Daily co-trimoxazole prophylaxis to prevent mortality in children with complicated severe acute malnutrition: a multicentre, double-blind, randomised placebo-controlled trial

James A Berkley, Moses Ngari, Johnstone Thitiri, Laura Mwalekwa, Molline Timbwa, Fauzat Hamid, Rehema Ali, Jimmy Shangala, Neema Mturi, Kelsey D J Jones, Hassan Alphan, Beatrice Mutai, Victor Bandika, Twahir Hemed, Ken Awuondo, Susan Morpeth, Samuel Kariuki, Gregory Fegan, James A Berkley, Moses Ngari, Johnstone Thitiri, Laura Mwalekwa, Molline Timbwa, Fauzat Hamid, Rehema Ali, Jimmy Shangala, Neema Mturi, Kelsey D J Jones, Hassan Alphan, Beatrice Mutai, Victor Bandika, Twahir Hemed, Ken Awuondo, Susan Morpeth, Samuel Kariuki, Gregory Fegan

Abstract

Background: Children with complicated severe acute malnutrition (SAM) have a greatly increased risk of mortality from infections while in hospital and after discharge. In HIV-infected children, mortality and admission to hospital are prevented by daily co-trimoxazole prophylaxis, despite locally reported bacterial resistance to co-trimoxazole. We aimed to assess the efficacy of daily co-trimoxazole prophylaxis on survival in children without HIV being treated for complicated SAM.

Methods: We did a multicentre, double-blind, randomised, placebo-controlled study in four hospitals in Kenya (two rural hospitals in Kilifi and Malindi, and two urban hospitals in Mombasa and Nairobi) with children aged 60 days to 59 months without HIV admitted to hospital and diagnosed with SAM. We randomly assigned eligible participants (1:1) to 6 months of either daily oral co-trimoxazole prophylaxis (given as water-dispersible tablets; 120 mg per day for age <6 months, 240 mg per day for age 6 months to 5 years) or matching placebo. Assignment was done with computer-generated randomisation in permuted blocks of 20, stratified by centre and age younger or older than 6 months. Treatment allocation was concealed in opaque, sealed envelopes and patients, their families, and all trial staff were masked to treatment assignment. Children were given recommended medical care and feeding, and followed up for 12 months. The primary endpoint was mortality, assessed each month for the first 6 months, then every 2 months for the second 6 months. Secondary endpoints were nutritional recovery, readmission to hospital, and illness episodes treated as an outpatient. Analysis was by intention to treat. This trial was registered at ClinicalTrials.gov, number NCT00934492.

Findings: Between Nov 20, 2009, and March 14, 2013, we recruited and assigned 1778 eligible children to treatment (887 to co-trimoxazole prophylaxis and 891 to placebo). Median age was 11 months (IQR 7-16 months), 306 (17%) were younger than 6 months, 300 (17%) had oedematous malnutrition (kwashiorkor), and 1221 (69%) were stunted (length-for-age Z score <-2). During 1527 child-years of observation, 122 (14%) of 887 children in the co-trimoxazole group died, compared with 135 (15%) of 891 in the placebo group (unadjusted hazard ratio [HR] 0·90, 95% CI 0·71-1·16, p=0·429; 16·0 vs 17·7 events per 100 child-years observed (CYO); difference -1·7 events per 100 CYO, 95% CI -5·8 to 2·4]). In the first 6 months of the study (while participants received study medication), 63 suspected grade 3 or 4 associated adverse events were recorded among 57 (3%) children; 31 (2%) in the co-trimoxazole group and 32 (2%) in the placebo group (incidence rate ratio 0·98, 95% CI 0·58-1·65). The most common adverse events of these grades were urticarial rash (grade 3, equally common in both groups), neutropenia (grade 4, more common in the co-trimoxazole group), and anaemia (both grades equally common in both groups). One child in the placebo group had fatal toxic epidermal necrolysis with concurrent Pseudomonas aeruginosa bacteraemia.

Interpretation: Daily co-trimoxazole prophylaxis did not reduce mortality in children with complicated SAM without HIV. Other strategies need to be tested in clinical trials to reduce deaths in this population.

Funding: Wellcome Trust, UK.

Copyright © 2016 Berkley et al. Open Access article distributed under the terms of CC BY. Published by Elsevier Ltd.. All rights reserved.

Figures

Figure 1
Figure 1
Trial profile *Includes severe congenital or acquired heart disease, malignancy, or probably terminal illness.
Figure 2
Figure 2
Cumulative hazard curves for time to death Kaplan-Meier curves are shown for time to death until 365 days. The time at risk observed was 763·3 and 763·3 child-years in the co-trimoxazole group and placebo group, respectively, of which 395·3 and 398·7 child-years, respectively, were during the first 6 months while receiving the study drug. HR=hazard ratio.
Figure 3
Figure 3
Anthropometry during follow-up Data are means; error bars indicate 95% CI. No individual timepoint comparisons between randomised groups were significant (appendix). MUAC=mid-upper-arm circumference.

References

    1. Bhutta ZA, Das JK, Rizvi A. Evidence-based interventions for improvement of maternal and child nutrition: what can be done and at what cost? Lancet. 2013;382:452–477.
    1. Trehan I, Maleta KM, Manary MJ. Antibiotics for uncomplicated severe malnutrition. N Engl J Med. 2013;368:2436–2437.
    1. Isanaka S, Langendorf C, Berthe F. Routine amoxicillin for uncomplicated severe acute malnutrition in children. N Engl J Med. 2016;374:444–453.
    1. WHO . Guideline: updates on the management of severe acute malnutrition in infants and children. World Health Organization; Geneva: 2013.
    1. WHO Pocket book for hospital care of children: guidelines for the management of common illness with limited resources. 2005. (accessed March 9, 2015).
    1. Bachou H, Tumwine JK, Mwadime RK, Ahmed T, Tylleskar T. Reduction of unnecessary transfusion and intravenous fluids in severely malnourished children is not enough to reduce mortality. Ann Trop Paediatr. 2008;28:23–33.
    1. Brewster DR. Inpatient management of severe malnutrition: time for a change in protocol and practice. Ann Trop Paediatr. 2011;31:97–107.
    1. Kerac M, Bunn J, Chagaluka G. Follow-up of post-discharge growth and mortality after treatment for severe acute malnutrition (FuSAM study): a prospective cohort study. PLoS One. 2014;9:e96030.
    1. Chisti MJ, Graham SM, Duke T. Post-discharge mortality in children with severe malnutrition and pneumonia in Bangladesh. PLoS One. 2014;9:e107663.
    1. Wiens MO, Pawluk S, Kissoon N. Pediatric post-discharge mortality in resource poor countries: a systematic review. PLoS One. 2013;8:e66698.
    1. Moisi JC, Gatakaa H, Berkley JA. Excess child mortality after discharge from hospital in Kilifi, Kenya: a retrospective cohort analysis. Bull World Health Organ. 2011;89:725–732. 732A.
    1. Chintu C, Bhat GJ, Walker AS. Co-trimoxazole as prophylaxis against opportunistic infections in HIV-infected Zambian children (CHAP): a double-blind randomised placebo-controlled trial. Lancet. 2004;364:1865–1871.
    1. Bwakura-Dangarembizi M, Kendall L, Bakeera-Kitaka S. A randomized trial of prolonged co-trimoxazole in HIV-infected children in Africa. N Engl J Med. 2014;370:41–53.
    1. Church JA, Fitzgerald F, Walker AS, Gibb DM, Prendergast AJ. The expanding role of co-trimoxazole in developing countries. Lancet Infect Dis. 2015;15:327–339.
    1. Craig JC, Simpson JM, Williams GJ. Antibiotic prophylaxis and recurrent urinary tract infection in children. N Engl J Med. 2009;361:1748–1759.
    1. Garly ML, Bale C, Martins CL. Prophylactic antibiotics to prevent pneumonia and other complications after measles: community based randomised double blind placebo controlled trial in Guinea-Bissau. BMJ. 2006;333:1245.
    1. Aguilar C, Malphettes M, Donadieu J. Prevention of infections during primary immunodeficiency. Clin Infect Dis. 2014;59:1462–1470.
    1. Gough EK, Moodie EE, Prendergast AJ. The impact of antibiotics on growth in children in low and middle income countries: systematic review and meta-analysis of randomised controlled trials. BMJ. 2014;348:g2267.
    1. Bhutta ZA. Antibiotics to promote growth in children? BMJ. 2014;348:g2624.
    1. WHO Guidelines of co-trimoxazole prophylaxis for HIV-related infections among children, adolescents and adults. 2006. (accessed April 21, 2015).
    1. Kenya National Bureau of Statistics Kenya Demographic and Health Survey. 2014. (accessed March 20, 2016).
    1. Dow A, Kayira D, Hudgens M. Effects of cotrimoxazole prophylactic treatment on adverse health outcomes among HIV-exposed, uninfected infants. Pediatr Infect Dis J. 2012;31:842–847.
    1. Sandison TG, Homsy J, Arinaitwe E. Protective efficacy of co-trimoxazole prophylaxis against malaria in HIV exposed children in rural Uganda: a randomised clinical trial. BMJ. 2011;342:d1617.
    1. Mbeye NM, Ter Kuile FO, Davies MA. Cotrimoxazole prophylactic treatment prevents malaria in children in sub-Saharan Africa: systematic review and meta-analysis. Trop Med Int Health. 2014;19:1057–1067.
    1. Coutsoudis A, Kindra G, Esterhuizen T. Impact of cotrimoxazole prophylaxis on the health of breast-fed, HIV-exposed, HIV-negative infants in a resource-limited setting. AIDS. 2011;25:1797–1799.
    1. Brent AJ, Ahmed I, Ndiritu M. Incidence of clinically significant bacteraemia in children who present to hospital in Kenya: community-based observational study. Lancet. 2006;367:482–488.
    1. Rudan I, O'Brien KL, Nair H. Epidemiology and etiology of childhood pneumonia in 2010: estimates of incidence, severe morbidity, mortality, underlying risk factors and causative pathogens for 192 countries. J Glob Health. 2013;3:010401.
    1. D'Acremont V, Kilowoko M, Kyungu E. Beyond malaria—causes of fever in outpatient Tanzanian children. N Engl J Med. 2014;370:809–817.
    1. Berkley JA, Munywoki P, Ngama M. Viral etiology of severe pneumonia among Kenyan infants and children. JAMA. 2010;303:2051–2057.
    1. Porco TC, Gebre T, Ayele B. Effect of mass distribution of azithromycin for trachoma control on overall mortality in Ethiopian children: a randomized trial. JAMA. 2009;302:962–968.
    1. Coles CL, Levens J, Seidman JC, Mkocha H, Munoz B, West S. Mass distribution of azithromycin for trachoma control is associated with short-term reduction in risk of acute lower respiratory infection in young children. Pediatr Infect Dis J. 2012;31:341–346.
    1. Gilliams EA, Jumare J, Claassen CW. Chloroquine-azithromycin combination antimalarial treatment decreases risk of respiratory- and gastrointestinal-tract infections in Malawian children. J Infect Dis. 2014;210:585–592.
    1. Taylor WR, Richie TL, Fryauff DJ. Malaria prophylaxis using azithromycin: a double-blind, placebo-controlled trial in Irian Jaya, Indonesia. Clin Infect Dis. 1999;28:74–81.
    1. Korpe PS, Petri WA., Jr Environmental enteropathy: critical implications of a poorly understood condition. Trends Mol Med. 2012;18:328–336.
    1. Kerac M, Blencowe H, Grijalva-Eternod C. Prevalence of wasting among under 6-month-old infants in developing countries and implications of new case definitions using WHO growth standards: a secondary data analysis. Arch Dis Child. 2011;96:1008–1013.

Source: PubMed

3
Iratkozz fel