating activities in vitro among Mycobacterium bovis bacillus Calmette-Guerin (BCG) substrains. FEMS Immunol. Med. Microbiol. 2009;56:116–128. doi: 10.1111/j.1574-695X.2009.00559.x.
    1. Rentsch C.A., Birkhauser F.D., Biot C., Gsponer J.R., Bisiaux A., Wetterauer C., Lagranderie M., Marchal G., Orgeur M., Bouchier C., et al. Bacillus Calmette-Guerin strain differences have an impact on clinical outcome in bladder cancer immunotherapy. Eur. Urol. 2014;66:677–688. doi: 10.1016/j.eururo.2014.02.061.
    1. Ritz N., Dutta B., Donath S., Casalaz D., Connell T.G., Tebruegge M., Robins-Browne R., Hanekom W.A., Britton W.J., Curtis N. The influence of bacille Calmette-Guerin vaccine strain on the immune response against tuberculosis: a randomized trial. Am. J. Respir. Crit. Care Med. 2012;185:213–222. doi: 10.1164/rccm.201104-0714OC.
    1. Shann F. Editorial commentary: different strains of Bacillus Calmette-Guerin vaccine have very different effects on tuberculosis and on unrelated infections. Clin. Infect. Dis. 2015;61:960–962. doi: 10.1093/cid/civ454.
    1. Roser M. Our World of Data. Covid-19 data explorer. 2022.
    1. Aronson N.E., Santosham M., Comstock G.W., Howard R.S., Moulton L.H., Rhoades E.R., Harrison L.H. Long-term efficacy of BCG vaccine in American Indians and Alaska Natives: a 60-year follow-up study. JAMA. 2004;291:2086–2091. doi: 10.1001/jama.291.17.2086.
    1. Cirovic B., de Bree L.C.J., Groh L., Blok B.A., Chan J., van der Velden W., Bremmers M.E.J., van Crevel R., Handler K., Picelli S., et al. BCG vaccination in humans elicits trained immunity via the hematopoietic progenitor compartment. Cell Host Microbe. 2020;28:322–334.e5. doi: 10.1016/j.chom.2020.05.014.
    1. Darrah P.A., Zeppa J.J., Maiello P., Hackney J.A., Wadsworth M.H., 2nd, Hughes T.K., Pokkali S., Swanson P.A., 2nd, Grant N.L., Rodgers M.A., et al. Prevention of tuberculosis in macaques after intravenous BCG immunization. Nature. 2020;577:95–102. doi: 10.1038/s41586-019-1817-8.
    1. Keefe R.C., Takahashi H., Tran L., Nelson K., Ng N., Kuhtreiber W.M., Faustman D.L. BCG therapy is associated with long-term, durable induction of Treg signature genes by epigenetic modulation. Sci. Rep. 2021;11:14933. doi: 10.1038/s41598-021-94529-2.
    1. Dias H.F., Kuhtreiber W.M., Nelson K.J., Ng N.C., Zheng H., Faustman D.L. Epigenetic changes related to glucose metabolism in type 1 diabetes after BCG vaccinations: a vital role for KDM2B. Vaccine. 2021 doi: 10.1016/j.vaccine.2021.04.011.
    1. Kuhtreiber W.M., Takahashi H., Keefe R.C., Song Y., Tran L., Luck T.G., Shpilsky G., Moore L., Sinton S.M., Graham J.C., Faustman D.L. BCG vaccinations upregulate Myc, a central switch for improved glucose metabolism in diabetes. iScience. 2020;23:101085. doi: 10.1016/j.isci.2020.101085.
    1. van der Heijden C., Noz M.P., Joosten L.A.B., Netea M.G., Riksen N.P., Keating S.T. Epigenetics and trained immunity. Antioxid Redox Signal. 2018;29:1023–1040. doi: 10.1089/ars.2017.7310.
    1. Fanucchi S., Dominguez-Andres J., Joosten L.A.B., Netea M.G., Mhlanga M.M. The intersection of epigenetics and metabolism in trained immunity. Immunity. 2021;54:32–43. doi: 10.1016/j.immuni.2020.10.011.
    1. Lunn D.J., Thomas A., Best N., Spiegelhalter D. WinBugs — a Bayesian modelling framework: concepts, structure, and extensibility. Stat. Comput. 2000;10:325–337.
    1. Polack F.P., Thomas S.J., Kitchin N., Absalon J., Gurtman A., Lockhart S., Perez J.L., Pérez Marc G., Moreira E.D., Zerbini C., et al. Safety and efficacy of the BNT162b2 mRNA Covid-19 vaccine. N. Engl. J. Med. 2020;383:2603–2615.

Source: PubMed

3
Subskrybuj