Next Article in Journal
Development of a Risk Score for the Prediction and Management of Pre-Eclampsia in Low-Resource Settings
Previous Article in Journal
Effects of Treatment on Structural and Functional Parameters of the Left Heart in Naïve Acromegaly Patients: Prospective Single-Centre Study: 12-Month Follow-Up
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Evaluating the Safety and Efficacy of Malaria Preventive Measures in Pregnant Women with a Focus on HIV Status: A Systematic Review and Network Meta-Analysis

by
Muayad Albadrani
1,2,*,
Heba M. Eltahir
3,
Ahmad Bakur Mahmoud
2,4 and
Mekky M. Abouzied
3,5
1
Department of Family and Community Medicine and Medical Education, College of Medicine, Taibah University, Madinah 42353, Saudi Arabia
2
Health and Life Research Center, Taibah University, Madinah 42353, Saudi Arabia
3
Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Madinah 42353, Saudi Arabia
4
Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taibah University, Madinah 42353, Saudi Arabia
5
Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2025, 14(10), 3396; https://doi.org/10.3390/jcm14103396
Submission received: 2 April 2025 / Revised: 5 May 2025 / Accepted: 8 May 2025 / Published: 13 May 2025
(This article belongs to the Section Infectious Diseases)

Abstract

:
Background and Objectives: Malaria poses significant threats to pregnant women, particularly in endemic regions. Preventive measures against it include insecticide-treated bed nets, intermittent preventive treatment, and various supplements. We aimed to assess and compare the safety and effectiveness of malaria preventive measures in pregnant women, considering their HIV status. Methods: We conducted a systematic search of PubMed, the Cochrane Library, Scopus, Embase, and Web of Science through January 2024. A network meta-analysis was performed using R 4.3.3 software on 35 studies (50,103 participants). Results: In HIV-positive pregnant women, Co-trimoxazole with dihydroartemisinin significantly reduced malaria incidence compared to Co-trimoxazole alone (RR = 0.45, 95% CI [0.30; 0.68]) and sulfadoxine–pyrimethamine (SP) (RR = 0.14, 95% CI [0.04; 0.48]). Mefloquine was also effective compared to controls and SP. In HIV-negative women, azithromycin–piperaquine significantly reduced infections compared to SP, bed nets, and controls (RR = 0.03, 95% CI [0.00; 0.83]; RR = 0.03, 95% CI [0.00; 0.86]; and RR = 0.03, 95% CI [0.00; 0.77], respectively). Conclusion: Different combinations of preventive measures show varying effectiveness based on HIV status. Co-trimoxazole with dihydroartemisinin and mefloquine are effective for HIV-infected pregnant women, while azithromycin–piperaquine and mefloquine work well for those without HIV. Customized prevention strategies considering HIV status are crucial for optimal protection.

1. Introduction

Malaria is a significant contributor to illness and death on a global scale, particularly impacting children below the age of five and pregnant women, who are the most vulnerable populations [1]. In 2022, the global number of malaria cases was estimated to be 249 million, surpassing the pre-pandemic figure of 233 million cases recorded in 2019 by an additional 16 million. Besides the challenges posed by the COVID-19 pandemic, the worldwide efforts to address malaria have encountered various emerging threats, including drug and insecticide resistance, humanitarian crises, limitations in resources, the impacts of climate change, and delays in implementing programs, especially in nations heavily burdened by the disease [1,2,3]. Malaria places a significant health and socioeconomic burden on global populations, with approximately 3.2 billion individuals facing the risk of malaria infection [4]. From 2000 to 2015, there was a 37% decline in global malaria incidence, progress attributed to economic development and urbanization in numerous endemic nations [4,5]. Additionally, there was a notable rise in investments aimed at addressing malaria, resulting in increased preventive measures, enhanced diagnostics, and improved treatment strategies [6].
Vector control is crucial in the efforts to control and eliminate malaria. The ability of vectors to transmit parasites and their susceptibility to control measures vary among mosquito species and are influenced by local environmental factors. Current prevention practices predominantly rely on personal preventive measures, which aim to minimize contact between adult mosquitoes and humans. Notably, these measures include two types of insecticide-treated nets (ITNs): long-lasting insecticidal nets (LLINs) with insecticide embedded during manufacturing for prolonged effectiveness, and regular ITNs requiring insecticide reapplication every six months. Another approach is indoor residual spraying (IRS), involving the application of insecticides on household walls [7].
Furthermore, anti-malarial chemoprophylaxis is employed for malaria prevention in children and pregnant women. Sulfadoxine–pyrimethamine (SP), mefloquine (MQ), amodiaquine (AQ), dihydroartemisinin–piperaquine (DP), and artesunate (AS) are commonly used prophylactic drugs, offering the advantage of achieving full prophylactic effects with a single dose [8,9]. Several less commonly employed measures in malaria prevention include insecticide-treated curtains (ITCs), mosquito coils, insecticide-treated hammocks, and insecticide-treated tarpaulins. Despite a global decrease in malaria incidence, the most effective common preventive interventions for malaria infection remain unclear. Identifying the most effective interventions is essential for prioritizing resources. A single comparative study evaluating preventive efficacy across insecticide-treated nets (ITNs), indoor residual spraying (IRS), and prophylactic drugs (PDs) found that IRS is as effective as ITNs in reducing malaria-attributable mortality in children [9]. While the WHO previously endorsed sulfadoxine–pyrimethamine (SP), the diminishing effectiveness of SP in addressing symptomatic malaria over the years has raised apprehensions regarding its appropriateness for extended use in intermittent preventive treatment.
Malaria during pregnancy is a significant global health problem, particularly in areas with moderate-to-high transmission. Pregnant women are more susceptible to malaria due to reduced immunity, and HIV co-infection further increases their vulnerability. The WHO recommends a package of interventions for preventing and controlling malaria during pregnancy. For pregnant women in areas with moderate-to-high transmission of Plasmodium falciparum, the WHO recommends intermittent preventive treatment with SP, starting in the second trimester. For HIV-positive pregnant women, daily Co-trimoxazole (CTX) prophylaxis is the standard. These recommendations are crucial for tailoring malaria preventive strategies to the specific needs of pregnant women, considering their HIV status. Preventive measures include ITNs, intermittent preventive treatment, and various supplements.
We aim to evaluate and compare the safety and efficacy of various preventive strategies employed to combat malaria in pregnant women, with a specific consideration of their HIV status. This assessment encompasses an exploration of different interventions, such as insecticide-treated nets (ITNs), indoor residual spraying (IRS), and anti-malarial chemoprophylaxis, in order to discern their comparative advantages and potential drawbacks in mitigating the risk of malaria infection during pregnancy. Additionally, we seek to examine how HIV status influences the effectiveness and safety profiles of these preventive measures, aiming to provide nuanced insights into the optimal strategies for malaria prevention in pregnant women living with HIV.

2. Methods

We conducted our systematic review and network meta-analysis in adherence with the preferred reporting items for systematic reviews and meta-analyses (PRISMA) guidelines for network meta-analysis. Also, we followed the guidelines outlined in the Cochrane Handbook for systematic reviews throughout this study [10,11].

2.1. Searching Databases and Keywords

We searched four databases in January 2024 (PubMed, Cochrane Library, Web of Science, and Scopus), and two individual authors searched. The detailed search string used for each database is provided in Supplementary Table S1. We carried out our search without imposing limitations on time or language and supplemented it by manually examining the references of the studies included in our analysis.

2.2. Eligibility Criteria and Study Selection

We included all eligible studies that used preventive measures against malaria in pregnant women with or without HIV. We included all possible interventions, and we compared the results of each intervention regarding maternal and neonatal outcomes, in addition to safety outcomes such as abdominal pain, dizziness, headache, nausea, vomiting, neonatal deaths, preterm birth, and stillbirth. We excluded cohorts, letters, abstracts that did not provide information, case controls, and case series. Titles and abstracts were initially screened, followed by a thorough examination of the full texts of potentially relevant studies to assess eligibility and determine the final set of included studies.

2.3. Data Extraction

We extracted the following data from the included studies. (A) Baseline data: study ID, follow-up duration site, study design, maternal age, gestational age, gravid. (B) Summary data, including arm description, diagnostic tools, primary endpoint, and conclusion. (C) The outcomes that we included in our analysis were as follows: 1—incidence of malarial infection; 2—maternal anemia at delivery; 3—low birth weight (less than 2.5 kg); 4—abdominal pain incidence; 5—headache; 6—nausea; 7—vomiting; 8—neonatal deaths; 9—preterm birth; and 10—stillbirth. Two reviewers independently extracted the data to ensure accuracy and consistency. Any discrepancies between the reviewers were resolved through consultation with a third reviewer. This approach ensured that the data extraction process was thorough and reliable, contributing to the robustness of our findings.

2.4. Quality Assessment

We employed the Cochrane risk of bias tool [12] to evaluate the quality of the included RCT studies, assessing various domains such as the random sequence generation, concealed allocation, blinding of participants and personnel, blinding of outcome assessors, handling of incomplete data, selective reporting, and other relevant aspects. Each domain was independently evaluated by two authors, and conflicts were resolved through consultation with a third author. We also used quasi-experimental study design risk of bias assessment to assess two studies [13].

2.5. Statistical Analysis

We utilized the netmeta package in R 4.3.3 software to perform a frequentist network meta-analysis. Network plots were created to visually display the interventions and their direct and indirect comparisons, helping to understand the structure and strength of the network. The assumptions of NMA, including transitivity, consistency, and similarity, were carefully considered. Transitivity was assumed based on the similarity of patient characteristics, interventions, and outcomes across studies, allowing for valid indirect comparisons. Consistency was assessed using node-splitting methods to compare direct and indirect evidence within the network, ensuring the robustness of our findings. Similarity was ensured by including studies that were comparable in design and execution. The reference treatment was selected based on its common use and relevance in the included studies, providing a stable and consistent comparator across the network. A random-effects model was used to account for variability among studies and ensure a comprehensive analysis. Outcomes were pooled using both direct and indirect evidence to provide a comprehensive estimate of the relative effects of the interventions. Heterogeneity among studies was assessed using the Chi-squared test (Q2) and I-squared test, with significant heterogeneity defined as I2 > 50% or a p-value < 0.1. A random-effects model was applied to address significant heterogeneity. P-scores were calculated to rank the interventions based on their effectiveness and safety profiles, providing a quantitative measure of the relative performance of each intervention. By incorporating these elements, we aimed to provide a robust and transparent analysis of the effectiveness and safety of malaria preventive measures in pregnant women.

3. Results

3.1. Literature Search

The initial database search yielded 20,903 records, reduced to 16,545 after removing 4358 duplicates. Subsequent title and abstract screening identified 139 studies for full-text assessment, ultimately including 35 studies [14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48] in the systematic review; 30 of these studies of were included in the analysis (Figure 1, PRISMA).

3.2. Summary and Baseline Characteristics of Included Studies

Our network meta-analysis comprised nine studies that were about pregnant women with HIV, while the remaining included studies were about HIV-negative pregnant women. The studies included a total of 50,103 participants, representing a diverse population from multiple countries, with a predominant focus on the African region. The countries included were Mali, Pakistan, Australia, Kenya, Thailand, the Republic of Congo, Gambia, Benin, Burkina Faso, Ghana, Nigeria, Tanzania, Mozambique, Uganda, and Malawi. Most participants were within the maternal and gestational age range of 20 to 30 years. The follow-up duration across the studies varied from three months to two years. Various diagnostic tools were employed: quantitative PCR, nested PCR, loop-mediated isothermal amplification, thick and thin blood smears stained with Giemsa stain, microscopic examination, and targeted next-generation sequencing for molecular markers. The primary endpoint in most studies was the incidence of malarial infections. The studies included in our analysis covered a period from 1993 to 2024 (Table 1 and Table 2).

3.3. Risk of Bias

Most of the RCTs were low-risk regarding randomization and allocation processes. Moreover, all the RCTs were low-risk regarding reporting bias. However, only two of the included studies were low-risk regarding all the aspects of risk of bias except for other bias [17,42]. They were both low-risk for all aspects of risk of bias except for one aspect, while most of the other included studies were at a high risk of bias. Analysis of the two other quasi-experimental studies showed that Kumar et al., 2020 [27], was fair in quality and Roh et al., 2022 [39], was good in quality. All the details about the risk of bias and quality assessment are presented in Figure 2 and Supplementary Table S2, respectively.

3.4. Outcomes

(A)
Preventive measures for pregnant women with HIV
  • Incidence of malarial infection.
The combination of CTX with DP caused a significantly lower incidence rate when compared to CTX alone (RR = 0.45, 95% CI [0.30; 0.68]) and SP doses (RR = 0.14, 95% CI [0.04; 0.48]). The MQ intervention had a significantly lower incidence of malarial infection when compared with controls and two SP doses, and the results were (RR = 0.47, 95% CI [0.27; 0.82]), and (RR = 0.19, 95% CI [0.27; 0.82]), respectively. Also, CTX had a significantly lower incidence rate than the two SP doses. Nevertheless, the results were heterogeneous. The best three treatments, according to p-score, were the combination of CTX with DP, MQ, and the combination of multivitamins and VA (Figure 3).
2.
Maternal anemia at delivery.
No significant difference was found among interventions regarding maternal anemia at delivery. According to the p-scores, the interventions that induced anemia the least were arranged as two SP doses, followed by SP, CTX, and MQ (Supplementary Figure S1).
3.
Low birth weight.
We found no significant difference between the different interventions regarding low birth weight. The lowest incidence of a low birth weight of less than 2.5 kg, according to p-score, was observed with AZ, followed by MQ, followed by two SP doses (Supplementary Figure S2).

3.5. Safety Outcomes in Pregnant Women with HIV and Preventive Measures

No significant difference was detected among the different interventions regarding dizziness and headache. A significantly lower incidence of vomiting was seen in CTX treatment compared to MQ (RR = 0.07, 95% CI [0.01; 0.30]) (Supplementary Figures S3–S5, respectively).
Regarding preterm birth and stillbirth, there was no significance among the interventions. According to the p-scores, SP was associated with the lowest incidence of preterm birth, while MQ was associated with the lowest stillbirth incidence (Supplementary Figures S6 and S7, respectively).
(B)
Pregnant women taking preventive measures without having HIV.
  • Incidence of malarial infection.
AZP significantly reduced infection when compared with two SP doses, bed nets, and controls; the results were (RR = 0.03, 95% CI [0.00; 0.83]), (RR = 0.03, 95% CI [0.00; 0.86]), and (RR = 0.03, 95% CI [0.00; 0.77]), respectively. Similarly, MQ significantly reduced infection when compared with two SP doses, bed nets, and controls; the results were (RR = 0.19, CI = 95% CI [0.05; 0.84]), (RR = 0.18, 95% CI [0.04; 0.77]), and (RR = 0.18, 95% CI [0.05; 0.63]), respectively. Nevertheless, the top treatments, according to p-score, that reduced malarial infection in patients without HIV were AZP, MQ, and DPm, followed by AQ. On the other hand, the least effective interventions were bed nets, two SP doses, and zinc (Figure 4).
2.
Maternal anemia at delivery.
Comparing the different interventions regarding maternal anemia at delivery, only MQ showed a significant decrease in the incidence compared to CQ (RR = 0.54, 95% CI [0.31; 0.94]) (Supplementary Figure S8).
3.
Low birth weight.
No significant difference was detected among the different interventions regarding neonatal birth weight. However, the lowest incidence of a neonatal birth weight of less than 2.5 kg, according to p-score, was found in IST DPm, followed by SST DPm and MQ (Supplementary Figure S9).

3.6. Safety Outcomes in Pregnant Women Taking Preventive Measures Without Having HIV

Regarding the incidence of abdominal pain, SP had a significantly lower incidence when compared to MQ, SPAQ, and AQ; the results were (RR = 0.51, 95% CI [0.27; 0.93]), (RR = 0.46, 95% CI [0.33; 0.66]), and (RR = 0.44, 95% CI [0.31; 0.62]), respectively (Figure 5).
Regarding the incidence of dizziness, AL was associated with the lowest incidence of dizziness and was significant compared to all arms except for DP. It should be noted that DP and SP were associated with lower incidences of dizziness than most other interventions (Figure 6). DPm had the highest incidence of nausea compared to other arms, where its effect was significantly different from SP (RR = 0.05), AQ (RR = 0.10), and AZP (RR = 0.11). Regarding vomiting, the intervention of SP was associated with the lowest incidence of vomiting, with a significant result compared to MQ and AQ, with (RR = 0.23) and (RR = 0.28), respectively. No significant difference could be detected among the interventions regarding headache incidence rate as a side effect (Supplementary Figures S10–S12, respectively).

3.7. Neonatal Deaths

Treatment with CQ resulted in the highest incidence of neonatal death. SST DPm showed a significantly lower incidence of neonatal death when compared with SP, AL, and MQ, and the results were (RR = 0.05, 95% CI [0.00; 0.90]), (RR = 0.04, 95% CI [0.00; 0.82]), and (RR = 0.04, 95% CI [0.00; 0.76]), respectively. It should be noted that the lowest incidence of neonatal deaths based on the p-score was observed with DPm treatment, followed by two SP doses, and then SST DPm (Figure 7).

3.8. Stillbirth

The highest incidence of stillbirth was observed in the case of MQ treatment. In contrast, both SST DPm and CQ Px showed significantly lower stillbirth incidence when compared with MQ, and the results were (RR = 0.04, 95% CI [0.00; 0.76]) and (RR = 0.4, 95% CI [0.17; 0.94]), respectively. The lowest incidence of stillbirth, according to p-score, was observed with SST DPm, followed by IST DPm and then DPm (Supplementary Figure S13).

4. Discussion

Travelers visiting high-risk malaria areas, particularly pregnant women with or without HIV, should consider taking anti-malarial medication. However, chemoprophylaxis is not advisable for destinations with sporadic malaria cases and a low transmission risk. The choice of medication depends on factors such as local drug resistance, travel duration, medical history, allergies, and potential side effects. Additionally, individuals can reduce infection risk by taking preventive measures, including limiting outdoor activities, using insect repellents, and using insecticide-treated bed nets. Our study emphasizes the effectiveness of various preventive measures against malaria in both HIV-positive and -negative individuals. Combinations like Co-trimoxazole with dihydroartemisinin and mefloquine demonstrate efficacy in reducing malaria incidence compared to other interventions. Meanwhile, azithromycin with piperaquine and dihydroartemisinin is effective in HIV-negative individuals. However, safety concerns exist for interventions like mefloquine in pregnant women. Multivitamin supplementation and azithromycin also hold promise. Overall, tailored preventive strategies considering factors like HIV status and pregnancy are crucial.
Before the widespread implementation of antiretroviral therapy, Co-trimoxazole (CTX) was a cost-effective, broad-spectrum antimicrobial medication extensively utilized in developing nations. It played a crucial role in decreasing morbidity and mortality among individuals, including both adults and children, living with HIV by preventing various infections such as bacterial infections, diarrhea, malaria, and Pneumocystis pneumonia, even in the face of prevalent microbial resistance [49]. According to previous studies, CTX prophylaxis significantly reduces early mortality rates [49,50,51]. Since 2001, the World Health Organization (WHO) has endorsed artemisinin-based combination therapies (ACTs) as the primary treatment for uncomplicated P. falciparum malaria [7]. Artemisinin and its derivatives are well known for their strong anti-malarial properties and have been widely adopted for clinical use in regions where malaria is endemic. In laboratory settings, the artemisinin concentration required to inhibit 50% of Plasmodium falciparum growth ranges from 3 to 30 μg/L [51]. The combination of CTX and artemisinin-based combination therapies for prevention and treatment has shown effectiveness against malaria in HIV-positive patients. CTX reduces morbidity and mortality in individuals with HIV by preventing various infections, including malaria. Meanwhile, ACTs, endorsed by the World Health Organization since 2001, are potent in treating uncomplicated P. falciparum malaria. Combining these drugs offers a synergistic approach, enhancing malaria management strategies, especially in endemic regions [42,52].
Mefloquine is widely recognized for its high efficacy in preventing and treating malaria. It is considered one of the most effective anti-malarial drugs available, particularly in regions where malaria parasites have not developed resistance to it. When used correctly and combined with other preventive measures, mefloquine can provide robust protection against malaria infection [53,54]. The World Health Organization (WHO) permits the use of mefloquine for pregnant women during the second and third trimesters, while some authorities, such as the Centers for Disease Control and Prevention (CDC), extend this approval to the first trimester [55]. In the event of accidental pregnancy while using mefloquine, termination is not recommended. Additionally, mefloquine chemoprophylaxis is considered safe during breastfeeding. Studies indicate that mefloquine is a viable option for other high-risk groups, including long-term travelers, visiting friends and relatives (VFR) travelers, and families with young children. Despite negative media portrayal, extensive pharmaco-epidemiological investigations have demonstrated that serious adverse events associated with mefloquine are rare [56]. In our study, we found that mefloquine is not highly ranked in terms of safety outcomes in pregnant women without HIV, and it may even increase the number of stillbirth infants significantly.
The use of multivitamin supplements containing vitamin B complexes, C, and E, has been observed to decelerate disease progression and lower the occurrence of HIV-associated complications such as dysentery and acute upper respiratory infections in HIV-positive women. However, it remains unclear whether multivitamins impact malaria susceptibility in HIV-positive women. Research conducted among children indicates that multivitamin supplementation may reduce the incidence of clinical malaria [46,57].
Azithromycin has been investigated as a potential anti-malarial agent due to its slow yet potent activity against malaria parasites, targeting the apicoplast organelle [58,59]. It is considered the most potent anti-malarial macrolide, demonstrating significant activity against cultured Plasmodium falciparum after extended in vitro exposure [59]. In treating uncomplicated falciparum malaria, combinations such as artesunate plus azithromycin have shown improved efficacy compared to artesunate alone. However, they are less effective than combinations including mefloquine or dihydroartemisinin [60]. Studies assessing azithromycin in combination with chloroquine have produced mixed results, with some showing promising efficacy while others find it inferior to alternative treatments like artemether–lumefantrine [60,61].
Our study possesses several strengths, notably in comprising most of our included studies, which were randomized controlled trials and considered the gold standard in evidence quality. Our study marks the first network meta-analysis to systematically compare various outcomes between pregnant women with HIV and those without HIV. With a substantial participant pool of 50,103 individuals across 35 studies, our study provides comprehensive insights into the efficacy and safety of preventive measures against malaria in pregnancy. Our findings promise to inform future decision-making regarding selecting appropriate preventive strategies for malaria infection. However, it is essential to acknowledge certain limitations. A prevalent risk of bias compromised the overall quality of the included studies. Furthermore, factors inherent to pregnancy may confound the association between preventive measures and malaria incidence. Adverse events observed during the study period may not solely be attributable to malaria infection or preventive measures but could also be influenced by the physiological changes associated with pregnancy. Additionally, variations in malaria detection techniques may introduce heterogeneity into our analysis.

5. Conclusions

Our study highlights the efficacy of various preventive measures against malaria in both HIV-positive and -negative individuals. Combinations like Co-trimoxazole with dihydroartemisinin and mefloquine show effectiveness in reducing malaria incidence compared to other interventions, while azithromycin with piperaquine and dihydroartemisinin are effective in HIV-negative individuals compared to other interventions. However, concerns exist regarding the safety of certain interventions, such as mefloquine, in pregnant women. Multivitamin supplementation and azithromycin also show promise, but further research is needed to confirm their effectiveness. Overall, tailored preventive strategies considering factors like HIV status and pregnancy are essential, with future research focusing on optimizing interventions while ensuring patient safety.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/jcm14103396/s1, Table S1: Detailed search strategy for retrieved databases. Table S2: Quality assessment of included quasi-experimental studies.; Figure S1: Incidence of maternal anemia at delivery in pregnant women with HIV. (A) Network graph showing direct evidence between evaluated interventions. (B) Forest plot comparing all interventions. (C) League table representing network meta-analysis estimates for all interventions’ comparisons.; Figure S2: Incidence of low birth weight in pregnant women with HIV. (A) Network graph showing direct evidence between evaluated interventions. (B) Forest plot comparing all interventions. (C) League table representing network meta-analysis estimates for all interventions’ comparisons.; Figure S3: Incidence of dizziness in pregnant women with HIV. (A) Network graph showing direct evidence between evaluated interventions. (B) Forest plot comparing all interventions. (C) League table representing network meta-analysis estimates for all interventions’ comparisons.; Figure S4: Incidence of headache in pregnant women with HIV. (A) Network graph showing direct evidence between evaluated interventions. (B) Forest plot comparing all interventions. (C) League table representing network meta-analysis estimates for all interventions’ comparisons.; Figure S5: Incidence of vomiting in pregnant women with HIV. (A) Network graph showing direct evidence between evaluated interventions. (B) Forest plot comparing all interventions. (C) League table representing network meta-analysis estimates for all interventions’ comparisons.; Figure S6: Incidence of preterm births in pregnant women with HIV. (A) Network graph showing direct evidence between evaluated interventions. (B) Forest plot comparing all interventions. (C) League table representing network meta-analysis estimates for all interventions’ comparisons.; Figure S7: Incidence of stillbirths in pregnant women with HIV. (A) Network graph showing direct evidence between evaluated interventions. (B) Forest plot comparing all interventions. (C) League table representing network meta-analysis estimates for all interventions’ comparisons.; Figure S8: Incidence of maternal anemia at delivery in pregnant women without HIV. (A) Network graph showing direct evidence between evaluated interventions. (B) Forest plot comparing all interventions. (C) League table representing network meta-analysis estimates for all interventions’ comparisons.; Figure S9: Incidence of low birth weight in pregnant women without HIV. (A) Network graph showing direct evidence between evaluated interventions. (B) Forest plot comparing all interventions. (C) League table representing network meta-analysis estimates for all interventions’ comparisons.; Figure S10: Incidence of nausea in pregnant women without HIV. (A) Network graph showing direct evidence between evaluated interventions. (B) Forest plot comparing all interventions. (C) League table representing network meta-analysis estimates for all interventions’ comparisons.; Figure S11: Incidence of vomiting in pregnant women without HIV. (A) Network graph showing direct evidence between evaluated interventions. (B) Forest plot comparing all interventions. (C) League table representing network meta-analysis estimates for all interventions’ comparisons.; Figure S12: Incidence of headache in pregnant women without HIV. (A) Network graph showing direct evidence between evaluated interventions. (B) Forest plot comparing all interventions. (C) League table representing network meta-analysis estimates for all interventions’ comparisons.; Figure S13: Incidence of stillbirths in pregnant women without HIV. (A) Network graph showing direct evidence between evaluated interventions. (B) Forest plot comparing all interventions. (C) League table representing network meta-analysis estimates for all interventions’ comparisons.

Author Contributions

Conceptualization, M.A.; methodology, H.M.E., M.M.A., A.B.M. and M.A.; formal analysis, H.M.E., M.M.A. and M.A.; data curation, H.M.E., M.M.A., A.B.M. and M.A.; writing—original draft, H.M.E., M.M.A., A.B.M. and M.A.; writing—review and editing, H.M.E., M.M.A., A.B.M. and M.A.; supervision, M.A. All authors have read and agreed to the published version of the manuscript.

Funding

This scientific paper was derived from a research grant funded by the Research, Development, and Innovation Authority (RDIA)—Kingdom of Saudi Arabia—with the grant number (12982-iau-2023-TAU-R-3-1-HW-).

Data Availability Statement

Data are available from the corresponding author upon reasonable request.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

SSTSingle screening and treatment
ISTIntermittent screening and treatment
IPTpIntermittent preventive treatment during pregnancy
DPmMonthly dihydroartemisinin
MQMefloquine
SPSulfadoxine and pyrimethamine
AQAmodiaquine
SPAQSulfadoxine and pyrimethamine plus amodiaquine
VAVitamin A
CQ PxProphylactic chloroquine
SPAZSulfadoxine and pyrimethamine plus azithromycin
DPAZDihydroartemisinin and azithromycin
AZ-PQ or AZPAzithromycin and piperaquine
MQASMefloquine–artesunate
ITNInsecticide-treated Nets
ALArtemether–lumefantrine
EFVEfavirenz
LLINsLong-lasting insecticide-treated bed nets
CTXCo-trimoxazole
TMP-SMXTrimethoprim-sulfa-methoxazole
AZAzithromycin

References

  1. WHO. Malria Report 2023. Available online: https://reliefweb.int/report/world/world-malaria-report-2023-enarruzh?gad_source=1&gclid=Cj0KCQjw-r-vBhC-ARIsAGgUO2Bxfw30urOvg7o_XLArrcglD9vsFqKveWSVAWOGspP_zKXr9KMC-LEaAtUvEALw_wcB (accessed on 5 January 2024).
  2. World Health Organization. WHO Guidelines for Malaria; World Health Organization: Geneva, Switzerland, 2022. [Google Scholar]
  3. Wang, Z.; Liu, Y.; Li, Y.; Wang, G.; Lourenço, J.; Kraemer, M.; He, Q.; Cazelles, B.; Li, Y.; Wang, R.; et al. The relationship between rising temperatures and malaria incidence in Hainan, China, from 1984 to 2010: A longitudinal cohort study. Lancet Planet Health 2022, 6, e350–e358. [Google Scholar] [CrossRef] [PubMed]
  4. Patel, P.; Bagada, A.; Vadia, N. Epidemiology and Current Trends in Malaria. Rising Contag. Dis. Basics Manag. Treat. 2024, 261–282. [Google Scholar] [CrossRef]
  5. Cotter, C.; Sturrock, H.J.; Hsiang, M.S.; Liu, J.; Phillips, A.A.; Hwang, J.; Gueye, C.S.; Fullman, N.; Gosling, R.D.; Feachem, R.G. The changing epidemiology of malaria elimination: New strategies for new challenges. Lancet 2013, 382, 900–911. [Google Scholar] [CrossRef]
  6. Murray, C.J.; Rosenfeld, L.C.; Lim, S.S.; Andrews, K.G.; Foreman, K.J.; Haring, D.; Fullman, N.; Naghavi, M.; Lozano, R.; Lopez, A.D. Global malaria mortality between 1980 and 2010: A systematic analysis. Lancet 2012, 379, 413–431. [Google Scholar] [CrossRef] [PubMed]
  7. WHO GS. World Malaria Report; WHO: Geneva, Switzerland, 2013. [Google Scholar]
  8. Greenwood, B. Anti-malarial drugs and the prevention of malaria in the population of malaria endemic areas. Malar. J. 2010, 9 (Suppl. S3), S2. [Google Scholar] [CrossRef]
  9. White, N.J. Intermittent Presumptive Treatment for Malaria. PLoS Med. 2005, 2, e3. [Google Scholar] [CrossRef] [PubMed]
  10. Cumpston, M.; Li, T.; Page, M.; Chandler, J.; Welch, V.; Higgins, J.P.; Thomas, J. Cochrane Handbook for Systematic Reviews of Interventions; Wiley: Hoboken, NJ, USA, 2019. [Google Scholar]
  11. Hutton, B.; Salanti, G.; Caldwell, D.M.; Chaimani, A.; Schmid, C.H.; Cameron, C.; Ioannidis, J.P.A.; Straus, S.; Thorlund, K.; Jansen, J.P.; et al. The PRISMA Extension Statement for Reporting of Systematic Reviews Incorporating Network Meta-analyses of Health Care Interventions: Checklist and Explanations. Ann. Intern. Med. 2015, 162, 777–784. [Google Scholar] [CrossRef]
  12. Boutron, I.P.M.; Higgins, J.P.T.; Altman, D.G.; Lundh, A.; Hróbjartsson, A. Chapter 7: Considering bias and conflicts of interest among the included studies. In Cochrane Handbook for Systematic Reviews of Interventions Version 6.2; Higgins, J.P.T., Thomas, J., Chandler, J., Cumpston, M., Li, T., Page, M.J., Welch, V.A., Eds.; Cochrane: London, UK, 2021; Available online: https://training.cochrane.org/handbook (accessed on 8 January 2024).
  13. Waddington, H.; Aloe, A.M.; Becker, B.J.; Djimeu, E.W.; Hombrados, J.G.; Tugwell, P.; Wells, G.; Reeves, B. Quasi-experimental study designs series—Paper 6: Risk of bias assessment. J. Clin. Epidemiol. 2017, 89, 43–52. [Google Scholar] [CrossRef]
  14. Ahmed, R.; Poespoprodjo, J.R.; Syafruddin, D.; Khairallah, C.; Pace, C.; Lukito, T.; Maratina, S.S.; Asih, P.B.S.; Santana-Morales, M.A.; Adams, E.R.; et al. Efficacy and safety of intermittent preventive treatment and intermittent screening and treatment versus single screening and treatment with dihydroartemisinin–piperaquine for the control of malaria in pregnancy in Indonesia: A cluster-randomized, open-label, superiority trial. Lancet Infect. Dis. 2019, 19, 973–987. [Google Scholar] [CrossRef]
  15. Briand, V.; Bottero, J.; Noël, H.; Masse, V.; Cordel, H.; Guerra, J.; Kossou, H.; Fayomi, B.; Ayemonna, P.; Fievet, N.; et al. Intermittent treatment for the prevention of malaria during pregnancy in Benin: A randomized, open-label equivalence trial comparing sulfadoxine-pyrimethamine with mefloquine. J. Infect. Dis. 2009, 200, 991–1001. [Google Scholar] [CrossRef]
  16. Clerk, C.A.; Bruce, J.; Affipunguh, P.K.; Mensah, N.; Hodgson, A.; Greenwood, B.; Chandramohan, D. A randomized, controlled trial of intermittent preventive treatment with sulfadoxine-pyrimethamine, amodiaquine, or the combination in pregnant women in Ghana. J. Infect. Dis. 2008, 198, 1202–1211. [Google Scholar] [CrossRef] [PubMed]
  17. Cox, S.E.; Staalsoe, T.; Arthur, P.; Bulmer, J.N.; Tagbor, H.; Hviid, L.; Frost, C.; Riley, E.M.; Kirkwood, B.R. Maternal vitamin A supplementation and immunity to malaria in pregnancy in Ghanaian primigravids. Trop. Med. Int. Health 2005, 10, 1286–1297. [Google Scholar] [CrossRef]
  18. Darling, A.M.; Mugusi, F.M.; Etheredge, A.J.; Gunaratna, N.S.; Abioye, A.I.; Aboud, S.; Duggan, C.; Mongi, R.; Spiegelman, D.; Roberts, D.; et al. Vitamin A and zinc supplementation among pregnant women to prevent placental malaria: A randomized, double-blind, placebo-controlled trial in Tanzania. Am. J. Trop. Med. Hyg. 2017, 96, 826–834. [Google Scholar] [CrossRef]
  19. Desai, M.; Gutman, J.; L’lanziva, A.; Otieno, K.; Juma, E.; Kariuki, S.; Ouma, P.; Were, V.; Laserson, K.; Katana, A.; et al. Intermittent screening and treatment or intermittent preventive treatment with dihydroartemisinin–piperaquine versus intermittent preventive treatment with sulfadoxine–pyrimethamine for the control of malaria during pregnancy in western Kenya: An open-label, three-group, randomized controlled superiority trial. Lancet 2015, 386, 2507–2519. [Google Scholar] [CrossRef] [PubMed]
  20. Divala, T.H.; Mungwira, R.G.; Mawindo, P.M.; Nyirenda, O.M.; Kanjala, M.; Ndaferankhande, M.; Tsirizani, L.E.; Masonga, R.; Muwalo, F.; Potter, G.E.; et al. A Randomized, Controlled Clinical Trial of Chloroquine as Chemoprophylaxis or Intermittent Preventive Therapy to Prevent Malaria in Pregnancy in Malawi. Lancet Infect. Dis. 2018, 18, 1097. [Google Scholar] [CrossRef] [PubMed]
  21. Dolan, G.; Ter Kuile, F.O.; Jacoutot, V.; White, N.J.; Luxemburger, C.; Malankirii, L.; Chongsuphajaisiddhi, T.; Nosten, F. Bed nets for the prevention of malaria and anaemia in pregnancy. Trans. R. Soc. Trop. Med. Hyg. 1993, 87, 620–626. [Google Scholar] [CrossRef]
  22. Filler, S.J.; Kazembe, P.; Thigpen, M.; Macheso, A.; Parise, M.E.; Newman, R.D.; Steketee, R.W.; Hamel, M. Randomized trial of 2-dose versus monthly sulfadoxine-pyrimethamine intermittent preventive treatment for malaria in HIV-positive and HIV-negative pregnant women in Malawi. J. Infect. Dis. 2006, 194, 286–293. [Google Scholar] [CrossRef] [PubMed]
  23. González, R.; Mombo-Ngoma, G.; Ouédraogo, S.; Kakolwa, M.A.; Abdulla, S.; Accrombessi, M.; Aponte, J.J.; Akerey-Diop, D.; Basra, A.; Briand, V.; et al. Intermittent preventive treatment of malaria in pregnancy with mefloquine in HIV-negative women: A multicentre randomized controlled trial. PLoS Med. 2014, 11, e1001733. [Google Scholar] [CrossRef]
  24. Jagannathan, P.; Kakuru, A.; Okiring, J.; Muhindo, M.K.; Natureeba, P.; Nakalembe, M.; Opira, B.; Olwoch, P.; Nankya, F.; Ssewanyana, I.; et al. Dihydroartemisinin-piperaquine for intermittent preventive treatment of malaria during pregnancy and risk of malaria in early childhood: A randomized controlled trial. PLoS Med. 2018, 15, e1002606. [Google Scholar] [CrossRef]
  25. Kayentao, K.; Kodio, M.; Newman, R.D.; Maiga, H.; Doumtabe, D.; Ongoiba, A.; Coulibaly, D.; Keita, A.S.; Maiga, B.; Mungai, M.; et al. Comparison of intermittent preventive treatment with chemoprophylaxis for the prevention of malaria during pregnancy in Mali. J. Infect. Dis. 2005, 191, 109–116. [Google Scholar] [CrossRef]
  26. Ter Kuile, F.O.; Kolczak, M.S.; Nahlen, B.L.; Friedman, J.F.; Shi, Y.P.; Phillips-Howard, P.A.; Hawley, W.A.; Lal, A.A.; Vulule, J.M.; Kariuki, S.K.; et al. Reduction of malaria during pregnancy by permethrin-treated bed nets in an area of intense perennial malaria transmission in western Kenya. Am. J. Trop. Med. Hyg. 2003, 68, 50–60. [Google Scholar] [CrossRef] [PubMed]
  27. Kumar, R.; Farzeen, M.; Hafeez, A.; Achakzai, B.K.; Vankwani, M.; Lal, M.; Iqbal, R.; Somrongthong, R. Effectiveness of a health education intervention on the use of long-lasting insecticidal nets for the prevention of malaria in pregnant women of Pakistan: A quasi-experimental study. Malar. J. 2020, 19, 232. [Google Scholar] [CrossRef]
  28. Lingani, M.; Zango, S.H.; Valéa, I.; Samadoulougou, S.; Somé, G.; Sanou, M.; Kaboré, B.; Rouamba, T.; Sorgho, H.; Tahita, M.C.; et al. Effects of maternal antenatal treatment with two doses of azithromycin added to monthly sulfadoxine-pyrimethamine for the prevention of low birth weight in Burkina Faso: An open-label randomized controlled trial. Malar. J. 2023, 22, 101. [Google Scholar] [CrossRef] [PubMed]
  29. Madanitsa, M.; Barsosio, H.C.; Minja, D.T.; Mtove, G.; Kavishe, R.A.; Dodd, J.; Saidi, Q.; Onyango, E.D.; Otieno, K.; Wang, D.; et al. Effect of monthly intermittent preventive treatment with dihydroartemisinin–piperaquine with and without azithromycin versus monthly sulfadoxine–pyrimethamine on adverse pregnancy outcomes in Africa: A double-blind randomized, partly placebo-controlled trial. Lancet 2023, 401, 1020–1036. [Google Scholar] [CrossRef] [PubMed]
  30. Mlugu, E.M.; Minzi, O.; Kamuhabwa, A.A.R.; Aklillu, E. Effectiveness of intermittent preventive treatment with dihydroartemisinin-piperaqunine against malaria in pregnancy in tanzania: A randomized controlled trial. Clin. Pharmacol. Ther. 2021, 110, 1478–1489. [Google Scholar] [CrossRef]
  31. Moore, B.R.; Benjamin, J.M.; Tobe, R.; Ome-Kaius, M.; Yadi, G.; Kasian, B.; Kong, C.; Robinson, L.J.; Laman, M.; Mueller, I.; et al. A randomized open-label evaluation of the anti-malarial prophylactic efficacy of azithromycin-piperaquine versus sulfadoxine-pyrimethamine in pregnant Papua New Guinean women. Antimicrob. Agents Chemother. 2019, 63, 10-1128. [Google Scholar] [CrossRef]
  32. Njagi, J.K.; Magnussen, P.; Estambale, B.; Ouma, J.; Mugo, B. Prevention of Anaemia in pregnancy using insecticide-treated bednets and sulfadoxine-pyrimethamine in a highly malarious area of Kenya: A randomized controlled trial. Trans. R. Soc. Trop. Med. Hyg. 2003, 97, 277–282. [Google Scholar] [CrossRef]
  33. Nosten, F.; ter Kuile, F.; Maelankiri, L.; Chongsuphajaisiddhi, T.; Nopdonrattakoon, L.; Tangkitchot, S.; Boudreau, E.; Bunnag, D.; White, N.J. Mefloquine prophylaxis prevents malaria during pregnancy: A double-blind, placebo-controlled study. J. Infect. Dis. 1994, 169, 595–603. [Google Scholar] [CrossRef]
  34. Labama Otuli, N.; Marini Djang’eing’a, R.; Losimba Likwela, J.; Bosenge Nguma, J.D.; Maindo Alongo, M.A.; Ahuka Ona Longombe, A.; Mbutu Mango, B.; Bono, D.M.N.; Mokili, J.L.; Manga Okenge, J.P. Efficacy and safety of malarial prophylaxis with mefloquine during pregnancy in Kisangani, Democratic Republic of Congo: A randomized clinical trial. Br. J. Clin. Pharmacol. 2021, 87, 3115–3126. [Google Scholar] [CrossRef]
  35. COSMIC Consortium. Community-based malaria screening and treatment for pregnant women receiving standard intermittent preventive treatment with sulfadoxine-pyrimethamine: A multicenter (The Gambia, Burkina Faso, and Benin) cluster-randomized controlled trial. Clin. Infect. Dis. 2019, 68, 586–596. [Google Scholar] [CrossRef]
  36. Steketee, R.W.; Wirima, J.J.; Hightower, A.W.; Slutsker, L.; Heymann, D.L.; Breman, J.G. The effect of malaria and malaria prevention in pregnancy on offspring birthweight, prematurity, and intrauterine growth retardation in rural Malawi. Am. J. Trop. Med. Hyg. 1996, 55 (Suppl. S1), 33–41. [Google Scholar] [CrossRef] [PubMed]
  37. Browne, E.N.L.; Maude, G.H.; Binka, F.N. The impact of insecticide-treated bednets on malaria and anaemia in pregnancy in Kassena-Nankana district, Ghana: A randomized controlled trial. Trop. Med. Int. Health 2001, 6, 667–676. [Google Scholar] [CrossRef]
  38. Kajubi, R.; Huang, L.; Jagannathan, P.; Chamankhah, N.; Were, M.; Ruel, T.; Koss, C.; Kakuru, A.; Mwebaza, N.; Kamya, M.; et al. Antiretroviral therapy with efavirenz accentuates pregnancy-associated reduction of dihydroartemisinin-piperaquine exposure during malaria chemoprevention. Clin. Pharmacol. Ther. 2017, 102, 520–528. [Google Scholar] [CrossRef] [PubMed]
  39. Roh, M.E.; Oundo, B.; Dorsey, G.; Shiboski, S.; Gosling, R.; Glymour, M.M.; Staedke, S.G.; Bennett, A.; Sturrock, H.; Mpimbaza, A. A quasi-experimental study estimating the impact of long-lasting insecticidal nets with and without piperonyl butoxide on pregnancy outcomes. Malar. J. 2022, 21, 5. [Google Scholar] [CrossRef] [PubMed]
  40. Akinyotu, O.; Bello, F.; Abdus-Salam, R.; Arowojolu, A. Comparative study of mefloquine and sulphadoxine–pyrimethamine for malaria prevention among pregnant women with HIV in southwest Nigeria. Int. J. Gynecol. Obstet. 2018, 142, 194–200. [Google Scholar] [CrossRef]
  41. Akinyotu, O.; Bello, F.; Abdus-Salam, R.; Arowojolu, A. A randomized controlled trial of azithromycin and sulphadoxine–pyrimethamine as prophylaxis against malaria in pregnancy among human immunodeficiency virus–positive women. Trans. R. Soc. Trop. Med. Hyg. 2019, 113, 463–470. [Google Scholar] [CrossRef]
  42. Barsosio, H.C.; Madanitsa, M.; Ondieki, E.D.; Dodd, J.; Onyango, E.D.; Otieno, K.; Wang, D.; Hill, J.; Mwapasa, V.; Phiri, K.S.; et al. Chemoprevention for malaria with monthly intermittent preventive treatment with dihydroartemisinin–piperaquine in pregnant women living with HIV on daily co-trimoxazole in Kenya and Malawi: A randomized, double-blind, placebo-controlled trial. Lancet 2024, 403, 365–378. [Google Scholar] [CrossRef]
  43. González, R.; Desai, M.; Macete, E.; Ouma, P.; Kakolwa, M.A.; Abdulla, S.; Aponte, J.J.; Bulo, H.; Kabanywanyi, A.M.; Katana, A.; et al. Intermittent preventive treatment of malaria in pregnancy with mefloquine in HIV-infected women receiving cotrimoxazole prophylaxis: A multicenter randomized placebo-controlled trial. PLoS Med. 2014, 11, e1001735. [Google Scholar] [CrossRef]
  44. Manirakiza, A.; Tondeur, L.; Ketta, M.Y.B.; Sepou, A.; Serdouma, E.; Gondje, S.; Bata, G.G.B.; Boulay, A.; Moyen, J.M.; Sakanga, O.; et al. Cotrimoxazole versus sulfadoxine–pyrimethamine for intermittent preventive treatment of malaria in HIV-infected pregnant women in Bangui, Central African Republic: A pragmatic randomized controlled trial. Trop. Med. Int. Health 2021, 26, 1314–1323. [Google Scholar] [CrossRef]
  45. Denoeud-Ndam, L.; Zannou, D.M.; Fourcade, C.; Taron-Brocard, C.; Porcher, R.; Atadokpede, F.; Komongui, D.G.; Dossou-Gbete, L.; Afangnihoun, A.; Ndam, N.T.; et al. Cotrimoxazole prophylaxis versus mefloquine intermittent preventive treatment to prevent malaria in HIV-infected pregnant women: Two randomized controlled trials. JAIDS J. Acquir. Immune Defic. Syndr. 2014, 65, 198–206. [Google Scholar] [CrossRef]
  46. Olofin, I.O.; Spiegelman, D.; Aboud, S.; Duggan, C.; Danaei, G.; Fawzi, W.W. Supplementation with multivitamins and vitamin A and incidence of malaria among HIV-infected Tanzanian women. Am. J. Ther. 2014, 67 (Suppl. S4), S173–S178. [Google Scholar] [CrossRef]
  47. Natureeba, P.; Kakuru, A.; Muhindo, M.; Ochieng, T.; Ategeka, J.; Koss, C.A.; Plenty, A.; Charlebois, E.D.; Clark, T.D.; Nzarubara, B.; et al. Intermittent preventive treatment with dihydroartemisinin-piperaquine for the prevention of malaria among HIV-infected pregnant women. J. Infect. Dis. 2017, 216, 29–35. [Google Scholar] [CrossRef] [PubMed]
  48. Nambozi, M.; Kabuya, J.-B.B.; Hachizovu, S.; Mwakazanga, D.; Mulenga, J.; Kasongo, W.; Buyze, J.; Mulenga, M.; Van Geertruyden, J.-P.; D’alessandro, U. Artemisinin-based combination therapy in pregnant women in Zambia: Efficacy, safety and risk of recurrent malaria. Malar. J. 2017, 16, 199. [Google Scholar] [CrossRef] [PubMed]
  49. Church, J.A.; Fitzgerald, F.; Walker, A.S.; Gibb, D.M.; Prendergast, A.J. The expanding role of co-trimoxazole in developing countries. Lancet Infect. Dis. 2015, 15, 327–339. [Google Scholar] [CrossRef] [PubMed]
  50. Berkley, J.A.; Ngari, M.; Thitiri, J.; Mwalekwa, L.; Timbwa, M.; Hamid, F.; Ali, R.; Shangala, J.; Mturi, N.; Jones, K.D.J.; et al. Daily co-trimoxazole prophylaxis to prevent mortality in children with complicated severe acute malnutrition: A multicentre, double-blind, randomised placebo-controlled trial. Lancet Glob. Health 2016, 4, e464–e473. [Google Scholar] [CrossRef]
  51. Nunn, A.J.; Mwaba, P.; Chintu, C.; Mwinga, A.; Darbyshire, J.H.; Zumla, A. Role of co-trimoxazole prophylaxis in reducing mortality in HIV infected adults being treated for tuberculosis: Randomized clinical trial. BMJ 2008, 337, a257. [Google Scholar] [CrossRef]
  52. Banda, C.G.; Nkosi, D.; Allen, E.; Workman, L.; Madanitsa, M.; Chirwa, M.; Kapulula, M.; Muyaya, S.; Munharo, S.; Tarning, J.; et al. Impact of dolutegravir-based antiretroviral therapy on piperaquine exposure following dihydroartemisinin-piperaquine intermittent preventive treatment of malaria in pregnant women living with HIV. Antimicrob. Agents Chemother. 2022, 66, e0058422. [Google Scholar] [CrossRef]
  53. Schlagenhauf, P.; Adamcova, M.; Regep, L.; Schaerer, M.T.; Rhein, H.-G. The position of mefloquine as a 21st century malaria chemoprophylaxis. Malar. J. 2010, 9, 357. [Google Scholar] [CrossRef]
  54. González, R.; Pons-Duran, C.; Piqueras, M.; Aponte, J.J.; Ter Kuile, F.O.; Menéndez, C. Mefloquine for preventing malaria in pregnant women. Cochrane Database Syst. Rev. 2015, 3, CD011444. [Google Scholar] [CrossRef]
  55. González, R.; Hellgren, U.; Greenwood, B.; Menéndez, C. Mefloquine safety and tolerability in pregnancy: A systematic literature review. Malar. J. 2014, 13, 75. [Google Scholar] [CrossRef]
  56. Ahmad, S.S.; Rahi, M.; Ranjan, V.; Sharma, A. Mefloquine as a prophylaxis for malaria needs to be revisited. Int. J. Parasitol. Drugs Drug Resist. 2021, 17, 23–26. [Google Scholar] [CrossRef] [PubMed]
  57. Bates, C.; Powers, H.; Lamb, W.; Gelman, W.; Webb, E. Effect of supplementary vitamins and iron on malaria indices in rural Gambian children. Trans. R. Soc. Trop. Med. Hyg. 1987, 81, 286–291. [Google Scholar] [CrossRef] [PubMed]
  58. Dahl, E.L.; Rosenthal, P.J. Apicoplast translation, transcription and genome replication: Targets for anti-malarial antibiotics. Trends Parasitol. 2008, 24, 279–284. [Google Scholar] [CrossRef]
  59. Dahl, E.L.; Rosenthal, P.J. Multiple antibiotics exert delayed effects against the Plasmodium falciparum apicoplast. Antimicrob. Agents Chemother. 2007, 51, 3485–3490. [Google Scholar] [CrossRef] [PubMed]
  60. Krudsood, S.; Silachamroon, U.; Wilairatana, P.; Singhasivanon, P.; Phumratanaprapin, W.; Chalermrut, K.; Phophak, N.; Popa, C. A randomized clinical trial of combinations of artesunate and azithromycin for treatment of uncomplicated Plasmodium falciparum malaria in Thailand. Southeast Asian J. Trop. Med. Public Health 2000, 31, 801–807. [Google Scholar]
  61. Chandra, R.; Ansah, P.; Sagara, I.; Sie, A.; Tiono, A.B.; Djimde, A.A.; Zhao, Q.; Robbins, J.; Penali, L.K.; Ogutu, B. Comparison of azithromycin plus chloroquine versus artemether-lumefantrine for the treatment of uncomplicated Plasmodium falciparum malaria in children in Africa: A randomized, open-label study. Malar. J. 2015, 14, 108. [Google Scholar] [CrossRef]
Figure 1. A flowchart depicting the selection process of the studies included in the meta-analysis. The diagram follows the PRISMA guidelines, illustrating the number of records identified, screened, excluded, and included in the final analysis.
Figure 1. A flowchart depicting the selection process of the studies included in the meta-analysis. The diagram follows the PRISMA guidelines, illustrating the number of records identified, screened, excluded, and included in the final analysis.
Jcm 14 03396 g001
Figure 2. Risk of bias of included RCTs. Symbols: green “+” = positive association; red “−” = negative association; yellow “?” = unclear or insufficient data.
Figure 2. Risk of bias of included RCTs. Symbols: green “+” = positive association; red “−” = negative association; yellow “?” = unclear or insufficient data.
Jcm 14 03396 g002
Figure 3. Incidence of malarial infection in pregnant women with HIV. (A) Network graph showing direct evidence between evaluated interventions. (B) Forest plot comparing all interventions. (C) League table representing network meta-analysis estimates for all interventions’ comparisons.
Figure 3. Incidence of malarial infection in pregnant women with HIV. (A) Network graph showing direct evidence between evaluated interventions. (B) Forest plot comparing all interventions. (C) League table representing network meta-analysis estimates for all interventions’ comparisons.
Jcm 14 03396 g003
Figure 4. Incidence of malarial infection in pregnant women without HIV. (A) Network graph showing direct evidence between evaluated interventions. (B) Forest plot comparing all interventions. (C) League table representing network meta-analysis estimates for all interventions’ comparisons.
Figure 4. Incidence of malarial infection in pregnant women without HIV. (A) Network graph showing direct evidence between evaluated interventions. (B) Forest plot comparing all interventions. (C) League table representing network meta-analysis estimates for all interventions’ comparisons.
Jcm 14 03396 g004
Figure 5. Incidence of abdominal pain in pregnant women without HIV. (A) Network graph showing direct evidence between evaluated interventions. (B) Forest plot comparing all interventions. (C) League table representing network meta-analysis estimates for all interventions’ comparisons.
Figure 5. Incidence of abdominal pain in pregnant women without HIV. (A) Network graph showing direct evidence between evaluated interventions. (B) Forest plot comparing all interventions. (C) League table representing network meta-analysis estimates for all interventions’ comparisons.
Jcm 14 03396 g005
Figure 6. Incidence of dizziness in pregnant women without HIV. (A) Network graph showing direct evidence between evaluated interventions. (B) Forest plot comparing all interventions. (C) League table representing network meta-analysis estimates for all interventions’ comparisons.
Figure 6. Incidence of dizziness in pregnant women without HIV. (A) Network graph showing direct evidence between evaluated interventions. (B) Forest plot comparing all interventions. (C) League table representing network meta-analysis estimates for all interventions’ comparisons.
Jcm 14 03396 g006
Figure 7. Incidence of neonatal deaths in pregnant women without HIV. (A) Network graph showing direct evidence between evaluated interventions. (B) Forest plot comparing all interventions. (C) League table representing network meta-analysis estimates for all interventions’ comparisons.
Figure 7. Incidence of neonatal deaths in pregnant women without HIV. (A) Network graph showing direct evidence between evaluated interventions. (B) Forest plot comparing all interventions. (C) League table representing network meta-analysis estimates for all interventions’ comparisons.
Jcm 14 03396 g007
Table 1. Baseline characteristics of included studies.
Table 1. Baseline characteristics of included studies.
Study IDStudy Arms,
n (%)
SiteStudy DesignMaternal Age, (Mean ± SD)
Year
Gestational Age, (Mean ± SD)
Weeks
Gravida, n (%)Follow-Up Duration (Months)Arm Description
Ahmed et al., 2019 [14]SST DPm, 744 (32.65)IndonesiaRCT (ISRCTN 34010937)27 ± 6.223.9 ± 51. G1, 201 (27)
2. G2, 194 (26.1)
3. ≥G3, 349 (46.9)
Mean (3.1)Single screening and treatment during pregnancy with dihydroartemisinin–piperaquine
IST DPm, 854 (37.47)26.7 ± 6.423.4 ± 4.81. G1, 264 (30.9)
2. G2, 284 (33.3)
3. ≥G3, 306 (35.8)
Intermittent screening and treatment during pregnancy with dihydroartemisinin–piperaquine
DPm, 681 (29.88)26.8 ± 6.123.9 ± 4.61. G1, 202 (29.7)
2. G2, 235 (34.5)
3. ≥G3, 244 (35.8)
Intermittent preventive treatment during pregnancy with dihydroartemisinin–piperaquine
Briand et al., 2009 [15]MQ, 802 (50.01)BeninRCT (NCT00274235)25 ± 5.424 ± 2.8G1, 216 (27)5.52 (SD 0.64)SP (1500 mg of sulfadoxine
and 75 mg of pyrimethamine per dose) plus daily ferrous (400 mg) and folic (5 mg) acid
SP, 799 (49.99)25 ± 5.424 ± 2.8G1, 215 (27)7.59 (SD 0.53)MQ (15 mg/kg per dose) plus daily ferrous (400 mg) and folic (5 mg) acid
Clerk et al., 2008 [16]SP, 693 (36.65)GhanaRCT (NCT00146783)21.1 ± 3.524 ± 3.9G1 or G2At least 1Single dose of SP (1500 mg
sulfadoxine and 75 mg pyrimethamine)
AQ, 503 (26.6)21.6 ± 3.524 ± 4Full treatment course of AQ (25 mg/kg) over 3 days
SPAQ, 695 (36.75)21.4 ± 3.523.9 ± 3.9SPAQ given over 3 days
Cox et al., 2005 [17]VA, 48 (48.98)GhanaRCT
(not registered)
21 ± 2.917 ± 4.3G1, 98 (100)Up to 3.68Capsules were given weekly containing 10,000 IU of vitamin A as retinyl palmitate in groundnut oil, plus tocopherol as preservative.
Control, 50 (50.02)21 ± 2.915 ± 5.6Groundnut oil and tocopherol only in placebo capsules
Darling et al., 2017 [18]VA, 697 (27.79)TanzaniaRCT (NCT0111478)23 ± 510 ± 2.4G1, 321 (46)Up to 102500 IU vitamin A
VA and Zinc, 707 (28.19)22.7 ± 3.710 ± 2.3G1, 346 (49)Both 2500 IU
vitamin A and 25 mg zinc
Zinc, 694 (27.67)23 ± 4.810.1 ± 2.4G1, 333 (48)25 mg zinc (as zinc sulfate)
No Zinc, 710 (28.31)22 ± 7.410 ± 2.3G1, 333 (47)Placebo
Desai et al., 2015 [19]IST DPm, 515 (33.34)KenyaRCT (NCT01669941)23.4 ± 5.922.9 ± 4.71. G1/G2, 267 (51.8)
2. G3+, 248 (48.2)
Up to 9Standard 3-day course
of DP (2, 3, or 4 tablets/day
for 24–35.9, 36–74.9, and ≥75 kg)
DPm, 514 (33.33)23.4 ± 5.523 ± 41. G1/G2, 263 (51.2)
2. G3+, 251 (48.8)
Standard 3-day course
of DP (40 mg/320 mg/tablet)
SP, 514 (33.33)23.5 ± 622.8 ± 4.41. G1/G2, 292 (56.8)
2. G3+, 222 (43.2)
Three tablets of quality-assured SP (500/25 mg/tablet)
Divala et al., 2018 [20]CQ Px, 300 (33.33)MalawiRCT (NCT01443130)20.4 ± 3.622.5 ± 2.2G1 or G2At least 6600 mg of chloroquine at enrolment followed by 300 mg once every week up to delivery
CQ, 300 (33.33)20.7 ± 3.222.2 ± 2.2Two treatments of chloroquine given as 600 mg on day 1, 600 mg on day 2, and 300 mg on day 3 at least four weeks apart during pregnancy
SP, 300 (33.34)20.4 ± 3.122 ± 2.11500 mg sulfadoxine and 75 mg pyrimethamine twice at least four weeks apart during pregnancy
Dolan et al., 1993 [21]Bed nets, 223 (65.4)ThailandRCT
(not registered)
26 ± 6.5NRG1, 40 (17.94)Up to 12Permethrin-impregnated bed net (PIB) or untreated bed
net (NIB)
Control, 118 (34.6)25.49 ± 6.04G1, 28 (23.73)No study bed net or family-sized non impregnated bed net
Filler et al., 2006
(Non-HIV) [22]
SP, 216 (50)MalawiRCT (NCT00126906)19.5 ± 2.622.3 ± 3.6G1, 131 (60.6)At least 6Two-dose SP, with directly observed treatment doses (1500 mg sulfadoxine and 75 mg pyrimethamine)
Two SP doses, 216 (50)19.9 ± 2.421.9 ± 3.7G1, 115 (53.2)Monthly SP, with directly observed treatment doses at enrollment and then monthly until delivery
Filler et al., 2006 (HIV) [22]SP, 135 (50.75)MalawiRCT (NCT00126906)21.6 ± 2.721.9 ± 3.8G1, 59 (43.7)At least 6Two-dose SP, with directly observed treatment doses (1500 mg sulfadoxine and 75 mg pyrimethamine)
Two SP doses, 131 (49.25)21.6 ± 3.822.0 ± 3.8G1, 56 (42.7)Monthly SP, with directly observed treatment doses at enrollment and then monthly until delivery
Gonza’lez et al., 2014 [23]SP, 1576 (33.21)Benin, Gabon, Mozambique, and TanzaniaRCT (NCT00811421)24.8 ± 6.321 ± 71. G1, 460 (29)
2. G1-G3, 778 (49)
3. ≥G4, 338 (21)
At least sixIPTp with SP
MQ, 3169 (66.79)24.6 ± 6.1521 ± 71. G1, 918 (28.97)
2. G1–G3, 1612 (50.87)
3. ≥G4, 639 (20.16)
IPTp with MQ (15 mg/kg) given once as full dose or IPTp with MQ (15 mg/kg) split over two days
Jagannathan et al., 2018 [24]SP, 100 (52.36)UgandaRCT (NCT02163447)21.4 ± 3.639.3 ± 1.81. G1, 35 (35)
2. G2, 33 (33)
3. >G3, 32 (32)
Up to 24Women: IPTp-SP8w;
children: DP every 12 weeks
Bimonthly DP, 44 (23.04)23 ± 4.139.1 ± 2.61. G1, 10 (22.7)
2. G2, 16 (36.4)
3. >G3, 18 (40.9)
Women: IPTp-DP8w; children: DP every 12 weeks
DPm, 47 (24.61)23 ± 3.840 ± 1.21. G1, 10 (21.3)
2. G2, 16 (34)
3. >G3, 21 (44.7)
Women: IPTp-DP8w, children: DP every 4 weeks.
Kayentao et al., 2004 [25]CQ Px, 394 (33.88)MaliRCT
(not registered)
19.4 ± 3.221.6 ± 3.3G1, 234 (59.4)At least 6Weekly CQ chemoprophylaxis (weekly CQ): treatment dose (25 mg/kg CQ base over 3 days) at first ANC visit, followed by weekly prophylaxis (300 mg CQ base per week)
CQ, 380 (32.67)19.1 ± 3.121.5 ± 3.1G1, 243 (64)Two-dose IPT with CQ (IPT/CQ): treatment doses of 25 mg/kg
of CQ base over 3 days at enrollment and again early in third trimester (28–30 weeks gestation)
SP, 389 (33.45)19.3 ± 3.321.8 ± 2.9G1, 244 (62.6)Two-dose IPT with SP (IPT/SP): treatment doses (1500 mg sulfadoxine and 75 mg pyrimethamine)
Kuile et al., 2004 [26]Bed nets, 381 (48.85)KenyaRCT
(not registered)
24.974 ± 7.2324.15 ± 14.661. G1–G4, 234 (61.42)
2. >G4, 147 (28.58)
At least 6Insecticide-treated bed nets
Control, 399 (51.15)26.999 ± 6.6224.4 ± 12.841. G1–G4, 229 (57.39)
2. >G4, 170 (42.61)
No nets
Kumar et al., 2020 [27]Bed nets, 100 (50)PakistanQuasi-experimental study
(not registered)
a. ≤25, 39 (39%)
b. 26–30, 39 (39%)
c. 31 and above, 22 (22%)
NRNRAt least 6Long-lasting insecticide-treated bed nets
Control, 100 (50)a. ≤25, 22 (22%)
b. 26–30, 53 (53%)
c. 31 and above, 25 (25%)
Core health workers
Lingani et al., 2023 [28]SPAZ, 450 (50.11)Burkina
Faso
RCT (PACTR 201808177464681)26 ± 622.4 ± 21. G1, 143 (28.8)
2. G2, 103 (20.8)
3. ≥G3, 250 (50.4)
At least 6Monthly sulfadoxine–pyrimethamine (1500/75 mg) and two grams azithromycin (1 g daily for 2 days) given at second and third trimesters of pregnancy
SP, 448 (49.89)25 ± 622.4 ± 21. G1, 149 (30)
2. G2, 121 (24.4)
3. ≥G3, 226 (45.6)
Monthly sulfadoxine–pyrimethamine (1500/75 mg) (IPTp-SP)
Madanitsa et al., 2023 [29]SP, 1561 (33.35)Tanzania, Kenya, and MalawiRCT (NCT03208179)24·9 ± 6.120.85 ± 3.431. G1, 493 (31·6)
2. G2, 373 (23·9)
3. G3 or more, 692 (44·4)
Median (4.3)Monthly IPTp with sulfadoxine (500 mg)–pyrimethamine (25 mg)
DPm, 1561 (33.35)25.1 ± 6.120.86 ± 3.41. G1, 473 (30·4)
2. G2, 393 (25·2)
3. G3 or more, 691 (44.4)
Monthly IPTp with
dihydroartemisinin (40 mg)–piperaquine (320 mg) plus single treatment course of placebo at enrolment
DPAZ, 1558 (33)24.9 ± 621 ± 3.51. G1, 435 (28)
2. G2, 429 (27·6)
3. G3 or more, 689 (44·4)
Monthly IPTp with dihydroartemisinin–piperaquine combined with single dose of azithromycin
Mlugu et al., 2021 [30]SP, 478 (50)TanzaniaRCT (PACTR 201612001901313)26.6 ± 721 ± 31. G1, 128 (26.8)
2. G2, 105 (22)
3. ≥G3, 245 (51.2)
At least 6Single dose of three tablets, each containing 500 mg sulfadoxine and 25 mg pyrimethamine
DPm, 478 (50)26.8 ± 822 ± 31. G1, 115 (24.1)
2. G2, 108 (22.6)
3. ≥G3, 255 (53.3)
40 mg dihydroartemisinin and 320 mg piperaquine daily for 3 consecutive days
Moore et al., 2019 [31]SP, 58 (48.74)AustraliaRCT
(not registered)
23 ± 3.04125 ± 4.561.67 (SD 1.52)At least 6Single-dose SP (three tablets
of 1500 mg sulfadoxine and 75 mg pyrimethamine
AZ-PQ, 61 (51.24)23 ± 4.5625.33 ± 4.562 (SD 1.52)Three daily doses (at 0, 24, and 48 h) of 1 g AZ (film-coated 500 mg tablets) given with 960 mg PQ tetraphosphate (three 320 mg tablets)
Nambozi et al., 2017 [48]AL, 300 (33.33)ZambiaRCT (NCT00852423)20.67 ± 4.471. 2nd TM, 150 (50%)
2. 3rd TM, 150 (50%)
1. G1, 100 (33.3)
2. G2, 91 (30.3)
3. ≥G3, 109 (36.3)
At least 320 mg artemether and 120 mg lumefantrine per tablet at 4 tablets twice per day over 3 days
MQAS, 300 (33.33)20.33 ± 4.471. 2nd TM, 150 (50%)
2. 3rd TM, 150 (50%)
1. G1, 107 (35.7)
2. G2, 91 (30.3)
3. ≥G3, 102 (34)
100 mg artesunate and 220 mg mefloquine per tablet at 3 tablets once per day over 3 days
DP, 300 (33.33)20.67 ± 4.471. 2nd TM, 131 (43.7%)
2. 3rd TM, 169 (56.3%)
1. G1, 94 (31.3)
2. G2, 96 (32)
3. ≥G3, 110 (36.7)
40 mg dihydroartemisinin
and 320 mg piperaquine phosphate per tablet, 3 tablets once per day over 3 days
Njagi et al., 2003 [32]ITN and SP, 198 (26.33)KenyaRCT
(not registered)
1. G1, 18.4 ± 2.2
2. G2, 21 ± 3.1
1. G1, 20.8 ± 3.5
2. G2, 20.5 ± 3.8
1. G1, 400 (53.19)
2. G2, 352 (46.81)
Rectangular blue or white polyester nets, measuring 190 × 180 × 150 cm dipped into cyfluthrin EW diluted with water to 5 mg/m2 concentration and SP
ITN, 192 (26.06)Rectangular blue or white polyester nets measuring 190 × 180 × 150 cm dipped into cyfluthrin EW diluted with water to 5 mg/m2 concentration
SP, 183 (24.34)Sulfadoxine–pyrimethamine tablets
Control, 175 (23.27)Identical placebo
Nosten et al., 1993 [33]MQ, 171 (50.44)ThailandRCT
(not registered)
26.4 ± 6.224.3 ± 3.33.7 (SD 2.6)Up to 24Mefloquine 500 mg base loading dose followed by 250 mg weekly for 4 weeks and thereafter 125 mg weekly until delivery
Control, 168 (49.56)26.5 ± 6.524.5 ± 3.43.9 (SD 2.8)Identical placebo
Otuli et al., 2020 [34]MQ, 156 (50.49)Republic of CongoRCT
(not registered)
a. ≤18, 39 (12.1%)
b. 19–34, 256 (79.2%)
c. ≥35, 28 (8.7%)
16 to 281. G1, 86 (26.63)
2. G2, 237 (73.37)
At least 61 tablet of 250 mg of mefloquine every 8 h at home and with meal
SP, 153 (49.51)4 doses of 1500 mg sulfadoxine and 75 mg pyrimethamine taken 4 weeks apart
COSMIC Consortium. 2018 [35]AL, 2448 (51.95)Gambia, Benin, and Burkina FasoRCT (NCT01941264)25.17 ± 7.01420.74 ± 3.791. G0, 528 (21.75)
2. G1, 412 (16.97)
3. G2, 373 (15.36)
4. G3, 353 (14.54)
5. ≥G4, 769 (31.67)
At least 6Artemether–lumefantrine
SP, 2264 (48.05)24.893 ± 6.920.77 ± 3.751. G0, 446 (19.7)
2. G1, 412 (18.2)
3. G2, 38,016.78)
4. G3, 348 (15.37)
5. ≥G4, 693 (30.61)
Sulfadoxine–pyrimethamine
Steeke et al., 1996 [36]CQ PxMalawiRCT
(not registered)
NRNRNRAt least 3Chloroquine (CQ) treatment dose of 25 mg of base/kg given as divided dose over two days, followed by 300 mg weekly
CQCQ treatment dose of 25 mg of base/kg given as divided dose over two days and repeated every four weeks
Weekly CQCQ, 300 mg of base weekly
MQMefloquine (MQ) treatment dose of 750 mg as a single dose followed by 250 mg weekly
Browne et al., 2001 [37]Bed nets, 1033 (52.68)GhanaRCT
(not registered)
NR1. 1st TM, 20 (1.9%)
2. 2nd TM, 388 (37.6%)
3. 3rd TM, 625 (60.5%)
1. G1, 204 (19.7)
2. G2, 168 (16.3)
3. ≥G3, 661 (64)
At least 6Insecticide-treated bed nets
Control, 928 (47.32)1. 1st TM, 18 (1.9%)
2. 2nd TM, 350 (37.7%)
3. 3rd TM, 560 (60.4%)
1. G1, 202 (21.8)
2. G2, 162 (17.5)
3. ≥G3, 564 (60.7)
No nets
Kajubi et al., 2017 [38]EFV, 27 (30.68)UgandaRCT (NCT02163447)30 ± 6.2512 to 28NRUp to 9EFV-based ART, standard single-tablet regimen of EFV (600 mg), tenofovir disoproxil fumarate (300 mg), and lamivudine (300 mg) once daily
Control, 31 (35.23)23 ± 3.25DHA–piperaquine, standard dose (3 tablets (40 mg DHA and 320 mg piperaquine) once daily) for 3 consecutive days with or without food
Non pregnant Control, 30 (30.09)24 ± 3.25NANADHA–piperaquine, standard dose (3 tablets (40 mg DHA and 320 mg piperaquine) once daily) for 3 consecutive days with or without food
Roh et al., 2022 [39]LLINs, 4207 (39)MulticenterQuasi-experimental study
(not registered)
24.3 ± 1NR25.8 (SD 10.8)At least 9Long-lasting insecticidal nets
PBO LLINs, 4473 (43.31)24.4 ± 1.923.6 (SD 9.4)Piperonyl butoxide long-lasting insecticidal nets
LLINs + PBO LLINs, 1828 (17.7)23.6 ± 0.632.1 (SD 4.6)Long-lasting insecticidal nets + piperonyl butoxide
Akinyotu et al., 2018 [40]MQ, 64 (48.85)NigeriaRCT (NCT02524444)34.67 ± 4.37At least 16a. <20, 24 (41)
b. 20–24, 30 (56)
c. >24, 10 (53)
At least 4Mefloquine (synthetic 4-quinoline methanol derivative related to quinine) administered in three doses of 250 mg at 4-week intervals
SP, 67 (51.15)32.12 ± 5.66a. <20, 34 (59)
b. 20–24, 24 (44)
c. >24, 9 (47)
Sulfadoxine–pyrimethamine, comprising 500 mg sulfadoxine and 25 mg pyrimethamine, also administered in three doses with 4-week intervals
Akinyotu et al., 2019 [41]SPAZ, 60 (48.78)NigeriaRCT
(not registered)
33.2 ± 4.92a. <20, 19 (31.7%)
b. 20–24, 35 (58.3%)
c. >24, 6 (10%)
1. G0, 6 (10)
2. G1, 21 (35)
3. G2, 27 (45)
4. ≥G3, 6 (10)
At least 6Monthly doses of SP (consisting of three tablets each containing 500 mg/25 mg) administered for 3 months as IPT-p with daily dose of AZ (consisting of one 500 mg tablet) administered for 3 d as IPT-p in HIV-positive pregnant women
SP, 63 (51.22)32.17 ± 5.64a. <20, 32 (50.8%)
b. 20–24, 22 (34.9%)
c. >24, 9 (14.3%)
1. G0, 19 (30.2)
2. G1, 21 (33.3)
3. G2, 14 (22.2)
4. ≥G3, 9 (14.3)
Monthly doses of SP (consisting of three tablets each containing 500 mg/25 mg) administered for 3 months as IPT-p
Barsosio et al., 2024 [42]DPm and CTX, 448 (49.56)MalawiRCT (NCT04158713)29.2 ± 5.622 ± 3.71. G1, 32 (7)
2. G2, 88 (20)
3. ≥G3, 328 (73)
At least 9Daily Co-trimoxazole combined
with monthly IPTp with active dihydroartemisinin–piperaquine
CTX, 456 (50.46)29.2 ± 5.722 ± 3.81. G1, 37 (8)
2. G2, 91 (20)
3. ≥G3, 328 (72)
Co-trimoxazole combined with
monthly identical placebo
Gonza’lez et al., 2014 [43]MQ, 534 (49.86)Kenya, Tanzania, and MozambiqueRCT
(NCT 00811421)
26.8 ± 5.821 ± 81. G1, 57 (11)
2. G1–G3, 341 (64)
3. ≥G4, 136 (25)
At least 9CTX (fixed combination of 800 mg trimethoprim and 160 mg
sulfamethoxazole/tablet) plus IPTp-MQ (250 mg of MQ base/tablet)
Control, 537 (50.14)26.6 ± 5.421 ± 71. G1, 51 (9)
2. G1–G3, 363 (68)
3. ≥G4, 122 (23)
CTX plus IPTp-placebo
(identical to MQ tablets in shape and color)
Manirakiza et al., 2021 [44]CTX, 47.77)Central African RepublicRCT (NCT01746199)27.167 ± 6.7821 ± 4.521G1, 7 (8)At least 6One daily tablet containing 160 mg of trimethoprim and 800 mg of sulfamethoxazole) was administered from 16 weeks until the end of pregnancy
SP, 98 (52.23)29.67 ± 6.78121.67 ± 6.02G1, 9 (9)Three doses of SP-IPTp (1500 mg sulfadoxine and 75 mg pyrimethamine per dose)
given under directly observed administration at one-month intervals from 16 weeks gestation
Ndam et al., 2013 [45]CTX, 152 (48.72)BeninRCT (NCT00970879)At least 1838.3 ± 1.82NRAt least 7CTX at daily dose of 800 mg
sulfamethoxazole and 160 mg trimethoprim
MQ, 160 (51.28)38.46 ± 1.5715 mg/kg MQ (Cipla, Mumbai, India) with rich-fat collation under direct observation
Olofin et al., 2014 [46]No Multivitamins, 522 (24.86)TanzaniaRCT
(not registered)
25.3 ± 4.820.3 ± 3.61. G0, 137 (26.3)
2. G1–3, 299 (57.3)
3. >G3, 86 (16.4)
At least 6Identical placebo
Multivitamins, 528 (25.14)25.4 ± 4.720.4 ± 3.21. G0, 142 (26.9)
2. G1–3, 318 (60.3)
3. >G3, 68 (12.8)
(20 mg vitamin B1, 20 mg B2,
25 mg B6, 100 mg niacin, 50 mg B12, 500 mg C, 30 mg E, and 800 mg folic acid) and Vitamin A
No Vitamin A, 521 (24.81)25.4 ± 4.820.5 ± 3.31. G0, 141 (27.1)
2. G1–3, 306 (58.7)
3. >G3, 74 (14.2)
(20 mg vitamin B1, 20 mg B2,
25 mg B6, 100 mg niacin, 50 mg B12, 500 mg C, 30 mg E, and 800 mg folic acid)
Vitamin A, 529 (25.19)25.3 ± 4.820.3 ± 3.41. G0, 139 (26.2)
2. G1–3, 311 (58.8)
3. >G3, 79 (15)
Vitamin A alone (30 mg
b-carotene with 5000 IU preformed vitamin A)
Natureeba et al., 2017 [47]TMP-SMX, 100 (50)UgandaRCT (NCT02282293)30.3 ± 5.819.2 ± 4.11. G1, 5 (5)
2. G2, 13 (13)
3. ≥G3, 82 (82)
At least 7Daily trimethoprim–sulfamethoxazole (160 mg/800 mg)
TMP-SMX and Monthly DP, 100 (50)29.8 ± 6.819.9 ± 4.51. G1, 13 (13)
2. G2, 12 (12)
3. ≥G3, 75 (75)
Daily trimethoprim–sulfamethoxazole (160 mg/800 mg) and DP (40 mg dihydroartemisinin plus
320 mg piperaquine
Abbreviations: RCT = randomized controlled trial; SD = standard deviation; NR = not reported; NA = not applicable; SST = single screening and treatment; IST = intermittent screening and treatment; IPTp = intermittent preventive treatment during pregnancy; DPm = monthly dihydroartemisinin; MQ = mefloquine; SP = sulfadoxine and pyrimethamine; AQ = amodiaquine; SPAQ = sulfadoxine and pyrimethamine plus amodiaquine; VA = vitamin A; CQ Px = prophylactic chloroquine; SPAZ = sulfadoxine and pyrimethamine plus Azithromycin; DPAZ = dihydroartemisinin and azithromycin; AZ-PQ = azithromycin and piperaquine; MQAS = mefloquine–artesunate; ITN = insecticide-treated net; AL = artemether–lumefantrine; EFV = efavirenz; LLINs = long-lasting insecticide-treated bed nets; CTX = Co-trimoxazole; TMP-SMX = trimethoprim-sulfa-methoxasole.
Table 2. Summary of included studies.
Table 2. Summary of included studies.
Study IDDiagnostic ToolsInclusion CriteriaPrimary EndpointsConclusion
Ahmed et al., 2019 [14]Quantitative
PCR [qPCR], nested PCR, and loop-mediated isothermal
amplification [LAMP] (Eiken Chemical Company, Japan)
1. Between 16 May 2013, and
21 April 2016
2. Pregnant women of any gravity
3. Viable pregnancy between 16 and 30 weeks gestation
4. Had given written informed consent
1. Malaria infections
2. Adverse events
“IST was associated with a lower prevalence of malaria than SST at delivery, but the prevalence of malaria in this group was also lower at enrolment, interpreting the effect of IST as challenging. Further studies with highly sensitive malaria rapid diagnostic tests should be considered. Monthly IPT with dihydroartemisinin–piperaquine is a promising alternative to SST in areas in the Asia-Pacific region with moderate or high malaria transmission”.
Briand et al., 2009 [15]Thick and thin blood smears stained with Giemsa stain1. In Benin from July 2005
through April 2008
2. Women of all gravidities of
16–28 weeks gestation
3. No history of neurologic or psychiatric disorder
4. Followed for at least 5.27 months
1. Malaria infections
2. LBW
3. Safety and adverse events
“MQ proved to be highly efficacious—clinically and parasitologically—for use as IPTp. However, its low tolerability might impair its effectiveness and requires further investigations”.
Clerk et al., 2008 [16]Thick blood
film
1. From June 2004 to
February 2007
2. Highly endemic area of malaria
3. Availability for follow-up during pregnancy
4. Willingness to comply with study procedures
1. Malarial infections
2. LBW
3. Safety and adverse events
“The effects of IPTp with AQ or SPAQ on maternal anaemia and LBW were comparable to the effects of IPTp with SP; however, IPTp regimens that contain AQ are unlikely to be useful as an alternative to IPTp with SP in Ghana because of a high frequency of associated adverse events”.
Cox et al., 2005 [17]Microscopic
examination of Giemsa-stained thick blood films
1. From March to June 2001
2. Primigravid pregnant women
3. Resident within study area
4. In good health and less than 24 weeks pregnant
5. Followed up for maximum 16 weeks
1. Malarial infections
2. LBW
“The data suggest that the reduction in the levels of anti-VSACSA antibodies to the known placental malaria isolate may reflect reduced intensity or duration of placental parasitemia in women receiving vitamin A supplementation. These observations are of potential public health significance and deserve further investigation”.
Darling et al., 2017 [18]Histopathology and polymerase chain reaction (PCR)1. Participants in
first trimester of pregnancy
2. Primigravida or secundigravida
3. Human immunodeficiency
virus (HIV)-negative
4. Intending to stay in Dar es Salaam for at least 6 weeks
after delivery
5. Followed up for at least
10 months
1. Malarial infections
2. LBW
3. Safety and adverse events
“No safety concerns were identified. We recommend additional studies in other geographic locations to confirm these findings”.
Desai et al., 2015 [19]PCR1. HIV-negative pregnant women
2. Between 16 and 32 weeks gestation
3. Viable pregnancy
4. No history of receiving IPTp-SP during pregnancy
Malarial infections“At the current levels of RDT sensitivity, ISTp is not a suitable alternative to IPTp-SP in the context of high SP resistance and malaria transmission. However, DP is a promising alternative drug to replace SP for IPTp. The efficacy, operational feasibility, and cost-effectiveness of IPTp-DP should be investigated further”.
Divala et al., 2018 [20]Histopathology, molecular results, or PCR1. Pregnant women in their first
or second pregnancy
2. Before 27th week
of gestation
3. Not yet taken routine SP IPTp
4. Hoped to remain in area until 14 weeks after delivery
1. Malaria infection
2. Maternal anemia
“Chloroquine administered as IPTp did not provide better protection from malaria and related adverse effects than SP-IPTp in this setting of high SP-resistance. Protocol-specified adjusted analyses suggest that chloroquine chemoprophylaxis may provide benefit in protecting against malaria during pregnancy”.
Dolan et al., 1993 [21]Blood taken by finger-prick for thick and thin filmsPregnant women given either permethrin-impregnated bed net (PIB),
an untreated bed net (NIB), or
no study bed net
1. Malaria infection
2. LBW
“PIB or FNIB reduce the adverse effects of malaria in pregnancy on the mother, and may also reduce subsequent infant morbidity and mortality”.
Filler et al., 2006
(Non-HIV) [22]
Thick blood smears stained
with Giemsa
1. Clinic patients seeking ANC
2. Women in their first and
second pregnancies
3. Between 16 and 28 weeks
of gestation
4. Had given informed consent
1. Malaria infection
2. Safety and adverse events
“In HIV-positive pregnant women, monthly SP IPTp is more efficacious than a 2-dose regimen in preventing placental malaria. The study also demonstrates the continued efficacy of SP for the prevention of placental malaria, even in the face of its decreasing efficacy for the treatment of malaria in children. In areas with intense transmission of falciparum malaria and a high prevalence of HIV infection, monthly SP IPTp should be adopted”.
Filler et al., 2006 (HIV) [22]Thick blood smears stained
with Giemsa
1. Clinic patients seeking ANC
2. Women in their first and
second pregnancies
3. Between 16 and 28 weeks
of gestation
4. Had given informed consent
1. Malaria infection
2. Safety and adverse events
“In HIV-positive pregnant women, monthly SP IPTp is more efficacious than a 2-dose regimen in preventing placental malaria. The study also demonstrates the continued efficacy of SP for the prevention of placental malaria, even in the face of its decreasing efficacy for the treatment of malaria in children. In areas with intense transmission of falciparum malaria and a high prevalence of HIV infection, monthly SP IPTp should be adopted”.
Gonza’lez et al., 2014 [23]Thick and thin blood
films stained
1. Pregnant women of all gravidities attending ANC
clinic for first time
2. Had not received IPTp during current pregnancy
3. Gestational age < 28 weeks
4. Negative HIV testing
at recruitment
5. Absence of history of allergy
to sulfa drugs or MQ
1. Malaria infection
2. LBW
3. Safety and adverse drug reactions
“Women taking MQ IPTp (15 mg/kg) in the context of long-lasting insecticide-treated nets had similar prevalence rates of low birth weight as those taking SP IPTp. MQ recipients had less clinical malaria than SP recipients, and the pregnancy outcomes and safety profile were similar. MQ had poorer tolerability even when splitting the dose over two days. These results do not support a change in the current IPTp policy”.
Jagannathan et al., 2018 [24]Blood collected for thick blood smear1. From June 2014 through
May 2017
2. Area of historically high malaria transmission intensity
3. Pregnant women of at least
16 weeks gestation
4. Negative HIV testing at recruitment
1. Malaria infection
2. Maternal anemia
3. Safety and adverse drug reactions
“Contrary to our hypothesis, preventing malaria in pregnancy with IPTp-DP in the context of chemoprevention with DP during infancy does not lead to a reduced incidence of malaria in childhood; in this setting, it may be associated with an increased incidence of malaria in females. Future studies are needed to better understand the biological mechanisms of in utero drug exposure on drug metabolism and how this may affect the dosing of antimalarial drugs for treatment and prevention during infancy”.
Kayentao et al., 2004 [25]Thick blood films stained with Giemsa1. Women with first or
second pregnancy
and between 16 and 26 weeks
of gestation
2. Had given written informed consent
Malaria infection“In Mali, IPT with SP appears more efficacious than weekly chloroquine chemoprophylaxis in preventing malaria during pregnancy. These data support World Health Organization recommendations to administer at least 2 doses of IPT during pregnancy. In intensely seasonal transmission settings in Mali, 12 doses may be required to prevent placental reinfection prior to delivery”.
Kuile et al., 2004 [26]Malaria thick and thin blood smears1. Pregnant women who
had parasitemia
2. Documented fever or
patients with history of fever
within previous 48 h treated with SP
3. At least 16 weeks gestation
4. Negative HIV testing at recruitment
1. Malaria infection
2. Maternal anemia
3. Safety and adverse events
“In areas of intense perennial malaria transmission, permethrin-treated bed nets reduce the adverse effect of malaria during the first four pregnancies”.
Kumar et al., 2020 [27]NRPregnant women
given either permethrin-impregnated bed net (PIB), untreated bed net (NIB), or no study bed net
Incidence of malaria infections“Results proved that health education could be an effective intervention for improving knowledge and usage of LLINs among pregnant women for the prevention of malaria. Such educational interventions have a positive potential to be implemented at a larger scale by incorporating them into routine health sessions provided by health
workers”.
Lingani et al., 2023 [28]Thick and thin blood smears stained with 5% Giemsa for 30 min1. Age of 16–35 years
2. A gestational age of
12–24 weeks
3. Negative HIV-testing
at recruitment
4. Willingness to adhere to study protocol and signed informed consent
1. Malaria infection
2. LBW
3. Safety and adverse events
“Adequate prevention regimen with monthly sulfadoxine-pyrimethamine given to all pregnant women has been proven to reduce the risk of LBW in malaria-endemic areas. Adding azithromycin to the regimen does not offer further benefits, as long as women receive a malaria prevention regimen early enough during pregnancy”.
Madanitsa et al., 2023 [29]Malaria microscopy, quantitative PCR (qPCR), and targeted next-generation sequencing for molecular markers1. From 29 March 2018 to
5 July 2019
2. Women of any age with
viable singleton pregnancy
3. Between 16 weeks and 28 weeks gestation confirmed by the US
4. Willingness to adhere to study protocol and signed informed consent
1. Malaria infection
2. LBW
3. Safety and adverse events
“Monthly IPTp with dihydroartemisinin–piperaquine did not improve pregnancy outcomes, and the addition of a single course of azithromycin did not enhance the effect of monthly IPTp with dihydroartemisinin–piperaquine. Trials that combine sulfadoxine-pyrimethamine and dihydroartemisinin–piperaquine for IPTp should
be considered”.
Mlugu et al., 2021 [30]RDT, microscopy, and PCR1. HIV-negative, age 16 years
or older
2. Malaria-negative (RDT)
3. Gestational age of ≥13 weeks
4. Willing and able to give informed consent
1. Malaria infection
2. LBW
3. Safety and adverse events
“However, the prevalence of LBW (4.6% versus 9.6%, p = 0.003) was significantly lower in IPTp-DHP compared to IPTp-SP. We report superior protective efficacy of monthly IPTp-DHP against malaria in pregnancy and LBW than IPTp-SP”.
Moore et al., 2019 [31]Thick/thin blood smears prepared for microscopy1. Pregnant women between
14 and 32 weeks of gestation
2. Not taken any study drugs in previous 28 days
3. No history of allergy to study drugs
4. Willing and able to give informed consent
1. Malaria infection
2. Maternal anemia
3. Safety and adverse events
“Further assessment of AZ-PQ (including alternative total dosing of AZ, with a focus on tolerability) should be undertaken in a variety of settings in which malaria is endemic, to ensure that this therapy would be accepted as an alternative to SP”.
Nambozi et al., 2017 [48]Giemsa-stained thick and thin blood filmsPregnant women with 20 weeks median gestational ageMalaria infection“As new infections can be prevented by a long-acting partner drug to the artemisinins, DHAPQ should be preferred in places such as Nchelenge district where transmission is intense while in areas of low transmission intensity AL or MQAS may be used”.
Njagi et al., 2003 [32]Giemsa-stained thick and thin blood films1. Pregnant women estimated
at gestational age between 12 and 24 weeks
2. Had given informed consent
Malaria infection“It was concluded that malaria is a major cause of anemia in primigravidae but that other causes play a more significant role in secundigravidae, and that intermittent treatment with SP or use of ITNs benefits primigravidae more than secundigravidae”.
Nosten et al., 1993 [33]Thick blood film stained with
Giemsa stain and examined
Pregnant women of at least
20 weeks gestation
1. Malaria infection
2. Maternal anemia
3. Safety and adverse events
“Mefloquine is safe and effective for antimalarial prophylaxis in the second half of pregnancy”.
Otuli et al., 2020 [34]Finger-prick performed on finger pulp and 5 μL blood dropped and spread on slide1. From 15 May to
30 November 2019
2. High rate of pregnant
women attending ANC
3. Between 16 and 28 gestational weeks
4. Not taking IPTp during current pregnancy
5. Willing and able to give informed consent
1. Malaria infection
2. Safety and adverse events
“Splitting dose and intake with a meal increased mefloquine tolerability while keeping its efficacy higher compared to sulfadoxine–pyrimethamine. Intermittent preventive treatment during pregnancy using mefloquine reduces the risk of placental malaria, maternal peripheral parasitemia, and low birth weight, compared to sulfadoxine–pyrimethamine. Thus, mefloquine is a good alternative to intermittent preventive treatment in pregnancy”.
COSMIC Consortium. 2018 [35]Giemsa-stained thick blood films1. Pregnant women in second or third trimester
2. Attending first ANC
3. Willing and able to give informed consent
1. Malaria infection
2. Maternal anemia
3. Safety and adverse events
“Adding CSST to existing IPTp-SP strategies did not reduce malaria in pregnancy. Increasing the number of IPTp-SP doses given during pregnancy is a priority”.
Steeke et al., 1996 [36]Thick blood smears,
hematocrit, and serum for anti-malarial antibody testing
1. Consecutive women of
any parity
2. During 1987–1990
3. Pregnant women in area
of high malaria endemicity
4. Willing and able to give informed consent
1. Malaria infection
2. LBW
“When evaluating antenatal care programs, health policymakers must consider providing an effective preventive drug (either MQ or other drugs identified in additional studies, e.g., sulfa-pyrimeth-amine compounds) to prevent low birth weight and its consequences”.
Browne et al., 2001 [37]Giemsa-stained thick and thin blood films1. Pregnant women
2. Special focus on primigravidae and secundigravidae
3. Willing and able to give informed consent
1. Malaria infection
2. LBW
3. Maternal anemia
“Chloroquine use in pregnancy was low and comparable in both groups. Implications of
findings for malaria control in pregnancy and further research are discussed”.
Kajubi et al., 2017 [38]Giemsa-stained thick and thin blood films1. HIV-negative pregnant women
2. HIV-positive pregnant women on EFV-based ART
3. HIV-negative non-pregnant women
4. Between 12 and 28 weeks gestation
5. Willing and able to give informed consent
Plasma concentration–time profile“Exposure to DHA and piperaquine were lower among pregnant women and particularly in women on efavirenz, suggesting a need for dose modifications. The study of modified dosing strategies for these populations is urgently needed”.
Roh et al., 2022 [39]NR1. Between March 2017 and
March 2018
2. Health facilities from each HSD
3. Government-operated
4. Included maternity ward
5. Located >5 km from neighboring HSD
6. had a mean delivery rate of >200/year
1. LBW incidence
2. Stillbirth incidence
“In this region of Uganda, where pyrethroid resistance is high, this study found that a mass LLIN campaign was associated with reduced stillbirth incidence. Effects of the campaign were greatest for women who would”.
Akinyotu et al., 2018 [40]Thick and thin blood smears1. Between 1 January and
31 August 2016
2. HIV-positive pregnant women
3. At least 16 weeks of gestation
4. Followed up for at least
four months
Malaria infection“Outcomes following prophylactic use of mefloquine for intermittent preventive therapy for malaria among pregnant women with HIV were comparable to sulphadoxine–pyrimethamine treatment; mefloquine is a feasible alternative therapy”.
Akinyotu et al., 2019 [41]Thick and thin blood smears1. Between 1 January and
31 August 2016
2. HIV-positive patients
3. At least 16 weeks of gestation
4. No history of AZ or SP use for 4 weeks prior to recruitment
1. Malaria infections
2. LBW
3. Safety and adverse events
“The findings suggest that AZ is comparable to SP in malaria prevention and safety in HIV-positive pregnant women”.
Barsosio et al., 2024 [42]Microscopy, PCR, and blood tests1. From 11 November 2019 to
3 August 2021
2. HIV-positive pregnant patients
3. Between 16 weeks and 28 weeks gestation
4. Willingness to give informed consent
1. Malaria infections
2. LBW
3. Safety and adverse events
“Addition of monthly intermittent preventive treatment with dihydroartemisinin–piperaquine to the standard of care with daily unsupervised co-trimoxazole in areas of high antifolate resistance substantially improves malaria chemoprevention in pregnant women living with HIV on dolutegravir-based cART and should be considered for policy”.
Gonza’lez et al., 2014 [43]Microscopy on Giemsa-stained blood films1. From March 2010 to April 2012
2. HIV-positive pregnant patients
3. Gestational age ≤ 28 weeks
4. Absence of history of allergy to sulfa drugs or MQ
5. Willingness to give informed consent
1. Malaria infections
2. Maternal anemia
3. Safety and adverse events
“An effective antimalarial added to CTXp and LLITNs in HIV-infected pregnant women can improve malaria prevention, as well as maternal health through reduction in hospital admissions. However, MQ was not well tolerated, limiting its potential for IPTp and indicating the need to find alternatives with better tolerability to reduce malaria in this particularly vulnerable group. MQ was associated with an increased risk of mother-to-child transmission of HIV, which warrants a better understanding of the pharmacological interactions between antimalarials and antiretroviral drugs”.
Manirakiza et al., 2021 [44]PCR1. HIV-positive pregnant patients
2. Between 16 and 28 weeks of gestation
3. CD4+ count ≥ 350 cells/mm3
4. Willingness to give informed consent
1. Malaria infections
2. Safety and adverse events
“Although our results do not allow us to conclude that CTX is more effective, drug safety and good compliance among women with this treatment favor its widespread use among HIV-infected pregnant women, as currently recommended by WHO”.
Ndam et al., 2013 [45]Blood samples collected in EDTA tubes then PCR1. HIV-positive pregnant women
2. Between 16 and 28 weeks
of gestation
3. Willingness to give informed consent
1. Malaria infections
2. Maternal anemia
“CTX alone provided adequate protection against malaria in HIV-infected pregnant women, although MQ-IPTp showed higher efficacy against placental infection. Although more frequently associated with dizziness and vomiting, MQ-IPTp may be an effective alternative given concerns about parasite resistance to CTX”.
Olofin et al., 2014 [46]Thick and thin films stained with 5%
Giemsa solution and examined
1. From April 1995 until
August 2003
2. HIV-positive pregnant patients
3. Between 16 and 28 weeks
of gestation
4. Willingness to give informed consent
1. Malaria infections
2. Maternal anemia
3. Safety and adverse events
“Multivitamin supplements have been previously shown to reduce HIV disease progression among HIV-infected women, and consistent with that, these supplements protected against development of symptomatic malaria. The clinical significance of the increased risk of malaria parasitemia among supplemented women deserves further research, however. Preventive measures for malaria are warranted as part of an integrated approach to the care of HIV-infected individuals exposed to malaria”.
Natureeba et al., 2017 [47]Blood smears stained with 2% Giemsa and read by experienced laboratory technologist1. Women ≥16 years of age
2. Positive for HIV-1
3. Between 16 and 28 weeks of gestation
4. Willingness to give informed consent
1. Malaria infections
2. Safety and adverse events
“Among HIV-infected pregnant women in the setting of indoor residual spraying of insecticide, adding monthly DP to daily TMP-SMX did not reduce the risk of placental or maternal malaria or improve birth outcomes”.
Abbreviations: RCT = randomized controlled trial; SD = standard deviation; NR = not reported; NA = not applicable; SST = single screening and treatment; IST = intermittent screening and treatment; IPTp = intermittent preventive treatment during pregnancy; DPm = monthly dihydroartemisinin; MQ = mefloquine; SP = sulfadoxine and pyrimethamine; AQ = amodiaquine; SPAQ = sulfadoxine and pyrimethamine plus amodiaquine; VA = vitamin A; CQ Px = prophylactic chloroquine; SPAZ = sulfadoxine and pyrimethamine plus azithromycin; DPAZ = dihydroartemisinin and azithromycin; AZ-PQ = azithromycin and piperaquine; MQAS = mefloquine–artesunate; ITN = insecticide-treated net; AL = artemether–lumefantrine; EFV = efavirenz; LLINs = long-lasting insecticide-treated bed nets; CTX = Co-trimoxazole; TMP-SMX = trimethoprim-sulfa-methoxasole.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Albadrani, M.; Eltahir, H.M.; Mahmoud, A.B.; Abouzied, M.M. Evaluating the Safety and Efficacy of Malaria Preventive Measures in Pregnant Women with a Focus on HIV Status: A Systematic Review and Network Meta-Analysis. J. Clin. Med. 2025, 14, 3396. https://doi.org/10.3390/jcm14103396

AMA Style

Albadrani M, Eltahir HM, Mahmoud AB, Abouzied MM. Evaluating the Safety and Efficacy of Malaria Preventive Measures in Pregnant Women with a Focus on HIV Status: A Systematic Review and Network Meta-Analysis. Journal of Clinical Medicine. 2025; 14(10):3396. https://doi.org/10.3390/jcm14103396

Chicago/Turabian Style

Albadrani, Muayad, Heba M. Eltahir, Ahmad Bakur Mahmoud, and Mekky M. Abouzied. 2025. "Evaluating the Safety and Efficacy of Malaria Preventive Measures in Pregnant Women with a Focus on HIV Status: A Systematic Review and Network Meta-Analysis" Journal of Clinical Medicine 14, no. 10: 3396. https://doi.org/10.3390/jcm14103396

APA Style

Albadrani, M., Eltahir, H. M., Mahmoud, A. B., & Abouzied, M. M. (2025). Evaluating the Safety and Efficacy of Malaria Preventive Measures in Pregnant Women with a Focus on HIV Status: A Systematic Review and Network Meta-Analysis. Journal of Clinical Medicine, 14(10), 3396. https://doi.org/10.3390/jcm14103396

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop