Effects of Mass Drug Administration of Azithromycin on Mortality and Other Outcomes Among 1-11 Month Old Infants in Mali (LAKANA)

November 28, 2023 updated by: Per Ashorn, Tampere University

LAKANA , a Cluster-randomized, Double-blinded, Parallel Group, Controlled Trial, Testing the Effects of Mass Drug Administration of Azithromycin on Mortality and Other Outcomes Among 1-11 Month Old Infants in Rural Mali.

The LAKANA trial will assess the impact on mortality and other health outcomes of quarterly and biannual azithromycin mass drug administration (MDA) when delivered to 1-11-month (29-364 days) old infants in a high-mortality setting where malaria is holoendemic but there is also a functioning seasonal malaria chemoprevention (SMC) program in place. The long-term goal is to more precisely define the role of mass azithromycin treatments as an intervention for reducing childhood mortality, and to determine the most effective treatment regimen. The main study hypotheses in terms of mortality effect are: i) Biannual azithromycin MDA to 1-11 month old infants reduces their mortality, ii) Quarterly azithromycin MDA to 1-11 month old infants reduces their mortality, iii) Quarterly azithromycin MDA has a bigger mortality effect than biannual MDA.

Study Overview

Status

Recruiting

Conditions

Intervention / Treatment

Detailed Description

Mass drug administration (MDA) of azithromycin has been shown to reduce under-5 mortality in some but not all sub-Saharan African settings. Because of the observed heterogeneity and possible effect modification by SMC or other co-interventions, further trials in new settings are needed in order to make evidence-based public health recommendations about the use of this treatment. The objectives of the LAKANA trial are:

  • To evaluate the impact of two azithromycin MDA regimens on infant mortality and other health outcomes, when provided in a rural West-African high-mortality context with an ongoing seasonal malaria chemoprevention program.
  • To evaluate the effect of alternative MDA frequencies on antimicrobial resistance (AMR) and host microbiota composition.
  • To test hypotheses that azithromycin MDA eliminates malaria parasitaemia and reduces systemic and intestinal inflammation in asymptomatic children and to collect and store biological samples for assessing other possible mechanisms of azithromycin effect.
  • To investigate the feasibility of alternative azithromycin MDA strategies, including economic analysis.

The LAKANA trial will be conducted in 1150 villages from 7-10 health districts in the Kayes, Kita and Koulikoro regions of Mali. LAKANA is a cluster-randomized, placebo-controlled, double-blinded, parallel-group, three-arm clinical trial, with adaptive design. Participating villages will be randomly allocated to three different intervention groups in a ratio of 3 : 2 : 4 (control : azithromycin quarterly : azithromycin biannually). Within each participating village, consenting households will be visited quarterly (at 3-month intervals), nine times. At the first eight of these visits, 1-11-month-old eligible infants (age 29-364 days), for whom there is a consent for study drug provision, will be given a single dose of study drug (azithromycin mixture or respective placebo mixture).

Mortality and serious adverse events (SAEs) data will be collected, and mortality-related questions answered using data from all the included 1150 villages. Mixed-effect Poisson regression model will be used to estimate the intervention effects on mortality, with random intercepts for the clusters. The investigators will explore effect modification by testing for interaction between the MDA intervention and the following variables:

  • Infant age at the time of MDA (1-5 months vs 6-11 months)
  • Infant weight-for-age at the time of MDA
  • Infant sex
  • Season of MDA dosing and time since the last SMC
  • Cluster level coverage of SMC
  • Cluster level baseline mortality (established at first census)
  • Cluster and individual level coverage and number of administered azithromycin MDA doses
  • District of residence
  • Distance from the nearest health facility
  • Household asset or income index
  • Household WASH index

The investigators will address the other study questions using a smaller separate secondary sample of 59 villages located around four selected health centers close to the city of Kita and a similar number of villages closer to Bamako, i.e. in Koulikoro or Kati (tertiary sample).

Study Type

Interventional

Enrollment (Estimated)

100000

Phase

  • Phase 3

Contacts and Locations

This section provides the contact details for those conducting the study, and information on where this study is being conducted.

Study Contact

Study Contact Backup

Study Locations

      • Bamako, Mali, BP 251
        • Recruiting
        • Center for Vaccine Development CVD-Mali
        • Contact:
        • Contact:
        • Principal Investigator:
          • Samba Sow, MD, MSc

Participation Criteria

Researchers look for people who fit a certain description, called eligibility criteria. Some examples of these criteria are a person's general health condition or prior treatments.

Eligibility Criteria

Ages Eligible for Study

1 year to 4 months (Child)

Accepts Healthy Volunteers

Yes

Description

Inclusion Criteria:

On a cluster (village) level:

  1. Location within Kayes, Kita, or Koulikoro region of Mali
  2. Considered accessible and safe by the local health authorities and research team
  3. Considered non-urban by the local health authorities and research team
  4. Permission from community leadership

On a household level (for trial enrollment):

  1. Location within a cluster that is included in the study
  2. Verbal consent from a head of household or an adult authorized by her / him

On a child level (for receiving study medication):

  1. Residence in a household enrolled in the trial
  2. Age between 29 and 364 days
  3. Verbal consent from at least one caregiver

Exclusion Criteria:

On child level (for not receiving study medication):

  1. Weight below 3.0 kg
  2. Known allergy to macrolides, as judged by a caregiver report of the infant experiencing an adverse reaction after oral ingestion of medication, which was deemed likely to be a macrolide by the interviewing data collector.

Study Plan

This section provides details of the study plan, including how the study is designed and what the study is measuring.

How is the study designed?

Design Details

  • Primary Purpose: Treatment
  • Allocation: Randomized
  • Interventional Model: Parallel Assignment
  • Masking: Quadruple

Arms and Interventions

Participant Group / Arm
Intervention / Treatment
Placebo Comparator: Control

Placebo mixture will be administered as a single dose in oral suspension form for children 1-11 months of age:

  1. Single-dose of 0.5 ml / kg child weight
  2. Participating households within villages allocated to this group will be visited quarterly (at 3-month intervals), for nine times. At the first eight of these visits, 1-11 month old eligible infants, for whom there is a consent for study drug provision, will be weighed and given a single dose of placebo mixture.
Placebo mixture will be administered as a single dose in oral suspension form for children 1-11 months of age. Weight-based dosing will be used: single-dose of 0.5 ml / kg child weight.
Active Comparator: Azithromycin-biannually (Azi-biannual)

Azithromycin or placebo will be administered as a single dose in oral suspension form for children 1-11 months of age:

  1. Single-dose of 0.5 ml / kg child weight
  2. Participating households within villages allocated to this group will be visited quarterly (at 3-month intervals), for nine times. At the first eight of these visits, 1-11 month old eligible infants, for whom there is a consent for study drug provision, will be weighed and given a single dose of study drug.
  3. Azithromycin will be given at quarterly visits between January and June, and Placebo mixture will be given at quarterly visits between July and December. Azithromycin dose will be 20 mg / kg.
Placebo mixture will be administered as a single dose in oral suspension form for children 1-11 months of age. Weight-based dosing will be used: single-dose of 0.5 ml / kg child weight.
Azithromycin will be administered as a single dose in oral suspension form for children 1-11 months of age. Weight-based dosing will be used: single-dose of 0.5 ml (20 mg) / kg child weight.
Active Comparator: Azithromycin-quarterly

Azithromycin will be administered as a single dose in oral suspension form for children 1-11 months of age:

  1. Single-dose of 0.5 ml (20 mg) / kg child weight.
  2. Participating households within villages allocated to this group will be visited quarterly (at 3-month intervals), for nine times. At the first eight of these visits, 1-11 month old eligible infants, for whom there is a consent for study drug provision, will be weighed and given a single dose of azithromycin.
Azithromycin will be administered as a single dose in oral suspension form for children 1-11 months of age. Weight-based dosing will be used: single-dose of 0.5 ml (20 mg) / kg child weight.

What is the study measuring?

Primary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Mortality
Time Frame: 3-month time interval (total of 8 intervals per cluster)
Mortality rate (deaths per 1,000 years at risk) among children 1-11 months of age.
3-month time interval (total of 8 intervals per cluster)

Secondary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Morbidity
Time Frame: 3-month time interval (total of 8 intervals per cluster)
Morbidity (14-day period prevalence of fever with respiratory symptoms (ARI), fever without respiratory symptoms (malaria), and diarrhea) in children aged 4-14 months assessed in each participating cluster (village) from the secondary outcome sample.
3-month time interval (total of 8 intervals per cluster)
Length-for-age Z-score
Time Frame: 3-month time interval (total of 3 intervals per cluster)
Length-for-age Z-score in 6-8 and 12-14 months old children from the secondary outcome sample. Length measures will be taken at 15, 18, and 21 months after the enrollment of the village into the trial. Length-for-age Z-score will be calculated using the WHO Child Growth Standards.
3-month time interval (total of 3 intervals per cluster)
Weight-for age Z-score
Time Frame: 3-month time interval (total of 3 intervals per cluster)
Weight-for age Z-score in 6-8 and 12-14 months old children from the secondary outcome sample. Weight measures will be taken at 15, 18, and 21 months after the enrollment of the village into the trial. Weight-for-age Z-score will be calculated using the WHO Child Growth Standards.
3-month time interval (total of 3 intervals per cluster)
Weight-for-length Z-score
Time Frame: 3-month time interval (total of 3 intervals per cluster)
Weight-for-length Z-score in 6-8 and 12-14 months old children from the secondary outcome sample. Length and weight measures will be taken at 15, 18, and 21 months after the enrollment of the village into the trial. Weight-for-length Z score will be calculated using the WHO Child Growth Standards.
3-month time interval (total of 3 intervals per cluster)
Mid-upper arm circumference
Time Frame: 3-month time interval (total of 3 intervals per cluster)
Mid-upper arm circumference in 6-8 and 12-14 months old children from the secondary outcome sample. Measures will be taken at 15, 18, and 21 months after the enrollment of the village into the trial.
3-month time interval (total of 3 intervals per cluster)
Percentage of moderate or severe stunting
Time Frame: 3-month time interval (total of 3 intervals per cluster)
Percentage of moderate or severe stunting (length-for-age Z-score < -2 / -3) in 6-8 and 12-14 months old children from the secondary outcome sample.
3-month time interval (total of 3 intervals per cluster)
Percentage of moderate or severe wasting
Time Frame: 3-month time interval (total of 3 intervals per cluster)
Percentage of moderate or severe wasting (Weight-for-length Z-score < -2 / -3) in 6-8 and 12-14 months old children from the secondary outcome sample.
3-month time interval (total of 3 intervals per cluster)
Prevalence of phenotypic and genotypic macrolide resistance
Time Frame: 12-month time interval (total of 3 intervals per cluster)

Prevalence of phenotypic and genotypic macrolide resistance among E. coli strains isolated from stool samples or S. pneumoniae strains isolated from nasopharyngeal swabs among among 4-14-month-old children, at one year after the MDA intervention has stopped (i.e., 36 months after MDA 1, the baseline).

Genetic markers of azithromycin and other antibiotic resistance (resistome) of intestinal microbiota among 4-14-month-old children, at one year after the MDA intervention has stopped (i.e., 36 months after MDA 1, the baseline).

(secondary outcome sample).

12-month time interval (total of 3 intervals per cluster)
Prevalence of phenotypic and genotypic macrolide resistance
Time Frame: 12-month time interval (total of 3 intervals per cluster)
Prevalence of phenotypic and genotypic macrolide resistance among E. coli strains isolated from stool samples or S. pneumoniae strains isolated from nasopharyngeal swabs among 49-59-month-old children, at 24 months after village enrolment to the trial, i.e., after 8 rounds of MDA. (secondary outcome sample).
12-month time interval (total of 3 intervals per cluster)
Prevalence of phenotypic and genotypic AMR resistance to other "ACCESS" group antibiotics
Time Frame: 12-month time interval (total of 3 intervals per cluster)
Prevalence of phenotypic and genotypic AMR against other antibiotics categorised by the World Health Organization (WHO) into "ACCESS" group, among azithromycin-resistant E. coli strains isolated from stool samples or azithromycin-resistant S. pneumoniae strains isolated from nasopharyngeal swabs (secondary outcome sample).
12-month time interval (total of 3 intervals per cluster)
Blood C-reactive protein concentration
Time Frame: Biological samples taken before and 14 days after the fourth MDA round, 9 months after enrollment.
Biological samples taken from 4-11 months old infants to assess effect of azithromycin MDA on systemic inflammation (blood C-reactive protein concentration) (secondary outcome sample).
Biological samples taken before and 14 days after the fourth MDA round, 9 months after enrollment.
Blood malaria parasitemia and hemoglobin concentration
Time Frame: Biological samples taken before and 14 days after the fourth MDA round, 9 months after enrollment.
Biological samples taken from 4-11 months old infants to assess effect of azithromycin MDA on malaria prevalence and blood hemoglobin concentration (secondary outcome sample).
Biological samples taken before and 14 days after the fourth MDA round, 9 months after enrollment.
Fecal neopterin, myeloperoxidase, and alpha-1-antitrypsin concentrations
Time Frame: Biological samples taken before and 14 days after the fourth MDA round, 9 months after enrollment.
Biological samples taken from 4-11 months old infants to assess effect of azithromycin MDA on intestinal inflammation (secondary outcome sample).
Biological samples taken before and 14 days after the fourth MDA round, 9 months after enrollment.
Incidence of Adverse events
Time Frame: 3-month time interval
Incidence of adverse events within 14 days of study drug administration. Data collected from 4-11 months old infants at 9 months after enrollment (secondary outcome sample).
3-month time interval
Incidence of Serious Adverse events
Time Frame: within 14 days after MDA round.
Incidence of serious adverse events (Death, life-threatening event, hospitalization, and other serious events as judged by a study physician) within 14 days of study drug administration, among 1-11 months old infants.
within 14 days after MDA round.
Mortality in children aged 12-59 month
Time Frame: 3-month time interval (total of 8 intervals per cluster)
Mortality rate (deaths per 1000 years at risk) among children who were 12-59 month old when the latest azithromycin MDA took place in their village of residence.
3-month time interval (total of 8 intervals per cluster)
Percentage of guardians and health care workers reporting MDA acceptable
Time Frame: 24 months after enrollment
Data collected through interviews of guardians and health care workers to assess acceptability of MDA.
24 months after enrollment
Percentage of the study population reached with MDA
Time Frame: 24 months after enrollment
Data collected on MDA distribution and through interviews of guardians and health care workers to assess equity of MDA.
24 months after enrollment
Cost and Cost-effectiveness of the intervention
Time Frame: 24 months after enrollment
Data collected on MDA distribution to assess the economic aspects of MDA.The effectiveness will be determined by the trial outcome - both the primary outcome of mortality and the secondary outcomes of morbidity and possible AMR effects. The effectiveness will be expressed in different units such as deaths averted, or disability adjusted life years (DALYs).
24 months after enrollment

Collaborators and Investigators

This is where you will find people and organizations involved with this study.

Investigators

  • Principal Investigator: Per Ashorn, MD, PhD, Center for Child Health Research, Tampere University
  • Principal Investigator: Ulla Ashorn, PhD, Center for Child Health Research, Tampere University
  • Principal Investigator: Samba Sow, MD, MSc, Center for Vaccine Development CVD-Mali
  • Principal Investigator: Nigel Klein, MBBS, PhD, University College London Hospitals
  • Principal Investigator: Camilla Ducker, MBBS, MSc, Tro Da Ltd, UK
  • Principal Investigator: Yin Bun Cheung, PhD, Centre for Quantitative Medicine, Duke-NUS Medical School

Study record dates

These dates track the progress of study record and summary results submissions to ClinicalTrials.gov. Study records and reported results are reviewed by the National Library of Medicine (NLM) to make sure they meet specific quality control standards before being posted on the public website.

Study Major Dates

Study Start (Actual)

October 15, 2020

Primary Completion (Estimated)

December 31, 2024

Study Completion (Estimated)

December 31, 2024

Study Registration Dates

First Submitted

May 22, 2020

First Submitted That Met QC Criteria

June 5, 2020

First Posted (Actual)

June 11, 2020

Study Record Updates

Last Update Posted (Actual)

November 29, 2023

Last Update Submitted That Met QC Criteria

November 28, 2023

Last Verified

November 1, 2023

More Information

Terms related to this study

Plan for Individual participant data (IPD)

Plan to Share Individual Participant Data (IPD)?

YES

IPD Plan Description

Individual participant data collected during the trial, after deidentification. (The details are to be determined).

IPD Sharing Time Frame

Starting 6 months after publication

IPD Sharing Access Criteria

(The details are to be determined)

IPD Sharing Supporting Information Type

  • STUDY_PROTOCOL
  • SAP
  • ICF
  • CSR

Drug and device information, study documents

Studies a U.S. FDA-regulated drug product

No

Studies a U.S. FDA-regulated device product

No

product manufactured in and exported from the U.S.

No

This information was retrieved directly from the website clinicaltrials.gov without any changes. If you have any requests to change, remove or update your study details, please contact register@clinicaltrials.gov. As soon as a change is implemented on clinicaltrials.gov, this will be updated automatically on our website as well.

Clinical Trials on Mortality

Clinical Trials on Placebo

3
Subscribe