ArticlePDF Available

Risks of miscarriage and inadvertent exposure to artemisinin derivatives in the first trimester of pregnancy: A prospective cohort study in western Kenya

Authors:

Abstract and Figures

Background: The artemisinin anti-malarials are widely deployed as artemisinin-based combination therapy (ACT). However, they are not recommended for uncomplicated malaria during the first trimester because safety data from humans are scarce. Methods: This was a prospective cohort study of women of child-bearing age carried out in 2011-2013, evaluating the relationship between inadvertent ACT exposure during first trimester and miscarriage. Community-based surveillance was used to identify 1134 early pregnancies. Cox proportional hazard models with left truncation were used. Results: The risk of miscarriage among pregnancies exposed to ACT (confirmed + unconfirmed) in the first trimester, or during the embryo-sensitive period (≥6 to <13 weeks gestation) was higher than among pregnancies unexposed to anti-malarials in the first trimester: hazard ratio (HR) = 1.70, 95 % CI (1.08-2.68) and HR = 1.61 (0.96-2.70). For confirmed ACT-exposures (primary analysis) the corresponding values were: HR = 1.24 (0.56-2.74) and HR = 0.73 (0.19-2.82) relative to unexposed women, and HR = 0.99 (0.12-8.33) and HR = 0.32 (0.03-3.61) relative to quinine exposure, but the numbers of quinine exposures were very small. Conclusion: ACT exposure in early pregnancy was more common than quinine exposure. Confirmed inadvertent artemisinin exposure during the potential embryo-sensitive period was not associated with increased risk of miscarriage. Confirmatory studies are needed to rule out a smaller than three-fold increase in risk.
This content is subject to copyright. Terms and conditions apply.
Dellicour et al. Malar J (2015) 14:461
DOI 10.1186/s12936-015-0950-6
RESEARCH
Risks ofmiscarriage andinadvertent
exposure toartemisinin derivatives inthe rst
trimester ofpregnancy: a prospective cohort
study inwestern Kenya
Stephanie Dellicour1*, Meghna Desai2, George Aol3, Martina Oneko3, Peter Ouma3, Godfrey Bigogo3,
Deron C. Burton2, Robert F. Breiman4, Mary J. Hamel2, Laurence Slutsker2, Daniel Feikin2, Simon Kariuki3,
Frank Odhiambo3, Jayesh Pandit5, Kayla F. Laserson2, Greg Calip6, Andy Stergachis7 and Feiko O. ter Kuile1
Abstract
Background: The artemisinin anti-malarials are widely deployed as artemisinin-based combination therapy (ACT).
However, they are not recommended for uncomplicated malaria during the first trimester because safety data from
humans are scarce.
Methods: This was a prospective cohort study of women of child-bearing age carried out in 2011–2013, evaluating
the relationship between inadvertent ACT exposure during first trimester and miscarriage. Community-based surveil-
lance was used to identify 1134 early pregnancies. Cox proportional hazard models with left truncation were used.
Results: The risk of miscarriage among pregnancies exposed to ACT (confirmed + unconfirmed) in the first trimester,
or during the embryo-sensitive period (6 to <13 weeks gestation) was higher than among pregnancies unexposed
to anti-malarials in the first trimester: hazard ratio (HR) = 1.70, 95 % CI (1.08–2.68) and HR = 1.61 (0.96–2.70). For
confirmed ACT-exposures (primary analysis) the corresponding values were: HR = 1.24 (0.56–2.74) and HR = 0.73
(0.19–2.82) relative to unexposed women, and HR = 0.99 (0.12–8.33) and HR = 0.32 (0.03–3.61) relative to quinine
exposure, but the numbers of quinine exposures were very small.
Conclusion: ACT exposure in early pregnancy was more common than quinine exposure. Confirmed inadvertent
artemisinin exposure during the potential embryo-sensitive period was not associated with increased risk of miscar-
riage. Confirmatory studies are needed to rule out a smaller than three-fold increase in risk.
Keywords: Anti-malarials, Pharmacovigilance, Drug safety in pregnancy, Teratogenicity, Miscarriage
© 2015 Dellicour et al. This article is distributed under the terms of the Creative Commons Attribution 4.0 International License
(http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium,
provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license,
and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/
publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
Background
Artemisinin-based combination therapy (ACT) anti-
malarials have been adopted as firstline treatment for
falciparum malaria in almost all endemic countries,
providing lifesaving benefits to children, adults and
pregnant women globally [1]. However, their safety is
uncertain when used in early pregnancy. Ascertainment
of risk from exposure to anti-malarials in the first trimes-
ter is difficult in resource-poor settings and data avail-
able for assessing risk are limited [2, 3]. Artemisinins
are embryo-toxic in several animal species, including
non-human primate models [4, 5]. Teratogenic effects
observed in mice and rabbits included death of the foe-
tus, malformations of the heart, great vessels, and limb
defects. Primate models exposed to prolonged courses
of ACT had high rates of foetal loss [6]. Animal mod-
els suggested that artemisinin embryo-toxicity targets
primitive erythroblasts, which are the primary form of
red blood cells in circulation between weeks 4 and 10
Open Access
*Correspondence: Stephanie.Dellicour@lstmed.ac.uk
1 Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3
5QA, UK
Full list of author information is available at the end of the article
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 2 of 9
Dellicour et al. Malar J (2015) 14:461
post-conception in humans. erefore the embryo-sen-
sitive period to artemisinin, if any, is thought to occur at
6–12 (inclusive) weeks’ gestation from the first day of the
last menstrual period (LMP) in humans [4, 5, 7, 8].
ere are limited data available to assess whether ACT
is embryo-toxic or teratogenic in humans; fewer than
700 exposures in the first trimester have been well doc-
umented [915]. After reviewing all existing evidence in
2003 and then in 2006, the World Health Organization
(WHO) recommended that artemisinins could be used
during the second or third trimesters of pregnancy and
that, due to insufficient safety data, treatment in the first
trimester was not recommended unless the life of the
mother is at risk, or oral quinine is not available [5, 16].
e recommended treatment for first trimester malaria
infections is seven days’ oral quinine alone or combined
with clindamycin [17]. However, as women may not be
aware of their pregnancy or do not declare an early preg-
nancy, and because clinic staff do not often assess for
pregnancy in women of child-bearing age (WOCBA), the
risk of exposure to drugs not recommended in pregnancy,
including to potential teratogens, is possible during this
period [18]. As ACT is increasingly available, a growing
number of women will be inadvertently exposed to an
artemisinin compound in early pregnancy, including dur-
ing the period when foetal organs and tissues are formed.
Malaria can have severe consequences to the health
of the pregnant woman and her unborn baby including
maternal anaemia, foetal loss, preterm birth, low birth
weight and perinatal mortality, and in some cases mater-
nal death. e impact of malaria infection in early preg-
nancy has been identified as a major knowledge gap for
estimating the burden of malaria in pregnancy. Recent
studies have provided insight into the potential adverse
consequences of malaria infections early in pregnancy,
showing a major impact on birth weight and maternal
anaemia [19, 20]. Findings from a retrospective analysis
from 25 years of data from the ai-Myanmar border,
where artemisinin deployment has been necessary for
many years because of multi-drug resistance, showed
that malaria infection in the first trimester (both sympto-
matic and asymptomatic) was a significant risk factor for
miscarriage. No association between first trimester arte-
misinin exposure and miscarriage was found. However
more data from a wider range of malaria-endemic coun-
tries are required to provide an increased level of reas-
surance that first trimester artemisinin exposure does
not significantly increase the risk of miscarriage or other
adverse pregnancy outcomes. e findings from a pro-
spective cohort study of WOCBA designed to examine
whether ACT exposure in the first trimester was associ-
ated with miscarriage are reported here.
Methods
Overview ofstudy design
is was a prospective cohort study conducted among
WOCBA (15–49 years of age) residing in a highly
malarious area in western Kenya with a population
under continuous health and demographic surveillance
system (HDSS) monitoring as part of the collabora-
tion between the Kenya Medical Research Institute
(KEMRI) and Centers for Disease Control and Pre-
vention (CDC) [21]. Participants received treatment
through the usual channels, including health facilities
and drug outlets.
Procedures
Recruitment ofwomen ofchild‑bearing age
andpregnancy detection
Between 15 February, 2011 and 15 February, 2013,
6010 WOCBA participating in an ongoing population-
based, infectious disease, surveillance project (PBIDS)
in rural Bondo District, western Kenya [22, 23] (Addi-
tional file1) were invited to participate in the ‘Evalua-
tion of Medications used in Early Pregnancy’ (EMEP)
prospective cohort study. EMEP staff visited all homes
in the PBIDS and enrolled consenting WOCBA who
met eligibility criteria for EMEP. WOCBA were eligi-
ble for EMEP if they were between 15 and 49years of
age and active participants of PBIDS. Exclusion cri-
teria included: inability to give informed consent or
provide an accurate medical history. WOCBA who
consented to participate were asked if they could be
pregnant and offered a pregnancy test at the time of
enrolment and again approximately every 3 months
thereafter. Any participant with a detected pregnancy
was referred to the antenatal clinic at Lwak Hospital
where trained EMEP nurses confirmed the pregnancy
(either by ultrasound if the women presented before
24weeks, or by palpation and by auscultation of the
foetal heart later in pregnancy) and offered free ante-
natal care (ANC). Additionally, all pregnant patients
presenting at the ANC clinic of Lwak Hospital were
enrolled if all criteria were met. EMEP nurses were not
involved in treatment of study participants.
Gestational age assessment
Gestational age was determined using the most accu-
rate measurement available for each participant in the
following order: ultrasound scan taken before 24weeks’
gestation performed by trained study nurses (Sonosite
180 plus portable ultrasound system), Ballard estimates
measured within 96h of birth, LMP or reported gesta-
tion at time of pregnancy loss, and, lastly gestational age
derived from fundal height assessment (Additional file1).
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 3 of 9
Dellicour et al. Malar J (2015) 14:461
Pregnancy outcome
Pregnancy outcomes were assessed using a combina-
tion of health facility- and home-based follow-up vis-
its. e latter is particularly relevant for miscarriages,
because the vast majority of these events occur in the
community, not in health facilities. Village-based staff
received monthly lists of participants with estimated
delivery dates and after visiting the participants’ homes
they informed study nurses of pregnancy outcomes. Fol-
low-ups by study staff were then arranged to administer
structured questionnaires about the delivery, outcome,
any illnesses and medication used during pregnancy.
Pregnancy outcomes captured included: pregnancy loss
(miscarriages, induced abortions and stillbirths), live
births and major congenital malformations detectable
at birth by surface examination. is analysis focuses on
miscarriage defined as spontaneous pregnancy loss at or
before 28 completed weeks’ gestation (2–28weeks inclu-
sive), which is considered the gestational age of viability
in resource-constrained settings.
Anti‑malarial drug exposure ascertainment
Drug exposure data were captured using three
approaches (Table 1): (a) interviews with pregnant
women visiting the antenatal clinic in Lwak Hospital and
at the time of pregnancy outcome follow-up (henceforth
referred to as EMEP data); (b) record linkage to data on
drugs prescribed to WOCBA at the outpatient depart-
ment in Lwak Hospital (henceforth referred to as Lwak-
OPD data); and, (c) weekly to twice monthly home visits
by fieldworkers as part of PBIDS.
Other covariates
Obstetric history and ANC laboratory information col-
lected routinely at antenatal booking (haemoglobin
level, HIV and syphilis testing, and malaria microscopy)
were extracted from the ANC records at Lwak hospital
or antenatal cards by study nurses. Demographic char-
acteristics and medical history, including illnesses (e.g.,
malaria) and drugs used during the current pregnancy
were collected at each EMEP study visit at ANC and dur-
ing pregnancy outcome follow-up visits. Household level
wealth quintiles were obtained from the HDSS [24].
Data analysis
Exposure denition
A trend of increase in risk of miscarriage with ACT expo-
sure during this artemisinin-specific, embryo-sensitive
period would corroborate the biological mechanism
observed in animal models and suggest a causal associa-
tion with ACT exposures. e analysis focused on two
exposure definitions: anti-malarial drug reported/pre-
scribed (1) ‘anytime’ in the first trimester, i.e., gestational
week 2 and 0days (day 14 since LMP) to week 13 and
6days (day 97 since LMP) post-LMP, and (2) between
weeks 6 day 0 (day 42 since LMP) to week 12 day 6
post-LMP (day 90 since LMP) (potential artemisinin
embryo-sensitive period as suggested by animal repro-
toxicology [8]). Unexposed was defined as no evidence
of anti-malarial or malaria exposure in any of the three
data sources. Confirmed exposures were defined as expo-
sures identified by at least two of the three data sources.
Confirmed + unconfirmed exposures were defined as
Table 1 Description ofdrug information sources used todetermine anti-malarial andmalaria exposure status
ANC antenatal care, EMEP evaluation of medications used in early pregnancy study, Lwak OPD Lwak hospital out-patient department, PBIDS population-based
infectious disease surveillance
Approach Format Drug information available
EMEP self-report Retrospective self-report of illness and medication used since
the beginning of the pregnancy collected at every ANC visit
and at pregnancy outcome follow-up visit. A general open
question about any drug use as well as a directed question
for specific anti-malarials were included as using medication/
indication-specific questions have been shown to improve
accuracy. Photographs of all anti-malarial drugs found in the
study area were used to facilitate recognition of drug names. A
calendar marking public holidays and school closures was also
used to enhance recall of dates
Drug name
Drug start date
Duration
Number of tablets per day
Indication and indication diagnosis
Drug source
Lwak-OPD records Prospective documentation by health facility clinic staff of
diagnosis and treatment prescribed at outpatient department
(OPD) whenever a PBIDS participant sought care at Lwak
Hospital for an infectious syndrome
Date of visit
Diagnosis
Prescribed treatment
PBIDS weekly and twice-monthly
home visits Self-report of symptoms, health-seeking behaviour and medica-
tion. This information was collected continuously on a weekly
(from 5 January, 2010 to 26 May, 2011) and then twice-
monthly basis (27 May, 2011 onwards). The same visual aids as
described above were used for recall of drug intake
Date of visit
Symptoms in previous week/2 weeks
Treatment taken for the symptoms including
drug name
If and where care was sought
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 4 of 9
Dellicour et al. Malar J (2015) 14:461
anti-malarial identified by at least one of the three data
sources.
Cox regression model
Analyses were performed using Stata v12.1 (StataCorp
LP, College Station, TX, USA). Cox proportional haz-
ard regression models with left truncation were fitted
to estimate the effect on miscarriage of ACT exposure
during the first trimester and during the artemisinin
embryo-sensitive period. Exposure was treated as a time-
dependent variable (Additional file 1). Known risk fac-
tors for miscarriage were considered and to determine
which variables remained in the final model, assessment
of confounding was based on the impact a variable had
on the hazard ratio, followed by the consideration of its
precision. If the HR changed by 10% the variable was
retained in the model [25, 26].
e primary analysis compared the hazard of miscar-
riage among pregnancies with confirmed ACT expo-
sures, either anytime during the first trimester or six to
12weeks post-LMP, with the hazard among women not
exposed to any anti-malarials anytime during the first tri-
mester or among women exposed to quinine anytime in
the first trimester or 6–12weeks post-LMP.
Secondary analyses consisted of similar models but
using (a) less restrictive exposure definitions, including
both confirmed and unconfirmed exposures, and, (b)
more restrictive exposure definitions where only ACT
exposures within estimated gestational age confidence
margins were included (Additional file2).
Ethical review andconsent
e EMEP study was approved by the ethics commit-
tees and institutional review boards of CDC (No. 5889),
KEMRI (No. 1752) and the Liverpool School of Tropi-
cal Medicine (No. 09.70). Written informed consent
or assent was obtained from each participant includ-
ing consent for record linkage with PBIDS and HDSS
databases.
Results
Participant characteristics
Out of 5911 eligible WOCBA, 5536 (94 %) consented
to participate and among them, 1453 pregnancies were
detected, and 1134 (78%) were included in the data anal-
ysis (Fig.1). e mean andmedian gestational age at time
of pregnancy detection was 13.3 (standard deviation 6.9)
and 12.1 (range 0–27.9) weeks (Table2). Overall, 62% of
deliveries took place at a health facility, and 25% of the
miscarriages. Overall, 67% of pregnancy outcomes were
captured within a week of the event; however, for miscar-
riages this was only 20%.
Prevalence ofrst trimester ACT andquinine exposure
Overall, 299 (26.4 %) of the 1134 pregnancies had evi-
dence of possible ACT exposure anytime in the first tri-
mester (confirmed + unconfirmed). For 77 (25.8 % of
exposures and 6.8% of all pregnancies) this could be con-
firmed by at least two of the three sources; 56 of these
confirmed exposures (18.7, 5.3 % of pregnancies) were
within the estimated gestational age confidence mar-
gins. For 212 out of 299 first trimester exposures (70.9%,
18.7% of pregnancies), the exposure occurred between
6 and 12weeks’ gestation; 47 of them were confirmed
exposures (Fig.2). Only 13 pregnancies were exposed to
quinine-alone anytime in the first trimester, and 11 dur-
ing the 6–12weeks’ gestational period.
Association betweenrst trimester ACT‑exposure
andmiscarriage
Conrmed exposure (primary analysis)
Compared to pregnancies without anti-malarial expo-
sure/malaria in the first trimester (793), the hazard
for miscarriage was non-significantly higher among
women with confirmed ACT exposures anytime in the
first trimester (77) [hazard ratio (HR) =1.24, 95% CI
(0.56–2.73)], and this was HR=1.72 (0.66–4.45) in mul-
tivariate analysis (Fig. 2). e corresponding values for
ACT exposure during the embryo-sensitive period (47)
were HR=0.73 (0.19–2.82) and HR=0.81 (0.21–3.03)
(Fig.2).
e values when compared against quinine (13) were:
HR=0.99 (0.12–8.33) and HR=0. 32 (0.03–3.61) (crude
analysis) for exposure anytime and six to 12weeks post-
LMP (Fig.2).
More restrictive definitions to define exposure within
the redefined margins for gestational age resulted in simi-
lar or lower effect estimates, but numbers of exposures
and events were limited (Additional file2). e method
used for missing value did not alter the conclusions
(Additional file3).
Conrmed+unconrmed exposure (secondary analysis)
When using a less restrictive definition of exposure by
including unconfirmed exposures as well, the risk of
miscarriages was significantly higher among the ACT-
exposed pregnancies relative to unexposed pregnan-
cies: adjusted HR=1.66 (1.04–2.67). is was HR=1.
61 (0.96–2.70) for the embryo-sensitive period. e HRs
when compared to quinine were HR=0.64 (0.08–4.91)
and HR=0.46 (0.05–4.44), respectively (Fig.2).
Discussion
Pregnancies exposed to ACT in the first trimester were
at increased risk of miscarriage compared to pregnancies
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 5 of 9
Dellicour et al. Malar J (2015) 14:461
not exposed to anti-malarials in the same gestation
period. is was only statistically significant at the 5%
level in the group with the less restrictive definition for
exposure (confirmed and unconfirmed) which had higher
number of events (29) and exposures (299) [adjusted
HR=1.66 95% CI (1.04–2.67)]. A similar effect measure
Analysis
Assessed for eligibility
N=6010
Excluded (n=474)
Not meeting inclusion criteria (n=99)
Declined to participate (n=375)
Consented (n=5536)
Pregnancies(n=1453)
Number of pregnancies per participants:
Single pregnancy (n=1266)
Two pregnancies(n=92)
Three pregnancies (n=1)
Follow-Up
Enrolment
Loss to follow-up(n=85)
Migrated (n=67)
Withdrawal or refused follow-up(n=13)
Maternal death (n=5)
Pregnancy Outcomes (n=1368)
Pregnancies included
(n=1134)
Excluded from analysis (n=319)
Detected at outcome(n=33)
Entered after 28 weeks (n=219)
No GA information (n=21)
Pregnancy end date error (n=5)
No follow-up (n=41)
Fig. 1 Study participant flow diagram from screening to inclusion in data analysis
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 6 of 9
Dellicour et al. Malar J (2015) 14:461
[HR=1.72 95% CI (0.66–4.45)] was obtained when the
analysis was restricted to those exposures that could be
confirmed by the OPD database or the ongoing house-
hold surveillance, which was the primary analysis. How-
ever the available exposures (77) and events (six) were
reduced markedly with this more restrictive analysis and
the difference was not statistically significant. When the
analysis was further restricted to exposures in the poten-
tial embryo-sensitive period in humans for the arte-
misinins, the effect estimates were again similar [HR=1.
61 95% CI (0.96, 2.70)] for confirmed + unconfirmed
exposures, but much lower for confirmed exposures
[HR = 0.73 95 % CI (0.19, 2.82)]. However this latter
analysis, which was also part of the primary analysis,
included only two events and 47 pregnancies exposed to
ACT. ere was no evidence for an increase in the risk
of miscarriage among women treated with ACT versus
women treated with oral quinine, but again the number
exposed to quinine alone was limited to 13 with only one
miscarriage.
It was expected that the risk of miscarriage would
be higher among women who received anti-malarials
than among women without anti-malarial exposure
early in pregnancy. is is related to the potential for
Table 2 Characteristics of1134 pregnancies byACT exposure status [n (%) otherwise stated]
ACT artemisinin combination therapy, SD standard deviation
*P values refer to Pearson Chi square test for categorical variables and ANOVA test for continuous variables
a Gestational age lowest estimate include 0 which reects inaccuracy in the gestational age measurements
b HIV status information was not available for 12% (129) of pregnancies that did not attend antenatal care or have the antenatal card for review. HIV status
information was complemented by HDSS and data which oered home-based HIV testing and counselling to PBIDS participants. Test results were linked to the
study participants using unique ID and missing data were updated if the test was performed before the pregnancy detection for HIV positive test results and for HIV
negative results if the test was performed maximum 3months before or after pregnancy detection. An additional 30 HIV status were ascertained while 8% (99) still
had no HIV status data
Overall
(N=1134) No ACT exposure
inthe rst trimester
(N=835)
Unconrmed ACT
exposure inthe rst
trimester (N=222)
Conrmed ACT
exposure inthe rst
trimester (N=77)
P values*
Age in years [mean (SD;
range)] 26.1 (6.8; 15–47) 26.1 (6.7; 15–45) 26.7 (7.2; 15–47) 25.2 (6.5; 16–41) 0.225
Gravidity Missing n = 16 Missing n = 14 Missing n = 1 Missing n = 1 0.065
Primigravidae 219 (19.6) 151 (18.4) 47 (21.3) 21 (27.6)
1–3 pregnancies 525 (47.0) 405 (49.3) 90 (40.7) 30 (39.5)
4+ pregnancies 374 (33.5) 265 (32.3) 84 (38.0) 25 (32.9)
Previous pregnancy loss 160 (14.3), Missing n = 17 118 (14.4), Missing n = 15 30 (13.6), Missing n = 1 12 (15.8), Missing n = 1 0.888
Gestational age at detec-
tion in weeks [mean (SD;
range)]a
13.3 (6.9; 0–27.9) 13.3 (7.0; 0–27.9) 13.0 (6.7; 0.3–27) 13.6 (7.1; 2.4–27.4) 0.770
Occupation Missing n = 31 Missing n = 28 Missing n = 1 Missing n = 2 0.191
Not working 379 (34.4) 281 (34.8) 68 (30.8) 30 (40.0)
Farming 369 (33.5) 268 (33.2) 80 (36.2) 21 (28.0)
Small business/Skilled
Labour 335 (30.4) 246 (30.5) 65 (29.4) 24 (32.0)
Other 20 (1.8) 12 (1.5) 8 (3.6) 0
Antenatal care summary
Number of ANC visit Missing n = 39 Missing n = 31 Missing n = 5 Missing n = 3 0.125
None 89 (8.1) 64 (8.0) 21 (9.7) 4 (5.4)
1 90 (8.2) 61 (7.6) 24 (11.1) 5 (6.8)
2 155 (14.2) 121 (15.1) 25 (11.5) 9 (12.2)
3 244 (22.3) 193 (24.0) 38 (17.5) 13 (17.6)
4+517 (47.2) 365 (45.4) 109 (50.2) 43 (58.1)
Gestational age at first
ANC visit in weeks
[mean (SD)]*
20.8 (7.8) range: 1.7–41.0 21.24 (7.8) range: 2.7–41.0 19.7 (7.6) range: 1.7–41.0 19.4 (7.7) range: 3.4-37.0 0.020
HIV positivebMissing n = 101 Missing n = 79 Missing n = 18 Missing n = 4 0.354
Negative 771 (74.4) 562 (74.3) 149 (73.0) 60 (82.2)
Positive 262 (25.4) 194 (25.7) 55 (27.0) 13 (17.8)
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 7 of 9
Dellicour et al. Malar J (2015) 14:461
confounding by indication, i.e., women treated with ACT
or quinine sought treatment because of their malaria
or other febrile illness, whereas women who did not
require anti-malarials did not. e comparison with
untreated women is therefore difficult to interpret as it
does not allow for the differentiation between the effects
of malaria and the drug treating it. Malaria itself, even
if it remains asymptomatic, is a known cause of miscar-
riage. A recent meta-analysis of five trials with malaria
chemoprophylaxis or intermittent preventive therapy in
2876 paucigravidae in sub-Saharan Africa showed that
women in the control arms were at a 1.54 95% CI (0.98–
2.44) higher risk of miscarrying than women protected
by chemoprevention [27]. Prospective studies in low
malaria-transmission areas in ailand also found that
asymptomatic malaria in the first trimester increased the
odds of miscarriage nearly three-fold and symptomatic
infections four-fold [13]. e 1.4- to 1.7-fold increased
risk for miscarriage among women exposed to ACT or
quinine relative to pregnancies not requiring treatment
observed in this study is thus within the expected range
of malaria-associated risk of miscarriage.
is study is underpowered to confidently detect or
exclude effects smaller than a three-fold increased risk of
miscarriage associated with ACT. Nevertheless no indi-
cation for such a potential association was found. First,
there was no indication that the effect size associated
with ACT exposure relative to unexposed women was
greater among women treated during the embryo-sensi-
tive period than at anytime during the first trimester. If
ACT was causing miscarriage through this mechanism,
the effect size would be expected to be highest for expo-
sures restricted to that embryo-sensitive period. No such
trend was observed. Secondly, the rates of miscarriage
in the quinine-only and ACT-exposed pregnancies were
similar. Although the comparison with quinine needs to
be interpreted with caution due to the small numbers of
quinine-only exposed women, these results are consist-
ent with observations from the ai-Burmese border by
McGready etal. ey also found no difference in the pro-
portions of pregnancies ending in miscarriages between
women treated with chloroquine (26%), quinine (27%)
or artesunate (31 %) [13]. A recent prospective study
from Tanzania reported higher risk of pregnancy loss
(miscarriage and stillbirth combined) in women exposed
to quinine compared to those exposed to ACT [14]. A
prospective study in Zambia found higher occurrence of
miscarriage in first trimester ACT-exposed pregnancies
(5%) compared to none in those exposed to sulfadox-
ine-pyrimethamine or quinine but the number exposed
to quinine (six) were too small to allow for a meaningful
comparison [12].
e small number of quinine exposures in the first
trimester in this study was surprising as this is the
Fig. 2 Miscarriage rate, unadjusted and adjusted hazard rates for the association between different anti-malarial exposure categories and miscar-
riage
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 8 of 9
Dellicour et al. Malar J (2015) 14:461
recommended first-line malaria treatment in the first tri-
mester. However these observations are consistent with
a recent study on malaria in pregnancy-prescribing prac-
tice carried out in the same area of western Kenya (Riley
etal., unpublished) and a study from Uganda [28]. ese
studies draw attention to the need to assess reasons for
poor adherence to quinine and malaria treatment guide-
lines. Poor tolerability and poor compliance to its seven-
day regimen is a known problem for treatment of malaria
with oral quinine [29, 30].
is study had several limitations that should be con-
sidered. First, the small number of quinine exposures
limited the ability to compare ACT-exposed pregnancies
to the purported ‘control’ drug (as quinine is not known
to cause miscarriages) [3]. Second, it was not possible to
control for confounding by indication (i.e., the disease
itself) because laboratory confirmation of malaria was
not available for most women. Controlling for malaria
and its severity is important, as malaria itself has been
suggested to reduce the potential risk of embryo-tox-
icity from artemisinin as was found in rat models [31].
ird, since induced abortions are illegal in Kenya, this
could have resulted in induced abortions being reported
as miscarriages. However since neither ACT nor qui-
nine exposures are perceived as indications for induced
abortion in this population, it is thus unlikely that such
misclassification would differ according to exposure sta-
tus. Fourth, it was not possible to account for exposure
misclassification due to lack of adherence to prescribed
medication (drug intake was not observed) or from coun-
terfeit anti-malarials [32], which could bias the estimate
towards the null. Fifth, the ability to confirm exposure
was limited because there was limited overlap in the
exposures ascertained in the three data sources. e
group at highest risk for bias are the unconfirmed expo-
sure cases as 32 first-trimester, ACT-exposures were only
reported after pregnancy outcome. Recall bias following
adverse pregnancy outcome has been well documented,
hence the focus in this study was to confirm ACT expo-
sures using prospective drug ascertainment approaches
through record linkage to minimize such bias [3335].
Another potential source of exposure misclassification is
gestational age measurement errors. e study could not
assess any dose–response effect of exposure.
Conclusion
e results presented here are consistent with two pre-
vious observational studies showing an increased risk of
miscarriage among women treated for malaria with ACT
in the first trimester versus unexposed women, and a
similar [13] or lower risk compared to oral quinine [14].
ese results also suggest that ACT use in the first tri-
mester is much more common than quinine. e risk
associated with malaria in early pregnancy, the compara-
ble observed risk between ACT and quinine exposures,
and the limited compliance to treatment with quinine
suggests a trial comparing ACT versus quinine for the
treatment of uncomplicated malaria in the first trimes-
ter may be merited. Before such a trial is considered,
further safety data on the association between ACT and
congenital malformations, that is forthcoming from stud-
ies conducted by the Malaria in Pregnancy Consortium
and WHO, should be reviewed and all available evidence
pooled to evaluate the evidence of the risk and benefits of
artemisinin use in early pregnancy.
Abbreviations
ACT: artemisinin-based combination therapy; ANC: antenatal care; CDC: US
Centers for Disease Control and Prevention; EMEP: evaluation of medications
used in early pregnancy study; HDSS: health and demographic surveillance
system; HR: hazard ratio; KEMRI: Kenya Medical Research Institute; LMP: last
menstrual period; OPD: outpatient department; PBIDS: population-based
infectious disease surveillance project; WHO: World Health Organization;
WOCBA: women of childbearing age.
Authors’ contributions
SD, FtK, AS, LS, and MJH conceived and designed the experiments. SD, GA, PO,
MO, and GB conducted field work. SD and GC analysed the data. GB, DF, RFB,
SK, DCB, FO, and FtK contributed data/analysis tools. SD, DCB, RFB, MJH, LS, DF,
SK, KL, AS, MD, and FtK interpreted the data. JP acted as Government liaison
and the Kenyan regulator. SD, FtK and MD rote the first draft of the manu-
script. All authors read and approved the final manuscript.
Author details
1 Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA,
UK. 2 Centers for Disease Control and Prevention, Atlanta, GA, USA. 3 Kenya
Medical Research Institute Centre for Global Health Research, Kisumu, Kenya.
4 Global Health Institute, Emory University, Atlanta, GA, USA. 5 Bayer Health-
care, Nairobi, Kenya. 6 Pharmacy Systems, Outcomes and Policy Department,
University of Illinois at Chicago, Chicago, USA. 7 Departments of Pharmacy
and Global Health, Schools of Pharmacy and Public Health, University of Wash-
ington, Seattle, USA.
Acknowledgements
The work presented in this paper was performed under the KEMRI and CDC
Collaboration in western Kenya. We are very grateful to all participants for
taking part in the study. We wish to thank the EMEP study team for their
perseverance and hard work. We are grateful to the International Emerging
Infection Program (IEIP) team for their help and collaboration. Furthermore
we thank the Asembo District health and medical team and the Lwak Mission
Hospital Board for their support. We also thank John Williamson and Jane
Bruce for the statistical support and advice. KEMRI/CDC HDSS is a member of
the INDEPTH Network. The findings and conclusions in this paper are those of
the authors and do not necessarily represent the views of the US Centers for
Disease Control and Prevention. This paper is published with the permission
of KEMRI Director. This work was partly supported by the Malaria in Pregnancy
(MiP) Consortium, which is funded through a grant from the Bill and Melinda
Gates Foundation to the Liverpool School of Tropical Medicine, UK and partly
Additional les
Additional le 1. Detailed description of study site and methodology.
Additional le 2. Description of sensitivity analysis looking at potential
gestational age measurement error.
Additional le 3. Description of sensitivity analysis using multiple impu-
tation for missing data.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 9 of 9
Dellicour et al. Malar J (2015) 14:461
by the US Centers for Disease Control and Prevention (CDC), Division of
Parasitic Diseases and Malaria through a cooperative agreement with Kenya
Medical Research Institute (KEMRI), Center for Global Health Research (CGHR),
Kisumu, Kenya. The funders had no role in study design, data collection and
analysis, decision to publish, or preparation of the manuscript.
Competing interests
The authors declare that they have no competing interests.
Received: 11 August 2015 Accepted: 21 October 2015
References
1. WHO. World Malaria Report: 2013. Geneva: World Health Organization
2013.
2. Phillips-Howard PA, Steffen R, Kerr L, Vanhauwere B, Schildknecht J, Fuchs
E, et al. Safety of mefloquine and other antimalarial agents in the first
trimester of pregnancy. J Travel Med. 1998;5:121–6.
3. Phillips-Howard PA, Wood D. The safety of antimalarial drugs in preg-
nancy. Drug Saf. 1996;14:131–45.
4. Clark RL. Embryotoxicity of the artemisinin antimalarials and potential
consequences for use in women in the first trimester. Reprod Toxicol.
2009;28:285–96.
5. WHO, TDR. Assessment of the safety of artemisinin compounds in
pregnancy: report of two joint informal consultations convened in 2006.
Geneva: World Health Organization 2007.
6. Clark R, Kumemura M, Makori N, Nakata Y, Bernard F, Harrell A, et al.
Artesunate: developmental toxicity in monkeys. Bir Birth Defects
Research (Part A). 2006;76:329.
7. Kelemen E, Calvo W, Fliedner T. Atlas of Human Hemopoietic Develop-
ment. Berlin: 1979.
8. White TE, Clark RL. Sensitive periods for developmental toxicity of orally
administered artesunate in the rat. Birth Defects Res B Dev Reprod Toxi-
col. 2008;83:407–17.
9. Adam I, Elhassan EM, Omer EM, Abdulla MA, Mahgoub HM, Adam GK.
Safety of artemisinins during early pregnancy, assessed in 62 Sudanese
women. Ann Trop Med Parasitol. 2009;103:205–10.
10. Deen JL, von Seidlein L, Pinder M, Walraven GE, Greenwood BM. The
safety of the combination artesunate and pyrimethamine-sulfadoxine
given during pregnancy. Trans R Soc Trop Med Hyg. 2001;95:424–8.
11. Dellicour S, Hall S, Chandramohan D, Greenwood B. The safety of arte-
misinins during pregnancy: a pressing question. Malar J. 2007;6:15.
12. Manyando C, Mkandawire R, Puma L, Sinkala M, Mpabalwani E, Njunju E,
et al. Safety of artemether-lumefantrine in pregnant women with malaria:
results of a prospective cohort study in Zambia. Malar J. 2010;9:249.
13. McGready R, Lee SJ, Wiladphaingern J, Ashley EA, Rijken MJ, Boel M, et al.
Adverse effects of falciparum and vivax malaria and the safety of anti-
malarial treatment in early pregnancy: a population-based study. Lancet
Infect Dis. 2012;12:388–96.
14. Mosha D, Mazuguni F, Mrema S, Sevene E, Abdulla S, Genton B. Safety
of artemether-lumefantrine exposure in first trimester of pregnancy: an
observational cohort. Malar J. 2014;13:197.
15. Rulisa S, Kaligirwa N, Agaba S, Karema C, Mens PF, de Vries PJ. Pharma-
covigilance of artemether-lumefantrine in pregnant women followed
until delivery in Rwanda. Malar J. 2012;11:225.
16. WHO. Assessment of the safety of artemisinin compounds in pregnancy.
Report of two informal consultations convened by WHO in 2002. Geneva:
World Health Organization; 2003.
17. WHO. Guidelines for the treatment of malaria. Third edition. Geneva:
World Health Organization; 2015.
18. Dellicour S, ter Kuile FO, Stergachis A. Pregnancy exposure registries for
assessing antimalarial drug safety in pregnancy in malaria-endemic coun-
tries. PLoS Med. 2008;5:e187.
19. Huynh BT, Fievet N, Gbaguidi G, Dechavanne S, Borgella S, Guezo-Mevo
B, et al. Influence of the timing of malaria infection during pregnancy
on birth weight and on maternal anemia in Benin. Am J Trop Med Hyg.
2011;85:214–20.
20. Cottrell G, Mary JY, Barro D, Cot M. The importance of the period of
malarial infection during pregnancy on birth weight in tropical Africa. Am
J Trop Med Hyg. 2007;76:849–54.
21. Odhiambo FO, Laserson KF, Sewe M, Hamel MJ, Feikin DR, Adazu K, et al.
Profile: the KEMRI/CDC Health and Demographic Surveillance System-
Western Kenya. Int J Epidemiol. 2012;41:977–87.
22. Bigogo G, Audi A, Aura B, Aol G, Breiman RF, Feikin DR. Health-seeking
patterns among participants of population-based morbidity surveillance
in rural western Kenya: implications for calculating disease rates. Int J
Infect Dis. 2010;14:e967–73.
23. Feikin DR, Audi A, Olack B, Bigogo GM, Polyak C, Burke H, et al. Evalua-
tion of the optimal recall period for disease symptoms in home-based
morbidity surveillance in rural and urban Kenya. Int J Epidemiol.
2010;39:450–8.
24. McKenzie D. Measuring inequality with asset indicators. Journal of Popu-
lation Economics. 2005;18:229.
25. Sonis J. A closer look at confounding. Fam Med. 1998;30:584–8.
26. Kleinbaum DG, Klein M. Chapter 6: Modeling Strategy Guidelines. Logistic
Regression: A Self-Learning Text. 3rd edition ed.: Springer; 2010.
27. Radeva-Petrova D, Kayentao K, ter Kuile FO, Sinclair D, Garner P. Drugs
for preventing malaria in pregnant women in endemic areas: any drug
regimen versus placebo or no treatment. Cochrane Database Syst Rev.
2014;10:CD000169.
28. Sangaré L, Weiss N, Brentlinger P, Richardson B, Staedke S, Kiwuwa M,
et al. Patterns of antimalarial drug treatment among pregnant women in
Uganda. Malar J. 2011;10:152.
29. Achan J, Talisuna AO, Erhart A, Yeka A, Tibenderana JK, Baliraine FN, et al.
Quinine, an old anti-malarial drug in a modern world: role in the treat-
ment of malaria. Malar J. 2011;10:144.
30. Piola P, Nabasumba C, Turyakira E, Dhorda M, Lindegardh N, Nyehangane
D, et al. Efficacy and safety of artemether-lumefantrine compared with
quinine in pregnant women with uncomplicated Plasmodium falciparum
malaria: an open-label, randomised, non-inferiority trial. Lancet Infect Dis.
2010;10:762–9.
31. Clark RL. Effects of artemisinins on reticulocyte count and relationship to
possible embryotoxicity in confirmed and unconfirmed malarial patients.
Birth Defects Res A Clin Mol Teratol. 2012;94:61–75.
32. Newton PN, Green MD, Mildenhall DC, Plancon A, Nettey H, Nyadong
L, et al. Poor quality vital anti-malarials in Africa—an urgent neglected
public health priority. Malar J. 2011;10:352.
33. Jong De, van den Berg LT, Feenstra N, Sorensen HT, Cornel MC. Improve-
ment of drug exposure data in a registration of congenital anomalies.
Pilot-study: pharmacist and mother as sources for drug exposure data
during pregnancy. EuroMAP Group. Europen Medicine and Pregnancy
Group. Teratology. 1999;60:33–6.
34. Rockenbauer M, Olsen J, Czeizel AE, Pedersen L, Sorensen HT, Euro MAPG.
Recall bias in a case-control surveillance system on the use of medicine
during pregnancy. Epidemiology. 2001;12:461–6.
35. de Jong-van den Berg LT, Waardenburg CM, Haaijer-Ruskamp FM, Dukes
MN, Wesseling H. Drug use in pregnancy: a comparative appraisal of data
collecting methods. Eur J Clin Pharmacol. 1993;45:9–14.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... One was unavailable for this meta-analysis, and one did not record maternal malarial infection by LM, PCR or histology 18,19 . Of the remaining 11 studies included, five were observational and six were clinical trials measuring the effect of chemoprevention or insecticide-treated nets [20][21][22][23][24][25][26][27][28][29][30][31] . Nine studies were conducted in sub-Saharan African countries and two in PNG. ...
Article
Full-text available
In areas of moderate to intense Plasmodium falciparum transmission, malaria in pregnancy remains a significant cause of low birth weight, stillbirth, and severe anaemia. Previously, fetal sex has been identified to modify the risks of maternal asthma, pre-eclampsia, and gestational diabetes. One study demonstrated increased risk of placental malaria in women carrying a female fetus. We investigated the association between fetal sex and malaria in pregnancy in 11 pregnancy studies conducted in sub-Saharan African countries and Papua New Guinea through meta-analysis using log binomial regression fitted to a random-effects model. Malaria infection during pregnancy and delivery was assessed using light microscopy, polymerase chain reaction, and histology. Five studies were observational studies and six were randomised controlled trials. Studies varied in terms of gravidity, gestational age at antenatal enrolment and bed net use. Presence of a female fetus was associated with malaria infection at enrolment by light microscopy (risk ratio 1.14 [95% confidence interval 1.04, 1.24]; P = 0.003; n = 11,729). Fetal sex did not associate with malaria infection when other time points or diagnostic methods were used. There is limited evidence that fetal sex influences the risk of malaria infection in pregnancy.
... As first-trimester pregnancy is the only indication for oral quinine, the supply of quinine and clindamycin is problematic in many countries; in some parts of sub-Saharan Africa, quinine is rarely available in public facilities and most first-trimester malaria is already treated with first-line ACTs. [53][54][55][56][57] Although a cost-effectiveness analysis is beyond the scope of this paper, our results suggest that artemetherlumefantrine, and possibly other ACTs, are likely to be more cost-effective than quinine-clindamycin because of the disability-adjusted life-year associated with poorly treated malaria, the simple antimalarial supply management, and the case management of women of childbearing age that does not require screening for pregnancy before treatment. ...
Article
Background Malaria in the first trimester of pregnancy is associated with adverse pregnancy outcomes. Artemisinin-based combination therapies (ACTs) are a highly effective, first-line treatment for uncomplicated Plasmodium falciparum malaria, except in the first trimester of pregnancy, when quinine with clindamycin is recommended due to concerns about the potential embryotoxicity of artemisinins. We compared adverse pregnancy outcomes after artemisinin-based treatment (ABT) versus non-ABTs in the first trimester of pregnancy. Methods For this systematic review and individual patient data (IPD) meta-analysis, we searched MEDLINE, Embase, and the Malaria in Pregnancy Library for prospective cohort studies published between Nov 1, 2015, and Dec 21, 2021, containing data on outcomes of pregnancies exposed to ABT and non-ABT in the first trimester. The results of this search were added to those of a previous systematic review that included publications published up until November, 2015. We included pregnancies enrolled before the pregnancy outcome was known. We excluded pregnancies with missing estimated gestational age or exposure information, multiple gestation pregnancies, and if the fetus was confirmed to be unviable before antimalarial treatment. The primary endpoint was adverse pregnancy outcome, defined as a composite of either miscarriage, stillbirth, or major congenital anomalies. A one-stage IPD meta-analysis was done by use of shared-frailty Cox models. This study is registered with PROSPERO, number CRD42015032371. Findings We identified seven eligible studies that included 12 cohorts. All 12 cohorts contributed IPD, including 34 178 pregnancies, 737 with confirmed first-trimester exposure to ABTs and 1076 with confirmed first-trimester exposure to non-ABTs. Adverse pregnancy outcomes occurred in 42 (5·7%) of 736 ABT-exposed pregnancies compared with 96 (8·9%) of 1074 non-ABT-exposed pregnancies in the first trimester (adjusted hazard ratio [aHR] 0·71, 95% CI 0·49–1·03). Similar results were seen for the individual components of miscarriage (aHR=0·74, 0·47–1·17), stillbirth (aHR=0·71, 0·32–1·57), and major congenital anomalies (aHR=0·60, 0·13–2·87). The risk of adverse pregnancy outcomes was lower with artemether–lumefantrine than with oral quinine in the first trimester of pregnancy (25 [4·8%] of 524 vs 84 [9·2%] of 915; aHR 0·58, 0·36–0·92). Interpretation We found no evidence of embryotoxicity or teratogenicity based on the risk of miscarriage, stillbirth, or major congenital anomalies associated with ABT during the first trimester of pregnancy. Given that treatment with artemether–lumefantrine was associated with fewer adverse pregnancy outcomes than quinine, and because of the known superior tolerability and antimalarial effectiveness of ACTs, artemether–lumefantrine should be considered the preferred treatment for uncomplicated P falciparum malaria in the first trimester. If artemether–lumefantrine is unavailable, other ACTs (except artesunate–sulfadoxine–pyrimethamine) should be preferred to quinine. Continued active pharmacovigilance is warranted. Funding Medicines for Malaria Venture, WHO, and the Worldwide Antimalarial Resistance Network funded by the Bill & Melinda Gates Foundation.
... If there is any doubt as to the microscopic diagnosis, alternative treatments include quinine and clindamycin, as per WHO guidelines for falciparum and vivax malaria (World Health Organization, 2021). These guidelines are based on published prospective data from 700 women exposed to ACT in the first trimester of pregnancy (Dellicour et al., 2015;McGready et al., 2012;Moore et al., 2016;Mosha et al., 2014), which indicate no adverse effects on the pregnancy or the health of the foetus or neonate, and are sufficient to exclude a ≥4.2-fold increase in risk of any major birth defect (background prevalence assumed to be 0.9%), if half the exposures occur during the embryo-sensitive period (4-9 weeks post conception) (World Health Organization, 2021). These data can be used to reassure women who are accidentally exposed to ACT during the first trimester of pregnancy. ...
Chapter
The zoonotic parasite Plasmodium knowlesi has emerged as an important cause of human malaria in parts of Southeast Asia. The parasite is indistinguishable by microscopy from the more benign P. malariae, but can result in high parasitaemias with multiorgan failure, and deaths have been reported. Recognition of severe knowlesi malaria, and prompt initiation of effective therapy is therefore essential to prevent adverse outcomes. Here we review all studies reporting treatment of uncomplicated and severe knowlesi malaria. We report that although chloroquine is effective for the treatment of uncomplicated knowlesi malaria, artemisinin combination treatment is associated with faster parasite clearance times and lower rates of anaemia during follow-up, and should be considered the treatment of choice, particularly given the risk of administering chloroquine to drug-resistant P. vivax or P. falciparum misdiagnosed as P. knowlesi malaria in co-endemic areas. For severe knowlesi malaria, intravenous artesunate has been shown to be highly effective and associated with reduced case-fatality rates, and should be commenced without delay. Regular paracetamol may also be considered for patients with severe knowlesi malaria or for those with acute kidney injury, to attenuate the renal damage resulting from haemolysis-induced lipid peroxidation.
... If there is any doubt as to the microscopic diagnosis, alternative treatments include quinine and clindamycin, as per WHO guidelines for falciparum and vivax malaria (World Health Organization, 2021). These guidelines are based on published prospective data from 700 women exposed to ACT in the first trimester of pregnancy (Dellicour et al., 2015;McGready et al., 2012;Moore et al., 2016;Mosha et al., 2014), which indicate no adverse effects on the pregnancy or the health of the foetus or neonate, and are sufficient to exclude a !4.2-fold increase in risk of any major birth defect (background prevalence assumed to be 0.9%), if half the exposures occur during the embryo-sensitive period (4-9 weeks post conception) (World Health Organization, 2021). These data can be used to reassure women who are accidentally exposed to ACT during the first trimester of pregnancy. ...
Chapter
Plasmodium knowlesi, a simian malaria parasite of great public health concern has been reported from most countries in Southeast Asia and exported to various countries around the world. Currently P. knowlesi is the predominant species infecting humans in Malaysia. Besides this species, other simian malaria parasites such as P. cynomolgi and P. inui are also infecting humans in the region. The vectors of P. knowlesi and other Asian simian malarias belong to the Leucosphyrus Group of Anopheles mosquitoes which are generally forest dwelling species. Continual deforestation has resulted in these species moving into forest fringes, farms, plantations and human settlements along with their macaque hosts. Limited studies have shown that mosquito vectors are attracted to both humans and macaque hosts, preferring to bite outdoors and in the early part of the night. We here review the current status of simian malaria vectors and their parasites, knowledge of vector competence from experimental infections and discuss possible vector control measures. The challenges encountered in simian malaria elimination are also discussed. We highlight key knowledge gaps on vector distribution and ecology that may impede effective control strategies.
... 1,2 This also occurs in prospective cohort studies where patients are enrolled after an initiating event of interest. 3 In time-to-event analyses, if a patient enters the study or database after the start of their follow-up or index time, they are said to have delayed entry. ...
Preprint
Full-text available
In real world data (RWD) studies, observed datasets are often subject to left truncation, which can bias estimates of survival parameters. Standard methods can only suitably account for left truncation when survival and entry time are independent. Therefore, in the dependent left truncation setting, it is important to quantify the magnitude and direction of estimator bias to determine whether an analysis provides valid results. We conduct simulation studies of common RWD analytic settings in order to determine when standard analysis provides reliable estimates, and to identify factors that contribute most to estimator bias. We also outline a procedure for conducting a simulation-based sensitivity analysis for an arbitrary dataset subject to dependent left truncation. Our simulation results show that when comparing a truncated real-world arm to a non-truncated arm, we observe the estimated hazard ratio biased upwards, providing conservative inference. The most important data-generating parameter contributing to bias is the proportion of left truncated patients, given any level of dependence between survival and entry time. For specific datasets and analyses that may differ from our example, we recommend applying our sensitivity analysis approach to determine how results would change given varying proportions of truncation.
Book
Full-text available
Based on a review of the evidence conducted in 2022, WHO now recommends artemether–lumefantrine, the ACT with the most human safety data available, as the preferred treatment for uncomplicated P. falciparum malaria in the first trimester of pregnancy. This document presents all relevant evidence on the effects and safety in early pregnancy of artemisinins and partner medicines used in ACTs from both studies in experimental animals and observational studies in humans.
Article
There have been recent calls for the use of artemisinin-based combination therapies (ACTs) for uncomplicated malaria in the first trimester of pregnancy. Nevertheless, the 2021 WHO Guidelines for Malaria reaffirmed their position that there is not adequate clinical safety data on artemisinins to support that usage. The WHO’s position is consistent with several issues with the existing clinical data. First, first trimester safety results from multiple ACTs were lumped in a meta-analysis which does not demonstrate that each of the included ACTs is equally safe. Second, safety results from all periods of the first trimester were lumped in the meta-analysis which does not demonstrate the same level of safety for all subperiods, particularly gestational Weeks 6 to 8 which is likely to be the most sensitive period. Third, even if there is evidence of a lack of an effect on miscarriage for a particular ACT, it does not follow then there are no developmental effects for any ACT. In monkeys, artesunate caused marked embryonal anemia leading to embryo death but the long-term consequences of lower levels of embryonal anemia are not known. Fourth, there have been advances in the sensitivity and usage of rapid diagnostic tests that will lead to diagnoses of malaria earlier in gestation which is less well studied and more likely sensitive to artemisinins. Any clinical studies of the safety of ACTs in the first trimester need to evaluate the results of treatment with individual ACTs during different 1- to 2-week periods of the first trimester.
Article
Malaria is a particular problem in pregnancy because of enhanced sensitivity, the possibility of placental malaria, and adverse effects on pregnancy outcome. Artemisinin‐containing combination therapies (ACTs) are the most effective antimalarials known. WHO recommends 7‐day quinine therapy for uncomplicated Plasmodium falciparum malaria in the first trimester despite the superior tolerability and efficacy of 3‐day ACT regimens because artemisinins caused embryolethality and/or cardiovascular malformations at relatively low doses in rats, rabbits, and monkeys. The developmental toxicity of artesunate, artemether, and DHA were similar in rats but artesunate was embryotoxic at lower doses in rabbits (5 mg/kg/day) than artemether (no effect level = 25 mg/kg/day). In clinical studies in Africa, treatment with artemether–lumefantrine in the first trimester was observed to be highly efficacious and the miscarriage rate (≤3.1%) was similar to no antimalarial treatment (2.6%). When data from the first‐trimester use of largely artesunate‐based therapies in Thailand were pooled together, there was no difference in miscarriage rate compared to quinine. However, individually, artesunate–mefloquine was associated with a higher miscarriage rate (15/71 = 21%) compared to other artemisinin‐based therapies including 7‐day artesunate + clindamycin (2/50 = 4%) and quinine (92/842 = 11%). Thus, appropriate statistical comparisons of individual ACT groups are needed prior to assuming that they all have the same risk for developmental toxicity. Current limitations in the assessment of the safety of ACTs in the first trimester are a lack of exposures early in gestation (gestational weeks 6–7), limited postnatal evaluation for cardiovascular malformations, and the pooling of all ACTs for the assessment of risk.
Article
Full-text available
Artemisinin combination therapy (ACT) is recommended by the World Health Organization (WHO) as first line treatment for uncomplicated malaria both in adults and children. During pregnancy, ACT is considered safe only in the second and third trimester, since animal studies have demonstrated that artemisinin derivatives can cause foetal death and congenital malformation within a narrow time window in early embryogenesis. During this period, artemisinin derivatives induce defective embryonic erythropoiesis and vasculogenesis/angiogenesis in experimental models. However, clinical data on the safety profile of ACT in pregnant women have not shown an increased risk of miscarriage, stillbirth, or congenital malformation, nor low birth weight, associated with exposure to artemisinins in the first trimester. Although further studies are needed, the evidence collected up to now is prompting the WHO towards a change in the guidelines for the treatment of uncomplicated malaria, allowing the use of ACT also in the first trimester of pregnancy.
Article
Introduction The association between pregnancy and altered drug pharmacokinetic (PK) properties is acknowledged, as is its impact on drug plasma concentrations and thus therapeutic efficacy. However, there have been few robust PK studies of antimalarial use in pregnancy. Given that inadequate dosing for prevention or treatment of malaria in pregnancy can result in negative maternal/infant outcomes, along with the potential to select for parasite drug resistance, it is imperative that reliable pregnancy-specific dosing recommendations are established. Areas covered PK studies of antimalarial drugs in pregnancy. The present review summarizes the efficacy and PK properties of WHO-recommended therapies used in pregnancy, with a focus on PK studies published since 2014. Expert opinion Changes in antimalarial drug disposition in pregnancy are well described, yet pregnant women continue to receive treatment regimens optimized for non-pregnant adults. Contemporary in silico modelling has recently identified a series of alternative dosing regimens that are predicted to provide optimal therapeutic efficacy for pregnant women.
Article
Full-text available
In western Kenya, maternal mortality is a major public health problem estimated at 730/100,000 live births, higher than the Kenyan national average of 488/100,000 women. Many women do not attend antenatal care (ANC) in the first trimester, half do not receive 4 ANC visits. A high proportion use traditional birth attendants (TBA) for delivery and 1 in five deliver unassisted. The present study was carried out to ascertain why women do not fully utilise health facility ANC and delivery services. A qualitative study using 8 focus group discussions each consisting of 8-10 women, aged 15-49 years. Thematic analysis identified the main barriers and facilitators to health facility based ANC and delivery. Attending health facility for ANC was viewed positively. Three elements of care were important; testing for disease including HIV, checking the position of the foetus, and receiving injections and / or medications. Receiving a bed net and obtaining a registration card were also valuable. Four barriers to attending a health facility for ANC were evident; attitudes of clinic staff, long clinic waiting times, HIV testing and cost, although not all women felt the cost was prohibitive being worth it for the health of the child. Most women preferred to deliver in a health facility due to better management of complications. However cost was a barrier, and a reason to visit a TBA because of flexible payment. Other barriers were unpredictable labour and transport, staff attitudes and husbands' preference. Our findings suggest that women in western Kenya are amenable to ANC and would be willing and even prefer to deliver in a healthcare facility, if it were affordable and accessible to them. However for this to happen there needs to be investment in health promotion, and transport, as well as reducing or removing all fees associated with antenatal and delivery care. Yet creating demand for service will need to go alongside investment in antenatal services at organisational, staffing and facility level in order to meet both current and future increase in demand.
Article
Full-text available
There is limited data available regarding safety profile of artemisinins in early pregnancy. They are, therefore, not recommended by WHO as a first-line treatment for malaria in first trimester due to associated embryo-foetal toxicity in animal studies. The study assessed birth outcome among pregnant women inadvertently exposed to artemether-lumefantrine (AL) during first trimester in comparison to those of women exposed to other anti-malarial drugs or no drug at all during the same period of pregnancy. Pregnant women with gestational age <20 weeks were recruited from Maternal Health clinics or from monthly house visits (demographic surveillance), and followed prospectively until delivery. 2167 pregnant women were recruited and 1783 (82.3%) completed the study until delivery. 319 (17.9%) used anti-malarials in first trimester, of whom 172 (53.9%) used (AL), 78 (24.4%) quinine, 66 (20.7%) sulphadoxine-pyrimethamine (SP) and 11 (3.4%) amodiaquine. Quinine exposure in first trimester was associated with an increased risk of miscarriage/stillbirth (OR 2.5; 1.3–5.1) and premature birth (OR 2.6; 1.3–5.3) as opposed to AL with (OR 1.4; 0.8–2.5) for miscarriage/stillbirth and (OR 0.9; 0.5–1.8) for preterm birth. Congenital anomalies were identified in 4 exposure groups namely AL only (1/164[0.6%]), quinine only (1/70[1.4%]), SP (2/66[3.0%]), and non-anti-malarial exposure group (19/1464[1.3%]). Exposure to AL in first trimester was more common than to any other anti-malarial drugs. Quinine exposure was associated with adverse pregnancy outcomes which was not the case following other anti-malarial intake. Since AL and quinine were used according to their availability rather than to disease severity, it is likely that the effect observed was related to the drug and not to the disease itself. Even with this caveat, a change of policy from quinine to AL for the treatment of uncomplicated malaria during the whole pregnancy period could be already envisaged.
Article
Full-text available
The KEMRI/Centers for Disease Control and Prevention (CDC) Health and Demographic Surveillance System (HDSS) is located in Rarieda, Siaya and Gem Districts (Siaya County), lying northeast of Lake Victoria in Nyanza Province, western Kenya. The KEMRI/CDC HDSS, with approximately 220 000 inhabitants, has been the foundation for a variety of studies, including evaluations of insecticide-treated bed nets, burden of diarrhoeal disease and tuberculosis, malaria parasitaemia and anaemia, treatment strategies and immunological correlates of malaria infection, and numerous HIV, tuberculosis, malaria and diarrhoeal disease treatment and vaccine efficacy and effectiveness trials for more than a decade. Current studies include operations research to measure the uptake and effectiveness of the programmatic implementation of integrated malaria control strategies, HIV services, newly introduced vaccines and clinical trials. The HDSS provides general demographic and health information (such as population age structure and density, fertility rates, birth and death rates, in- and out-migrations, patterns of health care access and utilization and the local economics of health care) as well as disease- or intervention-specific information. The HDSS also collects verbal autopsy information on all deaths. Studies take advantage of the sampling frame inherent in the HDSS, whether at individual, household/compound or neighbourhood level.
Article
Full-text available
The World Health Organization presently recommends Artemisinin-based combination therapy (ACT) as first-line therapy for uncomplicated P. falciparum malaria. Many malaria-endemic countries, including Rwanda, have adopted these treatment guidelines. The Artemisinin derivative Artemether, in combination with lumefantrine, is currently used in Rwanda for malaria during the second and third trimesters of pregnancy. Safety data on the use of ACT in pregnancy are still limited though and more data are needed. In this pharmacovigilance study, the exposed group (pregnant women with malaria given artemether-lumefantrine), and a matched non-exposed group (pregnant women without malaria and no exposure to artemether-lumefantrine) were followed until delivery. Data were collected at public health centres all over Rwanda during acute malaria, routine antenatal visits, after hospital delivery or within 48 hours after home delivery. Information gathered from patients included routine antenatal and peri-partum data, pregnancy outcomes (abortions, stillbirths, at term delivery), congenital malformations and other adverse events through history taking and physical examination of both mothers and newborns. The outcomes for the total sample of 2,050 women were for the treatment (n=1,072) and control groups (n=978) respectively: abortions: 1.3% and 0.4%; peri-natal mortality 3.7% and 2.8%; stillbirth 2.9% and 2.4%; neonatal death [less than or equal to]7 days after birth 0.5% and 0.4%; premature delivery 0.7% and 0.3%; congenital malformations 0.3% and 0.3%. A total of 129 obstetric adverse events in 127 subjects were reported (7.3% in the treatment group, 5.0% in the control group). In a multivariate regression model, obstetric complications were more frequent in the treatment group (OR (95% CI): 1.38 (0.95, 2.01)), and in primigravidae (OR (95% CI) 2.65 (1.71, 4.12) and at higher age (OR per year: 1.05 (1.01-1.09). There were no specific safety concerns related to artemether-lumefantrine treatment for uncomplicated falciparum malaria in pregnancy. However, more obstetric complications were observed in the treatment group. These increased occurrence of complications could, however, be caused by the malaria episode itself, but further assessment is required.
Article
Background: Pregnancy increases the risk of malaria and this is associated with poor health outcomes for both the mother and the infant, especially during the first or second pregnancy. To reduce these effects, the World Health Organization recommends that pregnant women living in malaria endemic areas sleep under insecticide-treated bednets, are treated for malaria illness and anaemia, and receive chemoprevention with an effective antimalarial drug during the second and third trimesters. Objectives: To assess the effects of malaria chemoprevention given to pregnant women living in malaria endemic areas on substantive maternal and infant health outcomes. We also summarised the effects of intermittent preventive treatment with sulfadoxine-pyrimethamine (SP) alone, and preventive regimens for Plasmodium vivax. Search methods: We searched the Cochrane Infectious Diseases Group Specialized Register, CENTRAL, MEDLINE, EMBASE, LILACS, and reference lists up to 1 June 2014. Selection criteria: Randomized controlled trials (RCTs) and quasi-RCTs of any antimalarial drug regimen for preventing malaria in pregnant women living in malaria-endemic areas compared to placebo or no intervention. In the mother, we sought outcomes that included mortality, severe anaemia, and severe malaria; anaemia, haemoglobin values, and malaria episodes; indicators of malaria infection, and adverse events. In the baby, we sought foetal loss, perinatal, neonatal and infant mortality; preterm birth and birthweight measures; and indicators of malaria infection. We included regimens that were known to be effective against the malaria parasite at the time but may no longer be used because of parasite drug resistance. Data collection and analysis: Two review authors applied inclusion criteria, assessed risk of bias and extracted data. Dichotomous outcomes were compared using risk ratios (RR), and continuous outcomes using mean differences (MD); both are presented with 95% confidence intervals (CI). We assessed the quality of evidence using the GRADE approach. Main results: Seventeen trials enrolling 14,481 pregnant women met our inclusion criteria. These trials were conducted between 1957 and 2008, in Nigeria (three trials), The Gambia (three trials), Kenya (three trials), Mozambique (two trials), Uganda (two trials), Cameroon (one trial), Burkina Faso (one trial), and Thailand (two trials). Six different antimalarials were evaluated against placebo or no intervention; chloroquine (given weekly), pyrimethamine (weekly or monthly), proguanil (daily), pyrimethamine-dapsone (weekly or fortnightly), and mefloquine (weekly), or intermittent preventive therapy with SP (given twice, three times or monthly). Trials recruited women in their first or second pregnancy (eight trials); only multigravid women (one trial); or all women (eight trials). Only six trials had adequate allocation concealment.For women in their first or second pregnancy, malaria chemoprevention reduces the risk of moderate to severe anaemia by around 40% (RR 0.60, 95% CI 0.47 to 0.75; three trials, 2503 participants, high quality evidence), and the risk of any anaemia by around 17% (RR 0.83, 95% CI 0.74 to 0.93; five trials,, 3662 participants, high quality evidence). Malaria chemoprevention reduces the risk of antenatal parasitaemia by around 61% (RR 0.39, 95% CI 0.26 to 0.58; seven trials, 3663 participants, high quality evidence), and two trials reported a reduction in febrile illness (low quality evidence). There were only 16 maternal deaths and these trials were underpowered to detect an effect on maternal mortality (very low quality evidence).For infants of women in their first and second pregnancies, malaria chemoprevention probably increases mean birthweight by around 93 g (MD 92.72 g, 95% CI 62.05 to 123.39; nine trials, 3936 participants, moderate quality evidence), reduces low birthweight by around 27% (RR 0.73, 95% CI 0.61 to 0.87; eight trials, 3619 participants, moderate quality evidence), and reduces placental parasitaemia by around 46% (RR 0.54, 95% CI 0.43 to 0.69; seven trials, 2830 participants, high quality evidence). Fewer trials evaluated spontaneous abortions, still births, perinatal deaths, or neonatal deaths, and these analyses were underpowered to detect clinically important differences.In multigravid women, chemoprevention has similar effects on antenatal parasitaemia (RR 0.38, 95% CI 0.28 to 0.50; three trials, 977 participants, high quality evidence)but there are too few trials to evaluate effects on other outcomes.In trials giving chemoprevention to all pregnant women irrespective of parity, the average effects of chemoprevention measured in all women indicated it may prevent severe anaemia (defined by authors, but at least < 8 g/L: RR 0.19, 95% CI 0.05 to 0.75; two trials, 1327 participants, low quality evidence), but consistent benefits have not been shown for other outcomes.In an analysis confined only to intermittent preventive therapy with SP, the estimates of effect and the quality of the evidence were similar.A summary of a single trial in Thailand of prophylaxis against P. vivax showed chloroquine prevented vivax infection (RR 0.01, 95% CI 0.00 to 0.20; one trial, 942 participants). Authors' conclusions: Routine chemoprevention to prevent malaria and its consequences has been extensively tested in RCTs, with clinically important benefits on anaemia and parasitaemia in the mother, and on birthweight in infants.
Article
We examined the possibilities of improving the retrospective collection of data an drug use during pregnancy. The European Registration of Congenital Anomalies (EUROCAT) has registered information on maternal drug exposure in the northern Netherlands through a question on the notification form for the registration of birth defects, filled out by physicians or midwives since 1981. Furthermore, hospital records are used and general practitioners are asked to add information on drug use. The present pilot study used pharmacy records and maternal questionnaires as well as maternal interview data to complete the data on drug exposure in the EUROCAT registration. Combined information from pharmacies, questionnaires, and interviews with the mother were used as the reference standard. Pharmacy records provided detailed data on 57% of drugs used (prescription drugs, mainly). Mothers were able to report 76% of drug groups used, but when only data on the exact name of the drug were studied, this figure was 52%. Of the drugs dispensed by the pharmacy, 6% were not used (false positives). We conclude that pharmacy records and maternal interviews are both indispensable sources of information on maternal drug exposure that provide much added value. (C) 1999 Wiley-Liss, Inc.
Article
We examined the possibilities of improving the retrospective collection of data on drug use during pregnancy. The European Registration of Congenital Anomalies (EUROCAT) has registered information on maternal drug exposure in the northern Netherlands through a question on the notification form for the registration of birth defects, filled out by physicians or midwives since 1981. Furthermore, hospital records are used and general practitioners are asked to add information on drug use. The present pilot study used pharmacy records and maternal questionnaires as well as maternal interview data to complete the data on drug exposure in the EUROCAT registration. Combined information from pharmacies, questionnaires, and interviews with the mother were used as the reference standard. Pharmacy records provided detailed data on 57% of drugs used (prescription drugs, mainly). Mothers were able to report 76% of drug groups used, but when only data on the exact name of the drug were studied, this figure was 52%. Of the drugs dispensed by the pharmacy, 6% were not used (false positives). We conclude that pharmacy records and maternal interviews are both indispensable sources of information on maternal drug exposure that provide much added value. Teratology 60:33–36, 1999. © 1999 Wiley-Liss, Inc.