A novel intervention combining supplementary food and infection control measures to improve birth outcomes in undernourished pregnant women in Sierra Leone: A randomized, controlled clinical effectiveness trial

David Taylor Hendrixson, Kristie Smith, Patrick Lasowski, Meghan Callaghan-Gillespie, Jacklyn Weber, Peggy Papathakis, Per Ole Iversen, Aminata Shamit Koroma, Mark J Manary, David Taylor Hendrixson, Kristie Smith, Patrick Lasowski, Meghan Callaghan-Gillespie, Jacklyn Weber, Peggy Papathakis, Per Ole Iversen, Aminata Shamit Koroma, Mark J Manary

Abstract

Background: Innovations for undernourished pregnant women that improve newborn survival and anthropometry are needed to achieve the Sustainable Development Goals 1 and 3. This study tested the hypothesis that a combination of a nutritious supplementary food and several proven chemotherapeutic interventions to control common infections would increase newborn weight and length in undernourished pregnant women.

Methods and findings: This was a prospective, randomized, controlled clinical effectiveness trial of a ready-to-use supplementary food (RUSF) plus anti-infective therapies compared to standard therapy in undernourished pregnant women in rural Sierra Leone. Women with a mid-upper arm circumference (MUAC) ≤23.0 cm presenting for antenatal care at one of 43 government health clinics in Western Rural Area and Pujehun districts were eligible for participation. Standard of care included a blended corn/soy flour and intermittent preventive treatment for malaria in pregnancy (IPTp). The intervention replaced the blended flour with RUSF and added azithromycin and testing and treatment for vaginal dysbiosis. Since the study involved different foods and testing procedures for the intervention and control groups, no one except the authors conducting the data analyses were blinded. The primary outcome was birth length. Secondary outcomes included maternal weight gain, birth weight, and neonatal survival. Follow-up continued until 6 months postpartum. Modified intention to treat analyses was undertaken. Participants were enrolled and followed up from February 2017 until February 2020. Of the 1,489 women enrolled, 752 were allocated to the intervention and 737 to the standard of care. The median age of these women was 19.5 years, of which 42% were primigravid. Twenty-nine women receiving the intervention and 42 women receiving the standard of care were lost to follow-up before pregnancy outcomes were obtained. There were 687 singleton live births in the intervention group and 657 in the standard of care group. Newborns receiving the intervention were 0.3 cm longer (95% confidence interval (CI) 0.09 to 0.6; p = 0.007) and weighed 70 g more (95% CI 20 to 120; p = 0.005) than those receiving the standard of care. Those women receiving the intervention had greater weekly weight gain (mean difference 40 g; 95% CI 9.70 to 71.0, p = 0.010) than those receiving the standard of care. There were fewer neonatal deaths in the intervention (n = 13; 1.9%) than in the standard of care (n = 28; 4.3%) group (difference 2.4%; 95% CI 0.3 to 4.4), (HR 0.62 95% CI 0.41 to 0.94, p = 0.026). No differences in adverse events or symptoms between the groups was found, and no serious adverse events occurred. Key limitations of the study are lack of gestational age estimates and unblinded administration of the intervention.

Conclusions: In this study, we observed that the addition of RUSF, azithromycin, more frequent IPTp, and testing/treatment for vaginal dysbiosis in undernourished pregnant women resulted in modest improvements in anthropometric status of mother and child at birth, and a reduction in neonatal death. Implementation of this combined intervention in rural, equatorial Africa may well be an important, practical measure to reduce infant mortality in this context.

Trial registration: ClinicalTrials.gov NCT03079388.

Conflict of interest statement

I have read the journal’s policy and the authors of this manuscript have the following competing interests: DTH, KS, PL, MCG, JW and MJM received research support from the Children’s Investment Fund Foundation and Hickey Family Foundation. DTH receives support from National Institute of Diabetes and Digestive and Kidney Diseases under Award Number T32 DK077653.

Figures

Fig 1. CONSORT flow diagram.
Fig 1. CONSORT flow diagram.
“Lost to follow-up” refers to individuals who were not seen at the time point indicated and subsequently never seen again. “Visit data unavailable” refers to individuals who were not seen at the time point indicated but subsequently seen. Deaths are reported as additional deaths between each time point.
Fig 2. Kaplan–Meier survival estimates.
Fig 2. Kaplan–Meier survival estimates.
Shown is the incidence of death among offspring assigned to the intervention group and those assigned to the standard of care. Values were calculated with the use of Kaplan–Meier methods and compared with Mantel–Haenszel test. Panel A demonstrates the survival curve for perinatal and neonatal mortality. Panel B demonstrates infant mortality from birth until 6 months of life.

References

    1. Desyibelew HD, Dadi AF. Burden and determinants of malnutrition among pregnant women in Africa: A systematic review and meta-analysis. PLoS ONE. 2019;14(9):e0221712. Epub 2019/09/07. doi: 10.1371/journal.pone.0221712.
    1. Elshibly EM, Schmalisch G. The effect of maternal anthropometric characteristics and social factors on gestational age and birth weight in Sudanese newborn infants. BMC Public Health. 2008;8:244. Epub 2008/07/22. doi: 10.1186/1471-2458-8-244.
    1. Ververs MT, Antierens A, Sackl A, Staderini N, Captier V. Which anthropometric indicators identify a pregnant woman as acutely malnourished and predict adverse birth outcomes in the humanitarian context? PLoS Curr. 2013;5. doi: 10.1371/currents.dis.54a8b618c1bc031ea140e3f2934599c8.
    1. Hambidge KM, Krebs NF, Garces A, Westcott JE, Figueroa L, Goudar SS, et al.. Anthropometric indices for non-pregnant women of childbearing age differ widely among four low-middle income populations. BMC Public Health. 2017;18(1):45. Epub 2017/07/26. doi: 10.1186/s12889-017-4509-z.
    1. Christian P, Lee SE, Donahue Angel M, Adair LS, Arifeen SE, Ashorn P, et al.. Risk of childhood undernutrition related to small-for-gestational age and preterm birth in low- and middle-income countries. Int J Epidemiol. 2013;42(5):1340–55. Epub 2013/08/08. doi: 10.1093/ije/dyt109 .
    1. Hoddinott J, Maluccio JA, Behrman JR, Flores R, Martorell R. Effect of a nutrition intervention during early childhood on economic productivity in Guatemalan adults. Lancet. 2008;371(9610):411–6. doi: 10.1016/S0140-6736(08)60205-6 .
    1. Katz J, Lee AC, Kozuki N, Lawn JE, Cousens S, Blencowe H, et al.. Mortality risk in preterm and small-for-gestational-age infants in low-income and middle-income countries: a pooled country analysis. Lancet. 2013;382(9890):417–25. doi: 10.1016/S0140-6736(13)60993-9 .
    1. Martorell R, Horta BL, Adair LS, Stein AD, Richter L, Fall CH, et al.. Weight gain in the first two years of life is an important predictor of schooling outcomes in pooled analyses from five birth cohorts from low- and middle-income countries. J Nutr. 2010;140(2):348–54. doi: 10.3945/jn.109.112300 .
    1. Hambidge KM, Westcott JE, Garces A, Figueroa L, Goudar SS, Dhaded SM, et al.. A multicountry randomized controlled trial of comprehensive maternal nutrition supplementation initiated before conception: the Women First trial. Am J Clin Nutr. 2019;109(2):457–69. Epub 2019/02/06. doi: 10.1093/ajcn/nqy228 .
    1. Callaghan-Gillespie M, Schaffner AA, Garcia P, Fry J, Eckert R, Malek S, et al.. Trial of ready-to-use supplemental food and corn-soy blend in pregnant Malawian women with moderate malnutrition: a randomized controlled clinical trial. Am J Clin Nutr. 2017;106(4):1062–9. Epub 2017/08/11. doi: 10.3945/ajcn.117.157198 .
    1. Das JK, Hoodbhoy Z, Salam RA, Bhutta AZ, Valenzuela-Rubio NG, Weise Prinzo Z, et al.. Lipid-based nutrient supplements for maternal, birth, and infant developmental outcomes. Cochrane Database Syst Rev. 2018;8:CD012610. Epub 2018/09/01. doi: 10.1002/14651858.CD012610.pub2.
    1. Goto E. Effectiveness of Prenatal Lipid-Based Nutrient Supplementation to Improve Birth Outcomes: A Meta-analysis. Am J Trop Med Hyg. 2019;101(5):994–9. Epub 2019/09/14. doi: 10.4269/ajtmh.19-0287 .
    1. Nosten F, McGready R, Simpson JA, Thwai KL, Balkan S, Cho T, et al.. Effects of Plasmodium vivax malaria in pregnancy. Lancet. 1999;354(9178):546–9. doi: 10.1016/s0140-6736(98)09247-2 .
    1. Umbers AJ, Aitken EH, Rogerson SJ. Malaria in pregnancy: small babies, big problem. Trends Parasitol. 2011;27(4):168–75. doi: 10.1016/j.pt.2011.01.007 .
    1. World Health Organization. WHO policy brief for the implementation of intermittent preventive treatment of malaria in pregnancy using sulfadoxine-pyrimethamine. Geneva, Switzerland: WHO; 2013.
    1. Christian P, Khatry SK, West KP Jr. Antenatal anthelmintic treatment, birthweight, and infant survival in rural Nepal. Lancet. 2004;364(9438):981–3. doi: 10.1016/S0140-6736(04)17023-2 .
    1. Torlesse H, Hodges M. Anthelminthic treatment and haemoglobin concentrations during pregnancy. Lancet. 2000;356(9235):1083. doi: 10.1016/S0140-6736(00)02738-0.
    1. Torlesse H, Hodges M. Albendazole therapy and reduced decline in haemoglobin concentration during pregnancy (Sierra Leone). Trans R Soc Trop Med Hyg. 2001;95(2):195–201. doi: 10.1016/s0035-9203(01)90164-6 .
    1. Mireku MO, Boivin MJ, Davidson LL, Ouedraogo S, Koura GK, Alao MJ, et al.. Impact of helminth infection during pregnancy on cognitive and motor functions of one-year-old children. PLoS Negl Trop Dis. 2015;9. doi: 10.1371/journal.pntd.0003463
    1. Hallamaa L, Cheung YB, Luntamo M, Ashorn U, Kulmala T, Mangani C, et al.. The impact of maternal antenatal treatment with two doses of azithromycin and monthly sulphadoxine-pyrimethamine on child weight, mid-upper arm circumference and head circumference: A randomized controlled trial. PLoS ONE. 2019;14(5):e0216536. Epub 2019/05/08. doi: 10.1371/journal.pone.0216536.
    1. Luntamo M, Rantala AM, Meshnick SR, Cheung YB, Kulmala T, Maleta K, et al.. The effect of monthly sulfadoxine-pyrimethamine, alone or with azithromycin, on PCR-diagnosed malaria at delivery: a randomized controlled trial. PLoS ONE. 2012;7(7):e41123. Epub 2012/07/26. doi: 10.1371/journal.pone.0041123.
    1. Hallamaa L, Cheung YB, Maleta K, Luntamo M, Ashorn U, Gladstone M, et al.. Child Health Outcomes After Presumptive Infection Treatment in Pregnant Women: A Randomized Trial. Pediatrics. 2018. Epub 2018/02/24. doi: 10.1542/peds.2017-2459.
    1. Chico RM, Mayaud P, Ariti C, Mabey D, Ronsmans C, Chandramohan D. Prevalence of malaria and sexually transmitted and reproductive tract infections in pregnancy in sub-Saharan Africa: a systematic review. JAMA. 2012;307(19):2079–86. Epub 2012/06/06. doi: 10.1001/jama.2012.3428 .
    1. Hauth JC, Goldenberg RL, Andrews WW, DuBard MB, Copper RL. Reduced incidence of preterm delivery with metronidazole and erythromycin in women with bacterial vaginosis. N Engl J Med. 1995;333(26):1732–6. doi: 10.1056/NEJM199512283332603 .
    1. McDonald HM, O’Loughlin JA, Vigneswaran R, Jolley PT, Harvey JA, Bof A, et al.. Impact of metronidazole therapy on preterm birth in women with bacterial vaginosis flora (Gardnerella vaginalis): a randomised, placebo controlled trial. Br J Obstet Gynaecol. 1997;104(12):1391–7. doi: 10.1111/j.1471-0528.1997.tb11009.x .
    1. Hendrixson DT, Koroma AS, Callaghan-Gillespie M, Weber J, Papathakis P, Manary MJ. Use of a novel supplementary food and measures to control inflammation in malnourished pregnant women in Sierra Leone to improve birth outcomes: study protocol for a prospective, randomized, controlled clinical effectiveness trial. BMC Nutrition. 2018;4(1):15. doi: 10.1186/s40795-018-0218-y
    1. Andersson R, Bergstrom S. Use of fundal height as a proxy for length of gestation in rural Africa. J Trop Med Hyg. 1995;98(3):169–72. Epub 1995/06/01. .
    1. WHO. WHO child growth standards and the identification of severe acute malnutrition in infants and children. Geneva: World Health Organization, United Nations Children’s Fund; 2009.
    1. Olsen SF, Halldorsson TI, Willett WC, Knudsen VK, Gillman MW, Mikkelsen TB, et al.. Milk consumption during pregnancy is associated with increased infant size at birth: prospective cohort study. Am J Clin Nutr. 2007;86(4):1104–10. Epub 2007/10/09. doi: 10.1093/ajcn/86.4.1104 .
    1. Mukhopadhyay A, Dwarkanath P, Bhanji S, Devi S, Thomas A, Kurpad AV, et al.. Maternal intake of milk and milk proteins is positively associated with birth weight: A prospective observational cohort study. Clin Nutr ESPEN. 2018;25:103–9. Epub 2018/05/22. doi: 10.1016/j.clnesp.2018.03.125 .
    1. Heppe DH, van Dam RM, Willemsen SP, den Breeijen H, Raat H, Hofman A, et al.. Maternal milk consumption, fetal growth, and the risks of neonatal complications: the Generation R Study. Am J Clin Nutr. 2011;94(2):501–9. Epub 2011/06/24. doi: 10.3945/ajcn.111.013854 .
    1. Lassi ZS, Padhani ZA, Rabbani A, Rind F, Salam RA, Das JK, et al.. Impact of Dietary Interventions during Pregnancy on Maternal, Neonatal, and Child Outcomes in Low- and Middle-Income Countries. Nutrients. 2020;12(2). Epub 2020/02/26. doi: 10.3390/nu12020531
    1. Ceesay SM, Prentice AM, Cole TJ, Foord F, Weaver LT, Poskitt EM, et al.. Effects on birth weight and perinatal mortality of maternal dietary supplements in rural Gambia: 5 year randomised controlled trial. BMJ. 1997;315(7111):786–90. Epub 1997/11/05. doi: 10.1136/bmj.315.7111.786 .
    1. Oza S, Lawn JE, Hogan DR, Mathers C, Cousens SN. Neonatal cause-of-death estimates for the early and late neonatal periods for 194 countries: 2000–2013. Bull World Health Organ. 2015;93(1):19–28. Epub 2015/01/06. doi: 10.2471/BLT.14.139790 .
    1. Sweeney EL, Dando SJ, Kallapur SG, Knox CL. The Human Ureaplasma Species as Causative Agents of Chorioamnionitis. Clin Microbiol Rev. 2017;30(1):349–79. Epub 2016/12/16. doi: 10.1128/CMR.00091-16 .
    1. Government of Sierra Leone. Sierra Leone Malaria Control Strategic Plan 2016–2020. In: Sanitation MoHa, editor. Freetown, Sierra Leone; 2015.
    1. Scaglione F, Rossoni G. Comparative anti-inflammatory effects of roxithromycin, azithromycin and clarithromycin. J Antimicrob Chemother. 1998;41(Suppl B):47–50. Epub 1998/05/14. doi: 10.1093/jac/41.suppl_2.47
    1. Tamaoki J. The effects of macrolides on inflammatory cells. Chest. 2004;125(2 Suppl):41S–50S; quiz 1S. Epub 2004/02/12. doi: 10.1378/chest.125.2_suppl.41s
    1. Ragsdale HB, Kuzawa CW, Borja JB, Avila JL, McDade TW. Regulation of inflammation during gestation and birth outcomes: Inflammatory cytokine balance predicts birth weight and length. Am J Hum Biol. 2019;31(3):e23245. Epub 2019/04/14. doi: 10.1002/ajhb.23245.
    1. Wilkinson AL, Pedersen SH, Urassa M, Michael D, Andreasen A, Todd J, et al.. Maternal systemic or cord blood inflammation is associated with birth anthropometry in a Tanzanian prospective cohort. Tropical Med Int Health. 2017;22(1):52–62. Epub 2016/10/21. doi: 10.1111/tmi.12799 .
    1. Doan T, Hinterwirth A, Worden L, Arzika AM, Maliki R, Abdou A, et al.. Gut microbiome alteration in MORDOR I: a community-randomized trial of mass azithromycin distribution. Nat Med. 2019;25(9):1370–6. Epub 2019/08/14. doi: 10.1038/s41591-019-0533-0 .
    1. van den Broek NR, White SA, Goodall M, Ntonya C, Kayira E, Kafulafula G, et al.. The APPLe study: a randomized, community-based, placebo-controlled trial of azithromycin for the prevention of preterm birth, with meta-analysis. PLoS Med. 2009;6(12):e1000191. Epub 2009/12/04. doi: 10.1371/journal.pmed.1000191.
    1. Luntamo M, Kulmala T, Cheung YB, Maleta K, Ashorn P. The effect of antenatal monthly sulphadoxine-pyrimethamine, alone or with azithromycin, on foetal and neonatal growth faltering in Malawi: a randomised controlled trial. Tropical Med Int Health. 2013;18. doi: 10.1111/tmi.12074
    1. Gray RH, Wabwire-Mangen F, Kigozi G, Sewankambo NK, Serwadda D, Moulton LH, et al.. Randomized trial of presumptive sexually transmitted disease therapy during pregnancy in Rakai. Uganda Am J Obstet Gynecol. 2001;185(5):1209–17. Epub 2001/11/22. doi: 10.1067/mob.2001.118158 .
    1. Unger HW, Ome-Kaius M, Wangnapi RA, Umbers AJ, Hanieh S, Suen CS, et al.. Sulphadoxine-pyrimethamine plus azithromycin for the prevention of low birthweight in Papua New Guinea: a randomised controlled trial. BMC Med. 2015;13:9. Epub 2015/01/17. doi: 10.1186/s12916-014-0258-3.
    1. Keenan JD, Bailey RL, West SK, Arzika AM, Hart J, Weaver J, et al.. Azithromycin to Reduce Childhood Mortality in Sub-Saharan Africa. N Engl J Med. 2018;378(17):1583–92. Epub 2018/04/26. doi: 10.1056/NEJMoa1715474 .
    1. Sie A, Bountogo M, Nebie E, Ouattara M, Coulibaly B, Bagagnan C, et al.. Neonatal azithromycin administration to prevent infant mortality: study protocol for a randomised controlled trial. BMJ Open. 2019;9(9):e031162. Epub 2019/09/07. doi: 10.1136/bmjopen-2019-031162.
    1. Roca A, Oluwalana C, Bojang A, Camara B, Kampmann B, Bailey R, et al.. Oral azithromycin given during labour decreases bacterial carriage in the mothers and their offspring: a double-blind randomized trial. Clin Microbiol Infect. 2016;22(6):565 e1–9. Epub 2016/03/31. doi: 10.1016/j.cmi.2016.03.005
    1. Bojang A, Camara B, Jagne Cox I, Oluwalana C, Lette K, Usuf E, et al.. Long-term Impact of Oral Azithromycin Taken by Gambian Women During Labor on Prevalence and Antibiotic Susceptibility of Streptococcus pneumoniae and Staphylococcus aureus in Their Infants: Follow-up of a Randomized Clinical Trial. Clin Infect Dis. 2018;67(8):1191–7. Epub 2018/04/03. doi: 10.1093/cid/ciy254 .
    1. Unger HW, Aho C, Ome-Kaius M, Wangnapi RA, Umbers AJ, Jack W, et al.. Impact of intermittent preventive treatment in pregnancy with azithromycin-containing regimens on maternal nasopharyngeal carriage and antibiotic sensitivity of Streptococcus pneumoniae, Haemophilus influenzae, and Staphylococcus aureus: a cross-sectional survey at delivery. J Clin Microbiol. 2015;53(4):1317–23. Epub 2015/02/13. doi: 10.1128/JCM.03570-14 .
    1. Fan H, Gilbert R, O’Callaghan F, Li L. Associations between macrolide antibiotics prescribing during pregnancy and adverse child outcomes in the UK: population based cohort study. BMJ. 2020;368:m331. Epub 2020/02/23. doi: 10.1136/bmj.m331.
    1. Bradshaw CS, Morton AN, Hocking J, Garland SM, Morris MB, Moss LM, et al.. High recurrence rates of bacterial vaginosis over the course of 12 months after oral metronidazole therapy and factors associated with recurrence. J Infect Dis. 2006;193(11):1478–86. Epub 2006/05/03. doi: 10.1086/503780 .
    1. Myer L, Kuhn L, Denny L, Wright TC Jr. Recurrence of symptomatic bacterial vaginosis 12 months after oral metronidazole therapy in HIV-positive and -negative women. J Infect Dis. 2006;194(12):1797–9. Epub 2006/11/17. doi: 10.1086/509625 .
    1. Bradshaw CS, Vodstrcil LA, Hocking JS, Law M, Pirotta M, Garland SM, et al.. Recurrence of bacterial vaginosis is significantly associated with posttreatment sexual activities and hormonal contraceptive use. Clin Infect Dis. 2013;56(6):777–86. Epub 2012/12/18. doi: 10.1093/cid/cis1030 .
    1. Lev-Sagie A, Goldman-Wohl D, Cohen Y, Dori-Bachash M, Leshem A, Mor U, et al.. Vaginal microbiome transplantation in women with intractable bacterial vaginosis. Nat Med. 2019;25(10):1500–4. Epub 2019/10/09. doi: 10.1038/s41591-019-0600-6 .
    1. Sobel JD, Ferris D, Schwebke J, Nyirjesy P, Wiesenfeld HC, Peipert J, et al.. Suppressive antibacterial therapy with 0.75% metronidazole vaginal gel to prevent recurrent bacterial vaginosis. Am J Obstet Gynecol. 2006;194(5):1283–9. Epub 2006/05/02. doi: 10.1016/j.ajog.2005.11.041 .
    1. United Nations General Assembly. Transforming our world: the 2030 Agenda for Sustainable Development. UN General Assembly; 2015.

Source: PubMed

3
Abonneren