Sulfadoxine-pyrimethamine Plus Dihydroartemisinin-piperaquine for Intermittent Preventive Treatment in Pregnancy (SAPOT)

October 19, 2023 updated by: Menzies School of Health Research

Intermittent Preventive Treatment in Pregnancy With Sulfadoxine-pyrimethamine Plus Dihydroartemisinin-piperaquine to Prevent Malaria Infection and Reduce Adverse Pregnancy Outcomes in Papua New Guinea - a Randomised Controlled Trial

This trial tests the hypothesis that intermittent preventive treatment in pregnancy (IPTp) with sulfadoxine-pyrimethamine (SP) plus dihydroartemisinin-piperaquine (DP) significantly reduces the risk of malaria infection (primary outcome) and adverse birth outcomes (key secondary outcome) in an endemic area of Papua New Guinea (PNG), compared to IPTp with SP alone (the current standard of care).

To test this hypothesis a double-blinded, placebo-controlled, phase-III, superiority trial will individually randomize 1,172 HIV-uninfected pregnant women enrolled from 12-26 gestational weeks in equal proportions to one of two IPTp arms: 1) SP given every for weeks, or 2) SP+DP given every 4 weeks. DP placebos will be used to ensure adequate blinding is achieved in the study and follow-up will end 28 days after giving birth.

Study Overview

Detailed Description

Plasmodium falciparum and P. vivax infections cause malaria, maternal anemia and interfere with the development of the fetus, thereby increasing the risks of adverse pregnancy outcomes such as miscarriage, stillbirth, premature birth, fetal growth restriction, low birth weight, and infant death. Infected pregnant women are frequently asymptomatic, and current point-of-care tests miss placental and low-density infections. Monthly intermittent preventive treatment in pregnancy (IPTp) with sulfadoxine-pyrimethamine (SP) is designed to clear asymptomatic infections and provide post-treatment prophylaxis. The World Health Organization recommends IPTp with SP and long-lasting insecticidal bed nets for the prevention of malaria in pregnancy in endemic areas of sub-Saharan Africa. However, the emergence and spread of high-grade parasite resistance to SP threatens to compromise this strategy. Dihydroartemisinin-piperaquine (DP) is a safe fixed-dose artemisinin-based combination therapy used for the management of uncomplicated P. falciparum and P. vivax malaria in pregnancy and has emerged as a potential candidate to replace SP for IPTp. In comparative trials conducted in high-transmission settings in sub-Saharan Africa IPTp with DP was safe and significantly reduced the risk of P. falciparum infection compared to IPTp with SP. IPTp with DP also reduced the risk of P. vivax parasitemia in Papua Indonesia when compared to a single screen and treat approach. However, DP's superior antimalarial efficacy in African studies did not translate to large reductions in adverse pregnancy outcomes in these trials. This suggests that SP, whilst failing as an antimalarial, may prevent adverse pregnancy events via potent non-malarial effects that are not inherent to DP. For example, SP may provide protection from pathogens other than malaria parasites that are directly or indirectly involved in the causation of adverse pregnancy outcomes.

Papua New Guinea (PNG) is characterized by moderate intensity co-transmission of P. falciparum and P. vivax and a high burden of adverse pregnancy outcomes. PNG is the only country outside of Africa that has a policy of IPTp with SP. However, P. vivax resistance to SP is now common, high-grade P. falciparum resistance to SP may be emerging, and DP could provide enhanced antimalarial protection. However, given the high burden of adverse pregnancy outcomes from malaria- and non-malaria related causes, simply replacing SP with DP for IPTp in PNG may not lead to a reduction in adverse birth outcomes. Instead, combining DP with SP for IPTp has the potential to substantially improve health outcomes by reducing the risk of malaria infection whilst harnessing the non-malaria-related benefits of SP.

A double-blinded randomized controlled clinical trial will (1) compare the risk of malaria infection among pregnant women randomized to receive monthly IPTp with SP vs. SP+DP; (2) compare the risk of adverse pregnancy outcomes among pregnant women randomized to receive monthly IPTp with SP vs. SP+DP; and (3) compare safety and tolerability of monthly IPTp with SP vs SP+DP. The findings of this trial may have important policy implications, and the evidence generated will inform practice for PNG and sub-Saharan Africa.

Study Type

Interventional

Enrollment (Estimated)

1172

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

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

16 years and older (Child, Adult, Older Adult)

Accepts Healthy Volunteers

Yes

Description

Inclusion Criteria:

  • Pregnant women between 12-26 weeks' gestation
  • 16 years of age or older
  • Viable singleton intrauterine pregnancy
  • Permanent resident of the study area
  • Willing to adhere to scheduled and unscheduled study visit procedures
  • Willing to birth in a study clinic or hospital
  • Able to provide written informed consent

Exclusion Criteria:

  • Multiple pregnancy (i.e. twins/triplets)
  • Known heart ailment or other chronic medical condition requiring frequent hospital care
  • Active medical problem requiring inpatient evaluation at the time of screening
  • Severe malformations or non-viable pregnancy if observed by ultrasound
  • Antimalarial therapy in the prior two weeks
  • Unable to provide written informed consent
  • Known allergy or contraindication to any of the study drugs
  • Early or active labour
  • Known HIV-positive status

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: Prevention
  • Allocation: Randomized
  • Interventional Model: Parallel Assignment
  • Masking: Quadruple

Arms and Interventions

Participant Group / Arm
Intervention / Treatment
Active Comparator: SP + DP placebo every 4 weeks
Control arm
SP (G-COSPE) will be supplied by Fosun Pharma, China. SP will be given as a single dose consisting of three 500mg/25mg tablets.
Other Names:
  • G-COSPE
Experimental: SP + DP given every 4 weeks
Intervention arm
SP (G-COSPE) will be supplied by Fosun Pharma, China. SP will be given as a single dose consisting of three 500mg/25mg tablets.
Other Names:
  • G-COSPE
DP (D-Artepp) will be supplied by Fosun Pharma, China. DP will consist of three 40mg/320mg) tablets given once a day for three consecutive days
Other Names:
  • D-Artepp

What is the study measuring?

Primary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Malaria infection in pregnancy
Time Frame: Starting two weeks after initial dose until and including delivery

'Malaria infection in pregnancy' is a composite outcome, defined as one or more episode of P. falciparum and/or P. vivax infection, detected by microscopy and/or qPCR in peripheral blood or placental blood, or P. falciparum and/or P. vivax infection, detected as active infection on placental histology. The surveillance period will run from two weeks after the first dose of the first monthly treatment up until and including delivery (numerator) in women who attend at least one scheduled or unscheduled visit during the surveillance period (denominator).

Proportion of women with 'malaria infection in pregnancy'

Starting two weeks after initial dose until and including delivery

Secondary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Other SAEs and AEs
Time Frame: 8 months from randomisation
Incidence of AEs and SAEs
8 months from randomisation
Dizziness
Time Frame: 6 months from randomisation
Prevalence of dizziness after a course of IP
6 months from randomisation
Gastrointestinal complaints
Time Frame: 6 months from randomisation
Prevalence of gastrointestinal complaints after a course of IP
6 months from randomisation
Adverse pregnancy outcome
Time Frame: Time of delivery up to 28 days postpartum

Composite adverse birth outcome is defined as the occurrence of any of the following:

  • Spontaneous miscarriage: Fetal loss <28 weeks of gestational age
  • Stillbirth: Infant born deceased at ≥28 weeks of gestational age
  • Low birth weight (LBW): Live birth with birth weight <2,500 grams
  • Preterm birth (PTB): Live birth <37 weeks gestational age
  • Small-for-gestational age (SGA): Live birth with birth weight-for-gestational-age <10th percentile of the INTERGROWTH-21st reference
  • Neonatal death: Live birth with neonatal death within the first 28 days of life

Prevalence of adverse pregnancy outcome

Time of delivery up to 28 days postpartum
Clinical malaria during pregnancy
Time Frame: Starting two weeks after initial dose until and including delivery
Incidence of new episodes of fever or history of fever plus positive RDT confirmed by microscopy and/or qPCR during pregnancy
Starting two weeks after initial dose until and including delivery
Parasitemia during pregnancy
Time Frame: Starting two weeks after initial dose until and including delivery
Proportion of samples with parasites detected in maternal peripheral blood samples by microscopy or qPCR
Starting two weeks after initial dose until and including delivery
Composite placental malaria detected by microscopy, qPCR or by histology
Time Frame: At time of delivery
Prevalence of placental parasites by microscopy, qPCR, or placental histology
At time of delivery
Placental malaria detected by microscopy
Time Frame: At time of delivery
Prevalence of parasites in placental blood by microscopy
At time of delivery
Placental malaria detected by qPCR
Time Frame: At time of delivery
Prevalence of parasites in placental blood by qPCR
At time of delivery
Active placental malaria detected by histology
Time Frame: At time of delivery
Prevalence of active infection (presence of parasites) on histology
At time of delivery
Past placental malaria detected by histology
Time Frame: At time of delivery
Prevalence of past infection (pigment only) on histology
At time of delivery
Placental malaria detected by histology
Time Frame: At time of delivery
Prevalence of placental infection (active or past) on histology
At time of delivery
Composite fetal loss and neonatal death
Time Frame: Time of delivery up to 28 days postpartum
Prevalence of fetal loss (spontaneous miscarriage or stillbirth) and neonatal death
Time of delivery up to 28 days postpartum
Composite of SGA-LBW-PTB
Time Frame: At time of delivery
Prevalence of small for gestational age, low birth weight, and preterm birth
At time of delivery
SGA
Time Frame: At time of delivery
Prevalence of small for gestational age using the new Intergrowth-21st population reference's 10th centile
At time of delivery
LBW
Time Frame: At time of delivery
Prevalence of low birth weight
At time of delivery
PTB
Time Frame: At time of delivery
Prevalence of preterm birth
At time of delivery
Birth weight
Time Frame: At time of delivery
Mean birthweight
At time of delivery
Neonatal length
Time Frame: At time of delivery
Neonatal length
At time of delivery
Maternal nutritional status
Time Frame: 8 months from randomisation
Changes in maternal body mass index (BMI)
8 months from randomisation
Maternal nutritional status
Time Frame: 6 months from randomisation
Changes in maternal mid-upper arm circumference (MUAC)
6 months from randomisation
Maternal anemia during pregnancy and at delivery
Time Frame: 6 months from randomisation
Proportion of routine haemoglobin measurements <100 g/L
6 months from randomisation
Maternal hemoglobin levels during pregnancy and at delivery
Time Frame: 6 months from randomisation
Mean hemoglobin (g/L) at the third trimester antenatal visit and at delivery
6 months from randomisation
Congenital anemia
Time Frame: At delivery
Prevalence of anaemia (Hb <130 g/L) from newborn cord blood
At delivery
Maternal gametocyte carriage during pregnancy and at delivery
Time Frame: 6 months from randomisation
Proportion of P. falciparum positive samples with gametocytes at the third trimester antenatal visit and at delivery, by light microscopy and RT-qPCR
6 months from randomisation
Molecular markers of DP resistance
Time Frame: 6 months from randomisation
Proportion of parasite positive samples with molecular markers of DP resistance
6 months from randomisation
Molecular markers of SP drug resistance
Time Frame: 6 months from randomisation
Proportion of parasite positive samples with molecular markers of SP resistance
6 months from randomisation
Maternal mortality
Time Frame: 8 months from randomisation
he death of a woman while pregnant or within 42 days of the end of pregnancy, irrespective of the duration and site of the pregnancy, but not from accidental or incidental causes
8 months from randomisation
Congenital malformations
Time Frame: 8 months from randomisation
Any visible external congenital abnormality on surface examination
8 months from randomisation
(History) of vomiting study drug
Time Frame: 6 months from randomisation
Prevalence of vomiting investigational product (IP) twice at the same IP administration visit
6 months from randomisation

Collaborators and Investigators

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

Investigators

  • Principal Investigator: Holger Unger, PhD MBChB, Menzies School of Health Research, Darwin, Australia

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)

August 31, 2022

Primary Completion (Estimated)

February 1, 2025

Study Completion (Estimated)

December 1, 2025

Study Registration Dates

First Submitted

June 14, 2022

First Submitted That Met QC Criteria

June 16, 2022

First Posted (Actual)

June 22, 2022

Study Record Updates

Last Update Posted (Actual)

October 23, 2023

Last Update Submitted That Met QC Criteria

October 19, 2023

Last Verified

October 1, 2023

More Information

Terms related to this study

Plan for Individual participant data (IPD)

Plan to Share Individual Participant Data (IPD)?

NO

Drug and device information, study documents

Studies a U.S. FDA-regulated drug product

No

Studies a U.S. FDA-regulated device product

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 Pregnancy Related

Clinical Trials on Sulfadoxine pyrimethamine (SP)

3
Subscribe