ArticlePDF Available

Safety of Daily Co-Trimoxazole in Pregnancy in an Area of Changing Malaria Epidemiology: A Phase 3b Randomized Controlled Clinical Trial

PLOS
PLOS ONE
Authors:
  • The Copperbelt University, School of Medicine, Ndola, Zambia
  • University of Zambia, School of Public Health

Abstract and Figures

Antibiotic therapy during pregnancy may be beneficial and impacts positively on the reduction of adverse pregnancy outcomes. No studies have been done so far on the effects of daily Co-trimoxazole (CTX) prophylaxis on birth outcomes. A phase 3b randomized trial was conducted to establish that daily CTX in pregnancy is not inferior to SP intermittent preventive treatment (IPT) in reducing placental malaria; preventing peripheral parasitaemia; preventing perinatal mortality and also improving birth weight. To establish its safety on the offspring by measuring the gestational age and birth weight at delivery, and compare the safety and efficacy profile of CTX to that of SP. Pregnant women (HIV infected and uninfected) attending antenatal clinic were randomized to receive either daily CTX or sulfadoxine-pyrimethamine as per routine IPT. Safety was assessed using standard and pregnancy specific measurements. Women were followed up monthly until delivery and then with their offspring up to six weeks after delivery. Data from 346 pregnant women (CTX = 190; SP = 156) and 311 newborns (CTX = 166 and SP = 145) showed that preterm deliveries (CTX 3.6%; SP 3.0%); still births (CTX 3.0%; SP 2.1%), neonatal deaths (CTX 0%; SP 1.4%), and spontaneous abortions (CTX 0.6%; SP 0%) were similar between study arms. The low birth weight rates were 9% for CTX and 13% for SP. There were no birth defects reported. Both drug exposure groups had full term deliveries with similar birth weights (mean of 3.1 Kg). The incidence and severity of AEs in the two groups were comparable. Exposure to daily CTX in pregnancy may not be associated with particular safety risks in terms of birth outcomes such as preterm deliveries, still births, neonatal deaths and spontaneous abortions compared to SP. However, more data are required on CTX use in pregnant women both among HIV infected and un-infected individuals. Clinicaltrials.gov NCT00711906.
Content may be subject to copyright.
Safety of Daily Co-Trimoxazole in Pregnancy in an Area
of Changing Malaria Epidemiology: A Phase 3b
Randomized Controlle d Clinical Trial
Christine Manyando
1,5
*, Eric M. Njunju
1,5
, David Mwakazanga
1
, Gershom Chongwe
1
,
Rhoda Mkandawire
2
, Davies Champo
1
, Modest Mulenga
1
, Maaike De Crop
3
, Yves Claeys
3
,
Raffaella M. Ravinetto
3,6
, Ch antal van Overmeir
3
, Umberto D’ Alessandro
3,4
, Jean-Pierre Van
geertruyden
5
1 Tropical Diseases Research Centre, Ndola, Zambia, 2 Choma District Health Management Team, Choma, Zambia, 3 Institute of Tropical Medicine, Antwerp, Belgium,
4 Medical Research Council, Fajara, The Gambia, 5 International Health Unit, University of Antwerp, Antwerp, Belgium, 6 Department of Pharmaceutical and
Pharmacological Sciences, KU Leuven, Leuven, Belgium
Abstract
Introduction:
Antibiotic therapy during pregnancy may be beneficial and impacts positively on the reduction of adverse
pregnancy outcomes. No studies have been done so far on the effects of daily Co-trimoxazole (CTX) prophylaxis on birth
outcomes. A phase 3b randomized trial was conducted to establish that daily CTX in pregnancy is not inferior to SP
intermittent preventive treatment (IPT) in reducing placental malaria; preventing peripheral parasitaemia; preventing
perinatal mortality and also improving birth weight. To establish its safety on the offspring by measuring the gestational
age and birth weight at delivery, and compare the safety and efficacy profile of CTX to that of SP.
Methods:
Pregnant women (HIV infected and uninfected) attending antenatal clinic were randomized to receive either daily
CTX or sulfadoxine-pyrimethamine as per routine IPT. Safety was assessed using standard and pregnancy specific
measurements. Women were followed up monthly until delivery and then with their offspring up to six weeks after delivery.
Results:
Data from 346 pregnant women (CTX = 190; SP = 156) and 311 newborns (CTX = 166 and SP = 145) showed that
preterm deliveries (CTX 3.6%; SP 3.0%); still births (CTX 3.0%; SP 2.1%), neonatal deaths (CTX 0%; SP 1.4%), and spontaneous
abortions (CTX 0.6%; SP 0%) were similar between study arms. The low birth weight rates were 9% for CTX and 13% for SP.
There were no birth defects reported. Both drug exposure groups had full term deliveries with similar birth weights (mean
of 3.1 Kg). The incidence and severity of AEs in the two groups were comparable.
Conclusion:
Exposure to daily CTX in pregnancy may not be associated with particular safety risks in terms of birth
outcomes such as preterm deliveries, still births, neonatal deaths and spontaneous abortions compared to SP. However,
more data are required on CTX use in pregnant women both among HIV infected and un-infected individuals.
Trial Registration:
Clinicaltrials.gov NCT00711906.
Citation: Manyando C, Njunju EM, Mwakazanga D, Chongwe G, Mkandawire R, et al. (2014) Safety of Daily Co-Trimoxazole in Pregnancy in an Area of Changing
Malaria Epidemiology: A Phase 3b Randomized Controlled Clinical Trial. PLoS ONE 9(5): e96017. doi:10.1371/journal.pone.0096017
Editor: Philip Bejon, Kenya Medical Research Institute (KEMRI), Kenya
Received January 13, 2014; Accepted March 31, 2014; Published May 15, 2014
Copyright: ß 2014 Manyando et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Funding: The study was funded under a program for institutional capacity strengthening, funded by the Belgian Development Cooperation and coordinated by
the Institute of Tropical Medicine in Antwerp. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the
manuscript.
Competing Interests: The authors have declared that no competing interests exist.
* E-mail: cmanyando@yahoo.com
Introduction
Malaria is one of the most important causes of morbidity and
mortality worldwide, with children and pregnant women being the
most severely affected groups [1]. In low transmission areas, all
pregnant women have little or no pre-existing immunity and
malaria can evolve towards severe disease with a higher risk of
maternal and perinatal mortality. Foetal and perinatal loss can be
as high as 60–70% [2–6]. In high transmission areas, primigrav-
idae are more at risk than multigravidae and malaria infection is
associated with maternal anaemia, low birth weight (LBW) and
stillbirth [7,8]. Annually, malaria during pregnancy is estimated to
account for 5% of the cases of severe anaemia in pregnant women,
approximately 35% of preventable low birth weight, 3–8% of
infant mortality and 5,000 to 200,000 infant deaths [9].
Anaemia, malnutrition and HIV infection are also common
events in malaria endemic areas and contribute to LBW. HIV
infection in pregnancy is associated with an increased risk of
malaria infection and higher parasite densities [10–12]. The risk
increment is more pronounced in multigravidae than in primi-
PLOS ONE | www.plosone.org 1 May 2014 | Volume 9 | Issue 5 | e96017
gravidae, indicating that HIV-1 hinders the development of
immunity [10,13,14]. HIV-1 infection is also associated with lower
birth weight [15], higher infant mortality and a 4-fold greater risk
of malaria attack in the new-born [16]. The increased risk of
placental malaria in HIV infected mothers is also associated with a
higher post-natal mortality [17]. The risk of anaemia suggests a
synergistic interaction between HIV and malaria, placing dually
infected women at very high risk of developing severe anaemia
[18,19].
The World Health Organization (WHO) recommends a
package of interventions for the prevention and control of malaria
in pregnancy. This comprises intermittent preventive treatment
(IPTp), use of insecticide treated materials (ITMs) [20,21], and
rapid access to effective case management for malaria illness and
anaemia [22]. Prevention with efficacious antimalarials can reduce
the incidence of placental malaria, LBW and maternal anaemia
[23,24]. IPTp with sulfadoxine/pyrimethamine (IPTp-SP) has
been proven to be an effective strategy to reduce the burden of
malaria during pregnancy. It is based on administering at least 2
treatment doses of SP to pregnant women after quickening
(around 18–20 weeks) at the Antenatal Care (ANC) centres
[25].The dose of SP should be given at not less than one month
interval as stipulated in the updated WHO IPTp guidelines, which
recommends IPTp at every scheduled antenatal visit [26]. HIV-1
infection may decrease the efficacy of IPTp-SP but 2 or more
doses of SP in the second and third trimester still reduce peripheral
and placental malaria, and maternal anaemia, including severe
anaemia, and increase birth weights [27,28]. Unfortunately, the
beneficial effect of IPTp-SP is currently threatened by increasing
resistance to SP. Therefore, other antimalarials to be used as IPTp
are currently investigated.
Cotrimoxazole (CTX) has been used for treating malaria in
children; recently, its daily use by non-pregnant HIV-infected
adults was associated with 70% reduction in the incidence of
clinical malaria [18,29,30]. Antibiotic therapy during pregnancy
may be beneficial and reduce some adverse pregnancy outcomes
[31]. CTX prophylaxis significantly improves birth outcomes in
HIV infected women with ,200 CD4 cells/ml with reduction of
chorionamnionitis, prematurity and neonatal mortality [32]. A
study conducted in Zambia using historical controls concluded
that antenatal provision of CTX was beneficial for HIV-infected
pregnant women with low CD4 count but not in women with $
200 CD4 cells/ml [32]. However, it is important to note that this
study was conducted in an area of very low malaria risk; CTX may
have had a different impact if malaria transmission had been
substantial. Though daily CTX could be considered as a potential
alternative for IPTp-SP, no information on its effectiveness in
preventing malaria infection during pregnancy and its conse-
quences (maternal anaemia and LBW) is available [33].
A clinical trial comparing the protective efficacy of daily CTX
versus IPTp-SP in pregnant women was carried out in southern
Zambia. Its primary objective was to test the hypothesis that CTX
prophylaxis is not inferior to SP prophylaxis in reducing placental
malaria. The secondary objectives were to evaluate efficacy of
CTX prophylaxis in preventing malaria peripheral parasitaemia,
perinatal mortality and in improving birth weight; to establish the
safety on the offspring by measuring the gestational age and birth
weight at delivery, and also to compare the safety and efficacy of
CTX prophylaxis to that of SP based on these parameters. The
study participants were stratified by HIV status in order to follow
the national preventive guidelines, taking note of the fact that
pregnant women with CD4 count ,200 cells/ml (at the time of the
trial as this has now been revised to ,350 cells/ml) are given daily
CTX and should not receive SP-IPTp. However, at the time of
implementation and thanks to the coordinated efforts for malaria
control, malaria transmission had become extremely low to the
extent that the study was prematurely stopped because it would
not have had sufficient power to meet the original objectives.
However, before interrupting the study, several pregnant women
were included and received either daily CTX or IPTp-SP in a 1:1
ratio. This provided the opportunity to report the safety of daily
CTX in an area of low malaria endemicity.
Methods
Study Area
Choma district is located in the Southern Province of Zambia.
Though malaria continued to be a key focus of public health
services in Choma as is the case in Zambia, there has been large
scale up of antimalarial interventions and considerable progress in
control of disease in recent years [22,34]. The malaria burden
decreased, in a population of 12 million, from 3.3 million reported
cases and 9,369 deaths in 2003 to 2.9 million cases and 3,862
deaths in 2009 [22].
The changing epidemiology was documented in Livingstone,
less that 200 km from Choma, where the number of malaria cases
declined from 8000 per quarter between 2004 and 2007[35] to
only 65 cases in the third quarter of 2008. Reported malaria
deaths declined from 60 in 2004 to zero in 2009 [35]. As Choma
and Livingstone are in the same epidemiological zone, these
reports retrospectively confirm that in 2009 the malaria risk in
Choma had decreased to an extremely low level and support the
decision of interrupting the clinical trial prematurely.
Decision to Stop the Trial in Choma
The study was initiated in Choma area in January, 2009, shortly
after a large observational study that was recruiting pregnant
women with history of clinical malaria and receiving antimalarial
treatment had just been concluded. The screening for the trial (the
Malcotrim study) included peripheral blood smear analysis. After 6
months of patient recruitment a review of the data on the
screening smear results revealed that only 4 of the 421 screened
patients had a malaria infection. This prompted the study director
in liaison with entire investigational team to inform the ethical
review committee at the Tropical Diseases Research Centre
(TDRC) and the National Ethical Review Committee, the sponsor
as well as the Zambia Medicines Regulatory Authority of the
extremely low malaria prevalence in the study area. As the risk of
malaria infection in pregnant women was much lower than
expected, the trial would not have had sufficient power to provide
the required answer, i.e. non-inferiority of the CTX arm in
preventing placental malaria as compared to SP intermittent
preventive treatment. The issue of changing the study site was
discussed also with the DSMB that received and reviewed on a
quarterly basis (unless differently requested by the DSMB
members) the updates on screening and recruitment and on losses
to follow-up, as well as the quarterly SAE reports routinely sent
also to the Ethical Committees. In addition, Serious Adverse
Events were also reported on a case-by-case basis, unless
differently requested by the DSMB members, in order to allow
them to promptly comment on specific events. The decision to
move the study site to another location known to have high
malaria transmission was taken by the research team in
consultation with the DSMB, ethics committees, the Regulatory
authority and the sponsor.
Safety of Daily Cotrimoxazole in Pregnancy: A Randomized Trial
PLOS ONE | www.plosone.org 2 May 2014 | Volume 9 | Issue 5 | e96017
Study Participants
Pregnant women between 16 and 28 weeks of gestation
attending the antenatal clinic of the Shampande Health Centre
or Choma Hospital in Choma District, accepting to be tested for
HIV and willing to participate in the study were enrolled. The
inclusion criteria were absence of symptoms consistent with
malaria at the time of recruitment (determined by asking them;
a positive blood smear was not an exclusion criterion although all
women found with a malaria infection at the time of recruitment
were also asked if they had any symptoms of malaria), willingness
to deliver at the health facility and willingness to adhere to study
requirements, e.g. monthly visits to the antenatal clinic. Exclusion
criteria were history of allergy to sulpha drugs, history or presence
of major illnesses likely to influence pregnancy outcome such as
diabetes, severe renal or heart disease or active tuberculosis; intent
to move outside the study catchment area before delivery, severe
anaemia (Hb, 7 g/dl) and previous history of unfavourable
pregnancy outcome (such as but not limited to pre-eclampsia,
caesarean section, stillbirth). Recruitment was conducted from
February to September, 2009.
Ethical Considerations
The study was sponsored by the Institute of Tropical Medicine,
Antwerp. It was approved by the Institutional Review Board at the
Institute of Tropical Medicine, Antwerp, and the Ethics commit-
tees at the University of Antwerp and at the Tropical Diseases
Research Centre, Ndola, Zambia. These bodies also approved the
Amendment 1.0 of 25 March 2009, which was issued to align the
study to the new management practices for prevention of mother
to child transmission (PMTCT) in Zambia (pregnant women to be
treated with anti-retroviral therapy (ART) if their CD4 count was
,350 cells/ml). All patients were informed about the study before
they were asked to sign the informed consent. Each study
participant was assigned a unique patient code that was used for
all subsequent forms in the study, so that confidentiality of all
subjects was guaranteed. A no fault study insurance was taken to
indemnify possible damage linked to a subjects participation in the
study. The trial was registered in the Clinicaltrials.gov registry,
identifier: NCT00711906, URL: http://clinicaltrial.gov/ct2/
show/NCT00711906.
Study Design
This was a phase 3 randomized open label clinical trial whose
planned primary endpoint was placental malaria. (The protocol
for this trial and supporting CONSORT checklist are available as
supporting information; see CONSORT Checklist S1 and
Protocol S1). The sample size calculation for the HIV negative
population was determined by assuming the prevalence of
placenta malaria at 10% in both groups with assumptions that
CTX prophylaxis was considered equivalent to SP if the two-sided
95% confidence interval was below 15% (90% power was required
for the primary hypothesis). Therefore, 788 pregnant women per
Figure 1. Study Design.
doi:10.1371/journal.pone.0096017.g001
Safety of Daily Cotrimoxazole in Pregnancy: A Randomized Trial
PLOS ONE | www.plosone.org 3 May 2014 | Volume 9 | Issue 5 | e96017
treatment arm were required with an assumed loss to follow up of
10%. For HIV positive women with CD4 count $200 cells/ml,
similar assumptions were considered though the two-sided 95%
confidence interval was expected to be below 20%. Therefore, 260
pregnant women per treatment arm were required with an
assumed loss to follow up of 10%. Therefore, total sample size
initially planned for the study was 2,096 pregnant women.
The study stratification by HIV status to correspond to the
study hypothesis and objectives (as the study aimed at describing
differences in pregnancy outcomes by HIV status) is shown in
figure 1.
All HIV negative and HIV positive with CD4 count $200
cells/ml were randomized to either three monthly doses of SP–IPT
(a product of Roche, Fansidar) or daily CTX prophylaxis (2 tablets
of 400 mg of sulphamethoxazole and 80 mg of trimethoprim; a
product of Roche, Bactrim). Therefore, the study intervention and
randomization did not vary by HIV status in these two groups of
women. Nevertheless, HIV positive pregnant women with CD4
count ,200 cells/ml and complying with all other entry criteria
were recruited in an observational cohort as they had to be given 2
tablets of CTX prophylaxis daily and antiretroviral treatment as
per standard guidelines. In Zambia CTX prophylaxis for these
women is part of the routine ANC care since November, 2003, in
accordance with the WHO policy [37]. Thus, those women
already on CTX prophylaxis and/or ARV treatment and
complying with all other entry criteria were also recruited in this
observational cohort.
At enrolment, a standardized questionnaire to collect demo-
graphic information, history of malaria episodes, past medical and
obstetric history was administered. Other risk factors (such as
smoking and drinking) that may affect pregnancy outcome were
directly recorded in the Case Record Form (CRF). The
socioeconomic status was also assessed on the basis of some key
household parameters and a physical examination was performed.
Laboratory tests at baseline included thick and thin blood smears
for the diagnosis of malaria infection and measurement of
haemoglobin. IPTp-SP administration was directly observed while
CTX intake was supervised only for the first dose, the rest being
taken at home until the next monthly visit when participants were
reviewed.
Randomization
The randomization was stratified by HIV status. Eligible
women were randomized to one of the two arms according to a
pre-defined randomization list prepared at ITM, Antwerp.
Participants were assigned sequential study numbers which were
matched with numbered envelopes containing the arm allocation
that were opened by the study nurses only after recruitment of the
study subject. There was no blinding as each of the study drugs
was openly administered.
Study Procedures
Women attended the antenatal clinic monthly for assessment of
efficacy and safety parameters. Safety parameters included adverse
events (AEs), serious adverse events (SAEs) and concomitant
medications. During the monthly visits, physical examination,
including foetal viability, was performed, information on bed net
use was recorded, the study drugs provided, and compliance
assessed by means of drug accountability. A blood sample for
haemoglobin measurement (only once early in the third trimester,
between 30–34 weeks), blood film for malaria parasites and a filter
paper for later molecular biological assessments were collected (for
efficacy assessment). Urine was analysed for glucose, proteins and
blood using a dipstick urine test. At delivery, similar information as
Figure 2. Trial Profile.
doi:10.1371/journal.pone.0096017.g002
Safety of Daily Cotrimoxazole in Pregnancy: A Randomized Trial
PLOS ONE | www.plosone.org 4 May 2014 | Volume 9 | Issue 5 | e96017
that collected at monthly visits was collected, including any AEs,
SAEs and use of concomitant medication. In addition, a placenta
blood sample for a thick blood film and later molecular analysis
(filter paper) was collected from the maternal surface of the
placenta. A biopsy was also obtained.
Women and their babies were seen at one and six weeks post-
delivery. Pregnancy specific assessments included rates of stillbirth
(.28 weeks gestation), neonatal mortality (#28 days of birth),
maternal mortality (up to 6 weeks post delivery), spontaneous
abortion (#28 weeks gestation), stillbirth, preterm delivery (#37
completed weeks), incidence of low birth weight, gestational age at
delivery (estimated from the last menstrual period [LMP], or by a
developmental score [36], if the LMP was unknown), incidence of
major and minor birth defects. Any infections that the patients
reported (or were treated for) were also recorded.
Data Management and Quality Control
Data were initially collected in a standardized source document
template, and then entered into an electronic case report form
(eCRF) using Macro 3.0 (InferMed Ltd, United Kingdom), a
CFR21Part11 compliant software with an in built audit trail,
password control and electronic signature. Each individual
involved in this process (the data manager, data entry clerk,
investigator and monitor) had a unique password-protected
database user profile.
The eCRF comprised a data entry interface which was an exact
but electronic copy of the source document (SD) and incorporated
pre-programmed checks and warnings for inconsistencies, omis-
sion and commission errors. Data were entered with single data
entry and then verified by the investigators who confirmed it with
an electronic signature. A study monitor performed several
external monitoring visits throughout the study, during which
partial source data verification (SDV) was done. In case of
inconsistencies, queries were raised in the eCRF. All findings were
discussed with the entire study team at the end of each visit, so that
corrective actions could be taken, and similar errors could be
prevented in the future. Similarly, the investigators based at
TDRC also performed SDV to resolve inconsistencies and answer
queries raised by monitors and TDRC based data managers along
with the study team on a monthly basis. In addition, a manual
review of the database was done on key variables such as safety-
and endpoint data. The system was set up on offline laptops and
had a facility for uploading and saving data at a central server at
the ITM. The data were extracted into SAS 9.2 (SAS Institute
Inc., Cary, NC, USA) for formatting and statistical analyses.
Statistical Analyses
Statistical analyses were based on the intention-to-treat princi-
ple (ITT). For continuous variables, tests of the normality of their
distributions were done. Descriptive statistics means, medians,
modes and standard deviations were determined as appropriate.
Table 1. Baseline, clinical and socio-economic characteristics of all patients.
Exposure groups
a
Cotrimoxazole (CTX Sulphadoxine-pyrimethamine (SP)
Baseline and clinical Characteristics N
b{
Mean Standard deviation N
b{
Mean Standard deviation
Age 181 24.8 5.6 165 25.1 5.6
Weight (Kg) 181 60.2 10.4 165 60.2 10.3
Height (M) 181 159.5 11.3 163 159.8 5.7
Gestational age (wks) 181 18.2. 3.0 163 19.3 4.0
Systolic pressure 181 108.7 11.1 165 106.9 10.4
Diastolic pressure 181 68.8 9.7 165 68.0 9.1
Haemoglobin (g/dl) 181 12.1 1.3 165 12.0 1.4
Gravidity 180 2.0 2.0 164 2.0 2.0
Socio-economic Characteristics N
b
% 95% CI N
b
% 95% CI
Married 154 81.5 75.2, 86.8 114 73.1 64.9, 79.4
Single 34 18.0 12.8, 24.2 41 26.3 19.4, 43.7
Able to read 176 93.1 88.5, 96.3 143 91.7 85.5, 95.0
Able to write 180 95.2 91.2, 97.8 149 95.5 90.2, 97.8
That own house of residence 73 38.6 31.7, 46.0 61 39.1 31.2, 47.0
That rent house of residence 115 60.8 53.5, 67.8 94 60.3 51.8, 67.6
Using electricity for power 70 37.0 30.1, 44.4 51 32.7 25.2, 40.4
Using wood and charcoal for power 115 60.8 53.5, 67.8 103 66.0 57.6, 73.0
Using communal taps and pumps for water 121 64.0 56.7, 70.9 104 66.7 58.3, 73.6
Using taps inside house for water 46 24.3 18.4, 31.1 44 28.2 21.2, 35.7
Using wells for water 19 10.1 6.2, 15.3 7 4.5 1.8, 9.0
a
Exposure groups represent the treatment given for prevention of malaria; Kg = Kilograms; M = meters; wks = weeks.
b
Enrolled pregnant women who gave informed consent,
{
Numbers varying due to missing values.
doi:10.1371/journal.pone.0096017.t001
Safety of Daily Cotrimoxazole in Pregnancy: A Randomized Trial
PLOS ONE | www.plosone.org 5 May 2014 | Volume 9 | Issue 5 | e96017
Table 2. Overall Pregnancy outcomes by exposure group.
Characteristic
Overall population HIV negative population
HIV positive population
(CD4$200cell/ ml)
HIV positive population (CD4,200cell/
ml)
CTX (N = 166) SP (N = 145) CTX (N = 129) SP (N = 121) CTX (N = 26) SP (N = 24) CTX (N = 11)
SP (Not Applicable-
(N/A))
Pregnancy outcomes n % n % n % n % n % n % N %
--
Full-term normal deliveries* 150 90.4 130 89.6 118 91.5 108 89.2 25 96.2 22 91.7 7 63.6 - -
Preterm deliveries 6 3.6 4 2.8 5 3.9 4 3.3 0 0 0 0 1 9.1 - -
Spontaneous abortions 1 0.6 0 0 1 0.8 0 0 0 0 0 0 0 0 - -
Stillbirths 5 3.0 3 2.1 2 1.6 1 0.8 1 3.8 2 8.3 2 18.2 - -
Caesarean sections 4 2.4 6 4.1 3 2.3 6 5.0 0 0 0 0 1 9.1 - -
Neonatal deaths 0 0 2 1.4 0 0 2 1.7 0 0 0 0 0 0 - -
CTX (N = 161
1
) SP (N = 142
**
) CTX (N = 127
**
) SP (N = 120
**
) CTX (N = 25**) SP (N = 22
**
) CTX (N = 9) SP (N/A)
Infant outcome n % n % n % n % n % n % n % - -
Low birth-weight 15 9.3 9 6.3 12 9.4 7 5.8 2 8.0 2 9.1 1 9.1 - -
Normal infant 146 90.7 133 93.7 115 90.6 113 94.2 23 92.0 20 90.9 8 90.9 - -
1
Live births including one set of twins;
**Live births only;
*Full-term normal deliveries = spontaneous vaginal deliveries at gestational age $37 completed weeks; N/A-Not applicable.
doi:10.1371/journal.pone.0096017.t002
Safety of Daily Cotrimoxazole in Pregnancy: A Randomized Trial
PLOS ONE | www.plosone.org 6 May 2014 | Volume 9 | Issue 5 | e96017
Frequency and percentage distributions were computed for
discrete variables. Additionally, asymptotic 95% confidence
intervals (95% CI) for means of continuous variables were
determined. Clopper-Pearson exact 95% confidence intervals
were determined for percentage distributions of count variables.
Missing data were assumed ignorable and thus all statistical
analyses included only complete cases. Maternal and foetal
outcomes such as still births and neonatal deaths; spontaneous
abortions (#28 weeks gestation); preterm deliveries (,37 com-
pleted weeks); neonatal mortality (within 28 days after birth) and
birth weight, including LBW (,2,500 grams), were outcomes of
interest.
For each of these variables, the treatment arms were analyzed
per stratum (i.e. HIV status).
Results
The Trial Profile
Four hundred and twenty one pregnant women were screened,
346 met the entry criteria and one additional patient was recruited
among those whose baseline visit was at delivery (Figure 2).
Gestational age #16 weeks or $28 weeks was the most frequent
reason for non-enrolment in the study. Two hundred and eighty
(280) HIV negative women and 52 HIV positive women with CD4
count $200/ml were randomized to either CTX (140 and 27
respectively) or SP (140 and 25 respectively). The 14 HIV positive
pregnant women with CD4 count ,200/ml continued on daily
CTX according to the National guidelines for prevention of
opportunistic infections in HIV infected pregnant women [37].
Due to loss to follow up and consent withdrawal (Figure 2), 166
pregnant women in the CTX arm and 145 in SP arm were
analyzed at delivery.
Demographic and Clinical Characteristics
Baseline characteristics including age, weight, height and
gestational age were similar between treatment arms (Table 1).
Most of women were married (although about a fifth of them
declared to be single) and literate (Table 1). There was no
difference in mean blood pressure and haemoglobin at baseline
between study arms (Table 1).
Pregnancy Outcomes
Most pregnancy outcomes were spontaneous vaginal deliveries
at gestational age of $37 weeks, with over 89% of the total
deliveries between the 2 arms (Table 2). There were 10 preterm
deliveries, 6 (3.6%) in the CTX and 4 (2.8%) in the SP arm, and
few adverse pregnancy outcomes and neonatal deaths, with no
difference between the study arms (table 2). Among the 10
caesarean sections, half of them were due to cephalo-pelvic
disproportion (CPD). The rest were due to transverse lie (1), foetal
distress (1), breech presentation (1) and pre-eclampsia (2).
No congenital malformation was detected. There were 24 (8%)
LBW babies, 15 (9.3%) in the CTX group and 9 (6.3%) in SP
group (table 2). The mean birth-weight did not differ between
study arms (3.1 kg; SD = 0.5) (Table 3). Pregnancy outcomes,
including mean birth weight, did not differ by HIV status nor in
HIV infected women by CD4 count (,200 cells/ml) (Tables 2 and
3). It is important to note that subgroup analyses were intended to
be purely descriptive and were not powered for between-groups
comparisons.
General Safety Outcomes
A total of 61 AEs were observed in the 346 enrolled patients and
there was no difference between study arms (Table 4). Most AEs
were mild, i.e. 19 of the 61 (31.2%, with 9 from CTX arm and 10
from SP arm), and moderate 24 (39.3%) (12 AEs on CTX arm and
12 from the SP arm). Almost all these were either definitely
unrelated or unlikely related to the study drug. Those reported to
be severe and life threatening were 11 (18.0%) and 7 (11.5%),
respectively and almost all were reported as SAEs (Table 4).
Thirty three AEs were defined as SAEs (Table 4) and included 9
preterm deliveries and 10 caesarean sections. Two preterm
deliveries in the CTX arm were classified as possibly related to
the study drug by the attending physicians who performed the
causality assessment, while 4 were unlikely to be related. Both
patients were HIV negative and the reason for considering them as
SAEs was related to the hospitalization.
Efficacy Outcomes
There were only 2 patients on the CTX arm and 1 on the SP
arm who had malaria positive peripheral smears at the end of
follow up of the 347 pregnant women recruited. During the
screening phase no patients were declared ineligible due to
confirmed clinical malaria (with positive peripheral smear).
Among the women screened, there were only 4 out of the 421
that were found to have malaria infection.
Among the placenta blood samples collected on filter paper and
analysed by PCR, only 1 sample out of 214 was positive for
malaria. Placenta histopathology was also performed and only one
sample out of 214 had malaria pigment. This positive sample by
histopathology was the same as the placenta blood positive by
PCR analysis.
Discussion
For all HIV-infected adults, including pregnant women, with
CD4 cell counts below a given threshold, WHO recommends the
use of CTX to prevent opportunistic infections [37]. However, the
Table 3. Birth-weight profiles across the exposure groups.
CTX SP Total
Birth weight in kilograms n** Mean (SD) n** Mean (SD) n** Mean (SD)
Overall cohort 161 3.1 (60.5) 142 3.1 (60.5) 303 3.1 (60.5)
HIV negative population 127 3.1 (60.5) 120 3.2 (60.5) 247 3.1 (60.5)
HIV positive CD4.200 cells/ml 25 3.1 (6 0.5) 22 3.2 (60.5) 47 3.1 (60.5)
HIV positive CD4,200 cells/ml 9 2.9 (6 0.5) - - 9 2.9 (60.5)
**Live births only;
SD = Standard deviation.
doi:10.1371/journal.pone.0096017.t003
Safety of Daily Cotrimoxazole in Pregnancy: A Randomized Trial
PLOS ONE | www.plosone.org 7 May 2014 | Volume 9 | Issue 5 | e96017
Table 4. Common adverse events by exposure group.
ADVERSE EVENTS AND SERIOUS ADVERSE EVENTS
Exposure group
CTX SP Total
1.0 Adverse events reporting by MedDRA Primary System Organ Class: preferred term
1.1 Infections and infestations
1
Malaria 213
Respiratory Tract infection 112
Urinary Tract Infection 202
Vaginal candidosis 202
PROM/Threatened abortion 112
1.2 Nervous system and other disorders
1
Headache 202
General body pains 202
Gastro-intestinal disorders 134
Hypertension 325
General (Weakness, Fever, hypotension) 224
2.0 Serious adverse events reported and their causal relationship to study drug
Serious adverse Event Causal relationship CTX SP Total
2.1. Preterm deliveries U 439
D 00
P 20
2.1. IUFD and stillbirth Unlikely 2 2 8
Definitely unlikely 1 1
possible 2 0
2.3. Caesarean section U 2110
D 25
P 00
2.4. Spontaneous abortion Unlikely 1 0 1
Definitely unlikely 0 0
possible 0 0
2.5. Neonatal death U 002
D 01
P 01
2.6. Pre-eclampsia Unlikely 1 0 1
Definitely unlikely 0 0
possible 0 0
2.7. Septicaemia with empetigo U 001
D 01
P 00
2.8. Tendinitis Unlikely 1 0 1
Definitely unlikely 0 0
possible 0 0
Subtotal SAEs 18 15 33
TOTAL (SAEs and AEs) 36 25 61
U = Unlikely related, D = Definitely unrelated, P = Possibly related; IUFD = Intrauterine Foetal death;
1
Mainly mild/moderate severity; PROM = Premature rupture of membranes.
doi:10.1371/journal.pone.0096017.t004
Safety of Daily Cotrimoxazole in Pregnancy: A Randomized Trial
PLOS ONE | www.plosone.org 8 May 2014 | Volume 9 | Issue 5 | e96017
recommendation also states that pregnant women should not
receive SP as IPTp together with CTX prophylaxis. There is
evidence that CTX is also an effective antimalarial [33][38],
though the effect of daily CTX prophylaxis during pregnancy on
malaria and birth outcomes has not been investigated in HIV-
infected or uninfected pregnant women [33][39]. This was the
reason for carrying out in Choma, Zambia, the clinical trial
described here.
The trial was aimed at determining the protective efficacy of
daily CTX against malaria, both in HIV infected and uninfected
pregnant women. Nevertheless, after starting the trial, it became
obvious that the risk of malaria infection in the study area had
decreased substantially, to the extent that that the trial could not
answer the primary question on the protective efficacy of CTX
prophylaxis. Therefore, the trial was prematurely terminated
though a substantial number of pregnant women had already been
recruited. This provided the opportunity of investigating the safety
of CTX prophylaxis in an area of extremely low malaria
transmission.
Short term treatments with CTX (10–14 days) have been
associated with birth defects when administered during the first
trimester [40]. In this study, CTX was administered in the second
and third trimester and pregnant women were prospectively
followed up. Antifolates such as trimethoprim have been known to
cause folate deficiency during pregnancy [41] and dietary folate
deficiency during the last two trimesters of pregnancy has been
linked to preterm delivery and maternal anaemia [42–44].
However, in our study daily CTX was not associated with a
higher occurrence of adverse pregnancy outcomes and was well
tolerated, irrespective of the HIV status. Besides pregnancy related
AEs, the other most commonly reported AEs and SAEs were
infections and infestations. There was no observable difference in
relation to the severity assessment between the study arms and the
causality link patterns of the AEs on the CTX arm were
comparable to those on the SP arm. Though no birth defect
was reported, the sample size was small and probably inadequate
to capture rare events, particularly those not immediately visible
such as congenital heart disease and other internal organ related
defects.
The rate of preterm deliveries using the LMP did not differ
between the CTX (combined group of HIV negative and positive
mothers) and SP arm. Similarly, the occurrence of preterm
deliveries, stillbirths, neonatal mortality, spontaneous abortions
and low birth weight among the HIV uninfected and infected with
CD4 count $200 cells/ml women did not differ between study
arms. For both study arms and irrespective of HIV status, the
majority of the infants had birth weights consistent with full term
delivery. These results are comparable to the findings from a
cohort study conducted in the same study area [45] which
observed still birth and neonatal death rates of about 2%, and a
higher preterm delivery rate (17%) for SP than that (2.1%)
observed in this study. However, our findings are based on the
sub-analysis of a sample size not sufficiently powered to interpret
between group comparisons but rather intended to be purely
descriptive of the outcomes. Nevertheless, even if the planned
number of pregnant women had been recruited, the study may not
have been able to detect rare adverse events. It is important to
note that as our findings here do not show an increase in safety
risks associated with CTX, we do not suggest that they are
conclusive of evidence of absence as explained by Altman et al
[46].
After stopping this trial, a similar trial (COTRIMAL
ClinTrialsGov number: NCT 01053325) was set up in an area
of high malaria transmission and data are currently being
analysed. Therefore, more information on the safety and efficacy
of CTX against malaria will be made available in due course.
Conclusions
Exposure to CTX during the latter part of pregnancy may not
be associated with increased safety risks when compared to SP.
However, considering the increasing resistance to SP, which is the
drug currently used for IPTp, and the need to prevent
opportunistic infections in HIV infected pregnant women by
administering daily CTX, there is need of exploring the possible
role CTX may have in preventing malaria in pregnant women,
both HIV infected and uninfected.
Supporting Information
Checklist S1 CONSORT Checklist.
(DOC)
Protocol S1 Trial Protocol.
(DOC)
Acknowledgments
We are grateful to the our patients for their participation in the study; the
study midwives Grace Chulu, Zodwa Hamweene, Alice Changuba, Lucia
Adams, Siphiwe Mwaba, Ida Nyanga; to the secretarial staff on the study,
Adah Biemba, Iness M. Simukonde and Esther Mashinde; laboratory staff,
Skitter Makwelele and Peter Mbewe. Thanks also to Joris Menten, Tom
Koyen, Danielle Van Melle, Celine Schurmans, Greta Gondol, Evi
Pockele and Harry Van Loen at the Institute of Tropical Medicine in
Antwerp.
Author Contributions
Conceived and designed the experiments: CM JPVG EMN UDA.
Performed the experiments: CM EMN GC JPVG UDA DM DC RM
MDC YC CVO RMR. Analyzed the data: CM DM JPVG UDA.
Contributed reagents/materials/analysis tools: DM CVO YC DC MM
JPVG UDA. Wrote the paper: CM JPVG UDA. Critical review of
manuscript: CM EMN GC RM MDC YC RMR JPVG UDA.
References
1. WHO (2012) World Malaria Report.
2. WHO (2006) Roll Back Malaria Partnership.
3. Shulman CE, Dorman EK (2003) Reducing childhood mortality in poor
countries - importance and prevention of malaria in pregnancy. Trans R Soc
Trop Med Hyg 97: 30–35.
4. Whitty CJ, Edmonds S, Mutabingwa TK (2005) Malaria in pregnancy. BJOG,
112: 1189–1195.
5. WHO: Assessment of the safety of artemisinin compounds in pregnancy [http: //
www.who.int/entity/malaria/publications/atoz/9789241596114/en/index.
html].Accessed 2014 April 15.
6. Nosten F, McGready R, Simpson JA, Thwai KL, Balkan S, et al (1999) Effects of
Plasmodium vivax malaria in pregnancy. Lancet 354: 546–549.
7. Steketee R, Wirima JJ HA, Slustker L, Heymann DL, Brenman JG (1996) The
effect of malaria and malaria prevention in pregnancy on offspring birthweight,
prematurity and intrauteri ne growth retardation in rural malawi. Am J Trop
Med Hyg 55: 33–41.
8. van Geertruyden JP, Thomas F, Erhart A, D’Alessandro U (2004) The
contribution of malaria in pregnancy to perinatal mortality. Am J Trop Med
Hyg 71: 35–40. 71/2_suppl/35 [pii].
9. Desai M, ter Kuile FO, Nosten F, McGready R, Asamoa K, et al (2007)
Epidemiology and burden of malaria in pregnancy. Lancet Infect Dis 7: 93–104.
S1473–3099(07)70021-X [pii]; 10.1016/S1473–3099(07)70021-X [doi].
10. Steketee R, Wirima JJ, Bloland PB, Chilima B, Mermin JH, et al (1996)
Impairment of a pregnant woman’s acquired ability to limit Plasmodium
falciparum by infection with human immunodeficiency virus type-1. Am J Trop
Med Hyg 55: 42–49.
Safety of Daily Cotrimoxazole in Pregnancy: A Randomized Trial
PLOS ONE | www.plosone.org 9 May 2014 | Volume 9 | Issue 5 | e96017
11. Allen S, Van de perre P, Serufilira A, Lepage P, Carael M, et al (1991) Human
Immunodeficiency virus and malaria in a representative sample of childbearing
women in Kigali, Rwanda. J infect Dis 164: 67–71.
12. ter Kuile FO, Parise ME, Verhoeff FH, Udhayakumar V, Newman RD, et al
(2004) The burden of co-infection with human immunodeficiency virus type 1
and malaria in pregnant women in sub-saharan Africa. Am J Trop Med Hyg 71:
41–54. 71/2_suppl/41 [pii].
13. Verhoeff FH, Brabin BJ, Hart CA, Chimsuku L, Kazembe P, et al (1999)
Increased prevalence of malaria in HIV-infected pregnant women and its
implications for malaria control. Trop Med Int Health 4: 5–12.
14. van Eijk AM, Ayisi JG, ter Kuile FO, Misore AO, Otieno JA, et al (2002) Risk
factors for malaria in pregnancy in an urban and peri-urban population in
western Kenya. Trans R Soc Trop Med Hyg 96: 586–592.
15. Ayisi JG, van Eijk AM, ter Kuile FO, Kolczak MS, Otieno JA, et al (2003) The
effect of dual infection with HIV and malaria on pregnancy outcome in western
Kenya. AIDS 17: 585–594. 10.1097/01.aids.0000042977.95433.37 [doi].
16. Ticconi C, Mapfumo M, Dorrucci M, Naha N, Tarira E, et al (2003) Effect of
maternal HIV and malaria infection on pregnancy and perinatal outcome in
Zimbabwe. J Acquir Immune Defic Syndr 34: 289–294.
17. Bloland PB, Wirima JJ, Steketee RW, Chilima B, Hightower A, et al (1995)
Maternal HIV infection and infant mort ality in Malawi: Evidence for increased
mortality due to placental malaria infection. AIDS 9: 721–726.
18. Anglaret X, Chene G, Attia A, Toure S, Lafont S, et al (1999) Early
chemoprophylaxis with trimethoprim-sulphamethoxazole for HIV-1-infected
adults in Abidjan, Cote d’Ivoire: a randomised trial. Cotrimo-CI Study Group.
Lancet 353: 1463–1468. S0140673698073991 [pii].
19. Brentlinger PE, Behrens CB, Micek MA (2006) Challenges in the concurrent
management of malarria and HIV in pregnancy in subsaharan Africa. Lancet
Infect Dis 6: 100–111.
20. WHO (2010) World Malaria Report. [www.who.int/entity/malaria/
publications/atoz/9789241564106/en/]. Accessed 2014 April 15.
21. WHO (2007) Assessment of the safety of artemisinin compounds in pregnancy.
[http:www.who.int/malaria/publications/atoz/9789241596114/en/index.
html]. Accessed 2014 April 15.
22. WHO (2010) World Malaria report: Summary. [http://www.who.int/malaria/
world_malaria_report_2010/malaria2010_summary_keypoints_en.pdf]. Ac-
cessed 2014 April 15.
23. Garner P GA (2003) Drugs for preventing malaria-related illness in pregnant
women and death in the newborn. 1:CD000169.
24. Cot M, Deloron P (2003) Malaria prevention strategies. Br Med Bull 67: 137–
148.
25. Peters PJ, Thigpen MC, Parise ME, Newman RD (2007) Safety and toxicity of
sulfadoxine/pyrimethamine: implications for malaria prevention in pregnancy
using intermittent preventive treatment. Drug Saf 30: 481–501. 3063 [pii].
26. WHO (2012) Intermittent Preventive Treatment of malaria in pregnancy using
Sulfadoxine-Pyrimethamine (IPTp-SP). [http://www.who.int/malaria/
publications/atoz/who_iptp_sp_policy_recommendation/en/]. Accessed 2014
April 15.
27. Parise ME, Ayisi JG, Nahlen BL, Schultz LJ, Roberts JM, et al (1998) Efficacy of
sulfadoxine-pyrimethamine for prevention of placental malaria in an area of
Kenya with a high prevalence of malaria and human immunodeficiency virus
infection. Am J Trop Med Hyg 59: 813–822.
28. Verhoeff FH, Brabin BJ, Chimsuku L, Kazembe P, Russell WB, et al (1998) An
evaluation of the effects of intermittent sulfadoxine-pyrimethamine treatment in
pregnancy on parasite clearance and risk of low birthweight in rural Malawi.
Ann Trop Med Parasitol 92: 141–150.
29. Omar SA, Bakari A, Owiti A, Adag u IS, Warhurst DC (2001) Co-trimoxazole
compared with sulfadoxine-pyrimethamine in the treatment of uncomplicated
malaria in Kenyan children. Trans R Soc Trop Med Hyg 95: 657–660.
30. Mermin J, Lule J, Ekwaru JP, Malamba S, Downing R, et al (2004) Effect of co-
trimoxazole prophylaxis on morbidity, mortality, CD4-cell count, and viral load
in HIV infection in rural Uganda. Lancet 364: 1428–1434.
S0140673604172255 [pii]; 10.1016/S0140–6736(04)17225–5 [doi].
31. Hauth JC, Goldenberg RL, Andrew WN, Dubard MB, Copper RL (1995)
Reduced incidence of preterm delivery with metronidazole and erytromycin in
women with bacterial vaginosis. N Engl J Med 333: 1732–1736.
32. Walter J, Mwiya M, Scott N, Kasonde P, Sinkala M, et al (2006) Reduction in
preterm delivery and neonatal mortality after the introduction of antenatal
cotrimoxazole prophylaxis among HIV-infected women with low CD4 cell
counts. J Infect Dis 194: 1510–1518. JID36726 [pii]; 10.1086/508996 [doi].
33. Manyando C, Njunju EM, D’Alessandro U, van Geertruyden JP (2013) Safety
and Efficacy of Co-Trimoxazole for Treatment and Prevention of Plasmodium
falciparum Malaria: A systematic Review. PLos one 8: e56916.
34. Chizema-Kawesha E, Miller JM, Steketee RW, Mukonka V, Mukuka C, et al
(2010) Scaling up malaria control in Zambia: progress and impact 2005–2008.
Am J Trop Med Hyg 83: 480–488.
35. Masaninga F, Sekeseke-Chinyama M, Malambo T, Moonga H, Babaniyi O, et
al (2012) Finding parasites and finding challenges: improved diagnostic access
and trends in reported malaria and anti-malarial drug use in Livingstone district,
Zambia. Malar J 11.
36. Dubowitz LMS, Dubowitz V, Goldberg C (1970) Clinical assessment of
gestational age in the new born infant. J Pediatr 77: 1–10.
37. WHO (2006) Guidelines on Cotrimoxazole prophylaxis for HIV-related
infections among children, adolescents and adults: Recommendations for a
public health approach. [http://www.who.int/hiv/pub/guidelines/ctx/en/].
Accessed 2014 Apr 15.
38. Thera MA, Sehdev PS, Coulibaly D, Traore K, Garba MN, et al (2005) Impact
of trimethoprim-sulfameth oxazole prophylaxis on falciparum malaria infection
and disease. J Infect Dis 192: 1823–1829. JID35552 [pii]; 10.1086/498249
[doi].
39. Meshnick SR, Mwapasa V, Rogerson S (2006) Protecting pregnant women from
malaria in areas of high HIv infection prevalence. J infect Dis 194: 273–275.
40. Czeizel AE, Rockenbauer M, Sorensen HT, Olsen J (2001) The teratogenic risk
of trimethoprim-sulfonamides: a population based case-control study. Reprod
Toxicol 15: 637–646. S0890623801001782 [pii].
41. Hernandez-Diaz S, Werler MM, Walker AM, Mitchell AA (2000) Folic acid
antagonists during pregnancy and the risk of birth defects. N Engl J Med 343:
1608–1614. 10.1056/NEJM200011 303432204 [doi].
42. Siega-Riz AM, Savitz DA, Zeisel SH, Thorp JM, Herring A (2004) Second
trimester folate status and preterm birth. Am J Obstet Gynecol 191: 1851–1857.
S0002937804008609 [pii]; 10.1016/j.ajog.2004.07.076 [doi].
43. Marti-Carvajal V, Pena-Marti, Comunian-Carraso G (2004) Prematurity and
maternal folate deficiency: anaemia during pregnancy study group results in
Valencia, Venezuela. Arch LatinoamNutri 54: 45–49.
44. Juarez-Vazquez J, Bonizzoni E SA (2002) Iron plus folate is more effective than
iron alone in the treatment of iron deficiency anaemia in pregnanct: a
randomised, bouble blind clinical trial. BJOG 109: 1009–1014.
45. Manyando C, Mkandawire R, Pum a L, Sinkala M, Mpabalwani E, et al. (2010)
Safety of artemether-lumefantrine in pregnant women with malaria: results of a
prospective cohort study in Zambia. Malaria J, 9: 249.
46. Altman DG, Bland JM (1995) Absence of evidence is not evidence of absence
BMJ, 311 (7003): 485.
Safety of Daily Cotrimoxazole in Pregnancy: A Randomized Trial
PLOS ONE | www.plosone.org 10 May 2014 | Volume 9 | Issue 5 | e96017
... 14 In a randomised study in Zambia on antenatal pregnant women who received either daily co-trimoxazole (non-IPTp-SP users) or routine sulphadoxine-pyrimethamine (IPTp-SP users), it was revealed that preterm deliveries (non-IPTp-SP users 3.6%; IPTp-SP 3.0%); still births (non-IPTp-SP 3.0%; IPTp-SP users 2.1%), neonatal deaths (non-IPTp-SP 0%; SP 1.4%), and spontaneous abortions (non-IPTp-SP users 0.6%; IPTp-SP users 0%) were similar between study arms. 15 Also, the low birth weight rates were 9% for non-IPTp-SP users and 13% for IPTp-SP users. 15 However, evaluation of the efficacy of IPT has not been updated in the study environment despite efforts being made on malaria prophylaxis. ...
... 15 Also, the low birth weight rates were 9% for non-IPTp-SP users and 13% for IPTp-SP users. 15 However, evaluation of the efficacy of IPT has not been updated in the study environment despite efforts being made on malaria prophylaxis. In addition, no previous study in Nigeria has evaluated the effects of IPT on rates of preterm delivery, spontaneous abortion, intra-uterine foetal death and low birth weight among IPTp-SP users when compared with nonusers of SP. ...
Article
Full-text available
Background: Following the World Health Organization (WHO) recommendations for 4-weekly antenatal intermittent preventive treatment of malaria in pregnancy using sulphadoxine-pyrimethamine (IPTp-SP), there is a need to evaluate the drug performance in order to determine their effectiveness as tools in malaria control policy. Objectives: To determine prevalence of cord blood malaria, compliance gap and adverse pregnancy outcomes (anaemia, preterm delivery, spontaneous abortion, intra-uterine foetal death and low birth weight) among antenatal IPTp-SP users compared with non-users. Methods: A cross-sectional analytical study was conducted among consenting 390 participants who were administered a questionnaire, and paired blood samples were collected from the venous blood of participants and neonatal cord immediately after delivery. The participants were categorised as IPTp-SP users and non-users. Adverse pregnancy outcomes were assessed. Neonatal birth weights were also measured within 1 h after delivery. Malaria parasitaemia and anaemia were analysed using standard parasitological and haematological methods of examination. Data were analysed using SPSS version 25 for Windows and p-value of < 0.05 considered significant. Results: Of 390 women, 336 (86.2%) were IPTp-SP users, while 54 (13.8%) were non-users. The compliance gap was 13.8%. Malaria parasitemia in pregnant women (21.7% versus 53.7%; p < 0.001) and their babies (12.2% versus 25.4%; p = 0.002) were observed for IPTp-SP users and non-users, respectively. The prevalence of maternal anaemia was 27(8.0%) in IPTp-SP users and 5 (9.3%) in non-users (p = 0.789). Mean parasite density was reduced in IPTp-SP users than in non-users (p < 0.001). Correlation of birth weight according to their sex showed a weak correlation [correlation coefficient (r) = 0.027; p = 0.736]. Pregnant women with preterm delivery, spontaneous abortion, intra-uterine foetal death, and low birth weight were significantly lower (p < 0.001, for all) in IPTp-SP users compared with non-users. Conclusion: Although the compliance gap was low, IPTp-SP users had significantly better pregnancy and foetal outcomes compared with non-users. Efforts should be intensified towards achieving total compliance in IPTp-SP usage by pregnant women.
... Materials 2021, 14, 6980 2 of 17 and pregnant women [8][9][10][11]. Tigecycline must be administered intravenously; therefore, its use is reserved for patients with severe and complicated malaria, with delayed action similar to doxycycline [7]. ...
... Anti-malarial activity has been described for other antibiotics such as co-trimoxazole, quinolones, tigecycline, mirincamycin, ketolides, fusidic acid, and thiopeptides [7]. Based on studies in Africa, co-trimoxazole can be used as an alternative in malaria prophylaxis, for children and adults, HIV (human immunodeficiency viruses) positive or negative patients on studies in Africa, co-trimoxazole can be used as an alternative in malaria prophylaxis, for children and adults, HIV (human immunodeficiency viruses) positive or negative patients and pregnant women [8][9][10][11]. Tigecycline must be administered intravenously; therefore, its use is reserved for patients with severe and complicated malaria, with delayed action similar to doxycycline [7]. ...
Article
Full-text available
Erythromycin A is an established anti-bacterial agent against Gram-positive bacteria, but it is unstable to acid. This led to an evaluation of erythromycin B and its derivatives because these have improved acid stability. These compounds were investigated for their anti-malarial activities, by their in silico molecular docking into segments of the exit tunnel of the apicoplast ribosome from Plasmodium falciparum. This is believed to be the target of the erythromycin A derivative, azithromycin, which has mild anti-malarial activity. The erythromycin B derivatives were evaluated on the multi-drug (chloroquine, pyrimethamine, and sulfadoxine)-resistant strain K1 of P. falciparum for asexual growth inhibition on asynchronous culture. The erythromycin B derivatives were identified as active in vitro inhibitors of asexual growth of P. falciparum with low micro-molar IC50 values after a 72 h cycle. 5-Desosaminyl erythronolide B ethyl succinate showed low IC50 of 68.6 µM, d-erythromycin B 86.8 µM, and erythromycin B 9-oxime 146.0 µM on the multi-drug-resistant K1 of P. falciparum. Based on the molecular docking, it seems that a small number of favourable interactions or the presence of unfavourable interactions of investigated derivatives of erythromycin B with in silico constructed segment from the exit tunnel from the apicoplast of P. falciparum is the reason for their weak in vitro anti-malarial activities.
... Table 1 shows at inclusion well-balanced characteristics of the 188 randomised women in the two arms. The median age of women was 28 (24)(25)(26)(27)(28)(29)(30)(31)(32)(33) years. In the CTX arm, women were included at 21 (18)(19)(20)(21)(22)(23)(24) weeks of gestation, and 8% (7/90) were primigravidae. ...
... This finding corroborates those of other studies on the safety of CTX during the second and third trimesters of pregnancy in HIV-infected women [28,29]. In a phase 3b randomised controlled trial, Manyando et al. found that exposure to daily CTX in pregnancy may not be associated with particular safety risks in terms of birth outcomes [30]. ...
Article
Full-text available
Objective The main objective of the MACOMBA (Maternity and Control of Malaria‐HIV co‐infection in Bangui) trial was to show that cotrimoxazole (CTX) is more effective than sulphadoxine–pyremethamine–IPTp (IPTp‐SP) to prevent placental malaria infection (primary end point) among HIV‐positive pregnant women with a CD4+ count ≥350 cells/mm³ in Bangui, CAR. Methods MACOMBA is a multicentre, open‐label randomised trial conducted in four maternity hospitals in Bangui. Between 2013 and 2017, 193 women were randomised and 112 (59 and 53 in CTX and IPTp‐SP arms, respectively) were assessed for placental infection defined by microscopic parasitaemia or PCR. Results Thirteen women had a placental infection: five in the CTX arm (one by microscopic placental parasitaemia and four by PCR) and eight by PCR in the SP‐IPTp (8.5% vs. 15.1%, p = 0.28). The percentage of newborns with low birthweight (<2500 g) did not differ statistically between the two arms. Self‐reported compliance to CTX prophylaxis was good. There was a low overall rate of adverse events in both arms. Conclusion Although our results do not allow us to conclude that CTX is more effective, drug safety and good compliance among women with this treatment favour its widespread use among HIV‐infected pregnant women, as currently recommended by WHO.
... Our study is in accord with data from several others regarding the benefits of antimalarial prophylaxis to reduce adverse birth outcomes [15][16][17][18][19][20]. significantly reduced the risk of preterm births compared to the previously recommended 2-dose regimen [15]. ...
Article
Full-text available
Objectives Until late 2015, Botswana recommended preventive treatment for pregnant women in malarial regions with chloroquine and proguanil (CP). The guideline change provided an opportunity to evaluate CP and adverse birth outcomes. Methods The Tsepamo Study performed birth outcomes surveillance at large delivery centres throughout Botswana. We evaluated adverse birth outcomes from 2015 to 2017 at three hospitals where 93% of CP use was recorded. Outcomes included neonatal death (NND), small for gestational age (SGA), very SGA, stillbirth (SB), preterm delivery (PTD) and very PTD. Logistic regression analysis (unadjusted and adjusted) was conducted for each adverse birth outcome. Results During the study period, 5883 (26%) of 23,033 deliveries were exposed to CP, with the majority (65%) in the most malaria‐endemic region. At this site, there was a trend or an association between CP use and reduction of three adverse birth outcomes: PTD (aOR 0.85, 95% CI 0.76–0.96), vPTD (aOR 0.83, 95% CI 0.68–1.01) and NND (aOR 0.65, 95% CI 0.42–1.00). However, at the least malaria‐endemic site, the association was in the opposite direction for SB (aOR 1.54, 95% CI 1.08–2.22), SGA (aOR 1.24, 95% CI 1.06–1.44) and vSGA (aOR 1.42, 95% CI 1.14–1.77). The association between CP and reduced PTD was present among women without HIV (aOR 0.77, 95% CI 0.67–0.89) but not among women with HIV (aOR 1.09, 95% CI 0.78–1.35). Conclusions Antimalarial prophylaxis was associated with improved birth outcomes in the most malaria‐endemic region of Botswana, but not elsewhere. This finding supports current WHO guidance to use prophylaxis strategies among pregnant women in highly malaria‐endemic regions. Further studies of the risks and benefits of specific antimalarial regimens in pregnancy are warranted, particularly in areas with lower incidence of malaria.
... CTXp was found to be safe and to have a similar efficacy as SP for preventing the prevalence of parasitemia, placental malaria and adverse neonatal outcomes such as prematurity, stillbirths and LBW [98]. Additionally, another study performed in Zambia among HIV-infected and HIV-uninfected women showed that exposure to daily CTXp is safe for mothers' and newborns' health [99]. ...
Article
Full-text available
Malaria infection during pregnancy is an important driver of maternal and neonatal health in endemic countries. Intermittent preventive treatment in pregnancy (IPTp) with sulfadoxine-pyrimethamine (SP) is recommended for malaria prevention at each scheduled antenatal care visit, starting at the second trimester, in areas of high and moderate transmission. However, the increased resistance to SP in some endemic areas challenges its effectiveness. Furthermore, SP is contraindicated in the first trimester of pregnancy and in HIV-infected women on co-trimoxazole prophylaxis due to potential drug–drug interactions. Thus, in recent last decades, several studies evaluated alternative drugs that could be used for IPTp. A comprehensive literature review was conducted to summarize the evidence on the efficacy and safety of antimalarial drugs being evaluated for IPTp. Chloroquine, amodiaquine, mefloquine and azithromycin as IPTp have proven to be worse tolerated than SP. Mefloquine was found to increase the risk of mother-to-child transmission of HIV. Dihydroartemisin-piperaquine currently constitutes the most promising IPTp drug alternative; it reduced the prevalence of malaria infection, and placental and clinical malaria in studies among HIV-uninfected women, and it is currently being tested in HIV-infected women. Research on effective antimalarial drugs that can be safely administered for prevention to pregnant women should be prioritized. Malaria prevention in the first trimester of gestation and tailored interventions for HIV-infected women remain key research gaps to be addressed.
... Maternal Outcomes. Nine articles containing aggregate data reported on maternal AEs from prenatal TMP-SMX exposure; no serious maternal AEs were reported [121,156,160,163,165,166,168,173,175]. Among 23 exposures during pregnancy from case reports, the only reported maternal AE was 1 case of esophageal candidiasis in a pregnant woman with human immunodeficiency virus (HIV) infection [47,152,[176][177][178][179][180][181][182][183][184][185][186]. ...
Article
Background: The safety profile of antimicrobials used during pregnancy is one important consideration in the decision on how to treat and provide postexposure prophylaxis (PEP) for plague during pregnancy. Methods: We searched 5 scientific literature databases for primary sources on the safety of 9 antimicrobials considered for plague during pregnancy (amikacin, gentamicin, plazomicin, streptomycin, tobramycin, chloramphenicol, doxycycline, sulfadiazine, and trimethoprim-sulfamethoxazole [TMP-SMX]) and abstracted data on maternal, pregnancy, and fetal/neonatal outcomes. Results: Of 13 052 articles identified, 66 studies (case-control, case series, cohort, and randomized studies) and 96 case reports were included, totaling 27 751 prenatal exposures to amikacin (n = 9), gentamicin (n = 345), plazomicin (n = 0), streptomycin (n = 285), tobramycin (n = 43), chloramphenicol (n = 246), doxycycline (n = 2351), sulfadiazine (n = 870), and TMP-SMX (n = 23 602). Hearing or vestibular deficits were reported in 18/121 (15%) children and 17/109 (16%) pregnant women following prenatal streptomycin exposure. First trimester chloramphenicol exposure was associated with an elevated risk of an undescended testis (odds ratio [OR] 5.9, 95% confidence interval [CI] 1.2-28.7). Doxycycline was associated with cardiovascular malformations (OR 2.4, 95% CI 1.2-4.7) in 1 study and spontaneous abortion (OR 2.8, 95% CI 1.9-4.1) in a separate study. First trimester exposure to TMP-SMX was associated with increased risk of neural tube defects (pooled OR 2.5, 95% CI 1.4-4.3), spontaneous abortion (OR 3.5, 95% CI 2.3-5.6), preterm birth (OR 1.5, 95% CI 1.1-2.1), and small for gestational age (OR 1.6, 95% CI 1.2-2.2). No other statistically significant associations were reported. Conclusions: For most antimicrobials reviewed, adverse maternal/fetal/neonatal outcomes were not observed consistently. Prenatal exposure to streptomycin and TMP-SMX was associated with select birth defects in some studies. Based on limited data, chloramphenicol and doxycycline may be associated with adverse pregnancy or neonatal outcomes; however, more data are needed to confirm these associations. Antimicrobials should be used for treatment and PEP of plague during pregnancy; the choice of antimicrobials may be influenced by these data as well as information about the risks of plague during pregnancy.
Preprint
Full-text available
Background Drug safety assessments in clinical trials present unique analytical challenges. Some of these include adjusting for individual follow-up time, repeated measurements of multiple outcomes and missing data among others. Furthermore, pre-specifying appropriate analysis becomes difficult as some safety endpoints are unexpected. Although existing guidelines such as CONSORT encourage thorough reporting of adverse events (AEs) in clinical trials, they provide limited details for safety data analysis. The limited guidelines may influence suboptimal analysis by failing to account for some analysis challenges above. A typical example where such challenges exist are trials of anti-malarial drugs for malaria prevention during pregnancy. Lack of proper standardized evaluation of the safety of antimalarial drugs has limited the ability to draw conclusions about safety. Therefore, a systematic review was conducted to establish the current practice in statistical analysis for preventive antimalarial drug safety in pregnancy. Methods The search included five databased (PubMed, Embase, Scopus, Malaria in Pregnancy Library and Cochrane Central Register of Controlled Trials) to identify original English articles reporting Phase III randomized controlled trials (RCTs) on anti-malarial drugs for malaria prevention in pregnancy published from January 2010 to July 2019. Results Eighteen trials were included in this review that collected multiple longitudinal safety outcomes including AEs. Statistical analysis and reporting of the safety outcomes in all the trials used descriptive statistics; proportions/counts (n=18, 100%) and mean/median (n=2, 11.1%). Results presentation included tabular (n=16, 88.9%) and text description (n=2, 11.1%). Univariate inferential methods were reported in most trials (n=16, 88.9%); including Chi-square/Fisher`s exact test (n=12, 66.7%), t-test (n=2, 11.1%) and Mann-Whitney/Wilcoxon test (n=1, 5.6%). Multivariable methods, including Poisson and negative binomial were reported in few trials (n=4, 22.2%). Assessment of a potential link between missing efficacy data and safety outcomes was not reported in any of the trials that reported efficacy missing data (n=7, 38.9%). Conclusion The review demonstrated that statistical analysis of safety data in anti-malarial drugs for malarial chemoprevention in pregnancy RCTs is inadequate. The analyses insufficiently account for multiple safety outcomes potential dependence, follow-up time and informative missing data which can compromise anti-malarial drug safety evidence development, based on the available data.
Preprint
Full-text available
Background Drug safety assessments in clinical trials present unique analytical challenges. Some of these include adjusting for individual follow-up time, repeated measurements of multiple outcomes and missing data among others. Furthermore, pre-specifying appropriate analysis becomes difficult as some safety endpoints are unexpected. Although existing guidelines such as CONSORT encourage thorough reporting of adverse events (AEs) in clinical trials, they provide limited details for safety data analysis. The limited guidelines may influence suboptimal analysis by failing to account for some analysis challenges above. A typical example where such challenges exist are trials of antimalarial drugs for malaria prevention during pregnancy. Lack of proper standardized evaluation of the safety of antimalarial drugs has limited the ability to draw conclusions about safety. We have therefore conducted a systematic review to establish the current practice in statistical analysis for antimalarial drug safety in pregnancy. Methods We searched PubMed, Embase, Scopus, Malaria in Pregnancy Library and Cochrane Central Register of Controlled Trials for original English articles reporting Phase III (randomized controlled trials) RCTs on antimalarial drugs for malaria prevention in pregnancy published from January 2010 to July 2019. Results Eighteen trials were included in this review that collected multiple longitudinal safety outcomes including AEs. Statistical analysis and reporting of the safety outcomes in all the trials used descriptive statistics; proportions/counts (n=18, 100%) and mean/median (n=2, 11.1%). Results presentation included tabular (n=16, 88.9%) and text description (n=2, 11.1%). Univariate inferential methods were reported in most trials (n=16, 88.9%); including Chi-square/Fisher`s exact test (n=12, 66.7%), t-test (n=2, 11.1%) and Mann-Whitney/Wilcoxon test (n=1, 5.6%). Multivariable methods, including Poisson and negative binomial were reported in few trials (n=4, 22.2%). Assessment of a potential link between missing efficacy data and safety outcomes was not reported in any of the trials that reported efficacy missing data (n=7, 38.9%). Conclusion The review demonstrated that statistical analysis of safety data in antimalarial drugs for malarial chemoprevention in pregnancy RCTs are inadequate. The analysis insufficiently account for multiple safety outcomes potential dependence, follow-up time and informative missing data which can compromise antimalarial drug safety evidence development, based on the available data.
Preprint
Full-text available
Background Drug safety assessments in clinical trials present unique analytical challenges. Some of these include adjusting for individual follow-up time, repeated measurements of multiple outcomes and missing data among others. Furthermore, pre-specifying appropriate analysis becomes difficult as some safety endpoints are unexpected. Although existing guidelines such as CONSORT encourage thorough reporting of adverse events (AEs) in clinical trials, they provide limited details for safety data analysis. The limited guidelines may influence suboptimal analysis by failing to account for some analysis challenges above. A typical example where such challenges exist are trials of antimalarial drugs for malaria prevention during pregnancy. Lack of proper standardized evaluation of the safety of antimalarial drugs has limited the ability to draw conclusions about safety. We have therefore conducted a systematic review to establish the current practice in statistical analysis for antimalarial drug safety in pregnancy. Methods We searched PubMed, Embase, Scopus, Malaria in Pregnancy Library and Cochrane Central Register of Controlled Trials for original English articles reporting Phase III (randomized controlled trials) RCTs on antimalarial drugs for malaria prevention in pregnancy published from January 2010 to July 2019. Results Eighteen trials were included in this review that collected multiple longitudinal safety outcomes including AEs. Statistical analysis and reporting of the safety outcomes in all the trials used descriptive statistics; proportions/counts (n=18, 100%) and mean/median (n=2, 11.1%). Results presentation included tabular (n=16, 88.9%) and text description (n=2, 11.1%). Univariate inferential methods were reported in most trials (n=16, 88.9%); including Chi-square/Fisher`s exact test (n=12, 66.7%), t-test (n=2, 11.1%) and Mann-Whitney/Wilcoxon test (n=1, 5.6%). Multivariable methods, including Poisson and negative binomial were reported in few trials (n=4, 22.2%). Assessment of a potential link between missing efficacy data and safety outcomes was not reported in any of the trials that reported efficacy missing data (n=7, 38.9%). Conclusion The review demonstrated that statistical analysis of safety data in antimalarial drugs for malarial chemoprevention in pregnancy RCTs are inadequate. The analysis insufficiently account for multiple safety outcomes potential dependence, follow-up time and informative missing data which can compromise antimalarial drug safety evidence development, based on the available data.
Article
Full-text available
Background: Drug safety assessments in clinical trials present unique analytical challenges. Some of these include adjusting for individual follow-up time, repeated measurements of multiple outcomes and missing data among others. Furthermore, pre-specifying appropriate analysis becomes difficult as some safety endpoints are unexpected. Although existing guidelines such as CONSORT encourage thorough reporting of adverse events (AEs) in clinical trials, they provide limited details for safety data analysis. The limited guidelines may influence suboptimal analysis by failing to account for some analysis challenges above. A typical example where such challenges exist are trials of anti-malarial drugs for malaria prevention during pregnancy. Lack of proper standardized evaluation of the safety of antimalarial drugs has limited the ability to draw conclusions about safety. Therefore, a systematic review was conducted to establish the current practice in statistical analysis for preventive antimalarial drug safety in pregnancy. Methods: The search included five databases (PubMed, Embase, Scopus, Malaria in Pregnancy Library and Cochrane Central Register of Controlled Trials) to identify original English articles reporting Phase III randomized controlled trials (RCTs) on anti-malarial drugs for malaria prevention in pregnancy published from January 2010 to July 2019. Results: Eighteen trials were included in this review that collected multiple longitudinal safety outcomes including AEs. Statistical analysis and reporting of the safety outcomes in all the trials used descriptive statistics; proportions/counts (n = 18, 100%) and mean/median (n = 2, 11.1%). Results presentation included tabular (n = 16, 88.9%) and text description (n = 2, 11.1%). Univariate inferential methods were reported in most trials (n = 16, 88.9%); including Chi square/Fisher's exact test (n = 12, 66.7%), t test (n = 2, 11.1%) and Mann-Whitney/Wilcoxon test (n = 1, 5.6%). Multivariable methods, including Poisson and negative binomial were reported in few trials (n = 3, 16.7%). Assessment of a potential link between missing efficacy data and safety outcomes was not reported in any of the trials that reported efficacy missing data (n = 7, 38.9%). Conclusion: The review demonstrated that statistical analysis of safety data in anti-malarial drugs for malarial chemoprevention in pregnancy RCTs is inadequate. The analyses insufficiently account for multiple safety outcomes potential dependence, follow-up time and informative missing data which can compromise anti-malarial drug safety evidence development, based on the available data.
Article
Full-text available
Cotrimoxazole (CTX) has been used for half a century. It is inexpensive hence the reason for its almost universal availability and wide clinical spectrum of use. In the last decade, CTX was used for prophylaxis of opportunistic infections in HIV infected people. It also had an impact on the malaria risk in this specific group. We performed a systematic review to explore the efficacy and safety of CTX used for P.falciparum malaria treatment and prophylaxis. CTX is safe and efficacious against malaria. Up to 75% of the safety concerns relate to skin reactions and this increases in HIV/AIDs patients. In different study areas, in HIV negative individuals, CTX used as malaria treatment cleared 56%-97% of the malaria infections, reduced fever and improved anaemia. CTX prophylaxis reduces the incidence of clinical malaria in HIV-1 infected individuals from 46%-97%. In HIV negative non pregnant participants, CTX prophylaxis had 39.5%-99.5% protective efficacy against clinical malaria. The lowest figures were observed in zones of high sulfadoxine-pyrimethamine resistance. There were no data reported on CTX prophylaxis in HIV negative pregnant women. CTX is safe and still efficacious for the treatment of P.falciparum malaria in non-pregnant adults and children irrespective of HIV status and antifolate resistance profiles. There is need to explore its effect in pregnant women, irrespective of HIV status. CTX prophylaxis in HIV infected individuals protects against malaria and CTX may have a role for malaria prophylaxis in specific HIV negative target groups.
Article
Full-text available
The non-equivalence of statistical significance and clinical importance has long been recognised, but this error of interpretation remains common. Although a significant result in a large study may sometimes not be clinically important, a far greater problem arises from misinterpretation of non-significant findings. By convention a P value greater than 5% (P>0.05) is called “not significant.” Randomised controlled clinical trials that do not show a significant difference between the treatments being compared are often called “negative.” This term wrongly implies that the study has shown that there is no difference, whereas usually all that has been shown is an absence of evidence of a difference. These are quite different statements.The sample size of controlled trials is generally inadequate, with a consequent lack of power to detect real, and clinically worthwhile, differences in treatment. Freiman et al1 found that only 30% of a sample of 71 trials published in the New England Journal of Medicine in 1978-9 with P>0.1 were large enough to have a 90% chance of detecting even a 50% difference in the effectiveness of the treatments being compared, and they found no improvement in a similar sample of trials published in 1988. To interpret all these “negative” trials as providing evidence of the ineffectiveness of new treatments is clearly wrong and foolhardy. The term “negative” should not be used in this context.2A recent example is given by a trial comparing octreotide and sclerotherapy in patients with variceal bleeding.3 The study was carried out on a sample of only 100 despite a reported calculation that suggested that 1800 patients were needed. This trial had only a 5% chance of getting a statistically significant result if the stated clinically worthwhile treatment difference truly existed. One consequence of such low statistical power was a wide confidence interval for the treatment difference. The authors concluded that the two treatments were equally effective despite a 95% confidence interval that included differences between the cure rates of the two treatments of up to 20 percentage points.Similar evidence of the dangers of misinterpretation of non-significant results is found in numerous metaanalyses (overviews) of published trials, when few or none of the individual trials were statistically large enough. A dramatic example is provided by the overview of clinical trials evaluating fibrinolytic treatment (mostly streptokinase) for preventing reinfarction after acute myocardial infarction. The overview of randomised controlled trials found a modest but clinically worthwhile (and highly significant) reduction in mortality of 22%,4 but only five of the 24 trials had shown a statistically significant effect with P<0.05. The lack of statistical significance of most of the individual trials led to a long delay before the true value of streptokinase was appreciated.While it is usually reasonable not to accept a new treatment unless there is positive evidence in its favour, when issues of public health are concerned we must question whether the absence of evidence is a valid enough justification for inaction. A recent publicised example is the suggested link between some sudden infant deaths and antimony in cot mattresses. Statements about the absence of evidence are common—for example, in relation to the possible link between violent behaviour and exposure to violence on television and video, the possible harmful effects of pesticide residues in drinking water, the possible link between electromagnetic fields and leukaemia, and the possible transmission of bovine spongiform encephalopathy from cows. Can we be comfortable that the absence of clear evidence in such cases means that there is no risk or only a negligible one?When we are told that “there is no evidence that A causes B” we should first ask whether absence of evidence means simply that there is no information at all. If there are data we should look for quantification of the association rather than just a P value. Where risks are small P values may well mislead: confidence intervals are likely to be wide, indicating considerable uncertainty. While we can never prove the absence of a relation, when necessary we should seek evidence against the link between A and B—for example, from case-control studies. The importance of carrying out such studies will relate to the seriousness of the postulated effect and how widespread is the exposure in the population.References↵Bailar JC, Mosteller FFreiman JA, Chalmers TC, Smith H, Kuebler RR.The importance of beta, the type II error, and sample size in the design and interpretation of the randomized controlled trial: survey of two sets of “negative” trials. In: Bailar JC, Mosteller F eds.Medical uses of statistics.2nd ed. Boston, MA: NEJM Books,1992: 357–73.↵Chalmers I.Proposal to outlaw the term “negative trial.”BMJ1985;290: 1002.↵Sung JJY, Chung SCS, Lai C-W, Chan FKL, Leung JWC, Yung M-L, Kassianides C, et al.Octreotide infusion or emergency sclerotherapy for variceal haemorrhage.Lancet1993;342:637–41.OpenUrlCrossRefMedlineWeb of Science↵Yusuf S, Collins R, Peto R, Furberg C, Stampfer MJ, Goldhaber SZ, et al.Intravenous and intracoronary fibrinolytic therapy in acute myocardial infarction: overview of results on mortality, reinfarction and side-effects from 33 randomized controlled trials.Eur Heart J1985;6:556–85.OpenUrlFREE Full Text
Article
The prevalence of infection with malarial parasites and the incidence of anaemia and delivery of infants with low birthweight (LBW) were investigated in 575 Malawian mothers who received one, two or three doses of sulfadoxine-pyrimethamine (SP) during pregnancy. All the subjects were enrolled at their first antenatal visit and all delivered at hospital. The prevalence of Plasmodium falciparum infection at first antenatal visit was 35.3% in primigravidae and 13.6% in multigravidae (P < 0.001). Mean haemoglobin concentration was significantly lower in primigravidae than in multigravidae (8.8 v. 9.5 g/dl; P< 0.001). Of the 233 women tested for HIV infection, 18.8% of the primigravidae and 23.7% of the multigravidae were seropositive. At delivery, there was no significant difference in parasite prevalence in peripheral or placental blood between women who had received one or two antenatal doses of SP. The multigravidae who had received two doses of SP had higher mean haemoglobin concentrations than those who had received just one (P = 0.009) [this difference was not seen in the primigravidae (P = 0.92)]. However, linear regression analysis indicated that the haematinic supplements given to the subjects contributed more to this increase in haemoglobin concentration than the SP. The mean birthweights were higher, and incidence of LBW lower in babies born to primi- and multi-gravidae who had received two or three doses of SP treatment than those seen in babies born to women who had had just one dose (P < 0.03 for each). The odds ratio for LBW in primigravidae compared with multigravidae decreased from 3.2 to 1.0 as the number of SP doses increased from one to three. The benefit of three doses (compared with none) was equivalent to the population-attributable risk of LBW in primigravidae being reduced from 34.6% to 0%. Subjects who were seropositive for HIV were twice as likely to give birth to LBW babies as the other subjects. The use of SP was not associated with maternal side-effects or perinatal complications. The present results indicate that multiple doses of SP taken during pregnancy will lead to a highly significant reduction in the incidence of LBW in infants born to primigravidae, even if the women have HIV infections. This reduction is observable even when parasite prevalence at delivery is high because of re-infections in late pregnancy; reduction in parasite prevalence earlier in pregnancy, as the result of SP treatment, leads to improved foetal growth.
Article
Malaria in pregnancy is one of the major causes of maternal morbidity worldwide, and leads to poor birth outcomes. There is a complex interaction between pregnancy and parasite-all favour the parasite and disadvantage the pregnant woman. Women who are semi-immune lose much of that immunity. They may present with placental malaria but with no parasites in their peripheral blood. A non-immune pregnant women and her fetus are at serious risk from falciparum malaria. The diagnosis and management of malaria in pregnancy, including the safety of antimalarial drugs and interactions of malaria with HIV in pregnancy are reviewed.
Article
Plasmodium falciparum infection during pregnancy is strongly associated with maternal anaemia and low birth weight, contributing to substantial morbidity and mortality in sub-Saharan Africa. Intermittent preventive treatment in pregnancy with sulfadoxine/pyrimethamine (IPTp-SP) has been one of the most effective approaches to reduce the burden of malaria during pregnancy in Africa. IPTp-SP is based on administering ≥2 treatment doses of sulfadoxine/pyrimethamine to pregnant women at predefined intervals after quickening (around 18–20 weeks). Randomised, controlled trials have demonstrated decreased rates of maternal anaemia and low birth weight with this approach. The WHO currently recommends IPTp-SP in malaria-endemic areas of sub-Saharan Africa. However, implementation has been suboptimal in part because of concerns of potential drug toxicities. This review evaluates the toxicity data of sulfadoxine/pyrimethamine, including severe cutaneous adverse reactions, teratogenicity and alterations in bilirubin metabolism. Weekly sulfadoxine/pyrimethamine prophylaxis is associated with rare but potentially fatal cutaneous reactions. Fortunately, sulfadoxine/pyrimethamine use in IPTp programmes in Africa, with 2–4 treatment doses over 6 months, has been well tolerated in multiple IPTp trials. However, sulfadoxine/pyrimethamine should not be administered concurrently with cotrimoxazole given their redundant mechanisms of action and synergistic worsening of adverse drug reactions. Therefore, HIV-infected pregnant women in malaria endemic areas who are already receiving cotrimoxazole prophylaxis should not also receive IPTp-SP. Although folate antagonist use in the first trimester is associated with neural tube defects, large case-control studies have demonstrated that sulfadoxine/pyrimethamine administered as IPTp (exclusively in the second and third trimesters and after organogenesis) does not result in an increased risk of teratogenesis. Folic acid supplementation is recommended for all pregnant women to reduce the rate of congenital anomalies but high doses of folic acid (5 mg/day) may interfere with the antimalarial efficacy of sulfadoxine/pyrimethamine. However, the recommended standard dose of folic acid supplementation (0.4 mg/day) does not affect antimalarial efficacy and may provide the optimal balance to prevent neural tube defects and maintain the effectiveness of IPTp-SP. No clinical association between sulfadoxine/pyrimethamine use and kernicterus has been reported despite the extensive use of sulfadoxine/pyrimethamine and related compounds to treat maternal malaria and congenital toxoplasmosis in near-term pregnant women and newborns. Although few drugs in pregnancy can be considered completely safe, sulfadoxine/pyrimethamine — when delivered as IPTp — has a favourable safety profile. Improved pharmacovigilance programmes throughout Africa are now needed to confirm its safety as access to IPTp-SP increases. Given the documented benefits of IPTp-SP in malaria endemic areas of Africa, access to this treatment for pregnant women should continue to expand.
Conference Paper
In 2010, there was an estimated 216 million episodes of malaria, of which approximately 81%, or 174 million cases occurred in the African region. Moreover, the estimated number of deaths due to malaria was 655 000, of which 91% still took place in Africa. Yet, results of effective intervention studies suggest that the true number could even be higher because of indirect effects of the disease on other health related issues like anemia. Therefore the aim of the study was to search for the correlation between the presences of malaria infection due to Plasmodium falciparum and anemia in a highly endemic area in children under 5 years of age. A cross sectional household based survey was conducted in the Health Zone of Mont Ngafula1, Kinshasa, during the dry season from April to August 2012. Informed consent, blood smears and the hemoglobin results were obtained from 700 children. The proportion of anemia in the global population was 51.1% (CI 95%: 47.4–54.9). Among the children with a positive blood smear, 76% presented anemia compared to 42% in the group with a negative blood smear (P < 0.000). The presence of anemia was also assessed within three different classes of parasitic density (<1000 p/ml, 1000–2000 p/ml, >2000 p/ml) but no significant statistical differences could be found. These results tend to show a correlation between the presence of malaria infection and the anemia status of children in a hyper endemic area for malaria.
Article
Background In sub-Saharan Africa, various bacterial diseases occur before pneumocystosis or toxoplasmosis in the course of HIV-1 infection, and are major causes of morbidity and mortality. We did a randomised, double blind, placebo-controlled clinical trial at community-health centres in Abidjan, Côte d'Ivoire, to assess the efficacy of trimethoprim-sulphamethoxazole (co-trimoxazole) chemoprophylaxis at early stages of HIV-1 infection. Methods 843 HIV-infected patients were screened and 545 enrolled in the study. Eligible adults (with HIV-1 or HIV-1 and HIV-2 dual seropositivity at stages 2 or 3 of the WHO staging system) received co-trimoxazole chemoprophylaxis (trimethoprim 160 mg, sulphamethoxazole 800 mg) daily or a matching placebo. The primary outcome was the occurrence of severe clinical events, defined as death or hospital admission irrespective of the cause. Analyses were by intention to treat. Findings Four of the randomised patients were excluded (positive for HIV-2 only). 120 severe events occurred among 271 patients in the co-trimoxazole group and 198 among 270 in the placebo group. Significantly fewer patients in the co-trimoxazole group than in the placebo group had at least one severe event (84 vs 124); the probability of remaining free of severe events was 63·7% versus 45·8% (hazard ratio 0·57 [95% CI 0·43–0·75], p=0·0001) and the benefit was apparent in all subgroups of initial CD4-cell count. Survival did not differ between the groups (41 vs 46 deaths, p=0·51). Co-trimoxazole was generally well tolerated though moderate neutropenia occurred in 62 patients (vs 26 in the placebo group). Interpretation Patients who might benefit from co-trimoxazole could be recruited on clinical criteria in community clinics without knowing the patients CD4-cell count. This affordable measure will enable quick public-health intervention, while monitoring bacterial susceptibility and haematological tolerance.