search
Back to results

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

Primary Purpose

Malaria, Malaria in Pregnancy, Pregnancy Related

Status
Recruiting
Phase
Phase 3
Locations
Papua New Guinea
Study Type
Interventional
Intervention
Dihydroartemisinin-Piperaquine (DP)
Sulfadoxine pyrimethamine (SP)
Sponsored by
Menzies School of Health Research
About
Eligibility
Locations
Arms
Outcomes
Full info

About this trial

This is an interventional prevention trial for Malaria

Eligibility Criteria

16 Years - undefined (Child, Adult, Older Adult)FemaleAccepts Healthy Volunteers

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

Sites / Locations

  • Papua New Guinea Institute of Medical ResearchRecruiting

Arms of the Study

Arm 1

Arm 2

Arm Type

Active Comparator

Experimental

Arm Label

SP + DP placebo every 4 weeks

SP + DP given every 4 weeks

Arm Description

Control arm

Intervention arm

Outcomes

Primary Outcome Measures

Malaria infection in pregnancy
'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

Adverse pregnancy outcome
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
Clinical malaria during pregnancy
Incidence of new episodes of fever or history of fever plus positive RDT confirmed by microscopy and/or qPCR during pregnancy
Parasitemia during pregnancy
Proportion of samples with parasites detected in maternal peripheral blood samples by microscopy or qPCR
Composite placental malaria detected by microscopy, qPCR or by histology
Prevalence of placental parasites by microscopy, qPCR, or placental histology
Placental malaria detected by microscopy
Prevalence of parasites in placental blood by microscopy
Placental malaria detected by qPCR
Prevalence of parasites in placental blood by qPCR
Active placental malaria detected by histology
Prevalence of active infection (presence of parasites) on histology
Past placental malaria detected by histology
Prevalence of past infection (pigment only) on histology
Placental malaria detected by histology
Prevalence of placental infection (active or past) on histology
Composite fetal loss and neonatal death
Prevalence of fetal loss (spontaneous miscarriage or stillbirth) and neonatal death
Composite of SGA-LBW-PTB
Prevalence of small for gestational age, low birth weight, and preterm birth
SGA
Prevalence of small for gestational age using the new Intergrowth-21st population reference's 10th centile
LBW
Prevalence of low birth weight
PTB
Prevalence of preterm birth
Birth weight
Mean birthweight
Neonatal length
Neonatal length
Maternal nutritional status
Changes in maternal body mass index (BMI)
Maternal nutritional status
Changes in maternal mid-upper arm circumference (MUAC)
Maternal anemia during pregnancy and at delivery
Proportion of routine haemoglobin measurements <100 g/L
Maternal hemoglobin levels during pregnancy and at delivery
Mean hemoglobin (g/L) at the third trimester antenatal visit and at delivery
Congenital anemia
Prevalence of anaemia (Hb <130 g/L) from newborn cord blood
Maternal gametocyte carriage during pregnancy and at delivery
Proportion of P. falciparum positive samples with gametocytes at the third trimester antenatal visit and at delivery, by light microscopy and RT-qPCR
Molecular markers of DP resistance
Proportion of parasite positive samples with molecular markers of DP resistance
Molecular markers of SP drug resistance
Proportion of parasite positive samples with molecular markers of SP resistance
Maternal mortality
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
Congenital malformations
Any visible external congenital abnormality on surface examination
Other SAEs and AEs
Incidence of AEs and SAEs
(History) of vomiting study drug
Prevalence of vomiting investigational product (IP) twice at the same IP administration visit
Dizziness
Prevalence of dizziness after a course of IP
Gastrointestinal complaints
Prevalence of gastrointestinal complaints after a course of IP

Full Information

First Posted
June 14, 2022
Last Updated
October 19, 2023
Sponsor
Menzies School of Health Research
Collaborators
Papua New Guinea Institute of Medical Research, University of Melbourne, Curtin University, Liverpool School of Tropical Medicine
search

1. Study Identification

Unique Protocol Identification Number
NCT05426434
Brief Title
Sulfadoxine-pyrimethamine Plus Dihydroartemisinin-piperaquine for Intermittent Preventive Treatment in Pregnancy
Acronym
SAPOT
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
Study Type
Interventional

2. Study Status

Record Verification Date
October 2023
Overall Recruitment Status
Recruiting
Study Start Date
August 31, 2022 (Actual)
Primary Completion Date
February 2025 (Anticipated)
Study Completion Date
December 2025 (Anticipated)

3. Sponsor/Collaborators

Responsible Party, by Official Title
Sponsor
Name of the Sponsor
Menzies School of Health Research
Collaborators
Papua New Guinea Institute of Medical Research, University of Melbourne, Curtin University, Liverpool School of Tropical Medicine

4. Oversight

Studies a U.S. FDA-regulated Drug Product
No
Studies a U.S. FDA-regulated Device Product
No
Data Monitoring Committee
Yes

5. 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.
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.

6. Conditions and Keywords

Primary Disease or Condition Being Studied in the Trial, or the Focus of the Study
Malaria, Malaria in Pregnancy, Pregnancy Related

7. Study Design

Primary Purpose
Prevention
Study Phase
Phase 3
Interventional Study Model
Parallel Assignment
Model Description
Double blinded randomized controlled trial
Masking
ParticipantCare ProviderInvestigatorOutcomes 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.
Allocation
Randomized
Enrollment
1172 (Anticipated)

8. Arms, Groups, and Interventions

Arm Title
SP + DP placebo every 4 weeks
Arm Type
Active Comparator
Arm Description
Control arm
Arm Title
SP + DP given every 4 weeks
Arm Type
Experimental
Arm Description
Intervention arm
Intervention Type
Drug
Intervention Name(s)
Dihydroartemisinin-Piperaquine (DP)
Other Intervention Name(s)
D-Artepp
Intervention Description
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
Intervention Type
Drug
Intervention Name(s)
Sulfadoxine pyrimethamine (SP)
Other Intervention Name(s)
G-COSPE
Intervention Description
SP (G-COSPE) will be supplied by Fosun Pharma, China. SP will be given as a single dose consisting of three 500mg/25mg tablets.
Primary Outcome Measure Information:
Title
Malaria infection in pregnancy
Description
'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'
Time Frame
Starting two weeks after initial dose until and including delivery
Secondary Outcome Measure Information:
Title
Adverse pregnancy outcome
Description
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 Frame
Time of delivery up to 28 days postpartum
Title
Clinical malaria during pregnancy
Description
Incidence of new episodes of fever or history of fever plus positive RDT confirmed by microscopy and/or qPCR during pregnancy
Time Frame
Starting two weeks after initial dose until and including delivery
Title
Parasitemia during pregnancy
Description
Proportion of samples with parasites detected in maternal peripheral blood samples by microscopy or qPCR
Time Frame
Starting two weeks after initial dose until and including delivery
Title
Composite placental malaria detected by microscopy, qPCR or by histology
Description
Prevalence of placental parasites by microscopy, qPCR, or placental histology
Time Frame
At time of delivery
Title
Placental malaria detected by microscopy
Description
Prevalence of parasites in placental blood by microscopy
Time Frame
At time of delivery
Title
Placental malaria detected by qPCR
Description
Prevalence of parasites in placental blood by qPCR
Time Frame
At time of delivery
Title
Active placental malaria detected by histology
Description
Prevalence of active infection (presence of parasites) on histology
Time Frame
At time of delivery
Title
Past placental malaria detected by histology
Description
Prevalence of past infection (pigment only) on histology
Time Frame
At time of delivery
Title
Placental malaria detected by histology
Description
Prevalence of placental infection (active or past) on histology
Time Frame
At time of delivery
Title
Composite fetal loss and neonatal death
Description
Prevalence of fetal loss (spontaneous miscarriage or stillbirth) and neonatal death
Time Frame
Time of delivery up to 28 days postpartum
Title
Composite of SGA-LBW-PTB
Description
Prevalence of small for gestational age, low birth weight, and preterm birth
Time Frame
At time of delivery
Title
SGA
Description
Prevalence of small for gestational age using the new Intergrowth-21st population reference's 10th centile
Time Frame
At time of delivery
Title
LBW
Description
Prevalence of low birth weight
Time Frame
At time of delivery
Title
PTB
Description
Prevalence of preterm birth
Time Frame
At time of delivery
Title
Birth weight
Description
Mean birthweight
Time Frame
At time of delivery
Title
Neonatal length
Description
Neonatal length
Time Frame
At time of delivery
Title
Maternal nutritional status
Description
Changes in maternal body mass index (BMI)
Time Frame
8 months from randomisation
Title
Maternal nutritional status
Description
Changes in maternal mid-upper arm circumference (MUAC)
Time Frame
6 months from randomisation
Title
Maternal anemia during pregnancy and at delivery
Description
Proportion of routine haemoglobin measurements <100 g/L
Time Frame
6 months from randomisation
Title
Maternal hemoglobin levels during pregnancy and at delivery
Description
Mean hemoglobin (g/L) at the third trimester antenatal visit and at delivery
Time Frame
6 months from randomisation
Title
Congenital anemia
Description
Prevalence of anaemia (Hb <130 g/L) from newborn cord blood
Time Frame
At delivery
Title
Maternal gametocyte carriage during pregnancy and at delivery
Description
Proportion of P. falciparum positive samples with gametocytes at the third trimester antenatal visit and at delivery, by light microscopy and RT-qPCR
Time Frame
6 months from randomisation
Title
Molecular markers of DP resistance
Description
Proportion of parasite positive samples with molecular markers of DP resistance
Time Frame
6 months from randomisation
Title
Molecular markers of SP drug resistance
Description
Proportion of parasite positive samples with molecular markers of SP resistance
Time Frame
6 months from randomisation
Title
Maternal mortality
Description
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
Time Frame
8 months from randomisation
Title
Congenital malformations
Description
Any visible external congenital abnormality on surface examination
Time Frame
8 months from randomisation
Title
Other SAEs and AEs
Description
Incidence of AEs and SAEs
Time Frame
8 months from randomisation
Title
(History) of vomiting study drug
Description
Prevalence of vomiting investigational product (IP) twice at the same IP administration visit
Time Frame
6 months from randomisation
Title
Dizziness
Description
Prevalence of dizziness after a course of IP
Time Frame
6 months from randomisation
Title
Gastrointestinal complaints
Description
Prevalence of gastrointestinal complaints after a course of IP
Time Frame
6 months from randomisation

10. Eligibility

Sex
Female
Minimum Age & Unit of Time
16 Years
Accepts Healthy Volunteers
Accepts Healthy Volunteers
Eligibility Criteria
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
Central Contact Person:
First Name & Middle Initial & Last Name or Official Title & Degree
Holger Unger, PhD MBChB
Phone
+61889468411
Email
holger.unger@menzies.edu.au
First Name & Middle Initial & Last Name or Official Title & Degree
Hellen Mnjala
Phone
+254722710920
Email
hellen.mnjala@menzies.edu.au
Overall Study Officials:
First Name & Middle Initial & Last Name & Degree
Holger Unger, PhD MBChB
Organizational Affiliation
Menzies School of Health Research, Darwin, Australia
Official's Role
Principal Investigator
Facility Information:
Facility Name
Papua New Guinea Institute of Medical Research
City
Madang
State/Province
Madang Province
ZIP/Postal Code
511
Country
Papua New Guinea
Individual Site Status
Recruiting
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Moses Laman, MBBS
Email
drmlaman@yahoo.com
First Name & Middle Initial & Last Name & Degree
Alice Mengi, MBBS
Email
alicemengi9@gmail.com

12. IPD Sharing Statement

Plan to Share IPD
No

Learn more about this trial

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

We'll reach out to this number within 24 hrs