Abstract

Background

Seasonal malaria chemoprevention (SMC) is a novel strategy to reduce malaria infections in children. Infection with Plasmodium falciparum results in immune dysfunction characterized by elevated expression of markers associated with exhaustion, such as PD1 and LAG3, and regulatory CD4+FOXP3+ T cells.

Methods

In the current study, the impact of seasonal malaria chemoprevention on malaria-induced immune dysfunction, as measured by markers associated with exhaustion and regulatory T cells, was explored by flow cytometry.

Results

Children that received seasonal malaria chemoprevention had fewer malaria episodes and showed significantly lower fold changes in CD4+PD1+ and CD4+PD1+LAG3+ compared to those that did not receive SMC. Seasonal malaria chemoprevention had no observable effect on fold changes in CD8 T cells expressing PD1 or CD160. However, children receiving SMC showed greater increases in CD4+FOXP3+ T regulatory cells compared to children not receiving SMC.

Conclusions

These results provide important insights into the dynamics of malaria-induced changes in the CD4 T-cell compartment of the immune system and suggest that the reduction of infections due to seasonal malaria chemoprevention may also prevent immune dysfunction.

Clinical Trials Registration

NCT02504918.

Malaria caused by Plasmodium falciparum continues to threaten global health with devastating consequences. Recent statistics from the World Health Organization (WHO) estimate that there were over 219 million malaria infections, resulting in 435 000 deaths, in 2017 [1], indicating a stall in progress in malaria reduction over the last 3 years. Over 90% of all malaria-related deaths occurred in sub-Saharan Africa.

In the absence of an efficacious and long-lasting malaria vaccine, other strategies to reduce malaria morbidity and mortality have been introduced. These include distribution of bednets, indoor residual spraying, and targeted interventions in the most vulnerable groups such as intermittent preventative treatment for pregnant mothers and seasonal malaria chemoprevention (SMC) for children under the age of 5 years [2]. SMC has been shown to effectively reduce infections as well as clinical malaria cases in the Sahel region of Africa [3], where the malaria transmission season coincides with the commencement of the rainy season. The WHO-recommended SMC regimen consists of monthly dosing with amodiaquine and sulfadoxine-pyrimethamine for at least 4 months during the transmission season [2]. In 2017, 15.7 million children in 12 countries in the African Sahel received SMC, although an additional 13.6 million children could have benefited if additional funding had been available. Mali was one of the first countries in the Sahel region to implement SMC in 2012, which has now been gradually rolled out to cover the entire country.

Sterilizing immunity against P. falciparum infection does not develop even against repeated infections [4]; instead, there appears to be gradual acquisition of clinical immunity. In malaria-endemic regions, children under the age of 5 years experience the highest burden of the most severe forms of the disease (manifested as severe malarial anemia and cerebral malaria), while the prevalence of asymptomatic P. falciparum infections is greater in older children and adults [5, 6]. While the markers of clinical immunity remain poorly defined, they have been associated with reduced inflammatory responses and acquisition of strain-specific immunity to blood-stage P. falciparum antigens [7–10].

Malaria infection leads to increases in CD4 T cells expressing the immune exhaustion markers such as programmed cell death protein-1 (PD1) and lymphocyte activation gene-3 (LAG3) on T and B cells [11–13]. In animal models, blockade of PD1, as well as LAG3, results in clearance of plasmodium parasites [11, 14], suggesting that these markers hinder an effective immune response. Similarly, malaria infections have been associated with altered T regulatory cells (Tregs). While there are many different types of Tregs, forkhead box P3 (FOXP3)-expressing Tregs have been the most widely studied in the context of malaria [15, 16]. Treg responses are variable in the literature; some studies have shown increases in Treg numbers after infections, others have highlighted a trend of reduced risk of symptomatic malaria in children living in a high transmission area [15–17]. In animal models, expansion of Tregs has been shown to limit the early immune effector response to malaria, thereby assisting the pathogen to establish infection [18]. Together, these studies suggest that malaria infection leads to generalized immune dysfunction that may have consequences for subsequent responses to malaria, other diseases, and vaccines.

The effect of SMC on clinical immunity to malaria (and on the immune system in general) has not been extensively studied. One study in Mali showed decreases in antibodies to blood-stage antigens in children receiving SMC [19], while another study in Senegal showed no difference in antibody titers to P. falciparum antigens between children receiving SMC versus controls [17].

In the current study, the impact of SMC treatment over a whole malaria transmission season on the T-cell compartment was assessed. More specifically, Tregs and immune exhaustion markers were compared in children residing in neighboring villages in Mali: Beneko (SMC+) where SMC was being implemented and Ferekoroba (SMC−) where SMC had yet to be introduced. Malaria infections were dramatically reduced in children enrolled from Beneko compared to Ferekoroba. Consequently, the fold change in CD4+PD1+ or CD4+PD1+LAG3+ was significantly lower in the SMC+ group than the SMC− group over the course of transmission season, whereas Tregs were increased in the SMC+ group. These results highlight the beneficial impact of SMC in reducing malaria-induced immune dysfunction.

METHODS

Ethics

The study protocol was approved by Ethics committees in Mali (FMPOS) and the United States (ClinicalTrials.gov identifier: NCT02504918). The National Institute of Allergy and Infectious Diseases Institutional Review Board/Ethics Committee approved the study protocol. Written informed consent was obtained from the parents/guardians of the children as required.

Study Site

The study was conducted in 2 neighboring villages, Beneko and Ferekoroba, in the health district center of Ouelessebougou in Mali. In 2008, the incidence rate of clinical malaria in children under the age of 5 years was 1.99 episodes/child/year in Ouelessebougou and the incidence of severe malaria (as defined by WHO criteria [20]) was between 1% and 2% during the transmission season [3]. During the study period, SMC was being implemented in Beneko but not in Ferekoroba (where SMC was rolled out in 2016 after completion of the study).

Study Population

Children between the ages of 12 and 59 months from Beneko or Ferekoroba were included in the study. Children were clinically evaluated and those with significant anemia, unable to give blood, with chronic or debilitating conditions, immunosuppression, or chronic infection, or on co-trimoxazole were excluded from the study. Characteristics of study subjects are shown in Table 1.

Table 1.

Characteristics of Study Subjects

CharacteristicsSMC+SMC−
Sex, n (%)
 Male27 (54.0)25 (50.0)
 Female23 (46.0)25 (50.0)
Age, mo, mean (SD)33.2 (11.1)33.4 (13.5)
Hb type, n (%)
 AA40 (80.0)34 (68.0)
 AC8 (16.0)5 (10.0)
 AS1 (2.0)9 (18.0)
 CC0 (0.0)1 (2.0)
 SC1 (2.0)1 (2.0)
Blood type, n (%)
 A21 (42.0)5 (10.0)
 AB1 (2.0)2 (4.0)
 B15 (30.0)15 (30.0)
 O13 (26.0)28 (56.0)
Infections >28 days apart, No.6397
Gametocytemia, mean (SD)10.18 (13.88)31.14 (82.62)
Parasitemia, mean (SD)1030.0 (1263)928.3 (1499)
CharacteristicsSMC+SMC−
Sex, n (%)
 Male27 (54.0)25 (50.0)
 Female23 (46.0)25 (50.0)
Age, mo, mean (SD)33.2 (11.1)33.4 (13.5)
Hb type, n (%)
 AA40 (80.0)34 (68.0)
 AC8 (16.0)5 (10.0)
 AS1 (2.0)9 (18.0)
 CC0 (0.0)1 (2.0)
 SC1 (2.0)1 (2.0)
Blood type, n (%)
 A21 (42.0)5 (10.0)
 AB1 (2.0)2 (4.0)
 B15 (30.0)15 (30.0)
 O13 (26.0)28 (56.0)
Infections >28 days apart, No.6397
Gametocytemia, mean (SD)10.18 (13.88)31.14 (82.62)
Parasitemia, mean (SD)1030.0 (1263)928.3 (1499)

Unit for Gametocytemia: /500 WBC; Unit for Parasitemia: /300 WBC.

Abbreviations: Hb, hemoglobin; SMC, seasonal malaria chemoprevention.

Table 1.

Characteristics of Study Subjects

CharacteristicsSMC+SMC−
Sex, n (%)
 Male27 (54.0)25 (50.0)
 Female23 (46.0)25 (50.0)
Age, mo, mean (SD)33.2 (11.1)33.4 (13.5)
Hb type, n (%)
 AA40 (80.0)34 (68.0)
 AC8 (16.0)5 (10.0)
 AS1 (2.0)9 (18.0)
 CC0 (0.0)1 (2.0)
 SC1 (2.0)1 (2.0)
Blood type, n (%)
 A21 (42.0)5 (10.0)
 AB1 (2.0)2 (4.0)
 B15 (30.0)15 (30.0)
 O13 (26.0)28 (56.0)
Infections >28 days apart, No.6397
Gametocytemia, mean (SD)10.18 (13.88)31.14 (82.62)
Parasitemia, mean (SD)1030.0 (1263)928.3 (1499)
CharacteristicsSMC+SMC−
Sex, n (%)
 Male27 (54.0)25 (50.0)
 Female23 (46.0)25 (50.0)
Age, mo, mean (SD)33.2 (11.1)33.4 (13.5)
Hb type, n (%)
 AA40 (80.0)34 (68.0)
 AC8 (16.0)5 (10.0)
 AS1 (2.0)9 (18.0)
 CC0 (0.0)1 (2.0)
 SC1 (2.0)1 (2.0)
Blood type, n (%)
 A21 (42.0)5 (10.0)
 AB1 (2.0)2 (4.0)
 B15 (30.0)15 (30.0)
 O13 (26.0)28 (56.0)
Infections >28 days apart, No.6397
Gametocytemia, mean (SD)10.18 (13.88)31.14 (82.62)
Parasitemia, mean (SD)1030.0 (1263)928.3 (1499)

Unit for Gametocytemia: /500 WBC; Unit for Parasitemia: /300 WBC.

Abbreviations: Hb, hemoglobin; SMC, seasonal malaria chemoprevention.

Study Visits, Administration of SMC, and Sample Collection

The study was conducted over a 5-month period starting in August 2015 (month 0) until December 2015 (month 4). Children recruited to the study were clinically examined at each study visit. Blood smears were prepared and used to determine malaria infections. Sulfadoxine-pyrimethamine plus amodiaquine were administered monthly from August to November 2015 as per the guidelines of the WHO [2]. Children with symptomatic malaria were treated with artemether-lumefantrine and resumed SMC treatment at their next monthly visit. The participants were also seen on unscheduled visits at any point during the study.

During each scheduled and unscheduled visit, a 5 mL venous blood sample was collected from study participants. An aliquot of this sample was used for ex vivo flow cytometry, preparation of blood smears, and for hemoglobin (Hb) measurements. The remaining blood was used for peripheral blood mononuclear cell isolation.

Laboratory Evaluation

Malaria parasitemia and gametocytemia was determined in Giemsa-stained blood smears and reported against 300 and 500 white blood cells (WBCs), respectively, by expert microscopists. When prompt blood smear microscopy was not available, such as during SMC delivery, a rapid diagnostic test was used for malaria diagnosis in order to allow prompt treatment. All diagnoses were later confirmed by blood smear microscopy, and only blood smear-positive results were considered as patent parasitemia for analysis purposes.

Flow Cytometry

Whole blood (150 µL) was used to stain for surface markers using conjugated monoclonal antibodies against anti-CD160-PE, anti-CD4-PE-CY5, anti-PD1-APC, anti-CD3-Alexa-700, anti-CD8- APC-CY7, and anti-LAG3-Pacific Blue. Cells were incubated for 20 minutes, washed with phosphate-buffered saline, fixed, permeabilized, and then stained with anti-FOXP3-FITC. All samples were acquired on a BD LSR II flow cytometer and analyzed using FlowJo software (version 10.1; Figure 1). The data from flow cytometry were expressed as fold change from the baseline (day 0).

Gating strategy used to identify CD4+PD1+, CD4+PD1+LAG3+, CD4+FOXP3+, CD8+PD1+, and CD8+CD160+ populations within the CD3+ gate. Abbreviations: FSC, forward scatter; FOXP3, forkhead box P3; LAG3, lymphocyte activation gene-3; PD1, programmed cell death protein-1; SSC, side scatter.
Figure 1.

Gating strategy used to identify CD4+PD1+, CD4+PD1+LAG3+, CD4+FOXP3+, CD8+PD1+, and CD8+CD160+ populations within the CD3+ gate. Abbreviations: FSC, forward scatter; FOXP3, forkhead box P3; LAG3, lymphocyte activation gene-3; PD1, programmed cell death protein-1; SSC, side scatter.

Statistical Analysis

Χ2 and time to parasitemia analyses (log-rank test) were performed using GraphPad Prism version 7.00 for Windows.

T-cell data were expressed as fold change from baseline and assessed for extreme outlying observations, defined as observations in excess of 3 standard deviations above the mean for that T-cell subset. None of the observations for CD4+PD1+, CD4+PD1+ LAG3+, or CD4+FOXP3+ were in excess of this cleaning cutoff.

Fold change of each variable was compared between the groups at each individual time point and analyzed using the nonparametric Kruskal-Wallis test. All P values were 2-sided and P values <.05 were considered to be statistically significant.

Generalized estimating equations (GEE) were used to model changes in T-cell subsets between the SMC+ and SMC− groups over the whole season. Univariate analysis was conducted for the following T-cell subsets: CD4+PD1+, CD4+PD1+LAG3+, CD4+FOXP3+, CD8+PD1+, CD8+CD160+, CD8+CD160+PD1+, and CD8+CD160PD1+ (Supplementary Table 1). Multivariate models were conducted for CD4+PD1+, CD4+PD1+LAG3+, and CD4+FOXP3+ with predictors for age, sex, Hb type, and blood group (Table 2). These analyses were done using SAS version 9.4.

Table 2.

Generalized Estimating Equation Output for Multivariate Models

T cellParametersβ EstimateSE95% CIP
Total
CD4+PD1+
QIC = 563.36
Treatment−0.26710.0681(−0.4006 to −0.1336)<.0001
Age−0.00160.0023(−0.0060 to 0.0028).4743
Sex (Female)−0.04530.0603(−0.1635 to 0.0728).4523
Hb (AA)0.09680.069(−0.0385 to 0.2321).1609
Blood type (O)−0.09590.0654(−0.2240 to 0.0323).1425
CD4+PD1+LAG3+
QIC = 551.4030
Treatment−1.47250.7264(−2.8962 to 0.0489).0426
Age−0.02070.0183(−0.0565 to 0.0151).2565
Sex (Female)−0.05010.5739(−1.1751 to 1.0748).9304
Hb (AA)0.7180.603(−0.4639 to 1.8999).2338
Blood type (O)−0.80970.6298(−2.0441 to 0.4247).1986
Total CD4+FOXP3+
QIC = 535.2059
Treatment0.26280.0828(0.1005 to 0.4252).0015
Age−0.0030.0029(−0.0086 to 0.0027).3075
Sex (Female)0.02260.0747(−0.0086 to 0.1689).7626
Hb (AA)0.09660.0894(−0.0787 to 0.2719).2804
Blood type (O)−0.03940.0834(−0.2027 to 0.1240).6367
T cellParametersβ EstimateSE95% CIP
Total
CD4+PD1+
QIC = 563.36
Treatment−0.26710.0681(−0.4006 to −0.1336)<.0001
Age−0.00160.0023(−0.0060 to 0.0028).4743
Sex (Female)−0.04530.0603(−0.1635 to 0.0728).4523
Hb (AA)0.09680.069(−0.0385 to 0.2321).1609
Blood type (O)−0.09590.0654(−0.2240 to 0.0323).1425
CD4+PD1+LAG3+
QIC = 551.4030
Treatment−1.47250.7264(−2.8962 to 0.0489).0426
Age−0.02070.0183(−0.0565 to 0.0151).2565
Sex (Female)−0.05010.5739(−1.1751 to 1.0748).9304
Hb (AA)0.7180.603(−0.4639 to 1.8999).2338
Blood type (O)−0.80970.6298(−2.0441 to 0.4247).1986
Total CD4+FOXP3+
QIC = 535.2059
Treatment0.26280.0828(0.1005 to 0.4252).0015
Age−0.0030.0029(−0.0086 to 0.0027).3075
Sex (Female)0.02260.0747(−0.0086 to 0.1689).7626
Hb (AA)0.09660.0894(−0.0787 to 0.2719).2804
Blood type (O)−0.03940.0834(−0.2027 to 0.1240).6367

Abbreviations: CI, confidence interval; Hb, hemoglobin; QIC, Quasilikelihood Information Criteria; SE, standard error.

Table 2.

Generalized Estimating Equation Output for Multivariate Models

T cellParametersβ EstimateSE95% CIP
Total
CD4+PD1+
QIC = 563.36
Treatment−0.26710.0681(−0.4006 to −0.1336)<.0001
Age−0.00160.0023(−0.0060 to 0.0028).4743
Sex (Female)−0.04530.0603(−0.1635 to 0.0728).4523
Hb (AA)0.09680.069(−0.0385 to 0.2321).1609
Blood type (O)−0.09590.0654(−0.2240 to 0.0323).1425
CD4+PD1+LAG3+
QIC = 551.4030
Treatment−1.47250.7264(−2.8962 to 0.0489).0426
Age−0.02070.0183(−0.0565 to 0.0151).2565
Sex (Female)−0.05010.5739(−1.1751 to 1.0748).9304
Hb (AA)0.7180.603(−0.4639 to 1.8999).2338
Blood type (O)−0.80970.6298(−2.0441 to 0.4247).1986
Total CD4+FOXP3+
QIC = 535.2059
Treatment0.26280.0828(0.1005 to 0.4252).0015
Age−0.0030.0029(−0.0086 to 0.0027).3075
Sex (Female)0.02260.0747(−0.0086 to 0.1689).7626
Hb (AA)0.09660.0894(−0.0787 to 0.2719).2804
Blood type (O)−0.03940.0834(−0.2027 to 0.1240).6367
T cellParametersβ EstimateSE95% CIP
Total
CD4+PD1+
QIC = 563.36
Treatment−0.26710.0681(−0.4006 to −0.1336)<.0001
Age−0.00160.0023(−0.0060 to 0.0028).4743
Sex (Female)−0.04530.0603(−0.1635 to 0.0728).4523
Hb (AA)0.09680.069(−0.0385 to 0.2321).1609
Blood type (O)−0.09590.0654(−0.2240 to 0.0323).1425
CD4+PD1+LAG3+
QIC = 551.4030
Treatment−1.47250.7264(−2.8962 to 0.0489).0426
Age−0.02070.0183(−0.0565 to 0.0151).2565
Sex (Female)−0.05010.5739(−1.1751 to 1.0748).9304
Hb (AA)0.7180.603(−0.4639 to 1.8999).2338
Blood type (O)−0.80970.6298(−2.0441 to 0.4247).1986
Total CD4+FOXP3+
QIC = 535.2059
Treatment0.26280.0828(0.1005 to 0.4252).0015
Age−0.0030.0029(−0.0086 to 0.0027).3075
Sex (Female)0.02260.0747(−0.0086 to 0.1689).7626
Hb (AA)0.09660.0894(−0.0787 to 0.2719).2804
Blood type (O)−0.03940.0834(−0.2027 to 0.1240).6367

Abbreviations: CI, confidence interval; Hb, hemoglobin; QIC, Quasilikelihood Information Criteria; SE, standard error.

RESULTS

Baseline Characteristics and Retention of Study Subjects

Fifty children from Beneko (SMC+) and 50 children from Ferekorobo (SMC−) villages were enrolled in the study that started in August 2015 (month 0), and baseline measurements were taken before SMC administration in Beneko. There was no difference in sex, age, or Hb variants in children recruited to the study between the SMC+ and SMC− groups. There was a greater number of blood group A children in the SMC+ versus SMC− group, while the SMC− group had a greater number of blood group O children (Table 1). Data from 3 volunteers in the SMC− group who only visited the clinic twice during the study period were excluded from the longitudinal analysis (Supplementary Figure 1). SMC treatment was well tolerated in the study population with only 7 reported instances of vomiting and only 1 of these was graded as a mild adverse event, from the 150 doses administered over the study period. Only 1 dose was considered unadministered as the child vomited just after receiving SMC as well as the replacement dose.

Comparison of Infections and Clinical Malaria in the SMC+ and SMCGroups

Malaria transmission in Mali is highly seasonal and its onset coincides with the rainy season. Over this season, the frequency of malaria infections was significantly lower in children in the SMC+ group compared to the SMC− group (63 versus 97, X2P < .001; Table 1 and Figure 2A). Similarly, the proportion of clinical malaria was significantly lower in the children in the SMC+ compared to the SMC− group (Figure 2B). Furthermore, the SMC+ group had a significantly greater time to first infection than the SMC− group (89 vs 31 days median time to infection, P < .0001 log-rank test; Figure 3A). Time to clinical malaria was also significantly increased in the SMC+ children (P < .0001 log-rank test; Figure 3B).

Proportion of malaria infections (A) and clinical malaria episodes (B) during the follow-up months in the children enrolled in the SMC+ and SMC− groups. Differences between the groups were evaluated using the X2 test. Abbreviation: SMC, seasonal malaria chemoprevention.
Figure 2.

Proportion of malaria infections (A) and clinical malaria episodes (B) during the follow-up months in the children enrolled in the SMC+ and SMC− groups. Differences between the groups were evaluated using the X2 test. Abbreviation: SMC, seasonal malaria chemoprevention.

Comparison of (A) time to first positive blood smear and (B) time to first clinical malaria infection in children in the SMC+)and control group, SMC−. Data were analyzed using the log-rank test. Abbreviation: SMC, seasonal malaria chemoprevention.
Figure 3.

Comparison of (A) time to first positive blood smear and (B) time to first clinical malaria infection in children in the SMC+)and control group, SMC−. Data were analyzed using the log-rank test. Abbreviation: SMC, seasonal malaria chemoprevention.

Reduction in CD4+PD1+ and CD4+PD1+LAG3+ T Cells in Children Receiving SMC

Ex vivo whole-blood staining was used to enumerate the percentage of CD4 and CD8 T cells expressing PD1, LAG3, FOXP3, and CD160 in all children and at each visit during the study period. There was no significant difference observed in CD8 T cells expressing PD1 at any time point (Supplementary Figure 2 and Supplementary Table 1) between the SMC+ and SMC− groups, while LAG3 and FOXP3 expression was only seen in CD4 T cells. Hence, the subsequent analysis focused on the CD4 T-cell compartment.

In the SMC− group, fold change from baseline in CD4+PD1+ T cells increased 1 month postenrollment followed by a gradual decrease in fold change. In the SMC+ group, CD4+PD1+ T-cells level remained similar 1 month postenrollment followed by a sharp decrease with a fold change of 0.5 after 2 doses of SMC.

Differences between SMC+ and SMC− groups achieved significance in month 2 and 3 (P < .0001 and .0001, respectively, Kruskal-Wallis test). Overall, during the study period the change in CD4+PD1+ T cells was 0.24-fold lower in the SMC+ group compared to the SMC− group (confidence interval [CI], 0.11–0.37; P = .0005; Figure 4A and Supplementary Table 1). A multivariate analysis was performed using fold change in CD4+PD1+ T-cell levels relative to baseline as the dependent variable and adjusted for age, sex, Hb type, and blood group. The point estimate (β coefficient) was −0.27 (CI, −0.40 to 0.13; P < .0001) when comparing the SMC+ to the SMC− group over the course of the study (Table 2).

Mean fold change from baseline of (A) CD4+PD1+ and (B) CD4+PD1+LAG3+ during the study period in the SMC+ and SMC− groups. Data are represented as mean fold change and error bars denote the 95% confidence interval. Generalized estimating equations were used to analyze differences between the groups. Abbreviations: LAG3, lymphocyte activation gene-3; PD1, programmed cell death protein-1; SMC, seasonal malaria chemoprevention.
Figure 4.

Mean fold change from baseline of (A) CD4+PD1+ and (B) CD4+PD1+LAG3+ during the study period in the SMC+ and SMC− groups. Data are represented as mean fold change and error bars denote the 95% confidence interval. Generalized estimating equations were used to analyze differences between the groups. Abbreviations: LAG3, lymphocyte activation gene-3; PD1, programmed cell death protein-1; SMC, seasonal malaria chemoprevention.

Fold change from baseline in CD4+PD1+LAG3+ T cells increased in both SMC+ and SMC− groups 1 month postenrollment but to a greater degree in the SMC− group with a mean fold change of 4.0 and 1.8 in SMC− and SMC+ groups, respectively. Although throughout the season fold change in CD4+PD1+LAG3+ T cells was higher in the SMC− compared to the SMC+ group, differences between the 2 groups achieved significance only in month 2 (P = .0389 Kruskal-Wallis test; Figure 4B). A multivariate analysis was performed using fold change in CD4+PD1+LAG3+ T-cell levels relative to baseline as the dependent variable and adjusted for age, sex, Hb type, and blood group. The point estimate was –1.47 (CI, −2.9 to 0.05; P < .04) when comparing the SMC+ to the SMC− group over the course of the study (Table 2).

Increase in CD4+FOXP3+T Cells in the SMC+ Versus the SMCGroup

Tregs, defined as CD4 T cells that coexpress the transcription factor FOXP3, can be induced during P. falciparum infections [16, 21]. In the SMC+ group, Tregs increased from baseline levels at each monthly interval, peaking at a mean fold increase of 1.53 by the end of the malaria transmission season in month 4. In contrast, in the SMC− group, Tregs decreased from baseline levels in months 1 and 2, spiked in month 3, and were 0.6-fold lower at the end of the malaria transmission season in month 4. Differences between SMC+ and SMC− groups achieved significance in months 2 and 4 (P < .0003 and <.0001, respectively, Kruskal-Wallis test; Figure 5). In a univariate analysis using fold change of CD4+FOXP3+ T cells from baseline, the point estimate was 0.37-fold (CI, 0.17–0.57; P = .0004) when comparing the SMC+ and SMC− groups (Supplementary Table 1). In a multivariate GEE analysis using fold change of CD4+FOXP3+ T cells from baseline, that adjusted for age, sex, Hb type, and blood group, the point estimate was 0.26 (CI, 0.1–0.43; P = .0015) when comparing the SMC+ to the SMC− group (Table 2).

Mean fold change from baseline of CD4+FOXP3+ during the study period in the SMC+ and SMC− groups. Error bars represent the 95% confidence intervals. Generalized estimating equations were used to analyze differences between the groups. Abbreviations: FOXP3, forkhead box P3; SMC, seasonal malaria chemoprevention
Figure 5.

Mean fold change from baseline of CD4+FOXP3+ during the study period in the SMC+ and SMC− groups. Error bars represent the 95% confidence intervals. Generalized estimating equations were used to analyze differences between the groups. Abbreviations: FOXP3, forkhead box P3; SMC, seasonal malaria chemoprevention

DISCUSSION

This study capitalized on the fact that SMC was rolled out at different times in 2 neighboring villages, in the health district center of Ouelessebougou in Mali, to compare the impact of P. falciparum infection on immune exhaustion and T regulatory markers. SMC administration in children resulted in a 37% reduction of malaria infections and almost halved the number of clinical cases. These results are consistent with previous studies that similarly showed a dramatic reduction in malaria cases in children receiving SMC [3, 22–25]. While these results were encouraging, almost half of children receiving SMC were still infected despite receiving prophylaxis, highlighting the need for multipronged approaches to achieve the goal of malaria eradication.

Malaria infections have been linked with immune dysfunction that results in greater susceptibility to bacterial infections [26] and loss of vaccine-induced immunity [27]. Malaria-induced upregulation of PD1 on CD4 T cells is well established [11, 12, 15]. The design of the current study allowed us to measure the impact of SMC on malaria-induced upregulation of PD1 and LAG3 on CD4 and CD8 T cells. Over the course of the study, children receiving SMC had lower increases from baseline of CD4+PD1+ and CD4+PD1+LAG3+ T cells compared to those in the control group. There were no differences observed in CD8+PD1+ T cells between the 2 groups. These findings suggest that malaria infections had a more pronounced effect on CD4 T cells than CD8 T cells. A potential caveat to the interpretation of this finding is the fact that γδ T cells can also express CD8 and PD1 [28–30] but they were not evaluated during the study due to technical constraints. Recent studies have shown that malaria exposure results in expansion of CD4+PD1+ T cells that coexpress another inhibitory ligand, cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), associated with high interleukin-10 (IL-10) production [31, 32]. Furthermore, malaria-induced PD1 upregulation has been linked with alterations in the memory B-cell compartment [12]. While the expression of CTLA-4, IL-10, or memory B cells was not measured in this study, it could be hypothesized that children receiving SMC would have lower levels of circulating IL-10 and preserved levels of memory B cells compared to the control group.

The frequency of Tregs, defined as CD4 T cells that coexpress FOXP3, was evaluated here. The suppressive activity of Tregs during a malaria infection may prevent hyperimmune activation but could result in a failure to generate immune memory that protects from future infections [18]. Furthermore, the dynamics of Tregs during malaria infection have been reported to be higher, lower, or unchanged [16, 33]. In the current study, children receiving SMC had a gradual increase in Tregs over the course of the season, while in the control group Tregs declined over the first 2 months. The sudden increase in Treg levels in the fourth month of the study was unexpected and the cause unknown. Unlike the CD4+PD1+ T-cell subset that was almost identical in the 2 groups at the end of the study, Treg levels were markedly higher in the SMC+ group compared to the SMC− group at the end of the malaria transmission season. These results suggest that Tregs are induced in P. falciparum-exposed children but not in children that develop active infections. In animal models, Tregs generated during malaria infections have been shown to impede both early and long-term responses [18]. These findings also raise the concern about whether children receiving SMC may have delayed acquisition of clinical immunity against malaria. These questions will be important to address as SMC coverage is expanded across the Sahel region.

While the current study highlights some important changes in T cells, there were some limitations. The study relied on phenotypic characterization of markers associated with immune exhaustion and regulation on CD4 and CD8 T cells. The current findings can serve as a primer for further functional characterization of specific immune subsets on specific P. falciparum immunity in these children. Future studies have been designed to evaluate these same markers throughout the dry season and the following transmission season in children as well as in adults living in Mali.

Overall, results show that introduction of SMC led to a reduction in CD4+PD1+ and an increase in CD4+FOXP3+ Tregs over the course of a malaria transmission season. These findings underscore the need to reduce malaria transmission and potential immune dysfunction in children living in endemic countries.

Supplementary Data

Supplementary materials are available at The Journal of Infectious Diseases online. Consisting of data provided by the authors to benefit the reader, the posted materials are not copyedited and are the sole responsibility of the authors, so questions or comments should be addressed to the corresponding author.

Notes

Presented in part: American Society of Tropical Medicine and Hygiene 65th Annual Meeting, 13–17 November 2016, Atlanta, Georgia.

Acknowledgment. The authors thank J. Patrick Gorres for assistance in writing and manuscript preparation.

Financial support. This work was supported by the Intramural Research Program of the National Institute of Allergy and Infectious Diseases, National Institutes of Health (ZIA AI001147-09) as well as the Bill and Melinda Gates Foundation (OPP1134361).

Potential conflicts of interest. All authors: No reported conflicts of interest. All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed.

References

1.

World Health Organization
.
World Malaria report 2018
.
Geneva
:
WHO
,
2018
.

2.

World Health Organization
.
Seasonal malaria chemoprevention with sulfadoxine-pyrimethamine plus amodiaquine in children: a field guide
.
Geneva
:
WHO
,
2013
.

3.

Dicko
A
,
Diallo
AI
,
Tembine
I
, et al.
Intermittent preventive treatment of malaria provides substantial protection against malaria in children already protected by an insecticide-treated bednet in Mali: a randomised, double-blind, placebo-controlled trial
.
PLoS Med
2011
;
8
:
e1000407
.

4.

Tran
TM
,
Li
S
,
Doumbo
S
, et al.
An intensive longitudinal cohort study of Malian children and adults reveals no evidence of acquired immunity to Plasmodium falciparum infection
.
Clin Infect Dis
2013
;
57
:
40
7
.

5.

Langhorne
J
,
Ndungu
FM
,
Sponaas
AM
,
Marsh
K
.
Immunity to malaria: more questions than answers
.
Nat Immunol
2008
;
9
:
725
32
.

6.

Portugal
S
,
Carret
C
,
Recker
M
, et al.
Host-mediated regulation of superinfection in malaria
.
Nat Med
2011
;
17
:
732
7
.

7.

Collins
WE
,
Jeffery
GM
.
A retrospective examination of secondary sporozoite- and trophozoite-induced infections with Plasmodium falciparum: development of parasitologic and clinical immunity following secondary infection
.
Am J Trop Med Hyg
1999
;
61
:
20
35
.

8.

Crompton
PD
,
Moebius
J
,
Portugal
S
, et al.
Malaria immunity in man and mosquito: insights into unsolved mysteries of a deadly infectious disease
.
Annu Rev Immunol
2014
;
32
:
157
87
.

9.

Dodoo
D
,
Omer
FM
,
Todd
J
,
Akanmori
BD
,
Koram
KA
,
Riley
EM
.
Absolute levels and ratios of proinflammatory and anti-inflammatory cytokine production in vitro predict clinical immunity to Plasmodium falciparum malaria
.
J Infect Dis
2002
;
185
:
971
9
.

10.

Jagannathan
P
,
Eccles-James
I
,
Bowen
K
, et al.
IFNγ/IL-10 co-producing cells dominate the CD4 response to malaria in highly exposed children
.
PLoS Pathog
2014
;
10
:
e1003864
.

11.

Butler
NS
,
Moebius
J
,
Pewe
LL
, et al.
Therapeutic blockade of PD-L1 and LAG-3 rapidly clears established blood-stage Plasmodium infection
.
Nat Immunol
2011
;
13
:
188
95
.

12.

Illingworth
J
,
Butler
NS
,
Roetynck
S
, et al.
Chronic exposure to Plasmodium falciparum is associated with phenotypic evidence of B and T cell exhaustion
.
J Immunol
2013
;
190
:
1038
47
.

13.

Liu
XQ
,
Stacey
KJ
,
Horne-Debets
JM
, et al.
Malaria infection alters the expression of B-cell activating factor resulting in diminished memory antibody responses and survival
.
Eur J Immunol
2012
;
42
:
3291
301
.

14.

Karunarathne
DS
,
Horne-Debets
JM
,
Huang
JX
, et al.
Programmed death-1 ligand 2-mediated regulation of the PD-L1 to PD-1 axis is essential for establishing CD4(+) T cell immunity
.
Immunity
2016
;
45
:
333
45
.

15.

de Jong
SE
,
Asscher
VER
,
Wammes
LJ
, et al.
Longitudinal study of changes in γδ T cells and CD4+ T cells upon asymptomatic malaria infection in Indonesian children
.
Sci Rep
2017
;
7
:
8844
.

16.

Finney
OC
,
Nwakanma
D
,
Conway
DJ
,
Walther
M
,
Riley
EM
.
Homeostatic regulation of T effector to Treg ratios in an area of seasonal malaria transmission
.
Eur J Immunol
2009
;
39
:
1288
300
.

17.

Ndiaye
M
,
Sylla
K
,
Sow
D
, et al.
Potential impact of seasonal malaria chemoprevention on the acquisition of antibodies against glutamate-rich protein and apical membrane antigen 1 in children living in Southern Senegal
.
Am J Trop Med Hyg
2015
;
93
:
798
800
.

18.

Kurup
SP
,
Obeng-Adjei
N
,
Anthony
SM
, et al.
Regulatory T cells impede acute and long-term immunity to blood-stage malaria through CTLA-4
.
Nat Med
2017
;
23
:
1220
5
.

19.

Mahamar
A
,
Issiaka
D
,
Barry
A
, et al.
Effect of seasonal malaria chemoprevention on the acquisition of antibodies to Plasmodium falciparum antigens in Ouelessebougou, Mali
.
Malar J
2017
;
16
:
289
.

20.

Severe malaria
.
Trop Med Int Health
2014
;
19
(
Suppl 1
):
7
131
.

21.

Walther
M
,
Tongren
JE
,
Andrews
L
, et al.
Upregulation of TGF-beta, FOXP3, and CD4+CD25+ regulatory T cells correlates with more rapid parasite growth in human malaria infection
.
Immunity
2005
;
23
:
287
96
.

22.

Cissé
B
,
Sokhna
C
,
Boulanger
D
, et al.
Seasonal intermittent preventive treatment with artesunate and sulfadoxine-pyrimethamine for prevention of malaria in Senegalese children: a randomised, placebo-controlled, double-blind trial
.
Lancet
2006
;
367
:
659
67
.

23.

Diawara
F
,
Steinhardt
LC
,
Mahamar
A
, et al.
Measuring the impact of seasonal malaria chemoprevention as part of routine malaria control in Kita, Mali
.
Malar J
2017
;
16
:
325
.

24.

Konaté
AT
,
Yaro
JB
,
Ouédraogo
AZ
, et al.
Intermittent preventive treatment of malaria provides substantial protection against malaria in children already protected by an insecticide-treated bednet in Burkina Faso: a randomised, double-blind, placebo-controlled trial
.
PLoS Med
2011
;
8
:
e1000408
.

25.

Wilson
AL
;
IPTc Taskforce
.
A systematic review and meta-analysis of the efficacy and safety of intermittent preventive treatment of malaria in children (IPTc)
.
PLoS One
2011
;
6
:
e16976
.

26.

Scott
JA
,
Berkley
JA
,
Mwangi
I
, et al.
Relation between falciparum malaria and bacteraemia in Kenyan children: a population-based, case-control study and a longitudinal study
.
Lancet
2011
;
378
:
1316
23
.

27.

Mooney
JP
,
Lee
SJ
,
Lokken
KL
, et al.
Transient loss of protection afforded by a live attenuated non-typhoidal Salmonella vaccine in mice co-infected with malaria
.
PLoS Negl Trop Dis
2015
;
9
:
e0004027
.

28.

Hayday
A
,
Gibbons
D
.
Regulated T-cell development: a victim of multiple conspiracies
.
Immunology
2001
;
104
:
8
10
.

29.

Hsu
H
,
Boudova
S
,
Mvula
G
, et al.
Prolonged PD1 expression on neonatal Vδ2 lymphocytes dampens proinflammatory responses: role of epigenetic regulation
.
J Immunol
2016
;
197
:
1884
92
.

30.

Iwasaki
M
,
Tanaka
Y
,
Kobayashi
H
, et al.
Expression and function of PD-1 in human γδ T cells that recognize phosphoantigens
.
Eur J Immunol
2011
;
41
:
345
55
.

31.

Mackroth
MS
,
Abel
A
,
Steeg
C
,
Schulze Zur Wiesch
J
,
Jacobs
T
.
Acute malaria induces PD1+CTLA4+ effector T cells with cell-extrinsic suppressor function
.
PLoS Pathog
2016
;
12
:
e1005909
.

32.

Abel
A
,
Steeg
C
,
Aminkiah
F
, et al.
Differential expression pattern of co-inhibitory molecules on CD4+ T cells in uncomplicated versus complicated malaria
.
Sci Rep
2018
;
8
:
4789
.

33.

Boyle
MJ
,
Jagannathan
P
,
Farrington
LA
, et al.
Decline of FoxP3+ regulatory CD4 T cells in peripheral blood of children heavily exposed to malaria
.
PLoS Pathog
2015
;
11
:
e1005041
.

This work is written by (a) US Government employee(s) and is in the public domain in the US.