Seasonal variation in the non-specific effects of BCG vaccination on neonatal mortality: three randomised controlled trials in Guinea-Bissau

Kristoffer Jarlov Jensen, Sofie Biering-Sørensen, Johan Ursing, Poul-Erik Lund Kofoed, Peter Aaby, Christine Stabell Benn, Kristoffer Jarlov Jensen, Sofie Biering-Sørensen, Johan Ursing, Poul-Erik Lund Kofoed, Peter Aaby, Christine Stabell Benn

Abstract

The BCG vaccine protects non-specifically against other diseases than tuberculosis. Three randomised controlled trials of early BCG in Guinea-Bissau found a 38% reduction in all-cause neonatal mortality. Little is known about the underlying mechanisms. In Guinea-Bissau, prevalent infectious diseases display distinct seasonality. Revisiting the three trials (>6500 infants) comparing early BCG versus no early BCG in low weight infants on all-cause neonatal mortality over 12 consecutive years, we explored the seasonal variation in BCG's effect on mortality. In a subgroup of participants, adaptive and innate cytokine responses were measured 4 weeks after randomisation. Consistently over the course of the three trials and 12 years, the effect of BCG on all-cause neonatal mortality was particularly beneficial when administered in November to January, coincident with peaking malaria infections. During these months, BCG was also associated with stronger proinflammatory responses to heterologous challenge. Recent studies have suggested a protective effect of BCG against malaria. BCG may also ameliorate immune-compromising fatal effects of placental malaria in the newborn.

Trial registration: ClinicalTrials.gov NCT00146302 NCT00625482.

Keywords: immunisation; malaria; maternal health; paediatrics; vaccines.

Conflict of interest statement

Competing interests: None declared.

© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
HR for neonatal death comparing early BCG with delayed BCG, per calendar month and by trial. For Trial II, no HR estimate for January and July due to no deaths in the early BCG group in January and delayed group in July, respectively. HR1 indicates lower mortality in the delayed BCG group.
Figure 2
Figure 2
HR for neonatal death comparing early BCG with delayed BCG, stratified by sex, trial and season. Estimated HR (with 95% CI) comparing early BCG to delayed BCG, stratified by season and trial. Season of BCG administration is defined as November to January versus February to October or dry (December to May) versus rainy (June to November). A HR of 1 is indicated with a vertical hatched line. Δp

Figure 3

Interferon (IFN)-γ responses to purified…

Figure 3

Interferon (IFN)-γ responses to purified protein derivative (PPD) and medium alone. Geometric means…

Figure 3
Interferon (IFN)-γ responses to purified protein derivative (PPD) and medium alone. Geometric means of IFN-γ concentrations in whole blood in vitro stimulated with PPD (10 μg/mL) 4 weeks after randomisation to early BCG or nothing (control), stratified by season (November to December vs March to October). Error bar is 95% CI.

Figure 4

The immunological effects of BCG…

Figure 4

The immunological effects of BCG on in vitro cytokine responses (collective test). Whole…

Figure 4
The immunological effects of BCG on in vitro cytokine responses (collective test). Whole blood from infants randomised to early BCG or nothing was collected 4 weeks after randomisation and stimulated with a panel of innate agonists and recall antigens (Jensen et al, J Infect D is 2015 211: 956–967), stratified by time (calendar month) of enrolment. The geometric mean ratios (GMRs) comparing early BCG vaccinated with non-vaccinated infants were analysed collectively for the non-specific responses to LPS, Pam3CSK4, PMA/ionomycin, CL075 and medium alone using Tobit regression. The study included infants randomised from March to December 2011. Season defined by the month of randomisation as November–December, n=101 versus March–October, n=366. *p<0.05; ***p<0.001 for test of BCG; ΔΔp<0.01 for interaction between season and BCG.
Figure 3
Figure 3
Interferon (IFN)-γ responses to purified protein derivative (PPD) and medium alone. Geometric means of IFN-γ concentrations in whole blood in vitro stimulated with PPD (10 μg/mL) 4 weeks after randomisation to early BCG or nothing (control), stratified by season (November to December vs March to October). Error bar is 95% CI.
Figure 4
Figure 4
The immunological effects of BCG on in vitro cytokine responses (collective test). Whole blood from infants randomised to early BCG or nothing was collected 4 weeks after randomisation and stimulated with a panel of innate agonists and recall antigens (Jensen et al, J Infect D is 2015 211: 956–967), stratified by time (calendar month) of enrolment. The geometric mean ratios (GMRs) comparing early BCG vaccinated with non-vaccinated infants were analysed collectively for the non-specific responses to LPS, Pam3CSK4, PMA/ionomycin, CL075 and medium alone using Tobit regression. The study included infants randomised from March to December 2011. Season defined by the month of randomisation as November–December, n=101 versus March–October, n=366. *p<0.05; ***p<0.001 for test of BCG; ΔΔp<0.01 for interaction between season and BCG.

References

    1. Higgins JPT, Soares-Weiser K, López-López JA, et al. . Association of BCG, DTP, and measles containing vaccines with childhood mortality: systematic review. BMJ 2016;355:i5170 10.1136/bmj.i5170
    1. Biering-Sorensen S, Aaby P, Lund N, et al. . Early BCG-Denmark and neonatal mortality among infants weighing. Clin Infect Dis 2017;65:1183–90.
    1. Biering-Sørensen S, Aaby P, Napirna BM, et al. . Small randomized trial among low-birth-weight children receiving Bacillus Calmette-Guérin vaccination at first health center contact. Pediatr Infect Dis J 2012;31:306–8. 10.1097/INF.0b013e3182458289
    1. Aaby P, Roth A, Ravn H, 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;204:245–52. 10.1093/infdis/jir240
    1. Schaltz-Buchholzer F, Biering-Sørensen S, Lund N, et al. . Early BCG vaccination, hospitalizations, and hospital deaths: analysis of a secondary outcome in 3 randomized trials from Guinea-Bissau. J Infect Dis 2019;219:624–32. 10.1093/infdis/jiy544
    1. Netea MG, Joosten LAB, Latz E, et al. . Trained immunity: a program of innate immune memory in health and disease. Science 2016;352:aaf1098 10.1126/science.aaf1098
    1. Fischer TK, Aaby P, Mølbak K, et al. . Rotavirus disease in Guinea-Bissau, West Africa: a review of longitudinal community and hospital studies. J Infect Dis 2010;202:S239–42. 10.1086/653568
    1. Weber MW, Milligan P, Sanneh M, et al. . An epidemiological study of RSV infection in the Gambia. Bull World Health Organ 2002;80:562–8.
    1. Fall A, Dia N, Cisse el HA, et al. . Epidemiology and molecular characterization of human respiratory syncytial virus in Senegal after four consecutive years of surveillance, 2012-2015. PloS one 2016;11:e0157163.
    1. Mølbak K, Højlyng N, Gottschau A, et al. . Cryptosporidiosis in infancy and childhood mortality in guinea Bissau, West Africa. BMJ 1993;307:417–20. 10.1136/bmj.307.6901.417
    1. Ursing J, Rombo L, Rodrigues A, et al. . Malaria transmission in Bissau, Guinea-Bissau between 1995 and 2012: malaria resurgence did not negatively affect mortality. PLoS One 2014;9:e101167 10.1371/journal.pone.0101167
    1. Jensen KJ, Larsen N, Biering-Sørensen S, et al. . Heterologous immunological effects of early BCG vaccination in low-birth-weight infants in Guinea-Bissau: a randomized-controlled trial. J Infect Dis 2015;211:956–67. 10.1093/infdis/jiu508
    1. Skogstrand K, Thorsen P, Nørgaard-Pedersen B, et al. . Simultaneous measurement of 25 inflammatory markers and neurotrophins in neonatal dried blood spots by immunoassay with xMAP technology. Clin Chem 2005;51:1854–66. 10.1373/clinchem.2005.052241
    1. Uh H-W, Hartgers FC, Yazdanbakhsh M, et al. . Evaluation of regression methods when immunological measurements are constrained by detection limits. BMC Immunol 2008;9:59 10.1186/1471-2172-9-59
    1. Andersen A, Benn CS, Jørgensen MJ, et al. . Censored correlated cytokine concentrations: multivariate Tobit regression using clustered variance estimation. Stat Med 2013;32:2859–74. 10.1002/sim.5696
    1. Nielsen BU, Byberg S, Aaby P, et al. . Seasonal variation in child mortality in rural Guinea-Bissau. Trop Med Int Health 2017;22:846–56. 10.1111/tmi.12889
    1. Veirum JE, Biai S, Jakobsen M, et al. . Persisting high hospital and community childhood mortality in an urban setting in Guinea-Bissau. Acta Paediatr 2007;96:1526–30. 10.1111/j.1651-2227.2007.00467.x
    1. Fischer TK, Valentiner-Branth P, Steinsland H, et al. . Protective immunity after natural rotavirus infection: a community cohort study of newborn children in Guinea-Bissau, West Africa. J Infect Dis 2002;186:593–7. 10.1086/342294
    1. Assane D, Makhtar C, Abdoulaye D, et al. . Viral and Bacterial Etiologies of Acute Respiratory Infections Among Children Under 5 Years in Senegal. Microbiol Insights 2018;11:117863611875865 10.1177/1178636118758651
    1. Kenmoe S, Tchendjou P, Vernet M-A, et al. . Viral etiology of severe acute respiratory infections in hospitalized children in Cameroon, 2011-2013. Influenza Other Respir Viruses 2016;10:386–93. 10.1111/irv.12391
    1. Feikin DR, Njenga MK, Bigogo G, et al. . Viral and bacterial causes of severe acute respiratory illness among children aged less than 5 years in a high malaria prevalence area of Western Kenya, 2007-2010. Pediatr Infect Dis J 2013;32:e14–19. 10.1097/INF.0b013e31826fd39b
    1. Pneumonia Etiology Research for Child Health (PERCH) Study Group Causes of severe pneumonia requiring hospital admission in children without HIV infection from Africa and Asia: the PERCH multi-country case-control study. Lancet 2019;394:30721–4. 10.1016/S0140-6736(19)30721-4
    1. Stensballe LG, Nante E, Jensen IP, et al. . 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;23:1251–7. 10.1016/j.vaccine.2004.09.006
    1. Parra M, Liu X, Derrick SC, et al. . Molecular analysis of non-specific protection against murine malaria induced by BCG vaccination. PLoS One 2013;8:e66115 10.1371/journal.pone.0066115
    1. Roth A, Gustafson P, Nhaga A, et al. . Bcg vaccination scar associated with better childhood survival in Guinea-Bissau. Int J Epidemiol 2005;34:540–7. 10.1093/ije/dyh392
    1. Walk J, de Bree LCJ, Graumans W, et al. . Outcomes of controlled human malaria infection after BCG vaccination. Nat Commun 2019;10:874 10.1038/s41467-019-08659-3
    1. Moya-Alvarez V, Abellana R, Cot M. Pregnancy-Associated malaria and malaria in infants: an old problem with present consequences. Malar J 2014;13:271 10.1186/1475-2875-13-271
    1. Ekanem AD, Anah MU, Udo JJ. The prevalence of congenital malaria among neonates with suspected sepsis in calabar, Nigeria. Trop Doct 2008;38:73–6. 10.1258/td.2007.005274
    1. Mwaniki MK, Talbert AW, Mturi FN, et al. . Congenital and neonatal malaria in a rural Kenyan district Hospital: an eight-year analysis. Malar J 2010;9:313 10.1186/1475-2875-9-313
    1. Moore KA, Fowkes FJI, Wiladphaingern J, et al. . Mediation of the effect of malaria in pregnancy on stillbirth and neonatal death in an area of low transmission: observational data analysis. BMC Med 2017;15:98 10.1186/s12916-017-0863-z
    1. Walther B, Miles DJC, Crozier S, et al. . Placental malaria is associated with reduced early life weight development of affected children independent of low birth weight. Malar J 2010;9:16 10.1186/1475-2875-9-16
    1. Le Hesran JY, Cot M, Personne P, et al. . Maternal placental infection with Plasmodium falciparum and malaria morbidity during the first 2 years of life. Am J Epidemiol 1997;146:826–31. 10.1093/oxfordjournals.aje.a009200
    1. Le Port A, Watier L, Cottrell G, et al. . Infections in infants during the first 12 months of life: role of placental malaria and environmental factors. PLoS One 2011;6:e27516 10.1371/journal.pone.0027516
    1. Rachas A, Le Port A, Cottrell G, et al. . Placental malaria is associated with increased risk of nonmalaria infection during the first 18 months of life in a Beninese population. Clin Infect Dis 2012;55:672–8. 10.1093/cid/cis490
    1. Kaestel P, Michaelsen KF, Aaby P, et al. . Effects of prenatal multimicronutrient supplements on birth weight and perinatal mortality: a randomised, controlled trial in Guinea-Bissau. Eur J Clin Nutr 2005;59:1081–9. 10.1038/sj.ejcn.1602215
    1. Berry I, Walker P, Tagbor H, et al. . Seasonal dynamics of malaria in pregnancy in West Africa: evidence for carriage of infections acquired before pregnancy until first contact with antenatal care. Am J Trop Med Hyg 2018;98:534–42. 10.4269/ajtmh.17-0620
    1. Desai M, ter Kuile FO, Nosten F, et al. . Epidemiology and burden of malaria in pregnancy. Lancet Infect Dis 2007;7:93–104. 10.1016/S1473-3099(07)70021-X
    1. Jayaraman K, Adhisivam B, Nallasivan S, et al. . Two Randomized Trials of the Effect of the Russian Strain of Bacillus Calmette-Guérin Alone or With Oral Polio Vaccine on Neonatal Mortality in Infants Weighing <2000 g in India. Pediatr Infect Dis J 2019;38:198–202. 10.1097/INF.0000000000002198
    1. Ritz N, Hanekom WA, Robins-Browne R, et al. . Influence of BCG vaccine strain on the immune response and protection against tuberculosis. FEMS Microbiol Rev 2008;32:821–41. 10.1111/j.1574-6976.2008.00118.x
    1. Ritz N, Dutta B, Donath S, et al. . The influence of Bacille Calmette-Guerin vaccine strain on the immune response against tuberculosis: a randomized trial. Am J Respir Crit Care Med 2012;185:213–22. 10.1164/rccm.201104-0714OC
    1. Funch KM, Thysen SM, Rodrigues A, et al. . Determinants of BCG scarification among children in rural Guinea-Bissau: a prospective cohort study. Hum Vaccin Immunother 2018;14:2434–42. 10.1080/21645515.2017.1421879
    1. Biering-Sørensen S, Jensen KJ, Aamand SH, et al. . Variation of growth in the production of the BCG vaccine and the association with the immune response. An observational study within a randomised trial. Vaccine 2015;33:2056–65. 10.1016/j.vaccine.2015.02.056
    1. Berendsen MLT, Øland CB, Bles P, et al. . Maternal priming: Bacillus Calmette-Guérin (BCG) vaccine scarring in mothers enhances the survival of their child with a BCG vaccine scar. J Pediatric Infect Dis Soc 2019;341 10.1093/jpids/piy142
    1. Stensballe LG, Ravn H, Birk NM, et al. . Bcg vaccination at birth and rate of hospitalization for infection until 15 months of age in Danish children: a randomized clinical multicenter trial. J Pediatric Infect Dis Soc 2018.
    1. Buffen K, Oosting M, Quintin J, et al. . Autophagy controls BCG-induced trained immunity and the response to intravesical BCG therapy for bladder cancer. PLoS Pathog 2014;10:e1004485 10.1371/journal.ppat.1004485
    1. Arts RJW, Carvalho A, La Rocca C, et al. . Immunometabolic pathways in BCG-induced trained immunity. Cell Rep 2016;17:2562–71. 10.1016/j.celrep.2016.11.011
    1. Moore SE. Early-Life nutritional programming of health and disease in the Gambia. Ann Nutr Metab 2017;70:179–83. 10.1159/000456555
    1. Moore SE, Goldblatt D, Bates CJ, et al. . Impact of nutritional status on antibody responses to different vaccines in undernourished Gambian children. Acta Paediatr 2003;92:170–6. 10.1111/j.1651-2227.2003.tb00522.x
    1. Walther B, Miles DJC, Waight P, et al. . Placental malaria is associated with attenuated CD4 T-cell responses to tuberculin PPD 12 months after BCG vaccination. BMC Infect Dis 2012;12:6 10.1186/1471-2334-12-6
    1. Brustoski K, Moller U, Kramer M, et al. . Reduced cord blood immune effector-cell responsiveness mediated by CD4+ cells induced in utero as a consequence of placental Plasmodium falciparum infection. J Infect Dis 2006;193:146–54. 10.1086/498578
    1. Brustoski K, Möller U, Kramer M, et al. . Ifn-Gamma and IL-10 mediate parasite-specific immune responses of cord blood cells induced by pregnancy-associated Plasmodium falciparum malaria. J Immunol 2005;174:1738–45. 10.4049/jimmunol.174.3.1738
    1. Bisseye C, van der Sande M, Morgan WD, et al. . Plasmodium falciparum infection of the placenta impacts on the T helper type 1 (Th1)/Th2 balance of neonatal T cells through CD4 + CD25 + forkhead box P3 + regulatory T cells and interleukin-10. Clin Exp Immunol 2009;158:287–93. 10.1111/j.1365-2249.2009.04014.x
    1. Ismaili J, van der Sande M, Holland MJ, et al. . Plasmodium falciparum infection of the placenta affects newborn immune responses. Clin Exp Immunol 2003;133:414–21. 10.1046/j.1365-2249.2003.02243.x
    1. Flanagan KL, Halliday A, Burl S, et al. . The effect of placental malaria infection on cord blood and maternal immunoregulatory responses at birth. Eur J Immunol 2010;40:1062–72. 10.1002/eji.200939638
    1. Malhotra I, Dent A, Mungai P, et al. . Can prenatal malaria exposure produce an immune tolerant phenotype? A prospective birth cohort study in Kenya. PLoS Med 2009;6:e1000116 10.1371/journal.pmed.1000116
    1. Cairo C, Longinaker N, Cappelli G, et al. . Cord blood Vγ2Vδ2 T cells provide a molecular marker for the influence of pregnancy-associated malaria on neonatal immunity. J Infect Dis 2014;209:1653–62. 10.1093/infdis/jit802
    1. Mazzola TN, Da Silva MTN, Moreno YMF, et al. . Robust gammadelta+ T cell expansion in infants immunized at birth with BCG vaccine. Vaccine 2007;25:6313–20. 10.1016/j.vaccine.2007.06.039
    1. Zufferey C, Germano S, Dutta B, et al. . The contribution of non-conventional T cells and NK cells in the mycobacterial-specific IFNγ response in Bacille Calmette-Guérin (BCG)-immunized infants. PLoS One 2013;8:e77334 10.1371/journal.pone.0077334
    1. Prahl M, Jagannathan P, McIntyre TI, et al. . Timing of in utero malaria exposure influences fetal CD4 T cell regulatory versus effector differentiation. Malar J 2016;15:497 10.1186/s12936-016-1545-6
    1. Prahl M, Jagannathan P, McIntyre TI, et al. . Sex Disparity in Cord Blood FoxP3+ CD4 T Regulatory Cells in Infants Exposed to Malaria In Utero. Open Forum Infect Dis 2017;4:ofx022 10.1093/ofid/ofx022
    1. Natama HM, Moncunill G, Rovira-Vallbona E, et al. . Modulation of innate immune responses at birth by prenatal malaria exposure and association with malaria risk during the first year of life. BMC Med 2018;16:198 10.1186/s12916-018-1187-3
    1. Gbédandé K, Varani S, Ibitokou S, et al. . Malaria modifies neonatal and early-life Toll-like receptor cytokine responses. Infect Immun 2013;81:2686–96. 10.1128/IAI.00237-13
    1. Zimmermann P, Curtis N. The influence of BCG on vaccine responses - a systematic review. Expert Rev Vaccines 2018;17:547–54. 10.1080/14760584.2018.1483727
    1. Marchant A, Goetghebuer T, Ota MO, et al. . Newborns develop a Th1-type immune response to Mycobacterium bovis Bacillus Calmette-Guérin vaccination. J Immunol 1999;163:2249–55.
    1. Smith SG, Zelmer A, Blitz R, et al. . Polyfunctional CD4 T-cells correlate with in vitro mycobacterial growth inhibition following Mycobacterium bovis BCG-vaccination of infants. Vaccine 2016;34:5298–305. 10.1016/j.vaccine.2016.09.002
    1. Freyne B, Donath S, Germano S, et al. . Neonatal BCG vaccination influences cytokine responses to Toll-like receptor ligands and heterologous antigens. J Infect Dis 2018;217:1798–808. 10.1093/infdis/jiy069
    1. Nissen TN, Birk NM, Blok BA, et al. . Bacillus Calmette-Guérin vaccination at birth and in vitro cytokine responses to non-specific stimulation. A randomized clinical trial. Eur J Clin Microbiol Infect Dis 2018;37:29–41. 10.1007/s10096-017-3097-2
    1. Garly ML, Balé C, Martins CL, et al. . Bcg vaccination among West African infants is associated with less anergy to tuberculin and diphtheria-tetanus antigens. Vaccine 2001;20:468–74. 10.1016/S0264-410X(01)00339-5
    1. Shaheen SO, Aaby P, Hall AJ, et al. . Cell mediated immunity after measles in Guinea-Bissau: historical cohort study. BMJ 1996;313:969–74. 10.1136/bmj.313.7063.969

Source: PubMed

3
Tilaa