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

Sponsor
Menzies School of Health Research (Other)
Overall Status
Not yet recruiting
CT.gov ID
NCT05426434
Collaborator
Papua New Guinea Institute of Medical Research (Other), University of Melbourne (Other), Curtin University (Other), Liverpool School of Tropical Medicine (Other)
1,172
2
40

Study Details

Study Description

Brief Summary

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.

Condition or Disease Intervention/Treatment Phase
  • Drug: Dihydroartemisinin-Piperaquine (DP)
  • Drug: Sulfadoxine pyrimethamine (SP)
Phase 3

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 Design

Study Type:
Interventional
Anticipated Enrollment :
1172 participants
Allocation:
Randomized
Intervention Model:
Parallel Assignment
Intervention Model Description:
Double blinded randomized controlled trialDouble blinded randomized controlled trial
Masking:
Quadruple (Participant, Care Provider, Investigator, Outcomes Assessor)
Masking Description:
Placebos will be used to mimic the identical dosing strategy such that every 4 weeks women will receive two drugs on day 1 (SP and placebo, or SP and DP) followed by one drug on days 2 and 3 (placebo or DP). One placebo that mimics the appearance of DP will be used. A randomization list will be computer generated by a member of the project who will not be directly involved in the conduct of the study. The randomization list will include consecutive treatment numbers with corresponding random treatment assignments. Randomized codes will correspond to the 2 treatment arms using permuted variable sized blocks.
Primary Purpose:
Prevention
Official Title:
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
Anticipated Study Start Date :
Aug 1, 2022
Anticipated Primary Completion Date :
Feb 1, 2025
Anticipated Study Completion Date :
Dec 1, 2025

Arms and Interventions

Arm Intervention/Treatment
Active Comparator: SP + DP placebo every 4 weeks

Control arm

Drug: Sulfadoxine pyrimethamine (SP)
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

    Drug: Dihydroartemisinin-Piperaquine (DP)
    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
  • Drug: Sulfadoxine pyrimethamine (SP)
    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
  • Outcome Measures

    Primary Outcome Measures

    1. Malaria infection in pregnancy [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'

    Secondary Outcome Measures

    1. Adverse pregnancy outcome [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

    2. Clinical malaria during pregnancy [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

    3. Parasitemia during pregnancy [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

    4. Composite placental malaria detected by microscopy, qPCR or by histology [At time of delivery]

      Prevalence of placental parasites by microscopy, qPCR, or placental histology

    5. Placental malaria detected by microscopy [At time of delivery]

      Prevalence of parasites in placental blood by microscopy

    6. Placental malaria detected by qPCR [At time of delivery]

      Prevalence of parasites in placental blood by qPCR

    7. Active placental malaria detected by histology [At time of delivery]

      Prevalence of active infection (presence of parasites) on histology

    8. Past placental malaria detected by histology [At time of delivery]

      Prevalence of past infection (pigment only) on histology

    9. Placental malaria detected by histology [At time of delivery]

      Prevalence of placental infection (active or past) on histology

    10. Composite fetal loss and neonatal death [Time of delivery up to 28 days postpartum]

      Prevalence of fetal loss (spontaneous miscarriage or stillbirth) and neonatal death

    11. Composite of SGA-LBW-PTB [At time of delivery]

      Prevalence of small for gestational age, low birth weight, and preterm birth

    12. SGA [At time of delivery]

      Prevalence of small for gestational age using the new Intergrowth-21st population reference's 10th centile

    13. LBW [At time of delivery]

      Prevalence of low birth weight

    14. PTB [At time of delivery]

      Prevalence of preterm birth

    15. Birth weight [At time of delivery]

      Mean birthweight

    16. Neonatal length [At time of delivery]

      Neonatal length

    17. Maternal nutritional status [8 months from randomisation]

      Changes in maternal body mass index (BMI)

    18. Maternal nutritional status [6 months from randomisation]

      Changes in maternal mid-upper arm circumference (MUAC)

    19. Maternal anemia during pregnancy and at delivery [6 months from randomisation]

      Proportion of routine haemoglobin measurements <100 g/L

    20. Maternal hemoglobin levels during pregnancy and at delivery [6 months from randomisation]

      Mean hemoglobin (g/L) at the third trimester antenatal visit and at delivery

    21. Congenital anemia [At delivery]

      Prevalence of anaemia (Hb <130 g/L) from newborn cord blood

    22. Maternal gametocyte carriage during pregnancy and at delivery [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

    23. Molecular markers of DP resistance [6 months from randomisation]

      Proportion of parasite positive samples with molecular markers of DP resistance

    24. Molecular markers of SP drug resistance [6 months from randomisation]

      Proportion of parasite positive samples with molecular markers of SP resistance

    25. Maternal mortality [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

    26. Congenital malformations [8 months from randomisation]

      Any visible external congenital abnormality on surface examination

    27. Other SAEs and AEs [8 months from randomisation]

      Incidence of AEs and SAEs

    28. (History) of vomiting study drug [6 months from randomisation]

      Prevalence of vomiting investigational product (IP) twice at the same IP administration visit

    29. Dizziness [6 months from randomisation]

      Prevalence of dizziness after a course of IP

    30. Gastrointestinal complaints [6 months from randomisation]

      Prevalence of gastrointestinal complaints after a course of IP

    Eligibility Criteria

    Criteria

    Ages Eligible for Study:
    16 Years and Older
    Sexes Eligible for Study:
    Female
    Accepts Healthy Volunteers:
    Yes
    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

    Contacts and Locations

    Locations

    No locations specified.

    Sponsors and Collaborators

    • Menzies School of Health Research
    • Papua New Guinea Institute of Medical Research
    • University of Melbourne
    • Curtin University
    • Liverpool School of Tropical Medicine

    Investigators

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

    Study Documents (Full-Text)

    None provided.

    More Information

    Publications

    None provided.
    Responsible Party:
    Menzies School of Health Research
    ClinicalTrials.gov Identifier:
    NCT05426434
    Other Study ID Numbers:
    • 2021-4107
    First Posted:
    Jun 22, 2022
    Last Update Posted:
    Jul 20, 2022
    Last Verified:
    Jul 1, 2022
    Individual Participant Data (IPD) Sharing Statement:
    No
    Plan to Share IPD:
    No
    Studies a U.S. FDA-regulated Drug Product:
    No
    Studies a U.S. FDA-regulated Device Product:
    No
    Additional relevant MeSH terms:

    Study Results

    No Results Posted as of Jul 20, 2022